Unit 3 - Drug Development Flashcards

1
Q

Outline the four stages of the drug development process (each requires a short definition)

A
  • Disease/target identification – to reveal a suitable target against a drug can be used to treat the disease
  • Drug Discovery and development – The development and screening of potential agents using in vitro and in vivo models
  • Clinical trials – to determine that a drug is effective against a specific disease with tolerable side-effects
  • Manufacturing and marketing – the up-scale of manufacture after clinical trials and associated packaging considerations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What are the 3 factors associated with drug development process:

A
  • It is long – on average 12 years – clinical trials, identifying a pathway if the disease is rare/not-well understood
  • It is expensive – on average £1.2bn
  • High rate of attrition – high-throughput screening can mean you start with 1000’s of molecules but end up with one drug
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is the longest and most expensive phase of drug development

A

Clinical trial phase

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

What does MHRA stand for?

A

Medicines and health care products regulatory authority

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

What things are good for a potential drug target be?

A
  • Must be potentially druggable (comes from good knowledge of structure/function usually from X-ray crystallography and modelling using in silico) – some sort of pocket that allows binding
  • Validated in disease pathogenesis – that its association with the disease is causative rather than correlative – so inhibition can effectively inhibit disease progression
  • Distinction from previously known targets and specificity to the disease – to limit side-effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Discuss some of the ways in which ‘new drugs’ can be developed/discovered?

A

Chemical Modification/me-too drugs
This is where an existing drug structure is modified slightly to improve its activity e.g., potency, faster on-set, fewer side-effects, improved selectivity (like regorafenib from sorafenib multi-kinase inhibitor).
Compound libraries
Uses a library of 1000’s of molecules with a similar structure which can be screened using high-throughput screening to select the drugs with the best characteristics (affinity, ability to inhibit a molecule/process, cell death… etc).
Rational Drug Discovery
Designing a drug against a specific molecule/pathway implicated in a disease – like Tratuzumab and pertuzumab in HER2 amplified breast cancer.
Serendipity
Molecules that have been observed to have a specific event by chances, but are then interpreted scientifically – penicillin inhibits the growth of bacteria or vincristine in cancer

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

What are the advantages and disadvantages of me-too drug discovery

A

It can potentially improve characteristics of the drug (potency, half-life, side-effects) and is cheap
But this may reduce the incentive for novel drug discovery and sometimes these changes may only be small!

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

What factors are investigated during development screening?

A

In vitro – binding, phenotypic changes, drug interactions (CYPs), permeability, mutagenicity, PgP inhibition, interacts with hERG
In vivo – Metabolic/pharmacokinetic prolife, toxicity, drug interactions, BBB
This may be followed by modification cycles

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

In drug development what is GLP

A

Good laboratory practice – must be abided by all those working in drug development – ensures data is comparable, reliable and robust

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

What factors are investigated in clinical trials

A
  • Pharmacokinetics - ADME
  • Pharmacodynamics
  • Safety and toxicology
  • Efficacy – usually compared to standard treatment options
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What does ADME stand for?

A

Adsorption, distribution, metabolism, excretion

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

Describe the phases of clinical trials

A

Phase 1 – Safety in healthy individuals
Phase 2- Safety and effectiveness in disease group
Phase 3- Effectiveness in larger disease groups
Phase IV – surveillance in large population after drug approval
Must abide by good clinical practice

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

What two phases in drug discovery require communication between pharmaceutical companies and regulatory agencies?

A
  • After preclinical (for Phase I) – IND (investigational new drug) or CTA (clinical trial application) based on this data
  • Seeking approval – NDA (new drug application) or MAA (marketing authorization application)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What factors may be involved in GMP?

A

Good manufacturing process- consistent and effective manufacturing, appropriate marketing, evaluation of the drug in Phase IV trial.

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

What must be done during the manufacturing and marketing phase of drug development?

A

Up-scale production – ensuring large quantities of this molecule can be made effectively and packaging considerations – light/temperature sensitivity, clear and understandable information.

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

What are the two types of bone metastases and what cells are involved in each and how these processes occur?

A
  • Osteolytic – degradation of bone (breast) ->osteoclasts
    Tumour cells secrete bone re-modelling factors like Cox-2 or IL-8(pre-metastatic niche) – which can acts directly on these osteoblasts or stimulate them to produce RANKL which interacts with RANK on osteoclast surface which leads to break down of the bone (and release of TFGβ which can stimulate breast cancers to produce more growth factors).
  • Osteoblastic – deposition of bone (prostate) – osteoblast
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What molecular changes are associated with bone metastases (not viscous cycle)?

A

Osteoclasts have ruffled membrane and secrete cathepsin K’s to degrade bone which require an acidic pH – which is what this ruffles membrane does – in producing an enclosed environment for bone breakdown.

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

Discuss the various options for targeting bone metastasis in cancer?

A

Targeting the cytoskeleton to prevent these ruffles/attach tightly to bone (creating microenvironment)
Bisphosphonates can be used which inhibit farnesyl-diphosphate-synthase (in membrane localisation of rho/rac). Two side groups R1- helps bind calcium component and R2 can dictate potency
Inhibiting cathepsins
Odanacatib is a molecule that inhibits these enzymes, but was associated with arterial fibrillation
Inhibiting the interaction between RANKL and RANK by the anti-RANKL antibody denosumab

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

Describe the ways in which you would carry out target identification/validation?

A
  • Look at its levels in tumour cells compared to normal cells; proteomics/gene microarray
  • Gene overexpression/inhibition studies to look at changes in cell phenotype
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is a pharmacophore?

A

The minimum region of a drug that is essential for activity

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

What is a pro-drug?

A

A drug not administered in its active form, but has to be activated usually through metabolic enzymes (an example is temozolomide). This might be due to stability/solubility parameters

22
Q

Why is knowing the structure of the active site so important in drug discovery?

A

It allows easier design of potential/lead agents based on shape/biochemical complementarity – meaning that drug design in cheaper and quicker.

23
Q

Why are natural products a valuable source of anti-cancer drugs?

A
  • Because the structure of these molecules is usually incredibly complex and something that would likely not be designed by a pharmacologist in the lab!! Make up around 50% of all current drugs.
  • Can be a good source due to the high competition and selection pressure in nature
  • Plus, can uses these molecules a s starting point to help produce more active molecules, for instance mitoxantrone from doxorubicin – which is easier to make and less cardiotoxic!!
    o Or you can produce from an intermediate from another species (paclitaxel via semi-synthesis) or use biotechnology to improve yield
    But these molecules are difficult to synthesise in the lab and there is often low yields from plants!!
24
Q

Give examples of natural products used in cancer therapy

A

Doxorubicin – from Streptomyces species – is a topoisomerase inhibitor
Paclitaxel from pacific yew tree – inhibits microtubule depolymerisation – terpenoids!!!!!
Vincristine – from Madagascan Periwinkle – can prevent microtubule polymerisation
Cytarabine from phylum porifera (sea sponges) – anti-metabolite which can be incorporated into DNA causing damage (semi-synthetic from 2-deoxycytidine)
Eribulin (sea sponge)– binds to plus end of microtubules (again is a derivate)
Trabectedin – from sea squirt (and some symbiotic bacteria)– binds to minor groover distorting structure and inhibits MDR1 induction
Combretastatin A4 – from South African Bushwillow – inhibits tubulin activity involved in inhibiting mitosis

25
Q

What are herbal medicines?

A

A preparation from a plant rather than a particular molecule! Complex mixture!

26
Q

What is phytochemistry?

A

The study of drugs extracted from plants

27
Q

What is oral bioavailability?

A

The fraction of the orally administered dose that reaches the circulation unchanged

28
Q

Why is it beneficial that a drug is given orally?

A

Higher chance of patient compliance due to fear of needles. This may also need to be given by a specialist in a hospital – whilst orally available drugs can be taken at home allowing patients to integrate treatment into their everyday routine.

29
Q

Explain the importance of Lipinski’s rule of 5

A
  • Molecular weight less than 500Da – higher weight will reduce solubility
  • Log(p) – ratio of its ability to dissolve in an organic solvent like octanol compared to water – has to be less than 5 – if too high wont be able to dissolve in blood (will clump) if too low wont be able to cross hydrophobic cell membrane and enter cells
  • No more than 5-hydrogen bonds doners (H-O/N)
  • No more than 10 hydrogen bond acceptors (O/N)
30
Q

Discuss ways in which compounds can get around issues with Lipinski’s rule of 5

A
  • Given as a salt – tamoxifen citrate
  • Give with albumin to help carry around the blood – abraxane (similar to paclitaxel) or a solubilising excipient (think of castor oil in paclitaxel)
31
Q

What modifications were made of chloromethine to make it less toxic -> melphalan?

A
  • Addition of phenylalanine group – more specific to cancer cells as proteins rapidly taken up
  • Addition of aromatic group – reduces reactivity
32
Q

What factors are associated with pro-drugs?

A
  • Masking of functional groups
  • Improved water solubility
  • Increase bioavailability
    Irinotectan (Camptothecin) – allows it to form a salt increasing solubility
33
Q

What is a pro-drug?

A

An inactive form of a drug converted to the active form after administration

34
Q

Describe differences between small molecule drugs and biologics

A
  • Small molecule drugs have a small Mwt (few 100) vs 100,000
  • Small molecule drugs are much cheaper
  • Small molecule drugs generally binding intracellular targets vs extracellular targets
35
Q

What are the three components of immunoconjugates

A
  • Monoclonal antibody to help specificity
  • Cleavage linker to separate the drug from the antibody
  • The drug
36
Q

Give examples of Antibody-drug-conjugates in cancer.

A

CD22 – inotuzumab ozogamicin in ALL (not effective) or CD33 – gemtuzumab ozogamicin in AML
Has the alkylating agent, calicheamicin, with Iodine atom, tri-sulphide sugar and enediyne group – I and sugar align with the major groove and then the tri-S group is cleaved, change in structure of molecule -> benzene di-radical DS break in DNA!!!
CD30 – Brentuximab vedotin
Vedotin (pseudo peptide- quite easy to make) from a marine sea hare with peptide linker

37
Q

What are the advantages and disadvantages of ADCs?

A

Advantages
Rational delivery system for highly toxic drugs
Selective for tumour tissue
Fewer side-effects
Stable until released at a precise location increasing the dose that reaches the target
Disadvantages
Delivery issues due to protein structure and molecular weight
Still expensive and cannot become generic – high cost and difficult of manufacture – inhibits tubulin similarity to vincristine/dolastatins

38
Q

What is BCL-3?

A

Is a protein involved in NF-KB signalling – has a transactivation domain but not DNA binding – hence can still promote expression of genes involved in inflammation (IL-1/8), myc and BCL-2

39
Q

Biomarker

A

An indicator used to determine (i) how well the body responds to a treatment for a disease or condition or (ii) how a particularly disease may progress.

40
Q

Formulation (of a drug)

A

Formulation is the mixing of compounds thatdo not react in order to get a mixture with the desired characteristics.The usefulness of a drug is based not only on its effectiveness in treating a condition but also on how readily it can be administered to the patient. The ideal drug is in tablet form and bland tasting. Tablets are formulated with additional ingredients to prevent stomach upset, give a timed release and hold the tablet together as a solid. Liquid medications must often be mixed with strong flavourings or alcohol to hide the taste of the medicine

41
Q

Mass balance studies

A

Used to determine the metabolite profile of a drug in laboratory animals and humans. These tests involve administration of a radio-labelled drug and collection of biological fluids which are then analysed for radioactivity and profiled for drug metabolites. These studies not only determine the rates and routes of excretion but also provide critical information on the metabolic pathways of drugs.

42
Q

Orphan drug

A

Drugs developed specifically to treat a rare medical condition

43
Q

Pharmacokinetics

A

the study of what the body does to a drug

44
Q

Pharmacodynamics

A

the study of what a drug does to the body.

45
Q

Pharmacogenetics

A

Pharmacogenetics is the study of how your genes affect the way your body responds to a medicine. Pharmacogenetics provides information that may help to provide better and safer drugs and/or a more appropriate dose of a medicine

46
Q

Surrogate Endpoint

A

biomarker intended to substitute for a clinical endpoint by measuring the efficacy of a drug substance indirectly, when direct measurement is either impossible or impractical. It is expected to predictclinical benefit (or lack of) but is not itself a measure of clinical benefit.

47
Q

What is adsorption?

A

The process by which the unchnaged drug proceeds from the adsorptive drug barrier to the circulatory system

48
Q

What is Distribution?

A

The process of drug transfer from immediate site to tissues

49
Q

What is Elimination

A

Process by which the drug is lost via metabolism or excretion

50
Q

Why might drugs be differently distributed in different tissues?

A

Plasma and tissue binding drugs

Tissues have different blood-flow and transporters which may affect distribution