The major difference between propensity score matching and synthetic control is that:

  • Journal List
  • Clin Epidemiol
  • v.12; 2020
  • PMC7218288

Clin Epidemiol. 2020; 12: 457–467.

Abstract

There has been a rapid expansion in the use of non-randomized evidence in the regulatory approval of treatments globally. An emerging set of methodologies have been utilized to provide greater insight into external control data used for these purposes, collectively known as synthetic control methods. Through this paper, we provide the reader with a set of key questions to help assess the quality of literature publications utilizing synthetic control methodologies. Common challenges and real-life examples of synthetic controls are provided throughout, alongside a critical appraisal framework with which to assess future publications.

Keywords: synthetic control, RCTs, real-world evidence

Current Challenges of Clinical Trial Investigations

Randomized clinical trials [RCTs] are the gold standard for the evaluation of experimental interventions. In RCTs, patients are usually randomized to either an experimental intervention arm or a control intervention arm that usually consists of placebo or standard-of-care [SOC]. Patient recruitment and retention are two key factors for successful RCTs. The use of placebo, however, can impose recruitment and retention challenges that can halt the timelines of these placebo-controlled trials, as patients have been shown to be less willing to participate in placebo-controlled RCTs.1 The intent of clinical trials is research and not medical care, but still, patients often hope for some level of treatment.2

While the use of active control [ie SOC treatment] has been suggested to address ethical and logistics challenges of associated with placebos, it often presents with similar challenges. In a rapidly progressing field such as oncology, it is not unusual for the SOC to become updated during the course of the trial. An updated SOC can ultimately challenge the fundamental ethical basis of RCTs in “clinical equipoise”, a genuine uncertainty within the scientific and medical community as to which intervention is clinically superior, that justify randomizing patients to the control group.3 In rare diseases, it can be difficult to determine what should be used as an active control, given that there are often no established treatments in these areas. Many clinical trials on rare diseases are conducted with very few patients, translating to insufficient statistical power, or are performed as single-arm trials that make it difficult to compare against other therapeutic options without synthetic control methods.4,5

With the rise in precision medicine, these challenges have been amplified, as more diseases are being diagnosed and classified according to their genetic make-up, resulting in increased sub-stratified disease definitions. For instance, epidermal growth factor receptor [EGFR] is a key mutation for non-small cell lung cancer [NSCLC] patients, and there have been several trial programs that have been based on EGFR mutations for this disease. However, it is important to recognize that only a proportion of NSCLC patients will have an EGFR mutation, so conducting clinical trials that only recruit EGFR-positive patients versus NSCLC patients based on a broader disease classification only will be much more challenging. With these granularities in how diseases are now being classified, ‘rare diseases’ have become more paradoxically common in oncology and other disease areas. Investigators are experiencing increasing challenges of enrolling a sufficiently large number of patients within a reasonable window of time for their clinical trials. While it is difficult to dispute the value of properly conducted RCTs, and the routine, successful implementation of studies utilizing either a placebo or SOC arm, the availability of data sources and methodologies developed to utilize external data have evolved dramatically over recent years. We can optimize the use of external data set with synthetic control methods, but as this is a new concept to many researchers, improving the literacy in these methods is important. For this discussion, we define external data as any source of clinical data from potentially relevant sources, inclusive of clinical trial data, routine health record data, insurance claims data or patient registries. Synthetic controls are defined as cohorts of patients from external data and adjusted using any of a variety of statistical methodologies.

Introduction to Synthetic Controls for Clinical Evaluation

The synthetic control methods are statistical methods that can be used to evaluate the comparative effectiveness of an intervention using external control data. The US Food and Drug Administration [FDA] and European Medicines Agency [EMA] have recognized these issues and taken several initiatives to allow for these novel approaches to external control data.6,7 The FDA approved cerliponase alfa for a specific form of Batten disease, based on synthetic control study that compared the data of 22 patients studied in a single-arm trial versus independent external control group data with 42 untreated patients.8 Across 20 European countries, alectinib, a non-small cell lung cancer treatment, had an expansion of label based on synthetic control study based on an external data set of 67 patients.9 A kinase inhibitor, palbociclib, also had an expanded indication for men with HR+, HER2-advanced or metastatic breast cancer on the basis of external control data.10 The use of non-comparative data is not unique to rare diseases alone, as more common chronic diseases such as hepatitis C and previously treated rheumatoid arthritis have had treatments approved based on non-comparative data.11 Moreover, a recent review of 489 pharmaceutical technologies assessed by the National institute for Health and Care Excellence [NICE] identified 22 submissions that used external data and synthetic control methods to establish clinical efficacy.11 Of these, 13 [59%] utilized published RCT data for their external control, and six [27%] utilized observational data. Over half of the applications were made in the last two recent years alone, further confirming the increasing attention paid by both drug manufacturers and health technology assessment agencies on this topic.

From the conventional evidence-based medicine, the use of external data to create synthetic controls for clinical evaluations represents a radical paradigm shift. A healthy degree of scepticism on the use of synthetic controls is thus expected from the scientific community. Nevertheless, it is likely that there will be an increasing number of clinical trials that use external data as a synthetic control, so it is important for researchers to comprehend the validity and reliability of synthetic control studies. Here in this paper, we provide guidance on what questions researchers must ask when interpreting and critically evaluating the evidence from synthetic control-based clinical trials. For a critical evaluation of synthetic control clinical trials, it will be important for researchers to evaluate the external data that is used itself and the statistical methods used to create a synthetic control group. We have outlined eleven key questions in Table 1 that researchers can ask regarding the validity and quality of trials utilizing external data and synthetic control trials.

Table 1

Synthetic Control Quality Checklist

Item NumberKey QuestionCriteria for Judgement
External Control Data Sources
1 Was the original data collection process similar to that of the clinical trial? State whether patients are from large well-conducted RCT[s] or high-quality prospective cohort studies, and whether patient characteristics are similar to the target population
2 Was the external control population sufficiently similar to the clinical trial population? State how the external population is similar with regards to key characteristics, such as [but not limited to]: age, geographic distribution, performance status, treatment history, sex etc.
3 Did the outcome definitions of the external control match those of that clinical trial? State whether the outcomes are measured similarly or not
4 Was the synthetic control data set sufficiently reliable and comprehensive? State whether there is sufficient sample sizes and covariates that can create comparable control groups
5 Were there any other major limitations to the dataset? State any other potential limitations of the dataset that would limit the reliability and validity of comparisons
Synthetic Control Methods
6 Did the clinical trial include a concurrent control arm, or is the synthetic control data the only control data? State the size of the concurrent control arm and whether the external data set is the only dataset being used or is being used to complement concurrent control arm[s]
7 How was the synthetic control data matched to the intervention group? State the analytical method[s] – eg propensity matching scores – used to create the synthetic control arm
8 Were the results robust to sensitivity assumptions and potential biases? State whether the sensitivity analyses were undertaken or reasons for not conducting sensitivity analyses, and compare whether the sensitivity analyses were comparable to the primary analyses.
9 Were synthetic control comparisons possible for all clinically important outcomes? State if all clinically important outcomes were considered for analyses. If not, state justifications for not including all important outcomes
10 Are the results applicable to your patients? State whether the synthetic control group created are similar to the patient group of interest
11 Were there any other major limitations to the synthetic control methods? State any other potential limitations of the statistical methods that would limit the reliability and validity of comparisons

“Synthetic” Control Data Set

It is important to consider the validity and reliability of the “synthetic” control data set that is used for clinical comparisons of different interventions. For this, it is important to consider the process of the original data collection, compare the populations of the datasets that are being compared, and the reliability and comprehensiveness of the datasets. We have outlined these important considerations in Table 1 and Figure 1.

Was the Original Data Collection Process Similar to That of the Clinical Trial?

Ideally, synthetic controls should be informed by external control data from recent RCTs answering as similar a question as possible, and using as similar designs and implementation processes as possible. Examples of such RCTs would be those investigating a less efficacious intervention in the same population as well as those investigating a broader population where subgroup control data on the target population are available. The data collection in RCTs generally adheres to a high level of stringency. Particularly for RCTs conducted within the same disease areas and within the past 5–10 years, one can usually be reasonably confident that clinical outcome and covariate definitions, value ranges, biomarker kits and thresholds, and others were reasonably similar.

Control data from well-designed observational cohorts may also be adequate, particularly if they have some link to RCTs such as concurrent SOC surveillance, prospective evaluation of efficiency, or were designed to be hypothesis generation for future RCTs. Conversely, control data retrieved from electronic medical records reflect clinical practice and not the controlled environment in which RCTs typically establish efficacy. Pertinent to many future FDA submissions, such data will likely come from large commercial entities selling “real-world data”. Particularly, the data collection and curation processes from such sources may be highly heterogeneous, leaving uncertainty of unknown biases and systematic missing data patterns. The same limitations apply to large case series. Methods exist to minimize these sources of heterogeneity as discussed below, but the resulting dataset is still susceptible to sources of bias.

While publications reporting a clinical trial making use of a synthetic control will rarely provide exhaustive details on the data curation processes from external control sources, a brief description of the external data source[s] as well as a justification for its use may often be available. If confidence in the similarity of data recording processes cannot be asserted from this information, a comparison of published trial protocols may be necessary. If the external data come from non-RCT sources, some additional information is required to assert that the reported data variables are, in fact, sufficiently similar to combine. These recommendations are in line with the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use [ICH] group harmonized tripartite guideline E10.12

Is the External Control Population Sufficiently Similar to the Clinical Trial Population?

Evaluating the similarity of the external control population to the clinical trial population is a multi-faceted exercise. There are many factors that may differ between external control sources and the clinical trial, but not all may matter. Further, not all important factors may be reported or quantifiable [ie unknown confounders]. The eligibility criteria for the considered external control sources should be similar, but this does not guarantee that key patient characteristics are similar. In the context of oncology, if two trials both recruited patients with stage III–IV cancer, but if one predominantly includes stage III patients and the other predominantly includes stage IV patients, these cannot be considered similar. Some account of similarities in the distributions of key baseline characteristics should, therefore, be provided by authors of clinical trials making use of synthetic controls. It is important to note that although patient characteristics may differ in the original external data set, this does not necessarily preclude their use for constructing control groups. Through appropriate statistical adjustments, subgroup analyses or sensitivity analyses, it may be possible to utilize the adjusted external data to create a synthetic control [see section: “Synthetic control methods”].

Other important factors may not be reported, either because of international shifts in clinical research practice or interventional guidelines. In these instances, it may not be possible to successfully generate synthetic controls if data are not available to bridge the gap between these shifts. For example, the World Health Organization previously recommended initiation of antiretroviral therapy [ART] for patients with HIV and a CD4+ cell count of

Chủ Đề