Konstantinos Beis - Antibodies + Drug Design Flashcards

You may prefer our related Brainscape-certified flashcards:
1
Q

What are antibodies?

A

An immunoglobulin protein produced by the immune system as an defense against foreign agents (known as antigens) any foreign molecule, virus, bacteria etc.

Each antibody has a region that binds specifically to a particular antigen which it neutralizes – can have a variety of different targets

It is typically made up of large heavy chains and small light chains

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

What is a catalytic antibody?

A

Catalytic Antibodies (abzyme) - Antibodies that behave like enzymes

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

Generally speaking how do humans produce antibodies? What are the two main systems at play?

A
  1. Humoral (antibody-mediated) immune system - antibodies produced in response to freely circulating pathogens - direct recognition of antigen by B cells which produces plasma cells (produce antibody) and memory cells
  2. Cellular (cell-mediated) immune system – antibodies produced in response to intracellular pathogens –> cell exposes antigen on surface (MHC – membrane protein complexed with antigen), which is recognized by T-Cell which activates/helps transform B cells into antibody providing plasma cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are the different immunoglobulin/antibody classifications?

A

5 different groups – IgG, IgA, IgM, IgD, IgE

Fakts:

  • IgM - is the first antibody to appear in response to initial exposure to antigen
  • IgE associated with allergic response
  • They exist in different states – monomers, dimer, pentamer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Outline the general structure of the IgG protein

A

IgG structure – Best characterized

Characteristic Y-shape – usually homodimeric - the two monomers are connected by disulphide bridges which connect the heavy chains together

Note - Disulphide bridges also exist between the Heavy and Light chain

Each monomer consists of a Heavy and Light chain

a) Heavy Chain – High Mw
b) Light Chain – Low Mw

Antibody is heavily glycosylated – meaning that the Fc region can itself initiate a immune response but this is not common

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

What are the different regions within the immunoglobulins e.g. IgG?

A

Constant region (denoted by C) – high degree of A.A. conservation

Hyper/Variable region (Denoted by V & HV) - when antigens bind + lower A.A. Conservation

The antibody can be further divided into the Fab and FC region

FC - easily crystallized

Fab - Antigen binding site

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

Breakdown the different parts of the IgG light chain

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

Breakdown the different parts of the IgG Heavy chain

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

Why is IgG normally used in the formation of catalytic antibodies?

A

Easily modified and the most common immunoglobulin, which is distributed between the blood and extravascular fluid.

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

What is the structure of the Immunoglobulin fold?

A

Two Beta sheets (built from anti-parallel Beta-strands) that are sandwiched together

Image - Loop region holds together the Variable and constant chain

In the variable domain you can see blue loop regions corresponds to the amino acid sequence that varies, providing specificity –> These loops are known as the complementary-determining regions (CDR) or Hypervariable Loops

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

How many CDRs/hyper-variable loops do the light and heavy chains have?

A

Each Light and Heavy chain variable domain will have 3 CDRs or hypervariable regions – CDR L1-3 or H1-3

All these CDR loops are all used to recognize an epitope/antigen

Across antibody species there is no conserved conformation within these regions –> Insertions, deletions and Amino acid sequence differ.

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

What does the following crystal structure show?

A

Example Crystal Structure of a Fab

Shows CDR H3 from the Heavy chain which extends significantly out – stabilized by a range of hydrogen bonds

Note - for high specificity we normally get the contribution of both Heavy and light chains CDRs

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

Why do we want to use catalytic antibodies and why do we not just use enzymes?

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

On a chemical level whats the benefit of using catalytic antibodies/abzymes?

A

Abzymes – Use antibodies to catalyze reactions

  1. Already present in body – doesn’t produce an immune response
  2. High affinity & specificity –> favours desired reaction coupled with less side reactions/effects

How do they act?

  • They have structural complementarity for the transition state –> reduce the activation energy to reach TS
  • Consequence? - strong binding of TS with high association constant, enhances the rate of reaction
  • Abzymes also reduce rotational entropy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is a problem associated with using an catalytic antibody that has a high specificity for the subtrate?

A

High affinity for substrate, hence it will help stabilize the Enzyme substrate complex (ES) (lower in energy) – increasing the energetic barrier to reach the TS.

Solution - We select antibodies for our transition state - Helps to stabilizes that rather than ES

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

Definition of antigen and hapten?

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

What is Cocaine?

A

Alkaloid from the coca plant

Cocaine acts as a serotonin–norepinephrine–dopamine reuptake inhibitor –> leads to increased extracellular concentrations of these neurotransmitters – keeps the neurons firing

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

How does cocaine acts as a serotonin–norepinephrine–dopamine reuptake inhibitor in the brain?

A

Normal Processing

  1. Dopamine released by vesicle into the synaptic cleft
  2. Binds to the dopamine receptor on the post-synaptic membrane – inducing a signal
  3. Dopamine is released
  4. Re-uptake transporter transports dopamine back into the pre-synaptic neuron where it is degraded by MAO to form monoamines which are no longer active

Cocaine action

  • Cocaine Binds to the dopamine re-uptake transporter –> preventing dopamine reuptake. Hence, it remains in the synaptic cleft where it can continuously stimulates the action of dopamine receptor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Outline how cocaine is normally degraded in the body

A

Cocaine Degradation –> Slow process performed by enzymes in the body (can be degraded prior to crossing BBB) - involves hydrolysis the benzoyl ester and the methyl ester

Different methods of degradation normally found in the body

  1. Enzyme found in the N.S. (Butyrylcholinesterase) – forms ecgonine methyl ester + Benzoic acid –> benzoyl ester hydrolysis
  2. Enzyme found in liver (liver carboxylesterase) –> methyl ester hydrolysis forming forming benzoylecgonine + methanol
  3. Enzyme found in the N.S –> Demethylation by N-demethylase followed by Butyrylcholinesterase action (benzoyl ester hydrolysis) to form Norecgonine methyl ester + benzoic acid

All of the products from Cocaine breakdown are harmless  which can be secreted out of the body

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

What is the role of Butyrylcholinesterase?

A

Butyrylcholinesterase (BChE) – Main enzyme involved in degradation

BChE hydrolyses butyrylcholine BUT It can also hydrolyse toxic compounds that contain an ester – Cocaine is one as well

Human esterases are slow to degrade cocaine

  • > 11 isoforms found in liver, brain, heart
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Outline Butyrylcholinestarase mechanism of action on butyrylcholine.

A

Acid-based catalysis with a glutamate, Histidine and serine

  1. Glutamate hydrogen bonds the histidine, allowing histidine to act as base to remove serine -OH hydrogen - driving nucleophilic attack to the ester bond (Carbonyl carbon)
  2. Formation of Transition state
  3. Collapse of the negative charges - histidine is now acting as a acid (donating it’s hydrogen)
  4. Incoming water displace choline
  5. Histidine acts a base removing a proton from water, allowing for the -OH to nucleophilically attack the carbonyl
  6. 2nd Tetrahedral intermediate formed followed by it’s collapse (collapse of negative charge)
  7. Release of the final product & active site returned to starting state
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Outline mechanism of Butyrylcholinestarase degradation of cocaine - general sequence of events

A
  1. Activation of serine to perform initial nucleophilic attack on carbonyl carbon
  2. Tetrahedral intermediate
  3. Collapse of negative charges
  4. Release of Ecgonine
  5. Incoming water molecule – activated by histidine
  6. -OH nucleophilically attack modified serine
  7. Tetrahedral intermediate
  8. Collapse of negative charge
  9. Release of benzoic acid a regeneration of active site
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

When designing the catalytic Abs against cocaine, what are some things to think about/take into consideration? What are we doing in practise?

A

Objective –> Ab recognizes, binds, and catalyzes cocaine. The products must be released so the Ab is recycled and free (unlike naturally occurring antibodies)

What do we have to do in practise?

  1. Predict transition states, from which we create TS analogs that match TS
  2. Produce a Hapten that is stable and mimic structural and electronic properties
  3. Epitope must be large enough to bind well with Abs
  4. Design and choice of Linker and carrier protein
  5. Hapten + carrier elicits immune response producing desried Abs which stabilize the TS of the reaction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is the transition state produced in cocaine hydrolysis which we can use for Abzyme production?

A

Transition state – Tetrahedral intermediate

In order to create an antibody that targets cocaine we need an antibody that binds and stabilizes a T.S. analogue

Basically need to re-create the T.S. so that an antibody against it can be created (we can’t capture the exact molecule – educational guess)

Example of an analog for Tetrahedral intermediate is replacing the carbon in the tetrahedral intermediate to a phosphate - cannot be attack nucleophilically by H2O – stable

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What are some examples of groups that can be used to re-create tetrahedral intermediates of transition states?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Once you have identified your TS analog, what do we do next?

A

Once we have identified T.S. analog we have our Hapten which can bind an antibody

BUT haptens are too small to illicit an immune response

Thus, in order to overcome this problem we design a linker to join our hapten to a carrier protein - big enough to illicit an immune response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Things to consider when designing a linker?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Apart from the linker design and carrier type, what other thing must you consider before conjugation?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What are the two methods we use to produce our desired catalytic antibodies?

A

Once we have our hapten conjugated to a carrier protein it is time to immunize

Conventional Antibody production

Inject/immunise mice with compound –> illicits an immune response –> kill the mice and remove spleen (B-cells located) –> isolate antibodies (using antiserum?) that can catalyze the reaction

Problem – Killed source - if you are successful in obtaining an antibody you can not make more

Monoclonal Antibody Production

Immunize mice –> kill mice –> remove spleen –> mix spleen cells and myeloma cells (M.C. LACK HGPRT – important for nucleic acid synthesis) in HAT medium –> unless fusion has taken place the cells will not survive –> isolate different clones –> test and identify clones that have catalytic activity

Useful technique as we retain a source of the antibodies –> immortal cell line

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

After isolating catalytic antibodies/abzymes, what is done next?

A

Determine kinetic parameters of the antibodies - assess which antibodiy is most catalytically active

Examine parameters such as…

Km – Antibodies binding Hapten – affinity

Kcat – reaction per unit time

Kcat/ Km – catalytic efficiency at low concentration

From the parameters we can identify 15A10 as having significant activity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

After identifying succesful abzymes, how are toxicity assays performed?

A

Toxicity assays - Cocaine Toxicity in the Rat

Question we are trying to answer - Do these antibodies help with survival rates?

Examine rats’ survival after infusion of an LD90 (lethal dose to kill 90% of mice) (16 mg/kg) cocaine with abzyme

The effect of mAb 15A10 on survival was significant at -

Graph shows that at higher concentrations of mAb 15A10 there was a higher survival rate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What did the crystal structure of reveal show about the binding of cocaine and TS analog with the abzyme?

A

Both cocaine and TS analog were crystallized in presence of eitehr cocaine and TS analog –> both cases they bound in the hypervariable regions - binding to CDR loops

Interestingly, they also allowed the antibody to react with cocaine for a few minutes before capturing X-ray diffraction data and they were able to show…

The crystal structure with the product – Ecgonine methyl ester and benzoate in the active site - breakdown products of cocaine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What was the proposed mechanism by which the abzyme degraded cocaine?

A

Examining active site of antibody there was a lack of normal residues associated with acid-base catalysis – instead replaced by tyrosine residues that form hydrogen bonds

Mechanism

  1. Cocaine enters active site - stabilized by hydrogen bonds
  2. Activated Water molecule performs nucleophilic attack on carbonyl carbon
  3. Tetrahedral intermediate formed – -ive charge is stabilized by tyrosine
  4. Followed by its collapse –> release of two products

Simpler mechanism than normal acid-base

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What is a prodrug and why is it beneficial to use?

A

Prodrug – Precusor molecule is introduced into the body which only forms the active drug once it is metabolised

  • Useful as drug itself can be highly toxic itself - limit side effects
  • Able to deliver high concentrations of active drug - protects against rapid metabolism and clearance
  • Improve targetting of drug to specific regions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

How can we combine both pro-drugs and catalytic antibodies?

A

Basic idea –> is that you introduce prodrug and catalytic antibody into patient –> the catalytic antibody is targeted towards a specific tissue, hence when the prodrug enters it can become metabolized by the catalytic antibody into the active form

Benefits?

  1. Minimizes toxicity
  2. Allows for drug targeting
  3. More complex chemistry to be performed (harder to do with normal chemistry) - more verstaile tool

In this case we would have to design a abzyme against the prodrug TS –> to favour formation of active drug

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What are the limitations of catalytic Abs?

A
  1. Haptens fail to generate antibodies that catalyse the desired reaction
  2. Haptens may closely resemble the final product (stabilize it) - slow product release or inhibition – preventing the antibody for catalyzing more reactions
  3. It might be difficult to synthesize the desired hapten (chemistry)
  4. The catalytic efficiency depends on the solvent exposed binding site?
  5. Introduction of foreign antibody might elicit immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

How can we create therapeutic/catalytic Abs that don’t elicit an immune response in the host?

A

Use chimeric or humanized antibodies

  1. Chimeric consist of variable regions (VL and VH) derived from a mouse and constant regions derived from human. ~65% human
  2. Humanized therapeutic mAbs are predominantly derived from a human source except for the CDRs, which are murine. >90% human
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Apart from acting as a catalytic antibody, what other useful functions can antibodies perform?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Summary of Catalytic antibody lecture content?

A
  1. Catalytic Abs can be raised against specific haptens
  2. Catalyse desired reactions
  3. Consider different analogues, linkers and carrier proteins
  4. Catalytic Abs have the potential to treat addictions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What are antibody-drug conjugates?

A

Antibody–Drug Conjugates (ADC) –> Antibodies used to deliver a drug to a specific site

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What are the current treatments of cancer and is the recurring problem?

A

Cancers (many different types!) can be treated by:

  1. Surgery
  2. Chemotherapy – several side effects and resistance to anticancer drugs
  3. Radiotherapy
  4. Hormone therapy
  5. Combination of therapies is usually required

Recurring problem - target/impact healthy cells – leads to side effects

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Why do antibody-drug conjugates provide a useful solution to treating cancer?

A

Antibody–Drug Conjugates (ADCs) are monoclonal antibodies or antibody fragments attached to biologically active molecules through chemical linkers with labile bonds

Advantage

Deliver highly cytotoxic agents directly to tumour cells without affecting other dividing cells in the body – targeting antigens on Tumour cells

When the drug is bound to the antibody, the chemotherapeutic ‘‘payload’’ no longer circulates systemically and is therefore doesn’t negatively impact healthy cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What is the general concept behind using antibody-drug conjugates?

A

Regular antibody conjugated to a drug with a linker

Antibody binds to cell surface receptor –> internalization –> release of payload –> block cellular process in nucleus or cytosol leading to apoptosis of cancer cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What are some examples of ADCs that have already been approved?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

General mechanism, requirements and componenets of a ADC?

A

Mechanism

An ADC binds to an antigen on the surface of a cancer cell and then internalises, after which the highly cytotoxic payload molecule is released, typically by lysosomal cleavage

Requirements

ADC needs to retain the selectivity of the original monoclonal antibody (bind to the specific antigen) while being able to release the attached cytotoxic payload in concentrations high enough to kill the targeted tumour cells.

Componenets

Three key components to an ADC: the antibody (which antigen will it bind), the linker (Cleaved/non-cleaved) and the payload (Pathway you desire to inhibit?)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Outline the steps by which ADCs target cells and become internalized

A
  1. ADCs are designed to directly target and kill cancer cells, and so the antibody has to be able to recognise and bind to its corresponding antigen localized on the tumour cell.
  2. Once bound to the antigen, the entire antigen–ADC complex is then internalized through receptor-mediated endocytosis.
  3. The internalization process proceeds with the formation of a clathrin-coated early endosome, containing the ADC–antigen complex –> release of the ADC from the receptor which is recycled
  4. Once inside the lysosome, the ADC is degraded and free cytotoxic payload released into the cell, leading to cell death.
  5. The mechanism of cell death will depend on the type of cytotoxic payload.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Why is it important to choose a payload that is sufficiently cytotoxic?

A

Another crucial requirement is to ensure that a sufficient concentration of payload reaches the interior of the cancer cells to guarantee their death

It is estimated that, even if the overall mechanism of action of an ADC works at an efficiency of 50%, only 1–2% of the administered payload will reach the tumour cells - a lot of it will be degraded

Therefore, it is important that the chosen payload is sufficiently cytotoxic to exert an effect at very low concentrations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

Benefits of using ADCs relative to chemotherapy?

A
  • Wider therapeutic windown –> less likley to be toxic as there is a higher maximum tolerated dose and lower minimum effective dose
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What are the three main things to consider when designing an ADC?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What are the major challenges that face researchers when designing ADCs?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

When designing a ADC linker what do we have to consider?

A
  1. Antigen Selection
  2. Cleavable linker
  3. Non-cleavable linker
  4. Site of conjugation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Why does the selection of target antigen potentially pose problems?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What is the relationship between DAR and the rate of degradation? What can we learn from this?

A

A/B) Shows relationship between the DAR and rate of degradation –> the more drugs we conjugate to the antibody – the quicker the rate of clearance (more susceptible to protease) but the more drug we attach the greater the payload –> trade-off

Hence, there is an optimal mid-point which yields that highest efficacy (DAR 3.5-4)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What are cleavable linkers? What are the different types?

A

Cleavable linkers

Cleavable linkers utilise the differences in conditions between the bloodstream and the cytoplasm within tumour cells

Take advantage of the change in environment once the ADC–antigen complex has internalized it triggers cleavage of the linker and release of the active payload

Cleavable linkers are divided into three main sub-categories:

(1) Acid-labile (e.g., hydrazones)
(2) Reducible (e.g., disulphides)
(3) Enzyme-cleavable (e.g., peptides).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

How do acid-labile linkers work? What problems are associated with it?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

How do reducible linkers work? What problems are associated with it?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

How do enzyme-cleavable linkers work? What problems are associated with it?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Apart from rely on Cathepsin, what other enzyme is used for Enzyme-cleavable linkers?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Outline the general mode of action of ADC’s with cleavable linkers, starting from antigen binding to apoptosis

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Why are non-cleavable linker used for ADCs?

A

Non-cleavable linkers – Why are they used?

  1. Drug (usually) remains attached to mAB - not effected by pH, reducing environment or enzymes
  2. In the lysosome – mAB is degraded by compound + linker is not!
  3. Increased plasma stability
  4. Lower risk of systemic toxicity due to premature release of the payload
  5. Better therapeutic window, with improved stability and tolerability
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Outline the mode of action of ADCs with non-cleavable linkers

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Example of a Non-cleavable Linker

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

When conjugating our drug to the antibody what do we have to consider?

A
  1. Prevent modifying antibody to the point where they induce an immune response
  2. DAR –> typical average DAR of 3.5 or 4
    - Species that have a DAR of more than 4 have been shown to lead to lower tolerability, higher plasma clearance rates and decreased efficacy in vivo
  3. Retaining antibody selectivity (antigen binding)

Conjugation Example:

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What are two examples of functional groups on our antibody which we tend to use for conjugation?

A
  1. Thiosuccinimide linkage, which is formed through the reaction of thiols and alkyl maleimides –> thiosuccinimide formation is slowly reversible under physiological conditions
  2. Conjugation to the terminal amines of lysine residues using an amide ester
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

How is conjugation with thiol groups performed?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What is one possible solution to thiol conjugations lack of DAR control?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

How is site-specific conjugation performed with THIOMAB technology?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

How is conjugation with lysine residues performed? What problems are associated with this form of conjugation?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What are some further potential strategies that could improve conjugation?

A
  1. Engineered Cys – Thiomab
  2. Insertion of unnatural amino acids – tight regulation of our payload with a specific amino acid that is not found naturally
  3. Enzyme-assisted ligation
  4. Glycan remodelling and glycoconjugation - remember that antibodies are glycosylated
  5. Amino‑terminal engineered serine –> problem may change overall properties of antibody
  6. Native cysteine rebridging
  7. Highly loaded ADCs at specific sites
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What is the toxic payload/warhead? What properties are we looking for? What cellular structures/organelles are we targetting?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What is the largest group of ADCs currently being trialed?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What are some other examples of drugs that are being used for ADCs?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What is the Bystander effect?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What are the 4 different forms of ADC toxicity (think good & bad)?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What do we have to remember when using antibodies (Think immune response)?

A
76
Q

Summary of things to consider with ADCs

A
77
Q

What is an example of ADC that is approved for targetting HER2-positive breast cancer treatment?

A

Market Name - Kadcyla

T-DM1 - DM1 conjugated with trastuzumab (antibody against HER2)

DM1 –> Binds tubulin to cause mitotic arrest and cell death

Research findings?

Conjugates were evaluated for in vivo pharmacokinetics and antitumor activity

In vivo studies revealed that the higher the linker stability the higher the antitumor activity

T-DM1 was approved in 2013 for HER2-positive breast cancer treatment

78
Q

Summary of ADCs

A
  1. ADCs are potent drug delivery tools
  2. Low toxicity – relative to other cancer treatments
  3. Care should be taken with linker design, payload choice
  4. Linker can be cleavable or not
  5. Bystander effect desirable or not
79
Q

Is there any particular reason why our cocaine T.S. analog is too small to elicit an immune response?

A

Remember that there is a large number of small molecules (e.g., vitamins, hormones) circulating through the body –> counterproductive for immune system to illicit an autoimmune response

80
Q

When using a Non-cleavable linker, why is more active drug compound delivered to the target?

A

Cannot no diffuse out through the membrane forcing the drug to remain in the cell + similarly it can’t diffuse into the nucleus (through the nuclear membrane) remains in the cytoplasm instead of moving out of the cell or into the nucleus – helps with the concentrating the active drug

Why?

Linker is fairly hydrophilic + increases size by 500-800 Da - repels from the hydrophobic membrane + the size inhibits free diffusion

81
Q

How do you prevent the aggregation of antibodies caused by payloads hydrophobicity?

A

Trial and error of changing DAR - how many drugs you load onto the antibody.

If we overload with hydrophobic molecules (ADC - drugs) – the antibodies will precipitate together

82
Q

What is one reason why cancer cells are often immune/resistant to chemotherapy/drugs?

A

Cancer cells have efflux protein in the membrane that can efflux the drugs –> problematic if our drug enters and does not induce apoptosis it allows our cancer cell to develop mutations (mutations that benefit cell survival) in the transporter which will allow it to be exported –> like antibiotic resistance idea

This is exacerbated by the fact that the cancer cells divide quickly facilitating this rapid evolutionary process

Thus….

making it favourable to use multiple therapeutics to maximize the effectiveness

83
Q

How to select appropriate conjugate?

A

You want to choose your conjugate so that it will be most effective at targeting the specific process that you are interested in inhibiting

For Example…

Target soluble enzyme –> you will want your conjugate drug to be hydrophilic

Target a membrane protein –> you will want your conjugate drug to be hydrophobic

Note - normally we are most interested in soluble enzymes within cancer cells.

84
Q

What happens to the drug once the cell undergoes apoptosis?

A

The drug will enter the metabolism –> The human body can detoxify these compounds – making them inactive

85
Q

How to identify target antigens on cancer cells?

A

Profiling of antigens that are expressed differentially in cancer cells relative to healthy cells –> Mass spec profiling

Also, worth looking at glycosylation – at the antigens tend to be heavily glycosylated - possible target

86
Q

Outline the different steps in the Drug Discovery proces/program?

A
87
Q

What are different way to identify new hits/drugs for a target molecule?

A
  1. High Throughput Screening (HTS) – wet lab setup - most commonly used by Pharma But…
    - Expensive
    - False positives
    - Assay variability or errors in data
  2. Virtual Screening (VS) –> main focus of lectures
  3. Combination of HTS and VS
88
Q

What are the limitations associated with HTS?

A
89
Q

What are some essential requirements when running a HTS screen?

A
  1. Expensive robotics are required to screen for candidates – large scale 107
  2. Very robust assay required – Biological activity meter
  3. Interaction analysis of successful hits - between drug and target
  4. Complement HTS with structural data and using in-silico methods
90
Q

What are the two methods that you can divide Computer-aided drug discovery (CADD) into?

A

CADD methods are classified into ligand-based methods (LBDD) and structure-based (SBDD)…

Structure-based methods require the 3D information of the target to be known - 3D conformation of target protein of interest

Ligand-based methods are used when the 3D structure of the target is not known. Rather…

They use information about the molecules that bind to the target of interest. Hits are identified, filtered and optimized to obtain potential drug candidates that will be experimentally tested in vitro - information about the substrate or inhibitor

91
Q

Difference between SBDD and LBDD?

A
92
Q

Outline the overall process of virtual HTS?

A
  1. Start with library of compounds
  2. Filter the library depending on Mw, physical properties, etc.
  3. Virtual HTS - LBDD or SBDD

LBDD - focus of lecture

  1. Searching for similarity in library towards ligand (known) of interest – identify hits
  2. Synthesize or purchase hits
  3. Evaluate in the lab
    a) Biological activity - refine
    b) No biological activity - Start again
93
Q

What type of information is used in LBDD to filter/search library?

A

Takes into account different kinds of molecular information of the known ligand such as

  1. 2D molecular fingerprint
  2. 3D molecular shape
  3. Molecular and electronic properties
  4. 3D pharmacophore

Image shows the two different LBDD strategies

94
Q

What are the three methods that LBDD searching can be divided into?

A
  1. Similarity searching
  2. Pharmacophore mapping
  3. Machine learning
95
Q

Outline what is meant by the LBDD similarity search method

A
96
Q

What filters are commonly used for a similarity search?

A
97
Q

Outline how 2D fingerprints of molecules are created?

A

Reasoning for using bits –> facilitates processing for computer

98
Q

Outline what is meant by Hashed fingerprinting (2D property filter)? What is meant by bit collision?

A
99
Q

What is the similarity coefficient used for ranking 2D fingerprints?

A
100
Q

What is scaffold hoping?

A
101
Q

What is meant by 3D fingerprinting?

A
102
Q

What is a pharmacophore?

A

Pharmacophore is the substructure of a molecule that is responsible for its pharmacological activity –> A set of geometrical constraints between specific functional groups that enable the molecule to have biological activity

Basically, similar functional groups in similar organisation but the scaffold of the protein changes –> this is how drug companies overcome patents

In this search both valence angles and torsion angles can be included

103
Q

Explain what is going on in the attached image (hint - 3D fingerprinting)

A
104
Q

After performing 3D fingerprinting, what do we do?

A
105
Q

When performing a search in a database, what should we look out for?

A

When searching we may have reference compounds of known activity, which we search against a database that contains other actives and decoy compounds using our filters

A good similarity measure will cluster the known actives at the top of the ranking –> appropriate filters

Known active compounds not appearing in ranked list –> suggestive that we have to adjust our search parameters

106
Q

Does 3D shape search require us to know the protein structure?

A

Nope!

We assume that all (or the majority) of the known actives bind to the same location of the protein

  1. Generate our pharmacophores –> identify pharmacophoric features (hydrogen bond donors and acceptors, lipophilic groups, charges) + their geometrical arrangement of all actives that match with a low-energy conformation
  2. Databse search –> find all molecules in a database that can match it in a low-energy conformation (energetically possible)

Scaffold-hopping possible –> needs to match the pharmacophore

107
Q

What is the most common pharmacophore model?

A
108
Q

When selecting a representative set of actives what should we consider?

A
  • Most methods assume similar binding modes
  • One or more rigid molecules are preferred
  • The ligands should be diverse (otherwise too many common features that are not involved in binding)

Procedure

  1. Prepare molecules (e.g. tautomeric form, protonation state), generate 3D structure and conformations (if required)
  2. Use pharmacophore software/tool to generate pharmacophores (depending on input it may be biased or unbiased)
  3. Select preferred pharmacophore model(s) and validate them experimentally –> are they really the core functional groups in binding our protein?
109
Q

Things to consider for database matches - i.e. how do we know if we have a good match?

A
  1. Pharmacophoric features in each ligand are identified (Donors, acceptors, hydrophobic groups, etc)
  2. Ligands aligned so the corresponding features are overlaid
  3. Conformational space explored
  4. Scoring system: number of features, goodness of fit to features, conformational energy, volume of the overlay, etc
110
Q

When searching a database, what should we keep in mind about the database itself?

A

Basically, the way in which we generate our model should resemble the way in which the database was created - same feature definitions (functional elements) + protocol used to define pharmacophores –> ensures accuracy

For example,

What tolerance should we use? What distance between two pharmacophores is acceptable?

111
Q

Case study (LBDD) - Inhibition of Human Thymidylate Synthase and Dihydrofolate Reductase Enzymes

Why do we care about these two enzymes? What known substrate was used for LBDD?

A
112
Q

What is the LBDD QSAR Model?

A

Quantitative-Structure Activity Relationships (QSAR) tries to establish quantitative relationships between descriptors and the target property capable of predicting activities of novel compounds

Basically using information about the chemical behaviour of each functional element to help predict activities of the new compounds

Require descriptors that accurately convey chemically-relevant information to the machine learning models

113
Q

What is the idea behind QSAR?

A

Takeaway message –> from assays and previous knowledge (chemically behaviour), the machine learning model will predict the activity of the novel compound

After the prediction you have to test it out experimentally

114
Q

Outline the general process of SBDD

A

Key difference between LBDD and SBDD is that the structure of the protein is known –> Either structure-based methods (X-ray etc.) or homology modelling can be used to determine structure

Note - More commonly used relative to LBDD

  1. Start with Library
  2. Filter compounds
  3. Virtual HTS – Use different types of docking
  4. Score Hits
  5. Evaluate using experimental techniques
  6. Process can be repeated for optimization
115
Q

What are the three main SBDD methods?

A
116
Q

Basic principles of SBDD?

A
  • Solve the structure of the target at high resolution – X-ray or Cryo-EM (resolution is improving) or alternatively use homology modelling (known homologous proteins)
  • Solve the structure in the presence of the substrate if possible –> determining binding interaction – Alternatively, use inhibitor
  • Use this structure as a template for screening of small molecules
  • Redesign the molecules to fit and/or bind better
117
Q

What does virtual HTS entail?

A

virtual HTS - Perform docking in-silico

After In-silico screening…

  • Test activity (enzymatic and pharmacokinetics)
  • Solve crystal structures to verify binding mode
118
Q

Examples of drugs designed using SBDD

A
119
Q

Main challenges when performing SBDD?

A
  • The energetics of protein-ligand interactions are complicated
  • Both proteins and ligands can be quite flexible - dynamics adds extra complexity
  • Many target-binding ligands in-silico are not good drug candidates
  • The structures of many important drug targets are difficult to determine – Lack of structural information e.g. Many GPCRs
  • Plus structures need to be high resolution - in order to determine all the Ligand-protein interactions
120
Q

What are the criteria/parameters/descriptors used for SBDD?

A
121
Q

What are Structure activity relationsips (SAR) and how can we identify them?

A

Correlations that are constructed between the features of chemical structure in a set of candidate compounds and biological activity, such as potency, selectivity and toxicity.

Basically… Linking chemical structurs/groups to biological activity - kinda like pharmacophores

How can these biologically relevant groups be identified?

  1. X-ray crystallography can be used to identify important interactions between drug and protein
  2. Test for biological activity with modified compounds and comparing them with the original compound – modify groups and check impact on activity
    a) Modification shows a significant lower activity, then the group that has been modified must be important
    b) If the activity remains similar, then the group is not essential
122
Q

What is Lipinski’s rule of Fives?

A

SBDD follows the Lipinski’s rule of Fives - Rule(s) of thumb that were created to predict the likeliness that a drug would be orally active in humans

  1. MW less than 500 – easier diffusion across membrane + easier to modify
  2. Fewer than 5 H-bond donating groups
  3. Fewer than 10 H-bond accepting groups
  4. LogP between -1 and +5 – experimentally determined using the partition of molecule in octanol and H2O – indicates hydrophobicity
123
Q

What are the two main parts of SBDD virtual screening (docking)?

A
  1. In silico to identify potential lead compounds from a database
    - From which we score, rank and filter a set of chemical structures
  2. Pose prediction – What will the algorithm – predict the interaction at the active site
    - Requires to have a high resolution structure
    - Identify key residues in the binding site
    - Dock compound by selecting proper stereochemistry
124
Q

What does the Rigid docking search algorithm do?

A

Ligand is treated as a rigid structure and only the translational and rotational degrees of freedom are considered

Means that…

Different ligand conformations are docked separately

plus…

Protein is considered rigid

Goal - satisfy as many of the positional and chemical descriptors of the active site as possible

Process

  1. Determine plausible conformations of ligand (prior to docking) - match binding site shape –> geometrical descriptors are combined with chemical and electrostatic descriptors - needs to match shape and chemical nature of active site
  2. Orient ligand in the active site using simple geometry descriptors, like spheres or triangles –> Satisfy stereochemistry, no clashes
  3. Rank/score the different poses based on receptor affinity
125
Q

What are the two different scoring methods for different ligand docking poses (SBDD)?

A

First principle scoring – Parameters - Molecular Mechanics force field.

Force fields typically contain intra-molecular terms from the ligand: Bond lengths, Bond angles, Dihedral terms

And inter-molecular terms: Van der Waals contacts (non-polar) Electrostatic interactions (polar)

All together - Ebind = EIntra + Enonpolar + Epolar

Empirical scoring – similar to first principal score - looks at DG

Score’s ligands very rapidly

ΔGbind= ΔG0 + ΔGpolar · Σf(Complex) + ΔGnon-polar · Σf(Complex) + ΔGrot · Nrotatable-bonds

  • ΔG0, ΔGpolar, ΔGnon-polar, and ΔGrot are empirically parameterised weights
  • f(Complex) is a penalty function aimed at penalising any unfavourable interactions
126
Q

Benefit of Rigid docking?

A

Fast as it is less computationally demanding – screen many ligands quickly

127
Q

What does the flexible docking search algorithm entail?

A

Flexible docking

Ligand flexibility along torsion angles is considered in the docking process –> increased accuracy ligand accommodation in binding site

Resulting in different binding models

  • Protein is considered “rigid”- small flexibility is allowed
    a) Large protein conformational changes challenging for docking
    b) Small conformational changes can be accommodated
128
Q

Before performing flexible docking (SBDD), what do we need to consider?

A

A) Structure are required to be high resolution structure since otherwise…

  1. Water molecules not resolved
  2. Electron density for flexible side chains might be weak
  3. Hydrogen bonds are absent - 1Å or better resolution is optimal but can be artificially added at lower resolutions (less accurate)
  4. pKa of the active site side chains - crystallisation buffer
  5. Hard to distinguish protonation state and tautomeric form of the ligand

B) Torsion angles provide flexibility to the ligand but too many rotatable bonds can be a problem E.g., Taxol – very computationally demanding

129
Q

What does De Novo screening (SBDD) entail?

A

Structure based fragment screening (de novo)

De novo synthesis - examine which regions in the active site are involved in binding from which we place fragments which can be linked to create biologically active molecules

Build into the active site and link fragments to create bioactive compounds

How can we identify the fragments?

  1. Examine possible poses & conformations (search algorithm) –> Orientation of molecule in binding site - More flexible as we are dealing with smaller molecules/fragments
  2. Predicting energetics of protein-ligand binding (scoring function) - Binding affinity & Thermodynamics
130
Q

What is Lead identification by Fragment evolution (De-novo synthesis technique?

A

Lead identification by Fragment evolution

Screen for fragments - Fragment is know to bind to the active site.

The fragment acts as a building block for the construction of the lead molecule - The lead molecule is evolved by building away

131
Q

What is Lead identification by Fragment linking (De-novo synthesis technique)?

A

Lead identification by Fragment linking - identify two fragments that bind and link them together

Fragment 1 binds to one site

Fragment 2 binds to an adjacent site

Fragments joined together by a linking group that allows the lead molecule to span both sites – increasing bioactivity

132
Q

What is Lead identification by Fragment self-assembly (De-novo synthesis technique)?

A

Lead identification by fragment self-assembly

This involves…

Identifying fragments that bind to the active site and are able to self-assemble into a single molecule - two fragments bind followed by a catalytic reaction performed by the enzyme, linking the two fragments together

Note - either one (e.g. turning it into a good nucleophile) or both can be modified for the reaction to occur

The protein is used to self-select or to catalyze the synthesis of its own inhibitor without covalent attachment of the protein to the inhibitor –> linking two fragments increased binding affinity (nM/µM –> fm)

Can only be done with proteins that possess enzymatic activity

133
Q

What is the last step for any De novo synthesis technique?

A

Lead progression by fragment optimization

  • Optimize or modify properties of the lead compound
  • Re-engineered to address optimization of a particular property (e.g. selectivity, cell-based activity, oral activity or efficacy)
134
Q

Difference between fragment based approahces (building into) and High throughput screening?

A
135
Q

Why is Thymidylate synthase an enzyme of interest for drug researchers?

A

Thymidylate synthase

This function mThymidilate synthase maintains the dTMP pool, critical for DNA replication and repair.

Cancer cells express elevated levels

The enzyme has been of interest as a target for cancer chemotherapeutic agents.

136
Q

Case-study - What in-silico docking steps were performed for screening for drug canditates against Thymidylate synthase?

A
  1. Researchers attempted to find molecules that resembled the co-factor tetrahydrofolate –> X-ray structure of the TS available allowing for identification of key interactions
  2. Identified potentual lead from docking
  3. They proceeded to modify the ligand in order to attempt to increase binding affinity and water solubility –> but in-silico binding did not match X-ray - lower affinity than expected
  4. Further redesigning - introduction of amide group –> Wet-lab results and in-silico predictions matched up
137
Q

Once a lead has been identified from screening, what criteria does the potential drug need to meet?

A

Lead is validated by re-testing checking for…

  1. X-ray Structure assesment
  2. Potency: the amount of drug required for its specific effect to occur; inverse of the EC50
  3. Efficacy: the maximum strength of the effect itself, at saturating drug concentrations.
  4. Pharmacokinetics: determining the fate of xenobiotics. Extend and rate of adsorption, distribution, metabolism, and excretion (ADME).
  5. Pharmacodynamics: determining the biochemical and physiological effects of drugs, the mechanism of drug action, and the relationship between drug concentration and effect.
  6. Chemical optimization
  7. Patentability
138
Q

Examples of two HIV-protease inhibitors designed using computer modelling?

A
139
Q

What is drug metabolism?

A

Drug metabolism is the metabolic breakdown of drugs by a living organisms –> metabolic products produced are less pharmacologically active in turn reducing the toxicity of the drug

  • Liver is the main site of drug metabolism but specific drugs may undergo biotransformation in other tissues
  • Drug metabolism uses pathways that are normally used for the biosynthesis of endogenous substrates - has to be a degree of resemblance to natural compound to enter the pathway
  • Drug metabolism has to be taken into account when designing novel drugs
140
Q

Why is it important to convert lipophilic compounds hydrophilic during drug metabolism?

A

Drug metabolism is required to convert lipophilic compounds into more hydrophilic compounds –> excretion

Why?

Lipid soluble non-polar compounds remain in the blood and tissues and maintain their pharmacological effects for much longer - result in undesirable side-effects

141
Q

What does Pharmacokinetics refer to?

A

Phatmacokinetics - Drug Absorbtion, Metabolism, Distribution, and Elimination (ADME)

e.g. Orally administered drug, dissolve in the GI tract and absorbed through the gut –> enter the liver and into the blood stream/circulation

142
Q

Why are pharmacokinetics important to conisder when thinking about the drug dosage?

A

The physical and chemical properties of a drug determine its success to reach its target

Chemical stability – e.g. stable in the stomach?

Metabolic stability – e.g. no interaction with other metabolites

Successful Absorption – e.g. cross membranes

  1. Volume of distribution (reach target) and clearance influence Half-Life
  2. Clearance and Absorption will influence oral bioavailability - how much reaches the blood stream
143
Q

What are the different phases in drug metabolism?

A
144
Q

Role of Phase I and II transformations in drug metabolism?

A

Phase I and Phase II Transformations - Role of detoxifying

Phase I transformations – introduce or unmask a functional group (make inactive), e.g., by oxygenation or hydrolysis (OH, -SH, -NH2, -COOH, etc.)

These metabolites are often inactive + Can be excreted readily

Phase II transformations – generate highly polar derivatives (conjugates) for excretion; glucoronide, sulfate, acetate, amino acids (large functional added so must be excreted through the feces)

145
Q

In phase I metabolism, what is the primary form of modification?

A

1o type of modification - Oxidation

  • Addition of oxygen or removal of hydrogen –> The first and most common step involved in the drug metabolism
  • Liver is the main organ where oxidation takes place by cytochrome P450
  • Increased polarity of the oxidized products; increased water solubility and excretion in urine

Oxidation, can occur on…

  • Aliphatic or aromatic hydroxylation
  • N-, or S-oxidation
  • N-, O-, S-dealkylation – e.g. removal of methyl
146
Q

Outline the reduction reactions that take place in phase I metabolism

A

Reduction

  • Removal of oxygen or addition of hydrogen
  • Less common than oxidation
  • Cytochrome P450 system is involved in some of these reactions. Other reactions are catalyzed by reductases present in different sites within the body.

Reduction can be classified into…

  1. Nitro reduction to hydroxylamine/ amine
  2. Carbonyl reduction to alcohol
147
Q

Outline the hydrolysis reactions that take place in phase I metabolism

A

Hydrolysis - reaction between a compound and water

Results in the addition of water – improves metabolites polarity

Different enzymes catalyze the hydrolysis of drugs:

a) Esterase
b) Amidase

Ester or amide to acid, alcohol or amine

148
Q

What are the main oxidation enzymes in drug metabolism?

A

Oxidation enzymes – all these enzymes found in the liver

  1. Cytochrome P450 monooxygenase system
  2. Alcohol dehydrogenase
  3. Aldehyde dehydrogenase
  4. Flavin-containing monooxygenase system
  5. Monoamine oxidase
149
Q

What are the main reduction enzymes in drug metabolism?

A

Reduction enzymes

  1. NADPH-cytochrome P450 reductase
  2. Reduced (ferrous) cytochrome P450
150
Q

What are the main hydrolysis enzymes in drug metabolism?

A

Hydrolysis enzymes

  1. Esterases and amidases
  2. Epoxide hydrolase
151
Q

What system does phase I metabolism normally rely on?

A

Phase I relies on microsomal mixed-function oxidase system (cytochrome P450 dependent)

The mixed-function oxidase is found in microsomes (endoplasmic reticulum) of many cells (liver, kidney, lung, and intestine)

P450 requires oxygen and a reducing system (NADPH)—one atom of oxygen is transferred to the substrate, and the other undergoes a two-electron reduction and is converted to water

152
Q

What is Cytochrome P450 (CYP) 3A4? Where is it located? What other enzyme does it require?

A

Cytochrome P450 (CYP) 3A4 - Catalyzes hydroxylation or epoxidation of various substrates

CYP requires another enzyme NADPH-cytochrome P450 reductase which is a flavoenzyme containing one molecule of flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN) –> used for electron transfer to CYP

Both Cytochrome P450 and NADPH-cytochrome P450 reductase are associated with the membrane

153
Q

What is the role of the heme group in Cytochrome P450?

A
154
Q

Can Cytochrome P450 accomodate a wide range of substrates?

A

Cytochrome P450 can accommodate a large variety of functional group as the cavity adjacent to the Heme is large, as shown below.

155
Q

Outline the reaction cycle of the heme-group in cytochrome P450

A
156
Q

What is the role of Phase II transformation?

A

When phase I products are not sufficiently hydrophilic or inactive to be eliminated, the drugs or metabolites formed from phase I reaction undergo phase II reactions

Phase I reactions provide a functional group in the molecule to undergo phase II reactions –> this group is modified in phase II via conjugation reactions forming more polar and water-soluble products.

Require coenzyme + transferases for conjugation (liver)

Note - most conjugates are inactive and not toxic –> highly polar and unable to pass through membrane

157
Q

What are the different chemical reactions that take place in Phase II metabolism?

A
  1. Glucuronidation by UDP-Glucuronosyltransferase (-OH, -COOH, -NH2, -SH)
  2. Sulfation by Sulfotransferase: (-NH2, -SO2NH2, -OH)
  3. Acetylation by acetyltransferase (-NH2, -SO2NH2, -OH)
  4. Amino acid conjugation (-COOH)
  5. Glutathione conjugation by Glutathione-S-transferase
  6. Fatty acid conjugation
  7. Condensation reactions
158
Q

What are the phase I and II steps performed by the body when processing aspirin?

A

Phase 1 - Prodrug converted to active compound Salicylic acid

Phase 2 - Conjugate to allow for excretion – different routes

159
Q

What is the most important phase II pathway for drugs and endogenous compounds?

A

Glucuronidation - transferring glucuronate sugar moiety (a-D-glucoronic acid)

Most important phase II pathway for drugs and endogenous compounds

Products are often excreted in the bile

Enterohepatic recycling may occur due to gut glucuronidases - recycling of glucuronate

Requires enzyme UDP-glucuronosyltransferase (UGT) to transfer sugar moiety on to target

160
Q

What are the two different types of glucoronidation?

A

N- or O-glucuronidation

N-glucuronidation can be added to amines, amides and sulphonamides which are added in phase 1

O-glucuronidation - creation of an ester or ether linkage using carboxylic acids, phenols and alcohols

161
Q

What should you know about Phase II metabolism sulfation?

A

Sulfation

Major pathway for phenols (also alcohols, amines and thiols).

Requires the enzyme PAPS (3’-Phosphoadenosine-5’-phosphosulfate)

Phenols can be modified by both Sulfation or Glucuronidation but usually…

a) Sulfation at low substrate concentrations
b) Glucuronidation at higher substrate concentrations

162
Q

What should you know about Phase II metabolism acetylation?

A

Acetylation

  • Targets aromatic amines and sulfonamides
  • Requires N-acetyltransferase and the co-factor acetyl-CoA

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 - can be more harmful so must be considered

163
Q

What should you know about Phase II metabolism Fatty Acid Conjugation?

A

Stearic and palmitic acids are conjugated to drug by esterification reaction

164
Q

What should you know about Phase II metabolism Amino Acid Conjugation?

A

Active CoA-amino acid conjugates that react with drugs via N-Acetylation –> Glycine, Glutamine, Arginine

165
Q

What should you know about Phase II metabolism Glutathione Conjugation?

A

Glutathione Conjugation

Tripeptide Gly-Cys-Glu; conjugated by glutathione-S-transferase (GST)

Conjugated compounds can subsequently be attacked by g-glutamyltranspeptidase and a peptidase product can be further acetylated  metabolite acts as a peptide than can be further digested and acetylated

166
Q

How can we make drugs more resistant to metabolism?

A

Remove functional groups susceptible to enzymes

E.g. Tolbutamide can be excreted directly whereas Chlorpropamide needs to enter phase I metabolism

167
Q

How can we make drugs less resistant to metabolism?

A

Making drugs less resistant to metabolism:

If a drug is too resistant to metabolism, it can pose problems as well (toxicity, long-lasting side effects).

Add functional groups that are susceptible to metabolic enzymes – allow it to easily enter phase I metabolism

168
Q

How are metabolites normally exported from the cell?

A

Efflux transporters detoxify cells from ‘toxic compounds’; eg P-glycoprotein

One of the primary proteins involved in multidrug resistance in the treatment of cancers –> cancer cell use the efflux transporters to drive out export of drugs

ATP-binding cassette (ABC) –superfamily- requires ATP hydrolysis to drive the export

169
Q

What are factors that can influence the metabolism of a single drug?

A

Rate and pathway of drug metabolism are affected by species, strain, sex, age, hormones, pregnancy, and liver diseases

Drug metabolism is stereospecific – drugs may contain different enantiomers – specific enantiomers may be more toxic (pre/post-metabolism)

For example…

Enantiomers act as two different xenobiotics – different metabolites and pharmacokinetics

Sometimes the inactive enantiomer produces toxic metabolites or may inhibit metabolism of active isomer

170
Q

Why is it important to consider the amount of drug that binds to plasma proteins?

A

Drugs can bind to plasma proteins – determines bioavailability as in the bound state the drug is not bioactive

Only unbound compound is available for distribution into tissues

Acidic drugs tend to bind to albumin. Basic drugs bind to alpha-1 acid glycoprotein.

a) 0-50% bound = negligible
b) 50-90% = moderate
c) 90-99% = high
d) >99% = very high

% of drug reaching bloodstream = bioavailability

171
Q

What stage of metabolism do pro-drugs rely on?

A

Drug metabolism of Prodrugs – rely on Phase I metabolism to active the drug

Prodrugs are compounds that are inactive, but are converted in the body to an active drug by metabolic enzymes

172
Q

How can we improve membrane permeability of drugs?

A

1. Improve membrane permebility using the addition of esters

Note - If a carboxylic acid is important for drug binding to its target, but it prevents the drug from crossing a membrane, temporarily “hide” it as an ester. Once in the blood, it is hydrolyzed to the active form by esterase’s

But….

There is a balance that must be reached as if it’s too hydrophobic it will prefer the membrane environment and not diffuse into the cell

  1. N-methylation - amines can be methylated to increase hydrophobicity allowing movement across membranes

- N-demethylation is a common liver metabolic reaction –> can be removed by liver

173
Q

Apart from increase membrane solubility to improve membrane permeability, can we also hijack transporters?

A

Membrane transporter - mimic substrates that have transporters to cross membrane

174
Q

How can pro-drug concept be used to extend the life of a drug?

A

Create a pro-drug that is slowly converted to active drug –> allows for slow and prolonged release of active drug into the body

Example - 6-mercaptopurine

Immune suppressant (organ transplants), but it is eliminated from the body quickly.

Hence, a prodrug is used as it is slowly converted to the drug allowing longer activity.

175
Q

How was aspirin made less toxic?

A

Salicylic acid is a painkiller, but phenolic -OH causes gastric bleeding.

Aspirin has an ester to mask this toxic group until it is hydrolyzed in the blood to form the active pro-drug salicylic acid

176
Q

General role of Phase I and II metabolism?

A

Phase I and II metabolism make drugs less toxic and more hydrophilic

177
Q

For Lipinski’s rule of five, is there any particular reason why fewer than 5 h bond donating groups and 10 h acceptor groups in the compound is preferred?

A

More H-bonds means that the ligand is likely to bind very tightly - making it hard to reverse the reaction (undseriable)

Hence, having 5H bonds allows for tight binding but not too tight to the point where the binding is hard to reverse

If you desire a tight binder than you would use more than 5H bonds

178
Q

What is the difference between monoclonal and polyclonal antibody?

A

Monoclonal - single type of antibody targeting a single epitope

Polyclonal - Several types of antibody for the same target but different epitope

179
Q

Can u explain in detail about bit collision? Why different structures can be represented by the same bit?

A

In 2D fingerprinting we are attempting to describe what groups/elements are present and how they are linked

Bit collision arises when you have similar chemical groups which can’t be distinguished unless you indicate the position of the group in the molecule

More bit collision - more false positive results –> reality to be worse hit than predicted

180
Q

For rigid docking, can you explain why some good candidates will be excluded due to stereochemistry? Why would they be good candidates if there is steric clash?

A

The structures present in database often come from crystal structure elucidation, in which the molecule adopts its lowest energy state

But this might not be ideal for docking in our protein –> removing it as a possible candidate

181
Q

Can you get bit collisions in both 2D and hashed fingerprinting?

A

2D fingerprints and hashed fingerprints are similar but hashed fingerprints includes structural information –> but you can still get bit collisions in both

182
Q

Why lead identification by fragment self-assembly sometimes useful?

A

Sometimes the linked ligand is unable to bind directly e.g. due to steric clashes

183
Q

In glutathione conjugation, how does digestion of glutathione by peptidases relate to acetylation of the products?

A

Glutathione resembles a tripeptide –> Peptidase recognize this Gly-Cys-Gln motif resulting in cleavage

This leaves an amine which can be acetylated - things to consider when thinking about drug metabolism

184
Q

In drug metabolism, can multiple modifications happen to a single molecule in phase 1?

A

Not common but It is possible if there are multiple functional groups that can be modified.

But we need to remember that the molecule needs to re-enter the P450 enzyme for example and this might not be possible after the initial modification

185
Q
A