ICH M4 eCTD Structure: Complete Implementation Guide
Step-by-step guide to structuring regulatory submissions using the ICH Common Technical Document format for global regulatory authorities
Table of Contents
Introduction to ICH M4
The ICH M4 guideline defines the Common Technical Document (CTD), a standardized format for organizing regulatory submission data. The CTD harmonizes the structure and format of applications across regulatory authorities in the US (FDA), Europe (EMA), Japan (PMDA), and other ICH regions.
Why ICH M4 Matters
- β Global Consistency: Submit the same structure to multiple agencies
- β Reduced Rework: Modules 2-5 are identical across regions
- β Reviewer Efficiency: Regulators know exactly where to find information
- β Industry Standard: Required by major regulatory authorities worldwide
ICH M4 Family of Guidelines
Overall structure and organization of the five modules
Module 3 (Quality) detailed structure and content requirements
Module 4 (Nonclinical) detailed structure and content
Module 5 (Clinical) detailed structure and content
eCTD vs. CTD
πCTD (Paper-Based)
- β’ Physical binders and paper documents
- β’ Sequential page numbering
- β’ No hyperlinks or navigation
- β’ Difficult to update
- β’ Largely obsolete
πΎeCTD (Electronic)
- β’ Electronic files (PDFs, XML)
- β’ Hierarchical folder structure
- β’ Hyperlinked navigation via XML backbone
- β’ Easy updates via sequences
- β’ Current standard
Note: This guide focuses on eCTD format as it is now required by FDA, EMA, PMDA, Health Canada, and most major regulatory authorities. The structural content follows ICH M4, but the technical implementation follows eCTD specifications (currently v4.0).
The Five-Module Structure
The CTD is organized into five modules. Modules 2-5 are common across all ICH regions, while Module 1 is region-specific.
CTD Module Overview
Module 1: Administrative Information & Prescribing Information
Region-SpecificForms, labels, administrative documents (varies by region)
Module 2: Common Technical Document Summaries
CommonQuality, Nonclinical, and Clinical overviews and summaries
Module 3: Quality
CommonDrug substance, drug product, and quality data
Module 4: Nonclinical Study Reports
CommonPharmacology, pharmacokinetics, and toxicology
Module 5: Clinical Study Reports
CommonClinical pharmacology, efficacy, and safety studies
Document Flow & Dependencies
The modules are designed to be read in a specific order, building from high-level summaries to detailed data:
Module 1: Administrative Information
Region-Specific Content
Module 1 varies significantly by regulatory authority
Module 1 contains administrative information and prescribing information specific to each region. Unlike Modules 2-5, Module 1 is NOT harmonized across ICH regions.
Module 1 by Region
πΊπΈModule 1 for FDA (United States)
Key Documents (Section 1.0 - 1.14):
- β’ 1.0: Cover letters
- β’ 1.2: Administrative information (FDA Form 356h, application type)
- β’ 1.3.1: FDA Form 1571 (for INDs) or Form 356h (for NDAs/BLAs)
- β’ 1.3.2: Other forms (3674, 3454, 3455, patent certifications)
- β’ 1.4: Debarment certification
- β’ 1.5: Field copy certification
- β’ 1.6: Patent information and exclusivity
- β’ 1.8: Financial disclosure
- β’ 1.11: Draft labeling (package insert, patient labeling)
- β’ 1.12: Risk evaluation and mitigation strategies (REMS)
- β’ 1.14: Environmental assessment or categorical exclusion
πͺπΊModule 1 for EMA (European Union)
Key Documents (Section 1.0 - 1.8):
- β’ 1.0: Cover letter, application form
- β’ 1.1: Comprehensive table of contents
- β’ 1.2: Application forms (varies by procedure: centralized, MRP, DCP)
- β’ 1.3: Product information (SmPC, labeling, package leaflet)
- β’ 1.4: Information about experts
- β’ 1.5: Specific requirements (orphan designation, pediatric investigation plan)
- β’ 1.6: Environmental risk assessment
- β’ 1.8: Information from reference member state (for MRP/DCP)
π―π΅Module 1 for PMDA (Japan)
Key Documents:
- β’ Application forms specific to PMDA
- β’ Proposed Japanese package insert (ζ·»δ»ζζΈ)
- β’ Documentation of GMP/GCP/GLP compliance
- β’ Foreign regulatory status
- β’ Re-examination period application
- β’ Documentation for bridging studies (if applicable)
Important: Always consult the specific regulatory authority's guidance for Module 1 requirements. The documents, forms, and organization vary significantly between regions.
Module 2: CTD Summaries
High-Level Overviews for Reviewers
Critical summaries that guide the review process
Module 2 provides comprehensive summaries of the quality, nonclinical, and clinical data. This is typically the first module reviewers read, so clarity and completeness are essential.
Module 2 Structure
2.1 CTD Table of Contents
Comprehensive table of contents for the entire CTD (Modules 2-5). Includes hyperlinks to all documents in the submission.
2.2 CTD Introduction
Brief introduction to the product, submission, and regulatory history:
- β’ Product name (INN, proprietary name)
- β’ Dosage form, route of administration, strength
- β’ Pharmacological class and therapeutic indication
- β’ Prior regulatory submissions and approvals
- β’ Rationale for current application
2.3 Quality Overall Summary (QOS)
Detailed summary of Module 3 content (typically 50-100 pages):
- β’ 2.3.S: Drug substance (API) summary
- β’ 2.3.P: Drug product (finished dosage form) summary
- β’ 2.3.A: Appendices (facilities, adventitious agents for biologics)
- β’ 2.3.R: Regional information (if applicable)
2.4 Nonclinical Overview
Integrated overview of nonclinical data (typically 30-40 pages):
- β’ Overview of nonclinical testing strategy
- β’ Integrated analysis of pharmacology, pharmacokinetics, and toxicology
- β’ Critical findings and their interpretation
- β’ Justification for clinical trial design
2.5 Clinical Overview
Integrated overview of clinical data (typically 50-100 pages):
- β’ Product development rationale
- β’ Overview of biopharmaceutics and clinical pharmacology
- β’ Overview of efficacy
- β’ Overview of safety
- β’ Benefit-risk assessment
- β’ Literature references
2.6 Nonclinical Written and Tabulated Summaries
Detailed tabulated summaries of all nonclinical studies:
- β’ 2.6.1: Introduction
- β’ 2.6.2: Pharmacology written summary
- β’ 2.6.3: Pharmacology tabulated summary
- β’ 2.6.4: Pharmacokinetics written summary
- β’ 2.6.5: Pharmacokinetics tabulated summary
- β’ 2.6.6: Toxicology written summary
- β’ 2.6.7: Toxicology tabulated summary
2.7 Clinical Summary
Detailed summary of clinical program (typically 100-200 pages):
- β’ 2.7.1: Summary of biopharmaceutic studies and associated analytical methods
- β’ 2.7.2: Summary of clinical pharmacology studies
- β’ 2.7.3: Summary of clinical efficacy
- β’ 2.7.4: Summary of clinical safety
- β’ 2.7.5: References
- β’ 2.7.6: Synopses of individual studies
Key Writing Principles for Module 2
Stand-Alone Readability
Module 2 summaries should be comprehensive enough that a reviewer can understand the entire submission without reading Modules 3-5. Include sufficient detail and context.
Critical Analysis, Not Just Description
Don't simply repeat data from Modules 3-5. Provide integrated analysis, interpretation, and discussion of findings. Address potential concerns proactively.
Liberal Use of Cross-References
Reference specific sections in Modules 3-5 to allow reviewers to find detailed data easily. Use hyperlinks in eCTD format.
Tables and Figures
Use tables and figures extensively to present data clearly. Summary tables should integrate data from multiple studies for easy comparison.
Module 3: Quality
Chemistry, Manufacturing, and Controls (CMC)
Comprehensive quality data for drug substance and drug product
Module 3 contains detailed information on the manufacture and control of the drug substance (API) and drug product (finished dosage form). This follows the ICH M4Q guideline.
Module 3 Structure: The "S-P-A-R" Organization
S3.2.S - Drug Substance (API)
- β’ 3.2.S.1: General Information (nomenclature, structure, properties)
- β’ 3.2.S.2: Manufacture (manufacturers, manufacturing process, control of materials)
- β’ 3.2.S.3: Characterization (structure elucidation, impurities)
- β’ 3.2.S.4: Control of Drug Substance (specification, analytical procedures, validation)
- β’ 3.2.S.5: Reference Standards or Materials
- β’ 3.2.S.6: Container Closure System
- β’ 3.2.S.7: Stability (stability studies, post-approval protocols, data)
P3.2.P - Drug Product (Finished Dosage Form)
- β’ 3.2.P.1: Description and Composition
- β’ 3.2.P.2: Pharmaceutical Development (formulation, overages, physicochemical properties)
- β’ 3.2.P.3: Manufacture (manufacturers, batch formula, manufacturing process, controls)
- β’ 3.2.P.4: Control of Excipients (specifications, analytical procedures)
- β’ 3.2.P.5: Control of Drug Product (specification, analytical procedures, validation)
- β’ 3.2.P.6: Reference Standards or Materials
- β’ 3.2.P.7: Container Closure System
- β’ 3.2.P.8: Stability (stability studies, post-approval protocols, data)
A3.2.A - Appendices
- β’ 3.2.A.1: Facilities and Equipment
- β’ 3.2.A.2: Adventitious Agents Safety Evaluation (for biologics)
- β’ 3.2.A.3: Excipients (additional information beyond 3.2.P.4)
R3.2.R - Regional Information
- β’ Region-specific CMC information (if applicable)
- β’ Typically not used in harmonized submissions
Critical Module 3 Documents
3.2.P.2: Pharmaceutical Development
The "story" of product development - critical for QbD approaches:
- β’ Components of drug product (selection rationale)
- β’ Drug product development (formulation, process)
- β’ Manufacturing process development
- β’ Container closure system development
- β’ Microbiological attributes
- β’ Compatibility with reconstitution diluents
3.2.P.8 & 3.2.S.7: Stability
Must follow ICH Q1A-F stability guidelines:
- β’ Long-term stability (25Β°C/60% RH or 30Β°C/75% RH)
- β’ Accelerated stability (40Β°C/75% RH)
- β’ Intermediate conditions (if needed)
- β’ Stress conditions (photostability, temperature)
- β’ Data from β₯3 batches (pilot or commercial scale)
- β’ Minimum 6 months data at filing (12 months ideal)
ICH Quality Guidelines: Module 3 should comply with all relevant ICH Q guidelines (Q1-Q14), including stability testing, analytical validation, impurities control, and pharmaceutical development. Reference specific ICH guidelines where applicable.
Module 4: Nonclinical Study Reports
Pharmacology, Pharmacokinetics, and Toxicology
Full study reports supporting safety of the product
Module 4 contains complete study reports for all nonclinical studies. Organization follows ICH M4S (Safety) guideline, with studies grouped by discipline.
Module 4 Structure
4.2.1 Pharmacology
- β’ 4.2.1.1: Primary pharmacodynamics
- β’ 4.2.1.2: Secondary pharmacodynamics
- β’ 4.2.1.3: Safety pharmacology (cardiovascular, CNS, respiratory)
- β’ 4.2.1.4: Pharmacodynamic drug interactions
4.2.2 Pharmacokinetics
- β’ 4.2.2.1: Analytical methods and validation reports
- β’ 4.2.2.2: Absorption studies
- β’ 4.2.2.3: Distribution studies
- β’ 4.2.2.4: Metabolism studies
- β’ 4.2.2.5: Excretion studies
- β’ 4.2.2.6: Pharmacokinetic drug interactions
- β’ 4.2.2.7: Other pharmacokinetic studies
4.2.3 Toxicology
- β’ 4.2.3.1: Single-dose toxicity
- β’ 4.2.3.2: Repeat-dose toxicity (duration supporting clinical studies)
- β’ 4.2.3.3: Genotoxicity (Ames, in vitro chromosomal aberration, in vivo micronucleus)
- β’ 4.2.3.4: Carcinogenicity (2-year rodent studies if applicable)
- β’ 4.2.3.5: Reproductive and developmental toxicity
- β’ 4.2.3.6: Local tolerance
- β’ 4.2.3.7: Other toxicity studies (antigenicity, immunotoxicity, etc.)
GLP Compliance Requirements
Which Studies Require GLP?
β GLP Required:
- β’ Safety pharmacology
- β’ All toxicology studies
- β’ Genotoxicity studies
- β’ Carcinogenicity studies
- β’ Reproductive toxicity
β GLP Not Always Required:
- β’ Primary pharmacodynamics
- β’ Secondary pharmacodynamics
- β’ Most pharmacokinetic studies
- β’ Exploratory studies
Note: Each study report must include a statement regarding GLP compliance and the role of the Quality Assurance Unit.
ICH Safety Guidelines Referenced in Module 4
ICH S1A/B
Carcinogenicity Studies
Need, conduct, and evaluation
ICH S2(R1)
Genotoxicity Testing
Battery of tests required
ICH S3A/B
Toxicokinetics & PK
Integration with toxicology
ICH S4A
Chronic Toxicity Testing
Duration of studies
ICH S5(R3)
Reproductive Toxicology
Fertility, embryo-fetal, pre/postnatal
ICH S6(R1)
Biotechnology Products
Species selection, immunogenicity
ICH S7A/B
Safety Pharmacology
Core battery, supplemental studies
ICH S8
Immunotoxicity Studies
When to conduct
ICH S9
Nonclinical for Anticancer
Special considerations for oncology
ICH S10
Photosafety Evaluation
Phototoxicity assessment
Module 5: Clinical Study Reports
Clinical Pharmacology, Efficacy, and Safety
Full clinical study reports supporting efficacy and safety
Module 5 contains complete clinical study reports for all clinical trials. Organization follows ICH M4E (Efficacy) guideline, with studies grouped by type and phase.
Module 5 Structure
5.2 Tabular Listing of All Clinical Studies
Complete list of all clinical studies (one line per study):
- β’ Study identifier, title, phase
- β’ Study population, number of subjects
- β’ Study design, objectives
- β’ Location in eCTD (cross-reference to Module 5 section)
5.3.1 Reports of Biopharmaceutic Studies
- β’ 5.3.1.1: Bioavailability (BA) study reports
- β’ 5.3.1.2: Comparative BA and bioequivalence (BE) study reports
- β’ 5.3.1.3: In vitro-in vivo correlation study reports
- β’ 5.3.1.4: Reports of bioanalytical and analytical methods
5.3.2 Reports of Studies Pertinent to Pharmacokinetics
- β’ Healthy subject PK studies
- β’ Patient PK studies
- β’ Intrinsic factor studies (renal/hepatic impairment, age, gender)
- β’ Extrinsic factor studies (drug-drug interactions, food effects)
- β’ Population PK studies
5.3.3 Reports of Human Pharmacodynamics Studies
- β’ Proof of concept studies
- β’ Dose-response exploration
- β’ Studies in healthy volunteers or patients
5.3.4 Reports of Human Pharmacokinetics and Pharmacodynamics
- β’ Integrated PK/PD studies
5.3.5 Reports of Efficacy and Safety Studies
The core clinical evidence (typically the largest section):
- β’ 5.3.5.1: Controlled studies pertinent to the claimed indication
- β’ 5.3.5.2: Uncontrolled studies
- β’ 5.3.5.3: Reports of analyses of data from more than one study
- β’ 5.3.5.4: Other clinical studies
5.3.6 Reports of Postmarketing Experience
- β’ For products already marketed in other regions
5.3.7 Case Report Forms and Individual Patient Listings
- β’ Case report forms (CRFs) - often provided electronically
- β’ Individual patient data listings for deaths, SAEs, discontinuations
ICH E3 Clinical Study Report Format
All clinical study reports in Module 5 should follow ICH E3 structure:
- 1. Title Page
- 2. Synopsis (2-3 page summary)
- 3. Table of Contents
- 4. List of Abbreviations
- 5. Ethics
- 6. Investigators and Study Administrative Structure
- 7. Introduction
- 8. Study Objectives
- 9. Investigational Plan
- 10. Study Patients
- 11. Efficacy Evaluation
- 12. Safety Evaluation
- 13. Discussion and Overall Conclusions
- 14. Tables, Figures, and Graphs Referred to but Not Included in the Text
- 15. Reference List
- 16. Appendices (protocol, sample CRF, technical details, patient data listings, etc.)
Integrated Summary of Safety (ISS) and Efficacy (ISE)
While not always required, integrated summaries provide powerful cross-study analyses:
- ISS:Integrated Summary of Safety - Pools safety data across studies to identify rare adverse events and characterize the overall safety profile. Follow ICH E2F guidance.
- ISE:Integrated Summary of Efficacy - Analyzes efficacy across studies to demonstrate consistency and robustness of treatment effect.
eCTD Technical Specifications
While ICH M4 defines the content structure, eCTD specifications define the technical format for electronic submission. The current standard is eCTD v4.0.
eCTD Folder Structure
[Application Root]
βββ index.xml (root XML file)
βββ util/
β βββ dtd/ (Document Type Definitions)
βββ m1/
β βββ us/ (or eu, jp, etc.)
β βββ 10-cover/
β βββ 12-application-information/
β βββ 13-forms/
β βββ ... (region-specific structure)
βββ m2/
β βββ 21-toc/
β βββ 22-introduction/
β βββ 23-quality-overall-summary/
β βββ 24-nonclinical-overview/
β βββ 25-clinical-overview/
β βββ 26-nonclinical-summaries/
β βββ 27-clinical-summary/
βββ m3/
β βββ 32s-drug-substance/
β βββ 32p-drug-product/
β βββ 32a-appendices/
β βββ 32r-regional/
βββ m4/
β βββ 42-study-reports/
β β βββ 421-pharmacology/
β β βββ 422-pharmacokinetics/
β β βββ 423-toxicology/
β βββ ...
βββ m5/
βββ 53-clinical-study-reports/
β βββ 531-biopharm-studies/
β βββ 532-pk-studies/
β βββ 533-pd-studies/
β βββ 534-pk-pd-studies/
β βββ 535-efficacy-safety/
β βββ 536-postmarketing/
β βββ 537-case-report-forms/
βββ ...XML Backbone
The eCTD XML backbone provides the structure and navigation for the submission:
- βindex.xml: Root file that references all modules and their contents
- βDocument nodes: Each document (PDF, XML, etc.) is represented as a leaf node
- βMetadata: Each node includes title, operation (new, replace, delete), checksum
- βHyperlinks: Cross-references within documents are maintained as hyperlinks
Sequence Concept
What is a Sequence?
An eCTD submission is organized into sequences. Each sequence represents a snapshot of the submission at a point in time:
- β’ Sequence 0000: Initial submission (IND, NDA, MAA)
- β’ Sequence 0001: First amendment/update
- β’ Sequence 0002: Second amendment/update
- β’ And so on...
Each new sequence only contains changed files (new, replace, delete operations), not the entire submission. Regulators build the complete submission by combining all sequences.
File Formats
Allowed Formats
- β’ PDF/A-1a or PDF/A-1b: Required for most documents
- β’ XML: For structured data (index.xml, study data)
- β’ MPEG-2, AVI, QuickTime: For videos
- β’ JPEG, PNG: For standalone images
- β’ RTF: For documents requiring editing (rare)
PDF Requirements
- β’ Must be searchable (OCR if scanned)
- β’ Hyperlinks must be functional
- β’ Bookmarks for navigation (optional but recommended)
- β’ No security restrictions (passwords, editing locks)
- β’ No JavaScript or executable content
Agency-Specific Requirements
While Modules 2-5 are harmonized, each agency has specific requirements for eCTD implementation:
πΊπΈFDA (United States)
- β’ Gateway: Electronic Submissions Gateway (ESG)
- β’ Mandatory: eCTD required for all NDAs, BLAs, ANDAs (since 2017-2018)
- β’ Validation: FDA provides free validation tools (Global Submission Tool - GST)
- β’ Study Data: CDISC datasets required (SDTM, ADaM) for Phase 2/3 studies
- β’ Granularity: Prefers individual study reports over compilations
πͺπΊEMA (European Union)
- β’ Gateway: EMA Gateway (CESP)
- β’ Mandatory: eCTD required for centralized procedure and many national procedures
- β’ Validation: Use EMA validation service before submission
- β’ Study Data: CDISC datasets increasingly expected
- β’ RMP: Risk Management Plan (RMP) required in Module 1.8
- β’ QRD: Product information must follow QRD template
π―π΅PMDA (Japan)
- β’ Gateway: PMDA eCTD Gateway
- β’ Mandatory: eCTD required for new applications
- β’ Validation: PMDA provides validation tools
- β’ Language: Module 1 in Japanese; Modules 2-5 can be English with Japanese summary
- β’ Bridging: Often requires ethnic factor studies comparing Japanese to non-Japanese data
- β’ Interview Form: Specific Japanese requirement for product information
π¨π¦Health Canada
- β’ Gateway: Common Electronic Submission Gateway (CESG)
- β’ Mandatory: eCTD required for most submission types
- β’ Module 1: Similar to FDA with Canadian-specific forms
- β’ Bilingual: Product monograph may need English and French versions
Best Practices & Common Pitfalls
Best Practices
βStart with Module 2
Write Module 2 summaries early in development. This forces critical thinking about data gaps and study design. Continuously update Module 2 as new data becomes available.
βUse Consistent Terminology
Use identical terminology across all modules. If Module 2 refers to "Study ABC-123," use that exact identifier in Module 5. Create a controlled vocabulary document.
βImplement Robust Cross-Referencing
Every claim in Module 2 should reference supporting data in Modules 3-5. Use hyperlinks in eCTD to make navigation seamless. Validate all hyperlinks before submission.
βValidate Early and Often
Use agency validation tools (FDA GST, EMA validator) throughout assembly, not just at the end. Fix structural issues early to avoid last-minute scrambles.
βMaintain Document Traceability
Keep detailed records of document versions, authors, review status. Use document management systems with audit trails to comply with 21 CFR Part 11.
Common Pitfalls to Avoid
βBroken Hyperlinks
One of the most common eCTD deficiencies. Always validate hyperlinks before submission. Broken links frustrate reviewers and delay approval.
βInconsistent Document Placement
Putting documents in wrong eCTD sections (e.g., stability data in 3.2.P.5 instead of 3.2.P.8). Follow ICH M4 structure precisely.
βNon-Searchable PDFs
Scanned documents without OCR are difficult for reviewers to search. Always ensure PDFs are text-searchable, either by creating from source documents or running OCR on scans.
βInadequate Module 2 Summaries
Module 2 that simply lists studies without analysis. Summaries should synthesize data, identify trends, address concerns, and tell a cohesive story.
βLast-Minute Assembly
Waiting until the end to compile the eCTD. Start early, build incrementally, and continuously validate. This reduces stress and improves quality.
Submission Checklist
Before submitting, verify:
Ready to Streamline Your eCTD Submissions?
See how DossiAIr automates ICH M4 eCTD structure creation with AI-powered intelligence