Emulated Clinical Trials from Longitudinal Real-World Data Efficiently Identify Candidates for Neurological Disease Modification: Examples from Parkinson’s Disease ===================================================================================================================================================================== * Daphna Laifenfeld * Chen Yanover * Michal Ozery-Flato * Oded Shaham * Michal Rozen-Zvi * Nirit Lev * Yaara Goldschmidt * Iris Grossman ## Abstract Real-world healthcare data hold the potential to identify therapeutic solutions for progressive diseases by efficiently pinpointing safe and efficacious repurposing drug candidates. This approach circumvents key clinical development challenges, particularly relevant for neurological diseases, concordant with the vision of the 21st Century Cures Act. However, to- date, these data have been utilized mainly for confirmatory purposes rather than as drug discovery engines. Here, we demonstrate the usefulness of real-world data in identifying drug repurposing candidates for disease-modifying effects, specifically candidate marketed drugs that exhibit beneficial effects on Parkinson’s disease (PD) progression. We performed an observational study in cohorts of ascertained PD patients extracted from two large medical databases, Explorys SuperMart (N=88,867) and IBM MarketScan Research Databases (N=106,395); and applied two conceptually different, well-established causal inference methods to estimate the effect of hundreds of drugs on delaying dementia onset as a proxy for slowing PD progression. Using this approach, we identified two drugs that manifested significant beneficial effects on PD progression in both datasets: rasagiline, narrowly indicated for PD motor symptoms; and zolpidem, a psycholeptic. Each confers its effects through distinct mechanisms, which we explored via a comparison of estimated effects within the drug classification ontology. We conclude that analysis of observational healthcare data, emulating otherwise costly, large, and lengthy clinical trials, can highlight promising repurposing candidates for common, late-onset progressive diseases for which disease-modifying therapeutic solutions are scarce. Key words * Real World Data * Clinical Evidence * Parkinson’s disease * Artificial Intelligence * Causal Inference * Rasagiline * Zolpidem * Disease Modifying Therapeutics * Repurposing. ## Introduction Repurposing of marketed drugs is an increasingly attractive prospect for drug developers and patients alike, given the ever-increasing costs of *de novo* drug development1. The rationale underlying the practice of drug repurposing is supported by the demonstration, in a multitude of disease areas, of a drug’s mechanistic and clinical utility for multiple indications, ranging from migraine to autoimmune diseases2–4. While the majority of repurposed drugs have been identified through serendipity, recent years have witnessed a growth in systematic efforts to identify new indications for existing drugs. These efforts include experimental screening approaches5–7 and *in silico* approaches in which existing data are used to discover repurposing candidates (see Ref. 2 for in depth review of these methods). Yet, key challenges in translating repurposing ideas into clinical application have hampered progress along this otherwise promising avenue. Validating the efficacy of a drug for any indication ultimately requires data from humans treated with said drug, traditionally acquired through clinical trials. In the last decade, new opportunities have emerged for acquiring clinical evidence in manners complementing clinical trials, with the growing availability of real-world data (RWD), specifically electronic health records (EHRs) and medical insurance claims data, together with the advent of state-of-the-art computational methodologies. EHRs record multiple health-related data types over time, including drug prescriptions, lab test results of varying nature, physician visits, and symptomology, allowing the relationships between these different features to be assessed. Medical insurance claims data, another form of health related RWD, capture complementary and partially overlapping information, including medical billing claims, enabling research of hospitalizations, doctor’s visits, drug prescription and purchasing, and clinical utilization. In the context of drug repurposing, there have been isolated attempts to use RWD in a confirmatory capacity, to support clinical incidental findings. For example, EHRs have been used to demonstrate an association between metformin and decreased cancer mortality8, and combined EHRs and claims data have been used to support the protective potential of L-DOPA against age-related macular degeneration (AMD)9. Here, we propose a novel approach in which, for the first time, retrospective RWD is used to “industrialize serendipity”. We therefore systematically emulate Phase IIb studies for all concomitant medications used in a disease (for other than disease modifying purposes), in order to identify potential unexpected beneficial effects. Further, investigating the effects of related drugs, e.g., sharing target profile or mechanism of action, allows the extraction of mechanistic explanations for drug effect. These effects, once validated in multiple independent sources of RWD, provide robust evidence on drug efficacy, tolerability, and safety, as well as mechanistic insight on disease modification. It is therefore envisaged that drug candidates identified in this manner will leapfrog into the registration trial phase, confirming aims stated in the USA’s 21st Century Cures Act10, and extending the European Medicines Agency (EMA) current use of RWD as an external control arm in rare disease clinical trials11. The complex nature and organ-inaccessibility of diseases related to the central nervous system (CNS) render them particularly attractive for an RWD-based approach of drug repurposing. For most CNS disorders, our understanding of pathophysiology and underlying etiology is still limited, resulting in poor availability of appropriate, mechanistically-relevant, animal models. Furthermore, clinical trials testing disease-modifying agents require lengthy and large studies, burdening the patient population and incurring high costs of development. Together, these limitations constrain the ability of field experts to rationally design drugs that target these devastating diseases. Thus, using RWD to robustly explore the relationship between various drugs and co-morbidities for which they are not prescribed can help mitigate the risk of lack of predictive animal models, alongside the lengthy clinical studies required to determine outcome in the human setting. An example of such an approach is described in Mittal et al.12 The authors used the Norwegian Prescription Database to demonstrate that individuals prescribed salbutamol (Beta2-adrenoceptor agonist) had a lower incidence of Parkinson’s disease (PD), while those prescribed propranolol (Beta2-antagonist) exhibited higher PD incidence. However, investigation of disease progression or severity was not pursued. PD is one of the most common neurodegenerative disorders, affecting 1 to 2 in 1,000 individuals worldwide and 1% of the population above 60 years of age13. To-date, no disease-modifying agents are approved for PD14, highlighting the need and potential for novel approaches utilizing RWD to bring new therapies to PD patients. One of the hallmark clinical pathologies of PD progression is PD dementia (PDD)15. An estimated 50-80% of PD patients experience dementia as their disease progresses, typically within 10 years of disease onset. It is therefore imperative to identify effective disease-modifying therapeutic agents. In this study, we used, for the first time, RWD from both EHRs and claims data to identify drugs associated with decrease in progression into PDD, as candidates for disease modification of PD. We applied a novel analytical framework of multiple, hierarchical “emulated clinical trials”, an approach that inherently proposes mechanistic rationale for these drugs. ## Methods ### Study design We used the drug repurposing framework16, emulating a randomized controlled trial (RCT) for each candidate drug, combining subject matter expertise with data-driven analysis, and applying a stringent correction for multiple hypotheses. Specifically, it follows the target trial emulation protocol described by Hernán and Robins17, which includes the following steps: define the study eligibility criteria; assign patients to treatment and control cohorts; list and extract a comprehensive set of per-patient baseline covariates; list and extract follow-up disease-related outcome measures; and, finally, use causal inference methodologies18 to retrospectively estimate drug effects on disease outcomes, correcting for confounding and selection biases. We next elaborate on each of these protocol components. #### Eligibility Criteria Patients were included in the PD cohort based primarily on diagnosis codes (Table S1), using the International Classification of Diseases (ICD) system (ICD-9 and ICD-10). We required a repeated PD diagnosis on two distinct dates and excluded patients with secondary parkinsonism or non-PD degenerative disorders. We further excluded early-onset (age <55) PD, and patients with metastatic tumors or those ineligible for prescription drugs through their medical insurance plans. *PD initiation date* was set to the earliest date of first PD diagnosis or a levodopa prescription within the year preceding the first diagnosis of the disease. Since PD is likely present latently before the first diagnostic or prescription record, we retracted the disease initiation date by additional six months. #### Treatment Assignment Each emulated RCT compared PD patients who initiated treatment with either the studied drug (treatment cohort) or an alternative drug (control cohort), comprising at least two prescriptions. The date of treatment initiation (first prescription) was termed *index date*. To avoid confounding by indication, we considered alternative drugs that shared the same (or similar) therapeutic class. Specifically, we first compared each studied drug to drugs taken from its second level Anatomical Therapeutic Chemical (ATC)19 class. Then, for top candidates, we expanded the analysis to control cohorts corresponding to ATC classes of all levels. We only included patients whose PD initiation date preceded the index date and had data history of at least one year prior to the index date. The effect of the drug was measured by the change in the prevalence of newly diagnosed dementia during a follow-up period of two years. Patients with a dementia-related diagnosis at baseline were excluded, as were control cohort patients who were prescribed the trial drug. #### Covariates and Outcomes We extracted hundreds of pre-treatment patient characteristics20 (throughout the one year preceding the index date). A subject matter expert then identified those potentially associated with confounding or selection bias. These included demographic attributes, co-morbidities and PD progression measures, prescribed drugs, healthcare services utilization and socioeconomics parameters (Table 1). The primary endpoint was newly diagnosed dementia as proxy for PD progression; other endpoints considered were fall and psychosis prevalence (see Table S3 for defining ICD codes). View this table: [Table 1.](http://medrxiv.org/content/early/2020/08/14/2020.08.11.20171447/T1) Table 1. Pre-treatment patient characteristics at baseline period #### Data Sources We analyzed two individual-level, de-identified medical databases. The IBM Explorys Therapeutic Dataset (“Explorys”; freeze date: August 2017) includes medical data of >60 million patients, pooled from multiple healthcare systems, primarily clinical EHRs. The IBM MarketScan Research Databases (“MarketScan”; freeze date: mid 2016) contain healthcare claims information from employers, health plans, hospitals, Medicare Supplemental insurance plans, and Medicaid programs, for ~120 million enrollees between 2011 and 2015. ### Statistical Analysis The effect of the trial drug on disease progression was evaluated as the difference between the expected prevalence of the outcome event for drug-treated patients and that in control patients during a complete follow-up period. Briefly, we corrected for potential confounding and selection biases, using two conceptually different causal inference approaches: (i) *balancing weights*, via Inverse Probability Weighting (IPW)21, which reweighs patients to emulate random treatment assignment and uninformative censoring; and (ii) *outcome model*, using standardization22 to predict counterfactual outcomes. We analyzed Explorys and MarketScan separately and report the overlapping candidates. This stringent approach bypasses the need to arbitrarily set aside one database as “confirmatory” and it extends more straightforwardly to >2 data resources. Finally, we used Benjamini and Hochberg’s23 method to correct for multiple hypothesis testing and considered adjusted P-values ≤ 0.05 as statistically significant. For a full description of the RWD-based drug repurposing framework see our methodological paper16. Ground truth effects (that is, RCT-validated) are, typically, unavailable for drug repurposing candidates; notably, however, the estimated effects showed significant correlation across different algorithms and data sources (adjusted P-value < 0.05 for all comparisons across outcomes, databases, and causal inference algorithms), attesting to the robustness of the framework. ## Results We first extracted cohorts of late-onset PD patients comprising 106,000 and 89,000 patients in MarketScan and Explorys, respectively, representing 0.09% and 0.15% of the total databases and consistent with recent epidemiological surveys13. Key characteristics of these separate cohorts (Table 2) exhibit high similarity in the average and range of age at first recorded diagnosis, the percentage of women, the fraction of patients with public insurance, and the baseline Charlson comorbidity index24. Notable dissimilarities between the two cohorts include the average total patient time in database, which was more than double in Explorys compared to MarketScan (Table 2). This dissimilarity stems from the different timespan covered in general by the two databases (average total patient timeline of 4.7±17.4 years in Explorys vs. 2.2±1.6 years in MarketScan). View this table: [Table 2.](http://medrxiv.org/content/early/2020/08/14/2020.08.11.20171447/T2) Table 2. PD cohort characteristics Overall, 218 drugs were prescribed to at least 100 PD patients in both the MarketScan and Explorys PD cohorts. We used this lower bound since many Phase III clinical trials, including those pursued in neurological indications, find 100 or less patients per arm to be satisfactory. Of these, we were able to balance the treatment and control cohorts (using IPW, see Methods) for 205 drugs (94%). Consequently, for each such drug we emulated an RCT, estimating its effect on the population-level prevalence of newly diagnosed dementia, in comparison to the level-2 Anatomical Therapeutic Chemical (ATC) control cohort. Using two independent causal inference methods, outcome model and balancing weights, our analysis identified, in both data sources, two candidate drugs estimated to significantly reduce dementia prevalence: rasagiline and zolpidem (see cohort characteristics in Tables S4-S7). Details of the emulated RCTs, estimating the effect of these drugs compared to their corresponding control ATC level-2 class, are shown in Table 3,4, S8 and S9. Figure 1 shows the prevalence of newly diagnosed dementia in the treatment and control cohorts throughout the follow-up period. Consistently, rasagiline is estimated to decrease the prevalence of newly diagnosed dementia by 7-9%, compared to symptomatic PD drugs. Zolpidem, compared to the class psycholeptics drugs, reduces dementia prevalence by 8-12%. View this table: [Table 3.](http://medrxiv.org/content/early/2020/08/14/2020.08.11.20171447/T3) Table 3. Rasagiline significantly attenuates PD progression View this table: [Table 4.](http://medrxiv.org/content/early/2020/08/14/2020.08.11.20171447/T4) Table 4. Zolpidem significantly attenuates PD progression ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/08/14/2020.08.11.20171447/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2020/08/14/2020.08.11.20171447/F1) Figure 1: Rasagiline and Zolpidem significantly delay the onset of dementia in PD patients in two independent datasets Kaplan-Meier plots comparing the prevalence of newly diagnosed dementia in the treatment and control cohorts, corrected with inverse probability weighting (IPW, dark color), or uncorrected (light color). Red and blue lines show the expected percentage of patients not yet diagnosed with dementia at each time point among the patients who take the drug and among the patients who take other ATC level 2 drugs (N04: symptomatic PD drugs; N05: Psycholeptics), respectively. The difference between each pair of red and blue lines correspond to the expected effect of the drug. Next, we expanded the analysis to consider all four ATC levels that include each drug, corresponding to anatomical main group (level 1), therapeutic subgroup (level 2), pharmacological subgroup (level 3), and chemical subgroup (level 4). Specifically, we compared each drug against all its encompassing ATC classes and additionally, each encompassing ATC class against all upper-level classes in the ATC hierarchy. The resulting set of RCTs estimates the effect of a target drug against drugs sharing its mechanism of action (MoA; e.g., rasagiline versus other monoamine oxidase B inhibitors, ATC class N04BD), as well as drugs conferring different MoAs (e.g., monoamine oxidase B inhibitors, N04BD, versus other dopaminergic agents, N04B), thus testing a set of related mechanistic hypotheses. This can be viewed as sensitivity analyses for the effect of a target drug. Table S11 shows the complete results of these emulated RCTs. ATC level-4 class N04BD, monoamine oxidase (MAO) B inhibitors, included only two drugs: rasagiline and selegiline. Therefore, the rasagiline versus N04BD emulated trial is essentially a head-to-head comparison between these two drugs. The results of the emulated trials in both MarketScan and Explorys suggest that the use of rasagiline reduces the prevalence of dementia compared to selegiline (Table 3; estimations using outcome model are significant). When compared to higher level ATC classes – specifically, dopaminergic agents, symptomatic PD drugs, and nervous system medications – all dominated by levodopa (77-82% of first prescriptions), rasagiline is estimated to significantly decrease dementia prevalence by 5-9% in both databases, using either causal inference approach (Table 3, and Table S8). We also estimated the effect of rasagiline on the prevalence of falls and psychosis: In MarketScan, rasagiline is estimated, by both causal inference algorithms, to decrease the population prevalence of falls compared to all its encompassing ATC classes; In Explorys, rasagiline is estimated to have a beneficial effect on the prevalence of psychosis (but only a subset of these estimands were significant). Zolpidem was estimated to have significant and beneficial effects on the prevalence of dementia only in comparison to its level-2 ATC class, psycholeptics (Table 4, and Table S9). The analysis in MarketScan suggests that zolpidem has a beneficial effect compared to other hypnotics and sedatives (N05C), but the different composition of the N05C control cohort in Explorys (dominated by midazolam) hinders conclusive results. Zolpidem was also estimated to have beneficial effects on the prevalence of falls and psychosis, compared to psycholeptics, but these effects were not significant. ## Discussion The present study used both EHRs and insurance claims data to assess the effects of hundreds of concomitant drugs on PD-associated dementia as one of the more common hallmarks of PD progression. Only those drugs for which a statistically significant effect was found independently in both EHR and claims data were further considered for their repurposing potential. Given the different nature of the data collected with each health data source and stringent statistical approach, the resultant repurposing candidates have a high likelihood of success in a Phase III prospective study. Our analysis unraveled therapeutic benefits of two drugs in decreasing the population-level incidence of PDD, representing slowing of PD disease progression. Thus, long-term treatment (24 months) with rasagiline, a MAO-B inhibitor narrowly indicated for PD motor symptoms, or with zolpidem, a gamma-aminobutyric acid (GABA)-A receptor modulator indicated for insomnia, is strongly associated with decreased PDD incidence in two separate large cohorts (N=195,262 in total). Indeed, the mechanistic, and at times clinical, support for the identified associations, as described below, not only provides support for the approach in identifying new drug repurposing candidates, but also a vehicle to bolster otherwise ambiguous results from RCTs. Cognitive impairment is highly prevalent in patients with progressive stages of PD and is associated with adverse health outcomes and increased mortality25. Cognitive deficits vary in quality and severity in different stages of disease progression in PD, ranging from subjective cognitive decline to mild cognitive impairment and to subsequent PDD. The latter is defined as acquired objective cognitive impairment in multiple domains, including attention, memory, executive and visuospatial ability26, and results in adverse alteration of activities of daily life27. A 78% eight-year cumulative prevalence of PDD development was reported in Norwegian PD patients28, and 83% over 20 years in Australia29. A single choline esterase inhibitor, rivastigmine, is approved by the US Food and Drug Administration (FDA) for the treatment of PDD, with modest efficacy30 resulting in a significant unmet medical need for additional pro-cognitive therapies31. Our finding that rasagiline slows PD progression is consistent with mechanistic evidence and extends prior clinical data. Clinical trials of rasagiline in PD patients implied possible disease-modifying effects, albeit inconclusively. Indeed, none of the studies reported to-date had the statistical power to support or refute slowing the progression of the disease. The largest study to assess disease-modifying effects of rasagiline was ADAGIO32, which failed to demonstrate a dose-dependent effect on the Unified Parkinson’s Disease Rating Scale (UPDRS) scores. This failure may be partly due to insufficient statistical power: the total number of participants in the ADAGIO study was N=1,176, much smaller than in our study (N=13,562 in Explorys; N=13,373 in MarketScan; See Table 3). Additionally, the ADAGIO study did not directly assess effects of rasagiline on cognition. Several recent studies addressed this hypothesis more directly, but were small (N=34-151) and short (3-6 months), yielding mixed results33-34. Importantly, many of the prior reports sought to demonstrate disease prevention/protection in as-yet-to-be-diagnosed patients, while we studied patients with confirmed PD diagnosis. Due to this important distinction, it can be expected that the class and specific agents reported, e.g., by Mittal et al35, to decrease (or increase) PD incidence did not show, in our analysis, similar effects. Overall, inadequate power and diverse study designs hampered conclusive therapeutic interpretation of the role of rasagiline, and the monoamine B class, as PD disease modifiers. Indeed, our approach directly resolved these shortcomings, dramatically increasing sample size and follow-up duration by virtue of the use of RWD, facilitating the discovery of rasagiline’s robust and consistent disease-modifying effects. Importantly, our analysis of proxy parameters supports the beneficial effects of rasagiline on PD progression beyond PDD, as reflected by a decrease in the population prevalence of falls and the trend reduction of psychosis (data not shown). Mechanistically, rasagiline has been suggested to have neuroprotective effects mediated by its ability to prevent mitochondrial permeability transition36. In addition, rasagiline induces anti-apoptotic pro-survival proteins, Bcl-2 and glial cell-line derived neurotrophic factor (GDNF) and increases expression of genes coding for mitochondrial energy synthesis, inhibitors of apoptosis, and the ubiquitin-proteasome system. Finally, systemic administration of selegiline and rasagiline increases neurotrophic factors in cerebrospinal fluid of PD patients and nonhuman primates37. The association between zolpidem, a non-benzodiazepine hypnotic drug used for the treatment of sleeping disorders, and decreased PDD incidence identified herein is a novel finding. In fact, a single prior report published more than two decades ago speculated that zolpidem would not be efficacious for PD, based on the limited clinical experience with the drug at the time, without specific consideration for cognition38. However, recent publications demonstrate Zolpidem’s ability to treat a large variety of neurologic disorders, most often related to movement disorders and disorders of consciousness, and suggest zolpidem induces transient effects on UPDRS39. Of note, several cross-sectional reports have raised concerns for increased risk of reversible dementia or Alzheimer’s diseases in the general population when exposed to zolpidem40,41. However, these reports considered only a handful of potential confounding biases, and applied regression-based methods, which unlike IPW, do not allow one to determine whether treatment and control biases were successfully eliminated21. Furthermore, neither report assessed impact on specific patient subsets, such as those diagnosed with PD. Indeed, a proof-of-concept clinical study is currently recruiting subjects in order to assess the benefits of low-dose zolpidem in late-stage PD ([NCT03621046](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT03621046&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom)), supporting the findings reported herein. Yet again, the limited sample size (N=28) in the recruiting study, together with the inclusion of cognition as a secondary (rather than primary) endpoint both pose a high risk for insufficient power and thus inconclusive results. Finally, latest literature reports on beneficial effects of zolpidem on renal damage and akinesia42 support a high benefit-risk profile of repurposing zolpidem for slowing or reversal of PD. Mechanistically, a structural relationship between the antioxidant melatonin and zolpidem suggests possible direct antioxidant and neuroprotective properties of zolpidem. Garcia-Santos et al43 demonstrated that zolpidem prevented induced lipid peroxidation in rat liver and brain homogenates, showing antioxidant properties similar to melatonin. Bortoli et al42 investigated *in silico* the antioxidant potential of zolpidem and identified it as an efficient radical scavenger similar to melatonin and Trolox. Although the mechanisms involved in the pathogenesis and progression of PD are not fully understood, there is overwhelming evidence that oxidative stress plays an important role in dopaminergic neuronal degeneration. Since the maintenance of reduction-oxidation reaction potential is an important determinant of neuronal survival44, its disruption ultimately leads to cell death. Accumulating evidence from patients and disease models indicate that oxidative and nitrative damage to key cellular components is important in the pathogenesis of PD progression45. Oxidative stress plays an important role in dopaminergic neuronal degeneration, triggering a cascade of events, including mitochondrial dysfunction, impairment of nuclear and mitochondrial DNA, and neuroinflammation, which in turn cause more reactive-oxygen species (ROS) production46. Genetic forms of PD, including those caused by mutations in *PARK7, PINK1, PRKN, SNCA* and *LRRK2*, also demonstrate the fundamental role that mitochondrial function plays in disease etiology45. Thus, the protective effects of zolpidem on the development of dementia could be explained by the antioxidant and neuroprotective capacities of the drug. In a preliminary method development study16, we validated the drug repurposing framework used here. We demonstrated that treatment effects estimated across different data sources and causal methodologies showed a high degree of agreement (P-value < 0.05 for all comparisons). Yet, the retrospective design of the study, combined with the use of RWD, introduces some limitations. Specifically, proxies with reliable representation in the data are required to emulate the endpoints in prospective clinical trials and need to be further assessed and refined in a controlled clinical environment47. Conversely, the mechanistic nature of the drug effects, and therefore potential utility in combination therapy for synergistic effects, require further assessment in a dedicated prospective study, consistent with the drug development paradigm. In addition, while RWD used in a retrospective manner enables the assessment of chronic processes, without the need for lengthy studies, they are bound by the length of follow-up data per individual. Finally, local healthcare practice may at times confound the analysis and requires in-depth understanding of such practices in data interpretation48. Notwithstanding these limitations, discoveries stemming from RWD of large, well-characterized patient populations can provide valuable clues to effective mechanisms and existing medications that may be beneficial in slowing disease progression, or potentially preventing it altogether. In the realm of CNS-related diseases, the extensive follow-up integral to medical-record tracking presents a well-suited setting for investigating the effects of concomitant interventions. The EMA has already employed RWD in lieu of control arms to support regulatory decisions either at authorization or for indication extension, in the context of rare, orphan diseases11. Similarly, the 21st Century Cures Act10 requires that the FDA establish a framework to evaluate the potential use of RWD in support of approval of new indications for approved drugs. In fact, successful examples are already being implemented49. Accordingly, the FDA allotted $100 million to build an EHR database of 10 million people as a foundation for more robust postmarketing studies. The current study provides evidence in support of such uses for RWD, accelerating the availability of solutions for patients in need. In conclusion, we demonstrated that emulating clinical trials based on observational healthcare data identifies promising repurposing drug candidates, efficiently relieving the societal burden of costly, large, and lengthy clinical trials. This approach is particularly relevant as a therapeutic discovery engine for common, late-onset progressive CNS diseases for which disease-modifying therapeutic solutions are scarce. The two drugs identified herein, rasagiline and zolpidem, both hold great promise as disease-modifying agents for PD, in general, and specifically in addressing aspects of cognitive impairment in PD. Further, these cognitive benefits may extend to other neurodegenerative diseases. The ability to systematically compare effects between various drug classes, as well as within classes, in patients in real-world settings is a significant step in accelerating patients’ access to safe and efficacious therapies. ## Data Availability The data analyzed in this paper is available in (i) IBM Explorys Therapeutic Dataset and (ii) IBM MarketScan Research Databases. [https://www.ibm.com/products/marketscan-research-databases](https://www.ibm.com/products/marketscan-research-databases) [https://www.ibm.com/watson-health/about/explorys](https://www.ibm.com/watson-health/about/explorys) ## Supplementary Information **Table S1**. ICD codes for PD cohort definition. **Table S2**. PD-indicated drugs and their corresponding ATC class names. **Table S3**. PD outcome definitions. **Table S4**. Rasagiline cohort characteristics. **Table S5**. Zolpidem cohort characteristics. **Table S6**. N04 cohort characteristics. **Table S7**. N05 cohort characteristics. **Table S8**. Estimated effects on dementia onset for emulated RCTs involving rasagiline and its encompassing ATC classes. **Table S9**. Estimated effects on dementia onset for emulated RCTs involving zolpidem and its encompassing ATC classes. **Table S10**. Trial drugs distribution. (Trial_drugs.xlsx) **Table S11**. Emulated RCTs outcomes and effects. (RCTs outcomes and effects.xlsx) ## Footnotes * **Conflict of Interest:** The authors declared no competing interests for this work. * **Funding:** No funding was received for this work. * Received August 11, 2020. * Revision received August 11, 2020. * Accepted August 14, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.Ashburn, T. T. & Thor, K. B. Drug repositioning: identifying and developing new uses for existing drugs. Nature Reviews Drug Discovery 3, 673 (2004). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrd1468&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15286734&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000223005000018&link_type=ISI) 2. 2.Cha, Y. et al. Drug repurposing from the perspective of pharmaceutical companies: Drug repurposing in pharmaceutical companies. British Journal of Pharmacology 175, 168–180 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/bph.13798&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28369768&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) 3. 3.Xiao, L. et al. Quetiapine facilitates oligodendrocyte development and prevents mice from myelin breakdown and behavioral changes. Molecular Psychiatry 13, 697–708 (2008). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/sj.mp.4002064&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17684494&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000256832700008&link_type=ISI) 4. 4.Yong, P. F. & D’Cruz, D. P. Mycophenolate mofetil in the treatment of lupus nephritis. Biologics 2, 297–310 (2008). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19707362&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) 5. 5.Buckley, C. E. et al. Drug reprofiling using zebrafish identifies novel compounds with potential pro-myelination effects. Neuropharmacology 59, 149–159 (2010). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuropharm.2010.04.014&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20450924&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000280220900003&link_type=ISI) 6. 6.Deshmukh, V. A. et al. A regenerative approach to the treatment of multiple sclerosis. Nature 502, 327–332 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature12647&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24107995&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000325706300044&link_type=ISI) 7. 7.Najm, F. J. et al. Drug-based modulation of endogenous stem cells promotes functional remyelination in vivo. Nature 522, 216–220 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature14335&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25896324&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) 8. 8.Xu, H. et al. Validating drug repurposing signals using electronic health records: a case study of metformin associated with reduced cancer mortality. Journal of the American Medical Informatics Association (2014).doi:10.1136/amiajnl-2014-002649 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1136/amiajnl-2014-002649&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25053577&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) 9. 9.Brilliant, M. H. et al. Mining Retrospective Data for Virtual Prospective Drug Repurposing: L-DOPA and Age-related Macular Degeneration. The American Journal of Medicine 129, 292–298 (2016). 10. 10.21st Century Cures Act, Pub. L. No. 114-255. (2016).at <[https://www.congress.gov/bill/114th-congress/house-bill/34/text](https://www.congress.gov/bill/114th-congress/house-bill/34/text)> 11. 11.Cave, A., Kurz, X. & Arlett, P. Real-World Data for Regulatory Decision Making: Challenges and Possible Solutions for Europe. Clin. Pharmacol. Ther. 106, 36–39 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/cpt.1426&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) 12. 12.Mittal, S. et al. β2-Adrenoreceptor is a regulator of the α-synuclein gene driving risk of Parkinson’s disease. Science 357, 891–898 (2017). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNTcvNjM1NC84OTEiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wOC8xNC8yMDIwLjA4LjExLjIwMTcxNDQ3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 13. 13.Tysnes, O.-B. & Storstein, A. Epidemiology of Parkinson’s disease. J Neural Transm (Vienna) 124, 901–905 (2017). 14. 14.Lang, A. E. & Espay, A. J. Disease Modification in Parkinson’s Disease: Current Approaches, Challenges, and Future Considerations: Disease Modification in PD. Movement Disorders 33, 660–677 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/mds.27360&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29644751&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) 15. 15.Hely, M. A., Reid, W. G. J., Adena, M. A., Halliday, G. M. & Morris, J. G. L. The Sydney multicenter study of Parkinson’s disease: The inevitability of dementia at 20 years: Twenty Year Sydney Parkinson’s Study. Movement Disorders 23, 837–844 (2008). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/mds.21956&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18307261&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000255600200010&link_type=ISI) 16. 16.Ozery-Flato, M., Goldschmidt, Y., Shaham, O., Ravid, S. & Yanover, C. Framework for Identifying Drug Repurposing Candidates from Observational Healthcare Data. *medRxiv* 2020.01.28.20018366 (2020). doi:10.1101/2020.01.28.20018366 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMC4wMS4yOC4yMDAxODM2NnYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDgvMTQvMjAyMC4wOC4xMS4yMDE3MTQ0Ny5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 17. 17.Hernan, M. A. & Robins, J. M. Using Big Data to Emulate a Target Trial When a Randomized Trial Is Not Available. Am J Epidemiol 183, 758–764 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/aje/kwv254&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26994063&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) 18. 18.Hernan, M. A. & Robins, J. M. Causal Inference. (Boca Raton: Chapman & Hall/CRC, 2019). 19. 19.World Health Organization WHO Collaborating Centre for Drug Statistics Methodology: ATC classification index with DDDs. Oslo, Norway: Norwegian Institute of Public Health (2020). 20. 20.Ozery-Flato, M. et al. Fast and Efficient Feature Engineering for Multi-Cohort Analysis of EHR Data. Stud Health Technol Inform 235, 181–185 (2017). 21. 21.Austin, P. C. An Introduction to Propensity Score Methods for Reducing the Effects of Confounding in Observational Studies. Multivariate Behav Res 46, 399–424 (2011). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/00273171.2011.568786&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21818162&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000291533400002&link_type=ISI) 22. 22.Hernan, M. A. & Robins, J. M. Causal Inference: What If. (Boca Raton: Chapman & Hall/CRC, 2020). 23. 23.Benjamini, Y. & Hochberg, Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. Journal of the Royal Statistical Society. Series B (Methodological) 57, 289–300 (1995). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2307/2346101&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=WOS:A1995QE4&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1995QE45300017&link_type=ISI) 24. 24.Charlson, M. E., Pompei, P., Ales, K. L. & MacKenzie, C. R. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis 40, 373–383 (1987). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/0021-9681(87)90171-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=3558716&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1987G855900002&link_type=ISI) 25. 25.Bäckström, D. et al. Early predictors of mortality in parkinsonism and Parkinson disease: A population-based study. Neurology 91, e2045-e2056 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1212/WNL.0000000000006576&link_type=DOI) 26. 26.Emre, M. et al. Clinical diagnostic criteria for dementia associated with Parkinson’s disease. Movement Disorders 22, 1689–1707 (2007). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/mds.21507&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17542011&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000249992700002&link_type=ISI) 27. 27.American Psychiatric Association Diagnostic and Statistical Manual of Mental Disorders. (American Psychiatric Association, 2013).doi:10.1176/appi.books.9780890425596 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.books.9780890425596&link_type=DOI) 28. 28.Aarsland, D., Andersen, K., Larsen, J. P., Lolk, A. & Kragh-Sørensen, P. Prevalence and characteristics of dementia in Parkinson disease: an 8-year prospective study. Arch. Neurol. 60, 387–392 (2003). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archneur.60.3.387&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12633150&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000181561500010&link_type=ISI) 29. 29.Hely, M. A., Reid, W. G. J., Adena, M. A., Halliday, G. M. & Morris, J. G. L. The Sydney multicenter study of Parkinson’s disease: The inevitability of dementia at 20 years: Twenty Year Sydney Parkinson’s Study. Movement Disorders 23, 837–844 (2008). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/mds.21956&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18307261&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000255600200010&link_type=ISI) 30. 30.Meng, Y., Wang, P., Song, Y. & Wang, J. Cholinesterase inhibitors and memantine for Parkinson’s disease dementia and Lewy body dementia: A meta-analysis. Experimental and Therapeutic Medicine (2018).doi:10.3892/etm.2018.7129 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3892/etm.2018.7129&link_type=DOI) 31. 31.Green, H., Tsitsi, P., Markaki, I., Aarsland, D. & Svenningsson, P. Novel Treatment Opportunities Against Cognitive Impairment in Parkinson’s Disease with an Emphasis on Diabetes-Related Pathways. CNS Drugs (2019).doi:10.1007/s40263-018-0601-x [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s40263-018-0601-x&link_type=DOI) 32. 32.Olanow, C. W. et al. A Double-Blind, Delayed-Start Trial of Rasagiline in Parkinson’s Disease. New England Journal of Medicine 361, 1268–1278 (2009). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa0809335&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19776408&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000270078700008&link_type=ISI) 33. 33.Hanagasi, H. A. et al. The effects of rasagiline on cognitive deficits in Parkinson’s disease patients without dementia: A randomized, double-blind, placebo-controlled, multicenter study. Mov. Disord. 26, 1851–1858 (2011). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/mds.23738&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21500280&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) 34. 34.Frakey, L. L. & Friedman, J. H. Cognitive Effects of Rasagiline in Mild-to-Moderate Stage Parkinson’s Disease Without Dementia. The Journal of Neuropsychiatry and Clinical Neurosciences 29, 22–25 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.neuropsych.15050118&link_type=DOI) 35. 35.Mittal, S. et al. β2-Adrenoreceptor is a regulator of the α-synuclein gene driving risk of Parkinson’s disease. Science 357, 891–898 (2017). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNTcvNjM1NC84OTEiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wOC8xNC8yMDIwLjA4LjExLjIwMTcxNDQ3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 36. 36.Naoi, M. & Maruyama, W. Functional mechanism of neuroprotection by inhibitors of type B monoamine oxidase in Parkinson’s disease. Expert Review of Neurotherapeutics 9, 1233–1250 (2009). 37. 37.Naoi, M. et al. Neuroprotection by propargylamines in Parkinson’s disease: intracellular mechanism underlying the anti-apoptotic function and search for clinical markers. J. Neural Transm. Suppl. 121-131 (2007). 38. 38.Lavoisy, J. & Marsac, J. Zolpidem in Parkinson’s disease. The Lancet 350, 74 (1997). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(05)66285-X&link_type=DOI) 39. 39.Bomalaski, M. N., Claflin, E. S., Townsend, W. & Peterson, M. D. Zolpidem for the Treatment of Neurologic Disorders: A Systematic Review. JAMA Neurology 74, 1130 (2017). 40. 40.Shih, H.-I. et al. An Increased Risk of Reversible Dementia May Occur After Zolpidem Derivative Use in the Elderly Population. Medicine (Baltimore) 94, (2015). 41. 41.Lee, J., Jung, S. J., Choi, J., Shin, A. & Lee, Y. J. Use of sedative-hypnotics and the risk of Alzheimer’s dementia: A retrospective cohort study. PLoS ONE 13, e0204413 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0204413&link_type=DOI) 42. 42.Bortoli, M. et al. Psychiatric Disorders and Oxidative Injury: Antioxidant Effects of Zolpidem Therapy disclosed In Silico. Computational and Structural Biotechnology Journal 17, 311–318 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.csbj.2019.02.004&link_type=DOI) 43. 43.García-Santos, G. et al. Antioxidant Activity and Neuroprotective Effects of Zolpidem and Several Synthesis Intermediates. Free Radical Research 38, 1289–1299 (2004). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/10715760400017343&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15763953&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) 44. 44.Puspita, L., Chung, S. Y. & Shim, J. Oxidative stress and cellular pathologies in Parkinson’s disease. Molecular Brain 10, (2017). 45. 45.Vera, D., Eunsung, J. & Maral, M. M. The Role of Oxidative Stress in Parkinson’s Disease. Journal of Parkinson’s Disease 461-491 (2013).doi:10.3233/JPD-130230 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3233/JPD-130230&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24252804&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000328332100002&link_type=ISI) 46. 46.Guo, J., Zhao, X., Li, Y., Li, G. & Liu, X. Damage to dopaminergic neurons by oxidative stress in Parkinson’s disease (Review). International Journal of Molecular Medicine (2018).doi:10.3892/ijmm.2018.3406 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3892/ijmm.2018.3406&link_type=DOI) 47. 47.Shivade, C. et al. A review of approaches to identifying patient phenotype cohorts using electronic health records. J Am Med Inform Assoc 21, 221–230 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1136/amiajnl-2013-001935&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24201027&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) 48. 48.Hersh, W. R. et al. Caveats for the Use of Operational Electronic Health Record Data in Comparative Effectiveness Research. Med Care 51, S30-S37 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/MLR.0b013e31829b1dbd&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23774517&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F14%2F2020.08.11.20171447.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000337921600005&link_type=ISI) 49. 49.Baumfeld Andre, E., Reynolds, R., Caubel, P., Azoulay, L. & Dreyer, N. A. Trial designs using real-world data: The changing landscape of the regulatory approval process. Pharmacoepidemiol Drug Saf pds.4932 (2019).doi:10.1002/pds.4932 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/pds.4932&link_type=DOI)