Drug Design_L2 Flashcards

1
Q

What are Antibody–Drug Conjugates (ADCs)?

A

(1)Antibody–Drug Conjugates (ADCs) are monoclonal antibodies or antibody fragments attached to biologically active molecules through chemical linkers with labile bonds, the benefit of applying the ADCs is the targeted antigens expressed exclusively or in extremely low abundance on the tumour cell surface

(2) Deliver highly cytotoxic agents directly to tumour cells without affecting other dividing cells in the body
while bound to the antibody, the chemotherapeutic ‘‘payload’’ no longer circulates systemically and is therefore well tolerated by healthy cells or tissues.

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

How is the antibody nature utilised by the ADCs?

A

(1) antibodies bind to the receptor of the cell specifically, 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) the cell internalises the receptor-antibody complex. Once bound to the antigen, the entire antigen–ADC complex is then internalized through receptor-mediated endocytosis. The internalization process proceeds with the formation of a clathrin-coated early endosome containing the ADC–antigen complex.
(3) attached pay-loader released/digested inside the lysozyme. Once inside the lysosome, the ADC is degraded and free cytotoxic payload released into the cell, leading to cell death. The mechanism of cell death will depend on the type of cytotoxic payload, the antibosy and linker are digested inside the endosome and the receptors are recylced to the cell surface membrane, payload molecule is released to cause cell death.
(4) act either inside the nucleus or in the cytosol

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

What are the limitations of ADCs? What strategies can be applied for improvement?

A

(1) 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, the ADC molecules kill the tumour cells, at the same time, somatic cells are killed as thwy often contain the same receptor as the cancer cells, moreover, the selection pressure posed on the tumour cells promotes the faster gene mutation.
(2) ADC needs to retain the selectivity of the original monoclonal antibody while being able to release the attached cytotoxic payload in concentrations high enough to kill the targeted tumour cells. Three key components to an ADC: the antibody, the linker and the payload, each of which requires optimisation.

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

What properties should the payloaders have in order to lead to sufficient cell death?

A

(1) cytoxic enough in low concentration, since the concentration applied to the cell does not mean the concentration reaching the cell, additionally, overloading the antibodies in the bloodstream can induce problems

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

challenges in designing ADCs?

A

(1) circulation of ADCs into the bloodstream: prevent/minimise the pre-mature release of payloader from the antibodies and the off-target toxicity
(2) binding: the mADC must retain high affinity to binding to the tumour cells
(3) inefficient internalisation due to limited target antigen level may prevent the cytokinetic from reaching the threshold concentration within the cell
(4) excessive binding to the FcRn can reduce the amount of toxic payload released into the cell
(5) ADC needs to release the payloader correctly in its active form
(6) potency of the payloader must be sufficient to kill the cell in the low concentration

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

What are the two major types of the linker present in the ADC cells?

A

(1) cleavable

(2) non-cleavable

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

What criteria should be based on for the tumour antigen selection?

A

(1) Requires substantial expression by tumour cells but limited expression by cells in normal tissues
(2) Limited number of antigens on the tumour cell surface (ranging from approximately 5,000– 106 antigens per cell) and the average drug-to-antibody ratio (DAR) of most current clinical-stage ADCs is limited to 3.5–4, the amount of the drug delivered by ADCs into tumour cells is low. However, the antibody based treatment is more effective in the bloodstream but not for the solid cancers.
(3) highly expressed on cancer cells and altogether absent on normal cells— is extremely rare, if not nonexistent, the cancer cell expression is higher than that of the normal cells, therefore, the side effects imposed on the normal counterparts are relatively small.

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

What criteria should be based on for the antigen-drug linker selection?

A

(1) Need to consider stability and drug release
(2) Stability typically refers to retention of drug by the antibody either ex vivo in buffers, plasma, or blood or in vivo after administration
(3) Antibody stability should also be considered upon conjugation with the drugs due to faster clearance: antobodies are more prone to degradation with more drug molecule attached, DAR is a good choice for the antibodies as it not only supports the drug transport and recognition but also facilitates in killing cancer cells.
(4) Stability of drug upon release

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

classfiy the cleavable linkers?

A

(1) Cleavable linkers utilise the differences in conditions between the bloodstream and the cytoplasm within tumour cells
(2) The change in environment once the ADC–antigen complex has internalized triggers cleavage of the linker and release of the active payload
(3) Cleavable linkers are divided into three main sub-categories: (1) Acid-labile (e.g.hydrazones), (2) Reducible (e.g.disulphides) and (3) Enzyme-cleavable (e.g., peptides, linkers include glucuronide-triggered linker/Val-Cit linker).

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

define the meaning of acid-labile linker

A

(1) Acid labile hydrazone linkers remain stable in neutral pH and utilize the low pH within the endosome/lysosomes to cleave the conjugation and release the drug from the ADC
(2) Clinical studies indicating that acid-cleavable linkers are associated with non-specific release of the payload, which can lead to systemic toxicities

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

define the meaning of reducible linker

A

(1) The principle behind this type of linker is the ability to respond to a difference in reduction potential between the intracellular compartment of a tumour cellversusthe blood based on glutathione concentration gradients, the environemnt inside the cancer cells has strong reducing power to enable the reduction of disulphide, as a result of the glutathione
(2) Disulfides are stable at physiologic pH but are susceptible to nucleophilic attack from thiols, such as glutathiol
(3) Abundance of thiol molecules within tumors, especially generated during stress like hypoxia, for thiols are involved in survival and growth of tumor cells

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

define the meaning of enzyme-cleavable linker

A

(1) This type of linker takes advantage of hydrolytic enzymes capable of recognising and cleaving particular peptide sequences contained within linkers, thus ensuring that the ADC only undergoes cleavage in the lysosomal environment and not in the plasma
(2) Enzyme-cleavable linker is based on the β-glucuronide moiety. The enzyme β-glucuronidase, which can release payloads from β-glucuronide-containing linkers, is present in lysosomes and is over-expressed in some tumour cell types. Hopefully, the tumour cells are specifically targeted. An important characteristic of a β-glucuronide linker is its hydrophilic properties, which can potentially reduce aggregation during conjugation compared with constructs containing dipeptide-based or other linker types

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

What is the mode of ADC action for ADCs with cleavage linker?

A

(1) binding to a specific Ag, followed by internalization of the ADC-Ag complex according to a clathrin dependent mechanism
(2) transfer into lysosomes
(3) lysosomal cleavage of the linker between the trigger and the spacer (if present), which then degrades to release the free drug
(4) binding of the drug (MMAE, MMAF, DM1, DM4) to tubulin or translocation of the drug (calicheamicin) into the nucleus
(5) Alternatively to 4, transfer of the drug to the cytosol
(6) diffusion of the drug to neighboring cancer cells to achieve bystander killing effect (all drugs except MMAF), it is most desirable if the neighbouring cell is a cancer cell and vice versa.
(7) MMAF, because of its charged character, cannot cross the membrane and therefore does not exhibit a bystander killing effect
(8) all the previous steps lead to cell death

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

define the meaning of non-cleavable linkers?

A

(1) Drug usually remains attached to mAB, the covalent linkage is not disruptable by pH, reducing strength and the enzyme cleavage
(2) Non-cleavable linkers engage lysosomal degradation only.
(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
(6) for example, the thioester bond is one of the non-cleavable linkers: -S-CH3-CH3-CO-

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

What is the mode of ADC action for ADCs with non-cleavage linker?

A

(1) ADC recognises a specific Ag, followed by internalization of the complex ADC-Ag through a clathrin-dependent mechanism
(2) transfer into lysosome
(3) complete digestion of the mAb to release the active corresponding metabolite amino-acid-NCL-drug
(4) transfer to tubulin or transfer to cytoplasm, becuase of the linker protein, the drug cannot be transported to the nucleus and acts in the nucleus
(5) Impossible to achieve bystander effect, therefore transferring to tubulin.
(6) all the previous steps lead to cell death of the Ag-positive cancer cell.

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

What is the site of conjugation?

A

(1) Second-generation ADCs are all controlled mixtures of different drug-loaded species (from 0 to 8 drug molecules per antibody)and have a typical average DAR of 3.5 or 4
(2) 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) Thiosuccinimide linkage, which is formed through the reaction of thiols and alkyl maleimides; thiosuccinimide formation is slowly reversible under physiological conditions
(4) Conjugation to the terminal amines of lysine residues with either disulphide bond or ester amide.
Lysine amines are very effective for payload conjugation as they are well-exposed and their amino groups are good nucleophiles. Lysine conjugation reaction involves formation of a stable amide bond using activated esters of the payload, typically an O –succinimide reagent such as NHS. Conjugation utilise maleimide, or haloacetamides.

17
Q

describe the inner linkage within the antibody?

A

(1) Monoclonal IgG antibodies contain multiple disulphide bonds: the disulphide bonds are required to be reduced for the activation of the antibodies in priority
(2) The inter-chain disulphide bonds are the most desired sites of attachment for cytotoxic payloads
(3) Disulphide bonds are typically reduced with agents such as tris(2-carboxyethyl) phosphine (TCEP), dithiothreitol (DTT) or 2-mercaptoethylamine (2-MEA) prior to conjugation
(4) React with a linker–payload complex containing a suitable electrophilic moiety; maleimide, (N-Hydroxysuccinimide) NHS, 3,4-dibromomaleimide in order to conjugate with the thiol group. Partial reduction of the disulphide bridges of an IgG1 antibody to generate two nucleophilic thiol groups which can then be reacted with an electrophilic linker–payload construct (DAR 2), in this case, the disulphide bond is reduced on the variable chain in order to react with the DAR molecules on conjugation with the thiol group
(5) Conjugates with mixed DARs and varying locations of payload, the number of broken disulphide bonds and the site of conjugation are completely random.

18
Q

In which way can the site-specific conjuagtion produced on the antibodies?

A

(1) Site-specific conjugation by the THIOMAB technology
(2) THIOMABs are antibodies with engineered reactive cysteine residues. Defined stoichiometry and sites for payloads without disruption of the interchain disulphide bonds
(3) workflow: 1. complete reduction of all the disulphide bonds on the antibody molecule by reducing agent
2. re-oxidation of the antibody, only cysteines far away from each other are unaffected by the oxidising agent, those regions are refered as the non-oxidative region
3. cytotoxic payload covalently attaches onto the cysteine in the thiol bond linkage.

19
Q

Further strategies to improve conjugation

A

(1) Engineered Cys
(2) Insertion of unnatural amino acids: tight regulation of toxicity of the payload by introducing the naturally uncommon amino acids
(3) Enzyme-assisted ligation: use enzyme to catalyse the ligation of the drug onto the surface of the antibodies
(4) Glycan remodelling and glycoconjugation: antibodies are easily glycosylated
(5) Amino‑terminal engineered serinewhich changes the overall property of the antibodies
(6) Native cysteine rebridging
(Highly loaded ADCsat specific sites)

20
Q

critise on the toxic payload or warheads

A

(1) High cytotoxicity is required since delivery is limited by antigen copy number; payloads need to be active in the low nanomolar or even picomolar region, as well as possessing favourable physicochemical properties, such as an acceptable hydrophilic/hydrophobic balance and good stability
(2) Clinical trials only use a limited number of families of cytotoxic drugs as warheads:
1. target DNA (these are cytotoxic for proliferating and non-proliferating cells)
2. microtubules (these are cytotoxic for proliferating cells) which has something to do with the metastasis
(3) Cytotoxic drugs that are used in ADCs are hydrophobic and tend to induce antibody aggregation(need to be optimised): hydrophobic surface shields away from the hydrophilic environment
(4) The drug must retain its potency when modified for linkage (or made ‘linkable’), show aqueous solubility and be stable in aqueous formulation as a conjugate

21
Q

give some example of toxic payload and their roles

A

(1) Auristatins. The largest group of ADCs in clinical trials are those based on monomethyl auristatin E (MMAE) and MMAF; synthetic analogues of dolastatin 10 (a natural antimitotic drug that is extracted from the sea hare Dolabella auricularia); too toxic to be used in its unconjugated form.
high potency, water solubility, stability under physiological conditions and suitability for the attachment of stable linkers
(2) Tubulysins. Tubulysins inhibit microtubule polymerization during mitosis to induce cell death
Calicheamicins. It binds to the minor groove of DNA and cleaves double-stranded DNA in a site-specific manner
(3) Duocarmycins. Duocarmycins are DNA minor groove-alkylating agents.
(4) Benzodiazepines. PBDs bind to the DNA minor groove in a sequence-specific manner.
(5) Doxorubicin. Doxorubicin is an actinomycete-derived antimitotic anticancer agent that is routinely used in the clinic.

22
Q

explain what is bystander effect

A

(1) Solid tumours often express the target antigen in a heterogeneous manner.ADCs that selectively kill only antigen-positive cells and spare neighbouring antigen-negative cancer cells may be ineffective in eradicating such tumours
(2) ADCs can be designed to kill not only antigen-positive cells but also other cells in the vicinity, without expression of the target antigen on their surface(bystander effect)
(3) Need to be able to cross biomembranes by diffusion and kill neighbouring cells in the condition that the linker is cleavable or hydrophobic
(4) Bystander effect is not always desirable; might choose a non-cleavable linker

23
Q

Why would the bystander effect negative?

A

(1) the ADC is recognised by the bound receptors and internalised into the cell by clathin-mediated transport
(2) lysosome fuses the ADC to enable digestion
(3) the antibody is separated from the cytoxic payload molecules which are released into the nucleus for cell death
(4) some payload molecules diffuse across the cell membrane to the nerighbouring antigen-negative cell to induce apoptosis
(5) the normal functions of cells are inhibited, which is clearly not desired.
(6) antibodies may be internalised without binding to the receptors
(7) the toxicity of the ADC brought to the cells should be tackled in the first place

24
Q

What changes are observed from the first generation to the third generation?

A

toxicity is reduced but at the same time the affinity is increased

25
Q

During the conjugation reaction, what components should be assessed and what aspects should be focued?

A

(1) Functionalization of the cytotoxic agent is required to enable the conjugation process
(2) enzymatic kinetics: how much ligands required to reach half activity
(3) the tight linkage between the ADCs and the antibodies