What Are Randomized Controlled Trials?

Published

A randomized controlled trial (RCT) is a type of scientific experiment which aims to reduce bias when testing a new treatment. The people participating in the trial are randomly allocated to either the group receiving the treatment under investigation or to a group receiving standard treatment (or placebo treatment) as the control.

Randomization minimises selection bias and the different comparison groups allow the researchers to determine any effects of the treatment when compared with the no treatment (control) group, while other variables are kept constant. The RCT is often considered the gold standard for a clinical trial.

Randomized controlled trials are often used to test the efficacy or effectiveness of various types of medical intervention and may provide information about adverse effects, such as drug reactions. Random assignment of intervention is done after subjects have been assessed for eligibility and recruited, but before the intervention to be studied begins.

Random Allocation

Random allocation in real trials is complex, but conceptually the process is like tossing a coin. After randomization, the two (or more) groups of subjects are followed in exactly the same way and the only differences between them is the care they receive.

For example, in terms of procedures, tests, outpatient visits, and follow-up calls, should be those intrinsic to the treatments being compared. The most important advantage of proper randomization is that it minimizes allocation bias, balancing both known and unknown prognostic factors, in the assignment of treatments.

The terms “RCT” and randomized trial are sometimes used synonymously, but the methodologically sound practice is to reserve the “RCT” name only for trials that contain control groups, in which groups receiving the experimental treatment are compared with control groups receiving no treatment (a placebo-controlled study) or a previously tested treatment (a positive-control study).

The term “randomized trials” omits mention of controls and can describe studies that compare multiple treatment groups with each other (in the absence of a control group). Similarly, although the “RCT” name is sometimes expanded as “randomized clinical trial” or “randomized comparative trial”, the methodologically sound practice, to avoid ambiguity in the scientific literature, is to retain “control” in the definition of “RCT” and thus reserve that name only for trials that contain controls.

Not all randomized clinical trials are randomized controlled trials (and some of them could never be, in cases where controls would be impractical or unethical to institute). The term randomized controlled clinical trials is a methodologically sound alternate expansion for “RCT” in RCTs that concern clinical research; however, randomized controlled trials are also employed in other research areas, including many of the social sciences.

Ethics Of Randomized Clinical Trials

Although the principle of clinical equipoise (“genuine uncertainty within the expert medical community… about the preferred treatment”) common to clinical trials has been applied to RCTs, the ethics of RCTs have special considerations. For one, it has been argued that equipoise itself is insufficient to justify RCTs.

For another, “collective equipoise” can conflict with a lack of personal equipoise (e.g., a personal belief that an intervention is effective). Finally, Zelen’s design, which has been used for some RCTs, randomizes subjects before they provide informed consent, which may be ethical for RCTs of screening and selected therapies, but is likely unethical “for most therapeutic trials.”

Although subjects almost always provide informed consent for their participation in an RCT, studies since 1982 have documented that RCT subjects may believe that they are certain to receive treatment that is best for them personally; that is, they do not understand the difference between research and treatment. Further research is necessary to determine the prevalence of and ways to address this “therapeutic misconception”.

The RCT method may also create cultural effects that have not been well understood. For example, patients with terminal illness may join trials in the hope of being cured, even when treatments are unlikely to be successful.

Classifications of Randomized Controlled Trials

By Study Design

One way to classify RCTs is by study design. From most to least common in the healthcare literature, the major categories of RCT study designs are:

  • Parallel-group – each participant is randomly assigned to a group, and all the participants in the group receive (or do not receive) an intervention.

  • Crossover – over time, each participant receives (or does not receive) an intervention in a random sequence.

  • Cluster – pre-existing groups of participants (e.g., villages, schools) are randomly selected to receive (or not receive) an intervention.

  • Factorial – each participant is randomly assigned to a group that receives a particular combination of interventions or non-interventions (e.g., group 1 receives vitamin X and vitamin Y, group 2 receives vitamin X and placebo Y, group 3 receives placebo X and vitamin Y, and group 4 receives placebo X and placebo Y).

An analysis of the 616 RCTs indexed in PubMed during December 2006 found that 78% were parallel-group trials, 16% were crossover, 2% were split-body, 2% were cluster, and 2% were factorial.

By Outcome (Efficacy vs. Effectiveness)

Randomized controlled trials can be classified as “explanatory” or “pragmatic.” Explanatory RCTs test efficacy in a research setting with highly selected participants and under highly controlled conditions.

In contrast, pragmatic RCTs test effectiveness in everyday practice with relatively unselected participants and under flexible conditions; in this way, pragmatic RCTs can “inform decisions about practice.”

By Hypothesis

Another classification of RCTs categorizes them as “superiority trials,” “noninferiority trials,” and “equivalence trials,” which differ in methodology and reporting. Most RCTs are superiority trials, in which one intervention is hypothesized to be superior to another in a statistically significant way.

Some RCTs are noninferiority trials “to determine whether a new treatment is no worse than a reference treatment.” Other RCTs are equivalence trials in which the hypothesis is that two interventions are indistinguishable from each other.

Randomization

The advantages of proper randomization in RCTs include:

  • “It eliminates bias in treatment assignment,” specifically selection bias and confounding.

  • “It facilitates blinding (masking) of the identity of treatments from investigators, participants, and assessors.”

  • “It permits the use of probability theory to express the likelihood that any difference in outcome between treatment groups merely indicates chance.”

There are two processes involved in randomizing patients to different interventions. First is choosing a randomization procedure to generate an unpredictable sequence of allocations; this may be a simple random assignment of patients to any of the groups at equal probabilities, may be “restricted,” or may be “adaptive.”

A second and more practical issue is allocation concealment, which refers to the stringent precautions taken to ensure that the group assignment of patients are not revealed prior to definitively allocating them to their respective groups. Non-random “systematic” methods of group assignment, such as alternating subjects between one group and the other, can cause “limitless contamination possibilities” and can cause a breach of allocation concealment.

However empirical evidence that adequate randomization changes outcomes relative to inadequate randomization has been difficult to detect empirically.

Procedures

The treatment allocation is the desired proportion of patients in each treatment arm.

An ideal randomization procedure would achieve the following goals:

  • Maximize statistical power, especially in subgroup analyses. Generally, equal group sizes maximize statistical power, however, unequal groups sizes maybe more powerful for some analyses (e.g., multiple comparisons of placebo versus several doses using Dunnett’s procedure ), and are sometimes desired for non-analytic reasons (e.g., patients maybe more motivated to enroll if there is a higher chance of getting the test treatment, or regulatory agencies may require a minimum number of patients exposed to treatment).

  • Minimize selection bias. This may occur if investigators can consciously or unconsciously preferentially enroll patients between treatment arms. A good randomization procedure will be unpredictable so that investigators cannot guess the next subject’s group assignment based on prior treatment assignments. The risk of selection bias is highest when previous treatment assignments are known (as in unblinded studies) or can be guessed (perhaps if a drug has distinctive side effects).

  • Minimize allocation bias (or confounding). This may occur when covariates that affect the outcome are not equally distributed between treatment groups, and the treatment effect is confounded with the effect of the covariates (i.e., an “accidental bias”). If the randomization procedure causes an imbalance in covariates related to the outcome across groups, estimates of effect may be biased if not adjusted for the covariates (which may be unmeasured and therefore impossible to adjust for).

However, no single randomization procedure meets those goals in every circumstance, so researchers must select a procedure for a given study based on its advantages and disadvantages.

Simple

This is a commonly used and intuitive procedure, similar to “repeated fair coin-tossing.”

Also known as “complete” or “unrestricted” randomization, it is robust against both selection and accidental biases. However, its main drawback is the possibility of imbalanced group sizes in small randomized controlled trials.

It is therefore recommended only for RCTs with over 200 subjects.

Restricted

To balance group sizes in smaller RCTs, some form of “restricted” randomization is recommended. The major types of restricted randomization used in RCTs are:

  • Permuted-block randomization or blocked randomization: a “block size” and “allocation ratio” (number of subjects in one group versus the other group) are specified, and subjects are allocated randomly within each block. For example, a block size of 6 and an allocation ratio of 2:1 would lead to random assignment of 4 subjects to one group and 2 to the other.

This type of randomization can be combined with “stratified randomization”, for example by center in a multicenter trial, to “ensure good balance of participant characteristics in each group.”

  • A special case of permuted-block randomization is random allocation, in which the entire sample is treated as one block. The major disadvantage of permuted-block randomization is that even if the block sizes are large and randomly varied, the procedure can lead to selection bias. Another disadvantage is that “proper” analysis of data from permuted-block-randomized RCTs requires stratification by blocks.

  • Adaptive biased-coin randomization methods (of which urn randomization is the most widely known type): In these relatively uncommon methods, the probability of being assigned to a group decreases if the group is overrepresented and increases if the group is underrepresented. The methods are thought to be less affected by selection bias than permuted-block randomization.

Adaptive

At least two types of “adaptive” randomization procedures have been used in RCTs, but much less frequently than simple or restricted randomization:

  • Covariate-adaptive randomization, of which one type is minimization: The probability of being assigned to a group varies in order to minimize “covariate imbalance.” Minimization is reported to have “supporters and detractors” because only the first subject’s group assignment is truly chosen at random, the method does not necessarily eliminate bias on unknown factors.[

  • Response-adaptive randomization, also known as outcome-adaptive randomization: The probability of being assigned to a group increases if the responses of the prior patients in the group were favorable. Although arguments have been made that this approach is more ethical than other types of randomization when the probability that a treatment is effective or ineffective increases during the course of an RCT, ethicists have not yet studied the approach in detail.

Allocation Concealment

Allocation concealment” (defined as “the procedure for protecting the randomization process so that the treatment to be allocated is not known before the patient is entered into the study”) is important in RCTs. In practice, clinical investigators in RCTs often find it difficult to maintain impartiality.

Stories abound of investigators holding up sealed envelopes to lights or ransacking offices to determine group assignments in order to dictate the assignment of their next patient. Such practices introduce selection bias and confounders (both of which should be minimized by randomization), thereby possibly distorting the results of the study.

Adequate allocation concealment should defeat patients and investigators from discovering treatment allocation once a study is underway and after the study has concluded. Treatment related side-effects or adverse events may be specific enough to reveal allocation to investigators or patients thereby introducing bias or influencing any subjective parameters collected by investigators or requested from subjects.

Some standard methods of ensuring allocation concealment include sequentially numbered, opaque, sealed envelopes (SNOSE); sequentially numbered containers; pharmacy controlled randomization; and central randomization.

It is recommended that allocation concealment methods be included in an RCT’s protocol, and that the allocation concealment methods should be reported in detail in a publication of an RCT’s results; however, a 2005 study determined that most RCTs have unclear allocation concealment in their protocols, in their publications, or both. On the other hand, a 2008 study of 146 meta-analyses concluded that the results of RCTs with inadequate or unclear allocation concealment tended to be biased toward beneficial effects only if the RCTs’ outcomes were subjective as opposed to objective.

Sample Size

The number of treatment units (subjects or groups of subjects) assigned to control and treatment groups, affects an RCT’s reliability. If the effect of the treatment is small, the number of treatment units in either group may be insufficient for rejecting the null hypothesis in the respective statistical test.

The failure to reject the null hypothesis would imply that the treatment shows no statistically significant effect on the treated in a given test. But as the sample size increases, the same RCT may be able to demonstrate a significant effect of the treatment, even if this effect is small.

Blinding

An RCT may be blinded, (also called “masked”) by “procedures that prevent study participants, caregivers, or outcome assessors from knowing which intervention was received.” Unlike allocation concealment, blinding is sometimes inappropriate or impossible to perform in an RCT; for example, if an RCT involves a treatment in which active participation of the patient is necessary (e.g., physical therapy), participants cannot be blinded to the intervention.

Traditionally, blinded RCTs have been classified as “single-blind,” “double-blind,” or “triple-blind”; however, in 2001 and 2006 two studies showed that these terms have different meanings for different people. The 2010 CONSORT Statement specifies that authors and editors should not use the terms “single-blind,” “double-blind,” and “triple-blind”; instead, reports of blinded RCT should discuss “If done, who was blinded after assignment to interventions (for example, participants, care providers, those assessing outcomes) and how.”

Randomized controlled trials without blinding are referred to as “unblinded”, “open”, or (if the intervention is a medication) “open-label”.

In 2008 a study concluded that the results of unblinded RCTs tended to be biased toward beneficial effects only if the RCTs’ outcomes were subjective as opposed to objective; for example, in an RCT of treatments for multiple sclerosis, unblinded neurologists (but not the blinded neurologists) felt that the treatments were beneficial.

In pragmatic RCTs, although the participants and providers are often unblinded, it is “still desirable and often possible to blind the assessor or obtain an objective source of data for evaluation of outcomes.”

Reporting Of Results

The CONSORT 2010 Statement is “an evidence-based, minimum set of recommendations for reporting RCTs.” The CONSORT 2010 checklist contains 25 items (many with sub-items) focusing on “individually randomised, two group, parallel trials” which are the most common type of RCT.

For other RCT study designs, “CONSORT extensions” have been published, some examples are:

Relative Importance And Observational Studies

Two studies published in The New England Journal of Medicine in 2000 found that observational studies and RCTs overall produced similar results. The authors of the 2000 findings questioned the belief that “observational studies should not be used for defining evidence-based medical care” and that RCTs’ results are “evidence of the highest grade.”

However, a 2001 study published in Journal of the American Medical Association concluded that “discrepancies beyond chance do occur and differences in estimated magnitude of treatment effect are very common” between observational studies and RCTs.

Two other lines of reasoning question RCTs’ contribution to scientific knowledge beyond other types of studies:

Advantages

Randomized controlled trials are considered to be the most reliable form of scientific evidence in the hierarchy of evidence that influences healthcare policy and practice because RCTs reduce spurious causality and bias.

Results of RCTs may be combined in systematic reviews which are increasingly being used in the conduct of evidence-based practice. Some examples of scientific organizations’ considering RCTs or systematic reviews of RCTs to be the highest-quality evidence available are:

As of 1998, the National Health and Medical Research Council of Australia designated “Level I” evidence as that “obtained from a systematic review of all relevant randomised controlled trials” and “Level II” evidence as that “obtained from at least one properly designed randomised controlled trial.”

Since at least 2001, in making clinical practice guideline recommendations the United States Preventive Services Task Force has considered both a study’s design and its internal validity as indicators of its quality.[75] It has recognized “evidence obtained from at least one properly randomized controlled trial” with good internal validity (i.e., a rating of “I-good”) as the highest quality evidence available to it.

The GRADE Working Group concluded in 2008 that “randomised trials without important limitations constitute high quality evidence.”

For issues involving “Therapy/Prevention, Aetiology/Harm”, the Oxford Centre for Evidence-based Medicine as of 2011 defined “Level 1a” evidence as a systematic review of RCTs that are consistent with each other, and “Level 1b” evidence as an “individual RCT (with narrow Confidence Interval).”

Notable RCTs with unexpected results that contributed to changes in clinical practice include:

  • After Food and Drug Administration approval, the antiarrhythmic agents flecainide and encainide came to market in 1986 and 1987 respectively. The non-randomized studies concerning the drugs were characterized as “glowing”, and their sales increased to a combined total of approximately 165,000 prescriptions per month in early 1989. In that year, however, a preliminary report of an RCT concluded that the two drugs increased mortality. Sales of the drugs then decreased.

  • Prior to 2002, based on observational studies, it was routine for physicians to prescribe hormone replacement therapy for post-menopausal women to prevent myocardial infarction. In 2002 and 2004, however, published RCTs from the Women’s Health Initiative claimed that women taking hormone replacement therapy with estrogen plus progestin had a higher rate of myocardial infarctions than women on a placebo, and that estrogen-only hormone replacement therapy caused no reduction in the incidence of coronary heart disease.

Possible explanations for the discrepancy between the observational studies and the RCTs involved differences in methodology, in the hormone regimens used, and in the populations studied. The use of hormone replacement therapy decreased after publication of the RCTs.

Disadvantages

Many papers discuss the disadvantages and limitations of RCTs. Among the most frequently cited drawbacks are:

Limitations Of External Validity

The extent to which RCTs’ results are applicable outside the randomized controlled trials varies; that is, randomized controlled trials’ external validity may be limited. Factors that can affect RCTs’ external validity include:

  • Where the RCT was performed (e.g., what works in one country may not work in another)

  • Characteristics of the patients (e.g., an RCT may include patients whose prognosis is better than average, or may exclude “women, children, the elderly, and those with common medical conditions”)

  • Study procedures (e.g., in an RCT patients may receive intensive diagnostic procedures and follow-up care difficult to achieve in the “real world”)

  • Outcome measures (e.g., randomized controlled trials may use composite measures infrequently used in clinical practice)

  • Incomplete reporting of adverse effects of interventions

However, all research suffers from limitations of external validity, whether observational or experimental, quantitative, or qualitative, since it is limited necessarily by the context in which it takes place.

Time And Costs

RCTs can be expensive; one study found 28 Phase III randomized controlled trials funded by the National Institute of Neurological Disorders and Stroke prior to 2000 with a total cost of US$335 million, for a mean cost of US$12 million per RCT.

Nevertheless, the return on investment of randomized controlled trials may be high, in that the same study projected that the 28 RCTs produced a “net benefit to society at 10-years” of 46 times the cost of the trials program, based on evaluating a quality-adjusted life year as equal to the prevailing mean per capita gross domestic product.

The conduct of an RCT takes several years until being published, thus data is restricted from the medical community for long years and may be of less relevance at time of publication.

It is costly to maintain RCTs for the years or decades that would be ideal for evaluating some interventions.

Interventions to prevent events that occur only infrequently (e.g., sudden infant death syndrome) and uncommon adverse outcomes (e.g., a rare side effect of a drug) would require RCTs with extremely large sample sizes and may therefore best be assessed by observational studies.

Due to the costs of running RCTs, these usually only inspect one variable or very few variables, rarely reflecting the full picture of a complicated medical situation; whereas the case report, for example, can detail many aspects of the patient’s medical situation (e.g. patient history, physical examination, diagnosis, psychosocial aspects, follow up).

Conflict Of Interest Dangers

A 2011 study done to disclose possible conflicts of interests in underlying research studies used for medical meta-analyses reviewed 29 meta-analyses and found that conflicts of interests in the studies underlying the meta-analyses were rarely disclosed. The 29 meta-analyses included 11 from general medicine journals; 15 from specialty medicine journals, and 3 from the Cochrane Database of Systematic Reviews.

The 29 meta-analyses reviewed an aggregate of 509 randomized controlled trials (RCTs). Of these, 318 RCTs reported funding sources with 219 (69%) industry funded. 132 of the 509 RCTs reported author conflict of interest disclosures, with 91 studies (69%) disclosing industry financial ties with one or more authors.

The information was, however, seldom reflected in the meta-analyses. Only two (7%) reported RCT funding sources and none reported RCT author-industry ties. The authors concluded “without acknowledgment of COI due to industry funding or author industry financial ties from RCTs included in meta-analyses, readers’ understanding and appraisal of the evidence from the meta-analysis may be compromised.”

Some RCTs are fully or partly funded by the health care industry (e.g., the pharmaceutical industry) as opposed to government, nonprofit, or other sources. A systematic review published in 2003 found four 1986-2002 articles comparing industry-sponsored and nonindustry-sponsored RCTs, and in all the articles there was a correlation of industry sponsorship and positive study outcome.

A 2004 study of 1999-2001 RCTs published in leading medical and surgical journals determined that industry-funded RCTs “are more likely to be associated with statistically significant pro-industry findings.” One possible reason for the pro-industry results in industry-funded published RCTs is publication bias.

Other authors have cited the differing goals of academic and industry sponsored research as contributing to the difference. Commercial sponsors may be more focused on performing trials of drugs that have already shown promise in early stage trials, and on replicating previous positive results to fulfill regulatory requirements for drug approval.

Jones, Byron; Kenward, Michael G. (2003)
Design and Analysis of Cross-Over Trials (Third ed.).
London: Chapman and Hall ISBN: 978-1439861424

John N.S. Matthews
Introduction to Randomized Controlled Clinical Trials (Second ed.)
Chapman and Hall ISBN: 978-1584886242