licensing a new drug Flashcards

1
Q

What molecular biology, in vitro studies & computer technology cannot do

A

• Integrated response
– molecule to man
• Reveal the unexpected
– secondary actions, selectivity. Doesn’t remove the risks
• Determine the therapeutic index- know the dose that you are taking into the clinical trail
• Assess importance of multiple mediators- seeing multiple effects
• Determine pharmacokinetics- hw drug is absorbed ,metabolised and excreted
• Assess safety & toxicology
• Set clinical dose range- need to know the dose you are treating with

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

what is meant by pharmacodynamics

A

• Is the study of the biochemical and physiological effects of drugs.
• Specifically those actions for which the drug was designed effect
• Information obtained includes:
– Lead optimization- make it as selective and as potent as possible
• Improve target specificity
• Improve target selectivity
– Efficacious dose range and therapeutic window
– Specificity
• Investigation of off target events

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

what is meant by pharmacokinetics

A

what happens to the drug after it has been administered?
• Is the determination of the fate of substances administered externally to a living organism. Need to monitor the patient depending on where the drug can affect more than one organ
– Absorption
– Distribution
– Metabolism
– Excretion

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

what is meant by absorption?

A

• The process by which drug proceeds from the site of administration to the site of measurement in the body i.e. blood stream. Need to know whether or not you may need to make a pro drug to form the active form.
• Varies due to administration
– IV drug is immediate and complete
– Oral drug delayed and incomplete. Goes through first pass metabolism so the dose will be lower than when it is first administered
• Relies on passage through membranes to reach the blood

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

what is meant by distribution?

A

• Determined by
– Partitioning across various membranes- does it cross the Blood Brain Barrier
– Binding to tissue components- does it bind to receptors or outside cells
– Binding to blood components- plasma protein binding
– Physiological volumes- the volumes of liquid in the tissue

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

what is meant by metabolism?

A

• Determined by the liver (mainly)
– Location of metabolism enzymes e.g. liver or kidney
– 1st pass metabolism
– Induction of drug metabolizing enzymes-can inhibit drug metabolizing enzymes
– Development of pro-drug

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

what is meant by elimination?

A

• Either by excretion unchanged or drug metabolites excreted
• Drug metabolism is usually associated with a chemical modification of the drug with the overall goal of getting rid of the drug, typically enzyme driven
• Excretion is the main process by which the body eliminates unwanted substances
– Most common route biliary or renal
– Other routes include lung, skin, etc need to know eg if it is ecreted whe breathing out

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

what is meant by toxicology?

A

• Concerned with the adverse effects of chemicals on living organisms.

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

what is

Toxicological profile

A
  • Single dose toxicity
  • Repeat dose toxicity
  • Genotoxicity- works in the genome
  • Carcinogenicity
  • Reproductive and developmental toxicity
  • Local tolerance
  • Environmental issues
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Molecular mechanisms of toxicity

A

• Allergic response
– Can lead to anaphylactic shock e.g. penacillin
– Deplete blood cell types e.g. sulfonamides can cause neutropenia
• Receptor, ion channel and enzyme mediated
– Animal toxins can block ion channels
• Biochemical pathways
– Inhibition of mitochondrial function
• Organ-directed toxicity
– Hepatotoxicity e.g. paracetamol affects liver depletes glutathione
– Nephrotoxicity
• Changing glomerular filtration rate e.g. ramapril increases it
• Chronic nephritis e.g. paracetamol
• Mutagenesis and carcinogenesis- drugs that can cause cancer and affect DNA
• Teratogenicity e.g. thalidomide (R-enatiomer sedative, S-enantiomer teratogen) affects the unborn fetus

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

what are the 2 testing methods?

A

• Preliminary toxicity testing
– Maximum non-toxic dose given for 28 days to 2 species before tissue damage assessment- see what possible damge there could be for long term use.
– LD50 test-the dose of drug which kills 50% of treated animals within a specified short amount of time

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

what does single dose studies/ acute toxicity studies entail?

A
  • Effect of single dose- most important stage as it determines the next stage
  • Carried out on two different animal species
  • Effects of dose observed for 14 days with any mortality recorded.
  • Morphological, biochemical, pathological and histological changes are investigated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Repeated dose studies/sub-actue or chronic studies

A

• Two mammalian species
• Long duration of studies (30-180 days)
• Dose is dependent on dose escalating studies
– High dose 10x the maximum clinical dose
– Low dose 2x clinical dose
– Medium dose midway between low and high dose
• Drug administered by clinical route
• Parameters monitored include
– Behavioral
– Physiological- does it affect breathing heart rate/ tachycardia
– Biochemical- liver, kidney
– Histological- look at every organ in the body

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

what is local toxicity studies?

A

• Route of administration dependent so for example
– Dermal toxicity studies- the dose is going to be highest at the point of administration
• Local signs (oedema, erythema)
• Histological studies
– Rectal tolerance studies- the dose is going to be highest at the point of administration so look for any signs of pain
• Signs of pain, blood or mucus
• Histological studies
– Parenteral drugs
• For IV, IM, SC, ID
• Sites of injection examined grossly and microscopically

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

Allergenicity/Hypersensitivity toxicological studies

A

• Guinea pig maximization test- skin on the ears is the same as the human skin
– Evaulation of erythema and oedema
– Determination of maximum non-irritant or minimum irritant dose
• Local lymph node assay
– Drug given on mouse ear skin
– 5 days treatment followed by auricular lymph node dissection
– Increase in tritiated-thymidine used for evaluation. Taken up by cells that are divining so it shows that there is an inflammatory reaction as the lymph node cells are dividing

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

Carcinogenicity/Oncogenicity studies

A
  • Life-time bioassays
  • Drug used for >6 months or frequent intermittent use for chronic diseases
  • Chemical structure of drug indicates carcinogenic potential
  • Therapeutic class of drugs which have produced positive carcinogenicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

what are the clinical signs of toxicity?

A

• Respiratory
– Dyspnea, abdominal breathing, gasping, cyanosis
• Motor activity
– Loss of righting reflex, ataxia, tremors
• Reflexes
– Pinneal, light, righting
• Ocular signs
– Lacrimation, miosis, iritis
• Cardio-vascular signs
– Bradycardia, tachycardia, vasodilation
• Autonomic signs
– Para/sympathomimetic actions or blockers
• Other signs
– Salivation, piloerection, GIT signs (emesis, diuresis)

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

what is Preliminary toxicity testing

A

NOAEL (no observed adverse effects level)
Highest concentration that does not a toxic response

LOAEL- lowest observed adverse effects level
Lowest concentration that produces a toxic response

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

what is the use for NOAEL ?

A

• Determine NOAEL
• Convert NOAEL to a human equivalent dose (HED)
– Adjust for anticipated exposure in humans
– Adjust for interspecies difference in affinity and potency
• Apply a >10 fold safety factor

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

how do you calculate

A

• Convert NOALs to HED based on body surface area
– Assumes there is a 1:1 relation between species when normalized to body surface area (BSA)
• HED (mg/kg)=NOAL (mg/kg) x Animal Km/Human Km
• Km=Body weight (kg)/BSA (m2)
– E.g. Human Km=37 (60 kg), mouse Km=3 (0.02 kg), Rat Km=6 (0.15 kg)

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

how do you calculate the therapeutic index?

A

The ratio of the dose of the drug that produces an unwanted
(toxic) effect to that producing a wanted (therapeutic) effect
= LD50 / ED50

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

Target related safety

A

• Tissue distribution
– If the target is highly expressed in organs other than those intended for therapeutic modulation

• KO animals
– Phenotypes observed in genetically manipulated animals are valuable in identifying potential issues

• siRNA approach
– Silencing the target in specific organs can identify toxicities
– Confirm the role of the target in a toxicological outcome

• Inactive enantiomers
– Inactive structure is a mirror image of the active isomer the potential for chemistry-related toxicity is equivalent
– Determines that the target is the cause of the toxicity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

what are the Chemistry related safety

A

• Chemical series
– Identify structural features associated with adverse effects

• Metabolites
– Is the drug metabolized into a chemical that causes adverse effects

• Isomers
– Does the drug have an isomer, does it show same activity/toxicity

• Impurities
– During the synthetic pathway what impurities are formed, what % remain in final formulation, do they cause any adverse effects

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

how to Identify physiological parameters for clinical monitoring of potential adverse effects

A
  • Adverse effects in animals for specific organs
  • Tissue expression of the target
  • Target itself e.g. receptor found in autonomic nervous system
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

what are the Hazard integration and risk assessment

A
•	Regarding patient safety
–	Co-morbidities
–	Co-medications
–	Age
•	Risk-benefit
–	Effect of drug on symptoms, disabilities and prognosis balanced against unwanted effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

what is meant by scientific study?

A

• Seeks to establish facts
– Things known to have occurred or be true (OED)
• Contributes to knowledge

• Confirms, adds to or establishes theory
– a conceptual framework that explains existing observations and predicts new ones

  • Should be controlled so as to be free of bias
  • Should be designed on a scale that the results can be statistically evaluated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Detailed scientific studies

A
•	Are expensive to conduct
•	Require specialised skilled operators to 
–	Design
–	Analyse 
–	Interpret the data
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Do all scientific studies provide evidence for clinical practice?

A
  • Animal studies

- In- vitro studies

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

what is meant by Epidemiology

A
  • Study of how often diseases occur in different groups of people and why
  • Used to plan and evaluate strategies to prevent illness and as a guide to the management of patients
  • Relate primarily to groups of people
  • Epidemiology is the method used to find the causes of health outcomes and diseases in populations. In epidemiology, the patient is the community and individuals are viewed collectively. By definition, epidemiology is the study (scientific, systematic, and data-driven) of the distribution (frequency, pattern) and determinants (causes, risk factors) of health-related states and events (not just diseases) in specified populations (neighborhood, school, city, state, country, global). It is also the application of this study to the control of health problems
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

list the different types of clinical studies?

A
  • Qualitative research
  • Focus groups
  • Individual interviews
  • Observational studies
  • Case studies
  • Cohort studies
  • Cross-sectional studies
  • Experimental studies
  • Clinical trials
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

what is meant by qualitative research?

A
•	Collecting and analysing non-numerical data
•	Understanding 
–	Concepts
–	Opinions
–	Experiences
•	Gather in-depth insights into problems
•	Generate new ideas for research
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

what is meant by observational studies?

A

• Researchers observe the effect of something on a population
– Disease
– Risk factor
– Diagnostic test
– Treatment or intervention
• Is not experimental
– Do not try to change who is or isn’t affected

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

what is meant by Cross-sectional studies

A
  • Measure the frequency of a disease or condition in a defined population at a given time
  • A census of occurrence of characteristic(s)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

what entails in a case study?

A

• Medical history of one (or small group of) patient
• No control group
– Anecdotal evidence

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

what is meant by case study control?

A

• Compares patients who have a condition with a group who do not have that condition
• Looks retrospectively at risk factors that may impact on the condition
– Also known as retrospective studies

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

what are the advantages and disadvantages of case control studies?

A
  • Good for studying rare conditions
  • Relatively quick to set up
  • Can look at multiple risk factors
  • Clinical records may be inconsistent
  • May rely on memory (recall bias)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

what is meant by cohort studies

A
  • Observational studies of subjects with a specific disease or characteristic
  • May be compared to a control group
  • Usually over a long time period
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

what is meant by prospective and retrospective studies?

A

n a retrospective cohort study, the group of interest already has the disease/outcome. In a prospective cohort study, the group does not have the disease/outcome, although some participants usually have high risk factors.

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

what is meant by experimental studies?

A
  • Introduce an intervention

* Study the effects on the treated population

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

what are the different categories of a clinical trail?

A
  • Specified condition
  • Specified treatment
  • Test and control group
  • Measure the outcomes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

what is meant by randomised controlled trails

A
  • Population of patients chosen
  • Divided into 2 groups
  • Trial group compared with control group
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

what is meant by Meta-analysis of RCTs

A
  • A type of systematic review that focuses on the numerical results
  • Aim is to combine the results to produce an estimate of the overall effect size
  • Data from several trials are pooled and averaged to estimate the overall effect
  • No new raw data collected
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

list in order the hierarchy of evidence

A
  • Systematic review / meta analysis
  • Randomised control trials
  • Cohort studies
  • Case control studies
  • Case series /reports
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

what do clinical trials test for?

A
•	Administration of new substances with therapeutic potential to people under controlled conditions for the purpose of determining
–	Efficacy
–	Bioavailability
–	Safety
–	Tolerability
–	Acceptability
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

what does the The Medicines for Human Use (Clinical Trials) Regulations 2004 say?

A
  • Good Clinical Practice
  • Good Manufacturing Practice
  • Regulatory inspection and enforcement
  • Protection of incapacitated adults
  • Protection of minors
  • Pharmacovigilance arrangements
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

what does ICH guidelines – good clinical practice

A
  • Risks and potential benefits must be assessed before trials are started
  • Interests of the individual study subject must take precedence over those of science or society
  • All trial subjects must give consent
  • Trials must be scientifically sound
  • Trials must have a clear protocol
  • Trials must be approved by a properly constituted ethics committee
  • Only properly qualified staff may be involved (including physicians to provide care if needed)
  • Should be adequate preclinical testing of the product
  • Product should be of adequate quality (as defined in ICH guidelines for medicines)
  • Trial subjects privacy and confidentiality must be respected and assured
  • Data must be recorded, handled and stored in a way that allows accurate reporting, interpretation and verification
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

what are the 4 clinical development phases?

A

phase 1: First administration and safety evaluation in man – usually healthy volunteers

phase 2: Early exploratory and dose-finding studies in patients

phase 3: Large scale studies in patients

phase 4: Post-marketing safety monitoring

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

what are some considerations of clinical trials

A
•	Most Phase I volunteers are men
–	Women of reproductive age?
–	In EU, only post-menopausal or surgically sterilised women can participate
•	Genetic polymorphisms
–	Fast or slow metabolisers
–	Accumulation of drug in slow metabolisers
–	Consider the target population
–	Where is the drug to be marketed?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

what does phase 1a single dose trials entail?

A

• 4-8 cohorts of 6-8 participants
• Escalating dose from one cohort to the next
• Maximum tolerated dose determined
– Before safety / tolerability issues arise
– For low toxicity drugs, the highest practicable dose

• Establishes pharmacokinetic parameters
– Cmax:Peak drug / metabolite concentration
– Tmax: Time to peak drug / metabolite concentration
– AUC: to infinity and to specified time
• Establishes bioavailability and total exposure
– T1/2: rate of elimination
– VD: Volume of distribution
– MRT: Mean residence time
• – average time the drug remains in the body after administration

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

what does phase 1b

A
  • Similar participants to Phase Ia
  • Escalating repeat-dose
  • Establishes ‘steady state’ dosing requirements
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

what does phase iia trails entail?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

what does phase iib trails entail?

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

what does phase iii trails entail?

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

why do trails fail?

A
•	Insufficient biological activity
•	Unacceptable toxicity
•	Problems in the design of the trial
–	Measurement criteria (endpoint)
–	Patient selection
–	Sample size
–	duration
•	Problems in the execution of the trial
–	Randomisation of patients
–	Analysis of data
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

what does phase iv trails entail

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

what is the yellow card scheme?

A
  • Reports made by patients and health professionals
  • Assessed at the MHRA – medics, pharmacists and other scientists
  • If a new side effect is identified, information is considered in the context of the overall side effect profile for the medicine, and how the side effect profile compares with other medicines used to treat the same condition.
  • The MHRA takes action to ensure that medicines are used in a way that minimizes risk, while maximizing patient benefit
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

what is the Black triangle (▼) drugs and products

A

• Black Triangle symbol (▼) indicated in
– British National Formulary (BNF)
– British National Formulary for Children (BNFC)
– Monthly Index of Medical Specialities (MIMS)
– Association of the British Pharmaceutical Industry (ABPI) Medicines Compendium
– on advertising material
in Drug Safety Update

58
Q

how often is the black triangle drugs and products monitored?

A
  • Monitored for 2 years (usually) after marketing
  • Healthcare professionals are asked to report all suspected adverse drug reactions to these products through the Yellow Card Scheme
59
Q

what does the black triangle drugs and products tell us?

A
  • Relatively limited information about safety of new drugs from clinical trials in the UK
  • Trials generally involve only small numbers of eligible patients who take the medicine for a relatively short period of time
  • Patients in clinical trials may not be fully representative of those who will use the medicine when it is marketed
60
Q

what information does the black triangle products tells us?

A
  • Contains a new combination of active substances
  • New route of administration or drug delivery system
  • Significant new indication which may alter the established risk/benefit profile of that drug
  • Established medicine which is to be used in a new patient population
61
Q

problems with drugs are revealed by…?

A
  • Information supplied by manufacturers, including Phase IV clinical trials
  • Side effects revealed through Yellow Card Scheme
  • Alerts on newly marketed drugs through the Black Triangle Scheme
  • Research stored in the General Practice Research Database
  • Quality checks on products, including labelling and packaging carried out by MHRA inspectors
  • Tip-offs about criminal activity, such as the sale of fake (counterfeit) medicines
  • Monitored by Commission on Human Medicines (CHM)
62
Q

what are the

MHRA’s Defective Medicines Report Centre (DMRC)

A
•	Issues alerts 
–	Healthcare professionals
–	Hospitals
–	GP surgeries
–	wholesalers 
•	Tells them when a medicine is being recalled or when there are concerns about the quality that will affect its safety or effectiveness
63
Q

what recall actions are taken by the regulator for each class of drug?

A

Class 1: Life threatening: immediate recall required
e.g. Nurofen Plus Tablets containing rogue Seroquel XL 50mg tablets capsules

class 2: Harmful: recall is required within 48 hours
e.g. Tramadol 50mg capsules - risk of fungal contamination
class 3:Unlikely to harm patients: action required within 5 days
e.g. certain batches of a cream/pessary, used to treat fungal infections, such as thrush (Gyno-Pevaryl), because of errors in the patient information leaflet
class 4: No threat to patient safety: caution advised
e.g. Xenidate XL 36mg prolonged-release tablets (methylphenidate hydrochloride) - incorrect number of tablets in packs from a specific batch
64
Q

GVK Biosciences: European Medicines Agency recommends suspending medicines over flawed studies

A
  • Inspection revealed data manipulations of electrocardiograms (ECGs) during the conduct of some studies of generic medicines
  • Manipulations appeared to have taken place over a period of at least five years
  • Their systematic nature, the extended period of time during which they took place and the number of members of staff involved cast doubt on the integrity of the way trials were performed at the site generally and on the reliability of data generated at that site
65
Q

New medicines

A
  • Why are they so expensive?
  • Is big pharma just there for the money?
  • How many new medicines disasters have there been since 1960’s?
  • http://bmjopen.bmj.com/content/3/2/e002088.full.pdf
  • Is society better or worse off for it?
66
Q

what does the marketing authorisation do?

A

• To place a medicinal product on the market requires authorisation from an appropriate regulatory authority
– Medicines and Healthcare Products Regulatory Agency (MHRA)
• Demonstrate evidence
– Quality
– Safety
– Efficacy

67
Q

how do you make sure that the product work?

A
  • Have taken a lead activity molecule
  • Made structural modifications to achieve best possible pharmacodynamic and pharmacokinetic properties
  • Formulated it into an elegant and acceptable medicine
68
Q

what is meant by efficacy?

A

• Have to demonstrate convincingly that the new product is efficacious
– Produces the desired effect
• It provides the means for a therapeutic intervention that will be of benefit to the patient
– Including tolerability and acceptability

69
Q

what things do you have to consider to reduce the risk of something happening?

A

• The risk of an event occurring in a specified population
– E.g. a myocardial infarction in hypertensive patients
• Event rate = number of people experiencing an event (e.g. myocardial infarction) as a proportion of the population (e.g. people with hypertension)
– Commonly expressed as a %

70
Q

what is meant by risk reduction?

A
  • In the high risk group there is an absolute risk reduction of 10%
  • The relative risk reduction is 25%
  • In the low risk group there is an absolute risk reduction of 2.5%
  • The relative risk reduction is still 25%
  • Which indicator would you choose?
71
Q

How do we quantify the benefit?

A

E.g. A RCT is designed to determine the reduction in the risk of stroke of a new anti-hypertensive drug (loTens)
Patients on active treatment (Drug loTens)
1800 stokes occurred in 15000 patients over an average follow up period of 5 years
Patients on placebo treatment
3000 stokes occurred in 15000 patients over an average follow up period of 5 years

72
Q

what is LoTens results

A

Absolute risk for the treatment group (proportion of events in treatment group - ART)
i.e. strokes who received drug loTens
ART = 1800/1500 = 0.12

Absolute risk for the control group (proportion of events in control group -ARC)
i.e. strokes who received placebo
ARC = 3000/1500 = 0.2

Absolute Risk Reduction (ARR)
ARR = ARC - ART
ARR = 0.2 - 0.12 = 0.08 (or 8%)
i.e. Patient who receives loTens is 8% less likely to have a stroke over a 5 year period than the patient on placebo

Relative Risk Reduction (RRR)
RRR = ARC - ART
ARC
RRR = 0.2 - 0.12 = 0.4 (OR 40%)
0.2
i.e. if patient took drug loTens for 5 years they are 40% less likely to have a stroke than a patient on placebo
Are measuring the reduction of risk of the treatment group relative to the control group

73
Q

what is meant by absolute and relative difference

A
  • Average mark for an assessment for students who attended lectures on a census day = 40.3%
  • Average mark for an assessment for students who did not attended lectures on a census day = 31.4%
  • Absolute difference: 8.9%
  • Relative difference: attending students achieved 28.3% higher on average than non-attending
  • Which message is correct?
74
Q

what is meant by number needed to treat?

A

• The number of patients who would have to receive treatment for 1 of them to benefit
• Calculated as 100/absolute risk reduction
– High risk group example: need to treat 10 people for 1 person to benefit from it
– Low risk group example: need to treat 40 people for 1 person to benefit

75
Q

how do you demonstrate efficacy

A

• Provide robust evidence that the product will do what it purports to do
• Evidence has to be capable of being verified
– Needs to be scientific
– Capable of statistical analysis

76
Q

what is meant by a placebo

A

• A placebo is an inert substance or dosage form which is identical in appearance, flavor and odour to the active substance or dosage form. It is used as a negative control in a bioassay or in a clinical study

77
Q

considerations of a placebo?

A
  • Requires a comparison between two groups

* Use mathematical calculations to establish whether the behaviour of the two groups is the same or different

78
Q

what is meant by null hypothesis?

A
  • An essential tool in hypothesis testing
  • Hypothesis: Patients receiving drug X will show an improved clinical outcome compared to those receiving placebo
  • Null hypothesis: The clinical outcome for patients receiving drug X will be not be better than those receiving placebo
79
Q

what is meant by statistical conclusions?

A
  • The outcome of the trial disproves the null hypothesis

* i.e. the outcomes for the test group are different from those of the control group

80
Q

How would we set up a clinical trial?

A

• Recruit a number of patients with a condition
• Divide them into two
– Test group
– Control group
• At the end of the trial, compare outcomes
• Simple!????

81
Q

how can trails be flawed?

A
•	Statistical errors
–	The hypothesis
–	The statistical set up – power of the study
–	Analysis of data
•	Bias
–	Setting up the groups (selection bias)
–	Observation bias 
–	Confounding factors
82
Q

Central assumption made on results

A

• The outcomes from a trial will be applicable to the treatment population as a whole

83
Q

Identify the trial population

A
  • MUST mirror the treatment population

* Need to have criteria in place to match the trial population to the treatment population

84
Q

Identify the treatment population

A
•	Nature of the condition
–	Inclusion and exclusion criteria
•	Demographics
–	Age
–	Gender
–	Ethnicity
–	Social background (if it may affect the disease)
85
Q

what is meant by Inclusion criteria

A

• Identify the condition to be treated precisely
• Use clinical diagnosis and standardised measurement scales
– ADAS-cog (Alzheimer’s Disease Assessment Scale-cognitive subscale) designed to measure the severity of the most important symptoms of Alzheimer’s disease
– American College of Rheumatology (ACR) Core Data Set2–4 includes seven measures of activity or activity and damage

86
Q

what is meant by exclusion criteria?

A

• Exclude those for whom the treatment would not apply
– E.g. - Exclude type-1 diabetics from a trial for a drug for type-2 diabetes
• Identify factors that might interfere with the results
– Co-morbidities
– Other medications
– Factors affecting adherence to protocol

87
Q

what is meant by normal distribution

A

• A population, when measured against a criterion, will usually show some level of variance
• This variance is likely to show a distribution around an average value
• Trial population MUST mirror the treatment population in all criteria
• Trial population is divided into two
– Test or intervention group
– Control group

88
Q

what is meant by randomisation?

A

• For the outcome to be true, the test group must be the same as the control group in all respects
• Best way to allocate to test and control is random
– Can involve use of a random number generator
– Should be done by an independent person not involved in the trial

89
Q

what is meant by outcomes?

A
  • Test and control groups are the same at the start of the trial
  • Outcomes at the end of the trial for test and control are different if this can be demonstrated statistically
90
Q

what is meant by blind and double blind?

A

• The operators should not know what treatment an individual has had
– Easier to do with medicines than with physical procedures
– Highlights importance of the placebo
• Double-blind: neither the operators nor the patients know the treatment

91
Q

what is meant by cross over trials?

A

• Half-way through the trial the test and control groups are swapped over
– Requires measurements all the way through
– Trial usually runs for some time
– Trial can be stopped if it becomes clear that one treatment is significantly better than the other
• Applicable to certain conditions
– Pain relief
– Insomnia
– Some chronic conditions

92
Q

what is meant by power calculations?

A
  • The trial must be designed to be capable of disproving the null hypothesis
  • Needs to be of a size large enough to show that the outcomes for the two comparison groups are statistically different from each other
  • Large enough to eliminate the play of chance
93
Q

how is power calculations determined?

A

• What kind of statistical test is being performed
• Sample size
– the larger the sample size, the larger the power
– increasing sample size = increased costs
• The size of experimental effects
– If the null hypothesis is wrong by a substantial amount, power will be higher than if it is wrong by a small amount.
• The level of error in experimental measurements
– Error = ‘noise’

94
Q

what is meant by intention to treat?

A

• ITT analysis includes all randomised patients in the groups to which they were assigned
– Regardless of treatment received or whether they completed the trial
– Participants could drop out due to adverse effects / unacceptability of treatment etc
• A high drop-out rate compromises the power of the trial and could hide a confounding factor
• ITT reduces the risk of bias in the analysis of data

95
Q

what is meant by on-treatment analysis?

A

• Seen in older clinical trials
• Analyse data from patients who complete the trial
– Drop-outs not accounted for

96
Q

what is meant by consistency in data collection?

A

• All participants must be treated in the same way
– Data collected according to the same protocol and at the same times
– Deviation can lead to bias

97
Q

what is meant by precision?

A

• How precise are the results?
• Data analysis should give statistically meaningful indications of the precision obtained
– Confidence intervals
– Coefficient of variance

98
Q

what is meant by the impact of the outcome of a trail?

A

• Outcome of the trial should relate to the overall health benefit of the therapy
– Relative risk reduction
– Absolute risk reduction
– Numbers needed to treat

99
Q

what is adverse events

A

• Can also be measured
• Number needed to harm
– Number of patients who would have to receive the treatment for 1 of them to experience an adverse event

100
Q

list the hierarchy of evidence?

A
  1. Systematic review of randomized trials
  2. Randomized trial or observational study with dramatic effect
  3. Non-randomized controlled cohort/follow-up study
  4. Case-series, case-control studies, or historically controlled studies
  5. Mechanism-based reasoning
101
Q

Sources of information

A
•	Primary and secondary literature
–	Requires subscriptions
–	Can be time consuming
–	Gives minutiae of detail 
•	NHS website  http://www.nhs.uk/pages/home.aspx 
–	Good at an overview level
•	Cochrane database
–	Source of detailed information on meta-analysis of clinical trials
•	NICE guidelines
102
Q

• what is

Critical appraisal skills programme (CASP)

A
  • How do we know which treatments or interventions really work?
  • How can you tell whether a piece of research has been done properly and that the information it reports is reliable and trustworthy?
  • How can health care commissioners know which treatments or services are truly worth funding?
  • How can patients decide whether the benefits of a particular intervention are likely to outweigh the harms and costs?
  • How can you decide what to believe when making a health care decision, when research on the same topic comes to different conclusions?
103
Q

what are the steps in making appraisal?

A
•	Is the study valid?
–	What type of study is it?
–	Is there evidence of bias?
–	Are the methods used OK?
•	What are the results?
–	Are the results significant?
•	Are the results useful?
–	Do they apply to your population of patients?
104
Q

what is the nature of the evidence?

A
•	A scientific study
–	Animals, in vitro
•	Observational studies
–	Case study
–	Cohort study
–	Cross sectional study
•	Clinical trial
•	Meta-analysis or systematic review
105
Q

CASP checklist for RCTs

A
•	Section A
–	Is the study design valid?
•	Section B
–	Is the study methodologically sound?
•	Section C
–	What are the results?
•	Section D
–	Will the results help locally?
106
Q
  1. Did the trial address a clearly focussed research question?
A

• Was the study designed to assess the outcomes of an intervention?
• Is the research question ‘focused’ in terms of:
– Population studied
– Intervention given
– Comparator chosen
– Outcomes measured

107
Q
  1. Was the assignment of patients to treatments randomised?
A
  • How was randomisation carried out? Was the method appropriate?
  • Was randomisation sufficient to eliminate systematic bias?
  • Was the allocation sequence concealed from investigators and participants?
108
Q
  1. Were all participants who entered the study accounted for at its conclusion?
A
  • Were losses to follow-up and exclusions after randomisation accounted for?
  • Were participants analysed in the study groups to which they were randomised (intention-to-treat analysis)?
  • Was the study stopped early? If so, what was the reason?
109
Q
  1. Was the study blinded?
A
  • Were the participants ‘blind’ to intervention they were given?
  • Were the investigators ‘blind’ to the intervention they were giving to participants?
  • Were the people assessing/analysing outcome/s ‘blinded’?
110
Q
  1. Were the study groups similar at the start of the randomised controlled trial?
A

• Were the baseline characteristics of each study group clearly set out?
– Age
– Sex
– Socio-economic group
• Were there any differences between the study groups that could affect the outcome/s?

111
Q
  1. Apart from the experimental intervention, did each study group receive the same level of care?
A

• Were they treated equally?
• Was there a clearly defined study protocol?
• If any additional interventions were given (e.g. tests or treatments), were they similar between the study groups?
– Tests
– Treatments
• Were the follow-up intervals the same for each study group?

112
Q
  1. Were the effects of intervention reported comprehensively?
A
  • Was a power calculation undertaken?
  • What outcomes were measured, and were they clearly specified?
  • How were the results expressed?
  • For binary outcomes, were relative and absolute effects reported?
  • Were the results reported for each outcome in each study group at each follow-up interval?
  • Was there any missing or incomplete data?
  • Was there differential drop-out between the study groups that could affect the results?
  • Were potential sources of bias identified?
  • Which statistical tests were used?
  • Were p values reported?
113
Q
  1. Was the precision of the estimate of the intervention or treatment effect reported?
A
  • What statistically rigorous measures of precision were used?
  • Were confidence intervals (CIs) reported?
114
Q
  1. Do the benefits of the experimental intervention outweigh the harms and costs?
A

• What was the size of the intervention or treatment effect?
• Were harms or unintended effects reported for each study group?
• Was a cost-effectiveness analysis undertaken?
– Comparison between different interventions used in the care of the same condition or problem

115
Q
  1. Can the results be applied to your local population/in your context?
A
  • Are the study participants similar to the people in your care?
  • Would any differences between your population and the study participants alter the outcomes reported in the study?
  • Are the outcomes important to your population?
  • Are there any outcomes you would have wanted information on that have not been studied or reported?
  • Are there any limitations of the study that would affect your decision?
116
Q
  1. Would the experimental intervention provide greater value to the people in your care than any of the existing interventions?
A
  • What resources are needed to introduce this intervention taking into account time, finances, and skills development or training needs?
  • Are you able to disinvest resources in one or more existing interventions in order to be able to re-invest in the new intervention?
117
Q

CASP checklist for meta-analyses

A
  1. Did the review address a clearly focused question?
  2. Did the authors look for the right type of papers?
  3. Do you think all the important, relevant studies were included?
  4. Did the review’s authors do enough to assess quality of the included studies?
  5. If the results of the review have been combined, was it reasonable to do so?
  6. What are the overall results of the review?
  7. How precise are the results?
  8. Can the results be applied to the local population?
  9. Were all important outcomes considered?
  10. Are the benefits worth the harms and costs?
118
Q

Key elements in RCTs

A
•	Choice of intervention and control
•	Choice of treatment population
–	Inclusion and exclusion criteria
•	Stratification and randomisation of participants
•	Outcome measures
•	Trial protocol
–	Intention to treat
•	Statistical analysis
–	Confidence intervals
•	Summary outcome data
–	Absolute and relative risk reduction, NNT
119
Q

what is the structure of a paper?

A
•	Abstract
	Summary of the paper
•	Methods
	What they did
•	Results
	What they found
•	Discussion
120
Q

the relevance of this work

A

Methods
• Why they did the trial
– Evaluate the effects of potential treatments in patients hospitalized with Covid-19
• Inclusion and exclusion criteria
– Hospitalized patients with clinically suspected or laboratory confirmed SARS-CoV-2 infection
– Excluding those with medical history that might put them at substantial risk

121
Q

the method section what does it contain?

A

• Consent
• Principles for conducting the trial, ethics approval
• Randomization
– 2 groups: standard care alone, or with addition of dexamethasone
• Procedures
– Recording of the outcomes
– Plus any other relevant information

122
Q

Methods -outcome measures

A
•	Primary outcome
–	all-cause mortality within 28 days
•	Secondary outcomes
–	Time until discharge
–	Additional treatments needed
•	Dialysis, haemofiltration, ventilation, cardiac arrythmias
–	Cessation of additional treatments
•	Ventilation
123
Q

Methods – statistical analysis

A

• Power calculation difficult to do from start
• Once trial was underway could estimate number of patients needed in order to achieve power of 90% and P value = 0.1
– If 28 day mortality was 20%, would need 2000 pts in intervention group and 4000 in the normal care group
• Specified statistical methods to be used

124
Q

what does the discussion contain?

A

• Was the intervention successful?
• Does the data provide compelling evidence for treatment?
• Would you recommend it?
• Can you work out some summary statistics?
– Risk reduction (absolute and relative)
– NNT

125
Q

name 3 regulators

A

MHRA

  • FDA
  • European medicines agency
126
Q

what 3 centres are formed in the MHRA?

A

– the Clinical Practice Research Datalink (CPRD), a data research service that aims to improve public health by using anonymised NHS clinical data
– the National Institute for Biological Standards and Control (NIBSC), a global leader in the standardisation and control of biological medicines
– the Medicines and Healthcare products Regulatory Agency (MHRA), the UK’s regulator of medicines, medical devices and blood components for transfusion, responsible for ensuring their safety, quality and effectiveness

127
Q

what does the MHRA do?

A
  • Ensuring that medicines, medical devices and blood components for transfusion meet applicable standards of safety, quality and efficacy
  • Ensuring that the supply chain for medicines, medical devices and blood components is safe and secure
  • Promoting international standardisation and harmonisation to assure the effectiveness and safety of biological medicines
  • Helping to educate the public and healthcare professionals about the risks and benefits of medicines, medical devices and blood components, leading to safer and more effective use
  • Supporting innovation and research and development that’s beneficial to public health
  • Influencing UK, EU and international regulatory frameworks so that they’re risk-proportionate and effective at protecting public health
128
Q

what do the FDA do?

A
  • Development of regulatory policy
  • Establishment of regulations and guidance document in support of the FDA act
  • Review and approval of pre-marketing submissions for drugs and devices
  • Inspection of facilitates and products with follow-up enforcement actions, where necessary
  • Ensuring that labeling, packaging and promotional material is truthful, informative and not misleading
  • Participation in international initiatives on global harmonisation
129
Q

what is the CDER responsible for?

A

• The centre is responsible for
– Reviewing application for new and generic pharmaceuticals
– Manages US GMP regulations for pharmaceutical manufacturing
– Determines which medications require a medical prescription
– Monitors advertising of approved medications
– Collects and analyses safety data about pharmaceuticals already in the market

130
Q

what does the EMA do?

A
  • facilitates developments and access to medicines
  • evaluate applications for marketing authorisation
  • provide reliable information on human and veterinary medicines in any language
131
Q

Requirement for an IND application

A

• An IND application is required if:
– A sponsor intends to conduct a human clinical investigation with an investigational new drug
• A pre-IND meeting can be arranged with the regulator to discuss:
– The design of animal research, which is required to lend support to clinical studies
– Intended protocol for conducting the clinical trial
– Chemistry, manufacturing and control of investigational drug

132
Q

IND content?

A

• IND application contains
– Cover sheet (Form FDA-1571)
– Table of contents
– Part A – Introductory statement and general investigational plan
– Part B – Protocols
– Part C – Chemistry, manufacturing and control information
– Part D – Pharmacology and toxicology information
– Part E – Previous human experience with the investigational drug
– Part F – Additional information

133
Q

Clinical Trials Authorisation (CTA) of Investigational Medicinal Products (CTIMP)

A

• “Do the data supplied support the use of this product, administered in this way, in the proposed dose for the proposed duration, to this ‘type’ of participant?”

134
Q

marketing authorisation

A

Common technical document (CTD) New drug application (NDA)

135
Q

Module 3: Quality

A
•	Identity
–	Recommended International Non-proprietary Name (INN)
•	Characterisation
–	Structural information
–	Chemical and physical properties
–	Spectral/chromatographic properties
•	Manufacturing process 
•	Control 
•	Stability
136
Q

Module 4: Non-clinical

A
•	Pharmacology
–	Primary, secondary pharmacology
–	Safety pharmacology
–	Pharmacological drug interactions
•	Pharmacokinetics (PK)
–	Analytical methods and validation
–	ADME
–	Pharmacokinetic drug interactions
137
Q

• Toxicology

A
–	Single and double dose
–	Genotoxicity: in vitro and in vivo
–	Carcinogenicity: medium to long-term studies
–	Reproductive and Development Toxicity
–	Local tolerance
–	Other toxicity studies
•	Antigenicity
•	Immunotoxicity
•	Metabolites and impurities
138
Q

Module 5: Clinical

A

• Biopharmaceutical studies
– Bioavailability
– Bioequivalence
– In vitro to in vivo correlations
– Bioanalytical methods for human studies
• Pharmacokinetics studies using human biomaterials
– Plasma protein binding studies
– Hepatic metabolism and drug interactions
– Using other human biomaterials

139
Q

Human pharmacokinetic (PK) studies

A
–	Subject PK and tolerability study reports
–	Intrinsic and extrinsic factor  
•	Human pharmacodynamic (PD) studies
–	Healthy subject PD
–	Patient PD
•	Efficacy and Safety reports
–	Controlled clinical studies to claimed indication
–	Uncontrolled clinical studies
–	Meta-analysis
140
Q

Module 2: Summaries

A
  • Non-clinical and clinical Overviews should present critical discussions and assessments of results with conclusions
  • Non-clinical and clinical Summaries should provide abbreviated factual data present as written summaries and support by summary tables