Facts Flashcards

1
Q

Stages of preclinical development

A

• M0: initial definition of a program;
• M1: selection of a drug candidate and of an indication;
• M2: first administration to humans;
• M3: first clinical efficacy data in humans (proof-of-concept

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

Washout period in cross over design

A

‘Sufficient washout periods ’ are often recommended to reduce or eliminate carryover effects. In practice it can be difficult to define sufficient. For a pharmacologic intervention, knowledge of kinetics can be valuable in the planning stages. For example, after seven half-lives less than 1% of the agent remains; at this time point meaningful pharmacologic carryover is removed for many drugs. If the effect of the agent is reasonably rapid and the outcome is closely related to physiological concentration then this might be all of the information that is needed. Other situations might be more complicated if pharmacodynamic effects persist beyond the physical elimination of the drug.

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

Types of two period designs to show disease modifying treatment effect

A

Withdrawal design
Delayed start design
Combination of withdrawal and delayed start design

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

Definition of term parameter

A

The term parameter refers to a numerical characteristic of a statistical population. In contrast, the term statistic refers to a value calculated in a sample. We are particularly interested in a type of statistic known as an estimate. A statistical estimate is a direct reflection of an underlying population parameter.

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

What is the statistical population

A

The statistical population is the entire collection of values (the “universe” of values) about which we want to draw conclusions.

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

Definition of power

A

Confidence is the complement of α, and power is the complement of β. Confidence ≡ 1 − α Power ≡ 1 − β

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

Selection of endpoints in middle stage development depends on what

A

Endpoint selection in early efficacy trials depends on the nature of the drug effect expected, previous experience with measurement scales used in the disease state, and the kind of decision problem faced by the study team. h e specii c endpoints selected should balance the need to measure the ef ect of the drug on the disease state, provide some initial reassurance that the drug ef ect is clinically meaningful, and have adequate operating characteristics for studies that typically are of somewhat smaller sample size.

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

Why is adequate washout period important for crossover design

A

Unwanted bias in the estimated treatment effect attributable to a carryover effect has already been described (‘Unequal carryover’).
The carryover effect can be found through data analysis, and when this effect is significant, it is difficult to interpret the treatment effect. Therefore, a protocol such as the above-mentioned washout period setting is an appropriate method to eliminate this effect.

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

Purpose of treatment withdrawal for checking disease modifying effects

A

h e purpose of the withdrawal maneuver is to determine whether any portion of the treatment ef ect that is evident at the end of Period 1 persists at er withdrawal of treatment, i.e., to distinguish between the short-term symptomatic ef ect and the disease-modifying ef ect.

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

What does the delayed start design share with the withdrawal design?

A

The delayed start design shares with the withdrawal design the potential problem that there is no blinding with respect to the treatment received during Period 2.

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

What are some limitations of two period designs

A

h ere are several limitations that accompany the use of two-period designs to determine whether or not an intervention has disease-modifying ef ects. Many of these have already been discussed, including the assumptions that: 1) Period 1 is long enough for a detectable disease-modifying ef ect to become apparent; 2) the disease-modifying ef ect acquired over the duration of Period 1 remains with the participant at least through the end of Period 2, but presumably longer; 3) Period 2 is long enough for the symptomatic ef ect from Period 1 to completely disappear by the end of Period 2; 4) withdrawal of active treatment does not modify (e.g., hasten) the disease process in some way (withdrawal design); and 5) the total (symptomatic + disease-modifying) ef ect of treatment received in Period 2 is independent of whether or not the participant received treatment during Period 1 (delayed start design), implying that Period 2 is long enough for the symptomatic ef ect of the treatment to become fully apparent.

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

Enrichment trial design

A

Clinical trials using enrichment designs involve at least two periods ( Figure 12.1 ). In the i rst period, the enrichment period, subjects are screened for their responsiveness according to predetermined criteria (e.g. a 30% reduction in baseline pain intensity). h ese criteria vary depending on the type of study being performed. Researchers ot en use the putative response to the treatment to be studied in the subsequent phase of the trial as a direct screening tool during the enrichment period. However, some researchers use other screening criteria such as biomarkers that may indicate potential response to the intervention. h is may be particularly useful when there is a biomarker that can be identii ed in the short term that predicts response to long-term treatment

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

Evidence that an association is cause and effect

A

What aspects of the research findings support cause and effect when only observational studies are available? In 1965, the British statistician Sir Austin Bradford Hill proposed a set of observations that taken together help to establish whether a relationship between an environmental factor and disease is causal or just an association.

Cause precedes effect
large relative risk
larger exposure to cause association with higher rates of disease
Reduction in exposure is followed by lower rates of disease
Repeatedly observed by different persons, in different places, circumstances and times
Makes sense according to biologic knowledge of the time
one cause leads to one effect
Cause and effect relationship already established for a similar exposure of disease

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

Definition of surrogate endpoint

A

A biomarker that is intended to substitute for a clinical endpoint. A surrogate endpoint is expected to predict clinical benei t or harm or lack of benei t or harm based on epidemiologic therapeutic pathophysiologic or other scientii c evidence. Surrogate endpoints are a subset of biomarkers. h e term surrogate literally means ‘substitute of ’ therefore the NIH working group discourages the use of the term surrogate marker because it suggests the substitution is for a marker rather than for a clinical endpoint.

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

Reasons why a surrogate endpoint may fail to be valid

A

h e i rst is that the surrogate outcome is not in the causal pathway between the disease and the true clinical outcome. h e second, related, possibility is that there is more than one causal pathway and the potential surrogate is only relevant to one of these pathways. h e third possibility is that the surrogate is not in the pathway of the intervention, or is insensitive to it. Finally, the intervention may have a mechanism of action that is independent of the disease process. h is last scenario may be most commonly observed in the case of (potentially harmful) side ef ects of treatment. A i t h possibility proposed by Frank and Hargreaves is that the biomarker may be overly sensitive and not correlated with a meaningful clinical phenotype [ 9 ] . In this case, improvements in the biomarker may be demonstrated, but would not be associated with health benei ts.

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

Considerations for developing drug in pediatric population

A

The decision to proceed with a pediatric development program for a medicinal product, and the nature of that program, involve consideration of many factors, including:
* The prevalence of the condition to be treated in the pediatric population
* The seriousness of the condition to be treated
* The availability and suitability of alternative treatments for the condition in the pediatric population, including the efficacy and the adverse event profile (including any unique pediatric safety issues) of those treatments
* Whether the medicinal product is novel or one of a class of compounds with known properties
* Whether there are unique pediatric indications for the medicinal product * The need for the development of pediatric-specific endpoints
* The age ranges of pediatric patients likely to be treated with the medicinal product
* Unique pediatric (developmental) safety concerns with the medicinal product, including any nonclinical safety issues
* Potential need for pediatric formulation development Of these factors, the most important is the presence of a serious or life-threatening disease for which the medicinal product represents a potentially important advance in therapy. This situation suggests relatively urgent and early initiation of pediatric studies.

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

Alternative hypothesis

A

The alternative hypothesis is closely aligned with the research hypothesis. The objective of the test is to seek evidence against the null hypothesis as a way of bolstering the alternative hypothesis, and thus the research hypothesis.

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

P value

A

We convert the z-statistic to a probability statement called the P-value. The P-value answers the question: “If the null hypothesis were true, what is the probability of the observed test statistic or one that is more extreme?” A small P-value indicates that observed data are unlikely to have come from the distribution suggested by H0.

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

One sample z-test

A

The one-sample z-test compares a mean from a single sample to an expected value. Several conditions must exist for the test to be used. These include: • The variable must be quantitative. • Measurements must be valid. •Data must be derived by an SRS or reasonable approximation thereof.

The standard deviation of the population (σ) must be known prior to data collection. •The sampling distribution of is approximately Normal.

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

Type 1 error

A

an erroneous rejection of a true null hypothesis

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

Type 2 error

A

an erroneous retention of a false null hypothesis

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

Confidence interval

A

Each of these confidence intervals has a 95% chance of capturing the population’s true mean weight μ.

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

What does confidence mean

A

What confidence means. The confidence level of an interval tells you how often the method will succeed in capturing μ in the long run. With repeating samples, (1 − α)100% of the intervals will capture μ and (α)100% will not.

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

Confidence interval reflects what?

A

Confidence interval length reflects the precision of the estimate. Narrow intervals reflect precision; wide intervals reflect imprecision.

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

Relationship Between Hypothesis Testing and Confidence Intervals

A

You can use a (1 − α)100% confidence interval for μ to predict whether a two-sided test of H0: μ = μ0 will be significant at the α-level of significance. When the value of the parameter identified in the null hypothesis (μ0) falls outside the interval, the results will be statistically significant (reject H0).

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

Accuracy

A

The accuracy of the measurement refers to how close the measured value is to the true value. It is otherwise called validity.

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

Precision

A

On the other hand, precision refers to how close a set or a group of measurements are together, which is otherwise called reliability.

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

Student t-distribution

A

When the sample size is small, the central limit theorem does not hold well. Exact sampling distributions are often derived for inferring from small samples. A classical example of the exact sampling distribution is Student’s t-distribution, which characterizes the sample mean for small samples.

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

Adverse event

A

An adverse event (AE), as defined by Good Clinical Practice, is any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease
having been absent at baseline, or, if present at baseline, appears to worsen AND is temporally associated with medical treatment or procedure, REGARDLESS of the attribution (i.e., relationship of event to medical treatment or procedure).
Adverse Event: FDA Definition

FDA defines an adverse events as any untoward medical occurrence associated with the use of a drug in humans,
whether or not considered drug related.

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

Phase II

A

Phase II studies may be designated ‘Phase IIa’ or ‘Phase IIb’. Phase IIa usually indicates a single-arm trial designed to show initial evidence of clinical efficacy (‘proof of concept’). Phase IIb indicates a more reliable assessment of efficacy in a larger number of patients, often randomized with a control therapy group.

Excerpt from: “Fast Facts: Clinical Trials in Oncology: The fundamentals of design, conduct and interpretation” by A. Hackshaw. Scribd.
This material may be protected by copyright.

Read this book on Everand: https://www.everand.com/book/490178989

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

Pick the winner

A

With this design, patients are randomly allocated to two or more experimental treatment arms, which could be for different drugs, combinations of drugs or doses of the same drug. The primary aim is to see which group has the highest response or most favorable markers of activity/efficacy, rather than to directly compare the experimental arms. The chosen arm (sometimes two arms) is likely to become the treatment arm for future trials, taking toxicity into account. Two-stage designs can also be used.

Excerpt from: “Fast Facts: Clinical Trials in Oncology: The fundamentals of design, conduct and interpretation” by A. Hackshaw. Scribd.
This material may be protected by copyright.

Read this book on Everand: https://www.everand.com/book/490178989

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

Placebo in phase II

A

Use of a placebo can strengthen Phase II trial results and conclusions, especially when there are subjective endpoints (for example, tumor response and disease progression) that can be influenced by the lack of blinding in open-label trials. However, the cost of producing and distributing placebo in a Phase II trial may outweigh the scientific benefits, and investigators accept the potential for bias, with the intention of using placebo in a subsequent Phase III study.

Excerpt from: “Fast Facts: Clinical Trials in Oncology: The fundamentals of design, conduct and interpretation” by A. Hackshaw. Scribd.
This material may be protected by copyright.

Read this book on Everand: https://www.everand.com/book/490178989

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

Basket trial

A

Basket trials involve testing a single drug targeted at a specific marker (for example, a mutation) in several tumor types or subtypes (one drug in several cancers).

Excerpt from: “Fast Facts: Clinical Trials in Oncology: The fundamentals of design, conduct and interpretation” by A. Hackshaw. Scribd.
This material may be protected by copyright.

Read this book on Everand: https://www.everand.com/book/490178989

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

Umbrella trial

A

Umbrella trials categorize patients with a single tumor type into different groups according to a biomarker or mutation; each group receives a targeted agent for that biomarker (several drugs in one cancer).

Excerpt from: “Fast Facts: Clinical Trials in Oncology: The fundamentals of design, conduct and interpretation” by A. Hackshaw. Scribd.
This material may be protected by copyright.

Read this book on Everand: https://www.everand.com/book/490178989

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

Analysis of single and pick the winner trials

A

Single-arm and ‘pick the winner’ randomized trials require mainly descriptive analyses, in which the efficacy endpoint data are summarized for each group separately.

Excerpt from: “Fast Facts: Clinical Trials in Oncology: The fundamentals of design, conduct and interpretation” by A. Hackshaw. Scribd.
This material may be protected by copyright.

Read this book on Everand: https://www.everand.com/book/490178989

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

Target validation

A

Employing genetically modified animals for target validation is an appealing methodology as it permits the scrutiny of the phenotypic consequences of gene manipulation. Development of knock-outs, knock-ins, conditional knock-outs, and transgenic animals are instances of genetically modified animals.

Excerpt from: “Pharmaceutical Medicine and Translational Clinical Research” by Academic Press. Scribd.
This material may be protected by copyright.

Read this book on Everand: https://www.everand.com/book/363365858

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Allosteric modulation

A

Allosteric modulation of drug targets is another novel approach towards drug discovery, wherein drugs bind at binding sites of the biological target—which are distinct from the active sites. A benefit with this approach is that while active sites might be common in several proteins, the allosteric sites might be unique, which allows for selective targeting and consequently either fewer or target-specific adverse effects.

Excerpt from: “Pharmaceutical Medicine and Translational Clinical Research” by Academic Press. Scribd.
This material may be protected by copyright.

Read this book on Everand: https://www.everand.com/book/363365858

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

DSUR

A

The Development Safety Update Report (DSUR) is used to provide periodic safety information during preapproval and for marketed drugs that are under further study, and the Periodic Safety Update Report (PSUR) is intended to provide an evaluation of the risk-benefit balance of a medicinal product at defined time points during the postapproval phase.

Excerpt from: “Pharmaceutical Medicine and Translational Clinical Research” by Academic Press. Scribd.
This material may be protected by copyright.

Read this book on Everand: https://www.everand.com/book/363365858

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

AEDR

A

Adverse event data review

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

dominant negative mutations

A

the product encoded by the mutated gene
adversely affects the usual gene product within the same cell (for example, in
Marfan syndrome, the abnormal fibrillin-1 protein encoded by the mutant allele
interferes with the protein encoded by the normal allele, with widespread effects
on connective tissue)

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

gain-of-function mutations

A

he encoded protein new or enhanced
activity (for example, gain-of-function mutations in STAT1 and STAT3, which en-
code signal transducer and activator of transcription 1 and 3, have been impli-
cated in different clinical phenotypes, including chronic mucocutaneous can-
didiasis, increased occurrence of viral, fungal and bacterial infections, and au-
toimmunity)

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

Gene silencing can be used in what diseases?

A

gain-of-function diseases to inhibit the func-
tioning of a mutated protein and so ameliorate disease symptoms. Certain classes
of silencing agent can also be used to produce truncated proteins that bypass the
pathogenic genetic information, producing a semifunctional copy of the affected
gene via exon skipping.

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

RNA interference

A

RNA interference (RNAi) can lead to the highly specific targeted knockdown of ex-
pression of a disease gene. RNAi is typically mediated by either chemically synthe-
sized double-stranded siRNA or vector-based short hairpin RNA (shRNA). Besides
more efficient and targeted delivery, the added advantages of shRNAs over siRNAs
are linked to enhanced specificity and the ability to use promoters to drive in-
ducible expression.

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

Mechanism of RNA interference

A

RNA interference (RNAi)-induced post-transcriptional
gene silencing. Viral vectors deliver short hairpin (sh)RNA transgenes into the nu-
cleus for expression and processing by the Drosha enzyme. The shRNA is exported
into the cytoplasm via exportin 5. Association with the Dicer enzyme leads to re-
moval of the loop sequence and a small interfering (si)RNA is produced. siRNAs
can also be directly delivered into the cytoplasm as synthetic short duplexes
through endocytosis. However it is generated, the siRNA is recognized by Dicer
and primed to associate with RNA-induced silencing complex (RISC). This acti-
vates argonaute, the RNAse component of the RISC, leading to destruction of one
of the RNA strands. Through design, the remaining single-stranded RNA will bind
to complementary messenger (m)RNA, forming a double-stranded RNA that
undergoes RISC degradation, resulting in targeted knockdown of the gene.

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

Two ways to use antisense
oligonucleotides to inhibit gene expression.

A

One is to target critical regulatory re-
gions, such as the translational start codon of the mature mRNA or the polyadeny-
lation signal in the 3’ UTR. An alternative method for inhibiting gene expression
would be to design antisense oligonucleotides to skip exons to result in an out-of-
frame transcript. This brings a PTC into frame, resulting in NMD of the transcript
and knockdown of pathogenic protein expression.

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

Targeting non-coding RNA

A

Small non-coding (nc)RNAs – miRNAs – and long
non-coding (lnc)RNAs can be used alone or together to regulate transcription and
post-transcriptional processes and to direct chromatin-modifying complexes.⁴
Chromatin regulates gene expression and normally acts to inhibit gene expression
through condensation of the DNA. Short (21–23 nucleotides) double-stranded
miRNAs manipulate gene expression in a similar way to siRNAs (see Figure 2.1) –
they act to silence gene expression through the degradation of RNA and prevention
of translation. An siRNA targets a single gene, while an miRNA has multiple gene
targets.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Do viral vectors for gene therapy cross the BBB?

A

A feature of some AAV (adeno-associated virus)
serotypes

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

Neuronal specificity of viral vectors?

A

A feature of some AAV
serotypes

Can help avoid
glial-cell-mediated immune
response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

Packing capacity of viruses

A

Adenovirus 7.5 kb
AAV 4.5 kb
Herpesvirus >30 kb
Lentivirus 8 kb

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

Advantages of adenovirus

A
  • DNA genome episomally resides in host nucleus, preventing insertional
    mutagenesis
  • Oncolytic*
  • Vaccination†
  • Able to transduce quiescent and proliferating human cells
  • Strong immunogenicity – can enhance antitumor immunity and therapeutic
    efficacy
  • Possible to produce 10–50 L
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

Advantages of AAV

A
  • Capsid determines tropism and transduction profile in different cell types
  • AAV9 can cross the blood–brain barrier and transduce cells of the CNS
    following a single iv injection
  • Can transduce both dividing and non-dividing cells
  • Recombinant viral genome stays in host nucleus, predominantly as episome
  • Single or multiple copies of vector genome can circularize in head-to-tail or
    head-to-head configurations in host nucleus, enhancing stability of the episomal
    viral DNA genome and mediating long-term transgene expression in non-dividing
    cells (e.g. muscle fibers)
  • No known disease associated with infection
  • Low toxicity
  • Long-term expression
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Advantages of Herpes Simplex Virus

A
  • Efficient infectivity in multiple cell types
  • Naturally neurotropic‡
  • Valuable for oncolytic virotherapy
  • Has carrying capacity for multiple genes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

Advantages of retroviruses

A
  • Effective integration into target cell chromatin
  • Envelope glycoproteins amenable to genetic modification to specify tissue
    and/or tropism
  • Genomic integration ensures the stability of transgene and persistent transgene
    expression in daughter cells following genome replication and cell division
  • Production can be scaled up and preparations banked (RCR free)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

Advantages of Lentiviruses

A
  • Efficient transduction in non-dividing cells
  • Long-term expression
  • Application in adoptive T-cell platforms
  • Envelope glycoproteins amenable to genetic modification to specify tissue
    and/or tropism
  • Genomic integration ensures the stability of transgene and persistent transgene
    expression in daughter cells following genome replication and cell division
  • Large-scale clinical production possible
    *Infect and kill tumor cells.
    †High immunogenicity induces a strong humoral T-cell response, tending toward
    a T helper 1 type response, to the transgene expressed by the vector.
    AAV, adeno-associated virus; CNS, central nervous system; iv, intravenous.
    ‡After initial infection via skin or mucous membranes, herpes simplex virus is
    taken up by sensory nerve terminals, travels along nerves to neuronal cell bodies
    and delivers its DNA genome into nuclei for replication.
    RCR, replication-competent retrovirus.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Disadvantages of adenoviruses

A
  • Pre-existing immune and inflammatory response
  • Complex for manipulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

Disadvantages of AAV

A
  • T-cell response
  • Antibody neutralization
  • Low DNA packaging capacity
  • Complex downstream processing complicates manufacturing
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

Disadvantages of herpes viruses

A
  • Lack of long-term expression
  • Poor patient experience
  • Low titers limit large-scale production
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Disadvantages of retroviruses

A
  • Insertional mutagenesis
  • Limited ex vivo application
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Disadvantages of lentiviruses

A
  • Insertional mutagenesis
  • Limited cassette size
    AAV, adeno-associated virus.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Major risk of AAV?

A

The major risk factor associated with adenoviral vectors is
the strong immune neutralization against the standard serotypes used. This could
be bypassed with adenoviral vectors derived from other primate species and these
are now being explored.
Certain serotypes of AAV vectors have restricted patient applicability; previous
infectious exposure to the viral capsid proteins results in seropositive neutralizing
antibody. Rare AAV serotypes are being identified and artificial serotypes engi-
neered to overcome this problem.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Transgene expression

A

The maximal therapeutic efficacy of a gene therapy is
achieved through controlled targeted expression, with the avoidance of side effects
associated with off-target and non-physiological expression. Modification of
tropism displayed by a vector is possible through engineering of the viral vector
surface proteins, together with the use of tissue-specific promoters. It is also pos-
sible to include elements within the expression cassette that allow regulation by the
host cell itself or that provide drug-inducible expression. It should be noted that
unique features, either structural or biochemical, displayed by diseased cells can
be exploited to restrict expression to where it is needed.

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

Basket trial

A

Basket trials refer to designs in which a targeted therapy is evaluated on multiple diseases that have common molecular alternations.

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

Umbrella trial

A

Umbrella trials, on the other hand, evaluate multiple targeted therapies for a single disease that is stratified into subgroups by molecular alternation.

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

Types of bias

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

ITT

A

In an ITT analysis, the study treatment group includes all patients who were assigned (usually randomized) to that study treatment group, even those who dropped out or received incomplete or inaccurate doses of the treatment. Even patients who never received a single dose of the treatment belong in the study group. There is a saying to describe ITT: “analyze as randomized.” So once patients are randomized, they are “stuck” in the analysis. Nothing that happens after randomization (e.g., noncompliance, protocol deviations, or dropping out) can exclude them being included in the analysis.

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

mITT

A

A modified ITT analysis includes all randomized patients that meet a specific minimum standard or simple set of criteria. The criteria should be simple, objective, and very straightforward. The criteria should not be related to the outcome (e.g., if you know that patients with lower socioeconomic status will be less likely to respond to your intervention, you cannot use socioeconomic status as a criterion). Using subjective criteria defeats the purpose of a modified ITT. The more common modified ITT will include all patients who received at least one dose of the study intervention (regardless of what happened to the patient after the initial dose). The study protocol should pre-specify a modified ITT analysis and its criteria. Otherwise, selection bias can occur (e.g., if you find that a certain characteristic correlates with poor intervention response, you could later exclude patients with that characteristic from the analysis.) Another common modified ITT analysis excludes all patients who had major protocol violations (e.g., did not meet the study selection criteria). Usually the primary analysis should be an ITT analysis, but in some rare cases (e.g., noninferiority or equivalence trials), a modified ITT may be appropriate.

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

Measuring CNS exposure and target engagement

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

MAD studies

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

When conduct phase I in patients

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

Dose response

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

FDA guidance

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

Max tolerated dose

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

Per protocol analysis

A

Unlike ITT analysis, per protocol analysis only includes patients who successfully adhered to the protocol (i.e., no major protocol violations). In many cases, per protocol analysis may serve as the primary data analysis (for clinical trials, FDA approval may be necessary). The major strength of per protocol analysis is the weakness of ITT analysis: it removes significant noise from study, noise that may prevent you from truly determining an intervention’s effects. Conversely, we covered the major weaknesses of per protocol analysis when discussing the strengths of ITT analysis.

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

Aim of single ascending dose studies?

A

he key objectives of single ascending dose stud- ies are to deine safety, tolerability, pharmacokinet- ics and pharmacodynamics of a drug. he dose range deployed usually covers approximately two logs and is framed by a starting dose that is a fraction of the preclinical pharmacologic no-pharmacologic efect dose (NOPED) in the most appropriate or sensitive species and limited to a top dose that is guided by the preclinical exposure (drug concentration in plasma) at the no-adverse efect level (NOAEL)

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

NOAEL

A

no-observed adverse effect level

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

MAD studies

A

Multiple ascending dose studies extend observa- tions on human pharmacology to longer periods of dosing. Again, the key objectives are to provide data on safety, tolerability and pharmacokinetics with pro- longed dosing. In most studies, the duration of dosing ranges from 7 to 14 days with dosing frequency deter- mined by the pharmacokinetic parameters deined in single-dose studies. Typically, 4–5 dose levels are examined in the single ascending dose study, with the dose range covering a little over 1 log.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What kind of studies are first conducted in patients in early development?

A

Some initial studies in humans can only be con- ducted in patients. Medications with substantial poten- tial toxicity risks such as cytotoxic or genotoxic drugs cannot be administered to normal volunteers and for this reason, early studies are conducted in patients. he most common setting where this occurs is in oncology drug development where initial single- and multiple- dose studies are virtually always conducted in cancer patients. Examples from neurological therapeutics include the use of speciic B-cell depleting therapies for multiple sclerosis and immunotherapeutic vaccines for A l z h e i m e r ’s d i s e a s e [ 1 , 2 ] .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What is critical to characterize during middle stage development?

A

During middle stage development it is critical to begin to characterize the dose-response relationship for eicacy and safety endpoints in the selected popula- tion. Determination of the likely efective and safe dose range is a critical objective of middle stage develop- ment that afects not only the design of later stage trials but other aspects of non-clinical development as well. An important study from the FDA showed that a sub- stantial percentage of new drugs approved were rela- beled to correct dose ranges, and the majority of these changes were for safety reductions [10]. Of all thera- peutic areas examined, drugs for nervous system indi- cations had the highest percentage of dosing changes. Establishment of the optimal dose range requires that substantial attention be paid to selection of the appro- priate patient population, eicacy endpoints, and safety evaluations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

How are PROs evaluated by regulators?

A

Regulatory agencies review and evaluate the suitability of PRO assessments based upon several characteristics including the medical condition and population for intended use, concepts being measured, number of items, conceptual frame- work, data collection method, scale administration, response options, scoring and weighting of items or domains, and availability of translations or cultural adaptations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What is important regarding PRO development?

A

Because the properties of a measurement instru- ment like a PRO need to be well understood prior to collecting deinitive eicacy data in pivotal conirma- tory trials, this important groundwork must be initi- ated and is oten largely completed during middle stage development. he FDA has published a useful guidance document that is aimed at ensuring that the process for evaluating new instruments is adequately understood by clinical researchers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

NOAEL

A

The common definition of NOAEL, “the highest experimental point that is without adverse effect,”

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

Lowest Observed Adverse Effect Level (LOAEL)

A

Lowest Observed Adverse Effect Level (LOAEL) is the lowest exposure level at which there are biologically significant increases in frequency or severity of adverse effects between the exposed population and its appropriate control group.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

Problems with historical control

A

Historical controls: may have received difering concomitant therapies; may have been treated by diferent physicians, using diferent protocols to manage therapy; may not have met all inclusion criteria for current study; may have difering distributions of prognostic factors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

Common types of bias

A

Selection bias occurs when there are systematic differences between groups. For example, if groups are not comparable on key demographic factors, then between-group differences in treatment outcomes cannot necessarily be attributed solely to the study intervention. RCTs attempt to address selection bias by randomly assigning participants to groups – but it is still important to assess whether randomization was done well enough to eliminate the influence of confounding variables.

Performance bias refers to systematic differences between groups that occur during the study. For example, if participants know that they are in the active treatment rather than the control condition, this could create positive expectations that have an impact on treatment outcome beyond that of the intervention itself. Ideally, participants and investigators should remain unaware of which group participants are assigned to. Of note, this is more easily achieved in medication trials, where the medication and the placebo appear identical, than in psychotherapy trials.

Detection bias refers to systematic differences in the way outcomes are determined. For example, if providers in a psychotherapy trial are aware of the investigators’ hypotheses, this knowledge could unconsciously influence the way they rate participants’ progress. It is crucial that psychotherapy RCTs address this by utilizing independent outcome assessors who are blind to participants’ assigned treatment groups and investigators’ expectations.

Attrition bias occurs when there are systematic differences between groups in withdrawals from a study. It’s common for participants to drop out of a trial before or in the middle of treatment, and researchers who only include those who completed the protocol in their final analyses are not presenting the full picture. Analyses should include all participants who were randomized into the study (intention to treat analysis), and not only participants who completed some or all of the intervention.

Reporting bias refers to systematic differences between reported and unreported data. One example is publication bias, which occurs because studies with positive results are more likely to be published, and tend to be published more quickly, than studies with findings supporting the null hypothesis. At the investigator level, outcome reporting bias can also occur when researchers only write about study outcomes that were in line with their hypotheses. Efforts to address this include requirements that RCT protocols be published in journals or on trial registry websites, which allows for confirmation that all primary outcomes are reported in study publications.

Other bias is a catch-all category that includes specific situations not covered by the above domains. This includes bias that can occur when study interventions are not delivered with fidelity by therapists, or when there is “contamination” between experimental and control interventions within a study (for example, participants in different treatment conditions discussing the interventions they are receiving with each other).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

How to select FIH dose

A

The EMA and the FDA have published guidance documents for investigators to follow when determining an appropriate starting dose. The EMA guidance assists in the transition from preclinical to early clinical development and covers many of the risks inherent to FIH trials and discusses mitigation strategies to manage these risks.2 The FDA guidance aims at avoiding toxicity at initial doses by using the generally accepted benchmark for safety, the NOAEL obtained from the most sensitive toxicology test species as a starting point for determining a reasonably safe starting dose.1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

Method of selecting FIH dose

A

Methods for calculating the starting dose are either empirical or mechanistic (Table 2). As mentioned, the most widely used empirical method follows the FDA guidance1 for estimating a maximum safe starting dose by converting the NOAEL to a human equivalent dose with the use of allometric scaling. However, this method has its disadvantages, including the use of a somewhat arbitrary safety factor to ensure safety of the starting dose, the dose is based on minimal risk of toxicity rather than based on pharmacologic activity, and it does not address dose escalation or the maximum allowable dose. The EMA guidance2 outlines a more mechanistic approach based on state-of-the-art modeling incorporating all relevant preclinical pharmacology data including ex vivo and in vitro studies in human tissues. This guidance highlights selection of a minimal anticipated biological effect level, as this is becoming increasingly important as more targeted therapies are being developed that are very specific and potent with predicted pharmacologically active doses well below a dose that is thought to be safe based on the NOAEL.11

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

Biomarker in phase 1

A

The FIH study represents the first opportunity that a biological readout can be obtained in humans. The measurement of biomarkers in conjunction with a drug treatment could be important both during drug development and clinical implementation after marketing approval. Biomarker readouts can help guide drug development: from discovery through target engagement assessment, to diagnostic use, and on to the promise of precision medicine and companion diagnostics in everyday clinical practice.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

Validity

A

Validity is a more complicated topic than reliability. Validity refers to the degree of correspondence between a test score and the construct (e.g., “hopelessness”) or diagnosis (e.g., “panic disorder”) that it was designed to measure. The absence of true gold standards for measuring psychiatric condition makes assessing validity even more difficult in our field. It may be best to think of validity as indicating the accuracy of a score. Basic forms of validity include content validity, criterion validity, and construct validity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

Content Validity

A

Content validity refers to the extent to which a scale adequately covers the important features of a condition or construct. For example an anxiety scale that did not con- tain a “worry” item may have questionable content validity. Evaluations of content validity tend to be rational and not empirically based.

90
Q

Criterion Validity

A

Criterion validity is measured by a correlation coefficient (like a reliability coeffi- cient) showing the correspondence between the test score and known or accepted markers of the target condition (for a new depression scale this might include scores on an existing measure of depression or receiving an independent diagnosis of major depressive disorder).

91
Q

Construct Validity

A

Construct validity represents the most comprehensive and complex assessment of score validity. Construct validity is the accumulated weight of evidence indicating that a measurement tool (1) adequately covers the full range of the target construct, (2) does so across a variety of clinical situations (inpatient and outpatient settings), and (3) does so across a wide range of subjects (e.g., young adults as well as older adults). There is no single measure of construct validity.

92
Q

Reliability

A

Reliability statistics provide a means for quantifying this degree of error contained in our measurement and indicating the consistency and stability of the score. Reli- ability is a necessary, but not a sufficient, quality of useful rating scales. The more reliable an instrument is, the more consistent a patient’s score will be over time or across different raters (in the case of self-report scales and clinician-administered scales, respectively, as shown in Table 1.1). Important reliability concepts include internal consistency, test–retest reliability, and inter-rater agreement.

93
Q

Internal Consistency

A

Internal consistency reflects the degree to which items in a scale measure a single property or dimension. Typically, instruments that are used clinically should have internal consistency coefficients (often expressed as “Coefficient Alpha”) of 0.80 or higher (often closer to 0.90). Scale length affects measures of internal consis- tency, so that briefer scales (10 items or less) often have lower internal consistency coefficients simply due to having fewer items.

94
Q

Test–Retest Reliability

A

Test–retest reliability reflects the stability of scores over repeated testings (across time) and is often slightly lower than internal consistency (stability coefficients between 0.75 and 0.85 would be considered acceptable). Knowing the test–retest time period is important when assessing a stability coefficient. A time interval of 2–3 weeks may be adequate for a test of depression (a state condition), while an interval of a few months would be appropriate for a measure of narcissistic personality (a trait condition).

95
Q

Inter-rater Reliability

A

For clinician-rated scales it is important to know the extent to which different clin- icians agree when assigning ratings; this is referred to as inter-rater agreement. Inter-rater agreement is typically reported as either a Kappa coefficient (for out- comes with two or three categories) or an intra-class correlation coefficient (for continuous outcomes). Kappa coefficients are often weighted to correct for chance agreement (i.e., given as “weighted Kappa”). Although there are no firm rules for interpreting Kappa values, Kappa coefficients ≤ 0.40 are usually considered “poor,” values between 0.60 and 0.70 are considered “good,” and coefficients > 0.70 are considered “excellent.” Kappa values can be affected by (1) the number of choices raters are required to make (e.g., “present or absent” versus “mild, moderate, or severe”) or (2) the base rate (prevalence) of the target condition.

96
Q

Database extraction

A
97
Q

Database cleaning

A
98
Q

Data cleaning

A

In the course of collecting and entering data into the database, there will be errors. The process of correcting the errors is called data cleaning. Data cleaning consists of three steps. First, the data quality and management plan is developed. This plan specifies the algorithm for detecting errors, for correcting errors, and the final standard that the data has to meet before it is consider to be clean. The algorithms might specify the highest acceptable value for data, such as “age below 18 or over 100 should be flagged,” or “if all the respiratory rate values from a site are 20, then query.” Some of these are called edit checks – simple queries that are automatically generated as the data is entered, to reduce the rate of frank typographical errors.

99
Q

Edit checks

A

The algorithms might specify the highest acceptable value for data, such as “age below 18 or over 100 should be flagged,” or “if all the respiratory rate values from a site are 20, then query.” Some of these are called edit checks – simple queries that are automatically generated as the data is entered, to reduce the rate of frank typographical errors.

100
Q

Queries

A

The query process starts with a query form, which specifies the questioned data point, such as “please verify that the patient visited the emergency room three times in two days.” The query is sent to the site, and the site fills out the query form. In general, most sponsors tend to over-query. The percent of queries that results in a change to the database should be greater than 50%, as a general rule. Obviously, EDC allows easier data cleaning and faster, real-time data cleaning.

101
Q

Soft database lock

A

In some cases, there might be a “soft” database lock to enable an interim analysis. The data up to a certain date or up to a certain visit is cleaned to a standard that is high but not as high as the final analysis, and the data analysis is performed on this partially cleaned database.

102
Q

CRF design

A

A CRF is a critical piece of the study. It defines the type of data collected, amount of data collected, and therefore directly impacts the cost of the study and the difficulty of monitoring the study. A study with a 20 page CRF is much easier to run and monitor than one with a 2000 page CRF. The CRF should collect all the data that the protocol says will be collected. In most cases, it should collect little else. The key to CRF design is to be brutal about limiting the amount of data collected. Each additional page of CRF can add hundreds of thousands of dollars to millions of dollars of additional cost. If the data is not absolutely necessary, it should not be collected. To do otherwise is to waste time and resources of the patient, physician, CRC, CRA, data manager, and other personnel. In general, it is desirable to limit open-ended questions, where the CRC can enter text. Check-boxes and fill-in questions are preferable. For example, rather than asking, “Please describe any hospitalization,” you should ask, “Was the patient hospitalized since the last visit? Yes/No.”

103
Q

When should SAP be finalized?

A

The importance of finalizing the SAP prior to unblinding is that once the data has been unblinded, it is impossible not to introduce a bias in designing the analysis plan. Almost inevitably, the analysis will be biased to show a positive result. The reason for this is that there are many judgment calls that have to be made with regard to the population to be included in the final analysis, imputation of missing values, etc. that can have an impact on the final results. And of course, once the data has been unblinded, it is always possible to design an analysis that will make the results look positive. In effect, a post hoc SAP turns a prospective study into a retrospective study, and makes it impossible to use the study to ascribe causality.

104
Q

Subgroup analyses

A

Subgroups are then often presented. These can be demographic subgroups, such as by race or age, and they can be by sites/geography, or they may be by pre-/ post-amendment or by patients enrolled in the first half of the study vs. second half.

105
Q

TLGs

A

As the first step in analysis, all data should be presented in tables, listings, and graphs (TLGs). This includes data by each visit, data by each patient, aggregate data, etc. Typically, the tables would include both standard deviations and confidence intervals. Means and standard deviations, or in nonparametric cases, medians and quartiles, should be presented. The programming for the TLGs should be prepared and validated in advance. Validation typically requires two independent sets of programmers to program the TLGs and test/dummy data set to be run with the programs. The output should match each other, and should match the expected output. Listings are lines of data, sometimes aggregate data, sometimes individual data and they should include the following.

106
Q

Type A FDA meeting

A

Type A meeting is for issues that are critical to resolve in order to proceed on a stalled development program. FDA should grant a meeting within 30 days of the request.

107
Q

Type B FDA meeting

A

Type B meeting includes certain milestone meetings such as pre-IND, end of Phase II, and pre-BLA/NDA meetings. FDA should grant a meeting within 60 calendar days. Typically, the agency will grant one Type B meeting per landmark.

108
Q

Type C FDA meeting

A

Type C meetings are all other meetings.

109
Q

Pre-IND Meeting

A

In the Pre-IND meeting, the planned pre-clinical studies, in particular the toxicology studies are discussed. It is a good idea to have this meeting in advance of starting the studies, because you don’t want to find out at the end of a 28-day study that you need to do a 6-month study. The FDA will need to understand the Phase I study design to weigh in on the proposal. Some pre-IND meeting will also focus on or include discussion about the CMC issues. This is less common, despite the fact that the regulations seem to indicate pre-IND meetings are really for CMC than toxicology studies.

110
Q

End of Phase II Meeting (EOP2)

A

There is typically an end of Phase II meeting (sometimes called pre-Phase III meeting). This is a critical meeting, where the sponsor and the FDA comes to an agreement on the design of Phase III study or studies as well as other clinical and nonclinical work that must be done before a BLA or an NDA can be filed. For example, discussion regarding if special population studies must be done (elderly patients, immunocompetency study more CMC work, etc.) will be had. The briefing package for the EOP2 meeting should include: • proposed indication and in some cases, claims; • Phase III protocol; • summary of pre-clinical and clinical data to date; • data analysis proposal. For a special protocol assessment, additional information that might be requested includes: • draft CRF; • draft IDMC charter; • draft SAP.

111
Q

Pre-BLA/NDA Meeting

A

After a successful Phase III study or studies, you should hold a pre-BLA or pre-NDA meeting with the FDA. The meeting should agree on the format and content of the submission. You should come to an agreement on the content of the data set, types of data tables that will be generated, types of analysis, and so on. For example, the sponsor might propose to include certain safety data in the 120-day safety update, and might want to make sure that it wouldn’t qualify as a major amendment that would trigger the resetting of the review clock. It is also a forum for discussing the possibility of a priority review, although the agency will often not commit to it. The meeting package should include: • topline efficacy and safety data; • description of the data set to be submitted; • types of tables that will be submitted; • indication and claims being sought.

112
Q

IVRS

A

The IVRS system is a generic term that is now used to refer to the method of randomization. It can be paper based, telephone based, fax based, or web based. The most sophisticated systems can handle dynamic randomization, and can be linked to drug shipment so that drug can be shipped automatically to replenish drug supply that is being used by the site. The IVRS system must be thoroughly validated (tested). It is unfortunately not uncommon for the system to be buggy. After all, it is software, and often it is custom-built software. It is, however, mission critical software, that can scotch the entire study if improperly programmed or configured. The most common IVRS is voice based. Typically, the site will call a number, and will be walked through a set of prompts. The IVRS will ask for the caller’s identification number, passcode, site identification number, confirmation of each inclusion and exclusion criteria (such as, “Is the patient over 18 years of age? Press 1 for yes, 2 for no.”), questions on the stratification criteria (such as “press 1 if the patient has anterior MI, 2 if the patient has posterior MI”), and so on. At the end, the patient will be assigned a patient number and a drug kit number.

113
Q

ICH Definitions An adverse event (AE) or adverse experience (AE)

A

An adverse event (AE) or adverse experience (AE) is “any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment.” In other words, an AE is anything bad that happens to the patient.

114
Q

ADR

A

An ADR has slightly different definition depending on whether it occurs in a pre-approval or post-approval setting. In the former, an ADR is “all noxious and unintended responses to a medicinal product related to any dose.” In the latter, an ADR is “a response to a drug which is noxious and unintended and which occurs at doses normally used in man for prophylaxis, diagnosis, or therapy of disease or for modification of physiological function.”

115
Q

An SAE is defined by ICH as

A

Any untoward medical occurrence that at any dose that meets any of the following criteria: • results in death; • is life-threatening (Note: The term “life-threatening” in the definition of “serious” refers to an event in which the patient was at risk of death at the time of the event; it does not refer to an event which hypothetically might have caused death if it were more severe.); • requires inpatient hospitalization or prolongation of existing hospitalization; • results in persistent or significant disability/incapacity; or • is a congenital anomaly/birth defect. Medical and scientific judgement should be exercised in deciding whether expedited reporting is appropriate in other situations, such as important medical events that may not be immediately life-threatening or result in death or hospitalisation but may jeopardise the patient or may require intervention to prevent one of the other outcomes listed in the definition above. These should also usually be considered serious. Examples of such events are intensive treatment in an emergency room or at home for allergic bronchospasm; blood dyscrasias or convulsions that do not result in hospitalisation; or development of drug dependency or drug abuse.

116
Q

A serious AE is different from a severe AE. ICH distinguishes between the two as follows.

A

To ensure no confusion or misunderstanding of the difference between the terms “serious” and “severe,” which are not synonymous, the following note of clarification is provided: The term “severe” is often used to describe the intensity (severity) of a specific event (as in mild, moderate, or severe myocardial infarction); the event itself, however, may be of relatively minor medical significance (such as severe headache). This is not the same as “serious,” which is based on patient/event outcome or action criteria usually associated with events that pose a threat to a patient’s life or functioning. Seriousness (not severity) serves as a guide for defining regulatory reporting obligations.

117
Q

Reporting SAEs

A

When there are SAEs, the knowledge of that should be disseminated as quickly as possible. That knowledge may have an important impact on the safety of the patients in the study. Therefore, there are some very strict guidelines on how and how rapidly such events need to be reported. All serious, unexpected, ADRs need to be reported in an expedited fashion. You do not have to report expected AEs. If it is listed in the IB, it is considered to be expected. There is some debate over whether an addendum to the IB can be used to make an unexpected AE into an AE so that the events need not be reported with each incident. An AE that is considered to be unrelated to the drug need not be reported. Of course, it is a judgment call whether the AE is related to the drug. There will usually be an assessment by the treating physician and another by the MM. In general, if either considers the AE to be related to the drug, or possibly related to the drug, then it should be reported. There are many different ways of describing the degree of likelihood: not related, possibly related, probably related, plausibly related, suspected to be related, definitely related, and so on. In general, only the ones that are definitely unrelated should be excluded from the reporting, although the regulations seem to allow a bit more latitude. In a safety reporting, it is always prudent to err on the side of caution.

118
Q

Minimum Criteria for Reporting

A

When does the clock start running for the reporting requirements? It starts running when the following minimum criteria are met: • An identifiable patient. • A suspect medicinal product. • An identifiable reporting source.

An event or outcome that can be identified as serious and unexpected, and for which, in clinical investigation cases, there is a reasonable suspected causal relationship. The clock starts running if any employee of the sponsor becomes aware of the event – even if the employee is not involved with the study or is not in the clinical group, the clock starts running. This would, for example, include a salesperson hearing about an event on a sales call or an administrative assistant hearing about an event over the radio. The CRO and the CRAs employed by them are considered to be agents of the sponsor and the clock starts running when they are aware of the event.

119
Q

How to Report SAE

A

An SAE should be reported as soon as possible by the site to the sponsor. During the training of the investigators, this should be made absolutely clear to them. Because of the time lag traditionally associated with paper-based CRFs most sponsors have a separate, faster, process for SAE reports. A site will typically fax a special SAE form to the sponsor, but may in some cases use an EDC system. Once the sponsor is notified, there should be a pre-defined process for processing the adverse events. The event should be logged in, and the data entered into a database. The SAE reports are typically submitted in a MedWatch format. There should be a clearly identified person who is responsible for each step of the processing and for each of the cases. There should be quality checks built into the system. There is usually a person assigned to query the site or to ask the CRA to query the site for additional information as needed. There should be follow-up with the site until the event has resolved. The MedWatch form is preferred by the FDA for reporting of unexpected SAEs. The CIOMS-I form is also widely used. The initial report may not contain all pertinent information. The form should be submitted with what data is available. Follow-up information should be actively sought and submitted as it becomes available. The report must include an assessment of the importance and implication of the findings, including relevant previous experience with the same or similar medicinal products. Key data elements for inclusion in expedited reports of serious ADRs (from ICH)

120
Q

Imputation

A

Imputation is finding a value to replace a missing or incorrect value.

121
Q

Chief problem with crossover design

A

Crossover designs have a number of problems that can invalidate their results. The chief difficulty concerns carryover, that is, the residual influence of treatments in subsequent treatment periods. In an additive model, the effect of unequal carryover will be to bias direct treatment comparisons. In the 2x2 design, the carryover effect cannot be statistically distinguished from the interaction between treatment and period and the test for either of these effects lacks power because the corresponding contrast is between subject. This problem is less acute in higher order designs, but cannot be entirely dismissed.

122
Q

additional problems that need careful attention in crossover trials

A

There are additional problems that need careful attention in crossover trials. The most notable of these are the complications of analysis and interpretation arising from the loss of subjects. Also, the potential for carryover leads to difficulties in assigning adverse events that occur in later treatment periods to the appropriate treatment. These and other issues are described in ICH E4. The crossover design should generally be restricted to situations where losses of subjects from the trial are expected to be small.

123
Q

Important disease considerations when using crossover design

A

The disease under study should be chronic and stable. The relevant effects of the medication should develop fully within the treatment period. The washout periods should be sufficiently long for complete reversibility of drug effect.

124
Q

A common generally satisfactory use of crossover design

A

A common, and generally satisfactory, use of the 2x2 crossover design is to demonstrate the bioequivalence of two formulations of the same medication. In this particular application in healthy volunteers, carryover effects on the relevant pharmacokinetic variable are most unlikely to occur if the wash-out time between the two periods is sufficiently long. However, it is still important to check this assumption during analysis on the basis of the data obtained, for example, by demonstrating that no drug is detectable at the start of each period.

125
Q

factorial design

A

In a factorial design, two or more treatments are evaluated simultaneously through the use of varying combinations of the treatments. The simplest example is the 2x2 factorial design in which subjects are randomly allocated to one of the four possible combinations of two treatments, A and B. These are: A alone; B alone; both A and B; neither A nor B. In many cases, this design is used for the specific purpose of examining the interaction of A and B. The statistical test of interaction may lack power to detect an interaction if the sample size was calculated based on the test for main effects. This consideration is important when this design is used for examining the joint effects of A and B, in particular, if the treatments are likely to be used together.

126
Q

Parallel Group Design

A

The most common clinical trial design for confirmatory trials is the parallel group design in which subjects are randomized to one of two or more arms, each arm being allocated a different treatment. These treatments will include the investigational product at one or more doses, and one or more control treatments, such as placebo and/or an active comparator. The assumptions underlying this design are less complex than for most other designs. However, as with other designs, there may be additional features of the trial that complicate the analysis and interpretation (e.g., covariates, repeated measurements over time, interactions between design factors, protocol violations, dropouts (see Glossary), and withdrawals).

127
Q

Internal validity

A

Internal validity is defined as the extent to which the observed results represent the truth in the population we are studying and, thus, are not due to methodological errors.

128
Q

External validity

A

External validity refers to the extent to which the results of a study are generalizable to patients in our daily practice, especially for the population that the sample is thought to represent.

129
Q

A non-inferiority trial is dependent on what?

A

NI study is dependent on knowing something that is not measured in the study, namely, that the active control had its expected effect in the NI study. When this occurs, the trial is said to have assay sensitivity, defined as the ability to have shown a difference from placebo of a specified size. A “successful” NI trial, one that shows what appears to be an acceptably small difference between treatments, may or may not have had assay sensitivity and therefore may or may not support a conclusion that the test drug was effective. Thus, if the active control had no effect at all in the NI trial (i.e., did not have any of its expected effect), then even ruling out a very small difference between control and test drug is meaningless and provides no evidence that the test drug is effective. (See Section III.D. for further discussion on assay sensitivity.) In the absence of a placebo arm, knowing whether the trial had assay sensitivity relies heavily on external (not within-study) information, giving NI studies some of the characteristics of a historically controlled trial.

130
Q

Objective of EOP2A

A

The main objectives of an EOP2A meeting are to help select the dosing regimens for the next phase (typically phases 2 and 3) of drug development and to design informative dose-response trials that will inform later phase clinical trials by best incorporating prior quantitative knowledge.

131
Q

Phase 2a and 2b

A

Phase II studies are sometimes divided into Phase IIa and Phase IIb. Phase 2a is specifically designed to assess dosing requirements (how much drug should be given), whereas Phase IIb is specifically designed to study efficacy (how well the drug works at the prescribed dose(s)).

132
Q

Typical primary endpoint for phase 2a

A

The primary endpoint of a Phase 2A trial is an efficacy endpoint often measured by a continuous biomarker.

133
Q

How does phase 2b differ from phase 2a study?

A

A typical Phase 2B trial is larger than a Phase 2A trial. It enrolls a few hundred patients, investigates several doses, and the treatment time is commonly longer than in a Phase 2A trial. Adverse events monitored in a Phase 2 trial comprise a secondary endpoint.

134
Q

minimum effective dose

A

The minimum effective dose (MED) is the smallest dose with a discernible useful effect (ICH E4 Guideline, 1994) [1]. The MED can be defined as the dose where the mean efficacy outcome is equal to a certain target, with the placebo used as a reference. Mean efficacy is usually assumed to be non-decreasing with dose. Both efficacy and safety endpoints are often taken into consideration when selecting a Phase 3 dose, as increasing the dose can result in both higher efficacy and increased adverse event rates.

135
Q

CTR

A

European Union (EU) pharmaceutical legislation known as the Clinical Trials Regulation entered into application on 31 January 2022. It aims to ensure the EU offers an attractive and favourable environment for carrying out clinical research on a large scale, with high standards of public transparency and safety for clinical trial participants.

On 31 January 2022, the Regulation repealed the Clinical Trials Directive (EC) No. 2001/20/EC and national implementing legislation in the EU Member States, which regulated clinical trials in the EU until the Regulation’s entry into application.

A transition period applies to clinical trial submission under the Regulation.

Consult the Regulation:

136
Q

Exit interview

A

Exit interviews conducted with patients who have completed (or terminated early) clinical trials have yielded valuable insights into patients’ experiences with a disease and with an investigational treatment.2,3,5 ,6 Such interviews are one way to highlight the meaningful changes patients experience during clinical trials, thus contextualizing and enriching clinical trial results. Further, interviews yield unique data to supplement clinical trial findings (e.g., regarding safety concerns or the benefits of treatment)

137
Q

kit numbers

A

When a patient enrolls into the trial, the investigator calls the IxRS and the IxRS randomizes the patient to a treatment arm, and tells the investigator the patient number and the kit IDs of the kits to dose the patient. Since the investigator and patient do not know the contents of the kits, they are blinded to the patient’s treatment arm. On return visits the investigator calls the IxRS with the patient number and the IxRS again tells the investigator the kit IDs of the kits to dose the patient. In order for this process to work the IxRS must track the kit inventory at each site. When the inventory is low at the site, the IxRS has the depot resupply the site. Additional descriptions of the IxRS processes are described in [1], [2].

The first batch of drug kits must be made before the trial begins. One of the first steps in the drug kit manufacturing process is to create the kit list. The kit list is used by manufacturing to label the kits, by supply chain to track the kits, and then by the IxRS to assign kits to patients. The seemingly mundane task of making a kit list, i.e., randomly assigning kit IDs to kit types, can have significant implications for blinding, trial integrity, and operational efficiency.

138
Q

Another name for CRA

A

Site monitor

139
Q

CAPA

A

Corrective action and preventative action

140
Q

priority review

A
141
Q

Aim of phase I studies

A

Dosing investigation comes in several phases, each fraught with different challenges. Phase I studies define acute toxicities, aim to generate enough data about the drug, and escalate the dose carefully to the maximally tolerated dose. When the maximally tolerated dose cannot be reached, the highest dose should be comfortably above the anticipated target dose. Phase I studies include single ascending dose (SAD) and multiple ascending dose (MAD) studies, which we discuss later in this chapter.

Chin, Richard; Lee, Bruce Y. Principles and Practice of Clinical Trial Medicine . Elsevier Science. Kindle Edition.

142
Q

Aim of phase 2

A

Phase II studies further investigate the dose to establish biological activity and the dose most likely to be useful. Typically you select 2–4 doses from the tested and tolerated doses in the SAD and MAD to be used for Phase II. Phase II studies aim to establish the dose(s) for the Phase III study and often perform testing on special subpopulations, such as renal failure patients.

Chin, Richard; Lee, Bruce Y. Principles and Practice of Clinical Trial Medicine . Elsevier Science. Kindle Edition.

143
Q

How to do you convert NOEAL to human equivalent dose (HED)

A

humans. The common method of converting the animal NOAELs to HEDS is normalizing the NOAEL to body surface area. Normalizing the NOAEL to body surface area means converting the NOAEL, which is in dose units per body weight (e.g., mg/kg), to dose units per body surface area (e.g., mg/m2). This method assumes that a given dose per body surface area has similar effects across different animal species.

Chin, Richard; Lee, Bruce Y. Principles and Practice of Clinical Trial Medicine . Elsevier Science. Kindle Edition.

144
Q
A
145
Q

Definition of therapeutic index

A
146
Q

Recommendations regarding pediatric Gaucher development

A

Enrolling pediatric patients who are
157 as homogenous as possible will increase the probability of detecting a treatment effect.
158 The need for a study to be conducted in a homogeneous population should not delay the
159 timely access to a drug product for age subgroups that are inherently harder to study
160 because of intrinsic heterogeneity.

147
Q

SmPC

A

Summary of product characteristics

148
Q

Allogenic transplant

A

Allotransplant is the transplantation of cells, tissues, or organs to a recipient from a genetically non-identical donor of the same species. The transplant is called an allograft, allogeneic transplant, or homograft. Most human tissue and organ transplants are allografts.

149
Q

CFR

A

Code of federal regulations

150
Q

GCP

A

GCP in the context of clinical trials stands for “Good Clinical Practice.” It is an international ethical and scientific quality standard for designing, conducting, recording, and reporting trials that involve the participation of human subjects. GCP ensures that the rights, safety, and well-being of trial subjects are protected and that the data generated from clinical trials are reliable and credible.

Compliance with GCP is essential for the acceptance of clinical trial data by regulatory authorities. Various regulatory agencies, including the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH), provide guidelines on Good Clinical Practice. Adhering to GCP helps maintain the integrity of clinical trials and fosters confidence in the results obtained during the drug development process.

151
Q

DOR

A

Delegation of responsibilities

152
Q

CAPA

A

Corrective and preventative actions

153
Q

CTIS

A

Clinical Trials Information System is a new platform for monitoring and assessing the process of clinical trials throughout the EU. CTIS contains a centralized EU portal and database for clinical trials. In addition to the European Databank on Medical Devices (Eudamed) and SPOR, CTIS is one of the large steps toward digitalization and harmonization for EMA and European Commission. CTIS will be available in January 2022, over 7 years after the functional specifications of the system were endorsed by EMA Management Board and almost 4 years later than originally scheduled.

154
Q

The Clinical Trials Regulation

A

The Clinical Trials Regulation (Regulation (EU) No 536/2014) will become effective on 31 January 2022. The regulation will change how clinical trials are conducted in the EU [i]. The regulation aims to harmonize the assessment and supervision of processes for clinical trials across the EEA. And this should happen thanks to the new platform CTIS.

155
Q

basic ideas behind CTIS

A

The basic ideas behind CTIS are familiar to anyone who has worked with any platform. Currently, sponsors must submit clinical trial applications (CTA) separately to national competent authorities (NCA) and ethics committees in each country to get regulatory approval to run a clinical trial (CT) [iii]. With CTIS, sponsors can apply for CTA (e-dossier) in all EEA countries with a single application. Thanks to CTIS, the sponsor and the authorities can collaborate on the maintenance of the application and trial information in one place.

The platform should also help with the different timelines applicable to different authorities. There is no need for a separate registration and submission of information to a public register of clinical trials (more on EudraCT below.)

156
Q

CTA

A

A Clinical Trial Application (CTA) is a Regulatory dossier that is submitted to the Health Authority (HA) of the country in which a sponsor would like to conduct clinical trials with Investigational Medicinal Products (IMPs) or with approved drugs to explore new indications. To obtain clinical trial authorization, a CTA application must be submitted with all the required documentation per the regulations of the competent HA. The regulations and safety reporting requirements for clinical trials vary from one country to another.

In the case of the US FDA, a Clinical Trial Application (CTA) is submitted in the form of an Investigational New Drug (IND) application, whereas for MHRA (UK) and the EU Member States, such requirements include the submission of the Investigational Medicinal Product Dossier (IMPD) along with the Clinical Trial Authorization application. In Canada and most of the world countries, a clinical trial authorization application is commonly referred to as a Clinical Trial Application (CTA).

157
Q

Clinical Trial Application (CTA) contains information on?

A

In general, a Clinical Trial Application (CTA) contains information on the quality, safety, and efficacy (or proposed therapeutic uses) of the investigational drug. The Chemistry, Manufacturing, and Controls (CMC) and safety/efficacy data requirements will vary based on the phase of the clinical trial conducted (i.e., Phase I, Phase II, and Phase III). Once submitted, the CTA shall be reviewed by the respective HA. Upon satisfactory review and evaluation of the information submitted in a CTA, the sponsor will receive formal authorization from HAs to conduct the proposed clinical trials. The Regulatory requirements for different phases of clinical trials and the type of study population are divergent in different countries of the world.

158
Q

Expected suspected adverse reactions

A

Restricted to suspected SAR‘s that were period previously observed with the IMP in more than one participant, and for which the sponsor has concluded that there is reasonable evidence of calls of relationship and develop the IMP at least once.

SAR that occurred only once maybe included in the section if there is a strong plausibility have a possible relationship with the IMP and a robust justification is provided

159
Q

Patient profiles

A

ll, most or just a selected portion of the data. It may be expressed in narrative English, collated or just a simple data
dump. A patient profile can be the key to identifying why a subject took a concomitant medication, the effects of an
unrelated surgical intervention or the sequence of adverse events which culminated in a serious adverse event. Or it
can simplify the task of verifying all of the dosing records for an individual subject. These different usages require a
certain amount of customized programming.
The first type of patient profile is the data dump. With this type of profile, it is important to see each and every field
that is collected on the Case Report Form (CRF). Once the data is clean and data standards have been applied, the
best method for medical review, discovery of subject abnormalities or preparation for the Clinical Study Report (CSR),
the narrative type of profile would best.

For a physician overseeing the clinical trial, the customization rises to another level. The starting point is that we
attempt to provide the equivalent of a briefing that they would receive from another physician as if the physician was
going to assume the responsibility for the care of that individual. With this in mind, three sections are prepared: an
English language introductory section, a brief baseline section and a schedule of events. Each is programmed and
generated differently.

160
Q
A
161
Q

CTMS

A

Clinical Trial Management System (CTMS)

162
Q

CMTL

A

Centralized monitoring tracking log

163
Q

AEDR

A

Adverse event dedicated review

164
Q

Dedicated AE review

A

Is specified in the central monitoring plan. It performed to comply with regulatory reporting of serious adverse events. Objective is to identify those events that should be categorized as SAEs or adverse events of special interest.

165
Q

Adverse Event (or Adverse Experience)

A

Any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment.

An adverse event (AE) can therefore be any unfavourable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product.

166
Q

Adverse Drug Reaction (ADR)

A

In the pre-approval clinical experience with a new medicinal product or its new usages, particularly as the therapeutic dose(s) may not be established: all noxious and unintended responses to a medicinal product related to any dose should be considered adverse drug reactions. The phrase “responses to a medicinal products” means that a causal relationship between a medicinal product and an adverse event is at least a reasonable possibility, i.e., the relationship cannot be ruled out.

Regarding marketed medicinal products, a well-accepted definition of an adverse drug reaction in the post-marketing setting is found in WHO Technical Report 498 [1972] and reads as follows: A response to a drug which is noxious and unintended and which occurs at doses normally used in man for prophylaxis, diagnosis, or therapy of disease or for modification of physiological function. The old term “side effect” has been used in various ways in the past, usually to describe negative (unfavourable) effects, but also positive (favourable) effects. It is recommended that this term no longer be used and particularly should not be regarded as synonymous with adverse event or adverse reaction.

167
Q

Unexpected Adverse Drug Reaction

A

An adverse reaction, the nature or severity of which is not consistent with the applicable product information (e.g., Investigator’s Brochure for an unapproved investigational medicinal product).

168
Q

serious adverse event (experience) or reaction

A

A serious adverse event (experience) or reaction is any untoward medical occurrence that at any dose:
* results in death,
* is life-threatening, NOTE: The term “life-threatening” in the definition of “serious” refers to an event in which the patient was at risk of death at the time of the event; it does not refer to an event which hypothetically might have caused death if it were more severe.
* requires inpatient hospitalisation or prolongation of existing hospitalisation,
* results in persistent or significant disability/incapacity, or
* is a congenital anomaly/birth defect. Medical and scientific judgement should be exercised in deciding whether expedited reporting is appropriate in other situations, such as important medical events that may not be immediately life-threatening or result in death or hospitalisation but may jeopardise the patient or may require intervention to prevent one of the other outcomes listed in the definition above. These should also usually be considered serious.

169
Q

STANDARDS FOR EXPEDITED REPORTING

A

All adverse drug reactions (ADRs) that are both serious and unexpected are subject to expedited reporting.

170
Q

Managing blind in the occurrence of a serious event

A

When the sponsor and investigator are blinded to individual patient treatment (as in a double-blind study), the occurrence of a serious event requires a decision on whether to open (break) the code for the specific patient. If the investigator breaks the blind, then it is assumed the sponsor will also know the assigned treatment for that patient. Although it is advantageous to retain the blind for all patients prior to final study analysis, when a serious adverse reaction is judged reportable on an expedited basis, it is recommended that the blind be broken only for that specific patient by the sponsor even if the investigator has not broken the blind. It is also recommended that, when possible and appropriate, the blind be maintained for those persons, such as biometrics personnel, responsible for analysis and interpretation of results at the study’s conclusion.

171
Q

Risk Reporting

A

The sponsor should describe the quality management approach implemented in the trial and summarize important deviations from the predefined quality tolerance limits and remedial actions taken in the clinical study report (ICH E3, Section 9.6 Data Quality Assurance).

172
Q

Hazard Ratio

A

The hazard ratio is the ratio of (chance of an event occurring in the treatment arm)/(chance of an event occurring in the control arm) (20). The HR has also been defined as, the ratio of (risk of outcome in one group)/(risk of outcome in another group), occurring at a given interval of time (21). In the situation where the hazard for an outcome is exactly twice in Group A than in Group B, the value of the hazard ratio can be either 2.0 or 0.5. The result of the calculation (whether HR=2.0 or 0.5) depends on whether the investigator chooses to calculate the ratio of hazards for (Group A)/(Group B) or, alternatively, to calculate the ratio of hazards for (Group B)/(Group A)

173
Q

Adverse Drug Reaction Reporting

A

The sponsor should expedite the reporting to all concerned investigator(s)/institutions(s), to the IRB(s)/IEC(s), where required, and to the regulatory authority(ies) of all adverse drug reactions (ADRs) that are both serious and unexpected.

174
Q

centralized monitoring can be used to

A

(a) identify missing data, inconsistent data, data outliers, unexpected lack of variability and protocol deviations. (b) examine data trends such as the range, consistency, and variability of data within and across sites. (c) evaluate for systematic or significant errors in data collection and reporting at a site or across sites; or potential data manipulation or data integrity problems. (d) analyze site characteristics and performance metrics. (e) select sites and/or processes for targeted on-site monitoring.

174
Q
A
174
Q
A
175
Q
A
176
Q

Statistical bias

A

Statistical bias, in the mathematical field of statistics, is a systematic tendency in which the methods used to gather data and generate statistics present an inaccurate, skewed or biased depiction of reality.

177
Q

Selection bias

A

Selection bias is the bias introduced by the selection of individuals, groups, or data for analysis in such a way that proper randomization is not achieved, thereby failing to ensure that the sample obtained is representative of the population intended to be analyzed.

178
Q

DRSR

A

Data review and surveillance report

179
Q

SAGA E2B

A

E2B is the electronic transmission of safety case using International Conference on Harmonization E2B standard. Through ARMADA, SAGA E2B notification is automatically forwarded to PV system to allow direct data import in PV database

180
Q

RDPM

A

Research & Development Portfolio Management

181
Q
A
182
Q

SmPC

A

Summary of product characteristics

183
Q

Database lock

A

Once the database is complete and clean the database is locked to prevent further changes to the data

Once a data cutoff is decided upon there may be a period where data needs to be cleaned

184
Q

DCR

A

Data change request

185
Q

E2B report

A

an electronic file transmitted to a health authority or company or elsewhere with all the elements of the ICSR. Sometimes cases are published as short reports in medical journals, some of which have been previously reported to health authorities and some not. These cases are picked up in the periodic review of the medical literature done by companies.

186
Q

Aggregate reports

A

There are multiple standard formats, of which the Periodic Safety Update Reports (PSUR) is the main one. The US aggregate reports are called, sometimes confusingly, NDA Periodic Reports, Periodic Reports, and PADERs (Periodic Adverse Drug Experience Reports). To worsen the situation, PADERs is also occasionally used to refer to PSURs. And the FDA also accepts PSURs.

187
Q

Causality

A

The FDA does not recommend any specific categorization of causality, but the categories probable, possible, or unlikely have been used. The WHO uses the following categories: certain, probably/likely, possible, unlikely, conditional/unclassified, and unassessable/unclassifiable.

A decision on causality for regulatory reporting is binary: yes or no, a causal relationship is possible or not. For other reasons, however, as explained below, various categories (usually three to six) are included in clinical trial protocols and case reports, as follows: ​Related ​Probably related ​Possibly related ​Weakly related ​Unrelated ​Unassessable This methodology is useful in later analyzing signals and in creating tables for investigator brochures, product labeling, and monographs to give a feel for the certainty or lack thereof about the causality of AEs by the drug in question.

188
Q

TPP

A

A TPP is the core document that outlines the anticipated profile of the drug once it is made available to patients. In essence, it is a condensed package insert that is anticipated. There are many different ways of writing a TPP. The specific format is less important than making sure that it clearly defines what the ultimate deliverable for the program is. Examples are given on the next page. The TPP is critical because the clinical development plan and the regulatory plan are built around achieving the TPP.

189
Q

Categorical measurements

A

Categorical measurements place observations into classes or groups. Examples of categorical variables are SEX (male or female), BLOOD_TYPE (A, B, AB, or O), and DISEASE_STATUS (case or noncase). Categorical measurements may occur naturally (e.g., diseased/not diseased) or can be created by grouping quantitative measurements into classes (e.g., classifying blood pressure as normotensive or hypertensive). Categorical variables are also called nominal variables (nominal means “named”), attribute variables, and qualitative variables.

190
Q

Ordinal measurements

A

Ordinal measurements assign observations into categories that can be put into rank order. An example of an ordinal variable is STAGE_OF_CANCER classified as stage I, stage II, or stage III. Another example is OPINION ranked on a 5-point scale (e.g., 5 = “strongly agree,” 4 = “agree,” and so on).

191
Q

Sensitivity analysis

A

Sensitivity analysis is the study of how the uncertainty in the output of a mathematical model or system (numerical or otherwise) can be divided and allocated to different sources of uncertainty in its inputs. A related practice is uncertainty analysis, which has a greater focus on uncertainty quantification and propagation of uncertainty; ideally, uncertainty and sensitivity analysis should be run in tandem.

The process of recalculating outcomes under alternative assumptions to determine the impact of a variable under sensitivity analysis can be useful for a range of purposes

192
Q

Smq

A

Standardized MedDRA Queries (SMQs) are groupings of terms from one or more MedDRA SOCs that relate to a defined medical condition or area of interest. They help in retrieving cases. This, of course, implies that they were coded correctly on data entry/input of the cases. The included terms may relate to signs, symptoms, diagnoses, syndromes, physical findings, and laboratory and other physiologic test data. Examples of SMQs that were developed and tested by CIOMS include cardiac arrhythmias, cardiac failure, cardiomyopathy, hepatic disorders, hostility/aggression, hyperglycemia/new onset diabetes mellitus, malignancies, and over 100 more. See the current list at the MedDRA website.

193
Q

excipients

A

An excipient is a substance formulated alongside the active ingredient of a medication. Excipients serve various purposes, including long-term stabilization, bulking up solid formulations containing potent active ingredients in small amounts (often referred to as “bulking agents”, “fillers”, or “diluents”), or enhancing the therapeutic properties of the active ingredient in the final dosage form

194
Q

Coefficient of variation

A

defined as
Standard deviation/sample mean

The CV removes the units of measure and expresses the standard deviation in relation to the size of the mean. This allows for direct comparison of numerical results.

195
Q
A
196
Q

DALA

A

Drug Abuse Liability Assessment Report

197
Q

Middle stage development

A

During middle stage development it is critical to begin to characterize the dose-response relationship for ei cacy and safety endpoints in the selected population. Determination of the likely ef ective and safe dose range is a critical objective of middle stage development that af ects not only the design of later stage trials but other aspects of non-clinical development as well. An important study from the FDA showed that a substantial percentage of new drugs approved were relabeled to correct dose ranges, and the majority of these changes were for safety reductions [ 10 ]. Of all therapeutic areas examined, drugs for nervous system indications had the highest percentage of dosing changes. Establishment of the optimal dose range requires that substantial attention be paid to selection of the appropriate patient population, ei cacy endpoints, and safety evaluations.

198
Q

SDV

A

Source data verification

199
Q

clinical endpoint:

A

A characteristic or variable that rel ects how a patient feels, functions, or survives. In a clinical trial, changes in a clinical endpoint may rel ect the ef ect of a therapeutic intervention. For the purpose of understanding the usefulness of a drug in a clinical setting, clinical endpoints are the most credible measure that can be assessed in a clinical trial.

200
Q

Biological marker

A

A characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes,processes, or pharmacologic response to a therapeutic intervention.

201
Q

Surrogate endpoint

A

A biomarker that is intended to substitute for a clinical endpoint. A surrogate endpoint is expected to predict clinical benei t or harm or lack of benei t or harm based on epidemiologic therapeutic pathophysiologic or other scientii c evidence. Surrogate endpoints are a subset of biomarkers. h e term surrogate literally means ‘substitute of ’ therefore the NIH working group discourages the use of the term surrogate marker because it suggests the substitution is for a marker rather than for a clinical endpoint.

202
Q

Time period effects

A

Random time-period effects are unexplained increases or decreases in the observed value for all individuals measured at a particular time point in a longitudinal study. They can be caused by learning effects, changes in equipment, personnel and overall subject co-operation

203
Q

AEDR

A

AE Dedicated Review

204
Q

Problems with crossover designs

A

However, there are also limitations in the crossover design. First, the conditions of the subjects must be stable throughout the study. In other words, a case is inappropriate for a study if the disease status changes over time, such as an acute cure, or when symptoms disappear or are cured by treatment in the first period. Second, a washout period may be necessary until the effect of the first treatment subsides. Therefore, if a treatment drug has an extended half-life, it may be difficult to conduct a study with a crossover design. Third, there is a burden that all treatments are carried out on one subject, which often causes ethical problems. Fourth, the processing of dropped or missing data is more problematic than in a parallel design, and the statistical analysis is complex

205
Q

Period effect

A

The period effect implies that the effect of the same treatment received at two different periods is different for each period and corresponds to Pj in Equation (1). Since the first and second treatments are inevitably separated in time, an effect may appear depending on the measurement period, not the treatment. Therefore, when comparing the value obtained by subtracting the first period from the second period of the AB sequence with the value obtained by subtracting the value of the first period from the second period of the BA sequence5), there should be no difference if there is no period effect.

206
Q

Carryover effect

A

The carryover effect, which corresponds to Cj-1,k in Equation (1), refers to the effect of the previous treatment or the change caused by the first treatment continues until the next period and alters the effect of the next treatment. Rather than determining that there is no carryover effect by statistical testing, it is better to select a crossover design when the possibility of a carryover effect is medically unlikely or when the effect can be eliminated through a washout period. Analyzing for the period-by-treatment interaction is used to determine whether the two treatment effects are different in the two periods, and it is difficult to distinguish the carryover effect from the period-by-treatment interaction; therefore, the carryover effect and the period-by-treatment interaction are often treated as identical. However, depending on which parameters are included in the crossover design model, the carryover effect may be embedded in parameters other than the period-by-treatment interaction6). In other words, it is difficult to analyze the carryover effect in the simplified 2 × 2 crossover design [5]; therefore, it is important in the study planning stage to design such that the carryover effect does not occur. For example, there is a method to set a sufficient washout period until the treatment effect or change disappears. In the case of drug studies, a washout period is sometimes set at 3–4 times or more of the blood plasma elimination half-life.

207
Q

Sequence effect

A

The fact that subjects are allocated to a particular sequence may affect the results. That is, when comparing the means of the dependent variables in the AB and BA sequences, there should be no difference if there is no sequence effect. This allows the assumption that there is no sequence effect by randomization in the AB/BA sequence. However, it should be noted that this assumption cannot be verified through statistical analysis

208
Q
A

Symptomatic, function, survival

209
Q

Bias

A
210
Q

Assay sensitivity

A
211
Q

Regression towards the mean

A
212
Q

Responsivity

A
213
Q

Internal validity

A
214
Q

Parametric vs nonparametric

A

So far in this book, we have dealt only with parametric techniques, which test hypotheses about population parameters, such as m and s2, and assume that the population being sampled has a particular distribution, such as a normal distribution. In this chapter, we consider nonparametric techniques, which test hypotheses about population distributions rather than about population parameters. Some nonparametric techniques are also distribution-free techniques, in that they do not require strict assumptions about the distribution of the population being s

215
Q
A
216
Q

O’Brien ordinary least square test

A
217
Q

O’Brien ordinary least square test

A
218
Q

Mixed model for repeated measures

A
219
Q

Anchor

A
220
Q

Mixed model for repeated measure

A