Elsevier

Drug Discovery Today

Volume 25, Issue 2, February 2020, Pages 414-421
Drug Discovery Today

Review
Informatics
Key indicators of phase transition for clinical trials through machine learning

https://doi.org/10.1016/j.drudis.2019.12.014Get rights and content

Highlights

  • Protocol features across therapeutic areas and trial phases linked to phase success.

  • Supervised machine learning predicts drug transition across clinical trial phases.

  • Clinical trials phase transitions predicted with an average accuracy of 80%.

  • Natural language algorithms to study eligibility criteria role on phase success.

  • Updated estimates for phase success and likelihood of approval.

A significant number of drugs fail during the clinical testing stage. To understand the attrition of drugs through the regulatory process, here we review and advance machine-learning (ML) and natural language-processing algorithms to investigate the importance of factors in clinical trials that are linked with failure in Phases II and III. We find that clinical trial phase transitions can be predicted with an average accuracy of 80%. Identifying these trials provides information to sponsors facing difficult decisions about whether these higher risk trials should be modified or halted. We also find common protocol characteristics across therapeutic areas that are linked to phase success, including the number of endpoints and the complexity of the eligibility criteria.

Introduction

High attrition in the drug development pipeline is well documented in the literature 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17. The most critical failures happen at later stages; ∼60–70% of Phase II trials and 30–40% of Phase III trials are unsuccessful; meaning that 60–70% of the drugs that make it to Phase II will not transition to Phase III. Similarly, of the drugs that make it to Phase III, 30–40% will not transition to New Drug Application (NDA)/Biologics License Application (BLA) submission 2, 4, 8. Overall, ∼11–19% of drugs in testing will make it from Phase I to final regulatory approval for a lead indication 2, 4, 8.

The biomedical research enterprise has made substantial technological and scientific advancements over the past few decades, yet, overall it is spending more on research and development (R&D) without seeing a comparable increase in novel therapeutics, as assessed by new molecular entities (NMEs) and new biologic entities (NBEs) or by life expectancy gains 3, 5, 6, 18, 19. Given the significant human and financial costs associated with bringing a drug to market 3, 8, 10, with estimates varying between US$600 million and US$2.8 billion, most stakeholders agree that the attrition rate is unsustainably high 2, 3, 4, 8, 18, 20, 21.

In an attempt to improve productivity and reduce R&D costs, numerous researchers and other stakeholders have tried to untangle the reasons for attrition by examining various molecular, strategic, financial, and regulatory factors. At a very basic level, researchers have found that most drugs fail because of lack of efficacy followed by safety issues 9, 11, 22; however, a large body of literature provides more detail and insight into the factors that potentially affect trial performance. Several studies consider the characteristics of the drug itself. For example, NMEs are more likely to fail compared with other drugs 8, 20, and biologics (or large molecules) have a higher success rate than small molecules in clinical trials 4, 6. Some research questions whether the target-based approach and/or drug-likeness approaches utilized earlier in the drug discovery process have led to more drugs entering the pipeline that later fail because of safety issues 6, 23.

Additional research looks beyond the drug itself and points to higher failure rates in trials for specific diseases and disorders, including chronic diseases such as Alzheimer’s and diabetes, whereas others point to higher failure across entire therapeutic areas, such as oncology, infectious disease, and central nervous system disorders 4, 5, 8, 11, 20. Further studies indicate that strategic factors might also have a role. For example, self-originated drugs fail at higher rates than those that are licensed-in 2, 4. Also, research shows that pharmaceutical companies have increasingly invested in less commercially crowded areas, such as immunomodulatory drugs, as well as in drugs with novel mechanisms of action, where there might be not only higher expected revenues, but also higher failure rates 2, 5, 6.

Some research indicates that trial protocol complexity, longer cycle times, and increased investigative site work burden also contribute to poor trial performance and failure 22, 24, 25. The type of company making strategic R&D decisions also appears to matter, because smaller companies are more likely to experience failure compared with larger companies; this is true whether company size is measured by pharmaceutical sales or by R&D budget 2, 8, 24. Finally, some research posits that barriers erected by regulatory agencies contribute to trial failure, although drugs with newer special designations, such as orphan status, might in fact improve success rates 6, 8.

In addition to this complex body of literature, recent research has attempted to use larger data sets 16, 26 and ML methodologies 24, 27, 28, 29, 30, 31, 32, 33 to analyze failures in greater detail and consider additional factors that might be contributing to pipeline performance. Some studies that have developed ML models 24, 27 present similar analyses to those presented here. However, they have focused on answering different questions. For instance, DiMasi et al. [24] developed a model to predict regulatory approval after Phase II for oncology trials using 98 oncology drugs from the top 50 pharmaceutical companies. They used logistic regression and ML methods [Random Forests (RF), particularly for variable selection purposes] to predict and identify the most important individual factors associated with regulatory approval. A similar study was done by Lo et al. [27], who, similar to DiMasi et al., analyzed the data of completed (finished) trials to predict drug approvals (probability of success) and variable importance using drug-development and clinical-trial data from 2003 to 2015. The analysis was performed by developing a set of distinct ML models, including RF, neural networks, and decisions trees, among others. Indeed, they found that k-nearest neighbor (k-NN) imputation with RF provided the best predictive performance. They also identified trial outcomes, trial status, and trial accrual rates as significant factors for prediction.

Given the extensive work using ML methods to investigate factors impacting pipeline performance, we similarly investigate factors before a trial begins that can explain trial outcome (defined as success or failure) and phase transition. Two of the most comprehensive data sets of clinical trial designs and drug characteristics available (the Aggregate Analysis of ClinicalTrials.gov, AACT [34], based on https://clinicaltrials.gov/ and Biomedtracker [26] data sets) were used for this case study. A series of supervised ML (SML) models, which are informed with clinical trial data, are used to forecast whether a drug will successfully transition from the beginning of one phase to next (e.g., if a drug entering Phase II will transition to III and Phase III to NDA/BLA submission or regulatory approval), and to identify which factors, if any, were associated with the chances of a drug advancing toward regulatory approval. A thorough literature search is then presented, which seeks to provide a better understanding of the importance of such factors.

The case study evaluates the capability of ML models to predict phase success and failure. Dozens of characteristics from individual clinical trials are used, including the design of the trial protocol (e.g., number of endpoints or number of study arms), operational characteristics (e.g., number of countries where trial is being conducted or stakeholder type serving as lead sponsor), and other potential covariates, such as the target enrollment of each trial (see S1 in the supplemental material online for the complete list of covariates). Several characteristics not utilized by previous studies are explored, including eligibility criteria complexity. Eligibility criteria and a significant number of other clinical trial characteristics exist only as unstructured and free text data in the available data sets. Therefore, to analyze the importance of eligibility criteria in trial success, a natural language processing (NLP) algorithm (based on pattern matching and replacement and character vector count) was used to generate a metric for eligibility criteria complexity (see Methods and supplemental material online for developmental details). Although NLP techniques have been widely used in systems biology [35], ontology for drug discovery [36] and in biomedical context 37, 38, 39, such as in drug name recognition in biomedical text [40], this case study is the first to use NLP to create and explore a metric for eligibility criteria complexity and to recognize that complexity as an important factor in trial outcome.

The SML models used in this study can identify the outcome of a trial phase with an average accuracy of 80% when analyzing specific therapeutic areas. Access to a model with this level of predictive accuracy offers insight in two distinct capacities. From a trialist’s perspective, the model can be used as an iterative tool offering counsel in the protocol design and operational characteristics that should be pursued, and those that should be avoided or altered. From a commercial perspective, the predictive capabilities of the model provide more information for decision-making when assessing portfolios and allocating resources.

Section snippets

Data set description: Likelihood of Approval and phase success

The SML models were built using clinical trial data aggregated from two distinct sources, ClinicalTrials.gov (https://clinicaltrials.gov/) 34, 41 and Biomedtracker 8, 26. ClinicalTrials.gov is a database of privately and publicly funded research studies conducted in the USA and >203 other countries. The current version contains information from >268 000 studies and is maintained by the National Library of Medicine at the National Institutes of Health as a publicly available database. Each

Supervised machine-learning model: Random Forest

We tested the capabilities of RF models to predict trial outcomes. Several phase-therapeutic area combinations, using Phase II and III and the therapeutic areas oncology, neurology, and cardiology, were selected for the study. The RF models were developed for each therapeutic area because there were noticeable differences in phase success across the therapeutic areas, which added confounded noise to the training process of the models. The confounding effect of therapeutic areas was more

Discussion and review

Here, we have reviewed the use of ML methods for understanding the factors associated with success and failure of clinical trials. We presented a case study with new estimates for phase success and the likelihood of approval and compared them with the existing literature. The estimates presented here contribute to the existing literature in that they are obtained from a larger set of trials that belong to a longer time frame, larger number of drugs, and larger set of companies than most

Concluding remarks

ML techniques offer the opportunity for all stakeholders to manage risk and cut attrition, which would in turn begin to address the significant human and financial costs associated with bringing a drug to market. An example of this, and a future research line that authors are exploring, is the usage of SML to provide adverse event (serious and nonserious) risk scores. Such scores can be based on either a classification task (whether a treatment arm is riskier than a placebo arm) or on a

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

Authors of this research received funding from Bloomberg Philanthropies, Argosy Foundation, and Blakely Investments. The research was in partnership with MIT Collaborative Initiatives.

References (53)

  • I. Harrow

    Ontology mapping for semantically enabled applications

    Drug Discov. Today

    (2019)
  • R. Mcentire

    Application of an automated natural language processing (NLP) workflow to enable federated search of external biomedical content in drug discovery and development

    Drug Discov. Today

    (2016)
  • R.A.A. Erhardt

    Status of text-mining techniques applied to biomedical text

    Drug Discov. Today

    (2006)
  • R. Rodriguez-Esteban et al.

    Text mining patents for biomedical knowledge

    Drug Discov. Today

    (2016)
  • L. He

    Drug name recognition in biomedical texts: a machine-learning-based method

    Drug Discov. Today

    (2014)
  • J.W. Andersen

    Factors associated with early study discontinuation in AACTG studies, DACS 200

    Contemp. Clin. Trials

    (2007)
  • I. Kola et al.

    Can the pharmaceutical industry reduce attrition rates?

    Nat. Rev. Drug Discov.

    (2004)
  • S.M. Paul

    How to improve RD productivity: the pharmaceutical industry’s grand challenge

    Nat. Rev. Drug Discov.

    (2010)
  • J.A. Dimasi

    Trends in risks associated with new drug development: success rates for investigational drugs

    Clin. Pharmacol. Ther.

    (2010)
  • F. Pammolli

    The productivity crisis in pharmaceutical R&D

    Nat. Rev. Drug Discov.

    (2011)
  • J.W. Scannell

    Diagnosing the decline in pharmaceutical R&D efficiency

    Nat. Rev. Drug Discov.

    (2012)
  • M. Ringel

    Does size matter in R&D productivity? If not, what does?

    Nat. Rev. Drug Discov.

    (2013)
  • M. Hay

    Clinical development success rates for investigational drugs

    Nat. Biotechnol.

    (2014)
  • M.J. Waring

    An analysis of the attrition of drug candidates from four major pharmaceutical companies

    Nat. Rev. Drug Discov.

    (2015)
  • R.K. Harrison

    Phase II and phase III failures: 2013-2015

    Nat. Rev. Drug Discov.

    (2016)
  • J.R. Empfield et al.

    Lessons learned from candidate drug attrition

    IDrugs

    (2010)
  • Cited by (30)

    • Artificial intelligence in vaccine development: Significance and challenges ahead

      2023, A Handbook of Artificial Intelligence in Drug Delivery
    • Artificial intelligence-based decision support model for new drug development planning

      2022, Expert Systems with Applications
      Citation Excerpt :

      They asserted that the results obtained from this study offer useful insights into the outcome of drug development because many of these are variables that had not been considered in prior studies. Feijoo et al. (Feijoo, Palopoli, Bernstein, Siddiqui, & Albright, 2020) also developed a model to predict the phase transition and LoA of clinical trials using supervised machine learning (SML) and Natural Language Processing (NLP) algorithms. The authors used the NLP algorithm to extract an indicator measuring the complexity of eligibility criteria from the text data of ClinicalTrials.gov., which had not been explored in previous studies.

    • Cooperation in R&D in the pharmaceutical industry: Technological and clinical trial networks in oncology

      2022, Technological Forecasting and Social Change
      Citation Excerpt :

      However, not all patents will become technological breakthroughs, and a CT is one of the necessary steps for new drugs to reach the market (Pereira et al., 2019). A CT evaluates the safety and efficacy of a new drug in humans in four distinct phases1 and upon reaching phase 3, approximately 65% will be approved by regulatory bodies (Feijoo et al., 2020). Recent investigations using patents and CTs aimed at CAR-T have identified an intense collaboration between organizations, and despite clinical studies with results leading to complete remissions, there are many challenges to be investigated, such as making them more accessible to the population (Picanço-Castro et al., 2020a).

    View all citing articles on Scopus
    View full text