Dmf
Dmf
Some images are taken – freely available from the internet for a
diagrammatic representation of the content and the source is
acknowledged
Production Team
Research n Development
IPR; Process; Analytical; Packaging Outsourced Facilities
CMO, CRO, Testing
APIs manufactured by
chemical synthesis,
the fermentation process
APIs derived from plants and animals (well-characterized) and
Semi-synthetic drug substances from plant and animal origin (well-
characterized) fall under the purview of one set of Guidance
EU – Multiple Agencies and multiple registration procedures for the drug
product (and reviewers from different National Agencies)
Need to have global approach to regulatory issues and build a strong working
knowledge of the countries and their regulators across cross functional teams
The ultimate goal is to sell the generic drug product to the human
population using “compliant” APIs on “Day 1”. The safety and efficacy of
the formulation is dependent upon the quality of the APIs and hence the
importance
Need for total co-operation and transparency between API Manufacturer and Drug
Product Manufacturer
Elemental Impurities
Need to follow the “commitments” made in submission strictly & any change
need to be reported through the appropriate regulatory procedure applicable
for the respective jurisdiction
©Raaj GPRAC, All rights reserved
CFR Regulations
What are the CFR Regulations?
GALP
GLP –Good Laboratory Practices Good Automated Laboratory Practices
21CFR Part 58 EPA Directive 2185 [ 1995 Ed.]
40CFR Part 160[EPA]
RISK MONITORING
Check if safety 26/
measures work
16 August 2020 Raaj DMF Workshop August 2020
Data integrity
Data integrity is critical to regulatory compliance, and the fundamental reason for
21 CFR Part 11. It means data should be
Attributable
Legible
Contemporaneous
Original
Accurate
Legible
Available
Retrievable
Complete
Consistent
*
16 August 2020 Raaj DMF Workshop August 2020 29
Acceptance Criteria
Acceptance Criteria for impurities are Continuously Evolving due to –
Increase in Scientific Knowledge wrt inherent properties of the API
Risk Assessment by various Approaches /Tools available on date
Inclusion of DS/DP monographs in different Pharmacopoeieas
Revision of existing Monographs following toxic qualification or re-assessment of say residual solvents
or metallic residues.
Case studies following an increasing number of Applications filed with varied ROSs of APIs following
deficiencies raised by Regulatory Agencies and Responses and Justifications provided by
Pharmaceutical Industry particularly to the Different Member states of the European Union
Dosage – MDD and Duration of Treatment
Development of More sophisticated Analytical techniques to detect, resolve and quantify Impurities
adequately with acceptable LOD/LOQs
©Raaj GPRAC, All rights reserved
Regulatory Expectations and Industry Thought
Processes – The Gaps
Ref . : ISPE Presentation on QbD – November 2016
Gaps between Regulatory Expectations and Industry Thought Processes
Raw Data
Software not validated
Raw Material Vendors
Equipment not validated/calibrated
Change Control/Unauthorized Access
Up dation of Validation - Most current
Reference & Working Standards
Pharmacopoiea & General Chapters for all tests
Clinical trials are a critical and well-known part of the drug discovery, development and
approval process. Given the importance of drug safety, there are a number of additional
testing processes for the assessment of drug safety and efficacy and they are equally
important. Stability testing is one such example.
As an important step in the drug approval process, stability testing assesses how the quality
of a drug substance/product (& its packaging) varies with time under the influence of
environmental factors, including temperature , humidity & light.
The process determines whether any physical, chemical or microbiological changes affect the
efficiency & integrity of the final product, thereby ensuring that a pharmaceutical product is
safe & effective, irrespective of the market it will be supplied.
©Raaj GPRAC, All rights reserved
Introduction & Background[Contd.]
The climatic data that defines these regions is related to the Mean Kinetic
Temperature (MKT), which is a widely used measure in the pharmaceutical industry to
express the overall effect of temperature fluctuations during storage or transit.
Ensuring the same is practiced, the United States Food and Drug
Administration (USFDA) has set forth deadline for DMF eCTD
submissions, i.e. 05/05/2018, post which submissions will not be
received. Other Agencies have also done or in the process of doing so.
Mandatory From:
New Market Authorizations - Dec 31, 2020
- Follow-up Submissions – Dec 31, 2025
As of May 5, 2018, the U.S. Food and Drug Administration (FDA) requires
electronic common technical document (eCTD) format for Type II, Type IV, and
Type V drug master file (DMF) submissions, including:
New DMFs
Annual Reports
Amendments
Supplemental Reports
FDA states that it will reject non-eCTD DMF submissions of these types.
The term Drug Master File (DMF) is commonly used to describe a submission to a
competent authority that may be used to provide certain confidential detailed
information about facilities, processes or articles used in manufacturing of the API
or other materials.
In the US, the DMF is generally submitted to support the IND (Investigational New
Drug), NDA (New Drug Application), ANDA (Abbreviated New Drug Application) or
other DMFs
In the EU, the DMF is generally submitted to support the MAA
(Marketing Authorization Application)
The review of the new DMF does not start automatically. The review
is triggered only after the submission of the drug product application
which refers the DMF
By submitting the LOA, the holder of the DMF basically authorizes the
respective authority to refer to the confidential information contained
in the DMF in support of the application submitted by the entity to
whom the LOA has been issued by the DMF holder
- General Information
- Manufacture
- Characterization
- Control of Drug Substance
- Reference Standards or Materials
- Container Closure System
- Stability
Filing of the DMF in the US is also at a cost now since October 2012
;in the EU, the CEP filing or the Amendment filing involves a certain
cost. Italy however charges for DMF filings in Europe.
Quality System assures overall compliance with cGMPs and internal procedures and
specifications
Packaging and Labeling System includes measures and activities that control
the packaging and labeling of intermediates and APIs
Letter of Access: In some cases a DMF holder will call the permission to reference a DMF a
“Letter of Access.” (Phrase used in Europe). In the US, this is called a “Letter of Authorization”
(LOA). An LOA does not permit anyone except FDA to “Access” i.e. “read” the DMF
Physical Media (CD-ROM, DVD or USB drive)–Holder sends the DMF on physical
medium in eCTD format to the address on previous slide
–See the Transmitting Specifications website when transmitting via physical
media (CD\DVD)
http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/Forms
SubmissionRequirements/ElectronicSubmissions/UCM163567.pdf
Acceptance of digital signatures the same as for any other submission to FDA
Follow the DMF Guidance and additional information on DMF Web site.
Pre-assigned Number
A pre-assigned number is required for an EDMF. May also be obtained for paper
DMF. See “Requesting a Pre-Assigned Application number”
http://www.fda.gov/Drugs/DevelopmentApprovalProcess/FormsSubmissionRequi
rements/Electronic Submissions/ucm114027.htm
Administrative information. For complete list of information to include see DMF Web site. Make
sure to include Telephone number, fax number and e-mail address for the responsible individual
(contact person)
NEW !! on Web site: List of Referenced applications e.g. DMF for intermediates
Technical information
http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInf
ormation/Guidances/UCM261078.pdf
Other guidances relevant to the specific market
Always refer to the most updated Guidance on the respective country site
©Raaj GPRAC, All rights reserved
Letter of Authorization (LOA)
The DMF will be reviewed ONLY when it is referenced in an
Application or another DMF.
An LOA does two things: Grants FDA authorization to review the
DMF
Grants the Authorized Party the right to incorporate the
information in the DMF by reference.
The holder MUST submit an LOA (2 copies for paper) to the DMF
THEN send a copy to the APPLICANT
APPLICANT submits copy of LOA in their Application. ONLY
mechanism to trigger complete technical review of the DMF.
Can convert paper DMF to EDMF but once electronic, cannot submit paper,
even for LOA.
ELECTRONIC DMFs
If changes have been made but not reported to DMF, reviewer can
waste valuable time (on the APPLICANT’s clock) reviewing obsolete
information.
Entry of a Reactivation into DARRTS changes status to “Active” and the DMF
is available for review.
If more information is needed to complete the review, a list of the information
needed is communicated to the holder in an Information Request (IR) Letter
If the information in the DMF cannot support approval of the application that
references it FDA sends a Deficiency Letter (DEF) or Complete Response (CR)
Letter (specific for Type II DMFs under GDUFA), and then [next slide]
The APPLICANT is notified that information has been requested for the DMF.
The letter to the APPLICANT is either an IR or CR Letter.
If no information needed for DMF, No letter to DMF holder except for “No
Further Comments” letter specific for Type II DMFs under GDUFA.
Do not use the word “authorize” in appointing an agent. This can be easily
confused with a Letter of Authorization. Use the word is “appoint.”
©Raaj GPRAC, All rights reserved
New GDUFA DMF Requirements
DMF fee – when and how to pay
Since GDUFA does not apply to any other type of DMF or to Type II
DMFs used to exclusively support NDAs or INDs, there are no fees for
these types of DMFs.
DMF Fee is a one-time fee & is different from the facility fee related to the drug
substance manufacturing facility
Payment of the DMF fee triggers the Completeness Assessment process in OGD
DMF Fee is required when referencing ANDA is submitted
DMF Fee may be paid independent of a referencing ANDA in order to get a
completeness assessment and be to listed on the FDA’s “Available for Reference”
webpage.
There are risks of delaying the ANDA filling if DMF fee has not been paid
The DMF letters will issue before the ANDA CR letter for a given review cycle to
comply with ANDA CR requirements.
DMF CR letters will be referred to in the ANDA CR letter along with instructions to
the applicant not to respond until the DMF holder has indicated that they have
submitted a complete response to the DMF CR letter.
DMF responses from the holder must address all issues raised in the Chemistry
and Microbiology sections of the letter or they will not be reviewed.
Note that statements in the DMF amendment deferring response to a future
submission are not acceptable.
DMF must notify the ANDA sponsor when they submit their response
Issued at approval of the ANDA, not when the DMF is first deemed adequate
Two things must happen for the DMF to be considered “available for reference”
DMF Fee must be paid
DMF must pass a “completeness assessment”
ANDAs can only be filed by OGD if all DMFs for the drug substance(s) are “available
for reference” .
However, to maintain the confidentiality and to refer the same information for
multiple drug product applications, as a practice, manufacturers of API/Excipients
file the information as Drug Master Files (DMFs), Active Substance Master Files
(ASMFs), and Certificates of Suitability (CEPs) as per the region-specific
requirements.
Once API/Excipient information is filed with HAs and is accepted, same can be
referred through Letter of Access (LoA) to multiple applications to avoid
duplication of API information for different drug product submissions.
©Raaj GPRAC, All rights reserved
New Market Authorizations - APIs
Regulatory submission requirements for API (Drug Substance)/ Excipients are very
specific and different for every Health Authorities .
Hence, expertise and experience in filing DMFs, ASMFs, CEPs, and active
ingredient master files to global Health Authorities help in quick review approval
of drug product applications.
Organizations need very strong Regulatory team with expertise in handling new
submissions to all major Health Authorities.
The Japanese DMF system is similar to the one run by the US Food and Drug
Administration (FDA). The Japanese DMF system makes it possible for API
manufacturers to register their products with the PMDA directly. In this way, API
manufacturers avoid having to turn over sensitive information when finished drug
manufacturers use their product. For each finished drug application involving the
API, the PMDA simply pulls the information it needs from the DMF.
In Japan, the API or excipient manufacturer is in charge of DMF registration. If the
manufacturer lacks its own office in Japan, it must appoint a person or a business
entity called an In-Country Caretaker (ICC) to apply for them. The ICC must fill out
the DMF application and answer all PMDA questions, both during and after API
registration.
Finished drug manufacturers must cite all relevant DMFs during their own drug
registration. They are encouraged to attach DMF registration certificate copies to
their own applications. In the course of the PMDA review for the finished drug
product, all questions about the DMF products are directed to either the DMF
license holder or to the ICC.
In Japan, the Drug Master File (DMF) is called “Master File” or “MF”.
New excipients and pre-mix excipients with different composition ratios from the
existing ones
In China, API applications may be submitted together with applications for the
finished drug product. When this occurs, the API application may pull clinical data
and test results from the application for the finished drug product.
However, foreign manufacturers of finished drugs that are imported do not have
to submit their foreign DMFs to the CFDA. If they so choose, they may provide
information from foreign DMF filings to take the place of their CPP (Certificate of
Pharmaceutical Product).
Finally, the draft regulations also require that domestic API manufacturers
periodically conduct quality audits of their suppliers. These audit reports would
need to be included along with the DMF application.
CFDA, now NMPA, had a pre-market approval policy that allowed APIs, excipients
and packaging materials to be registered separately with pharmaceutical product
application. The process usually took longer and was expensive. The new system
allows these manufacturers to file the DMF reducing the approval time and
respective cost involved. Pharmaceutical manufacturers can refer to the DMF
code while submitting the drug application making the process more efficient.
With respect to drug packaging, the Announcement amends the registration dossier
requirements.
Notably, data requirements for certain categories may be reduced for drug
packaging materials where the data needed for registration are determined
according to the risk level and use information about a particular drug packaging
material.
For example, if a drug packaging material has not been used in pharmaceutical
applications but has lawful use in direct contact with food as food packaging
materials, there may be a reduction in data requirements.
Some regulatory guidance on specification limits for residues of metal catalysts and reagents is in
EMA only. An ICH guideline Q3D ensures that new requirements have the necessary input of the
regional regulatory authorities, to the benefit of regulators, industry, and public health.
The existing ICH Q3A guideline classifies impurities as organic, inorganic, and residual
solvents.
The Q3A and Q3B guidelines effectively address the requirements for organic impurities.
An additional guideline Q3C was developed to provide clarification of the requirements
for residual solvents.
The new guideline Q3D provides similar clarification of requirements for metals, which
are included in the ICH inorganic impurities classification.
©Raaj GPRAC, All rights reserved
Q3D: Impurities: Guideline for Metal Impurities
A harmonised approach for control of metal impurities, including the
list of specific metals to be limited.
These metal impurities may arise from the drug substances, excipients, or
manufacturing processes used for drug products, and may include catalysts,
reagents, ligands, heavy metals or other residual metals, such as those due to the
material source (e.g. Pb, Hg, As,Cd).
Although the risk factors for metal contamination have changed dramatically, the
standards for their control have changed little for more than 50 years, and most Heavy
Metals limits have little basis in toxicology.
So, arose the potential need and benefit of a harmonised guideline for metal impurities.
Previously, the Expert Working Group on Quality developed the Q3A, Q3B,and Q3C
guidelines in order to provide a harmonised approach to limiting impurities. Toxicologists
also provided input on the subject of impurities, particularly with regard to toxicity of
process-related impurities and residual solvents.
©Raaj GPRAC, All rights reserved
Regulatory requirements- Key Guidances and
References
CTD sections related to Stability Testing
General Structure/Drug Product 3.2 P Drug Product 3.2 P.1 Description and
Composition of the Drug Product 3.2 P.2 Pharmaceutical Development 3.2 P.3
Manufacture 3.2 P.4 Control of Excipients 3.2 P.5 Control of Drug Product 3.2 P.6
Reference Standards or Materials 3.2 P.7 Container Closure System 3.2 P.8 Stability
4) ICH Q1D: Bracketing and matrixing designs for stability testing of new drug substances and
products
- General Information
- Manufacture
- Characterization
- Control of Drug Substance
- Reference Standards or Materials
- Container Closure System
- Stability
In the year 1995, the European Union (EU) established an organization called the
European Medicines Evaluation Agency (EMEA, now known as EMA) for a
centralized approval process. The EMEA co-ordinates drug license applications
within the European Union (EU)
For the EU, it is for the drug products filed through the following procedures:
- DCP (Decentralized Procedure)
- Mutual Recognition Procedure (MRP)
- CP (Centralized Procedure)
- National Procedure