Original Paper
Abstract
Background: Clinical trials are vital for developing new therapies but can also delay drug development. Efficient trial data management, optimized trial protocol, and accurate patient identification are critical for reducing trial timelines. Natural language processing (NLP) has the potential to achieve these objectives.
Objective: This study aims to assess the feasibility of using data-driven approaches to optimize clinical trial protocol design and identify eligible patients. This involves creating a comprehensive eligibility criteria knowledge base integrated within electronic health records using deep learning–based NLP techniques.
Methods: We obtained data of 3281 industry-sponsored phase 2 or 3 interventional clinical trials recruiting patients with non–small cell lung cancer, prostate cancer, breast cancer, multiple myeloma, ulcerative colitis, and Crohn disease from ClinicalTrials.gov, spanning the period between 2013 and 2020. A customized bidirectional long short-term memory– and conditional random field–based NLP pipeline was used to extract all eligibility criteria attributes and convert hypernym concepts into computable hyponyms along with their corresponding values. To illustrate the simulation of clinical trial design for optimization purposes, we selected a subset of patients with non–small cell lung cancer (n=2775), curated from the Mount Sinai Health System, as a pilot study.
Results: We manually annotated the clinical trial eligibility corpus (485/3281, 14.78% trials) and constructed an eligibility criteria–specific ontology. Our customized NLP pipeline, developed based on the eligibility criteria–specific ontology that we created through manual annotation, achieved high precision (0.91, range 0.67-1.00) and recall (0.79, range 0.50-1) scores, as well as a high F1-score (0.83, range 0.67-1), enabling the efficient extraction of granular criteria entities and relevant attributes from 3281 clinical trials. A standardized eligibility criteria knowledge base, compatible with electronic health records, was developed by transforming hypernym concepts into machine-interpretable hyponyms along with their corresponding values. In addition, an interface prototype demonstrated the practicality of leveraging real-world data for optimizing clinical trial protocols and identifying eligible patients.
Conclusions: Our customized NLP pipeline successfully generated a standardized eligibility criteria knowledge base by transforming hypernym criteria into machine-readable hyponyms along with their corresponding values. A prototype interface integrating real-world patient information allows us to assess the impact of each eligibility criterion on the number of patients eligible for the trial. Leveraging NLP and real-world data in a data-driven approach holds promise for streamlining the overall clinical trial process, optimizing processes, and improving efficiency in patient identification.
doi:10.2196/50800
Keywords
Introduction
Background
Clinical trials are crucial for developing new therapies, but they require significant resources and can introduce delays in drug development, leading to increased costs [
, ]. Complex and restrictive eligibility criteria hinder patient enrollment, impacting target goals, timelines, and ultimately patient well-being [ - ]. This issue is particularly notable in cancer trials with poor recruitment and high failure rates [ - ] because >80% of the trials fail to meet their initial target accruals and timelines [ , ]. In addition, overly restrictive eligibility criteria limit the representation of the broader patient population, reducing real-world applicability and treatment impact [ - ]. Nonetheless, the practice of trials reusing complicated eligibility criteria without a clear rationale is a common one [ ], despite the minimal impact on trial outcomes [ ]. Liu et al [ ] demonstrated that broadening eligibility criteria using a data-driven approach can benefit initially excluded patients. A comprehensive and standardized eligibility criteria knowledge base that is compatible with real-world data can address these challenges. Such a knowledge base optimizes trial protocol design, improves patient enrollment, enhances the reliability and applicability of evidence synthesis, and fosters the efficient development of new therapies. Furthermore, it enables opportunities such as generating synthetic control arms (SCAs) for single-arm clinical trials using electronic health records (EHRs) [ - ].The importance of semantically representing eligibility criteria interoperable with EHRs has been highlighted in multiple studies [
- ]. Converting free-text eligibility criteria to computable formats poses challenges, addressed by a range of natural language processing (NLP) techniques and transformer models [ - ]. An NLP interface, Criteria2Query, enables computable queries for eligible cohort identification using EHRs [ ]. This tool supports clinical trial knowledge base development, enhancing EHR interoperability and scalability for efficient eligibility criteria knowledge engineering [ ]. Manually annotated data sets such as “Chia, a large annotated corpus of clinical trial eligibility criteria” [ ] and the “Leaf Clinical Trials corpus, the largest and most comprehensive human-annotated corpus of publicly available clinical trials eligibility criteria” [ ] have significantly enhanced NLP model training and the development of effective query structures. Despite significant progress in bridging the gap between eligibility criteria and EHRs, limitations persist in accurately representing the granularities of eligibility criteria and real-time eligible patient number checks [ , , ]. Using varying hierarchical levels of medical concepts, whether as hypernyms or hyponyms, presents one of the challenges when aligning eligibility criteria with EHRs; for instance, numerous trial eligibility criteria use hypernyms, which encompass a group of related medical concepts, such as cardiovascular disease. Conversely, the patient problem list within the EHR specifies particular medical conditions or diseases (hyponyms), such as myocardial infarction. Establishing a standardized eligibility criteria knowledge base by transforming ambiguous hypernym concepts into computable hyponyms can enhance optimizing trial protocol design and identifying eligible patients through seamless integration with EHR data.Objectives
In this study, we aim to create a standardized eligibility criteria knowledge base that seamlessly integrates with EHRs. By using deep learning–based NLP techniques, hypernym concepts in eligibility criteria will be converted to their EHR-compatible hyponyms along with their corresponding values. In addition, the prototype user interface will be developed as a pilot study, enabling the data-driven optimization of clinical trial protocols and the identification of eligible patients through the integration of the eligibility criteria knowledge base and EHRs.
Methods
Data Set
We obtained the data from ClinicalTrials.gov, specifically industry-sponsored phase 2 or 3 interventional clinical trials initiated between January 2013 and May 2020. A total of 3281 trials were identified: 817 (24.9%) for non–small cell lung cancer (NSCLC), 649 (19.78%) for prostate cancer (PCa), 1057 (32.22%) for breast cancer (BCa), 447 (13.62%) for multiple myeloma (MM), 160 (4.88%) for ulcerative colitis (UC), and 151 (4.6%) for Crohn disease (CD).
For the development of the prototype interface, we selected a subgroup of patients (n=2775) diagnosed with NSCLC from a previously curated cohort of patients with lung cancer. This cohort was established using the data from Mount Sinai-Sema4 Health System data [
], and patient information was deidentified for the purposes of this study.Deep Learning–Based NLP Pipeline Development
Our NLP pipeline consists of 3 modules: ontology construction and manual annotation, model training and pipeline evaluation, and application.
Ontology Construction and Manual Annotation
To construct our ontology, we randomly selected 425 eligibility criteria from diverse cancer trials and manually analyzed entities and relations. This manual analysis focused on identifying entities and their relationships. Entities were subsequently categorized into primary and modifier groups, with detailed examples provided in
and . The primary groups included demographic, diagnosis, biomarker, disease status, prior therapy, comorbidity, laboratory test, vital, procedure, and other medication, while the modifier groups included value, condition, evidence, lines of therapy, negation, exception, grade, dose, and temporal. Any entities that did not fall into the primary groups were classified as other observation. Furthermore, we defined relations between the entities. The commonly detected relationships between the primary and modifier groups were (1) has_value_limit between demographic (age) or vital lorlaboratory test and value, (2) has temporal limit between comorbidity or other medication or procedure and temporal, (3) has_negation between observation or biomarker or prior therapy and negation, and (4) has_exception between comorbidities or biomarker or diagnosis and exception. Other relationships included has_dose limit, has_line of therapy limit, has_grade_limit, has_condition, and need_evidence. The applicability of the ontology was tested on 60 UC and CD trials. Next, we manually annotated 246 eligibility criteria from NSCLC trials and performed model training using Clinical Language Annotation, Modeling, and Processing, which is an NLP toolkit [ ].Model Training and Pipeline Evaluation
A multilayer deep learning architecture was implemented for NLP modeling. The first step involved transforming the text into sequential vectors of characterization during the embedding process. These vectors were subsequently input into a bidirectional long short-term memory network, which is an artificial neural network designed for text classification. The bidirectional long short-term memory network was used to recognize patterns in both forward and backward directions [
]. The identified patterns were then passed to the next layer, which used a conditional random field model to compute the prediction probability [ ]. The NLP model was trained using annotated criteria, with 80% of the manually annotated gold standard data allocated for training. Model performance was evaluated on a separate validation set (20%) using precision, recall, and F1-score values:Precision = TP / (TP + FP) (1)
Recall = TP / (TP + FN) (2)
F1-score = 2 × (Precision × Recall) / (Precision + Recall) (3)
In equations 1 and 2, TP stands for true positives, FP for false positives, and FN for false negatives.
The manual annotation and training processes were iteratively performed with additional manually annotated notes until the model achieved a F1-score of >0.8 in the test set (
). To tailor the pipeline for specific cancer types, a preannotation method using the NSCLC pipeline was implemented for PCa, BCa, and MM for common eligibility criteria such as laboratory test values and comorbidities. Specific eligibility criteria such as biomarkers and treatments were manually annotated for each cancer type: PCa with 124 trials, BCa with 73 trials, and MM with 60 trials.Application
The fully trained named entity recognition and relation models were integrated and applied to annotate the remaining eligibility criteria for the 4 types of cancer studied (BCa, MM, NSCLC, and PCa). The output data included sentences, tokens, parts of speech, entities, negations, and relations.
Construction of Standardized Eligibility Criteria Knowledge Base Table
The standardized knowledge base was constructed in an EntityGroup-AttributeName-Value format, involving 2 key steps: attribute normalization and transforming hypernyms to hyponyms with corresponding values.
Attribute Normalization
To normalize attributes, we used a 3-step approach. First, we assigned a Unified Medical Language System concept unique identifier to map synonyms of an entity, such as estrogen receptor-positive, ER-positive, and ER+ to the Unified Medical Language System concept unique identifier C0279754. Second, we developed a set of rules (
) to map abbreviations (eg, CrCl to creatinine clearance) and different phrases with the same meaning (eg, ≥1.5x ULN [where ULN stands for upper limit of normal], greater than or equal to 1.5x ULN, and ≥1.5x upper limit of normal) back to their original text. Finally, 2 domain experts manually curated unnormalized entities.Rule and attributes from eligibility criteria | Normalized attributes | ||
Rules for mapping synonyms | |||
ASTa, SGOTb, aspartate aminotransferase, serum AST | AST | ||
ALTc, SGPTd, alanine aminotransferase, serum ALT | ALT | ||
Total bilirubin, serum bilirubin, total bilirubin level, bilirubin level | Total bilirubin | ||
Hgbe, hemoglobin | Hgb | ||
HbA1cf, hemoglobinA1c | HbA1c | ||
serum creatinine, creatinine, creatinine levels, creatinine level | Serum creatinine | ||
ANCg, absolute neutrophil count, absolute neutrophil counts, neutrophil count, neutrophil counts, absolute neutrophil | ANC | ||
WBCh, white blood cells, white blood cell, WBC count, white blood cell count, white blood count, leucocytes | WBC | ||
platelets, platelet, platelet count, platelet counts | Platelets | ||
CrCli, creatine clearance | CrCl | ||
ALPj, alkaline phosphatase | ALP | ||
ULNk, upper limit of normal | ULN | ||
LLNl, lower limit of normal | LLN | ||
Rules related to unit and temporal modifier | |||
less than or equal to, ≤ | ≤ | ||
greater than or equal to, ≥ | ≥ | ||
greater than, > | > | ||
less than, < | < | ||
within 4 weeks, within 28 days | within 4 weeks | ||
within 2 weeks, within 14 days | within 2 weeks | ||
within 3 weeks, within 21 days | within 3 weeks | ||
last 6 months, past 6 months, within 6 months, within six months | within 6 months | ||
last 3 months, past 3 months, within 3 months, within three months | within 3 months | ||
within 2 years, last 2 years, past 2 years | within 2 years | ||
within 3 years, last 3 years, past 3 years | within 3 years | ||
within 5 years, last 5 years, past 5 years | within 5 years | ||
109/L, 109/L, 103/uL, 103/microliter, 1000/uL, 1000/microliter, K/microliter, 103/mm3 | 103/uL | ||
Other miscellaneous rules | |||
Case insensitive | —m | ||
Remove spaces | — |
aAST: aspartate aminotransferase.
bSGOT: serum glutamic oxaloacetic transaminase.
cALT: alanine transaminase.
dSGPT: serum glutamic pyruvic transaminase.
eHgb: hemoglobin.
fHbA1c: glycated hemoglobin.
gANC: absolute neutrophil count.
hWBC: white blood cell.
iCrCl: creatinine clearance.
jALP: alkaline phosphatase.
kULN: upper limit of normal.
lLLN: lower limit of normal.
mNot applicable.
Transforming Hypernyms to Hyponyms Along With Corresponding Values
To formalize hypernyms, identified in primary groups such as laboratory test, comorbidity, biomarker, prior therapy, and other medication, we used the following approaches: (1) for adequate organ functionlaboratory test values, we determined prevalent laboratory test values by analyzing the unique laboratory test values for each test across the trials of the same cancer type that defined the normal organ function; and (2) for comorbidity, biomarker, prior therapy, and other medication hypernyms, we collected all example hyponyms described across the trials of the same cancer type.
Creation of a Prototype Interface for Enhancing Trial Protocol Design Optimization
We developed a prototype interface using the R programming language (R Foundation for Statistical Computing) and the Shiny package to enhance trial protocol design optimization. The interface allows users to simulate the number of eligible patients based on specific criteria, including a combination of criteria such as histology, stages, laboratory test values, performance scores, prior line of therapy, and comorbidities. For this pilot study, a subset of patients with NSCLC (n=2775) was selected and deidentified. To ensure consistency and accuracy, we standardized the sample entities found in both the eligibility criteria knowledge base and EHRs using concept codes such as the International Classification of Diseases; Logical Observation Identifiers, Names, and Codes (LOINC); and normalized medical prescription codes. In addition, we converted the patients’ absolute laboratory test values to either the upper limit of normal (ULN) or the lower limit of normal based on the provided normal ranges for each specific test.
and and present some examples of normalized concepts and their codes.Laboratory test | LOINCa code | Normal range |
ALTb (SGPTc; U/L) | 1742-6 | 7-56 |
ASTd (SGOTe; U/L) | 1920-8 | 10-40 |
Total bilirubin in serum (mg/dL) | 1975-2 | 0.1-1.2 |
Direct (conjugated) bilirubin in serum (mg/dL) | 1968-7 | <0.3 |
Serum creatinine (mg/dL) | 2160-0 | 0.6-1.2 (male), 0.5-1.1 (female) |
CrClf (mL/min) | 2164-2 | 97-137 (male), 88-128 (female) |
ANCg (cells/µL) | 26499-4 | >90 mL/min/1.73 m2 |
Platelets (cells/µL) | 777-3 | 150,000-450,000 |
Hemoglobin (g/dL) | 718-7 | 12-18 |
aLOINC: Logical Observation Identifiers, Names, and Codes.
bALT: alanine transaminase.
cSGPT: serum glutamic pyruvic transaminase.
dAST: aspartate aminotransferase.
eSGOT: serum glutamic oxaloacetic transaminase.
fCrCl: creatinine clearance.
gANC: absolute neutrophil count.
Disease | ICD-10 codes | ICD-9 codes |
Congestive heart failure | I50.2, I50.3, I50.4 | 428.[2-4][0-3] |
Unstable angina | I20.0 | 411.1 |
Acute myocardial infarction | I21 | 410.9[0-2] |
Arrythmia | I49 | 429.9 |
Torsade de pointes | I45.81 | 426.82 |
Long QT syndrome | I45.81 | 426.82 |
Atrial fibrillation and flutter | I48 | 427.3[1-2] |
Symptomatic bradycardia | R00.1 | 427.89 |
Uncontrolled hypertension | I10 | 401.[09] |
Heart aneurysm | I25.3 | 414.1[09] |
Coronary heart disease | I25.1 | 414.01 |
Cardiomyopathy | I42.9 | 425.[49] |
Vasculitis, or angiitis | I77.6 | 447.6 |
Pericardial effusion | I31.3 | 423.9 |
Peripheral vascular disease | I73.9 | 443.9 |
Drug and RxNORM code
- Bortezomib: 356733
- Carfilzomib: 1302966
- Ixazomib: 1723735
- Lenalidomide: 342369
- Pomalidomide: 1369713
The interface uses a rule-based algorithm to match patients’ EHR data with the criteria. The comprehensive rules for matching EHR data with criteria have been described in our previous studies [
]; for instance, we defined the following rules to map each laboratory test in EHRs to 1 corresponding LOINC code:- Mapping the laboratory test in the LOINC dictionary to the laboratory test in the EHR, based on the popularity rank available in the LOINC dictionary
- Mapping the laboratory test for serum or plasma samples in the LOINC dictionary to the laboratory test in the EHR when the popularity rank is not available in the LOINC dictionary
- If one-to-one mapping is not feasible using rule 1 and rule 2, the test unit (eg, gram is preferred ovemolar) is considered to facilitate the mapping
- When one-to-one mapping is not attainable using rule 1, rule 2, and rule 3, preference is given to the laboratory test that lacks information about the method for mapping
We associated medication classes with their respective medications; for instance, we extended the annotation “post-menopausal not older than 60 years and taking LHRH [luteinizing hormone–releasing hormone] agonist” to include “post-menopausal not older than 60 years and taking goserelin, leuprolide, or other LHRH agonists.” To achieve this, we used both our in-house knowledge bases and standard resources, such as the National Comprehensive Cancer Network’s Clinical Practice Guidelines in Oncology.
Users can specify different criteria and combinations, such as different laboratory test values with specific conditions such as no brain metastasis to determine the number of qualified patients. The algorithm matches each patient’s EHR data with the selected criteria and calculates the number of matched patients for each criterion. The performance of the interface was evaluated by comparing it to the manual patient selection process conducted by experienced clinical domain experts.
Ethical Considerations
This study was confirmed and approved by the Program for the Protection of Human Subjects at the Mount Sinai School of Medicine (IRB-17-01245)
Results
Development of Eligibility Criteria–Specific Ontology
Our analysis of cancer clinical trials revealed that hormone therapy was the most frequently applied modality (1470/2970, 47.37%), primarily in BCa and PCa trials, followed by targeted therapy (753/2970, 25.35%) and immunotherapy (691/2970, 23.26%). Chemotherapy alone was used in 3.8% (113/2970) of the clinical trials. We developed an eligibility criteria ontology applicable to all cancer trials by manually analyzing 425 eligibility criteria (
). Entities were categorized into 10 primary groups (inside the blue dotted box) and 9 modifier groups based on semantic types and relations. Entities falling outside the blue dotted box were classified as other observation. The inclusion criteria mainly involved entities in the demographic, diagnosis, laboratory test, and vital groups, while the exclusion criteria commonly included entities in the comorbidity, procedure, and other medication groups. Entities in the biomarker, prior therapy, and disease status groups appeared in both the inclusion and exclusion criteria. Relationships originated from the primary groups and terminated in the modifier groups, except for the has outcome relationship, which started and ended in the primary group ( ). To assess the applicability of the cancer eligibility criteria ontology in a different disease context, we conducted a manual analysis of 60 trials related to UC and CD. For reference, the computable formats of the manually annotated 485 trials can be found in - .NLP Pipeline Quality Metrics
To evaluate the quality of our NLP pipeline, we computed precision, recall, and F1-score measures. For the primary group entities, the average scores were 0.91 (precision), 0.79 (recall), and 0.83 (F1-score).
presents the range of precision, recall, and F1-score values of 17 primary group entities.Primary group and attribute group::name | Precision | Recall | F1-score | ||||
Demographic | |||||||
Demographic::age | 1.000 | 0.923 | 0.960 | ||||
Demographic::gender | 1.000 | 0.870 | 0.931 | ||||
Diagnosis | |||||||
Diagnosis::histology | 1.000 | 1.000 | 1.000 | ||||
Diagnosis::stage | 0.667 | 1.000 | 0.800 | ||||
Biomarker | |||||||
Biomarker::biomarker | 1.000 | 0.800 | 0.889 | ||||
Disease status | |||||||
Clinical status::disease status | 0.737 | 0.684 | 0.709 | ||||
Prior therapy | |||||||
Prior therapy::chemotherapy | 0.944 | 0.895 | 0.919 | ||||
Prior therapy::targeted therapy | 1.000 | 0.786 | 0.880 | ||||
Prior therapy::immunotherapy | 0.897 | 0.788 | 0.839 | ||||
Prior therapy::radiotherapy | 0.682 | 0.682 | 0.682 | ||||
Prior therapy::adjuvant therapy | 1.000 | 0.571 | 0.727 | ||||
Prior therapy::neoadjuvant therapy | 1.000 | 0.500 | 0.667 | ||||
Comorbidity | |||||||
Comorbidity::disease | 0.842 | 0.762 | 0.800 | ||||
Laboratory test | |||||||
Laboratory test::test | 0.871 | 0.818 | 0.844 | ||||
Vital | |||||||
Vital::vital | 1.000 | 1.000 | 1.000 | ||||
Procedure | |||||||
Procedure::procedure | 1.000 | 0.600 | 0.750 | ||||
Other medication | |||||||
Other medication::medication | 0.800 | 0.727 | 0.762 |
Eligibility Criteria Attribute Extraction and Classification
The integrated named entity recognition and relation model extracted 9090 NSCLC, 7427 PCa, 10,217 BCa, 6803 MM, 1565 CD, and 1586 UC entities along with their attribute relations. After normalization and manual curation processes, the eligibility criteria knowledge base for each disease type was established in the EntityGroup-AttributeName-Value format (
- ). The number of unique EntityGroup-AttributeName-Value combinations varied across disease types, with 494 from 817 NSCLC trials, 471 from 649 PCa trials, 525 from 1057 BCa trials, 389 from 447 MM trials, 231 from 160 UC trials, and 230 from 151 CD trials. Notably, UC and CD trials had a smaller number of unique EntityGroup-AttributeName-Value combinations compared to cancer trials, indicating the presence of more complicated eligibility criteria in cancer trials.and show the distribution of EntityGroup-AttributeName-Value combinations in each primary group from different diseases and provide examples. The laboratory test, prior therapy, and comorbidity groups exhibited a high number of EntityGroup-AttributeName-Value combinations, followed by the biomarker and other medication groups. Variations were observed between solid cancers and hematologic cancers, with higher numbers of EntityGroup-AttributeName-Value combinations in solid cancer types for prior therapy and biomarker, while laboratory test and comorbidity were comparable. The diagnosis group exhibited varying numbers of EntityGroup-AttributeName-Value combinations across all 4 cancer types (BCa, MM, NSCLC, and PCa). EntityGroup-AttributeName-Value in the biomarker, diagnosis, and prior therapy groups were specified per indication, while shared EntityGroup-AttributeName-Value were found in other primary groups.
Primary group | Number of attributes | Example attributes: group, name, value (with or without condition) | ||||
NSCLCa | BCab | PCac | MMd | |||
Demographic | 11 | 12 | 10 | 5 | Demographic, age, ≥18 y | |
Diagnosis | 18 | 31 | 17 | 4 | Stage, TNMe system, T2bf | |
Biomarker | 49 | 72 | 42 | 14 | Biomarker, HER2g mutation, L755Ph | |
Disease status | 11 | 11 | 13 | 9 | Disease status, relapsed, yes | |
Prior therapy | 114 | 108 | 93 | 50 | LOTi, prior LOT, ≥2 | |
Comorbidity | 105 | 96 | 97 | 108 | Cardiovascular disease, arrhythmia, yes (≤3 mo) | |
Laboratory test | 103 | 103 | 110 | 119 | Test, ASTj, ≤2.5x ULNk | |
Vital | 18 | 22 | 21 | 18 | Vital, ECOGl, ≥2 | |
Procedure | 6 | 6 | 6 | 8 | Procedure, organ transplantation, yes | |
Other medication | 45 | 45 | 45 | 46 | Other medication, use of anticoagulants, warfarin (<4 wk) |
aNSCLC: non–small cell lung cancer.
bBCa: breast cancer.
cPCa: prostate cancer.
dMM: multiple myeloma.
eTNM: tumor, nodes, metastasis.
fT2b: a moderately advanced tumor in terms of size and extent but not the most advanced stage; specific implications can vary based on the type of cancer being described.
gHER2: human epidermal growth factor receptor 2.
hL755P: a reference to a specific mutation in the HER2 gene, with “L” standing for leucine, “755” being the position of the amino acid in the protein, and “P” standing for proline.
iLOT: line of therapy.
jAST: aspartate aminotransferase.
kULN: upper limit of normal.
lECOG: Eastern Cooperative Oncology Group.
Transformation of Umbrella Terms Into Computable Attributes With Representative Values
Overview
The conversion of hypernym concepts into computable attributes along with their corresponding values was carried out.
provides some examples of converted attributes and their corresponding values for each hypernym. All lists can be found in - .Entity and subgroup entity in eligibility criteria and converted attribute | Corresponding values | |||
Adequate organ function | ||||
Normal hepatic function | ||||
ASTa | ≤2.5x ULNb | |||
ALTc | ≤2.5x ULN | |||
Total bilirubin | ≤1.5x ULN | |||
Normal renal function | ||||
Creatinine | ≤1.5x ULN | |||
Normal hematologic function | ||||
ANCd | ≥1500 cells/uL | |||
Platelets | ≥100,000 cells/uL | |||
Hemoglobin | ≥9 mg/dL | |||
Comorbidities | ||||
Second malignancy | ||||
All cancers | Yes, with exceptions | |||
Infectious disease | ||||
HIV | Yes | |||
HBVe | Yes | |||
HCVf | Yes | |||
TBg | Yes | |||
Cardiovascular disease | ||||
CHFh | Yes | |||
MIi | Yes | |||
Angina | Yes | |||
Arrhythmia | Yes | |||
Autoimmune disease | ||||
UCj | Yes | |||
CDk | Yes | |||
Systemic lupus erythematosus | Yes | |||
Rheumatoid arthritis | Yes | |||
Systemic sclerosis | Yes | |||
Graves disease | Yes | |||
Guillain-Barré syndrome | Yes | |||
Antiphospholipid syndrome | Yes | |||
Sjogren syndrome | Yes | |||
Biomarker | ||||
EGFRl mutation sensitive to TKIm | ||||
Exon 19 deletion | Yes | |||
Exon 21 L858R | Yes | |||
Exon 21 L861Q | Yes | |||
Exon 18 G719C | Yes | |||
Exon 18 G719X | Yes | |||
Amplification | Yes | |||
EGFR mutation resistant to TKI | ||||
Exon 20 T790M | Yes | |||
Exon 20 C797S | Yes | |||
Exon 20 S768I | Yes | |||
Exon 20 insertion | Yes | |||
Mismatch repair deficient | ||||
MSH2, MSH6, MLH1, PMS2, or EXO1 gene mutation | Yes | |||
MLH1 hypermethylation | Yes | |||
Prior therapy (targeted) | ||||
First-generation EGFR inhibitor | ||||
Gefitinib | Yes | |||
Erlotinib | Yes | |||
Vandetanib | Yes | |||
Second-generation EGFR inhibitor | ||||
Afatinib | Yes | |||
Dacomitinib | Yes | |||
Poziotinib | Yes | |||
Tesevatinib | Yes | |||
Third-generation EGFR inhibitor | ||||
Osimertinib | Yes | |||
Lazertinib | Yes | |||
Rociletinib | Yes | |||
Tarloxotinib | Yes | |||
Proteasome inhibitor | ||||
Bortezomib based | Yes | |||
Carfilzomib based | Yes | |||
Ixazomib based | Yes | |||
Oprozomib based | Yes | |||
Prior therapy (hormone) | ||||
First-generation antiandrogen | ||||
Bicalutamide | Yes | |||
Nilutamide | Yes | |||
Flutamide | Yes | |||
Second-generation antiandrogen | ||||
Abiraterone | Yes | |||
Enzalutamide | Yes | |||
Darolutamide | Yes | |||
Apalutamide | Yes | |||
Androgen deprivation therapy | ||||
Leuprolide | Yes | |||
Goserelin | Yes | |||
Degarelix | Yes | |||
5-α reducing agent | ||||
Finasteride | Yes | |||
Dutasteride | Yes | |||
Megestrol acetate | Yes | |||
Other medication | ||||
Current use of antibiotics | ||||
Rifabutin | Yes | |||
Clarithromycin | Yes | |||
Azithromycin | Yes | |||
Imipenem | Yes | |||
Current use of antiarrhythmic agents | ||||
Propafenone | Yes | |||
Procainamide | Yes |
aAST: aspartate aminotransferase.
bULN: upper limit of normal.
cALT: alanine transaminase.
dANC: absolute neutrophil count.
eHBV: hepatitis B virus.
fHCV: hepatitis C virus.
gTB: tuberculosis.
hCHF: congestive heart failure.
iMI: myocardial infarction.
jUC: ulcerative colitis.
kCD: Crohn disease.
lEGFR: epidermal growth factor receptor.
mTKI: tyrosine kinase inhibitor.
Adequate Organ Function
Adequate organ function criteria were defined using various laboratory tests. Normal ranges and eligible values for alanine transaminase (ALT)/aspartate aminotransferase (AST), total bilirubin, serum creatinine, creatinine clearance, absolute neutrophil count, platelets, and hemoglobin were determined. Representative values for adequate organ/hematologic function included ≤2.5x ULN for ALT/AST, ≤1.5x ULN for total bilirubin/serum creatinine, ≥1500 cells/uL for absolute neutrophil count, ≥100,000 cells/uL for platelets, and ≥9 ng/dL for hemoglobin.
A-3H display the laboratory test value range and trial counts for each value in BCa and NSCLC clinical trials. The trends observed are similar in both cancer types.Comorbidities
The presence of comorbidities is a common exclusion criterion in clinical trials; however, natural language descriptions of comorbidities, such as “uncontrollable cardiovascular diseases,” “pulmonary diseases,” and “autoimmune diseases,” can be ambiguous and need domain knowledge to interpret them. We analyzed the hypernyms and their corresponding hyponyms used in BCa trial eligibility criteria.
shows the collected hyponyms for each comorbidity class. The presence of second primary malignancies was excluded in almost all trials.Prior Therapy, Other Medication, and Biomarker
By combining all examples of each hypernym, we broke down these hypernyms into actual medication and mutation hyponyms; for instance, we collected procainamide or propafenone for current use of antiarrhythmic medication. Similarly, we collected epidermal growth factor receptor (EGFR) exon 20 T790M, T797S, S768I, or insertion for EGFR mutations resistant to EGFR inhibitors.
Development of a Prototype Interface for the Optimization of Protocol Design
Our study investigated the impact of various criteria on the number of eligible patients. We developed a prototype interface that uses real-world patient information. Using a subset of deidentified cohorts of patients with NSCLC (n=2775), we deployed an eligibility criteria knowledge base that we had constructed in the interface.
A displays the selected criteria list, B shows the corresponding patient number, and C illustrates the distribution of patient numbers in each group.Sequentially incorporating criteria such as nonsquamous histology and stages III and IV criteria, we identified 2166 (78.05%) and 426 (15.35%) eligible patients, respectively, from the total pool of 2775 patients with NSCLC. The inclusion of AST and ALT ≤2.5x ULN criteria yielded 363 (13.08%) eligible patients from the pool of 2775 patients. Limiting AST and ALT to ≤1.0x ULN resulted in a decreased number of eligible patients (315/2775, 11.35%;
D). In addition, we explored the influence of Eastern Cooperative Oncology Group (ECOG) performance status as an additional criterion. With histology, stage, and ALT/AST laboratory test values (<2.5x ULN) as fixed criteria, by introducing ECOG scores of 0 to 2 or 0 to 1, we identified 194 (6.99%) and 151 (5.44%) eligible patients, respectively, from the pool of 2775 patients ( E).Patient-matching performance was evaluated using precision, recall, and F1-score performance metrics across specific clinical attributes. The average F1-score, computed across 10 attributes from 8 domains (other primary malignancy, congestive heart failure, squamous NSCLC, organ/tissue transplantation, platelets, programmed death-1 antibody therapy, programmed cell death protein-1 or programmed cell death program-ligand 1 positive, stage groups, prior LOT [line of therapy], and ECOG), was 0.94 (range 0.82-1.00 [
]).Discussion
Principal Findings
The challenge of achieving a high success rate in clinical trials is an ongoing issue [
, ]. Our study demonstrates the feasibility of a data-driven approach to optimize trial protocols and efficiently identify eligible patients by constructing a comprehensive, EHR-interoperable eligibility criteria knowledge base and integrating EHR data. To accomplish this, we analyzed 3281 clinical trials using our customized deep learning NLP model. We extracted all entities with their attributes and converted the hypernym concepts used in eligibility criteria to EHR-compatible hyponyms along with their corresponding values. We also evaluated the feasibility of optimizing the trial protocol design on the interface we developed. This interface offers an efficient and effective approach for assessing the number of eligible patients across various combinations of eligibility criteria such as different laboratory test values as well as combinations that account for vital signs.We developed an eligibility criteria–specific ontology by manually scrutinizing 425 eligibility criteria to be used as a reference for manual annotation during NLP model training. Accurately identifying intricate semantic relationships among entities within eligibility criteria is crucial for constructing an appropriate ontology for precise information extraction, including temporal, arithmetic values, Boolean values, and negation modifiers [
]. Our customized NLP pipeline based on the eligibility criteria–specific ontology that we created enabled us to efficiently extract all pertinent attributes across different modalities and diseases, allowing for a more accurate definition of the trial population. To determine the applicability of our ontology generated using cancer clinical trials to other disease domains, we compared the concepts and relations in clinical trials of inflammatory bowel diseases. We observed very similar trends, suggesting that our eligibility criteria–specific ontology can be extended to other types of disease trials.Moreover, the corpus of 485 manually annotated and standardized trials in a computable format can be used in eligible patient identification in EHRs.
Liu et al [
] conducted a thorough analysis of 352,100 clinical trials across various disease domains and constructed a knowledge base of clinical trial eligibility criteria. Their comprehensive knowledge base and user-friendly interface showcased the potential of advanced NLP techniques in enhancing eligibility criteria analysis and retrieval. Fang et al [ ] also adopted a data-driven approach to optimizing clinical trial eligibility criteria in the context of Alzheimer disease and pancreatic cancer domains. Building upon these efforts, our study aimed to further narrow the gap between eligibility criteria and EHRs in multicancer domains, specifically in representing the granularities of eligibility criteria for identifying eligible patients and optimizing protocol designs. This was achieved by transforming hypernyms in the criteria into EHR-compatible hyponyms. We found that most of the primary groups include umbrella terms such as prior therapy (eg, proper prior therapy for actionable mutations) and biomarker (eg, EGFR inhibitor–resistant mutations). Our study also addressed the challenge of standardizing ambiguous clinical concepts in eligibility criteria for EHR interoperability and patient matching. To overcome this challenge, we converted hypernyms to the Entity-Attribute-Value format using prevailing values across different cancer types and modality therapies. We believe that our EHR-interoperable standardized eligibility criteria knowledge base and interface, integrating real-world EHR data, have the potential to improve the automatic screening system. This improvement has the potential to significantly reduce manual extraction efforts. Moreover, specific, computable criteria reduce ambiguity in patient identification and enable the inclusion of a broader range of patients who may qualify for the trial but could be excluded when using more general terms. This can increase patient trial enrollment, ultimately improving the overall success rate of trials. Notably, patients who were given the option to participate in a trial by their physicians demonstrated a significantly higher participation rate of 55% [ ] compared to the current average of 5% to 8% among patients with cancer [ , ]. The implementation of our hypernym/hyponym semantic terminology model can likewise improve the effectiveness of information retrieval from EHRs and other clinical databases in the context of real-world evidence studies.Certain criteria such as histology, stage, previous treatment, or biomarker are difficult to modify, while others such as vital signs or laboratory test values can be adjusted during the protocol design [
]. Our study revealed the impact of modifying laboratory test values while keeping other criteria constant, resulting in fluctuations in the number of eligible patients. Our findings, which demonstrate both the number of trials for different laboratory test value ranges and eligible patient numbers, offer insights for optimizing future protocol design and refining patient selection criteria. Seeking future collaboration with clinicians to conduct a direct comparison between the patient identification results by clinical domain experts and those generated by our prototype holds promise for a more comprehensive and informative evaluation of the prototype’s performance and its potential to enhance patient identification for clinical trials. Furthermore, a careful examination of the cases identified by the prototype can provide an understanding of the nature of false positives and false negatives. This will provide insights into how the prototype may differ in its patient identification results compared to manual extraction. Our eligibility criteria knowledge base can also be leveraged for generating SCAs using EHRs. SCAs, derived from real-world evidence, are regarded as substitutes for experimental control arms in trials [ - ]. The integration of SCAs into single-arm trial data or replacing traditional control arms with SCAs can alleviate the burden of target accrual in trials with low eligible patient numbers, such as rare disease or oncology trials with specific biomarkers. The Food and Drug Administration’s approval of the palbociclib inhibitor for male patients with metastatic BCa based on real-world evidence demonstrates the potential and relevance of SCAs in improving trial design and outcomes [ ].Limitations
Our study has several limitations to consider. First, we focused on a limited scope, analyzing only 4 different cancer types and exploring extendibility in the context of inflammatory bowel diseases. Future studies should encompass a wider range of cancer types and disease domains for a more comprehensive analysis. Second, while most attributes were well defined, some umbrella terms lacked clear examples in other cancer types, potentially affecting result accuracy. Further manual annotation using knowledge bases could enhance the precision of the attribute tables. Third, our data set may be biased because we solely included industry-sponsored trials, potentially limiting the generalizability of our findings. In addition, the NLP training and test data sets in this study can display similarities owing to the shared attributes among different cancer trials, which heightens concerns regarding potential overfitting. Fourth, we did not address entity logic, and establishing the logic between entities would enhance cohort definition accuracy. Fifth and last, our interface feasibility testing was limited to small cohorts of patients with NSCLC, and the generalizability of our findings to other populations or disease conditions may vary. Furthermore, we did not perform a quantitative evaluation of the accuracy of matched patients although domain experts checked whether the patient information matched the eligibility criteria manually. While our model serves as a valuable illustration of how NLP can contribute to the design of trials across different diseases, we fully acknowledge the indispensable role of clinicians and biomedical researchers in ensuring the integrity of trial criteria. Clinical trials vary in their objectives, encompassing assessments of treatment end points, effectiveness, and other specific goals. The process is far more nuanced than merely adjusting laboratory test values because such modifications can have a substantial impact on the pool of eligible patients. Therefore, a comprehensive approach, considering both the clinical and biomedical aspects, is imperative for robust trial design.
Conclusions
Our study using an EHR-executable eligibility criteria knowledge base and real-world patient information provides valuable insights into the influence of different criteria on the number of eligible patients during the protocol design. The findings highlight the potential of using a data-driven approach that incorporates NLP and EHRs in clinical research.
Data Availability
The patient data sets analyzed during this study are not publicly available due to patient privacy, security and the Health Insurance Portability and Accountability Act of 1996 (HIPAA) requirement. To enable a complete run of the code shared in this study, a minimum amount of desensitized sample data could be shared with the sharing agreement. Relevant requests should be addressed to ZL. The Prototype interface is available on the internet [
]; the R scripts are available on the internet [ ].Authors' Contributions
KL, ZL, and XW designed the study and wrote the manuscript. KL, YM, and XW reviewed the literature, clinical trial eligibility criteria, and patient notes and constructed the eligibility criteria–specific ontology. KL, ZL, MM, YM, and TW were responsible for the model training, the postprocessing procedure, and the data analysis. TJ, LA, EC, WO, GS, ES, and XW discussed the project and reviewed the manuscript.
Conflicts of Interest
KL, ZL, TJ, MM, YM, TW, LA, EC, GS, and XW are employees of Sema4. WO and ES are employees of the Icahn School of Medicine at Mount Sinai. WO holds stock in GeneDx. ES was employed by Sema4 and held stock in the company at the time the work in this paper was carried out.
Classification of attributes into primary entities and their subgroups.
PNG File , 79 KBClassification of attributes into modifier entities and their subgroups.
PNG File , 62 KBThe scheme for the natural language processing–assisted clinical trial analysis.
PNG File , 50 KBManually annotated non–small cell lung cancer trials.
XLSX File (Microsoft Excel File), 184 KBManually annotated breast cancer trials.
XLSX File (Microsoft Excel File), 133 KBManually annotated prostate cancer trials.
XLSX File (Microsoft Excel File), 209 KBManually annotated multiple myeloma trials.
XLSX File (Microsoft Excel File), 57 KBManually annotated ulcerative colitis and Crohn disease trials.
XLSX File (Microsoft Excel File), 136 KBNon–small cell lung cancer attributes.
XLSX File (Microsoft Excel File), 28 KBBreast cancer attributes.
XLSX File (Microsoft Excel File), 28 KBProstate cancer attributes.
XLSX File (Microsoft Excel File), 26 KBMultiple myeloma attributes.
XLSX File (Microsoft Excel File), 23 KBUlcerative colitis attributes.
XLSX File (Microsoft Excel File), 29 KBCrohn disease attributes.
XLSX File (Microsoft Excel File), 29 KBClinical trial counts with each unique laboratory test value defining normal organ function. (A-B) Alanine transaminase (ALT) and aspartate aminotransferase (AST): normal ranges from ≤1x upper limit of normal (ULN) to ≤3x ULN, with exceptions for liver diseases (eg, liver metastasis and Gilbert syndrome [GS]) allowing values of up to ≤5x ULN. (C) Total bilirubin: normal ranges from ≤1x ULN to ≤2.5x ULN, with exceptions for liver diseases (eg, liver metastasis and GS) allowing values of up to ≤3x ULN. (D) Serum creatinine: normal ranges from ≤1x ULN to ≤2.5x ULN. (E) Creatinine clearance: normal ranges from ≥30 to ≥60 mL/min. (F) Hemoglobin: normal ranges from ≥8.0 to ≥11.0 ng/dL. (G) Absolute neutrophil count (ANC): normal ranges from ≥750 to ≥1500 cells/uL. (H) Platelets: normal ranges from ≥50,000 to ≥100,000 cells/uL. BCa: breast cancer; NSCLC: non–small cell lung cancer.
PNG File , 122 KBScreenshots from a prototype interface. (A-B) The selected criteria list and the corresponding number of patients. (C) The distribution of patient numbers in each group. (D) Displayed are eligible patient numbers after sequentially incorporating criteria such as non-squamous histology and stage III and IV, with the further inclusion of aspartate aminotransferase (AST) and alanine transaminase (ALT) laboratory test values of either ≤2.5x upper limit of normal (ULN) or ≤1.0x ULN. (E) The influence of Eastern Cooperative Oncology Group (ECOG) performance status as an additional criterion. Displayed are eligible patient numbers after introducing ECOG scores of 0 to 2 or 0 to 1, with histology, stage, and ALT/AST laboratory test values (<2.5x ULN) as fixed criteria. ANC: absolute neutrophil count; CrCl: creatinine clearance; EGFR: epidermal growth factor receptor; NSCLC: non–small cell lung cancer; PD-1 ab: programmed cell death protein-1.
PNG File , 330 KBReferences
- Dickson M, Gagnon JP. Key factors in the rising cost of new drug discovery and development. Nat Rev Drug Discov. May 2004;3(5):417-429. [CrossRef] [Medline]
- Wouters OJ, McKee M, Luyten J. Estimated research and development investment needed to bring a new medicine to market, 2009-2018. JAMA. Mar 03, 2020;323(9):844-853. [FREE Full text] [CrossRef] [Medline]
- Jin S, Pazdur R, Sridhara R. Re-evaluating eligibility criteria for oncology clinical trials: analysis of investigational new drug applications in 2015. J Clin Oncol. Nov 20, 2017;35(33):3745-3752. [FREE Full text] [CrossRef] [Medline]
- Garcia S, Bisen A, Yan J, Xie XJ, Ramalingam S, Schiller JH, et al. Thoracic oncology clinical trial eligibility criteria and requirements continue to increase in number and complexity. J Thorac Oncol. Oct 2017;12(10):1489-1495. [FREE Full text] [CrossRef] [Medline]
- Osarogiagbon RU, Vega DM, Fashoyin-Aje L, Wedam S, Ison G, Atienza S, et al. Modernizing clinical trial eligibility criteria: recommendations of the ASCO-friends of cancer research prior therapies work group. Clin Cancer Res. May 01, 2021;27(9):2408-2415. [FREE Full text] [CrossRef] [Medline]
- Stensland KD, McBride RB, Latif A, Wisnivesky J, Hendricks R, Roper N, et al. Adult cancer clinical trials that fail to complete: an epidemic? J Natl Cancer Inst. Sep 2014;106(9):dju229. [CrossRef] [Medline]
- Fogel DB. Factors associated with clinical trials that fail and opportunities for improving the likelihood of success: a review. Contemp Clin Trials Commun. Sep 2018;11:156-164. [FREE Full text] [CrossRef] [Medline]
- Nipp RD, Hong K, Paskett ED. Overcoming barriers to clinical trial enrollment. Am Soc Clin Oncol Educ Book. May 2019;39:105-114. [CrossRef]
- Brøgger-Mikkelsen M, Ali Z, Zibert JR, Andersen AD, Thomsen SF. Online patient recruitment in clinical trials: systematic review and meta-analysis. J Med Internet Res. Nov 04, 2020;22(11):e22179. [FREE Full text] [CrossRef] [Medline]
- Naci H, Ioannidis JP. How good is "evidence" from clinical studies of drug effects and why might such evidence fail in the prediction of the clinical utility of drugs? Annu Rev Pharmacol Toxicol. 2015;55:169-189. [CrossRef] [Medline]
- Heneghan C, Goldacre B, Mahtani KR. Why clinical trial outcomes fail to translate into benefits for patients. Trials. Mar 14, 2017;18(1):122. [FREE Full text] [CrossRef] [Medline]
- Van Spall HG, Toren A, Kiss A, Fowler RA. Eligibility criteria of randomized controlled trials published in high-impact general medical journals: a systematic sampling review. JAMA. Mar 21, 2007;297(11):1233-1240. [CrossRef] [Medline]
- Sen A, Ryan PB, Goldstein A, Chakrabarti S, Wang S, Koski E, et al. Correlating eligibility criteria generalizability and adverse events using Big Data for patients and clinical trials. Ann N Y Acad Sci. Jan 2017;1387(1):34-43. [FREE Full text] [CrossRef] [Medline]
- Begg CB. Cancer clinical trials in the USA: patient eligibility, generalizability of results and technology transfer. Bull Cancer. 1987;74(2):197-203. [Medline]
- Liu R, Rizzo S, Whipple S, Pal N, Pineda AL, Lu M, et al. Evaluating eligibility criteria of oncology trials using real-world data and AI. Nature. Apr 2021;592(7855):629-633. [FREE Full text] [CrossRef] [Medline]
- Ko YA, Chen Z, Liu C, Hu Y, Quyyumi AA, Waller LA, et al. Developing a synthetic control group using electronic health records: application to a single-arm lifestyle intervention study. Prev Med Rep. Dec 2021;24:101572. [CrossRef] [Medline]
- Carrigan G, Whipple S, Capra WB, Taylor MD, Brown JS, Lu M, et al. Using electronic health records to derive control arms for early phase single-arm lung cancer trials: proof-of-concept in randomized controlled trials. Clin Pharmacol Ther. Feb 2020;107(2):369-377. [FREE Full text] [CrossRef] [Medline]
- Thomas DS, Lee AY, Müller PL, Schwartz R, Olvera-Barrios A, Warwick AN, et al. Contextualizing single-arm trials with real-world data: an emulated target trial comparing therapies for neovascular age-related macular degeneration. Clin Transl Sci. May 2021;14(3):1166-1175. [FREE Full text] [CrossRef] [Medline]
- Weng C, Tu SW, Sim I, Richesson R. Formal representation of eligibility criteria: a literature review. J Biomed Inform. Jun 2010;43(3):451-467. [FREE Full text] [CrossRef] [Medline]
- Chondrogiannis E, Andronikou V, Tagaris A, Karanastasis E, Varvarigou T, Tsuji M. A novel semantic representation for eligibility criteria in clinical trials. J Biomed Inform. May 2017;69:10-23. [FREE Full text] [CrossRef] [Medline]
- Su Q, Cheng G, Huang J. A review of research on eligibility criteria for clinical trials. Clin Exp Med. Oct 2023;23(6):1867-1879. [FREE Full text] [CrossRef] [Medline]
- Tu SW, Peleg M, Carini S, Bobak M, Ross J, Rubin D, et al. A practical method for transforming free-text eligibility criteria into computable criteria. J Biomed Inform. Apr 2011;44(2):239-250. [FREE Full text] [CrossRef] [Medline]
- Weng C, Wu X, Luo Z, Boland MR, Theodoratos D, Johnson SB. EliXR: an approach to eligibility criteria extraction and representation. J Am Med Inform Assoc. Dec 2011;18 Suppl 1(Suppl 1):i116-i124. [FREE Full text] [CrossRef] [Medline]
- Kang T, Zhang S, Tang Y, Hruby GW, Rusanov A, Elhadad N, et al. EliIE: an open-source information extraction system for clinical trial eligibility criteria. J Am Med Inform Assoc. Nov 01, 2017;24(6):1062-1071. [FREE Full text] [CrossRef] [Medline]
- Tian S, Erdengasileng A, Yang X, Guo Y, Wu Y, Zhang J, et al. Transformer-based named entity recognition for parsing clinical trial eligibility criteria. ACM BCB. Aug 2021;2021:49. [FREE Full text] [CrossRef] [Medline]
- Kreimeyer K, Foster M, Pandey A, Arya N, Halford G, Jones SF, et al. Natural language processing systems for capturing and standardizing unstructured clinical information: a systematic review. J Biomed Inform. Sep 2017;73:14-29. [FREE Full text] [CrossRef] [Medline]
- Yuan C, Ryan PB, Ta C, Guo Y, Li Z, Hardin J, et al. Criteria2Query: a natural language interface to clinical databases for cohort definition. J Am Med Inform Assoc. Apr 01, 2019;26(4):294-305. [FREE Full text] [CrossRef] [Medline]
- Liu H, Chi Y, Butler A, Sun Y, Weng C. A knowledge base of clinical trial eligibility criteria. J Biomed Inform. May 2021;117:103771. [FREE Full text] [CrossRef] [Medline]
- Kury F, Butler A, Yuan C, Fu LH, Sun Y, Liu H, et al. Chia, a large annotated corpus of clinical trial eligibility criteria. Sci Data. Aug 27, 2020;7(1):281. [FREE Full text] [CrossRef] [Medline]
- Dobbins NJ, Mullen T, Uzuner Ö, Yetisgen M. The Leaf Clinical Trials Corpus: a new resource for query generation from clinical trial eligibility criteria. Sci Data. Aug 11, 2022;9(1):490. [FREE Full text] [CrossRef] [Medline]
- Ross J, Tu S, Carini S, Sim I. Analysis of eligibility criteria complexity in clinical trials. Summit Transl Bioinform. Mar 01, 2010;2010:46-50. [FREE Full text] [Medline]
- Sim I, Olasov B, Carini S. An ontology of randomized controlled trials for evidence-based practice: content specification and evaluation using the competency decomposition method. J Biomed Inform. Apr 2004;37(2):108-119. [FREE Full text] [CrossRef] [Medline]
- Lee K, Liu Z, Chandran U, Kalsekar I, Laxmanan B, Higashi MK, et al. Detecting ground glass opacity features in patients with lung cancer: automated extraction and longitudinal analysis via deep learning–based natural language processing. JMIR AI. Jun 1, 2023;2:e44537. [CrossRef]
- Soysal E, Wang J, Jiang M, Wu Y, Pakhomov S, Liu H, et al. CLAMP - a toolkit for efficiently building customized clinical natural language processing pipelines. J Am Med Inform Assoc. Mar 01, 2018;25(3):331-336. [FREE Full text] [CrossRef] [Medline]
- Alfattni G, Peek N, Nenadic G. Attention-based bidirectional long short-term memory networks for extracting temporal relationships from clinical discharge summaries. J Biomed Inform. Nov 2021;123:103915. [FREE Full text] [CrossRef] [Medline]
- Xu J, Li Z, Wei Q, Wu Y, Xiang Y, Lee HJ, et al. Applying a deep learning-based sequence labeling approach to detect attributes of medical concepts in clinical text. BMC Med Inform Decis Mak. Dec 05, 2019;19(Suppl 5):236. [FREE Full text] [CrossRef] [Medline]
- Lee K, Mai Y, Liu Z, Raja K, Higashi MK, Jun T, et al. Establishing the automatic identification of clinical trial cohorts from electronic health records by matching normalized eligibility criteria and patient clinical characteristics. medRxiv. Preprint posted online April 09, 2024. [FREE Full text] [CrossRef]
- Hay M, Thomas DW, Craighead JL, Economides C, Rosenthal J. Clinical development success rates for investigational drugs. Nat Biotechnol. Jan 2014;32(1):40-51. [CrossRef] [Medline]
- Wong CH, Siah KW, Lo AW. Estimation of clinical trial success rates and related parameters. Biostatistics. Apr 01, 2019;20(2):273-286. [FREE Full text] [CrossRef] [Medline]
- Fang Y, Liu H, Idnay B, Ta C, Marder K, Weng C. A data-driven approach to optimizing clinical study eligibility criteria. J Biomed Inform. Jun 2023;142:104375. [CrossRef] [Medline]
- Unger JM, Hershman DL, Till C, Minasian LM, Osarogiagbon RU, Fleury ME, et al. "When offered to participate": a systematic review and meta-analysis of patient agreement to participate in cancer clinical trials. J Natl Cancer Inst. Mar 01, 2021;113(3):244-257. [FREE Full text] [CrossRef] [Medline]
- Unger JM, Vaidya R, Hershman DL, Minasian LM, Fleury ME. Systematic review and meta-analysis of the magnitude of structural, clinical, and physician and patient barriers to cancer clinical trial participation. J Natl Cancer Inst. Mar 01, 2019;111(3):245-255. [FREE Full text] [CrossRef] [Medline]
- Unger JM, Cook E, Tai E, Bleyer A. The role of clinical trial participation in cancer research: barriers, evidence, and strategies. Am Soc Clin Oncol Educ Book. May 2016;35:185-198. [FREE Full text] [CrossRef] [Medline]
- Wedam S, Fashoyin-Aje L, Bloomquist E, Tang S, Sridhara R, Goldberg KB, et al. FDA approval summary: palbociclib for male patients with metastatic breast cancer. Clin Cancer Res. Mar 15, 2020;26(6):1208-1212. [CrossRef] [Medline]
- Clinical trial eligibility design. Shiny Apps. URL: https://zongzhi-liu.shinyapps.io/trialdesign/ [accessed 2024-06-18]
- trialdesign. GitHub. URL: https://github.com/zz2liu/trialdesign [accessed 2024-06-18]
Abbreviations
ALT: alanine transaminase |
AST: aspartate aminotransferase |
BCa: breast cancer |
CD: Crohn disease |
ECOG: Eastern Cooperative Oncology Group |
EGFR: epidermal growth factor receptor |
EHR: electronic health record |
LOINC: Logical Observation Identifiers, Names, and Codes |
MM: multiple myeloma |
NLP: natural language processing |
NSCLC: non–small cell lung cancer |
PCa: prostate cancer |
SCA: synthetic control arm |
UC: ulcerative colitis |
ULN: upper limit of normal |
Edited by K El Emam, B Malin; submitted 16.07.23; peer-reviewed by MO Khursheed, D Chrimes, P Han, L Guo; comments to author 18.10.23; revised version received 07.11.23; accepted 23.03.24; published 29.07.24.
Copyright©Kyeryoung Lee, Zongzhi Liu, Yun Mai, Tomi Jun, Meng Ma, Tongyu Wang, Lei Ai, Ediz Calay, William Oh, Gustavo Stolovitzky, Eric Schadt, Xiaoyan Wang. Originally published in JMIR AI (https://ai.jmir.org), 29.07.2024.
This is an open-access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work, first published in JMIR AI, is properly cited. The complete bibliographic information, a link to the original publication on https://www.ai.jmir.org/, as well as this copyright and license information must be included.