Fact-checked by Grok 2 weeks ago

Common Technical Document

The Common Technical Document (CTD) is a standardized, harmonized format developed by the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) for organizing the technical documentation required for the registration of pharmaceutical products, including , , and data, to facilitate efficient regulatory review across multiple regions. Introduced to address the inefficiencies of region-specific submission formats, the CTD was finalized through ICH guidelines in the late 1990s and early 2000s, becoming mandatory for submissions in the and in 2003, and strongly recommended in the United States since 2003, with the electronic version (eCTD) becoming mandatory for the U.S. (FDA) in 2017, thereby reducing the time and resources needed to compile applications while promoting consistent information exchange among regulatory authorities. The CTD's core objective is to enable a common structure for dossiers submitted to agencies like the (EMA), FDA, and Japan's (PMDA), minimizing reformatting efforts and supporting good review practices through unambiguous presentation of data. The CTD is organized into five modules, with Modules 2 through 5 providing a universal framework applicable across ICH regions, while Module 1 accommodates region-specific administrative and prescribing information: Module 2 contains high-level summaries such as the Quality Overall Summary and clinical overviews; Module 3 details quality aspects like drug substance and product ; Module 4 covers nonclinical study reports on and ; and Module 5 includes clinical study reports on and in humans. This modular design ensures transparency and granularity, particularly in its electronic variant, the electronic Common Technical Document (eCTD), which has become the standard for digital submissions since its implementation in the early 2000s and is now mandatory for most applications to the FDA's Center for Drug Evaluation and Research (CDER) and Center for Biologics Evaluation and Research (CBER), including new drug applications (NDAs), abbreviated new drug applications (ANDAs), and investigational new drug applications (INDs). Overall, the CTD has significantly enhanced global regulatory , with ongoing updates like ICH M4(R4) refining its to better support submissions and lifecycle management of pharmaceutical approvals.

Introduction

Definition and Purpose

The Common Technical Document (CTD) is an internationally format for organizing the technical documentation required for pharmaceutical registration applications, encompassing data on the , , and of medicines for use. Developed through the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Use (ICH), the CTD provides a standardized structure that ensures consistency in presenting administrative, summary, and detailed scientific information across regulatory jurisdictions. The primary purpose of the CTD is to streamline the regulatory review process by facilitating efficient communication between applicants and authorities, as well as enabling the exchange of information among regulatory bodies. It aims to reduce the time and resources needed for compiling submissions, minimize duplication of data, and promote mutual recognition of scientific evaluations internationally, thereby accelerating global access to safe and effective medicines. By replacing disparate national formats—such as those previously used in , , and the —the CTD addresses inefficiencies in the pre-harmonization era, where varying requirements often led to redundant efforts and delayed approvals. At its core, the CTD embodies principles of to standardize technical requirements while allowing flexibility for region-specific needs, ultimately supporting a more cohesive global regulatory framework.

Scope and Applicability

The Common Technical Document (CTD) is mandatory for new drug applications (NDAs), abbreviated new drug applications (ANDAs), biologics license applications (BLAs), and their supplements and amendments in International Council for Harmonisation (ICH) regions, including the , , and . It also applies to variations and post-approval changes that require detailed technical submissions, facilitating lifecycle management of approved products. Outside ICH regions, CTD adoption is optional but widely encouraged for harmonized regulatory submissions to streamline international reviews. The CTD covers pharmaceuticals for human use, including chemically synthesized , biotechnology-derived biologics, and certain combination products that incorporate drug components with medical devices. It excludes veterinary medicinal products, which fall under separate harmonization efforts like the Veterinary International Conference on Harmonisation (VICH) guidelines. Primary target users include pharmaceutical and biotechnology companies preparing regulatory dossiers, as well as agencies such as the U.S. (FDA), (EMA), and Pharmaceuticals and Medical Devices Agency (PMDA) in , enabling efficient multi-regional evaluations. The format supports applicants in organizing quality, safety, and efficacy data across its modules for consistent review. The CTD format for applications, such as investigational new drug () submissions, is required for electronic submissions to the FDA's CDER and CBER as of September 2024, mandatory for commercial INDs and optional for noncommercial ones, often utilizing partial CTD module structure. Minor post-approval changes not requiring comprehensive resubmission, like administrative updates or low-risk modifications, typically do not necessitate the complete CTD format.

History and Development

Origins in ICH Guidelines

The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) was established in 1990 through collaboration among regulatory authorities and pharmaceutical industry representatives from the (EU), , and the (US). This founding aimed to harmonize technical requirements for the registration of pharmaceutical products, thereby streamlining global , reducing redundant testing, and enhancing efficiency in regulatory approvals while maintaining high standards of , , and quality. The initiative built on earlier bilateral discussions in the 1980s, including talks between the EU and on regulatory alignment, as well as broader trilateral exchanges involving the US, which highlighted the need for coordinated approaches to avoid fragmented international standards. Prior to the CTD, pharmaceutical submissions faced significant inconsistencies across regions, complicating global development efforts. In the EU, the Committee for Proprietary Medicinal Products (CPMP) required detailed formats under its guidelines, while Japan's Ministry of Health, Labour and Welfare (MHLW) mandated region-specific structures, and the US Food and Drug Administration (FDA) followed the (NDA) format, often leading to duplicated data preparation, increased costs, and delays in market access. These disparities stemmed from divergent national priorities and formats, prompting the ICH to prioritize a unified submission standard as part of its early harmonization agenda. The initial development of the Common Technical Document (CTD) emerged directly from ICH's multidisciplinary efforts, with the ICH M4 guideline finalizing its basic framework in the late 1990s following preparatory work that began around 1989. This guideline outlined a standardized, modular organization for regulatory dossiers, influenced by the aforementioned 1980s harmonization talks and the need to integrate quality, safety, and efficacy data cohesively across jurisdictions. Key stakeholders, including the European Federation of Pharmaceutical Industries and Associations (EFPIA), the Japan Pharmaceutical Manufacturers Association (JPMA), and the Pharmaceutical Research and Manufacturers of America (PhRMA), played pivotal roles in drafting the CTD through their participation in ICH Expert Working Groups, providing scientific expertise to ensure practical applicability for industry submissions.

Key Revisions and Milestones

The International Council for Harmonisation (ICH) finalized the Common Technical Document (CTD) guideline, ICH M4, in November 2000, establishing a standardized format for submitting quality, safety, and efficacy data to regulatory authorities. This approval marked the culmination of efforts to harmonize regulatory submissions across ICH regions, with initial implementations occurring between 2001 and 2003, including mandatory adoption for new drug applications in the and by 2003. In 2002 and 2003, the CTD underwent early refinements to enhance clarity and consistency, particularly in Module 2 summaries. The September 2002 re-edition updated numbering and section headers across the document, while specific guidelines like ICH M4Q for quality aspects and for were adopted in July 2003 and February 2003, respectively, improving the organization and presentation of summary information. During the 2010s, revisions focused on adapting the CTD to emerging product types, including alignment with biosimilars through the ICH M4Q guideline and its Quality Overall Summary (QOS), which facilitates comparability assessments by structuring in Modules 2 and 3. In 2016, ICH M4(R4) was finalized, introducing updates to Module 3 on , such as refined levels to support submissions and better of and control information. More recent milestones include the 2020 ICH endorsement of the M4Q(R2) revision proposal, which expands the CTD's applicability to non-traditional products like biologicals and therapies by modernizing Module 3 structures for comprehensive quality information across chemical and biological entities. In May 2025, ICH released a draft of the M4Q(R2) guideline for , which closed in October 2025, aiming to further enhance registration efficiency and ization. Ongoing efforts emphasize CTD evolution for digital submissions, with eCTD v4.0 specifications released in 2016 and subsequent updates to accommodate structured data and lifecycle management. These revisions have been driven by regulatory from submission reviews, which highlighted needs for improved and clarity, as well as technological advances such as the transition to CTD (eCTD) formats that enable better and global harmonization.

Overall Structure

Hierarchical Organization

The Common Technical Document (CTD) is structured hierarchically into five that provide a logical progression from administrative details to detailed scientific data, facilitating efficient regulatory review across jurisdictions. Module 1 contains region-specific administrative and prescribing information, while 2 through 5 form the harmonized core applicable to all ICH regions: Module 2 offers high-level summaries, Module 3 addresses quality aspects, Module 4 covers nonclinical study reports, and Module 5 includes clinical study reports. This ensures that the presents information in a standardized manner, allowing regulators to navigate from overviews to supporting evidence without redundancy. Within each module, the hierarchy employs varying levels of granularity to organize data logically, using numbered sections, subsections, and appendices that build from summaries to raw data and detailed reports. For instance, Module 2 typically uses up to four levels of headings for its summaries, while Modules 3, 4, and 5 extend to five or more levels, including appendices for supplementary materials such as raw data tables or protocols. This structure promotes a flow where high-level analyses in Module 2 reference deeper details in subsequent modules, enabling reviewers to access information progressively based on need. The granularity is defined to balance comprehensiveness with manageability, with guidance specifying whether single or multiple files are appropriate at each level. Navigation within the CTD is supported by a comprehensive (TOC) that outlines the hierarchy down to relevant depths—such as third or fourth levels for Module 2 and study-specific levels for Modules 4 and 5—ensuring easy location of sections in both paper and electronic formats. In the (eCTD), hyperlinks integrated into the XML backbone further enhance accessibility, allowing direct jumps between referenced elements across modules. This aids reviewers in tracing connections, such as cross-references from summaries to full reports, without manual searching. The CTD can be submitted in or , with paper versions using standardized page sizes ( in the EU and Japan, or 8.5x11 inches in the US) and fonts like 12-point for legibility. Electronic submissions, now mandatory in many regions, utilize PDF files within the eCTD framework for lifecycle management. For complex drug applications, the total often exceeds 100,000 pages, encompassing extensive data from development stages.

Granularity and Navigation

The Common Technical Document (CTD) incorporates a internal within its modules to enhance and allow regulatory reviewers to complex submissions efficiently. Granularity in the CTD is defined as the appropriate division of content into discrete documents, where each document comprises a set of sequentially numbered pages or an electronic file, often separated by tabs in paper-based formats. This approach ensures that information is presented at a level of detail that balances comprehensiveness with accessibility, as specified in the ICH M4 guideline. Standardized headings provide a consistent framework for labeling sections across Modules 2 through 5, such as 2.1 for the Table of Contents in Module 2 or 3.2.S for drug substance-related content in Module 3, facilitating uniform structure in submissions to international regulatory authorities. These headings follow a hierarchical numbering system that aligns with the overall module organization, enabling quick location of specific topics without ambiguity. To prevent redundancy, cross-referencing is employed throughout the CTD, where references point to pertinent data in other modules or supporting materials rather than repeating information, thereby streamlining the dossier while maintaining completeness. Appendices and annexes serve as dedicated placements for supplementary elements like , protocols, and detailed supporting studies, allowing the main body to focus on synthesized analysis. For instance, nonclinical study reports in Module 4 may separate core text from voluminous appendices containing raw observations or methodologies, preserving the document's focus and readability. Best practices for navigation emphasize logical sequencing from general overviews to specific details within each , coupled with detailed Tables of Contents that extend to the third or fourth level in Module 2 and up to the fifth level in Module 3. Additionally, ICH Q&A documents recommend consistent pagination—starting at page one for each document—and precise cross-references that include numbers alongside brief contextual descriptions to aid reviewer orientation. Subnumbering within individual documents, such as 3.2.P.3.3.1, is permitted to further refine granularity without altering the primary heading structure. These practices ensure the CTD's internal navigation supports both initial review and ongoing lifecycle management of pharmaceutical applications.

Core Modules

Module 1: Administrative and Prescribing Information

Module 1 of the Common Technical Document (CTD) serves as the administrative front matter for regulatory submissions, encompassing region-specific documents that provide essential legal, procedural, and prescribing details to support the review process by authorities such as the U.S. Food and Drug Administration (FDA) and the (EMA). Unlike the harmonized Modules 2 through 5, Module 1 is not standardized across International Council for Harmonisation (ICH) regions, allowing each jurisdiction to define its required contents and format to align with local regulations. This module ensures that submissions include necessary administrative paperwork and prescribing information, facilitating efficient regulatory evaluation while addressing unique regional mandates. The primary purpose of Module 1 is to furnish the regulatory and legal context for the overall CTD submission, including application forms, cover letters, and certifications that verify compliance with submission requirements. It also incorporates prescribing information, such as proposed product labels and summaries of product characteristics, to outline safe and effective use for healthcare professionals and patients. By centralizing these elements, Module 1 streamlines the initial administrative review, enabling regulators to quickly assess procedural completeness before delving into scientific data in subsequent modules. Key contents of Module 1 typically include a comprehensive for the entire submission, application forms tailored to the product type (e.g., new drug applications), and cover letters that summarize the submission's purpose and contents. Additional elements often encompass environmental assessments or risk evaluations to address potential ecological impacts of the pharmaceutical product, as well as patent information detailing status where applicable. Prescribing information forms a critical component, featuring labels, package inserts, and summaries of product characteristics that must undergo readability testing to ensure clarity. Examples of supporting documents include debarment certifications, which affirm that no disqualified individuals are involved in the application process, and financial disclosure statements from clinical investigators to mitigate conflicts of interest. Regional customization is a defining feature of Module 1, with contents varying significantly by jurisdiction to reflect local legal frameworks. For FDA submissions , Module 1 requires specific forms such as Form FDA 356h for application cover sheets, alongside environmental assessments under the and detailed patent certifications under the Hatch-Waxman Act. Debarment and financial disclosure requirements are mandatory, with the latter covering compensation, equity interests, and proprietary data arrangements for investigators in clinical studies. In contrast, for EMA submissions in the , Module 1 follows the structure outlined in the EU Module 1 eCTD Specification, including an electronic application form per Directive 2001/83/EC, product information aligned with the European Public Assessment Report (EPAR) format, and an environmental risk assessment for active substances. The Summary of Product Characteristics (SmPC) and package leaflet must be provided in all official EU languages, emphasizing plans and paediatric specifics if applicable under Regulation (EC) No 1901/2006. This lack of harmonization necessitates applicants to prepare region-specific versions of Module 1, often using electronic CTD (eCTD) formats for validation and submission.

Module 2: Common Technical Document Summaries

Module 2 of the Common Technical Document (CTD) serves as the integrative summary section, providing high-level overviews and analyses of the quality, nonclinical, and clinical data to facilitate efficient regulatory review. It condenses the detailed information from Modules 3, 4, and 5 into concise narratives and tables, emphasizing critical assessments rather than exhaustive data reproduction. This module enables reviewers to the overall rationale, benefits, and risks of the pharmaceutical product without delving into full reports. The structure of Module 2 is standardized into seven sections, beginning with navigational aids and progressing to specialized summaries. Section 2.1, the , outlines the contents of Modules 2 through 5, typically down to the third or fourth level of (e.g., 2.3.S for drug substance subsections), ensuring easy navigation across the . Section 2.2, the , offers a brief general overview of the pharmaceutical, including its pharmacologic class, , proposed clinical use, and development context, limited to approximately one page to set the stage for subsequent summaries. Section 2.3, the Quality Overall Summary (QOS), provides a comprehensive yet concise integration of the quality information from Module 3, focusing on processes, controls, and product specifications. Guided by ICH M4Q, it is organized into subsections mirroring Module 3: 2.3.S for drug substance (covering manufacture, characterization, control, reference standards, and ), 2.3.P for drug product (including description, pharmaceutical development, , control, and ), 2.3.A for appendices (e.g., facilities/), and 2.3.R for regional information. The QOS highlights critical aspects such as , batch analyses, profiles, and data, with justifications for specifications and cross-references to detailed Module 3 data. It employs tables and figures for clarity, typically limited to 40 pages (text only, excluding visuals) for conventional products or up to 80 pages for complex biologics. As of 2025, ICH M4Q is under revision as M4Q(R2), which proposes updates to the structure of section 2.3 and Module 3 to enhance support for lifecycle management and digital submissions; the revision is in phase and not yet adopted. Section 2.4, the Nonclinical Overview, delivers a critical, integrated analysis of nonclinical , , and data from Module 4, emphasizing the testing strategy, key findings, and their implications for clinical development. As per ICH M4S, it includes discussions on pharmacodynamic effects, /// (ADME), toxic effects (e.g., dose-dependency, reversibility, ), and integrated risk assessments, with cross-references to specific Module 4 study reports. Literature references and considerations for impurities or degradants are incorporated, using tables for interspecies comparisons. This section is recommended to be approximately 30 pages, prioritizing narrative interpretation over rote summarization. Section 2.5, the Clinical Overview, offers a critical evaluation of the clinical data from Module 5, narrating the product development rationale, biopharmaceutics, , , and to support benefit-risk conclusions. Structured under ICH M4E, it comprises subsections such as 2.5.1 (product development rationale, including indication and program overview), 2.5.2 (biopharmaceutics, analyzing factors like food effects), 2.5.3 (, covering / in special populations), 2.5.4 (, assessing study designs and results across indications), 2.5.5 (, evaluating adverse events and ), 2.5.6 (benefits and risks, integrating findings in therapeutic context), and 2.5.7 (references). Cross-references to Module 5 reports are essential, with the overview limited to about 30 pages, enhanced by graphs and tables for key metrics like exposure-response relationships. Section 2.6, Nonclinical Written and Tabulated Summaries, furnishes detailed, factual synopses of nonclinical data, organized by , , and , to complement the overview in 2.4. Following ICH M4S, it includes written narratives for each category (e.g., primary/secondary , studies, single/repeat-dose toxicity, , carcinogenicity, ) alongside tabulated overviews with study details like , doses, durations, and no-observed-adverse-effect levels (NOAELs). Tables follow standardized templates, with in vitro data preceding , and cross-references to Module 4 reports. The total length is generally 100-150 pages, using appendices for extensive tables to maintain readability. Finally, Section 2.7, the Clinical Summary, presents an integrated, detailed factual compilation of clinical information from 5, avoiding (reserved for 2.5). Per ICH M4E, it is divided into 2.7.1 (biopharmaceutic studies, summarizing /equivalence and analytical methods), 2.7.2 ( studies, detailing /, interactions, and ), 2.7.3 (clinical , covering study results, comparisons, and dosing per indication), 2.7.4 (clinical , including exposure, adverse events, data, and special populations), 2.7.5 (references), and 2.7.6 (synopses of individual studies, limited to 3-10 pages each). Tables illustrate cross-study comparisons, such as endpoints or incidences, with cross-references to full 5 reports. Length varies from 50 to 400 pages based on data volume, one document per indication. Throughout Module 2, summaries cross-reference Modules 3-5 to avoid redundancy, adhering to ICH guidelines for consistent formatting, such as PDF files with hyperlinks and standardized numbering. Each summary is typically constrained to 30-50 pages for overviews to promote conciseness, while tabulated sections allow for more extensive data presentation.

Module 3: Quality

Module 3 of the Common Technical Document (CTD) presents a detailed quality dossier encompassing the chemistry, manufacturing, and controls () for both the drug substance and drug product, ensuring their , , and are rigorously documented for regulatory review. This module follows the overarching structure outlined in ICH M4Q, which harmonizes the presentation of quality data across regulatory authorities. It includes the body of data (3.2), appendices (3.2.A), and regional information (3.2.R), focusing exclusively on processes, , and controls without overlapping into nonclinical or clinical aspects. As of 2025, ICH M4Q is under revision as M4Q(R2), which proposes updates to the structure of Module 3 and section 2.3 to enhance support for lifecycle management and digital submissions; the revision is in phase and not yet adopted. The substance section (3.2.S) provides in-depth information on the manufacture, , and of the . Manufacture details include the description of the manufacturing process, critical steps, and controls, often supported by flow diagrams and justifications for reprocessing, with guidance on development drawn from ICH Q11. covers the elucidation of the substance's structure, physicochemical properties, and impurity profiles, where impurities are classified, identified, and qualified in accordance with ICH Q3A to ensure they do not pose risks. Control aspects encompass specifications, analytical procedures, validation of those procedures, and batch analyses, referencing ICH Q6A for setting acceptance criteria. data are included to demonstrate the consistency and reproducibility of the manufacturing process. The drug product section (3.2.P) addresses , , and of the finished . It details pharmaceutical , including selection, compatibility studies, and process optimization, again informed by ICH Q11 principles for understanding formulation impacts on quality. Components, such as the drug substance and excipients, are described with their specifications and functions, while manufacturing information outlines the process, controls, and validation data to ensure product consistency. Stability studies are a core element, conducted under defined conditions (e.g., temperature, humidity) as per ICH Q1A(R2), providing evidence of how the drug product's quality varies over time and supporting proposed shelf-life claims through accelerated and long-term testing protocols. Regional information (3.2.R) accommodates jurisdiction-specific requirements, such as batch analyses, certificates of analysis, and comparative profiles where applicable, ensuring with local standards without duplicating core data. Appendices support the main body with additional details: 3.2.A.1 on facilities and equipment describes sites, utilities, and measures to prevent cross-contamination; 3.2.A.2 evaluates adventitious agents , particularly viral risks for biotechnology-derived products, following ICH Q5A(R1) through testing of cell substrates, virus clearance studies, and risk assessments; and 3.2.A.3 covers novel excipients if used. These elements collectively form a robust , enabling regulators to assess reliability and product .

Module 4: Nonclinical Study Reports

Module 4 of the Common Technical Document (CTD) contains detailed reports from nonclinical studies conducted to assess the , , and of a pharmaceutical, primarily using systems and animal models. These reports provide the foundational and data supporting clinical development, with full study protocols, methodologies, results, and included to allow regulatory . Unlike the summaries in Module 2, this module presents the complete, uncondensed study documentation, often emphasizing (GLP) compliance for pivotal studies. The structure begins with 4.1 Table of Contents, which lists all nonclinical study reports and their locations within the CTD for easy navigation. This is followed by 4.2 Study Reports, organized into three main categories: , , and . In (4.2.1), reports cover primary pharmacodynamics (effects on intended targets), secondary pharmacodynamics (off-target effects), pharmacology (vital organ functions per ICH S7 guidelines), and pharmacodynamic drug interactions. reports (4.2.2) detail analytical methods and validation, absorption, distribution, metabolism, excretion (), pharmacokinetic drug interactions, and other relevant studies. reports (4.2.3) are the most extensive, including single-dose and repeat-dose toxicity by species and route (with toxicokinetics), (in vitro and in vivo assays per ICH S2), carcinogenicity (long-term studies per ICH S1, unless waived), reproductive and developmental toxicity (fertility, embryo-fetal development, prenatal/postnatal per ICH S5, and juvenile animal studies), local tolerance, and other studies like immunotoxicity. For oncology pharmaceuticals, the ICH S9 guideline modifies these requirements, exempting and carcinogenicity studies from routine conduct for marketing in advanced cancer patients, focusing instead on core battery toxicity and only at marketing authorization, while maintaining GLP for studies. All pivotal nonclinical studies in Module 4 must comply with GLP unless scientifically justified otherwise, ensuring and ; deviations or non-GLP status are noted in the reports. Tabular summaries of these studies, including key findings and interpretations, are integrated into the Nonclinical Overview and Summary in Module 2, but Module 4 owns the , individual animal observations, and statistical analyses. The module concludes with 4.3 Literature References, a compilation of published supporting or supplementing the conducted studies, with each reference justified for its relevance (e.g., bridging data gaps where studies are limited). This section allows regulators to external evidence without duplicating full texts.

Module 5: Clinical Study Reports

Module 5 of the Common Technical Document (CTD) encompasses the detailed clinical study reports and associated data that support the and profile of a medicinal product, focusing on clinical trials from phases 1 through 3. This module provides regulators with comprehensive evidence derived from biopharmaceutic, pharmacokinetic, pharmacodynamic, and therapeutic studies to evaluate the benefit-risk balance. Unlike summaries in Module 2, Module 5 includes full technical reports formatted according to the ICH guideline, which standardizes the structure and content of individual clinical study reports to ensure consistency across ICH regions. The module begins with section 5.1, a outlining all included items, followed by section 5.2, a tabular listing of all clinical studies with details on , objectives, numbers, and status (completed or ongoing). Section 5.3 contains the core clinical study reports, subdivided into categories such as 5.3.1 for biopharmaceutic studies (e.g., and ), 5.3.2 for pharmacokinetic studies using biomaterials, 5.3.3 for pharmacokinetic studies, 5.3.4 for pharmacodynamic studies, and 5.3.5 for and studies. These reports adhere to ICH , incorporating elements like study synopses, title pages, statements, investigator details, objectives, , demographics, and evaluations, statistical methods, discussions, and appendices with protocols and amendments. Protocols and their amendments are fully included in Appendix 16.1.1 of each report, with statistical analyses detailed in sections on methods and results, including sample size justifications, planned analyses, and p-values. Within 5.3.5, reports cover controlled and uncontrolled clinical studies pertinent to the claimed indication, alongside other therapeutic studies, with full integration of phase 1-3 data. A key component is subsection 5.3.5.3, which houses the Integrated Summary of Efficacy (ISE) and Integrated Summary of Safety (ISS), providing cross-study analyses such as meta-analyses, event rates, dose-response relationships, and subgroup evaluations to synthesize overall effectiveness and safety. The ISE focuses on efficacy outcomes across studies, including statistical pooling where appropriate, while the ISS addresses adverse events, laboratory abnormalities, and exposure data, often exceeding 1,000 pages with extensive appendices. These summaries integrate data from all relevant clinical study reports in Module 5, differing from the concise overviews in Module 2's clinical summary. Additional subsections include 5.3.6 for post-marketing experience reports and 5.3.7 for case report forms and individual patient listings, ensuring traceability of raw data. For special populations, such as pediatric subsets, reports under 5.3.5 follow ICH E11 principles, with dedicated analyses of and in children, including strategies when adult suffice, and placement in 5.3.5.4 for other studies if not directly tied to the primary indication. Similarly, geriatric are evaluated per ICH E7, integrated into and reports with subgroup breakdowns by age. Section 5.4 compiles literature references supporting the clinical , listed numerically and cross-referenced to relevant reports, drawing from published studies on , , or . This module's content directly informs the clinical overview in Module 2, providing the evidentiary foundation for regulatory review.

Electronic Common Technical Document

Format Specifications

The (eCTD) employs a standardized XML-based format to ensure across regulatory authorities, with the backbone serving as the central navigational structure that links all submission components. This backbone is defined by a (DTD) file, which specifies the hierarchical organization of modules 2 through 5 according to ICH guidelines, while regional variations handle module 1. The eCTD backbone primarily consists of the index.xml file, which provides for the entire submission, including titles, references to individual files, and operation attributes (such as new, , or replace) to manage lifecycle. Leaf files, the core elements, are typically in PDF format for , though other types like datasets or images are permitted, and each is referenced within the XML backbone to maintain the without the directly. Submissions are organized into sequences, numbered sequentially (e.g., for initial, 0001 for amendments), allowing regulators to track incremental updates efficiently. Regional backbones, such as the EU's regional.dtd or the FDA's us-regional.xml, extend the core ICH specification to accommodate jurisdiction-specific administrative data in module 1, as outlined in ICH M2 guidelines. Validation of eCTD submissions relies on conformance to these DTDs and ICH M8 specifications for formats, with tools assessing XML syntax, file integrity, and metadata completeness to prevent submission errors. The current eCTD version, 3.2.2, was finalized in 2008 building on the initial release, emphasizing modular XML for common technical sections while supporting regional adaptations. Version 4.0, under development since 2016, introduces enhanced lifecycle management features like granular updates and RESTful APIs, with initial implementations accepted by agencies like the FDA starting in 2024.

Implementation and Validation

The implementation of the Electronic Common Technical Document (eCTD) involves a structured process to ensure submissions align with the Common Technical Document (CTD) framework and meet regulatory requirements for electronic filing. Preparation begins with mapping source documents to the CTD's modular structure (Modules 1–5), where files are organized into predefined directories such as m1 for administrative information or m5 for clinical study reports, using standardized folder and file naming conventions outlined in the ICH eCTD Specification. This mapping is facilitated through the XML backbone file, which defines the hierarchy and references each document's location via leaf elements, ensuring logical navigation across the dossier. Hyperlinking is then incorporated, primarily in PDF files, to enable internal and cross-document navigation; relative paths are used to maintain link integrity, with bookmarks limited to four hierarchical levels for optimal viewer performance. Finally, the submission is compiled into sequences—self-contained units numbered sequentially (e.g., 0000 for initial, 0001 for the first update)—each containing an index.xml file, MD5 checksums for integrity verification, and all mapped files zipped for transmission. Validation of eCTD submissions focuses on technical compliance to prevent rejection, primarily through checking adherence to (DTD) schemas that govern the XML structure and content. The XML backbone and regional files (e.g., us-regional.dtd for FDA) must parse without errors against these schemas, verifying element attributes, hyperlinks, and file references. Specialized tools automate this process; for instance, GlobalSubmit VALIDATE employs a robust engine to scan for compliance with ICH and regional standards, flagging issues like malformed XML or invalid paths before submission, and supports validation for multiple authorities including FDA and . Additional checks include file size limits (e.g., PDFs under 100 MB), embedded fonts to avoid substitution, and checksums to confirm no alterations during transfer. Common errors in eCTD submissions often stem from technical oversights that trigger automated rejections, such as invalid XML structures failing DTD validation or missing metadata in leaf elements, including attributes like 'name', 'manufacturer', or dosage form details. Another frequent issue is the absence of required files, exemplified by the missing ts.xpt dataset in study data submissions, which accounted for 58% of FDA rejections between February 2022 and February 2023, or mismatched Study Tagging Files (STFs) where IDs do not align with dataset identifiers. Remediation follows the ICH and electronic submissions (ES) framework, involving the use of operation attributes in a new sequence to delete erroneous leaves, correct metadata or files, and resubmit without duplicating unchanged content; for example, the 'delete' operator removes invalid entries, while 'replace' updates datasets, ensuring traceability and minimizing dossier bloat. The eCTD lifecycle is managed through incremental sequences that accommodate post-approval changes like variations and supplements, allowing regulators to track the evolving dossier without full resubmissions. Each update constitutes a new sequence with an incremented number, referencing prior sequences via the XML envelope to maintain continuity. Operation attributes such as add new leaves for supplemental data, 'replace' overwrites existing ones for revisions (e.g., updated clinical reports), and 'delete' removes obsolete elements, all while preserving hyperlinks and metadata for seamless integration. This approach supports regulatory activities like amendments or annual reports, coded appropriately in Module 1, and enables advanced management in eCTD v4.0 contexts where contexts of use can be replaced or grouped for complex lifecycles.

Global Adoption and Variations

Regulatory Acceptance by Region

The Common Technical Document (CTD) format was developed under the International Council for Harmonisation (ICH) to standardize regulatory submissions for pharmaceuticals. In the ICH regions, adoption occurred through phased implementation. For Japan, the Pharmaceuticals and Medical Devices Agency (PMDA) made the CTD mandatory for new drug applications since 2003, following voluntary adoption starting in 2001. In the European Union, the European Medicines Agency (EMA) required the CTD as the mandatory format for new drug applications from July 2003. For the United States, the Food and Drug Administration (FDA) designated the CTD as the preferred format in 2003 but mandated it for electronic submissions (eCTD) in 2017, specifically requiring eCTD for New Drug Applications (NDAs), Abbreviated New Drug Applications (ANDAs), and Biologics License Applications (BLAs) starting May 5, 2017. Beyond the ICH regions, the CTD has been widely embraced by non-ICH regulatory authorities to facilitate international harmonization. The (WHO) incorporates the CTD format into its prequalification program for medicines and , requiring submissions in CTD structure to ensure consistency with global standards and reduce duplication for manufacturers. In , began accepting CTD-formatted submissions in 2004 and mandated electronic CTD (eCTD) for most regulatory activities, including drug submissions, starting January 1, 2018. Australia's (TGA) adopted the CTD in 2004, making it the required format for all prescription medicine dossiers, with electronic submissions following suit in subsequent years. Implementation of the CTD across regions has involved phased transitions to allow stakeholders to adapt. For instance, the FDA's shift to mandatory eCTD included a voluntary period from 2003, with full enforcement by 2017 to streamline reviews and enhance . Similar transitions occurred in other jurisdictions, such as Health Canada's progressive mandates from 2004 onward, culminating in eCTD requirements by 2018. These timelines reflect a global effort to balance innovation with regulatory efficiency. By 2025, the CTD format is accepted in numerous countries worldwide, including all ICH members and many others, promoting broader harmonization and expediting access to safe pharmaceuticals.

Regional Adaptations and Differences

While the core modules (2 through 5) of the (CTD) are harmonized internationally under ICH guidelines, Module 1 is adapted to incorporate region-specific administrative and prescribing requirements to meet local regulatory needs. In the , the () extends Module 1 to include the Summary of Product Characteristics (SmPC), which provides detailed prescribing information for healthcare professionals, and the (RMP), a standalone document outlining the medicine's safety profile, activities, and risk minimization measures. The RMP references SmPC sections, such as those on special warnings (4.4) and undesirable effects (4.8), to ensure alignment between product labeling and post-authorization safety monitoring, without duplicating content. These elements are submitted as part of the initial marketing authorization application or updates via the eCTD format. For the , the (FDA) customizes Module 1 with requirements such as environmental assessments under section 1.12.14, which evaluate the potential ecological impact of the drug product in accordance with 21 CFR 25.15(d) for abbreviated new drug applications (ANDAs) and 21 CFR 314.50(d)(1)(iii) for new drug applications (NDAs) or biologics license applications (BLAs). Additionally, certifications appear in section 1.3.5.2, detailing statements on validity and infringement under 21 CFR 314.50(i), 314.52(e) for NDAs/BLAs, and 314.94(12) for ANDAs, supporting exclusivity claims and regulatory review processes. These components are integrated into the eCTD backbone for electronic submissions. In , the (PMDA) employs a region-specific approach with a dedicated J-module equivalent within Module 1 for administrative data, while Modules 1 and 2 were traditionally prepared in to facilitate local review; however, since September 2024, English is accepted under specific conditions (e.g., for applicants without a branch). PMDA mandates inclusion of local data, particularly bridging studies in Module 5, to demonstrate and in the population, addressing ethnic differences in and as per regulatory expectations for new drug applications. Among emerging markets, Brazil's National Health Surveillance Agency (ANVISA) adapts the CTD by requiring additional local stability data in Module 3, aligned with WHO Zone IVb conditions (30°C/75% RH) to account for impacts on products, beyond standard ICH guidelines. As a full ICH member since 2022, ANVISA participates in harmonization efforts, incorporating CTD modules while mandating specific formats for stability studies, such as explicit short-term and freeze-thaw assessments, to ensure product integrity in local conditions.

Benefits and Challenges

Advantages for Stakeholders

The Common Technical Document (CTD) provides significant advantages to regulatory authorities by standardizing the format of drug submissions, which accelerates the review process and reduces the for assessors. Prior to , reviewers faced challenges in familiarizing themselves with unique application formats; the CTD's consistent structure mitigates this, allowing for more efficient evaluation of technical data across quality, nonclinical, and clinical modules. Furthermore, it enables easier comparative assessments between similar products or applications, fostering better decision-making and resource allocation among agencies like the FDA, , and PMDA. In targeted areas such as pharmaceutical development sections (e.g., CTD 3.2.S.2), ICH guidelines associated with the CTD have demonstrated reductions in review queries, directly saving assessor time and minimizing follow-up requests. Pharmaceutical industry stakeholders benefit from the CTD through substantial reductions in preparation costs and efforts, as the modular format permits extensive reuse of content across ICH regions without major reformatting. This eliminates the redundancies of preparing separate dossiers for the , , and , streamlining global development pipelines and enabling companies to allocate resources more effectively toward research and innovation. Surveys of industry participants in harmonized registration initiatives, such as the ZaZiBoNa collaborative in , confirm high satisfaction with these efficiencies, with a majority reporting time and resource savings alongside improved multi-market access for their products. Patients ultimately gain from the CTD's , which expedites regulatory approvals and enhances the of and data for more robust post-market . By shortening timelines in collaborative frameworks—such as achieving approval times of 12 months in the ZaZiBoNa initiative—the format accelerates access to safe and effective medicines worldwide. These advantages are amplified by the electronic CTD (eCTD), which supports automated validation and lifecycle management for even greater efficiency. Overall, ICH evaluations highlight the CTD's role in delivering substantial economic impacts, including amortized costs for guideline implementation through reduced queries and streamlined submissions, though specific per-drug figures vary by context and region. Recent updates, such as the ICH M4Q(R2) guideline finalized in June 2025, further refine the quality module to enhance digital compatibility and global .

Common Implementation Hurdles

One major hurdle in implementing the Common Technical Document (CTD) is organizing and mapping data from documents to the standardized five-module , which requires meticulous of quality, nonclinical, and clinical information to avoid misplacement and submission rejections. This process is particularly challenging for companies with extensive historical archives, as converting paper-based or non-standard digital files to the electronic CTD (eCTD) format demands significant effort to ensure and , often leading to delays in regulatory filings. Additionally, staff training is essential to build competency in this restructuring, as unfamiliarity with module-specific requirements can result in errors that compromise integrity. The financial and resource demands of CTD adoption pose substantial barriers, especially for small pharmaceutical companies, where initial investments in eCTD software, validation tools, and system setup can be prohibitive. timelines typically span 9 to 18 months, involving ongoing costs for maintenance and , which strain limited budgets and personnel in resource-constrained settings. These challenges are amplified for smaller firms lacking dedicated regulatory teams, potentially delaying market entry and increasing operational inefficiencies. Compliance with evolving CTD guidelines presents ongoing difficulties, as frequent revisions to International Council for Harmonisation (ICH) standards require continuous updates to submission processes, while regional discrepancies—such as varying Module 1 requirements across agencies like the FDA, , and those in emerging markets—often necessitate rework and multiple versions. For instance, unique local mandates in countries like demand additional documentation placements not aligned with core ICH formats, leading to inconsistencies and heightened rejection risks. These variations, rooted in differing interpretations of harmonized guidelines, can result in non-compliance notices and extended review times. To address these hurdles, organizations can leverage ICH-provided resources and workshops to enhance expertise in CTD structuring and compliance. Partnering with specialized software vendors for eCTD publishing and validation tools helps streamline and reduce technical errors, while adopting a phased implementation approach—starting with core modules and gradually incorporating regional adaptations—minimizes resource strain and facilitates smoother transitions. In low-resource environments, international technical assistance programs further support to overcome infrastructural gaps.

References

  1. [1]
    CTD - ICH
    This guideline presents the agreed upon common format for the preparation of a well-structured Common Technical Document (CTD) for applications that will be ...
  2. [2]
    [PDF] ICH guideline M4 (R4) on common technical document (CTD) for the ...
    Mar 19, 2021 · This guideline presents the agreed upon common format for the preparation of a well- structured. Common Technical Document for applications ...
  3. [3]
    Electronic Common Technical Document (eCTD) - FDA
    Oct 4, 2024 · The eCTD is the standard format for submitting applications, amendments, supplements, and reports to FDA's Center for Drug Evaluation and Research (CDER)
  4. [4]
    Understanding the Common Technical Document (CTD)
    Jun 16, 2014 · The Common Technical Document (CTD) is a set of specifications for a dossier for the registration of medicines.Purpose · Terminology · Module 2: Common technical... · Module 3: Quality
  5. [5]
    [PDF] VICH and its role for authorisation of veterinary medicinal products
    May 5, 2010 · An application for approval of a new veterinary medicinal product undergoes an investigation by a committee of the Pharmaceutical Affairs Sub- ...
  6. [6]
    [PDF] Postapproval Changes to Drug Substances Guidance for Industry
    Comments and suggestions regarding this draft document should be submitted within 60 days of publication in the Federal Register of the notice announcing ...
  7. [7]
    ICH Official web site : ICH
    - **Formation**: ICH was established in 1990 as the International Conference on Harmonisation.
  8. [8]
    [PDF] ICH Overview - FDA
    Launched in 1990 by the US, EU, and Japan. Canada, Swissmedic and WHO as observers. • Well-defined objectives: – To improve efficiency of new drug development ...
  9. [9]
    How Did We Get Here? A History of eCTD and Prospects for eCTD 4.0
    The CTD became the mandatory or strongly recommended format for most types of drug applications in the US, Europe, and Japan over the following years and ...Missing: inconsistencies CPMP
  10. [10]
    ICH M4 Common technical document (CTD) for the registration of ...
    Feb 1, 2004 · Guidance on the organisation of the information to be presented in registration applications for new pharmaceuticals (including biotechnology-derived products).<|control11|><|separator|>
  11. [11]
    [PDF] Overview of recent developments in ICH - Thomas Lonngren
    ICH Structure. The six parties: Committed to implementation of guidelines. Regulators : EC (EU), MHLW (JP), FDA (US). Industry: EFPIA, JPMA, PhRMA. Secretariat ...
  12. [12]
    ICH: M 4 Q: Common technical document for the registration of ...
    Jul 1, 2003 · Adopted Reference Number: CPMP/ICH/2887/99 Legal effective date: 01/07/2003. English (EN) (269.43 KB - PDF). First published: 01/07/2003 Last ...
  13. [13]
    [PDF] M4Q_R1_Guideline.pdf - ICH
    This document is intended to provide guidance on the format of a registration application for drug substances and their corresponding drug products as defined ...
  14. [14]
    [PDF] This Guideline has been developed by the appropriate ICH Expert ...
    Jun 15, 2016 · This Guideline has been developed by the appropriate ICH Expert Working Group and has been subject to consultation by the regulatory parties ...
  15. [15]
    ICH Quality Guidelines: Present Initiatives & ISPE Involvement
    This document is intended to provide guidance regarding Good Manufacturing Practice (GMP) for the manufacturing of Active Pharmaceutical Ingredients (APIs) ...
  16. [16]
    [PDF] Electronic Common Technical Document (eCTD) v4.0 ... - FDA
    REVISION HISTORY. DATE. VERSION SUMMARY OF REVISIONS. 2019-12-03 1.0. Initial Revision. 2021-01. 1.1. Revisions based on comments received during a public ...Missing: milestones | Show results with:milestones
  17. [17]
    Digital Innovation in Medicinal Product Regulatory Submission ... - NIH
    Learnings from CTD and eCTD inform us that the path to a new model will not be quick nor linear, however, with digital technology available the pace of change ...<|control11|><|separator|>
  18. [18]
    [PDF] The Comprehensive Table of Contents Headings and Hierarchy - FDA
    Jul 6, 2005 · 3.2.S.3.1 Elucidation of Structure and other Characteristics. 3.2.S ... Synopsis (ICH E3, section 2). Study report body (E3 1, 3 to 15).
  19. [19]
    [PDF] eCTD specification 3.2.2 - ICH
    Jul 16, 2008 · The XML eCTD DTD describes the hierarchical structure according to the CTD as defined by the ICH M4. Expert Working Group. It includes multiple ...
  20. [20]
    Easy as eCTD? - The Medicine Maker
    ... 100,000 pages. To standardize the application dossier, the Common Technical Document (CTD) was developed by the International Conference on Harmonization of ...
  21. [21]
    [PDF] Guidance for Industry M4: The CTD - General Questions and Answers
    This is one in a series of guidances that provide recommendations for applicants preparing the. Common Technical Document for the Registration of ...Missing: formatting | Show results with:formatting
  22. [22]
    [PDF] M4E(R2): The CTD — Efficacy Guidance for Industry - FDA
    Module 1: Administrative Information and Prescribing Information. 1.1 Table of Contents of the Submission Including Module 1. 1.2 Documents Specific to Each ...
  23. [23]
    the Common Technical Document (CTD) : Module 1 region specific
    The module contains the following elements: 1.0 Cover letter. 1.1 Comprehensive table of contents. 1.2 Application form. 1.3 Product information documents. 1.4 ...
  24. [24]
    CTD Module 1: Administrative information and prescribing ...
    Sep 28, 2007 · This guidance explains the administrative and prescribing (dossier) information you need to support the registration or variation of a prescription medicine.
  25. [25]
    [PDF] Guidance for Industry - FDA
    Diagrammatic Representation of the ICH Common Technical. Document. Not Part of the CTD. 5. Page 11 ... length will usually range from 50 to 400 pages. Table of ...
  26. [26]
    [PDF] M4S_R2_Guideline.pdf - ICH
    MODULE 2. ORGANISATION OF MODULE 4. Current Step 4 version dated 20 December 2002. This Guideline has been developed by the appropriate ICH Expert Working ...
  27. [27]
    [PDF] EFFICACY - M4E(R2) - ICH
    Jun 15, 2016 · MODULE 2 : COMMON TECHNICAL DOCUMENT SUMMARIES ... The periodic safety update reports can be included in Module 5. Details of the ...<|separator|>
  28. [28]
    [PDF] development and manufacture of drug substances (chemical entities ...
    May 1, 2012 · ICH Q11 is a guideline for the development and manufacture of drug substances, recommended for adoption to the EU, Japan, and USA regulatory ...
  29. [29]
    [PDF] Impurities in new drug substances Q3A (R2) - ICH
    Oct 25, 2006 · Impurities in new drug substances are addressed from two perspectives: Chemistry Aspects include classification and identification of ...
  30. [30]
    [PDF] Q1A(R2) Guideline - ICH
    The purpose of stability testing is to provide evidence on how the quality of a drug substance or drug product varies with time under the influence of a ...
  31. [31]
    [PDF] Q5A(R1) - ICH
    This document is concerned with testing and evaluation of the viral safety of biotechnology products derived from characterised cell lines of human or animal.
  32. [32]
    [PDF] ich-m-4-s-common-technical-document-registration ...
    Note: The sequence of M4 Common Technical Document for the Registration of Pharmaceuticals for Human Use is: M4. - Organisation of Common Technical Document.Missing: size | Show results with:size
  33. [33]
    [PDF] S9 Step 5 Nonclinical evaluation for anticancer pharmaceuticals
    The purpose of this guidance is to provide information to assist in the design of an appropriate program of nonclinical studies for the development of ...
  34. [34]
    [PDF] Structure And Content of Clinical Study Reports E3 - ICH
    The objective of this guideline is to allow the compilation of a single core clinical study report acceptable to all regulatory authorities of the ICH regions.
  35. [35]
    [PDF] Guidance for Industry - Integrated Summaries of Effectiveness ... - FDA
    Generally, the Module 2 clinical summary sections (hereafter clinical summary sections) follow the outline of the ISE and ISS described in ICH M4E; however, ...<|control11|><|separator|>
  36. [36]
    Electronic Common Technical Document (eCTD) v3.2.2 - ICH
    The Electronic Common Technical Document (eCTD) allows for the electronic submission of the Common Technical Document (CTD) from applicant to regulator.Missing: hierarchical | Show results with:hierarchical
  37. [37]
    [PDF] eCTD Backbone File Specification for Modules 2 through 5-3.2.2 - FDA
    Jul 16, 2008 · The XML eCTD DTD describes the hierarchical structure according to the CTD as defined by the ICH M4. Expert Working Group. It includes multiple ...
  38. [38]
    [PDF] The eCTD Backbone Files Specification for Module 1 - FDA
    Nov 1, 2012 · xml) includes administrative information and information for each file submitted in Module 1. The backbone file contains an XML element named.
  39. [39]
  40. [40]
    [PDF] M2 eCTD: Electronic Common Technical Document Specification
    The specification for the eCTD is based upon content defined within the CTD issued by the ICH M4 EWG. The CTD describes the organization of modules, sections ...
  41. [41]
    ICH electronic Common Technical Document - eCTD v4.0
    As of May 2024, the Implementation Guide and Controlled Vocabulary documents were split into two different packages to enable Controlled Vocabulary Versioning.
  42. [42]
    eCTD Submission Standards for eCTD v4.0 and Regional M1 - FDA
    CDER and CBER are accepting new regulatory applications in eCTD v4.0 format as of September 16, 2024. More information can be found on the eCTD page.<|control11|><|separator|>
  43. [43]
    [PDF] eCTD Technical Conformance Guide - FDA
    Future revisions will be posted directly on the eCTD website3 and the revision history page of this document will contain sufficient information to indicate ...
  44. [44]
    GlobalSubmit™ eCTD Software - Certara
    Publish, validate, and review your eCTD submissions with GlobalSubmit eCTD Software, eliminating risk and expediting regulatory acceptances.GlobalSubmit eCTD ソフトウェア · GlobalSubmit™ REVIEW
  45. [45]
    [PDF] Most Common Submission Issues and FDA Plans for eCTD v4.0
    Rejected Submissions: February 15, 2022 – February 15, 2023. ❖ 1734 (58%) is the most common error and rejection reason for a missing ts.xpt. Notes: Metrics ...
  46. [46]
    [PDF] CTD General Questions and Answers
    Sep 1, 2002 · Are expert reports still required for submissions under the CTD format? No. Expert Reports are replaced by Module 2. (N.B. For specific European ...
  47. [47]
    [PDF] An overview of the Common Technical Document (CTD) regulatory ...
    The aim of the CTD was simple – it would provide a common format for the technical documentation that would significantly reduce the time and resources needed ...
  48. [48]
    CTD Preparation & Submission
    The CTD is mandatory for WHO prequalification, with modules 1-5. Submission deadlines are Jan 31, May 31, and Sept 30. Manufacturers must engage with NRAs.
  49. [49]
    [PDF] WHO Guidelines on submission of documentation for the pilot ...
    These guidelines present the agreed-upon common format for the preparation of a well-structured. CTD for PDs that will be submitted to WHO. A common format for ...
  50. [50]
    Filing submissions electronically - Canada.ca
    Companies are required to file submissions electronically to Health Canada in either Electronic Common Technical Document (eCTD) format or non-eCTD format.
  51. [51]
    Health Canada confirms mandatory use of eCTD format from 1st ...
    Health Canada confirms mandatory use of eCTD format from 1st January 2018. From the beginning of next year, Health Canada will only accept submissions for ...
  52. [52]
    NeeS AU module 1 and regional information v2.0
    Jan 1, 2018 · All dossiers must be structured in accordance with the Common Technical Document (CTD) format, a format Australia initially adopted in 2004.
  53. [53]
    FDA & HC eCTD Deadlines: Quick Submission Overview - Freyr.
    eCTD Submissions – A quick look over the FDA & HC deadlines · New Drug Applications (NDAs) – 5th May 2017 · Biologics License Application (BLAs) – 5th May 2017 ...<|control11|><|separator|>
  54. [54]
    Health Canada updated notice for Mandatory use of eCTD Format
    Feb 6, 2019 · From since 2004, Health Canada is accepting regulatory activities in eCTD format. As of December 2018, 93 percent of regulatory activities (Part ...Missing: CTD | Show results with:CTD
  55. [55]
    Tag: CTD global requirements - Pharma Regulatory
    Aug 10, 2025 · CTD: Adopted in ICH regions (US, EU, Japan) and recognized in more than 100 countries globally. Module 1 varies by country but Modules 2–5 ...
  56. [56]
    [PDF] Guidance on the format of the risk management plan (RMP) in the EU
    Oct 31, 2018 · An RMP includes a safety specification, pharmacovigilance plan, and risk minimisation plan. It should be concise, scientifically based, and ...
  57. [57]
    [PDF] Electronic Common Technical Document Specification
    This module contains administrative information that is unique for each region. There will be local requirements for both the content and electronic component ...Missing: J- | Show results with:J-
  58. [58]
    Japan (PMDA) - Pharma Regulatory
    Aug 2, 2025 · Local Clinical Data Requirement: PMDA often requires local trial data (bridging studies) to confirm efficacy and safety in Japanese populations.
  59. [59]
    ANVISA: an introduction to a new regulatory agency with many ...
    Dec 12, 2018 · Although Brazil is now an ICH official member, ANVISA requires WHO Zone IVb for stability storage conditions, including Active Pharmaceutical ...
  60. [60]
    [PDF] COMPARITIVE STUDY BETWEEN ICH GUIDELINE AND ANVISA ...
    The ICH Q1 guideline provides guidance on the core stability data, which are required for new drug substances and products. The currently valid Brazilian ...
  61. [61]
    [PDF] 30th Anniversary Publication - Portal Gov.br
    Common Technical Document (CTD). The agreement to assemble all Quality, Safety and Efficacy information in a common format has revolutionised the regulatory.<|control11|><|separator|>
  62. [62]
    The Complexity of eCTD Compliance: Common Challenges and ...
    Oct 2, 2024 · eCTD compliance challenges include understanding the structure, regional variations, converting legacy data, managing lifecycle submissions, ...
  63. [63]
    [PDF] ELECTRONIC COMMON TECHNICAL DOCUMENT (eCTD)
    It was seen as a way for. FDA reviewers to have rapid access to report and data together, in a format that allowed efficient and high-quality analysis of data.<|separator|>
  64. [64]
    None
    ### Challenges in Implementing CTD in Developing Countries
  65. [65]
    A Review Article on the Common Technical Document (CTD ...
    Aug 9, 2025 · However, challenges remain in areas such as region-specific requirements and the adaptation of legacy documentation. As global regulatory ...<|control11|><|separator|>
  66. [66]
    Common Technical Document (CTD) Challenges in Emerging Markets
    Jun 20, 2024 · Having a universal and uniform Common Technical Document (CTD) for all markets, major and emerging, is an aspiration of all pharmaceutical companies.