pathogenetic : it impairs the pathogenetic mechanism(s)
symptomatic : it doesn't remove cause(s), but just symptom(s) and/or
sign(s)
monotherapy : treatment of a condition by means of a single drug.
Net effect of a therapy =
placebo effect :
the sum total of all nonspecific effects, both good and adverse, of medical
treatment, primarily psychological and psychophysiological effects associated
with the physician-patient relationship and the patient's expectations
and apprehensions concerning the treatment. In 1978, scientists demonstrated
that an opioid blocker called naloxone could abolish the placebo effect
in a pain experimentref.
The simple interaction between patient and practitioner has measurable
effects in the brain : "open" injections of analgesics (patient-witnessed)
were significantly more effective and less variable than "hidden" injections
(patients were ignorant of injections). Furthermore, they showed that by
blocking the opioids, naloxone greatly reduces this open-injection placebo
effect, suggesting that the therapist-patient interaction activates the
endogenous opioid systemsref.
Placebo and opioid analgesia may have a common neural mechanism in activation
of the rostral anterior cingulate cortex (rAAC) for both treatments, though
analgesic provide more pain reliefref
:
Both severely depressed men given either placebo or fluoxetine exhibit
increased activity in the cortex and decreased activity in limbic regions,
but only patients given fluoxetine experienced changes in brain stem, striatum,
and hippocampusref.
The release of dopamine, may be triggered by the placebo effect in patients
with Parkinson diseaseref.
To uncover regions of the brain that show decreased activity during the
placebo effect, in one trial, researchers told subjects that they were
administering a powerful analgesic cream. In another, the subjects received
the same cream but were told it has no effect. When subjects were experiencing
the placebo effect, a subset of known pain-sensitive brain regions showed
a signal reduction of 20% to 25%ref.
In a subsequent study, patterns of neuronal firing, not visible via neuroimaging,
corresponding to the previous findings were observed performing single-neuron
recording via deep brain stimulation of subthalamic nuclei in patients
with Parkinson disease who had been administered a sham treatmentref.
The brain areas involved in anticipation of pain relief have significant
overlap with those involving empathyref.
Reductions in pain ratings when administered a placebo with expected analgesic
properties have been described and hypothesized to be mediated by the pain-suppressive
endogenous opioid system. Using molecular imaging techniques, the activity
of the endogenous opioid system on µ-opioid
receptors
was directly examined in humans in sustained pain with and without the
administration of a placebo. Significant placebo-induced activation of
µ-opioid receptor-mediated neurotransmission was observed in both
higher-order and sub-cortical brain regions, which included the pregenual
and subgenual rostral anterior cingulate, the dorsolateral prefrontal cortex,
the insular cortex, and the nucleus accumbens. Regional activations were
paralleled by lower ratings of pain intensity, reductions in its sensory
and affective qualities, and in the negative emotional state of the volunteers.
These data demonstrate that cognitive factors (e.g., expectation of pain
relief) are capable of modulating physical and emotional states through
the site-specific activation of µ-opioid receptor signaling in the
human brainref.
Placebo responses are often larger in studies of alternative therapiesref1,
ref2.
This may, in part, be due to elevated expectations of patients.
The nocebo effect is the phenomenon whereby a patient who believes
that a harmless substance cause harm actually experiences adverse effects
due to negative expectations or the psychological condition of the patient.
Web resources : The
science of the placebo: Toward an interdisciplinary research agenda
+ pharmacological effect
Drug development
empiric or focused screening => optimization via chemistry
preclinical testing according to good laboratory practice (GLP)
and standard operating procedure (SOP)
(1-5 years : average 2.6 years) :
studies in vitro to define binding properties
testing on model organisms with experimental diseases (rare for psychiatric
disorders, except anxiety)
short-term preclinical animal studies
acute (single dose) toxicity testing in several species using route of
administration for clinical use
subacute or subchronic toxicity studies ranging from 1 week to 3 months
mutagenicity/gene tox : short term tests (in vitro or in vivo)
which may predict carcinogenic potential
initiation of reproduction study (segment I)
long-term preclinical animal studies
chronic toxicity (6-12 months) for kidney, liver, nervous system and blood
reproductive/teratology studies are typically conducted into 3 phases or
segments:
segment I - fertility and general reproductive performance
study of fertility and general reproductive performance, e.g., mating behavior,
gonadal function, conception, and early gestation
required to initiate short term trials in females
species studied include male and female rats
segment II
measure the embryo toxic (embryo lethality) and/or teratogenic (external
visceral and skeletal defects) effects caused by administration of new
drug to pregnant females during organogenesis
2 species are studied, the rat and rabbit (mouse and primate are also studied
if indicated)
segment III - perinatal and postnatal study
study of perinatal and postnatal effects, e.g., on late stage fetal development,
labor, delivery, lactation, viability, and growth of the newborn
assessments include observations of labor; deliver; lactation; viability,
growth, development, and weaning of newborn
species studied include the rat (mouse or primate if indicated)
carcinogenicity
pre-IND meeting
investigational new drug
(IND) : the notification of data relating to a new drug, which must
be made to the FDA or local bioethic committee, which then review its safety
before it may be administered to man (30 days). Chemistry, manufacturing
and control (CMC) data are the chemical and pharmaceutical data which
are included in an IND or an NDA. The clinical protocol must include :
background and rationale
objectives
patient selection criteria
study design and treatment
expected toxicities and dose a
clinical and lab monitoring
criteria to evaluate endpoints
statistical considerations
references
informed consent
data form
investigators
clinical testing in human
beings (2-10 years : average 5.6 years)
Principles of drug testing prior to trials in humans :
exact composition of drug should be known; if not, method of preparation
acute toxicity studies in animals of different species
chronic toxicity experiments at varying doses in different species for
cumulative effects
careful and frequent observations of animals, to develop a composite picture
of clinical effects
careful pathological examinations of tissues with appropriate stains
effects of drugs on excretory or detoxifying organs, expecially kidney
and liver
rate of absorption and elimination, path and manner of excretion, concentration
in blood and tissues at varying times
possible influence of other drugs and foodstuffs
careful examination for any idiosyncrasies or untoward reactions
the voluntary consent of the human subject is absolutely essential
the experiment should be such a sto yield fruitful results for the good
of society, unprocurable by other methods or means of study, and not random
and unnecessary in nature
the experiment should be so designed and based on the results of animal
experimentation and a knowledge of the natural history of the disease or
other problem under study that the anticipated results will justify the
performance of the experiment
the experiment should be so conducted as to avoid all unnecessary physical
and mental suffering and injury
no experiment should be conducted where there is an a priori reason to
believe that death or disabling injury will occur; except, perhaps, in
those experiments where the experimental physicians also serve as subjects
the degree of risk to be taken should never exceed that determined by the
humanitarian importance of the problem to be solved by the experiment
proper preparations should be made and adequate facilities provided to
protect the experimental subject against even remote possibilities of injury,
disability or death
the experiment should be conducted only by scientifically qualified persons.
The highest degree of skill and care should be required through all stages
of the experiment of those who conduct or engage in the experiment
during the course of the experiment the human subject should be at liberty
to bring the experiment to an end if he has reached the physical or mental
state where continuation of the experiment seems to him to be impossible
during the course of the experiment the scientist in charge must be prepared
to terminate the experiment at any stage, if he has probable cause to believe,
in the exercise of the good faith, superior skill and careful judgement
required of him, that a continuation of the experiment is likely to result
in injury, disability or death to the experimental project.
Kind of trials :
n of 1 trial : a specific therapy
of uncertain efficacy can be compared with a placebo or alternative therapy
in a double-blind design with well-defined endpoints for a patient with
stable clinical symptomatology
clinical trials
phase I clinical testing is lead by clinical
pharmacologists
Objectives : test drug interactions, pharmacokinetics
(drug absorption, distribution, metabolism, and elimination (ADME)),
and safe dosage range appropriate for use in phase II trials (maximum
tolerated dose (MTD) and recommended dose (RD)
Patient selection criteria : 50-100 closely
monitored healthy volunteers or special populations (e.g. patients with
renal and hepatic impairment, advanced cancer
resistant to standard therapies but with "normal" organ function, HIV-infected
patients)
Starting dose : for INDs, 1/10 of LD10
in the most sensitive species.
Study design : the initial types of phase
I studies include :
phase Ia
single dose
ascending dose tolerance : cohorts of 3-6 pts for each dose level up to
dose-limiting
toxicities (DLT) in 33% of pts (the so-called maximum tolerated
dose (MTD)) (Fibonacci's scheme). When 1 out of 3 patients experience
DLT, other 3 patients are tested, and only if none experiences DLT the
upper dose can be tested in the next group.
phase Ib : relation between dose and biological
effect
Main of issues of concern to be included in a risk analysis of a new compound.
This analysis assumes acceptable and stable pharmaceutical and chemical
quality :
phase II clinical testing is led by clinical
pharmacologists and investigators to correlate blood levels with pharmacologic
activity
(a surrogate endpoint, i.e., ability to induce those pharmacodynamic modifications
by which it is presumed clnical benefit can arise) and adverse events.
Patient selection criteria : same disease,
histology, stage and previous therapies, normal organ function; if IND,
resistance to (or not available) standard therapies is required
phase IIa : studies in a small number (100-200)
of patients to make a preliminary determination of safety and first signs
of efficacy of a new drug to provide proof of the concept.
phase IIb : studies in a larger number of
patients to determine the range of doses to be used in the phase III clinical
trials, establish dose and overall efficacy/safety properties, and the
initial risk/benefit ratio
If pharmacological activity is enormously higher than for currently available
therapies, a phase III trial may become unethical and an NDA can be directly
submitted (e.g. what occurred for imatinib
mesylate)
phase III / pivotal studies
clinical testing is led by clinical investigators who test effectiveness
(ie. ability to induce clinical benefits), monitor effects from long term
use, establish labeling requirements, and determine overall risk/benefit
ratio on several hundred to several thousand (inversely related to
the width of the clinical benefit you aim to demonstrate, usually + 5-10%
with respect to previously available therapies) of selected patients prior
to seeking marketing approval and change guidelines for clinical practice.
Lack of correlation with activity may be due to side effects or post-therapeutical
rebound.
requirements
specific outcomes
well-defined clinical end-point(s)
should be used rather than an intermediate end-point / "surrogate" marker
(i.e. a clinical sign or laboratory test that correlates with the clinical
outcome of a disease). E.g. :
freedom from progression (FFP) / freedom from disease progression (FFDP)
/ freedom from treatment failure (FFTF)
local freedom from progression (LFFP)
no evidence of disease (NED)
symptom-free day (SFD)
minimum clinically important differences (MCID)
response :
clinical response or remission
histologic response or remission
continuous complete histologic remission (CCR)
haematological response or remission
complete hematological response (CHR)
cytogenetic response or remission
major cytogenetic response (MCR)
complete cytogenetic response (CCR)
partial cytogenetic response
minimal cytogenetic response (mCR)
molecular response or remission
complete response or remission (CR)
near-complete response or remission (nCR)
partial response or remission (PR)
very good partial response or remission (VGPR)
stable disease (SD)
progressive disease (PD)
survival :
overall survival (OS) : death from any cause since entry onto trial
cause-specific survival
cause-specific mortality : death related to disease
disease-free survival (DFS) / time to relapse (TTR) / freedom from recurrence
(FFR) / time to progression (TTP) / response duration / duration of response
(DR) : time to relapse or progression since first documentation of
response
disease-specific survival (DSS)
event-free survival (EFS) / time-to-treatment failure (TTF) : failure
or death from any cause since entry onto trial
failure-free survival (FFS)
relapse-free survival (RFS)
progression-free survival (PFS) : disease progression or death from
disease since entry onto trial
time to next treatment : time when new treatment is needed since
entry onto trial
metameter is the measurement or transformation of the measurement
used in evaluating biological tests.
examples of metameters of dose include "milligrams," "moles," "log milligrams,"
"log milligrams per kilogram of body weight," etc.
examples of metameters of response include "increase in blood pressure,
in mmHg," "maximum blood pressure achieved, in mmHg," and "percent increase
in blood pressure."
Metameters are frequently and erroneously chosen only to facilitate statistical
summary and analysis of data; the metameter used may also obscure or influence
the biological interpretation of the data in a manner not intended or expected
by the investigator. For example, implicit in the calculation of "percent
change in blood pressure" is the statement that the final state of the
system is a function of the initial state which may or may not be true.
accuracy of diagnosis and the severity of the disease must be comparable
inn the groups being contrasted. This is a particular issue in developing
therapies for syndromes whose aetiology and/or pathogenesis is poorly understood.
dosages must be chose in a manner that allows relative efficicacy
to be compared at equivalent toxicities or allows relative toxicities to
be compared at equivalent efficacies
randomized controlled
trials (RCTs)
randomized grouping
cross-over experiment : each subject receives the test preparation
at least once, and every test preparation is administered to every subject.
At successive experimental sessions each preparation is "crossed-over"
from
one subject to another. The purpose of the cross-over experiment is to
permit the effects of every preparation to be studied in every subject,
and to permit the data for each preparation to be similarly and equally
affected by the peculiarities of each subject. In a well-designed cross-over
experiment, if it is at all possible, the sequence in which the test preparations
are administered is not the same for all subjects, in order to avoid bias
in the experiment as a result of changes in the behavior of the subjects
that are a function of time rather than of drug administration, or a function
of drug interactions. At least, the cross-over design permits detecting
such biases when they occur. The preparations under test in a cross-over
experiment may - ideally, should - include :
one or more doses, of an experimental or "unknown" drug
one or more doses of a dummy or placebo medication (negative control
drugs)
placebo control : a medicine or preparation with no inherent pertinent
pharmacologic activity which is effective only by virtue of the factor
of suggestion attendant upon its administration
dummy control : a form of placebo mimicking in every way (dosage
form, route of administration, etc.) the purportedly active ingredient
upon which the effectiveness of the active treatment is expected to depend
In therapeutic trials involving life-threatening diseases for which there
already is an effective therapy, the use of a placebo is unethical, and
new treatments must be compared with current therapies.
one or more doses of a "standard" drug, the actions of which are expected
to be similar to those of the "unknown" (positive control drug).
open experiment : pertaining to a clinical trial or other experiment
in which both the subjects and the persons administering the test are aware
of which treatment is administered to which subject.
single-blind experiment : 1 participant - usually the subject -
is left uninformed
double-blind experiment / randomized double-blind controlled trial (RDBCT)
: 2 participants - usually the subject and observer - are uninformed
triple-blind experiment : pertaining to a clinical trial or other
experiment in which neither the subject nor the person administering treatment
nor the person evaluating the response to treatment knows which treatment
any particular subject is receiving. The term triple mask is sometimes
preferred to avoid confusion associated with the use of the term ?blind.?
subjects compliance with the experimental
regimens. Adherence to (or compliance with) a medication regimen is generally
defined as the extent to which patients take medications as prescribed
by their health care providers. The word "adherence" is preferred by many
health care providers, because "compliance" suggests that the patient is
passively following the doctor's orders and that the treatment plan is
not based on a therapeutic alliance or contract established between the
patient and the physician. Both terms are imperfect and uninformative descriptions
of medication-taking behaviorref.
sample size enough to find statistically significant differences
number needed to treat (NNT)
number needed to harm (NNH)
limitations
patients for experimental trials often are selected to eliminate coexisting
diseases and concomitant therapy
trials usually assess the effect of only 1 or 2 drugs, not the many that
might be given to or taken by the same patient under the care of a physician
limited periods of time
compliance may be better controlled than it can be in practice
small numbers of patients => undetected rare side effects
most medications have not been evaluated in women, children or elderlies
Cumulative analysis of many phase III trials with same endpoints (metaanalysis)
is required if results are unconclusive.
During advanced clinical trials, neither doctors nor patients know
whether they are using the drug or the placebo. But this is often not the
case in earlier stages of trials, so patients could draw conclusions about
the success of a prototype drug from their own response to it. Patients
in clinical trials might be using information about the drug they are taking
to trade illegally in pharmaceutical stocks : some patients, who pick up
confidential information about the success of the drug they are taking,
use this inside knowledge to trade in biotech or pharmaceutical company
stocks. This may be illegal under trading rules. So far, there is only
anecdotal evidence that this actually occurs, but the practice could be
common among patients whose friends or family routinely invest in the market.
If pervasive, steps might be introduced to prevent it, such as asking patients
to sign forms that forbid them to release information or trade in any relevant
stock during the trial. But any information patients might have is of little
use to investors, because it could be overturned once all the data are
analysedref Evidence-based medicine (EBM)ref1,
ref2,
ref3,
ref4,
ref5,
ref6,
ref7,
ref8,
ref9,
ref10,
ref11,
ref12,
ref13,
ref14,
ref15,
ref16,
ref17,
ref18,
ref19,
ref20
(based on US Agency for Health Care Policy and Research (AHCPR) 1992) :
level of evidence :
the AHCPR classification is most appropriate for questions of causal relationships,
and is usually used to assign studies, dealing with causal relationships,
to levels of evidence.
level I : intervention is useful and effective (should be administered
unless contraindicated)
level Ib : evidence obtained from at least one properly designed randomised
controlled trial
level IIa : weight of evidence/opinion is in favor of usefulness/efficacy;
evidence obtained from at least one well designed controlled study without
randomisation
level IIb : usefulness/efficacy is less well established by evidence/opinion;
evidence obtained from at least one other type of well designed quasi-experimental
study
level III : intervention is not useful/effective and may be harmful (should
not be administered); evidence obtained from well designed non experimental
descriptive studies, such as comparative studies with historical control,
two or more single-arm studies, or interrupted time series without a parallel
control group, correlation studies and case studies
level IV : evidence obtained from expert committee reports or opinions
and/or clinical experiences of respected authorities on case-series, either
post-test, or pre-test and post-test
This classification however is proving to be increasingly problematic in
guideline development that considers issues beyond intervention, and more
specifically pharmacological interventions, and is also increasingly inadequate
for assigning levels of evidence even to pharmacological intervention evidence.
For example, there is no specific evidence level for systematic reviews
(rather than meta-analysis). In terms of randomised controlled trials (RCTs)
there is no recognition of whether the trial is considered adequately powered,
or other indicators of quality. In non-pharmacological intervention evidence
the problems are considerable. RCTs may contain other information that
is useful. For example, in this guideline valuable epidemiological data
was available from a large RCT (the UKPDS). But it is not obvious how the
evidence classification deals with this. (In this guideline we have given
a level III to the epidemiological evidence from the UKPDS, and level Ia
or Ib to intervention evidence). The classification of studies that are
concerned for example with screening for a condition/disease are not well
served by this classification. None of the screening studies could achieve
a level of evidence higher than III using this system, no matter how good
the screening study. Similarly differentiation between high quality and
lower quality screening studies are obscured by giving all screening studies
level III as this system requires. Although other taxonomies for classifying
and assigning evidence levels to other types of study (with different research
questions as their starting point) have been developed they are not yet
widely used (e.g. evidence levels for studies concerned with diagnosis
and prognosis are detailed in Canadian Medical Association, 1998, S3).
E1 / level I : evidence obtained from a systematic review of all relevant
randomised controlled trials.
E2 / level II : evidence obtained from at least one properly designed randomised
controlled trial.
E31 / level III-1 : evidence obtained from well-designed pseudo-randomised
controlled trials (alternate allocation or some other method).
E32 / level III-2 : evidence obtained from comparative studies
with concurrent controls and allocation not randomised (cohort studies),
case-control studies, or interrupted time series without a parallel control
group.
E33 / level III-3 : evidence obtained from comparative studies
with historical control, two or more single-arm studies, or interrupted
time series without a parallel control group.
E4 / level IV : evidence obtained from case-series, either post-test, or
pre-test and post-test.
US Preventive Services Task Force Rating System of Quality of Scientific
Evidence (US Preventive Services Task Force. Appendix A: task force ratings.
In: Guide to Clinical Preventive Services. 2nd ed. Alexandria, VA: International
Medical Publishing; 1996:861 ?865)
I : evidence obtained from at least 1 properly designed, randomized, controlled
trial
II-1 : evidence obtained from well-designed controlled trials without randomization
II-2 : evidence obtained from well-designed cohort or case-control analytic
studies, preferentially from >1 center or group
II-3 : evidence obtained from multiple time series with or without the
intervention or dramatic results in uncontrolled experiments (such as the
results of the introduction of penicillin treatment in the 1940s)
III : opinions of respected authorities, based on clinical experience,
descriptive studies, or reports of expert committees
grading of recommendations
:
grade A : directly based on category I evidence
grade B : directly based on category II evidence, or extrapolated recommendation
from category I evidence
grade C : directly based on category III evidence, or extrapolated recommendation
from category I or II evidence
grade D : directly based on category IV evidence, or extrapolated recommendation
from category I, II or III
generalizability index :
1 : very likely that results generalize to women
2 : somewhat likely that results generalize to women
3 : unlikely that results generalize to women
0 : unable to project whether results generalize to women
The arguments in favor of the registration of clinical trials are now familiarref1,
ref2,
ref3,
ref4.
Chief among these addresses the practice of selective reporting, whereby
negative or detrimental studies are not brought into the public domain,
which experts on the subject of clinical trials consider an important form
of scientific misconductref.
This practice, as illustrated in a number of high-profile examples, have
increased the demand for the mandatory public registration of clinical
trials. Registration of trials should improve the completeness, reliability,
and quality of the interpretation of clinical research. This need for registration
prompts the question of where trials should be registered. It seems obvious
that, to avoid conflicts of interest and to increase the public trust,
the entities that establish and manage registries should meet certain requirements.
One set of such requirements, as established by the International Committee
of Medical Journal Editors (ICMJE) on September 2005ref,
requires that registries be owned and operated by not-for-profit entities,
that they contain a minimal, clinically directive data setref,
and that they be electronically searchable, without charge, by any interested
party. In October, the US Senate and Congress introduced legislation
seeking mandatory registration of clinical trials.
Through the European Clinical Trials Directive (Directive 2001/20/EC),
the European Union introduced legislation requiring the registration of
"clinical trials on medicinal products for human use" in a European database.
Since 1 May 2004, all clinical trials conducted in member states of the
European Union had to be registered in the EudraCT
database, supervised by the European Medicines Agency (EMEA). This registry
is confidential and open only to regulatory agencies and organizations
that provide funding for research.
The United Kingdom?based Current
Controlled Trials'
metaRegister
of Clinical Trials (mRCT) is an example of a registry that may
compile the data from smaller registries. The World Health Organization
(WHO) is working together with the ISRCTN and ClinicalTrials.gov to develop
a common scheme to reduce duplicate registrations and publications and
to establish the unambiguous identification of trials with the use of a
unique
numbering systemref.
This is an important standard that must be met if registries are to be
maximally effective. But the WHO does not have the last word on this subject.
For example, the Ottawa statementref
on trial registration goes much further than the WHO minimal data set.
Its principles are to disclose the protocol details up front, to disclose
amendments along the way, and to post the results at the end. The principles
underlying this statement have been endorsed by > 100 persons and organizations
on all 5 continents, but not by a single pharmaceutical company. This strategy
is more than wishful thinking. The ultimate goal should be to make trial
protocols publicly available in their entirety, including any financial
arrangements and agreements with regard to publication, so that patients
can be sure that the results will become available. At the same time, it
is the responsibility of those who set the rules and establish the registries
to make them practical to use and understandable for all kinds of research
groups, both small and large. Only then will the use of registries become
sufficiently comprehensive. The widespread use of trial registries will
not prevent negative trial results or unwelcome outcomes from remaining
unpublished. The current demand for the public registration of all clinical
trials at their inception must therefore be followed by a demand for the
addition of all results after each trial is complete and a certain amount
of time has elapsed, to allow the researchers to publish in a journalref.
the International
Standard Randomised Controlled Trial Number (ISRCTN) Register scheme
was launched on a pilot basis in 2000 and formally began operations in
May 2003. The goal of this system is to simplify the identification of
trials and provide a unique number that can be used to track all publications
and reports resulting from each trial. This registry charges a minimal
fee to registrants but is free to all who search its contents. This past
September, ownership of the database was transferred to a not-for-profit
entity, and it now meets all the ICMJE registration requirements. In addition
to EudraCT and ISRCTN, publicly accessible national registries of clinical
trials have been established in several European countries, Japan, and
Australia. There has also been a proliferation of registries based on trials
involving specific diseases. In addition to public registration, the confidential
registration of most clinical trials already exists in France, Italy, Spain,
UK (National Clinical Research Network) and the Netherlands. If these databases
were open to the public, a wealth of information would be unearthed. Although
the desire for regional and specialized registries may be understandable,
it is important for all such registries to contain uniform data elements
and to be linked electronically ? and even better, to share data ? so that
a search for trials that meet certain criteria would automatically cover
all trial registries.
ClinicalTrials.gov,
a public registry that was set up to fulfill the legislative requirement
mandating the registration of U.S. clinical trials involving patients with
serious and life-threatening diseases, was the first to met these requirements.
At first, this database was limited to clinical trials sanctioned by
a U.S. entity, but as of the fall of 2004, it began including clinical
trials from anywhere in the world. Even so, many European researchers
were reluctant to use ClinicalTrials.gov as a locus for trial registration.
Sponsors, principal investigators, or other persons or organizations with
primary responsibility for a given clinical trial (called "data providers")
can register with
ClinicalTrials.gov
through a Web-based systemref.
In some instances, "intermediary trial registries," such as that of the
National
Cancer Institute, provide trial data. The database uses both open-ended
responses and menu-based options, and terms from the National Library of
Medicine Unified Medical Language Systemref1,
ref2
are used to facilitate subsequent information retrieval. Trials with the
same protocol that are conducted at multiple sites are considered one trial
in the registry. The complete entry in the registry for a given trial is
referred to as a record in the database. ClinicalTrials.gov includes both
mandatory and optional data elements. Trials cannot be registered without
the completion of all mandatory data elements, which include both FDAMA
113 and registry-imposed requirements. In addition, the ICMJE requires
completion of some of the optional data elements. Members of the National
Library of Medicine staff manage the quality of information in the registry
by rejecting records that do not have all required fields completed, reviewing
entries for appropriate content and internal consistency, ensuring that
links are active and relevant, checking contact information for recruiting
studies, and confirming that approval from an institutional review board
has been obtained. In addition, sponsoring organizations must electronically
sign off on all entries (and subsequent revisions) before they are made
available on the Web siteref.
Many investigators already register their trials in national or European
databases, but unfortunately, no pan-European agreement has been struck
with regard to how to move forward with public registration of clinical
trialsref.
One measure of medical progress is new treatments. The discovery of a novel
therapy takes time and money, but more important, it requires the mutual
effort of groups that, while they share the common goal of improved treatment,
often have fundamentally competing interests. These interests intersect
at the clinical trial. Patients who are looking for more effective and
safer treatment agree to take part in a clinical trial in the hope that
they will benefit from such treatment or that others with similar conditions
will benefit later. The company developing the new therapy shares the hope
that the trial will be successful, because it wants to market the tested
therapy exclusively and profitably for as long as possible before its competitors
can launch a similar therapy into the marketplace. These goals, though
overlapping, are inevitably in conflict and will generate tension. Such
tension has been thrown into sharp relief over the past 15 months by the
push for clinical trial registration. The academic establishment and patients
have argued that when patients, motivated by altruism, participate (or
even consider participating) in a clinical trial, they are entitled to
understand fully all the options available to them in the various trials
that are currently recruiting subjects. In addition, their participation
in a clinical trial should result in generalizable knowledge that will
be available to future patients and investigators to improve patient care.
This can happen only when appropriate details of the clinical trial are
made available to the public in a timely fashion. The Internet and public
registries have made this possible. Some in industry have argued that to
open their portfolio of clinical trials to public scrutiny, particularly
the scrutiny of other drug companies, would put them at such a competitive
disadvantage that they would be unable to bring new products to market.
Congress, however, decided to encourage openness by enacting on November
21, 1997, Section 113 of the Food and Drug Administration Modernization
Act (FDAMA 113) (Public Health Service Act, 42 U.S.C. 282(j).). Section
113 ultimately created ClinicalTrials.gov
as an Internet-based public resource for information on studies of drugs,
including biologic drug products, that are conducted under the FDA's investigational-new-drug
regulations (Investigational New Drug Application, 21 C.F.R. 312) and involve
the treatment of serious or life-threatening diseases and conditions. In
September 2004, the International Committee of Medical Journal Editors
(ICMJE) announced that its journals would not publish the results of any
ongoing trial that had not been appropriately registered in ClinicalTrials.gov
or another qualified public registry by September 13, 2005ref1,
ref2.
In this issue of the Journal, Zarin et al.5 provide a report card on compliance
with these legislative and ICMJE requirements and the quality of the reporting
that occurred before and after the ICMJE deadline for clinical trial registration.
This report card examines whether the data fields required by FDAMA 113
have been completed in a meaningful fashion, with details about the drug
or other intervention being studied and the prespecified measure of the
trial's primary outcome. Without such critical data, registration becomes
meaningless. Zarin et al. show that there was a dramatic change in the
number of trials registered during the summer of 2005. There can be no
doubt that this spike was related to the ICMJE statement and deadline,
because the rate of registration fell (though to a rate higher than that
before the statement) after the deadline for registration passed. In addition,
they show that the Intervention Name field was universally completed in
a meaningful fashion when the trials were sponsored by academic institutions
or the National Institutes of Health. In contrast, among trials registered
by commercial sponsors, compliance with this field was variable. Here the
message is more nuanced. The vast majority of commercial entities provided
meaningful data in most of their entries before the ICMJE statement and
continued to do so during the summer of 2005. However, in the spring, some
companies, such as Merck, GlaxoSmithKline, and Pfizer, provided meaningful
entries in the Intervention Name field in an astonishingly low number of
registrations. During the summer, Merck amended most of its meaningless
entries to include clinically useful information in this field; by October
they were in compliance in > 99% of their registrations. GlaxoSmithKline
and Pfizer are still using meaningless entries in the Intervention Name
field in 21% and 11% of entries, respectively. This is puzzling, since
most other companies are able to comply fully with the requirements of
FDAMA 113. The second critical measure examined by Zarin et al. was the
number of records with the Primary Outcome field. Here the data are
less reassuring, and the performance of some companies remains abysmal.
Of note, by October 2005, Novartis had completed this field only 3% of
the time, and Merck only 20% of the time. Again, many of their competitors
were in virtually full compliance, undercutting any argument that this
failure reflects a commercial imperative. The ICMJE requirement that clinical
trials be registered if they are to be considered for publication has been
a resounding success. But the report cards for some companies would read
improved but could do better. We demand complete compliance, because trial
registration makes moral sense. When patients put themselves at risk to
participate in clinical trials, they do so with the tacit understanding
that their risk is part of the public record, not merely the secret record
of the sponsor. In our opinion, it is unacceptable for a trial sponsor
not to register its trial in a complete, meaningful, and timely fashion.
We call for all clinical investigators and patients to participate only
in fully registered trials. This call has recently been echoed by the major
organization representing academic medical centers in the USA ? the Association
of American Medical Collegesref.
If a company continues to register trials using meaningless data, with
no respect for the registration process and the patients who participate
in those trials, investigators and patients should refuse to participate.
If a company realizes that secrecy and failure to register mean that it
cannot meet its recruitment goals, it will quickly recognize that the consequence
of that secrecy is commercial failure, not competitive success. We must
monitor the companies that are currently noncompliant to ensure that all
live up to the spirit of the law as reflected in meaningful clinical trial
registrationref Society risked taking for granted the huge successes brought to us
through randomised clinical trials. The vast bureaucracy that now governs
clinical trials is creating a culture in which it is harder, not easier,
to conduct research. The result is not good for patients. Although
much of the over-regulation?in adjudication committees, complex monitoring
arrangements, and demands for voluminous data by national agencies?is done
in the name of patients' safety, few researchers dare to say that this
absurd regulatory hypertrophy is killing more than scientific curiosity.
If
investigators are dissuaded from doing experimental human research, the
plain fact is that patients will die unnecessarily thanks to a diminution
in the rate at which our clinical knowledge advances. The public could
be forgiven for thinking otherwise. Britain's two biggest selling daily
newspapers ran frightening headlines in March this year when TeGenero's
phase I trial of an antibody treatment in healthy volunteers at Northwick
Park Hospital went badly wrong??We saw human guinea pigs explode? (Parker
N, Wheeler V. . The Sun March 16 2006; 1) and ?Hell of human guinea pigs?
(Seamark M, Hope J. . Daily Mail March 17 2006; 1). The idea that doctors
and clinical scientists see patients as little more than guineapigs is
seriously mistaken. Yet this message is transmitted almost with glee by
some influential sections of the mass media when an unforeseen experimental
adverse event takes place. Such a response will surely damage perceptions
of scientific integrity. It is vital that scientists and policymakers do
all they can to protect this integrity and to secure its fragile roots
in our society. This flourishing of clinical research for the benefit of
patients is the overarching objective of WHO's effort to gain agreement
on a practicable way of registering all clinical trials, from the moment
of their inception and for a minimum set of scientifically and ethically
essential elements. 2 principles must underpin trial registrationref
: all interventional trials, including early-phase studies (such as
TeGenero's), should be registered, and all elements of the 20-item minimum
datasetref
must be disclosed at the time of registration.
Moreover, registration of all trials can help countries better manage,
understand, and monitor their clinical research activities?especially given
the trend towards greater conduct of early trials in the developing world
(Vince G. Drug trials enter a new phase. New Scientist 2006; 189: 56-59),
where regulation and oversight can be inadequate. The potential in such
countries for exploitation of vulnerable populationsref
and for not reporting unfavourable trial results is a cause for concern.
Lastly, trials of new interventions, including early trials, are often
terminated for economic reasonsref.
Registration of all trials will ensure that these results are not lost
to the body of general medical knowledge. Arguably, the registration of
early-phase trials (and not only late-phase trials) is particularly indicated,
to ensure that information about the risks of new interventions will be
publicly availableref.
Therefore the Registry Platform considers it a basic principle that all
interventional trials, including early trials in patients or healthy volunteers,
be registered. These positions are consistent with the Declaration
of Helsinkiref
requirement that ?the design of all studies should be publicly available?,
as well as with the recognition by the Declaration of Helsinki and the
Nuremburg Code that the rights of trial participants hold primacy over
commercial and career interests.
These principles have been arrived at after extensive consultation
with all interested parties, including the pharmaceutical industry. WHO's
latest statement therefore sets an important global ethical standard to
which all those involved in clinical research now have an obligation to
adhere. It represents an important step forward in the move to register
trials, efforts that have stumbled on many occasions in the past. Details
of the registration process remain to be resolved. For example, how will
national or disease-specific registers be quality assured? What criteria
will be applied for participating registers to be recognised? How will
results be linked to the original record in the trial register? How can
the minimum dataset for trial registration be expanded to include further
elements that many experts believe are important for the patient to understand
fully the meaning and value of a study? These questions will be answered
over the next 12 months. And WHO would welcome support and commitment to
ensure the successful implementation of the registry platform. But for
now, WHO must begin to build a wide coalition of support for its two principles.
Such an alliance is critically important to the success of this initiative.
Two especially important partners have been the US National Library of
Medicine's ClinicalTrials.gov and the UK's Current Controlled Trials International
Standard Randomised Controlled Trial Number (ISRCTN) register. The Australian
Clinical Trials Registry (ACTR) has also become a vital contributor
to this emerging movement. Most member states of WHO and many pharmaceutical
companies back the agency's plans. Disappointingly, a few have signalled
their opposition to WHO's strong endorsement of transparency surrounding
clinical trials. The Director-General, J W Lee, has successfully resisted
this pressure so far. But he needs the continuing support of patients'
groups and clinical researchers to convert sound principles into real practice.
Journal editors can help. Just as the editors of general medical journals
played a part in raising the profile of trial registrationref,
so editors could again help to move the current debate beyond merely an
aspiration and towards a necessity. Results disclosure will be the next
major step on the road to full transparency of all relevant information
about a particular trial. Editors could drive that disclosure process by
insisting that trialists and sponsors deposit key results information into
a publicly accessible database, akin to GenBank. But, as GlaxoSmithKline's
Frank Rockhold and Ronald Krall point out in this issueref,
some editors are operating policies that inhibit rapid disclosure of trial
data. Editors have been tough on pharmaceutical companies resistant to
registering their trials. They have urged companies, such as GlaxoSmithKline,
to put public interest before commercial interest. Yet journals now find
themselves in a similar position to industry, but with respect to results
disclosure. They?we?have a self-interested motivation to delay full disclosure
of results until publication of a final paper?the Ingelfinger rule.
In the past, editors have sought to control access to research results
by insisting on a journal's priority in releasing data in advance of any
other venue. Happily, that rule has broken down in the face of multiple
outlets for data?in particular, presentation at scientific conferences.
Given the ever-widening flow of information, it would be only a small step
to recognise posting of trials data on an independent results database
as an ethical imperative (Sim I, Owens DK, Lavori DW, Rennels GD. Electronic
trials banks: a complementary method for reporting randomised trials. Med
Decision Making 2000; 20: 440-450). GenBank is the National Institute of
Health's genetic sequence database. It contains an annotated collection
of publicly available DNA sequences. Many journals now require authors
to send their sequence data to GenBank before publication. In that way
an accession number can be published in the final paper and linked electronically
to the scientific record. This has become an ethical norm for genetic sequencing
studies. The same principle could be envisaged for trials. A trials results
bank could contain minimum data derived from a randomised study. Journals
could require that information be submitted to the trials results bank
before publication. Trials registers might seamlessly overlap with the
trials bank. This mechanism would not compromise the publication process.
It would only add to transparency?helping to secure trust and credibility
in the global clinical trials enterprise, an objective vital to patient's
interests.
The industry's share of total biomedical research increased from nearly
a third in 1980 to nearly two-thirds in 2000. Nonetheless, a report documenting
combined data from > 1,100 studies showed that industry-sponsored clinical
trials are "significantly more likely to reach conclusions that were favorable
to the sponsor than were non-industry studies", possibly because of publication
bias or selection of an inappropriate comparator to the drug being evaluatedref.
Within academic institutions, for example, trials funded by industry but
initiated by the investigators typically have 10?20% lower overhead costs.
They tend to restrict industry sponsors to ensuring adherence to good clinical-practice
guidelines and to checking data for serious adverse events, notation errors
and omissions. Hence industry has very limited influence on the conduct
of the trial. In contrast, trials that are both initiated and funded by
industry within academic institutions have higher overhead costs, usually
in the range of 30?40%, paid to the academic institution. Such trials allow
greater involvement by the industry funder, for example in writing the
protocol, training and assisting investigators, and helping in data collection
and analysis.
Web resources :
data sheet : a summary of the information about a drug that is provided
to doctors and pharmacists to enable the drug to be prescribed appropriately.
It contains the uses, doses, contraindications, warnings and precautions
that must be taken into account.
pre-NDA meeting
new drug application (NDA)
(for non-biologicals) or product license application (PLA) (for
biologicals) review by Food and Drug Administration (FDA) or marketing
authorisation application (MAA) by drug regulatory authority in Europe
for approval to market the drug (average 12 months)
supplemental NDA (sNDA) may be submitted for a variety of reasons
such as labeling changes, a new or expanded clinical indication, or a new
dosage form presentation
product license applications (PLA)
biologics license applications (BLA)
phase IIIb : differentiation from other
treatments, exploring use in additional patient populations, seeking new
indications for the study, or exploring AEs.
phase IV / marketing
support trial : these are marketing oriented trials which may extend
the recommended duration of treatment or they may be primarily instructive
in nature to help familiarize a larger number of practitioners with the
drug's efficacy and side effects (seeding studies)
limited market release surveillance study (LMRSS)
postmarketing surveillance
(PMS)ref1,
ref2,
ref3,
ref4,
ref5,
ref6,
ref7,
ref8,
ref9
: all physicians check adverse
drug reactions (ADR),
patterns of drug utilization, and additional indications discovered on
patients given drug for therapy, often in comparison with other treatments.
3 spontaneous signalations are considered an alarm signal, and may require
an observational study to verify, confirm and quantify the reaction. Unfortunately
animal testing is poorly predictive and trials are conducted on a few selected
patients (excluding babies, elderlies and pregnants) for a limited period,
in conditions differing signaficatively from clinical practice and without
concurrent drug interactions. PMS should be practiced at a district level
to face pharmacogenomical issues. Number of signalations per million inhabitants
range from 370 in Sweden to 80 in Italy in 2000, 94,000 in Ireland, 97,295
in UK, 97, 290 in France > Denmark > Germany > Norway > Switzerland > Spain
> Finland > The Netherlands > Italy > Austria > Portugal > Greece). The
last drugs in old classes should never be extensively used at the beginning
of marketing. The strength of evidence for an ADR comes from :
case reports and cases series (82%), including positive dechallenge and
rechallenge
observation studies (18%) : e.g. for oral
contraceptives
and thromboembolism, b-blockers
and sudden death, stilbestrol and vaginal carcinoma, glophenin hypersensitivity,
phenfluramine and interstitial pneumonia, chlormezanone and TEN
The ADR may be :
type A : dose-dependent (e.g. NSAIDs
and SSRIs
and gastrointestinal bleeding, third-generation oral contraceptives and
venous thromboembolism)
type B : dose-independent (e.g. nifedipine
and aplastic anemia, cisapride
and torsade de pointes syndrome when associated with CYP3A4 inhibitors
(e.g. macrolides, antiproteases, azoles), astemizole
and ventricular arrhytmias, alendronate
and gastroesophageal reflux (prevented by assumption with 180 ml of water
and remaining upright for 30' to allow gastric emptying and prevent direct
first contact with gastric mucosa), nimesulide
and hepatotoxicity in Spain and Finland when associated with ethanol)
Waxman-Hatch Act is a 1984 act of Congress that extends patent exclusivity
in marketing a drug for the same number of years that the regulatory process
required (3-4 years). Also defined bioequivalence of some generics and
eliminated many obstacles to generic product introduction in the market.