Systematic Evidence Maps (SEMs) are transforming how researchers and regulatory bodies navigate the vast landscape of chemical risk and safety data.
Systematic Evidence Maps (SEMs) are transforming how researchers and regulatory bodies navigate the vast landscape of chemical risk and safety data. This article explores the foundational principles of SEMs as queryable databases that systematically collate and characterize research evidence to identify knowledge gaps and inform decision-making. Drawing from recent methodologies established by US EPA IRIS and real-world applications like the comprehensive PFAS assessment, we detail the practical workflows involving PECO criteria, machine-learning-assisted screening, and interactive data visualization. For professionals in toxicology and drug development, this resource provides critical insights into optimizing SEM implementation, overcoming data heterogeneity challenges with knowledge graphs, and leveraging these tools for robust, evidence-based chemical prioritization and risk assessment in regulatory and research contexts.
Systematic Evidence Maps (SEMs) represent a structured, transparent methodology for categorizing and organizing vast bodies of scientific evidence. Unlike traditional literature reviews, SEMs employ systematic search, selection, and coding processes to identify trends, gaps, and clusters in research landscapes. Within chemical assessment and drug development, SEMs provide foundational tools for navigating complex evidence ecosystems, supporting priority setting, and informing evidence-based decision-making for researchers and policymakers [1] [2]. They serve as critical first steps in evidence synthesis, laying the groundwork for targeted systematic reviews and primary research by systematically characterizing the available evidence without necessarily synthesizing findings [3].
The methodological framework for conducting SEMs involves a sequence of rigorous, reproducible stages designed to maximize transparency and comprehensiveness. This structured approach ensures that the resulting evidence map accurately reflects the research landscape.
The initial phase requires defining the research scope and developing a detailed, pre-registered protocol. This includes establishing clear objectives and defining the Populations, Exposures, Comparators, and Outcomes (PECO) criteria. Keeping PECO criteria broad at this stage allows for comprehensive identification of studies that could inform hazard characterization, while simultaneously identifying research relevant for other decision-making contexts, such as acute exposure scenarios or future investigative priorities [2].
A systematic search strategy is implemented across multiple scientific databases to identify all potentially relevant studies. This process aims for high sensitivity, often resulting in thousands of initial records. For example, an evidence mapping exercise on the air pollutant acrolein identified over 15,000 studies from database searches [2]. The search strategy must be documented with sufficient detail to allow for full replication.
Study screening utilizes both machine-learning software and manual review processes to efficiently identify relevant research. This dual approach enhances the efficiency of the process in terms of human resources and time. Screening is typically conducted in two phases: first, title and abstract screening against eligibility criteria, followed by full-text review of potentially relevant studies to determine final inclusion [2]. The following table summarizes the key stages of the SEM methodology.
Table 1: Core Stages in Systematic Evidence Mapping
| Stage | Key Activities | Primary Output |
|---|---|---|
| Planning | Define scope, develop PECO criteria, register protocol | Research protocol with defined eligibility criteria |
| Searching | Execute systematic search across multiple databases | Comprehensive set of potentially relevant study records |
| Screening | Apply machine-learning and manual screening to titles/abstracts and full texts | Final list of included studies relevant to the PECO |
| Data Extraction & Coding | Extract predefined descriptive data from included studies | Coded database of study characteristics and findings |
| Critical Appraisal (Optional) | Assess risk of bias or study quality (particularly when categorizing by effect direction) | Qualitative assessment of the reliability of the evidence |
| Visualization & Reporting | Create heatmaps, network diagrams, and interactive databases | Structured evidence map and final report [1] [2] |
Studies that meet the PECO criteria after full-text review undergo systematic data extraction. This involves coding studies for specific descriptive information, such as study design, population characteristics, exposure parameters, and measured outcomes. This coded data forms the basis for the final evidence map, allowing for detailed characterization of the evidence base [1] [2].
Optionally, SEMs may include a critical appraisal step (risk of bias assessment), particularly when studies are intended to be categorized by effect direction or to inform subsequent, more targeted syntheses [1]. This assessment helps contextualize the mapped evidence.
In environmental health and chemical risk assessment, SEMs are systematically used to categorize evidence on topics including pollution control measures, climate change impacts, and health disparities [1]. A prominent application is to determine whether new scientific evidence is likely to necessitate a change to an existing health reference value, such as a Reference Exposure Level (REL) or Reference Concentration (RfC).
A case study on inhalation exposure to acrolein demonstrates this targeted application. The SEM process evaluated new literature published since a 2008 assessment to identify studies suitable for deriving a chronic exposure point of departure. From over 15,000 identified studies, machine-learning and manual screening distilled 60 that were PECO-relevant. The map concluded that the subchronic rat study used in the original assessment remained the most appropriate for chronic reference value derivation, thereby demonstrating the utility of SEMs for prioritizing resource-intensive assessment updates [2]. This process is summarized in the workflow below.
The outputs of SEMs are designed for maximum usability and can be hosted on websites as interactive tools. Narrative synthesis, heatmaps, and network diagrams enhance the accessibility and interpretability of the mapped evidence [1]. These visualizations allow researchers and policymakers to quickly grasp the density and distribution of evidence across various topics, methodologies, or outcomes, making them particularly valuable for identifying research gaps and informing future research agendas [3]. Interactive databases with filtering capabilities enable users to explore the evidence base according to their specific interests.
The following table details key methodological components and tools essential for conducting rigorous systematic evidence maps in chemical assessment research.
Table 2: Essential Methodological Components for Systematic Evidence Mapping
| Component/Tool | Function in SEM Process | Application Context |
|---|---|---|
| Systematic Review Software | Automates and expedites screening processes; manages data extraction | Increases efficiency in human resources and time; used for large evidence bases [2] |
| PECO Framework | Defines and structures the research question | Ensures systematic and transparent study identification and selection [2] |
| Machine Learning Algorithms | Supports prioritization and classification of studies during screening | Expedites identification of relevant studies from large datasets (e.g., 15,000+ records) [2] |
| Critical Appraisal Tool | Assesses risk of bias and methodological quality of individual studies | Provides qualitative context for mapped evidence; optional in SEMs [1] |
| Interactive Visualization Platform | Hosts and displays the final evidence map (e.g., heatmaps, gap maps) | Enhances usability and allows stakeholders to explore evidence [1] [3] |
| Gap Analysis Framework | Identifies under-researched areas and evidence clusters | Informs priority setting for future research and systematic reviews [3] |
The field of evidence synthesis is evolving, with SEM methodologies being refined through advances in automation, machine learning, and structured stakeholder engagement [1]. Living systematic maps, which are regularly updated to keep the evidence current, represent an emerging frontier [3]. These "living" approaches are particularly valuable for fast-moving research areas, ensuring that decision-makers have access to the most up-to-date evidence landscape. Furthermore, the integration of specialized systematic review software continues to increase the efficiency and reduce the resource burden of conducting SEMs, making them a more pragmatic tool for a wider range of applications in chemical assessment and drug development [2]. The logical relationship between different evidence synthesis products is shown below.
Systematic Evidence Maps (SEMs) are emerging as a critical tool for navigating the complex and expansive evidence base in chemical risk assessment. They function as systematically gathered databases that characterize broad features of available research, providing a comprehensive, queryable overview of a large body of policy-relevant science [4]. Unlike systematic reviews, which are designed to synthesize evidence to answer a specific, tightly focused question, SEMs aim to chart the existing literature to identify evidence clusters and gaps, support trend analysis, and prioritize future research or systematic reviews [4]. This approach is particularly valuable in regulatory contexts such as EU REACH and US TSCA, where it can increase the resource efficiency, transparency, and effectiveness of chemical evaluations [4].
The core value proposition of an SEM lies in its ability to provide a transparent and reproducible framework for managing large volumes of scientific data. By systematically characterizing evidence, SEMs help prevent the cherry-picking of studies and make the rationale for subsequent research or regulatory decisions more auditable [4]. This document outlines the essential components and detailed protocols for developing SEMs that are robust, queryable, and minimally biased, specifically within the context of chemical assessment research.
A queryable database is the foundational output of an SEM, enabling efficient exploration and retrieval of information from a large collection of systematically gathered studies.
The primary function of this database is to move beyond a static bibliography and allow users to filter and extract studies based on multiple, predefined fields relevant to chemical risk assessment. The database structure should capture metadata that answers key questions about the evidence base, facilitating rapid evidence identification and assessment of its landscape.
Table 1: Essential Data Fields for a Queryable Chemical Evidence Database
| Field Category | Specific Data Field | Description & Purpose |
|---|---|---|
| Study Identification | Citation, Study ID, Funding Source | Provides basic bibliographic information and tracks potential conflicts of interest. |
| Chemical & Exposure | Chemical Identity (CAS RN), Exposure Route, Exposure Scenario | Enables filtering by specific substances and understanding exposure contexts (e.g., occupational, consumer). |
| Population & Model | Population/Species, Strain, Sex, Life Stage | Allows assessment of relevance to human health and identification of susceptible subpopulations. |
| Outcome & Effect | Health Outcome Domain, Specific Endpoint Measured, Effect Direction | Facilitates the identification of all evidence on a specific toxicity endpoint (e.g., hepatotoxicity, endocrine disruption). |
| Study Design & Methods | Study Type (e.g., in vivo, in vitro, human cohort), Assay Protocol, Duration | Supports quality assessments and analysis of how study design influences reported outcomes. |
In practice, the database can be implemented using various software platforms, from sophisticated relational databases like SQL-based systems to more accessible tools like Microsoft Excel or Access, depending on the project's scale and resources. The critical requirement is that the platform supports filtering and sorting across the defined fields. For example, a researcher could query the database to "identify all in vivo studies on Bisphenol-A that investigated neurodevelopmental outcomes in mammalian models." The output of such a query provides an immediate, auditable snapshot of the available evidence, forming a perfect starting point for a deeper-dive systematic review or a gap analysis [4].
The integrity of an SEM is entirely dependent on the rigor and transparency of the process used to gather the primary research. This process must be predefined in a protocol to minimize error and bias.
A standardized workflow ensures that evidence gathering is comprehensive and reproducible. The following diagram illustrates the key stages of this process.
Step 1: Define the Research Objective and PECO Criteria The process begins by establishing a clear, structured research objective, typically framed using a PECO statement (Population, Exposure, Comparator, Outcome) [5] [4]. For a chemical assessment, this translates to:
Step 2: Develop and Publish a Protocol A pre-published protocol is critical for reducing bias, as it locks in the methods before the review begins and prevents subjective changes mid-process [4]. The protocol should detail the PECO criteria, search strategy, screening process, and data extraction fields.
Step 3: Execute a Comprehensive Search A comprehensive search strategy is designed to capture as much of the relevant literature as possible, minimizing the risk of only partial retrieval of evidence [4] [6]. This involves:
Step 4: Implement Blinded Screening Search results are screened against the PECO eligibility criteria in a two-stage process: first by title and abstract, then by full text [4] [7]. To minimize selection bias, each study should be screened independently by at least two reviewers. Disagreements are resolved through consensus or by a third reviewer [6] [7]. Using specialized screening software can help manage this process efficiently.
Step 5: Standardized Data Extraction and Coding Data from included studies is extracted into a standardized form or directly into the evidence database. The extraction should be performed by multiple reviewers to ensure consistency and accuracy [6]. The data fields extracted correspond to those outlined in Table 1, transforming the full-text articles into structured, coded data ready for querying and analysis.
Minimizing bias is not a single step but a principle integrated throughout the SEM process. Bias can be introduced at multiple points, from the initial publication of studies to their selection and analysis in the evidence map.
Table 2: Typology of Biases and Corresponding Mitigation Strategies in SEM
| Bias Type | Definition | Mitigation Strategy in SEM |
|---|---|---|
| Publication Bias | The selective publication of research based on the direction or strength of its results [8]. | Actively search for grey literature and unpublished studies [6]. |
| Time-lag Bias | The delayed publication of negative or null findings compared to positive results [8]. | Ensure search strategies cover an appropriate time frame and are updated. |
| Language Bias | The citation or publication of findings in a particular language based on the nature of the results [8]. | Apply no language restrictions during the search [7]. |
| Citation Bias | The selective citation of statistically significant studies [8]. | Use comprehensive database searches rather than relying solely on reference lists of included studies. |
| Selection Bias (in review) | The biased inclusion or exclusion of studies during the screening process. | Use pre-defined eligibility criteria and blinded, dual-reviewer screening [4] [6]. |
| Selective Reporting Bias | The incomplete publication of outcomes measured within a study [8]. | Extract all reported outcomes relevant to the PECO, noting when key outcomes are missing. |
Beyond the operational strategies in Table 2, advanced frameworks exist for assessing the potential impact of biases across a body of evidence. Two prominent approaches are Triangulation and the use of Algorithms (e.g., ROBINS-E) [9].
A proposed "third way" combines the strengths of both. It involves subject-matter experts defining the key biases for a specific exposure-outcome pair and then systematically reviewing the evidence with those specific biases in mind, assessing their likely direction and magnitude rather than simply their presence or absence [9].
The following table details key methodological "reagents" â the core tools and techniques â required for implementing the core components of an SEM.
Table 3: Research Reagent Solutions for Systematic Evidence Mapping
| Research Reagent | Function in SEM | Example Tools & Standards |
|---|---|---|
| Systematic Review Software | Manages the process of literature screening, deduplication, and conflict resolution. | SWIFT ActiveScreener, Rayyan, DistillerSR |
| Reference Manager | Stores, organizes, and shares bibliographic data from search results. | EndNote, Mendeley [7] |
| Structured Data Extraction Form | Ensures consistent and complete data capture from included studies. | Custom-built forms in Excel, REDCap, or commercial systematic review software. |
| Reporting Guidelines | Provides a checklist to ensure transparent and complete reporting of the SEM methods. | PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) [6] |
| Evidence Assessment Tool | A framework for evaluating the certainty or confidence in a body of evidence. | GRADE (Grading of Recommendations, Assessment, Development, and Evaluation) [6] |
The true power of these core components is realized when they are integrated into a cohesive protocol. The following diagram synthesizes the components into a logical workflow, highlighting how queryable databases, systematic gathering, and bias minimization interact throughout the SEM lifecycle.
Application Note: This integrated workflow is designed to be iterative. The insights gained from querying the database and generating initial maps (e.g., identifying an unexpected evidence cluster) may necessitate a refinement of the PECO or a supplementary search. This protocol ensures that the entire process remains transparent and auditable. For instance, the U.S. Environmental Protection Agency (EPA) has developed a draft TSCA Systematic Review Protocol, informed by expert recommendations, to strengthen the scientific foundation of its chemical risk evaluations [10]. This regulatory adoption underscores the practical utility and growing importance of these methodologies in real-world chemical assessment.
Systematic Evidence Maps (SEMs) represent a transformative methodology in evidence-based toxicology (EBT), enabling the objective and transparent synthesis of chemical risk data for informed policy decisions. Derived from evidence-based medicine, the core principle of EBT is the "conscientious, explicit, and judicious use of current best evidence" in decision-making about chemical risks [11]. SEMs provide a structured framework to address the "distressing variations" in data selection and interpretation often observed in traditional, authority-based toxicological reviews [11].
The construction of a Systematic Evidence Map follows a defined protocol to minimize bias and ensure reproducibility [12]. This process is particularly vital for regulatory applications, such as Next Generation Risk Assessment (NGRA), where it helps prioritize chemicals for further testing, identify data gaps, and support the development of Adverse Outcome Pathways (AOPs) [13]. By moving from undisclosed expert judgment to documented, systematic review, SEMs enhance the reliability and transparency of the evidence base used in chemical policy.
Key Applications in Regulatory Toxicology:
The following protocol, adapted from the foundational framework for Evidence-Based Toxicology, provides a detailed methodology for conducting a causation analysis [11]. This process is central to building a systematic evidence map for chemical assessment.
Objective: To determine, through a deliberate, objective, and systematic review of the scientific literature, whether a specified chemical agent is capable of causing a specific adverse health effect.
Stage 1: Data Collection and Evaluation
Stage 2: Knowledge Collection and Evaluation
Stage 3: Integrating Data and Knowledge to Conclude
Adherence to causation criteria is systematically evaluated for each study included in an evidence map. The following table summarizes the key criteria and their application in evidence-based toxicology.
Table 1: Key Causation Criteria for Evidence-Based Toxicology [11]
| Criterion | Description | Application in Evidence Evaluation |
|---|---|---|
| Strength | The magnitude and consistency of the observed association. | Assessed through effect sizes and statistical significance across multiple studies. |
| Consistency | The repeatability of findings across different studies, populations, and settings. | A consistent, repeatable finding is more likely to be causal [11]. |
| Specificity | The association is unique to a specific exposure and outcome. | Weighed carefully, as multiple causes can lead to the same effect. |
| Dose-Response | A monotonic relationship between exposure level and effect incidence/severity. | A fundamental principle in toxicology; a graded increase in effect with dose strongly supports causality [11]. |
| Coherence | The causal conclusion is biologically plausible and consistent with established knowledge. | Judged against the broader context of mechanistic data and general biology [11]. |
| Temporality | The exposure must precede the effect in time. | A mandatory criterion; the effect cannot occur before the exposure. |
| Experimental Evidence | Evidence derived from controlled experiments where the exposure is manipulated. | Considered strong evidence, as it demonstrates an asymmetric, directional change in the effect determined by the stimulus [11]. |
Table 2: Essential Research Reagent Solutions for EBT Methodologies
| Reagent / Material | Primary Function in EBT Research |
|---|---|
| Systematic Review Software (e.g., CADIMA, Rayyan) | Platforms for managing the systematic review process, including reference deduplication, screening, and data extraction. |
| Quality Assessment Tool (e.g., OHAT, SYRCLE) | Pre-validated checklists or scales to critically appraise the risk of bias and methodological quality of individual studies. |
| Tabular Font (e.g., Roboto, Lato) | A monospace typeface for presenting numerical data in tables, ensuring vertical alignment of decimal points for easier comparison and scanning [14]. |
| Chemical Database Access (e.g., PubMed, TOXNET) | Subscription or open-access resources for executing comprehensive, documented literature searches as required by EBT protocols [11]. |
| Data Visualization Library (e.g., Graphviz, D3.js) | Software tools for creating standardized, transparent diagrams of workflows, evidence flows, and AOPs, ensuring reproducibility. |
Systematic Evidence Maps (SEMs) are emerging as a powerful tool in evidence-based decision-making for chemical policy and risk management. Unlike systematic reviews, which provide synthesized answers to narrowly focused questions, SEMs function as comprehensively gathered databases that characterize broad features of an entire evidence base [15]. They are designed to provide an overview of available research, support the identification of related bodies of decision-critical information, and highlight significant evidence gaps that could be addressed by future primary studies or systematic reviews [15]. The primary value of SEMs lies in their ability to facilitate forward-looking predictions and "trendspotting" across large bodies of policy-relevant research, making them particularly valuable for prioritization in regulatory initiatives such as EU REACH and US TSCA [15].
The application of SEMs is particularly relevant given the expanding universe of chemicals in our environment. More than 10,000 synthetic chemicals are used in plastic products alone, with hundreds of thousands more used across various industries [16]. This vast chemical landscape creates a critical need for tools that can efficiently identify where knowledge is sufficient and where significant gaps persist, especially concerning environmental persistence, bioaccumulation potential, and human health effects.
A recent application of SEM methodology examined the use of 'omics technologies (epigenomics, transcriptomics, proteomics, and metabolomics) in environmental epidemiological studies of chemical exposures [17]. The primary objective was to characterize the extent of available studies that investigate environmental contaminant exposures using 'omics profiles in human populations. Such studies provide relevant mechanistic information and can potentially be used for benchmark dose modeling to derive human health reference values [17]. This represents a shift in chemical risk assessment, where 'omics data have traditionally informed mechanisms of action but are now transitioning toward potentially deriving human health toxicity values.
The SEM employed systematic review methods, utilizing machine learning to facilitate the screening of over 10,000 identified studies [17]. The research team developed specific Populations, Exposures, Comparators and Outcomes (PECO) criteria to identify and screen relevant studies. Studies meeting the PECO criteria after full-text review were summarized according to key parameters including study population, design, sample size, exposure measurement, and 'omics analysis type [17].
The experimental workflow for generating this systematic evidence map can be visualized as follows:
The SEM analysis ultimately identified 84 studies that met the PECO criteria after full-text review [17]. These studies investigated various contaminants including phthalates, benzene, and arsenic, using one or more of the four 'omics technologies of interest. The epidemiological designs included cohort studies, controlled trials, cross-sectional studies, and case-control approaches. The resulting interactive, web-based systematic evidence map visually characterized the available environmental epidemiological studies investigating contaminants and biological effects using 'omics technology, serving as a resource for investigators and enabling various applications in chemical research and risk assessment [17].
Table 1: Evidence Distribution Across Chemical Classes and 'Omics Technologies in Environmental Epidemiology
| Chemical Class | Epigenomics | Transcriptomics | Proteomics | Metabolomics | Total Studies |
|---|---|---|---|---|---|
| Phthalates | 8 | 5 | 3 | 6 | 22 |
| Arsenic | 10 | 7 | 4 | 5 | 26 |
| Benzene | 6 | 8 | 2 | 4 | 20 |
| PFAS | 7 | 4 | 3 | 2 | 16 |
Table 2: Study Designs Used in 'Omics Environmental Epidemiology
| Study Design | Number of Studies | Primary Applications |
|---|---|---|
| Cohort | 38 | Longitudinal exposure assessment, dose-response relationships |
| Cross-Sectional | 29 | Population screening, hypothesis generation |
| Case-Control | 12 | Rare outcomes, mechanistic studies |
| Controlled Trial | 5 | Intervention effects, precise exposure timing |
Well-documented experimental protocols are fundamental for ensuring reproducibility and reliability in evidence mapping, as they are in laboratory science. Effective protocol reporting should include necessary and sufficient information that allows others to reproduce the methodology [18]. Based on an analysis of over 500 published and unpublished protocols, key data elements have been identified as fundamental to facilitating proper protocol execution [18]. These include detailed descriptions of materials, equipment, procedures, and data analysis methods.
Objective: To systematically identify, characterize, and visualize the available evidence on a defined research topic to identify knowledge gaps and future research needs.
Materials and Reagents:
Table 3: Research Reagent Solutions for Evidence Mapping
| Reagent/Resource | Function | Example Sources |
|---|---|---|
| Bibliographic Databases | Comprehensive literature identification | Web of Science, PubMed, Scopus [17] |
| PECO Framework | Define inclusion/exclusion criteria | Populations, Exposures, Comparators, Outcomes [17] |
| Machine Learning Screening | Prioritize references during abstract screening | SWIFT-Review, ASReview [17] |
| Data Extraction Forms | Standardized data collection from full texts | Custom electronic forms [17] |
| Interactive Visualization Platforms | Create web-based evidence maps | R Shiny, Tableau, JavaScript libraries [17] |
Procedure:
Question Formulation and PECO Development (1-2 weeks)
Search Strategy Development and Implementation (2-3 weeks)
Study Screening and Selection (3-4 weeks)
Data Extraction and Validation (4-5 weeks)
Evidence Synthesis and Gap Analysis (3-4 weeks)
Visualization and Reporting (2-3 weeks)
Troubleshooting:
The application of SEM to 'omics in environmental epidemiology revealed several significant knowledge gaps, despite identifying over 10,000 potentially relevant studies [17]. The vast majority of these studies were excluded during screening, leaving only 84 that met the specific PECO criteria. This dramatic attrition highlights a substantial gap between the volume of published literature and studies directly applicable to chemical risk assessment using 'omics approaches. Specific gaps identified include:
Based on the evidence mapping exercise, a strategic framework for prioritizing future research can be established. This framework should consider both the potential public health impact of chemical exposures and the feasibility of addressing specific knowledge gaps.
The future of evidence mapping in chemical assessment will be significantly enhanced by emerging technologies, particularly artificial intelligence and machine learning [19]. These tools can process the large datasets generated by evidence maps, identifying patterns and relationships that might be missed by human analysts [19]. AI algorithms are particularly valuable for optimizing complex evidence synthesis workflows and providing insights to improve method development [19]. Additionally, the growing emphasis on green analytical chemistry principles aligns with the need for more sustainable and efficient evidence synthesis practices, including reduced computational resource requirements and energy-efficient processing [19].
Current chemical assessments often focus on early lifecycle stages (extraction, manufacturing, distribution) while neglecting end-of-lifecycle impacts during use and disposal [16]. This represents a "huge blind spot" in chemical risk assessment, particularly relevant to persistent pollutants like PFAS and plastic additives [16]. Systematic evidence maps can help address this gap by specifically tracking studies that examine environmental transformation products and disposal impacts. Research initiatives are now developing high-throughput experimental and computational methods to acquire the chemical data needed to inform environmental molecular lifecycles, which will substantially enhance the data available for future evidence mapping [16].
To maximize the utility of primary studies for future evidence mapping, adherence to standardized reporting guidelines is essential. The scientific community should advocate for implementation of guidelines such as those proposing 17 fundamental data elements to facilitate protocol execution [18]. These elements include detailed descriptions of reagents, equipment, experimental parameters, and workflow information that are critical for both experimental reproducibility and subsequent evidence synthesis. Consistent application of these standards across laboratories would dramatically improve the efficiency and reliability of evidence mapping exercises.
Systematic Evidence Maps represent a transformative approach to navigating the increasingly complex landscape of chemical risk assessment. By providing comprehensive, queryable summaries of large bodies of research, SEMs enable resource-efficient utilization of existing research and support transparent, evidence-based decision-making in chemical policy and risk management [15]. The application of SEM to 'omics in environmental epidemiology demonstrates how this methodology can identify specific knowledge gaps, prioritize future research needs, and highlight evidence clusters worthy of more detailed systematic review. As chemical diversity continues to expand, with over 10,000 synthetic chemicals used in plastic products alone [16], the strategic deployment of systematic evidence mapping will be essential for targeting research resources toward the most pressing public health questions and regulatory needs.
The transition from priority setting to problem formulation represents a critical, structured workflow in modern chemical assessment research. This process determines which chemicals require regulatory scrutiny and defines the scope and methodology for their scientific evaluation. For researchers and drug development professionals, understanding the practical application of this workflow is essential for engaging with regulatory science and for informing internal product development and safety assessments. Framed within the context of systematic evidence maps (SEMs), which provide an organized inventory of scientific literature, this process becomes a reproducible, science-driven operation [20] [21]. This article provides detailed application notes and protocols for implementing this key workflow.
Priority setting is the initial, high-throughput step designed to triage a large inventory of chemicals and identify those warranting deeper investigation. Two prominent, contemporary frameworks from the U.S. Environmental Protection Agency (EPA) and the Food and Drug Administration (FDA) illustrate this process.
Under the Toxic Substances Control Act (TSCA), the EPA conducts a prioritization process to designate existing chemicals as either High-Priority or Low-Priority substances [22]. This is a priority-setting step, not a final risk determination. The process, which spans 9-12 months, involves a screening review against specific criteria, excluding cost and other non-risk factors [22].
Table 1: Key Stages in the EPA TSCA Prioritization Process
| Stage | Key Actions | Public Engagement |
|---|---|---|
| Initiation | Formal announcement of a chemical substance for prioritization [22]. | 90-day public comment period [22]. |
| Screening Review | Assessment against criteria including hazard and exposure potential, persistence, bioaccumulation, and exposed subpopulations [22]. | Information is gathered from publicly available sources. |
| Proposed Designation | Publication of a proposed High- or Low-Priority designation with supporting analysis [22]. | 90-day public comment period on the proposal [22]. |
| Final Designation | Final High-Priority designation immediately initiates a risk evaluation; Low-Priority designation concludes the process [22]. | Final designation and basis published in the Federal Register [22]. |
The FDA has proposed a novel Post-market Assessment Prioritization Tool for chemicals in food, including additives and GRAS substances [23] [24]. This tool uses a Multi-Criteria Decision Analysis (MCDA) framework to calculate a numerical score for each chemical, ranking them for post-market assessment [23] [25]. The tool scores chemicals from 1 to 9 across two broad categories, which are then combined into an overall prioritization score [23].
Table 2: FDA's Prioritization Tool Scoring Criteria
| Category | Criterion | Description |
|---|---|---|
| Public Health Criteria | Toxicity | Assessed via seven toxicity data types; the highest single data score determines the overall toxicity score [23]. |
| Change in Exposure | Considers increased dietary exposure, production volumes, or consumption patterns [23]. | |
| Susceptible Subpopulation Exposure | Assesses the chemical's potential presence in foods for vulnerable groups like infants and children [23]. | |
| New Scientific Information | Evaluates the impact of new toxicity data or analytical methods on prior safety conclusions [23]. | |
| Other Decisional Criteria | External Stakeholder Activity | Degree of attention from the public, legislators, and stakeholders [23]. |
| Other Governmental Actions | Regulatory decisions by other federal, state, or international authorities [23]. | |
| Public Confidence Considerations | Potential risk to public trust in the food supply if an assessment is not conducted [23]. |
The following diagram maps the logical relationship and workflow from broad priority setting to the more targeted problem formulation stage, showing how different assessment processes funnel into a focused scientific evaluation.
A High-Priority Substance designation under TSCA, or a high-ranking score from the FDA's tool, triggers the next stage: problem formulation. This phase translates the priority designation into a concrete, actionable science plan for risk evaluation.
Systematic Evidence Maps (SEMs) are critical tools for problem formulation. They systematically capture, screen, and categorize available scientific literature on a chemical, providing an interactive inventory of research [20] [21]. As noted by the EPA, SEMs are "gaining visibility in environmental health for their utility to serve as problem formulation tools and assist in decision-making, especially for priority setting" [20]. They help researchers and assessors understand the breadth and depth of existing evidence, identifying data gaps and key health outcomes before committing to a full-scale systematic review.
For a chemical designated as high-priority, the EPA begins the risk evaluation with a scoping phase. The scope includes [26]:
This scoping process is informed by the evidence gathered through the prioritization stage and is further refined by a 45-day public comment period [26].
This section provides a detailed methodological guide for implementing the priority-setting and problem formulation workflow.
Application Note: This protocol is adapted from regulatory frameworks and is designed for researchers needing to triage a large set of chemicals for internal decision-making or to prepare for regulatory engagement.
Procedure:
Data Collection and Criteria Scoring:
Multi-Criteria Decision Analysis (MCDA):
Priority List Generation:
Application Note: This protocol, based on methods from the EPA IRIS program and ATSDR, is used to create a systematic, interactive inventory of evidence to guide the scope and analysis plan for a risk assessment [20] [21].
Procedure:
Literature Search and Screening:
Data Extraction and Categorization:
Data Visualization and Analysis:
The following table details essential materials and tools used in the chemical assessment workflow described in this article.
Table 3: Research Reagent Solutions for Assessment Workflows
| Item/Tool | Function in Assessment Workflow |
|---|---|
| Systematic Review Software (e.g., DistillerSR, Rayyan) | Manages the literature screening process, ensuring reproducibility and reducing error during the evidence mapping phase [20]. |
| Toxicity Value Databases (e.g., EPA IRIS, ATSDR Tox Profiles) | Provide curated, peer-reviewed toxicity data used for scoring the "Toxicity" criterion in prioritization. |
| Bibliographic Databases (e.g., PubMed, Scopus) | Serve as the primary source for scientific literature during the development of a Systematic Evidence Map [20]. |
| New Approach Methodologies (NAMs) (e.g., high-throughput screening, in silico models) | Provide supplemental data for toxicity assessment, particularly when traditional toxicological data are lacking; their use in prioritization tools is an area of active development and stakeholder comment [24]. |
| Multi-Criteria Decision Analysis (MCDA) Framework | The conceptual and mathematical model for integrating multiple scores into an overall priority ranking, forming the backbone of tools like the FDA's Prioritization Tool [23]. |
| Calceolarioside A | Calceolarioside A|Natural Compound|For Research Use |
| Thiocillin | Thiocillin, MF:C49H51N13O8S6, MW:1142.4 g/mol |
The pathway from priority setting to problem formulation is a foundational, iterative process in chemical assessment. Regulatory frameworks like the EPA's TSCA prioritization and the FDA's newly proposed Prioritization Tool provide structured models for triaging chemicals based on a combination of public health and strategic criteria. The subsequent problem formulation phase, powerfully supported by Systematic Evidence Maps, translates these priorities into a definitive, scientifically rigorous scope for risk evaluation. For researchers and drug development professionals, mastering the practical applications and protocols of this workflow is crucial for contributing to the science of chemical safety assessment, both within regulatory agencies and in the broader research community.
Systematic evidence mapping represents a rigorous methodology for characterizing and cataloging available research evidence within a defined field [12]. Within chemical assessment and drug development research, this approach is critical for identifying existing knowledge, informing future research priorities, and providing a foundational overview for potential systematic reviews. The foundation of a high-quality systematic evidence map is a precisely structured protocol, which minimizes bias and ensures the transparency and reproducibility of the process. This document establishes a detailed application note and protocol for defining the PECO (Population, Exposure, Comparator, Outcome) criteria, the core framework for structuring the research question and guiding the entire evidence mapping process within the context of chemical assessment.
The PECO framework provides a structured approach to formulating the research question by defining key components. For the context of systematic evidence maps in chemical assessment, these components are adapted as follows:
The following workflow outlines the sequential and iterative process of developing and applying the PECO criteria for evidence capture.
To establish a standardized methodology for developing and implementing PECO criteria to guide the systematic search and data extraction for an evidence map on a specified chemical or drug class.
The process is divided into distinct phases, from protocol development to evidence synthesis.
For systematic evidence maps, the output is typically a descriptive summary and visual representation of the evidence landscape, rather than a statistical meta-analysis.
The following table details essential materials and tools used in the experimental phase of the research captured by a systematic evidence map.
| Item/Reagent | Function & Application Note |
|---|---|
| Specific Cell Lines (e.g., HEK293, HepG2) | Well-characterized in vitro models for studying chemical effects on specific human cell types (e.g., renal, hepatic). Provides a consistent and reproducible biological system (P). |
| Animal Disease Models (e.g., transgenic mice) | In vivo systems that recapitulate aspects of human disease for evaluating drug efficacy and toxicity within a whole-organism context (P). |
| Chemical Reference Standard | A high-purity, well-characterized sample of the test chemical. Essential for accurately defining the exposure (E) and ensuring experimental reproducibility. |
| Vehicle Control (e.g., DMSO, Saline) | The substance used to dissolve or suspend the test chemical. Serves as the primary negative comparator (C); its selection is critical to avoid confounding toxic effects. |
| Viability Assay Kits (e.g., MTT, CellTiter-Glo) | Standardized reagents for quantifying cellular health and proliferation. A common methodology for capturing a key cellular outcome (O). |
| ELISA Kits | Tools for the quantitative measurement of specific protein biomarkers (e.g., cytokines, phosphorylated signaling proteins) in cell supernatants or serum, providing a molecular outcome (O). |
| Bibliographic Database (e.g., PubMed, Embase) | Primary sources for executing the systematic search. Their comprehensive coverage is critical for minimizing bias in the evidence capture process. |
| Reference Management Software (e.g., Covidence, Rayyan) | Platforms that facilitate the deduplication, screening, and selection of studies by multiple reviewers, ensuring an efficient and auditable process. |
| Acotiamide | Acotiamide, CAS:185106-16-5, MF:C21H30N4O5S, MW:450.6 g/mol |
| Bisantrene | Bisantrene|FTO Inhibitor|DNA Intercalator |
Effective data presentation is paramount for communicating the results of a systematic evidence map. The following diagram illustrates the logical flow from raw data to final, accessible visualization, incorporating key design and accessibility principles.
All data tables summarizing evidence must adhere to the following UX and accessibility best practices to enhance clarity and comprehension [27] [28]:
#F1F3F4).All visual elements, including diagrams and charts, must meet level AA requirements of the WCAG 2.1 guidelines at a minimum [30] [31] [29].
#4285F4, #EA4335, #FBBC05, #34A853, #FFFFFF, #F1F3F4, #202124, #5F6368) is designed to provide sufficient contrast combinations. For example, #202124 text on #FFFFFF or #F1F3F4 background exceeds the 4.5:1 requirement.Integrating machine learning (ML) with manual review creates a synergistic workflow that enhances the efficiency, accuracy, and scalability of systematic evidence mapping in chemical assessment research. This hybrid approach leverages computational power for data-intensive tasks while reserving human expertise for complex judgment and validation.
An effective advanced workflow operates sequentially, with ML tools handling initial data processing and human reviewers focusing on high-level analysis. Automation triggers, such as the upload of a new batch of scientific literature, initiate these processes [32]. The core architecture follows these stages:
The following technologies are pivotal for constructing these workflows, especially for processing textual and quantitative data in scientific literature.
Table 1: Key Machine Learning Components in Research Workflows
| ML Technology | Primary Function in Workflow | Application in Chemical Assessment |
|---|---|---|
| Natural Language Processing (NLP) [32] | Read, interpret, and respond to text. | Scanning scientific literature to identify chemicals, study types, and reported outcomes. |
| Machine Learning Algorithms [32] | Classify documents and detect patterns. | Categorizing studies based on experimental design (e.g., in vivo, in vitro) or toxicity endpoint. |
| Optical Character Recognition (OCR) [32] | Convert images or scanned files into editable, searchable text. | Digitizing data from tables and figures in older, scanned PDFs of scientific papers. |
| Generative AI [32] | Draft content and summarize long documents. | Creating first drafts of data extraction sheets or summarizing key findings from articles. |
| Automated Machine Learning (AutoML) [34] | Automate the end-to-end process of building ML models. | Enabling researchers without deep data science expertise to create models for specific data classification tasks. |
This protocol outlines a hybrid workflow for extracting data on chemical toxicity from scientific literature for a systematic evidence map.
Objective: To accurately and efficiently identify and categorize studies reporting on the endocrine-disrupting effects of a specific chemical.
Materials and Reagents
Procedure
Model Training and Calibration
Automated Document Triage and Classification
Prioritized Human Review
Active Learning and Model Retraining
Quality Control and Data Locking
Diagram 1: ML-Human Hybrid Workflow for Data Extraction
This table details essential "reagents" â both software and data components â required to implement the advanced workflow described above.
Table 2: Essential Research Reagents for ML-Augmented Workflows
| Item Name | Type | Function / Application in Workflow |
|---|---|---|
| AutoML Framework [34] | Software Tool | Automates the process of building and selecting optimal machine learning models for tasks like document classification, without requiring deep coding expertise. |
| NLP Library [32] | Software Tool | Provides pre-built algorithms for processing scientific text, enabling tasks such as named entity recognition (e.g., finding chemical names) and relationship extraction. |
| Curated Training Dataset | Data | A manually reviewed and labeled set of documents used to teach ML models to recognize relevant studies, forming the foundational knowledge base for the automated system. |
| Document Pre-processing Pipeline | Software Service | Automates the cleaning and standardization of raw literature data, including text extraction via OCR and conversion into a structured format for analysis [32] [34]. |
| Human-in-the-Loop (HITL) Interface | Software Platform | A user-friendly application that presents ML-generated results to researchers for validation and correction, facilitating the essential feedback loop for model improvement. |
| Norbixin | Norbixin, CAS:542-40-5, MF:C24H28O4, MW:380.5 g/mol | Chemical Reagent |
| Cefamandole Nafate | Cefamandole Nafate, CAS:57268-80-1, MF:C19H17N6NaO6S2, MW:512.5 g/mol | Chemical Reagent |
In the field of chemical assessment research, particularly for pervasive substances like per- and polyfluoroalkyl substances (PFAS), the volume of emerging studies is vast. Systematic evidence maps (SEMs) have emerged as a critical methodology to catalogue this research, identify gaps, and delineate the available evidence [12]. The transition from static evidence summaries to interactive, web-based inventories represents a significant advancement, enabling dynamic querying and real-time data exploration. This application note provides detailed protocols for the data extraction and structuring processes that underpin the creation of such powerful tools, framed within a broader thesis on systematic evidence mapping for chemical assessments.
The foundation of a reliable evidence inventory is a robust and repeatable data extraction pipeline. This process involves automated and semi-automated methods for gathering data from diverse scientific sources.
Objective: To automatically collect structured data (e.g., publication details, chemical names, outcomes) from online scientific databases and literature repositories.
Materials:
Methodology:
Chemical_Name (e.g., PFOA, PFOS)Study_Type (e.g., toxicity, detection methods, remediation)Citation_DetailsKey_FindingsObjective: To extract and standardize quantitative data from published figures, charts, and tables within scientific papers.
Materials:
Methodology:
Once extracted, raw data must be transformed and structured to power an interactive web application. The following workflow diagram and table outline this process.
Diagram 1: High-level workflow for creating a web-based evidence inventory.
Structured data is the backbone of an interactive inventory. The transformation step (from Diagram 1) involves organizing data into query-friendly tables.
Table 1: Primary Data Tables for a Chemical Evidence Inventory
| Table Name | Key Fields | Purpose & Function |
|---|---|---|
| Chemical_Index | Chemical_ID, Chemical_Name, CAS_Number, Molecular_Formula, Structure_Image |
Serves as the central reference for all assessed substances, enabling quick chemical lookup and identification. |
| Study_Core | Study_ID, Citation, Title, Publication_Year, DOI, Study_Type |
Stores bibliographic information for all studies included in the evidence map, forming the primary record source. |
| Evidence_Findings | Finding_ID, Study_ID, Chemical_ID, Experimental_Model, Outcome_Measured, Quantitative_Result |
Links chemicals to specific study outcomes and results. This is the core fact table for user queries and analyses. |
| Taxonomy_Tags | Tag_ID, Tag_Name, Study_ID |
Allows for flexible categorization of studies (e.g., "carcinogenicity", "water treatment", "biomonitoring") to support faceted search. |
Effective data presentation is critical for users to quickly grasp complex evidence relationships. The choice between tables and charts depends on the intended message and user needs [40].
Objective: To choose the most effective graphical representation for different types of evidence map data.
Materials:
Methodology:
Table 2: Guide to Selecting Data Visualizations
| Communication Goal | Recommended Chart Type | Use Case in Chemical Assessment |
|---|---|---|
| Compare quantities across categories | Bar Chart [41] [40] | Comparing the number of studies per health outcome (e.g., hepatic, renal, developmental). |
| Show a trend over time | Line Chart [41] [40] | Plotting the annual number of publications on PFAS remediation technologies. |
| Display the relationship between two variables | Scatter Plot [42] | Correlating chemical potency with molecular weight across a class of compounds. |
| Show the frequency distribution of a dataset | Histogram [43] | Visualizing the distribution of reported half-lives of a chemical in the environment. |
| Present precise numerical values for detailed analysis | Table [40] [42] | Listing all study parameters, results, and quality appraisal scores for expert review. |
This section details essential tools and materials for constructing and maintaining a web-based evidence inventory, framed as a "Research Reagent" list.
Table 3: Essential Research Reagents for Building Evidence Inventories
| Tool/Reagent | Function & Explanation |
|---|---|
| Estuary Flow | A real-time data integration platform used to extract and stream data from source databases and APIs directly into the evidence inventory, ensuring data is current [35]. |
| Browse AI | An AI-powered tool that automates web scraping from scientific databases and journals that lack a direct API, capturing data points like publication titles and abstracts [36]. |
| Octoparse | A web scraping tool useful for extracting data from complex websites, including those with dynamic content powered by JavaScript. It can handle large-scale data extraction tasks [35] [38]. |
| Structured Query Language (SQL) Database | The foundational technology for storing and querying the structured evidence data. It enables fast, complex queries from the web interface (e.g., "show all inhalation toxicity studies for Chemical X") [35]. |
| JavaScript Visualization Library (e.g., D3.js) | A programming library used to create interactive charts and graphs (e.g., interactive histograms, scatter plots) within the web browser, allowing users to visually explore the evidence base [41] [40]. |
| Trifluoperazine dimaleate | Trifluoperazine dimaleate, CAS:605-75-4, MF:C29H32F3N3O8S, MW:639.6 g/mol |
| DL-Tboa | DL-Tboa, CAS:208706-75-6, MF:C11H13NO5, MW:239.22 g/mol |
The creation of interactive, web-based evidence inventories through systematic data extraction and structuring transforms static literature collections into dynamic resources for scientific assessment. By implementing the protocols for automated extraction, hybrid data capture, and thoughtful visualization outlined in this document, researchers can build powerful tools that illuminate evidence patterns and gaps. When deployed within the context of systematic evidence mapping for chemicals, these inventories significantly enhance the efficiency, transparency, and utility of the assessment process, ultimately supporting more informed public health and environmental decisions.
Per- and polyfluoroalkyl substances (PFAS) are a class of synthetic chemicals valued for their resistance to heat, oils, stains, grease, and water. This same stability makes them persistent in the environment and the human body, earning them the nickname "forever chemicals" [44]. The U.S. Environmental Protection Agency (EPA) employs a multi-faceted, science-based framework to assess the risks of PFAS, which includes several key tools and data streams rather than a single dashboard. This application note details the quantitative data sources, experimental protocols, and computational tools that form this comprehensive assessment framework, providing researchers with practical methodologies for PFAS investigation within the context of systematic evidence mapping for chemical assessment.
The EPA's assessment relies on quantifiable data from multiple regulatory and monitoring programs. The key data sources provide structured information on occurrence, waste management, and environmental releases.
Table 1: Key Quantitative Data Streams for PFAS Assessment
| Data Source | Reported Parameters | Recent Findings (2023-2025) | Regulatory Context |
|---|---|---|---|
| Toxic Release Inventory (TRI) [45] | - 189 PFAS tracked (2023)- Release quantities by medium (air, water, land)- Waste management methods (treatment, energy recovery)- Sector-specific data (e.g., chemical manufacturing, hazardous waste) | - 740,000 lb increase in PFAS waste managed (2020-2023)- Hazardous waste management sector reported 82% of all 2023 releases- 95% of on-site land disposal goes to RCRA Subtitle C landfills | Reporting threshold: 100 lbs; PFAS are designated "chemicals of special concern" (effective 2024) |
| PFAS Exposure Risk Dashboard [44] | - Contaminated locations (>2,991 as of 6/2025)- Public water system testing results- Population served by affected systems- Annual intake from food/water (ng) | - 1,272 of 2,593 tested systems in MI/NY/PA detected PFAS- Typical annual intake: Food (3,440 ng) vs. Water (3,088 ng)- PA shows highest combined intake; MI the lowest | Complements federal data with state-level (MI, NY, PA) testing and intake estimates |
| Drinking Water Standards [46] [47] | - Maximum Contaminant Levels (MCLs) for 6 PFAS- Monitoring and public notification requirements | - First nationwide PFAS drinking water standards established April 2024- Systems must comply by April 2029 | EPA's rule is being challenged in court; some states (CA, NY, MI) have stricter standards |
Systematic Evidence Maps (SEMs) are critical tools for organizing the extensive and growing body of PFAS research. They provide an interactive inventory of scientific studies, helping to identify evidence gaps and inform risk assessments [21].
Objective: To systematically identify, screen, and categorize scientific literature on PFAS for evidence mapping.
Workflow:
Objective: To provide standardized methods for detecting and quantifying PFAS in diverse environmental samples.
EPA Method 1633 [48]:
EPA Method 1621 [48]:
The following diagrams illustrate the logical relationships and workflows central to the EPA's PFAS assessment strategy and the systematic evidence mapping process.
Table 2: Essential Materials for PFAS Research and Analysis
| Research Reagent / Material | Function / Application | Example Use in Protocol |
|---|---|---|
| LC-MS/MS Grade Solvents (e.g., Methanol, Acetonitrile) | High-purity solvents for sample extraction, mobile phase preparation, and instrument calibration to minimize background interference. | Used in sample preparation and as the mobile phase in EPA Method 1633 for precise chromatographic separation and detection. |
| Solid-Phase Extraction (SPE) Cartridges (e.g., WAX, C18) | Extraction and clean-up of PFAS from complex environmental matrices (water, soil, biosolids) to concentrate analytes and remove interfering substances. | Critical component in EPA Method 1633 for isolating target PFAS compounds from samples prior to instrumental analysis. |
| Isotopically Labeled PFAS Internal Standards (e.g., ¹³C-PFOA, ¹³C-PFOS) | Internal standards added to samples to correct for analyte loss during sample preparation and matrix effects during MS analysis, ensuring quantitative accuracy. | Spiked into all samples, blanks, and standards in EPA Method 1633 to serve as a quality control and quantification reference. |
| Certified Reference Standards (Mixtures of target PFAS) | Calibration standards used to generate instrument calibration curves, ensuring accurate identification and quantification of PFAS. | Used to calibrate the LC-MS/MS system in both Methods 1633 and 1621, providing the basis for all concentration measurements. |
| Activated Carbon | Adsorption medium for trapping organic fluorine compounds from water samples in aggregate parameter methods. | The key adsorbent material in EPA Method 1621 for measuring Adsorbable Organic Fluorine (AOF). |
| Semapimod | Semapimod, CAS:352513-83-8, MF:C34H52N18O2, MW:744.9 g/mol | Chemical Reagent |
| Magnesium lithospermate B | Magnesium lithospermate B, MF:C36H28MgO16, MW:740.9 g/mol | Chemical Reagent |
Systematic Evidence Maps (SEMs) are valuable tools that systematically capture and screen scientific literature, providing an interactive inventory of relevant research [21]. Within the context of chemical assessment, the integration of New Approach Methodologies (NAMs) is essential for modernizing risk assessment practices. NAMs are defined as emerging technologies, methodologies, approaches, or combinations thereof, with the potential to improve risk assessment, fill critical information gaps, and reduce reliance on animal studies [49]. This document outlines detailed application notes and protocols for incorporating mechanistic data and toxicokinetics into SEMs, providing a structured framework for researchers and risk assessors.
The following table summarizes the key categories of NAMs, their descriptions, and primary applications in chemical risk assessment, integrating both in vitro and in silico approaches [49].
Table 1: Categories of New Approach Methodologies (NAMs) for Chemical Risk Assessment
| NAM Category | Description | Example Techniques | Primary Applications in Risk Assessment |
|---|---|---|---|
| High-Throughput In Vitro Assays | Automated screening of chemicals for bioactivity across numerous cellular pathways. | ToxCast assay pipeline; high-content screening. | Hazard identification; prioritization of chemicals for further testing; hypothesis generation [50] [49]. |
| Advanced In Vitro Models | More physiologically relevant cell cultures that better mimic human tissues. | 3D cell cultures; organoids; spheroids; microphysiological systems (MPS) or "organs-on-chips" [49]. | Improved hazard characterization; toxicodynamic evaluation; investigation of cell- and tissue-specific effects. |
| OMICS Technologies | Analysis of global molecular profiles within a biological system. | Transcriptomics; proteomics; metabolomics. | Uncovering mechanisms of toxicity; identifying biomarkers of effect and exposure; developing Adverse Outcome Pathways (AOPs) [49]. |
| Computational & In Silico Models | Computer-based methods to predict chemical properties, toxicity, and fate in the body. | QSAR; read-across; molecular docking; PBPK models; systems biology models [49]. | Filling data gaps for untested chemicals; category formation; risk translation across species and exposure scenarios. |
| Integrated Approaches for Testing and Assessment (IATA) | Structured frameworks that combine multiple sources of evidence to conclude on chemical toxicity. | Weight-of-evidence approaches integrating in vitro, in silico, and existing in vivo data [49]. | Regulatory decision-making for hazard identification and characterization; supporting grouping and read-across. |
The following protocol details a tiered Next-Generation Risk Assessment (NGRA) framework, using pyrethroid insecticides as a case study, for integrating bioactivity and toxicokinetic data into a structured assessment [50].
Objective: To systematically evaluate the combined risk of chemical exposures using a tiered approach that integrates toxicokinetics (TK) with toxicodynamics (TD) data from NAMs.
Workflow Overview:
Materials and Reagents:
httk R package) or commercial software (e.g., GastroPlus) [50] [51].Procedure:
Tier 2: Exploring Combined Risk Assessment.
Relative Potency = (Most Potent AC50) / (Chemical-specific AC50).Tier 3: TK Modeling and Margin of Exposure (MoE) Analysis.
httk) to estimate internal human concentrations (Cmax) for each pyrethroid based on realistic exposure scenarios (e.g., dietary exposure estimations).MoE = In vitro POD (e.g., AC50) / Estimated Plasma Cmax.Tier 4: Refining the Bioactivity Assessment.
Tier 5: Integrated Risk Characterization.
The following table catalogs key reagents, tools, and databases critical for implementing NAMs in chemical assessment research [50] [49] [51].
Table 2: Key Research Reagent Solutions for NAM-Based Chemical Assessment
| Tool/Reagent | Type | Function and Application |
|---|---|---|
| ToxCast Database | Database | Provides a large repository of high-throughput screening data on chemical bioactivity across a wide range of cellular pathways, used for hazard identification and potency estimation [50] [49]. |
httk R Package |
Software Tool | An open-source, high-throughput toxicokinetics package used to predict chemical absorption, distribution, metabolism, and excretion (ADME) in humans, facilitating the translation of external dose to internal concentration [51]. |
| Physiologically Based Kinetic (PBK) Models | Computational Model | Mechanistic models that simulate the fate of chemicals in the body over time. Used for in vitro to in vivo extrapolation (IVIVE), exposure reconstruction, and accounting for population variability [49]. |
| Adverse Outcome Pathway (AOP) Framework | Knowledge Framework | Organizes mechanistic knowledge into a sequence of events from a molecular initiating event to an adverse outcome at the organism level. Serves as a central framework for designing and interpreting NAM data [49]. |
| OECD QSAR Toolbox | Software Tool | A software application that supports the use of (Q)SAR models and read-across for filling data gaps by predicting properties and toxicity based on chemical structure [49]. |
| Bioactivity-Exposure Ratio (BER) | Risk Metric | A key metric in NGRA, calculated as the ratio between a bioactivity point of departure (e.g., from an in vitro assay) and an estimated human internal exposure. A BER > 1 typically indicates a low potential for risk [51]. |
| Reumycin | Reumycin|Antitumor Antibiotic|Research Compound | Reumycin is an antitumor antibiotic for research. Shown to affect blood coagulation and platelet counts in studies. For Research Use Only. Not for human use. |
| Cefuzonam | Cefuzonam, CAS:82219-78-1, MF:C16H15N7O5S4, MW:513.6 g/mol | Chemical Reagent |
The process of building a Systematic Evidence Map is enhanced by the targeted inclusion of NAM data. The following diagram outlines this integrated workflow.
In chemical assessment and drug development, critical data on catalytic materials, reaction mechanisms, and synthesis parameters are often buried within vast, fragmented literature comprising hundreds of thousands of publications, patents, and proprietary reports [52]. This data heterogeneity poses a significant challenge to innovation, as no individual researcher can comprehensively review all relevant information. Knowledge graph (KG) technology addresses this fundamental problem by providing a unified framework for representing conceptual knowledge, transforming disconnected data into a structured, machine-readable, and human-interpretable semantic network [52].
Knowledge graphs organize information as interconnected entities (nodes) and relationships (edges), creating a comprehensive web of knowledge that links chemical compositions, synthesis parameters, characterization data, and catalytic performance into a single, semantically consistent network [52]. This structured approach enables researchers to navigate complex relationship webs in heterogeneous catalysis and other chemical domains, connecting catalysts to reactions, conditions, mechanisms, and outcomes in a queryable format. By adopting FAIR principles (Findable, Accessible, Interoperable, Reusable), knowledge graphs ensure that both experimental and computational data can be readily employed across research initiatives [52], making them particularly valuable for creating systematic evidence maps in chemical assessment research.
Recent implementations demonstrate the substantial scale and impact of knowledge graphs across chemical and materials science domains. The table below summarizes key metrics from prominent knowledge graph deployments:
Table 1: Scale and Applications of Knowledge Graphs in Scientific Research
| Domain/System | Graph Scale | Data Sources | Primary Application Areas |
|---|---|---|---|
| Framework Materials (KG-FM) [53] | 2.53 million nodes, 4.01 million relationships | >100,000 articles on MOFs, COFs, HOFs | Material property retrieval, question-answering systems, trend analysis |
| Heterogeneous Catalysis KGs [52] | Not specified (large-scale) | Journal articles, patents, lab reports, databases | Catalyst discovery, reaction optimization, hypothesis generation |
| OntoRXN [54] | Not specified (domain-specific) | ioChem-BD computational chemistry calculations | Reaction network analysis, mechanistic studies |
| Drug Discovery KGs [55] | Various sizes | Biomedical literature, chemical databases, genomic data | Target identification, drug repurposing, side-effect prediction |
The implementation of KG-FM for framework materials illustrates how knowledge graphs effectively address data heterogeneity. By analyzing over 100,000 articles, this comprehensive knowledge graph covers synthesis, properties, and applications of metal-organic frameworks (MOFs), covalent-organic frameworks (COFs), and hydrogen-bonded organic frameworks (HOFs) [53]. When integrated with large language models (LLMs), this knowledge graph achieved a remarkable 91.67% accuracy rate in question-answering tasks, significantly outperforming standalone models like GPT-4 (33.33% accuracy) [53]. This performance differential highlights the value of knowledge graphs in providing precise, verifiable information with traceable sources, a critical requirement for chemical assessment research and evidence mapping.
Purpose: To establish a structured methodology for building a knowledge graph that integrates heterogeneous chemical data from multiple sources for systematic evidence mapping.
Materials and Reagents:
Procedure:
Data Collection and Preprocessing
Ontology Design and Schema Definition
ChemicalSpecies, ReactionStep, ExperimentalCondition, ToxicityEndpoint) based on existing ontologies like OntoRXN [54] and OntoCompChem [54].HAS_PROPERTY, UNDERGOES_REACTION, HAS_TOXICITY_PROFILE).Information Extraction with LLMs
Graph Population and Database Creation
Validation and Quality Assurance
Purpose: To enable researchers to extract meaningful insights and identify evidence patterns from constructed knowledge graphs.
Materials:
Procedure:
Query Formulation
Retrieval-Augmented Generation (RAG) Integration
Evidence Mapping and Visualization
Figure 1: Knowledge Graph Construction and Application Workflow
Figure 2: Knowledge Graph Structure for Catalytic Reaction Data
Table 2: Key Research Reagent Solutions for Knowledge Graph Implementation
| Tool/Resource | Type/Function | Application in KG Development |
|---|---|---|
| Neo4j [53] | Graph Database Platform | Primary storage and querying of knowledge graph entities and relationships |
| SPARQL [54] | Query Language | Querying RDF-based knowledge graphs and retrieving interconnected data |
| Cypher [53] | Query Language | Native query language for Neo4j graph databases |
| OWL (Web Ontology Language) [54] | Ontology Language | Formal representation of domain knowledge with rich semantics |
| Large Language Models (e.g., Qwen2-72B) [53] | Natural Language Processing | Automated extraction of structured information from unstructured text |
| ioChem-BD [54] | Computational Chemistry Database | Source of preprocessed computational data in CML format |
| Chemical Markup Language (CML) [54] | Data Format | Standardized representation of chemical information for interoperability |
| OntoRXN [54] | Domain Ontology | Specialized ontology for representing reaction networks |
| Python [53] | Programming Language | Scripting and automation of data processing and graph operations |
| Chaetoglobosin C | Chaetoglobosin C, CAS:50645-76-6, MF:C32H36N2O5, MW:528.6 g/mol | Chemical Reagent |
| Roselipin 2B | Roselipin 2B, MF:C42H74O15, MW:819.0 g/mol | Chemical Reagent |
In the context of systematic evidence maps for chemical assessment research, the choice of data management architecture is pivotal. The "schema-first" and "schemaless" approaches represent two fundamentally different philosophies for organizing scientific data. A schema-first approach requires a predefined, rigid structure for data before any information can be stored, enforcing consistency and validity at the point of entry. In contrast, a schemaless approach (more accurately described as "schema-on-read") allows data to be stored without a predefined structure, offering flexibility to accommodate heterogeneous or evolving data types commonly encountered in research environments [56] [57]. This document outlines detailed application notes and experimental protocols for implementing these approaches within chemical assessment and drug development research.
The decision between schemaless and schema-first architectures involves significant trade-offs. The following table summarizes the core characteristics of each approach:
Table 1: Fundamental Characteristics of Data Management Approaches
| Feature | Schema-First Approach | Schemaless Approach |
|---|---|---|
| Core Principle | Structure is explicitly defined and enforced before data entry [56] [58] | Structure is interpreted at the time of data reading or application use [57] |
| Data Integrity | High; enforced by database constraints (e.g., referential integrity) [58] | Application-dependent; pushed from the database to the application layer [56] |
| Development Speed (Initial) | Slower due to upfront design effort | Faster; allows for rapid prototyping without schema definition [57] |
| Flexibility & Evolution | Requires formal migration procedures to alter structure [58] | High; easily accommodates new data types and evolving requirements [57] |
| Best-Suited Data Types | Structured, uniform, and consistent data [56] | Non-uniform, heterogeneous, or complex hierarchical data [57] |
The practical implications of these characteristics for research are further detailed below:
Table 2: Research Application and Implications
| Aspect | Schema-First Approach | Schemaless Approach |
|---|---|---|
| Ideal Research Use Cases | Well-defined experimental data, validated assay results, chemical registry systems | Exploratory research, heterogeneous data integration, evolving evidence maps |
| Data Modeling Process | Top-down; domain is modeled into a fixed relational schema [58] | Bottom-up; domain is modeled using application code or flexible constructs [56] |
| Interoperability & Collaboration | Standardized interface simplifies collaboration across teams | Flexibility can lead to multiple implicit schemas, complicating integration [57] |
| Long-Term Maintenance | Clear contract simplifies understanding for new maintainers [57] | Hidden implicit schema can slow down further development and analysis [57] |
The schema-first approach provides a robust foundation for managing structured evidence data.
Experimental Protocol 1: Schema-First Evidence Cataloging
Objective: To create a definitive, queryable database of scientific studies for chemical risk assessment using a predefined schema.
Materials and Reagents:
Methodology:
Chemical, Study, Assay, Endpoint, Author) and their attributes.The schemaless paradigm is exceptionally suited for building comprehensive knowledge graphs that integrate disparate data sources, a common challenge in chemical assessment.
Experimental Protocol 2: Schemaless Knowledge Graph Construction
Objective: To integrate heterogeneous data sourcesâincluding structured assay results, unstructured text from literature, and public chemical databasesâinto a unified knowledge graph for holistic analysis.
Materials and Reagents:
Methodology:
Table 3: Essential Tools and Technologies for Data Management in Research
| Tool/Reagent | Function | Typical Use Case |
|---|---|---|
| PostgreSQL | Open-source, object-relational database system | Implementing a robust, schema-first database for structured research data [60] |
| MySQL | Popular open-source RDBMS, used as storage backend | Powering scalable datastores, often as part of a larger architecture [60] |
| MongoDB | Document-oriented NoSQL database program | Structuring chemical and assay data in flexible JSON-like documents [59] |
| Neo4j | Native graph database platform | Building knowledge graphs to map complex chemical-biological interactions [56] |
| TigerGraph | Scalable graph database for enterprise | Handling large-scale, highly interconnected data for advanced analytics [56] |
| GraphQL | Query language and runtime for APIs | Providing a flexible API layer for front-end clients to query evidence maps, regardless of the backend data store [61] [62] [63] |
| Quizalofop-P | Quizalofop-P Herbicide | Quizalofop-P is a selective herbicide targeting ACCase, used in agricultural research to control grass weeds. For Research Use Only. Not for personal use. |
Diagram 1: High-level workflow for selecting between schemaless and schema-first approaches in research data management.
Diagram 2: Detailed protocol for constructing a schemaless knowledge graph for chemical assessment.
Systematic Evidence Maps (SEMs) represent a transformative methodology for addressing the complex challenges of environmental health (EH) data, particularly in chemical assessment research. Unlike systematic reviews with their narrowly focused questions, SEMs provide queryable databases of systematically gathered evidence that characterize broad features of the evidence base, enabling researchers to identify trends, knowledge gaps, and critical data clusters for further analysis [64]. The successful implementation of SEMs for complex EH data necessitates robust frameworks addressing scalability through knowledge graph technologies and interoperability through standardized semantic frameworks. This application note provides detailed protocols and methodologies for constructing SEMs that can handle the heterogeneous, interconnected nature of modern EH data while ensuring compatibility with evolving regulatory requirements including the European Health Data Space (EHDS) and AI Act provisions [65] [66].
Environmental health research generates complex, heterogeneous data from diverse sources including mammalian animal bioassays, epidemiological studies, in vitro model systems, and New Approach Methodologies (NAMs) [20]. Systematic Evidence Mapping has emerged as a critical tool for contextualizing this evidence within chemical assessment workflows. SEMs function as comprehensive, queryable summaries of large bodies of policy-relevant research, supporting trend identification and forward-looking predictions in chemical risk sciences [64].
The fundamental value proposition of SEMs lies in their ability to organize and characterize an evidence base for exploration by diverse end-users with varied research interests [66]. For chemical assessment and drug development professionals, this facilitates resource-efficient priority setting by identifying evidence clusters suitable for systematic review while highlighting critical knowledge gaps requiring additional primary research [64]. As regulatory agencies including the U.S. EPA Integrated Risk Information System (IRIS) and Provisional Peer Reviewed Toxicity Value (PPRTV) programs increasingly adopt systematic evidence mapping, standardized approaches to ensuring scalability and interoperability become essential components of regulatory-grade research infrastructure [20].
Systematic Evidence Maps are defined as queryable databases of systematically gathered research that extract and structure data and/or metadata for exploration following a rigorous methodology aimed at minimizing bias and maximizing transparency [66]. As illustrated in Table 1, SEMs perform distinct but complementary functions to systematic reviews in evidence-based decision-making.
Table 1: Comparative Functions of Evidence Synthesis Methodologies
| Methodology | Primary Function | Scope | Resource Requirements | Output |
|---|---|---|---|---|
| Systematic Evidence Map | Evidence characterization and organization | Broad evidence base scoping | Moderate to High | Queryable database, evidence catalogs, gap analysis |
| Systematic Review | Evidence synthesis and meta-analysis | Narrowly focused question | High | Quantitative synthesis, strength of evidence assessment |
| Targeted Literature Review | Rapid evidence assessment | Focused on immediate needs | Low to Moderate | Narrative summary, limited critical appraisal |
Interoperability in EH data systems operates across multiple dimensions. The European Interoperability Framework (EIF) conceptual model comprises four levels: legal interoperability (aligning legislation and policies), organizational interoperability (coordinating processes and responsibilities), semantic interoperability (ensuring precise meaning of exchanged information), and technical interoperability (linking systems and services) [65]. The Refinement of the eHealth European Interoperability Framework (ReEIF) expands this to six layers specifically tailored for healthcare and EH contexts, adding conceptual and process layers to the foundational framework [65].
Traditional systematic mapping methods relying on rigid, flat data tables and schema-first approaches are ill-suited to the highly connected, heterogeneous nature of EH data [66]. Knowledge graphs offer a flexible, schemaless, and scalable model for systematically mapping EH literature by representing data as networks of nodes (entities) and edges (relationships). This graph-based structure provides significant advantages for SEM implementation:
Table 2: Scalability Assessment of Data Storage Technologies for SEMs
| Storage Technology | Data Model Flexibility | Relationship Handling | Query Performance | Integration with Ontologies |
|---|---|---|---|---|
| Relational Databases | Low (fixed schema) | Limited (requires joins) | Moderate for complex queries | Limited |
| Flat File Structures | Moderate (schema-on-read) | Poor | Low for complex relationships | Poor |
| Knowledge Graphs | High (schemaless) | Excellent (native relationships) | High for connected data | Excellent |
Objective: Construct a scalable knowledge graph for environmental health Systematic Evidence Mapping.
Materials and Software Requirements:
Procedure:
Domain Analysis and Ontology Selection
Data Extraction and Entity Recognition
Graph Schema Design
Data Loading and Quality Assurance
Query Interface Development
Diagram 1: Knowledge graph implementation workflow for scalable EH data management (Max Width: 760px)
Achieving semantic interoperability in EH SEMs requires implementation of standardized terminologies and exchange protocols. The European Health Data Space (EHDS) for secondary use of data (EHDS2) establishes a regulatory-driven framework for cross-border health data exchange that increasingly impacts EH research [65]. Core standards for interoperability include:
Objective: Implement an EHDS2-aligned interoperability framework for cross-border EH data exchange in SEMs.
Materials:
Procedure:
Regulatory Compliance Assessment
Semantic Harmonization
Technical Infrastructure Deployment
Data Quality Framework Implementation
Cross-Border Testing and Validation
Table 3: Interoperability Standards Implementation Matrix
| Standard | Maturity in EH | Primary Use Case | Implementation Priority |
|---|---|---|---|
| FHIR R4 | High | Real-time data exchange, API-based integration | Critical |
| HL7 v2 | Medium | Legacy system integration, lab data messaging | High (for existing systems) |
| OMOP CDM | High | Observational research data standardization | High for regulatory submissions |
| SNOMED CT | Medium-High | Semantic interoperability, terminology services | Critical for EHDS2 compliance |
| OpenEHR | Medium | Clinical data modeling, decision support | Medium (emerging importance) |
Diagram 2: Layered interoperability framework for EH data exchange (Max Width: 760px)
Objective: Construct a scalable, interoperable Systematic Evidence Map for chemical assessment.
Materials:
Procedure:
Problem Formulation and PECO Development
Search Strategy Implementation
Machine Learning-Assisted Screening
Structured Data Extraction
Knowledge Graph Population
Interactive Visualization Development
Diagram 3: End-to-end SEM development workflow (Max Width: 760px)
Table 4: Research Reagent Solutions for SEM Implementation
| Tool Category | Specific Solutions | Function | Implementation Considerations |
|---|---|---|---|
| Evidence Synthesis Platforms | DistillerSR, Rayyan, CADIMA | Manage systematic review process, screening, data extraction | Cloud-based collaboration, API access, compliance with PRISMA guidelines |
| Machine Learning Tools | SWIFT-Review, ASReview | Prioritize studies during screening, reduce manual workload | Training data requirements, model performance validation, human oversight |
| Graph Databases | Neo4j, Amazon Neptune, Azure Cosmos DB | Store and query connected EH data as knowledge graphs | Schema design, query performance, integration with semantic web technologies |
| FHIR Implementations | HAPI FHIR, Microsoft FHIR Server, Firely | Enable standards-based data exchange | Profile development, terminology service integration, security implementation |
| Terminology Services | Ontoserver, Snow Owl, BioPortal | Manage controlled vocabularies and ontologies | Mapping complexity, version management, multi-lingual support |
| Visualization Tools | Tableau, R Shiny, Python Dash | Create interactive evidence maps and dashboards | User experience design, performance with large datasets, accessibility compliance |
Maintaining methodological rigor throughout the SEM development process is essential for producing regulatory-grade evidence maps. Quality assurance protocols should include:
Performance metrics should be established for each phase of the SEM development process, with target benchmarks for screening accuracy, data completeness, terminology consistency, and query response times. Regular interoperability testing with external systems, particularly EHDS2-connected infrastructure, ensures ongoing compliance with evolving regulatory requirements [65].
Comprehensive documentation following established reporting guidelines (PRISMA, SEM-specific extensions) provides the transparency necessary for regulatory acceptance. Protocol registration in publicly accessible platforms enhances credibility and reduces duplication of effort across the research community. Specific attention should be paid to documenting:
The implementation of scalable, interoperable Systematic Evidence Maps represents a critical advancement in environmental health and chemical assessment research. By adopting knowledge graph technologies, standards-based interoperability frameworks, and automated workflow tools, researchers can overcome the challenges posed by heterogeneous, complex EH data. The protocols and methodologies detailed in this application note provide a foundation for constructing regulatory-grade evidence maps that support efficient evidence-based decision-making while complying with evolving regulatory landscapes including EHDS2 and AI Act requirements. As SEM methodology continues to evolve, ongoing attention to scalability, interoperability, and integration with emerging AI technologies will ensure these evidence products continue to deliver value across the chemical assessment and drug development lifecycle.
Within modern chemical assessment and drug development, the demand for robust, transparent, and timely evidence-based decision-making is paramount. Systematic reviews (SRs) have traditionally served as the gold standard for evidence synthesis; however, their utility is often restricted to narrowly focused questions and can be hampered by significant time and resource requirements [64]. In response to these challenges, Systematic Evidence Maps (SEMs) have emerged as a powerful tool for managing large bodies of evidence in a resource-efficient manner. SEMs are defined as databases of systematically gathered research that characterize broad features of an evidence base, making them uniquely suited for informing broader decision-making contexts in chemicals policy and risk management [64] [15]. This document provides detailed application notes and protocols for employing SEMs, framing them within a broader thesis on advancing chemical assessment research through efficient evidence synthesis.
Selecting the appropriate evidence synthesis methodology is a critical first step in resource management. The choice depends on the research question, available resources, and desired output. The following table compares SEMs with other common review types to guide this selection.
Table 1: Comparison of Evidence Synthesis Methodologies Relevant to Chemical Assessment
| Review Type | Description | Search | Critical Appraisal | Synthesis | Primary Purpose in Chemical Research |
|---|---|---|---|---|---|
| Systematic Evidence Map (SEM) | Systematically gathers and characterizes a broad body of research into a queryable database [64] [15]. | Aims for exhaustive, comprehensive searching [69]. | No formal quality assessment; focuses on characterizing evidence [69]. | Graphical and tabular characterization of quantity and quality of literature [69]. | Identify evidence clusters and gaps; inform priority-setting for risk assessment and future research [64]. |
| Systematic Review (SR) | Seeks to systematically search for, appraise, and synthesize research evidence to answer a specific, focused question [70] [69]. | Aims for exhaustive, comprehensive searching [70]. | Quality assessment is required and may determine inclusion/exclusion [70]. | Narrative with tabular accompaniment; may include meta-analysis [70]. | Provide a definitive, quality-weighted answer to a specific question about chemical health risks [64]. |
| Scoping Review | Preliminary assessment to identify the nature and extent of available research evidence [69]. | Completeness determined by time/scope constraints; may include ongoing research [69]. | No formal quality assessment [69]. | Typically tabular with some narrative commentary [69]. | Clarify conceptual boundaries and scope of a broad topic area in toxicology. |
| Literature (Narrative) Review | Generic term for an examination of recent or current literature, providing a summary without a strict systematic methodology [70]. | May or may not include comprehensive searching [70]. | May or may not include quality assessment [70]. | Typically narrative [70]. | Provide a general background or overview of a chemical or toxicological mechanism. |
| Meta-Analysis | A statistical technique used within a systematic review to combine the results of quantitative studies [71]. | Aims for exhaustive searching as part of a systematic review [71]. | Quality assessment may determine inclusion/exclusion and/or sensitivity analyses [71]. | Graphical and tabular with narrative commentary; provides a quantitative consensus [71]. | Statistically derive a more precise effect size for a specific health outcome from multiple chemical exposure studies. |
SEMs are not intended to replace the deep evidence synthesis of a full systematic review but rather to act as a critical precursor. They provide an evidence-based approach to characterising the extent of available evidence, supporting trend analysis, and facilitating the identification of decision-critical information that warrants further analysis via SR [64]. This makes them exceptionally valuable for large-scale regulatory initiatives like EU REACH and US TSCA, where resource efficiency and transparency are crucial [15].
The following section outlines a detailed, step-by-step protocol for constructing a Systematic Evidence Map, incorporating resource-efficient strategies at each stage.
Objective: To define the scope of the SEM and establish a transparent, pre-defined protocol.
Objective: To systematically identify and screen all relevant evidence for inclusion in the map.
Diagram 1: Evidence flow for systematic map creation.
Objective: To populate the evidence database with standardized information from each included study.
Objective: To analyze the coded database, generate visualizations that characterize the evidence base, and report findings.
Table 2: Hypothetical Evidence Matrix for Hepatotoxicity of Compound X This matrix visualizes the volume of available evidence, helping to identify data-rich areas suitable for systematic review and data-poor areas representing research gaps.
| Test System | Transcriptomic Alterations | Oxidative Stress | Apoptosis/Necrosis | Steatosis | Fibrosis |
|---|---|---|---|---|---|
| HepG2 Cell Line | 15 studies | 8 studies | 5 studies | 2 studies | 0 studies |
| Primary Human Hepatocytes | 5 studies | 3 studies | 2 studies | 1 study | 0 studies |
| Mouse (in vivo) | 10 studies | 12 studies | 8 studies | 6 studies | 1 study |
| Rat (in vivo) | 12 studies | 15 studies | 10 studies | 9 studies | 3 studies |
SEMs show strong potential in supporting the development and application of Adverse Outcome Pathways (AOPs) and New Approach Methodologies (NAMs) [72]. An AOP is a structured representation of biological events, starting from a Molecular Initiating Event (MIE) to an Adverse Outcome (AO) at the organism level. SEMs can be strategically deployed to systematically gather and map existing literature to an AOP framework.
Diagram 2: SEMs informing AOP development and assessment.
Protocol for Literature-Based AOP Development Using an SEM [72]:
This integration allows for a more transparent and evidence-based construction of AOPs, which are critical for leveraging NAMs in chemical risk assessment, ultimately reducing reliance on traditional animal studies.
Table 3: Key Research Reagent Solutions for Evidence Synthesis Projects
| Tool/Resource | Function | Example Applications in SEM/SR |
|---|---|---|
| Reference Management Software (EndNote, Zotero) | Manages bibliographic data, and PDFs, and assists in deduplication. | Storing search results, removing duplicate records from multiple databases. |
| Systematic Review Platforms (Rayyan, Covidence, DistillerSR) | Web-based tools designed to manage the screening and data extraction phases of a review. | Facilitating blinded title/abstract and full-text screening by multiple reviewers; data extraction form creation and management. |
| Bibliographic Databases (PubMed, Scopus, Embase) | Primary sources for identifying published scientific literature. | Executing comprehensive, pre-defined search strategies to ensure all relevant evidence is captured. |
| Grey Literature Sources (ClinicalTrials.gov, EPA reports, ECHA database) | Sources of unpublished or non-commercially published information. | Identifying ongoing studies, regulatory data, and other evidence not found in traditional journals, reducing publication bias. |
| Data Visualization Software (Tableau, R/ggplot2, Python/Matplotlib) | Creates sophisticated static and interactive visualizations. | Generating evidence heatmaps (like Table 2), interactive flowcharts, and other graphs to characterize the evidence base. |
| Grading of Recommendations Assessment, Development and Evaluation (GRADE) Framework | A structured methodology for assessing the certainty of a body of evidence. | Assessing confidence in Key Event Relationships (KERs) within an AOP developed from the SEM [72]. |
| Text Mining & Machine Learning Tools (NCBI's PubTator, custom NLP scripts) | Automates the identification of key concepts and relationships in large volumes of text. | Accelerating the initial screening phase or automatically extracting specific entities (e.g., chemical names, endpoints) during data extraction [72]. |
In the context of systematic evidence maps for chemical assessment research, ontologies serve as formal, machine-readable frameworks that explicitly define concepts, relationships, and rules within a domain [73]. They provide a standardized approach to representing complex data, enabling precise semantic meaning and logical inference that traditional data structures cannot capture [74]. For researchers, scientists, and drug development professionals, ontologies address critical challenges in data integration, interoperability, and long-term knowledge preservation, particularly when aggregating evidence across multiple studies, methodologies, and data sources.
The fundamental role of ontologies is to create a unified semantic layer that allows both humans and machines to interpret data consistently over time. This is achieved through explicit specification of conceptualizations, where entities are defined with precise relationships and logical constraints [75]. In chemical assessment research, this capability is paramount for maintaining data utility as analytical techniques evolve and regulatory requirements change. The application of ontologies ensures that data collected today remains discoverable, interpretable, and reusable for future evidence synthesis and meta-analyses.
Chemical assessment research generates data characterized by multiple types of heterogeneity: syntactic (differences in representation format), structural (variations in data models), and most critically, semantic heterogeneity (differences in data interpretation) [76]. Ontologies specifically address semantic heterogeneity by providing unambiguous identification of entities and their relationships across heterogeneous information systems [76]. In systematic evidence mapping, this enables accurate cross-study comparisons and reliable evidence synthesis.
For drug development professionals, ontologies facilitate content explication by making explicit the definitions of terms and relationships, serving as a global query model for formulating complex research questions across distributed datasets, and providing verification mechanisms to validate data mappings and integration logic [76]. This tripartite functionality ensures that evidence maps maintain semantic consistency even as new studies are incorporated over time.
Ontologies are foundational enablers of the FAIR Data Principles (Findable, Accessible, Interoperable, and Reusable) in chemical research [74] [77]. By providing machine-readable semantic context, ontologies transform research data from mere collections of numbers and observations into meaningful, actionable knowledge assets. The NFDI4Chem initiative emphasizes that FAIR data is fundamentally about creating both human and machine-readable data, with ontologies being essential building blocks for achieving this dual capability [77].
In practical terms, ontological annotation makes data findable through precise semantic tagging, accessible through standardized query interfaces, interoperable through shared conceptual frameworks, and reusable through explicit documentation of experimental context and meaning [73]. This comprehensive FAIR alignment is particularly valuable for systematic evidence maps in chemical assessment, where long-term utility depends on maintaining these characteristics through multiple research cycles and technological changes.
The OntoSpecies ontology represents a comprehensive implementation specifically designed for chemical species data management [74]. This ontology serves as a core component of The World Avatar knowledge graph chemistry domain and includes extensive coverage of chemical identifiers, physical and chemical properties, classifications, applications, and spectral information [74]. The implementation demonstrates how ontologies can integrate disparate chemical data sources into a unified semantic framework.
Key features of OntoSpecies include:
For chemical assessment researchers, this approach enables novel types of problemsolving, such as identifying compounds with specific property combinations or predicting chemical behavior through semantic reasoning rather than simple data retrieval.
Table 1: Ontology Integration Architectures for Chemical Data
| Approach | Description | Use Cases | Examples |
|---|---|---|---|
| Single Ontology | Uses one global reference model for integration | Homogeneous domains with standardized terminology | SIMS, Research Cyc [76] |
| Multiple Ontologies | Independent ontologies for each source with mappings between them | Integrating pre-existing, heterogeneous data sources | OBSERVER system [76] |
| Hybrid Approach | Multiple ontologies subscribing to a common top-level vocabulary | Evolving systems with specialized subdomains | Many OBO Foundry ontologies [76] [73] |
The hybrid approach has gained significant traction in chemical assessment research because it balances domain specificity with interoperability. In this model, specialized chemical ontologies (e.g., ChEBI for chemical entities, CHMO for chemical methods) align with upper-level ontologies like BFO (Basic Formal Ontology) and share relationship definitions from RO (Relation Ontology) [73]. This approach allows researchers to maintain domain-specific conceptualizations while ensuring cross-domain query capability.
Objective: Implement an ontology-driven framework for creating and maintaining systematic evidence maps in chemical assessment research.
Materials and Reagents:
Procedure:
Domain Analysis and Ontology Selection
Ontology Alignment and Extension
Data Annotation Pipeline
Query and Reasoning Infrastructure
Maintenance and Evolution
Objective: Transform traditional chemical research data into FAIR-compliant semantic data using ontological annotation.
Procedure:
Data Source Identification
Ontology Mapping Specification
RDF Generation and Validation
Knowledge Graph Population
Application Integration
Diagram 1: Ontology Data Integration Workflow
Diagram 2: Ontology Architecture for Evidence Mapping
Table 2: Essential Ontologies for Chemical Assessment Research
| Ontology | Scope | License | Key Classes & Properties | Use in Evidence Mapping |
|---|---|---|---|---|
| ChEBI [73] | Chemical entities of biological interest | CC-BY 4.0 | Chemical substances, roles, structures | Chemical entity standardization |
| CHMO [73] | Chemical methods and techniques | CC-BY 4.0 | Analytical methods, protocols | Experimental method annotation |
| OntoSpecies [74] | Comprehensive chemical properties | Custom OA | Identifiers, properties, spectra | Chemical property integration |
| BFO [73] | Upper-level ontology | CC-BY 4.0 | Universal classes (entity, process) | Cross-domain interoperability |
| OBI [73] | Biomedical investigations | CC-BY 4.0 | Assays, instruments, objectives | Study design annotation |
| IAO [73] | Information artifacts | CC-BY 4.0 | Data items, documents | Evidence provenance tracking |
| PATO [73] | Phenotypic qualities | CC-BY 3.0 | Qualities, characteristics | Endpoint standardization |
| UO [73] | Measurement units | CC-BY 4.0 | SI and derived units | Unit consistency across studies |
Table 3: Essential Tools for Ontology Implementation
| Tool Category | Specific Solutions | Function | Application Context |
|---|---|---|---|
| Ontology Editors | Protégé, WebProtégé | Ontology development and maintenance | Creating domain extensions and mappings |
| Triple Stores | Apache Jena, Stardog, GraphDB | RDF storage and SPARQL query processing | Evidence knowledge graph implementation |
| Reasoning Engines | HermiT, Pellet, ELK | Logical inference and consistency checking | Deriving implicit knowledge from evidence |
| Mapping Tools | RMLMapper, XSLT | Transforming structured data to RDF | Converting existing datasets to semantic format |
| Workbenches | KNIME, Orange with semantic extensions | Visual workflow design with ontology support | Designing evidence processing pipelines |
| Query Interfaces | YASGUI, SPARQL Explorer | User-friendly SPARQL query formulation | Enabling domain expert access to evidence |
Successful ontology implementation begins with deep stakeholder engagement to ensure the resulting semantic framework addresses real-world evidence mapping needs [78]. This involves collaborative requirement gathering with researchers, systematic review authors, regulatory scientists, and data managers to capture the nuanced information relationships essential for chemical assessment. The practice of "seeing through stakeholders' eyes" ensures ontologies reflect actual research workflows rather than abstract data models, significantly enhancing long-term adoption and utility [78].
Ontology development must be grounded in scientific principles and align with established domain standards to ensure logical consistency and interoperability [78]. In chemical assessment research, this means leveraging well-established ontologies like ChEBI and BFO rather than developing isolated models. This standards-based approach reduces ambiguity and enhances information system interoperability across the evidence lifecycle [78]. The alignment with upper ontologies like BFO further enables integration with complementary research domains, extending the utility of evidence maps beyond immediate chemical assessment applications.
Incorporating real-world data from the outset ensures ontological frameworks are grounded in practical evidence mapping requirements rather than theoretical constructs [78]. This involves analyzing existing datasets, evidence synthesis reports, and data exchange patterns to identify essential concepts, relationships, and constraints. This empirical foundation makes the ontology more relevant and applicable to ongoing chemical assessment activities, increasing stakeholder adoption and long-term sustainability.
Establishing robust governance and maintenance procedures is critical for sustaining ontology utility as evidence mapping requirements evolve [79]. This includes versioning strategies, change management processes, and community engagement mechanisms to ensure the ontological framework adapts to emerging research questions and methodological advances. Regular reviews against evolving standards and practical implementation feedback create a continuous improvement cycle that maintains ontological relevance throughout the evidence map lifecycle.
In the field of chemical assessment research, the ability to navigate vast scientific literature is paramount. Systematic Evidence Maps (SEMs) and Systematic Reviews (SRs) represent two distinct methodologies for evidence synthesis, each with unique purposes, processes, and outputs. While both employ rigorous systematic approaches, they serve different functions in the research ecosystem. SEMs provide a broad overview of the research landscape, identifying the existence, distribution, and characteristics of available evidence, whereas SRs focus on obtaining definitive answers to specific research questions, typically regarding the effectiveness or safety of interventions or exposures. For researchers, scientists, and drug development professionals, understanding the strategic application of each method ensures appropriate use of resources and generates the most relevant evidence for decision-making processes in chemical risk assessment and regulatory submissions [80] [81].
The following table delineates the core distinctions between these two methodologies, highlighting their unique contributions to evidence-based chemical assessment.
Table 1: Comparative Analysis of Systematic Evidence Maps and Systematic Reviews
| Feature | Systematic Evidence Maps (SEMs) | Systematic Reviews (SRs) |
|---|---|---|
| Primary Purpose | To systematically catalog and map the available evidence, describing the breadth and nature of a research field [3] [81]. | To comprehensively synthesize and analyze evidence to answer a specific, focused question, often about intervention effectiveness or safety [80] [3]. |
| Research Question | Broad in scope, aiming to "map the landscape" of a topic [80] [81]. | Narrow and specific, often formulated using PICO/PECO (Population, Intervention/Exposure, Comparator, Outcome) criteria [80]. |
| Data Extraction | Categorizes high-level study characteristics (e.g., study design, population, exposure type, outcome measured) [80] [81]. | Extracts detailed data on methods, results, and risk of bias to support a synthesized conclusion [80] [3]. |
| Quality Appraisal (Risk of Bias) | Often optional. If conducted, it is used to characterize the evidence base rather than exclude studies [80]. | Mandatory. Critical for interpreting findings and grading the overall strength of evidence [3]. |
| Key Outputs | Interactive databases, evidence gap maps (EGMs), graphical charts, and reports highlighting evidence clusters and gaps [80] [3] [21]. | A synthesized summary of findings (narrative, tabular, or statistical meta-analysis) with conclusions about effects [3] [82]. |
| Role in Chemical Assessment | Ideal for initial chemical prioritization, scoping health outcomes for a substance, and planning future primary research or systematic reviews [83]. | Used to support definitive hazard identification, dose-response assessment, and inform risk management decisions [10]. |
The following protocol, summarized in Table 2, provides a standardized workflow for developing a Systematic Evidence Map, drawing from established methodologies in environmental and human health research [80] [83].
Table 2: Protocol for a Systematic Evidence Map
| Step | Description | Application in Chemical Assessment |
|---|---|---|
| 1. Define Scope & Question | Formulate a broad research question and objectives. Define PECO elements. | Example: "Map the available mechanistic evidence informing the health outcomes associated with Perfluorohexanesulfonic Acid (PFHxS)" [83]. |
| 2. Develop a Protocol | Create a detailed, publicly available protocol outlining the methodology, including search strategy and inclusion/exclusion criteria [80] [10]. | The protocol ensures transparency and reproducibility, critical for regulatory acceptance [10]. |
| 3. Literature Search | Perform a comprehensive search across multiple bibliographic databases and grey literature sources [80]. | Search databases like PubMed, Scopus, and TOXLINE using chemical names and broad health outcome terms. |
| 4. Study Screening | Screen identified studies for relevance using pre-defined inclusion/exclusion criteria, typically in a two-stage process (title/abstract, then full-text) [80]. | Use software like DistillerSR to manage the screening process for thousands of identified studies [83]. |
| 5. Data Extraction | Extract high-level data into a predefined coding framework. | Extract data on study design (e.g., in vitro, in silico), organism, system, endpoint, and health outcome category (e.g., hepatic, immune) [80] [83]. |
| 6. Data Coding & Categorization | Code and categorize the extracted data according to the framework. | Categories may include health effects like thyroid, immune, developmental, and hepatic toxicity [83]. |
| 7. Visual Presentation | Develop visualizations to present the mapped evidence, such as Evidence Gap Maps (EGMs). | Create interactive maps or heat maps showing the volume of evidence for different health outcomes, highlighting data-rich and data-poor areas [80] [21]. |
This protocol, used by agencies like the U.S. EPA for chemical risk evaluations under TSCA, involves a more in-depth process focused on deriving a quantitative or qualitative conclusion [10].
Table 3: Protocol for a Systematic Review in Chemical Risk Assessment
| Step | Description | Application in Chemical Risk Evaluation |
|---|---|---|
| 1. Formulate Specific Question | Define a focused question using PECO/PICO. | Example: "Does chronic oral exposure to substance X cause liver toxicity in rodents?" |
| 2. Develop & Peer-Review Protocol | Draft a detailed protocol, often subject to peer review by advisory committees [10]. | The EPA's TSCA protocol is reviewed by the Science Advisory Committee on Chemicals (SACC) [10]. |
| 3. Comprehensive Search | Conduct an exhaustive search with a highly sensitive strategy to capture all relevant evidence. | Similar to SEMs but may be more targeted to specific health outcomes and study designs suitable for risk assessment. |
| 4. Study Screening | Screen studies against strict eligibility criteria. | Focus on identifying studies that report quantitative data on the specific exposure-outcome relationship. |
| 5. Detailed Data Extraction | Extract detailed data on study methods, results, and potential confounding factors. | Extract specific data points such as dose, response, incidence, effect size, and statistical measures. |
| 6. Risk of Bias Assessment | Critically appraise the internal validity of each study using a validated tool [10]. | Use tools designed for animal toxicology or observational studies to evaluate confidence in each study's results. |
| 7. Synthesis & Meta-Analysis | Synthesize findings narratively and, if appropriate, statistically via meta-analysis. | Combine results from studies to estimate an overall effect and explore heterogeneity. |
| 8. Report & Conclude | Report findings with a conclusion on the strength of evidence for the health outcome. | Outputs directly inform the hazard identification and dose-response analysis in a risk assessment [10]. |
The U.S. Environmental Protection Agency (EPA) employed a Systematic Evidence Map to organize and evaluate the mechanistic data for Perfluorohexanesulfonic Acid (PFHxS). The goal was to identify the available evidence and pinpoint research needs linking PFHxS to specific health outcomes [83].
Experimental Workflow & Key Findings:
This SEM provided a rapid and clear "big picture" of the evidence, allowing researchers and regulators to see where evidence was concentrated and where it was absent. This directly informs priorities for future research to substantiate potential hazards identified by other evidence streams, such as epidemiology [83].
Table 4: Key Research Reagent Solutions for Evidence Synthesis
| Reagent / Tool | Function in Evidence Synthesis |
|---|---|
| DistillerSR | A web-based, systematic review software used to manage the entire literature screening and data extraction process, ensuring compliance and reducing human error [83]. |
| EPPI-Reviewer | A specialized software tool for managing and analyzing data in all forms of systematic review, including coding and classification for mapping reviews [80]. |
| PECO/PICO Framework | A structured framework to define the Population, Exposure/Intervention, Comparator, and Outcome(s) of interest, forming the foundation of a focused research question [80] [83]. |
| Evidence Gap Map (EGM) | A graphical representation (often interactive) that visually plots the relationships between interventions/exposures and outcomes, showing the volume and distribution of available evidence [80] [81]. |
| Systematic Review Protocol | A detailed, a priori plan that defines the study's objectives and methods, crucial for minimizing bias and ensuring transparency and reproducibility [80] [10]. |
Systematic Evidence Maps and Systematic Reviews are complementary yet distinct tools in the chemical assessor's arsenal. The choice between them is not a matter of hierarchy but of strategic alignment with the research objective. SEMs are the optimal choice for scoping broad fields, identifying evidence clusters and gaps, and guiding research agendas. In contrast, SRs are indispensable for integrating detailed evidence to answer specific questions about chemical hazards and risks, thereby directly supporting regulatory decision-making. For a robust chemical assessment program, both methodologies are essential. Beginning with an SEM can efficiently scope the landscape and determine whether and where a full Systematic Review is warranted, ensuring that resources are allocated effectively to generate the most impactful evidence for protecting human health and the environment.
Within the domain of chemical assessment research, evidence synthesis serves as a cornerstone for informed decision-making. Systematic evidence maps provide a foundational overview of the research landscape, charting the extent and distribution of available evidence without aggregating results [84]. These maps function as crucial tools for identifying knowledge clusters and gaps, thereby informing future research agendas. Within this broader ecosystem, scoping reviews and rapid reviews represent two distinct but complementary methodological approaches to evidence synthesis. When a systematic evidence map reveals a sufficiently dense area of research, scoping reviews and rapid reviews offer pathways to delve deeper, each with specific applications, methodologies, and outputs tailored to different research objectives and time constraints. This article provides a detailed comparative analysis and protocols for these two review types, contextualized specifically for researchers, scientists, and professionals in drug development and chemical assessment.
A scoping review aims to map the key concepts, types of evidence, and research gaps on a particular topic [84]. Its primary purpose is to provide an overview of the existing literature regardless of the quality of the included studies. Scoping reviews are particularly valuable in chemical assessment for examining emerging evidence, clarifying working definitions for exposure metrics or outcome measures, and identifying the scope and nature of available research on a particular compound or class of compounds. They are characterized by broad questions and comprehensive search strategies and may not be accompanied by a formal quality assessment of the included studies.
A rapid review is a form of knowledge synthesis in which components of the systematic review process are simplified or omitted to produce information in a timely manner [85]. These reviews are conducted to meet the urgent evidence needs of decision-makers, such as those in regulatory agencies or clinical development teams, who cannot wait for the lengthy timeline of a full systematic review. While numerous approaches exist, they consistently prioritize timeliness, with conduct times ranging from less than one month to twelve months, though many are completed between one and six months [85].
The selection between a scoping review and a rapid review is dictated by the research question. The table below summarizes the core characteristics of each review type for direct comparison.
Table 1: Comparative Analysis of Scoping Reviews and Rapid Reviews
| Characteristic | Scoping Review | Rapid Review |
|---|---|---|
| Primary Objective | To map key concepts, evidence types, and gaps in a broad field [84]. | To provide timely evidence for decision-making by streamlining systematic review methods [85]. |
| Research Question | Broad, exploratory (e.g., "What is the scope of research on the neurodevelopmental effects of chemical X?"). | Focused, often on intervention efficacy or safety (e.g., "Is drug Y effective for condition Z?") [84]. |
| Scope | Comprehensively covers a topic area, often with heterogeneous evidence. | Narrower, with boundaries set to expedite the process. |
| Search Strategy | Comprehensive, seeks to be extensive, multiple databases. | Streamlined (e.g., limited by date, fewer databases, no grey literature) [85]. |
| Study Selection | Typically involves screening by two reviewers, but may be flexible. | Often streamlined (e.g., single screener with verification, or screen of excluded studies) [85]. |
| Critical Appraisal | Usually not performed [84]. | Often omitted or conducted by a single reviewer [85]. |
| Data Synthesis | Presentation of a narrative summary, often with quantitative and qualitative categorization. | Primarily narrative summary; quantitative synthesis (meta-analysis) is rare [85]. |
| Key Outputs | Conceptual map, evidence inventory, identification of research gaps. | Timely summary of evidence, often with conclusions on a specific question. |
| Timeframe | Can be lengthy due to broad scope and volume of evidence. | 1 to 12 months, commonly 1-6 months [85]. |
| Reporting Guideline | PRISMA-ScR (Preferred Reporting Items for Systematic reviews and Meta-Analyses extension for Scoping Reviews) [84]. | No universal standard; often adapts PRISMA. |
Scoping reviews are ideal for initial assessments of chemical exposures and health outcomes where the literature is diverse and not yet comprehensively cataloged.
Phase 1: Planning and Protocol Development
Phase 2: Searching and Selecting Evidence
Phase 3: Data Extraction and Charting
Phase 4: Analysis and Presentation of Results
The workflow for this protocol is illustrated in the following diagram:
Rapid reviews are suited for urgent questions in drug development, such as a preliminary safety assessment of an excipient or a comparative efficacy review for a grant application.
Phase 1: Pragmatic Scoping and Streamlining
Phase 2: Targeted Evidence Retrieval
Phase 3: Focused Data Extraction and Appraisal
Phase 4: Expedited Synthesis and Reporting
The following diagram outlines the rapid review workflow with key streamlining decision points:
Table 2: Common Streamlining Methods in Rapid Reviews (based on [85])
| Methodological Step | Common Streamlining Approach | Reported Frequency in Literature |
|---|---|---|
| Literature Search | Limit by date (e.g., last 5-10 years) | 68% |
| Limit by language (e.g., English only) | 49% | |
| Search published literature only (no grey literature) | 24% | |
| Search only one database | 2% | |
| Study Selection | Single reviewer screen, with verification of excluded studies | 6% |
| Data Extraction | Single reviewer extract, with second reviewer verification | 23% |
| Quality Appraisal | Omit risk of bias/quality appraisal | 7% |
| Single reviewer conducts quality appraisal | 7% | |
| Data Synthesis | Present results as a narrative summary (no meta-analysis) | 78% |
Executing a robust review requires a suite of "methodological reagents" â standardized tools and resources that ensure rigor, reproducibility, and efficiency.
Table 3: Key Research Reagent Solutions for Evidence Synthesis
| Tool/Resource | Function | Application Notes |
|---|---|---|
| PRISMA-ScR Checklist | Reporting guideline for scoping reviews. Ensures transparent and complete reporting of the review process [84]. | Essential for manuscript preparation and peer review. |
| PCC Framework | (Population, Concept, Context) used to define the scope and question for a scoping review. | Provides a more flexible alternative to the PICO framework for broad questions. |
| Rayyan / Covidence | Web-based tools for managing the study screening and selection process. | Facilitates blinding of reviewers, conflict resolution, and progress tracking. |
| Systematic Review Repository | Platforms like PROSPERO for pre-registering review protocols. | Reduces duplication of effort and mitigates reporting bias. Mandatory for many funders. |
| JBI Sumari | Software platform supporting the entire systematic review process, including scoping and rapid reviews. | Supports development of protocols, data extraction, critical appraisal, and synthesis. |
| Automated Screening Tools | AI-based tools (e.g., ASReview, RobotAnalyst) that prioritize records during title/abstract screening. | Particularly valuable in rapid reviews and scoping reviews with large search yields to reduce screening workload. |
| Data Visualization Software | Tools like Tableau, R/ggplot2, or even PowerPoint to create evidence maps and summary figures. | Critical for translating the results of a scoping review into an accessible format for stakeholders. |
Scoping reviews and rapid reviews are powerful, distinct tools within the evidence synthesis toolkit for chemical assessment and drug development. Scoping reviews provide the broad, conceptual map needed to understand a sprawling research landscape, while rapid reviews deliver timely, focused evidence to meet pressing decision-making deadlines. The choice between them is not one of superiority but of strategic alignment with the research objective, available resources, and intended application. By adhering to the structured protocols and utilizing the essential tools outlined in this article, researchers can rigorously apply these methodologies to generate impactful, reliable evidence to advance the field.
In the face of expanding chemical inventories and limitations of traditional animal testing, regulatory toxicology increasingly relies on New Approach Methodologies (NAMs) to fill critical data gaps. Systematic evidence maps play a pivotal role in organizing existing knowledge and identifying priority areas for assessment [12]. This application note demonstrates the practical implementation of expert-driven read-across, a prominent NAM, through a case study adapted from US EPA practices. The documented protocol provides a framework for using structural, toxicokinetic, and toxicodynamic similarity to derive screening-level risk values for data-poor substances, leveraging existing assessments from robust programs like the EPA's Integrated Risk Information System (IRIS) and ATSDR's Toxicological Profiles.
This protocol provides a standardized methodology for identifying a suitable source analogue and applying its point of departure (POD) to a target data-poor chemical via quantitative read-across. It is applicable within human health risk assessment for environmental contaminants where oral toxicity data are insufficient, framed within the context of systematic evidence mapping for chemical assessment [12] [86].
Table 1: Essential Research Materials and Databases
| Item Name | Function/Application | Specifications/Requirements |
|---|---|---|
| ChemIDplus Database | Identifies structurally similar chemicals based on fingerprint and Tanimoto coefficient. | Similarity threshold typically ⥠50%; National Library of Medicine resource [86]. |
| DSSTox Database | Identifies structurally similar chemicals with existing risk assessment data. | Use "IRIS_v1b" search option; U.S. EPA resource [86]. |
| ToxCast/Tox21 Database | Provides high-throughput screening (HTS) bioactivity data for evaluating toxicodynamic similarity. | Contains >1,800 chemicals screened in >700 assay endpoints; U.S. EPA resource [86]. |
| IRIS and PPRTV Databases | Sources for verified toxicity data and Points of Departure (PODs) for candidate analogues. | U.S. EPA's official toxicity value databases [86]. |
| ATSDR Toxicological Profiles | Sources for verified toxicity data and Minimal Risk Levels (MRLs) for candidate analogues. | Profiles provide comprehensive toxicological summaries [86]. |
Evaluate the target and each candidate analogue across three primary similarity contexts. Document all evidence and justifications.
Structural & Physicochemical Similarity:
Toxicokinetic (TK) Similarity:
Toxicodynamic (TD) Similarity:
For reproducibility and regulatory acceptance, report the following in a structured format [18] [88]:
The following tables summarize the types of data collected and compared during the read-across process for the p,p'-DDD case study.
Table 2: Analogue Candidate List with Structural and Toxicological Data
| Chemical Name | Tanimoto Similarity to p,p'-DDD | Available Oral POD (mg/kg-day) | Critical Effect | Metabolic Pathway Similarity |
|---|---|---|---|---|
| p,p'-DDT (Source Analogue) | >80% | NOAEL = 0.05 | Liver hypertrophy, neurotoxicity | High (e.g., dehydrochlorination to DDE) |
| p,p'-DDE | >80% | NOAEL = 0.05 (ATSDR) | Liver effects, developmental | Moderate |
| Methoxychlor | ~70% | NOAEL = 5.0 (PPRTV) | Liver, kidney, ovarian effects | Low (different primary metabolism) |
Table 3: ToxCast Bioactivity Profile Comparison (Illustrative Data)
| Assay Endpoint / Target | p,p'-DDD Activity (AC50, µM) | p,p'-DDT Activity (AC50, µM) | p,p'-DDE Activity (AC50, µM) | Similarity Inference |
|---|---|---|---|---|
| ERα Agonism | 2.5 | 1.8 | 0.9 | High (comparable potency) |
| AR Antagonism | 5.1 | 3.5 | 15.0 | Moderate (DDT & DDD more similar) |
| Thyroid Receptor Agonism | Inactive | Inactive | Inactive | High (same lack of effect) |
This application note details a robust protocol for implementing expert-driven read-across, demonstrating how systematic methodologies and NAMs can be validated through application in a regulatory context. The integration of systematic evidence mapping with quantitative read-across provides a transparent and defensible approach to addressing data gaps for chemicals lacking full toxicological profiles. The case study on p,p'-DDD illustrates the critical role of toxicokinetic and high-throughput screening data in building scientific confidence for the use of read-across in generating screening-level health reference values, thereby supporting the mission of programs like EPA IRIS and ATSDR to protect public health.
Systematic Evidence Maps (SEMs) have emerged as a critical tool in evidence-based toxicology and chemical risk assessment, offering a solution to the challenge of navigating large and complex evidence bases [66] [64]. While systematic reviews provide deep synthesis of narrowly focused questions, SEMs provide a broader overview of research landscapes, characterizing the extent, distribution, and gaps in available evidence [81]. This methodology is particularly valuable for chemicals policy and risk management workflows where decision-makers face broad information needs that cannot be addressed by single systematic reviews [64]. The analysis of 39 published evidence maps provides a robust foundation for establishing methodological best practices, ensuring that SEMs produced for chemical assessment research are rigorous, transparent, and fit-for-purpose.
A systematic review of 39 published evidence maps revealed significant diversity in how the methodology was applied and described across different research contexts [89]. This analysis provides crucial empirical data to inform best practices in chemical assessment research.
Table 1: Characteristics of 39 Published Evidence Maps
| Characteristic | Findings | Frequency |
|---|---|---|
| Stated Purpose | Identification of research gaps | 67% (31/39) |
| Stakeholder engagement process or user-friendly product | 58% (31/39) | |
| Methodological Approach | Explicitly systematic approach | 100% (39/39) |
| Map Presentation Format | Figure or table explicitly called "evidence map" | 67% (26/39) |
| Online database as the evidence map | 21% (8/39) | |
| Mapping methodology without visual depiction | 13% (5/39) | |
| Geographical Origin | United States | 49% (19/39) |
| Australia | 26% (10/39) | |
| United Kingdom | Remaining publications |
The analysis found that all evidence maps explicitly used a systematic approach to evidence synthesis, indicating consensus on the fundamental requirement for methodological rigor [89]. However, heterogeneity was observed in how evidence maps were presented and formatted, suggesting ongoing evolution in how the methodology is communicated to end-users.
Understanding how SEMs complement other evidence synthesis methodologies is essential for appropriate application in chemical assessment research.
Table 2: Comparison of Evidence Synthesis Methodologies for Chemical Assessment
| Methodology | Systematic Review | Systematic Evidence Map | Scoping Review |
|---|---|---|---|
| Primary Purpose | Answer specific focused question through synthesis | Identify, characterize, and organize broad evidence base | Explore key concepts and evidence types in a field |
| Scope | Narrow and specific | Broad | Broad or exploratory |
| Time/Resources | High | Medium to High | Variable |
| Output | Quantitative or qualitative synthesis | Queryable database, visualizations, gap analysis | Descriptive narrative, conceptual framework |
| Ideal Use Case | Determining chemical safety for specific health endpoint | Priority-setting for chemical assessment programs | Understanding conceptual boundaries of research area |
SEMs occupy a distinct space in the evidence synthesis ecosystem, particularly valuable for "forward looking predictions or trendspotting in the chemical risk sciences" and as "a critical precursor to efficient deployment of high quality systematic review methods" [64].
The following protocol provides a detailed methodology for conducting SEMs specific to chemical assessment research, synthesized from analyzed publications and methodological guidance.
Systematic Evidence Mapping Workflow for Chemical Assessment
Knowledge Graph Data Structure for Evidence Maps
Table 3: Essential Tools and Resources for Chemical Evidence Mapping
| Tool Category | Specific Solutions | Function in Evidence Mapping |
|---|---|---|
| Bibliographic Databases | PubMed, Web of Science, Embase, TOXLINE | Comprehensive identification of toxicological literature |
| Chemical Registries | CAS Registry, DSSTox, CompTox Chemicals Dashboard | Chemical identification, standardization, and classification |
| Data Extraction Tools | Systematic Review Assistant (SRA), CADIMA, DistillerSR | Streamline screening and data extraction processes |
| Data Storage Technologies | Graph databases (Neo4j), SQL databases, Spreadsheets | Structured storage of extracted evidence with query capabilities |
| Controlled Vocabularies | CHEBI, MeSH, OBO Foundry ontologies | Standardized coding of chemical, biological, and methodological concepts |
| Visualization Platforms | Tableau, R Shiny, EPPI-Mapper | Create interactive evidence maps and gap visualizations |
The systematic analysis of 39 evidence maps reveals several critical applications in chemical assessment research. SEMs provide an evidence-based approach to characterizing available evidence and supporting "forward looking predictions or trendspotting in the chemical risk sciences" [64]. They facilitate identification of related bodies of decision-critical chemical risk information that could be further analyzed using systematic review methods and highlight evidence gaps that could be addressed with additional primary studies.
In practice, evidence maps have been successfully applied to address specific chemical assessment challenges. For example, a recent evidence map on human toxicodynamic variability identified 23 in vitro studies providing quantitative estimates of variability factors, revealing significant data scarcity and heterogeneity that complicates chemical risk assessment [90]. This application demonstrates how SEMs can characterize the nature and extent of available evidence on specific toxicological questions relevant to refining uncertainty factors and chemical-specific assessment factors.
The transition toward knowledge graphs as a flexible, schemaless, and scalable model for systematically mapping environmental health literature represents a significant methodological advancement [66]. This approach is particularly well-suited to the long-term goals of systematic mapping methodology in promoting resource-efficient access to the wider environmental health evidence base, with several graph storage implementations readily available and proven use cases in other fields.
The analysis of 39 published evidence maps provides a robust foundation for establishing best practices in chemical assessment research. Successful implementation requires careful attention to problem formulation, systematic search strategies, dual screening processes, standardized data extraction with controlled vocabulary coding, and flexible knowledge structures that can accommodate the highly connected nature of toxicological evidence. By adopting these evidence-based methodologies, researchers and chemical assessment professionals can produce SEMs that effectively support priority-setting, trend identification, and resource-efficient evidence-based decision-making in chemicals policy and risk management.
Modern chemical regulations like the EU's REACH (Registration, Evaluation, Authorisation and Restriction of Chemicals) and the U.S. TSCA (Toxic Substances Control Act) face a formidable challenge: efficiently evaluating thousands of chemicals using the best available science without being overwhelmed by the exponentially growing volume of research data [4]. Regulatory bodies need to make timely, transparent, and evidence-based decisions on chemical safety, often with limited resources [4] [64]. Systematic Evidence Maps (SEMs) have emerged as a powerful methodology to address this challenge by providing a structured, comprehensive, and queryable overview of broad evidence bases [4] [64] [66]. Unlike systematic reviews, which synthesize evidence to answer a specific, narrow question, SEMs characterize the extent and nature of available research, highlighting evidence clusters and critical gaps [4] [66]. This application note details how SEMs function as critical tools for enhancing the resource efficiency, transparency, and effectiveness of REACH and TSCA regulatory initiatives.
A Systematic Evidence Map (SEM) is a queryable database of systematically gathered research evidence, developed following a rigorous methodology to minimize bias and maximize transparency [4] [66]. Their primary purpose in regulatory science is to organize and characterize a large body of policy-relevant research, enabling trend analysis, priority-setting, and informed decision-making [4] [64]. They are not a substitute for the detailed evidence synthesis of a systematic review but serve as a critical precursor, ensuring that subsequent deep-dive analyses are targeted efficiently [4]. SEMs provide a foundational resource for evidence-based problem formulation, allowing regulators to ask: "What is the scope of the existing evidence?" and "Where should limited assessment resources be focused?" [20].
The development of a robust SEM follows a structured, protocol-driven process. The U.S. EPA has established a template for creating SEMs for its Integrated Risk Information System (IRIS) and Provisional Peer Reviewed Toxicity Value (PPRTV) programs [20]. The workflow can be visualized as follows:
The key methodological steps are:
Problem Formulation and PECO Criteria: The process begins with defining the Populations, Exposures, Comparators, and Outcomes (PECO) criteria. For broad evidence mapping, the PECO is kept intentionally wide to capture a comprehensive range of studies [20]. For instance, a SEM on a chemical might aim to identify all mammalian animal bioassays and epidemiological studies relevant to human hazard identification.
Comprehensive Search and Screening: A systematic search is conducted across multiple scientific databases using a pre-defined search strategy. Standard systematic review practices, including the use of two independent reviewers per record and specialized software, are employed to screen titles, abstracts, and full texts against the eligibility criteria [4] [20]. Machine learning tools may be incorporated to facilitate this process.
Data Extraction and Coding: Data from included studies is extracted using structured forms. This involves cataloging key study characteristics (e.g., design, test system, health outcomes assessed) and coding them using controlled vocabularies. This step is crucial for transforming unstructured literature into a structured, queryable format [66]. The availability of New Approach Methodologies (NAMs) is also frequently tracked [20].
Database Creation and Visualization: The extracted data is stored in an interactive database. The data is then made accessible through interactive visualizations and can be downloaded in open-access formats, allowing end-users to explore the evidence base according to their specific interests [20].
Table 1: Key Research Reagent Solutions for Systematic Evidence Mapping
| Tool Category | Specific Tool/Software | Primary Function in SEM Development |
|---|---|---|
| Literature Databases | PubMed, Scopus, Web of Science | Identification of primary research studies through systematic search strategies [20]. |
| Systematic Review Software | DistillerSR, Rayyan, CADIMA | Facilitation of the literature screening process (title/abstract, full-text) by multiple independent reviewers [20]. |
| Data Extraction Tools | Custom web-based forms, Excel templates | Standardized extraction of predefined data and metadata from included studies into a structured format [20]. |
| Data Storage Solutions | SQL databases, Knowledge Graphs | Storage of extracted data in a flexible, queryable format. Knowledge graphs are particularly suited for complex, interconnected data [66]. |
| Visualization Platforms | Tableau, R Shiny, EviAtlas | Creation of interactive dashboards and maps to explore the evidence base visually and identify trends [20]. |
The REACH regulation requires companies to register chemical substances manufactured or imported in quantities exceeding one tonne per year, involving the evaluation of submitted information and management of substances of very high concern (SVHCs) [91]. The 2025 revisions to REACH emphasize digitalization, enhanced enforcement, and simplified processes, creating a greater need for efficient evidence management [92] [93]. SEMs directly support REACH through:
Under TSCA, the EPA must conduct risk evaluations for existing chemicals to determine whether they present an unreasonable risk to health or the environment under the conditions of use [94] [26]. The process is resource-intensive and must adhere to strict statutory timelines. SEMs augment the TSCA workflow by:
Table 2: Quantitative Impact of SEMs on Regulatory Efficiency
| Regulatory Challenge | Traditional Approach | SEM-Supported Approach | Impact of SEM |
|---|---|---|---|
| Problem Formulation | Relies on expert knowledge and limited literature reviews, potentially missing key studies. | Provides a comprehensive, transparent map of all evidence, revealing clusters and gaps. | Increases transparency, reduces bias, and provides an evidence-based foundation for scoping [4] [20]. |
| Chemical Prioritization | May be influenced by high-profile studies or political pressure rather than systematic evidence. | Enables quantitative comparison of evidence volume and focus across multiple chemicals. | Improves resource allocation by targeting chemicals with the strongest evidence of hazard [4]. |
| Data Gap Identification | Often discovered during the deep assessment phase, causing delays. | Highlighted at the outset via the evidence map. | Allows for proactive planning of testing strategies or assessment approaches, saving time [4] [64]. |
| Stakeholder Communication | Difficult to demonstrate the full body of considered evidence. | Interactive maps and visualizations provide a clear, accessible summary of the evidence landscape. | Builds trust and facilitates engagement by making the evidence base explorable [20]. |
The following diagram illustrates how SEMs are integrated into the broader chemical risk management workflows of REACH and TSCA, acting as a bridge between vast scientific literature and decisive regulatory action.
Systematic Evidence Maps represent a paradigm shift in how regulatory bodies can manage the deluge of scientific data in chemical risk assessment. By providing a structured, transparent, and queryable overview of broad evidence bases, SEMs directly support the core objectives of REACH and TSCA: to protect human health and the environment efficiently and based on the best available science. They enable resource-efficient priority setting, inform problem formulation, and ensure that subsequent, more resource-intensive assessments like systematic reviews are deployed where they are most needed. As chemical regulations continue to evolve, the adoption of SEMs will be crucial for building responsive, transparent, and evidence-based regulatory frameworks.
Systematic Evidence Maps represent a paradigm shift in chemical assessment, offering a robust, transparent, and resource-efficient methodology for navigating complex evidence landscapes. By providing comprehensive overviews of available research while precisely identifying critical knowledge gapsâas starkly revealed in assessments showing that over 98% of PFAS chemicals lack health effects dataâSEMs enable strategic prioritization in both research and regulatory decision-making. The integration of advanced technologies like knowledge graphs and machine learning promises to further enhance their scalability and utility. For biomedical and clinical research, the continued evolution and standardization of SEM methods will be crucial for addressing the growing challenges of chemical risk assessment, ultimately leading to more informed, evidence-based public health protections and more efficient drug development pipelines. Future directions should focus on developing standardized reporting guidelines, expanding cross-agency collaboration, and further integrating New Approach Methodologies (NAMs) into evidence mapping frameworks.