Guidelines For Registration of Allopathic Drugs CTD
Guidelines For Registration of Allopathic Drugs CTD
Version No. : 02
FDA/DRI/DER/GL-RAD/2013/02
ACKNOWLEDGEMENT
The Food and Drugs Authority (FDA) acknowledges the technical support of the World
Health Organization (WHO) and the International Conference on Harmonisation (ICH) in
the development of this guideline.
i
FDA/DRI/DER/GL-RAD/2013/02
TABLE OF CONTENTS
1.2.13 Name and complete address(es) of the manufacturer(s) of the FPP ....................... 8
1.2.14. Manufacturing and registration/international registration status .............................. 9
ii
FDA/DRI/DER/GL-RAD/2013/02
iii
FDA/DRI/DER/GL-RAD/2013/02
iv
FDA/DRI/DER/GL-RAD/2013/02
MODULE 4: Nonclinical Study Reports For New Chemical Entities Only .................................... 69
4.1 Table of Contents of Module 4 ....................................................................................................... 69
4.2 Study Reports ................................................................................................................................... 69
4.2.1 Pharmacology........................................................................................................................ 70
4.2.2 Pharmacokinetics.................................................................................................................. 70
4.2.3 Toxicology .............................................................................................................................. 70
v
FDA/DRI/DER/GL-RAD/2013/02
ONLY) ........................................................................................................................................... 82
5.3.1 Requirements for Animal Safety ..................................................................................... 82
5.3.2 Requirements for Human Safety .................................................................................... 83
4. LANGUAGE ........................................................................................................................... 85
5. DATA PRESENTATION ......................................................................................................... 85
6. OFFICIAL REFERENCES AND TEXTS ................................................................................. 85
7. SUBMISSION OF APPLICATION .......................................................................................... 86
7.1 New applications for registration .................................................................................................... 86
7.2 Applications for Renewal of Registration ...................................................................................... 87
7.3 Application for Variation of a registered medicinal product ....................................................... 87
8. PAYMENT OF FEES .............................................................................................................. 87
9. AN OUTLINE OF THE EVALUATION PROCESS ................................................................. 88
9.1 Receiving of new applications ........................................................................................................ 88
vi
FDA/DRI/DER/GL-RAD/2013/02
vii
FDA/DRI/DER/GL-RAD/2013/06
1.0. INTRODUCTION
This guideline applies only to allopathic drugs. This guideline prescribes the minimum
information required for submission of documentation as well as the appropriate format
and organisation of the requisite data.
In this new format, each application is organised into modules, together with the
associated technical guidelines. Applicants should not modify the overall organisation of
this format.
Applicants are requested to carefully read this guideline, fill in application form, prepare
dossier and submit two electronic copies (in a Portable Document Format (PDF), on a
(CD-Rom) and should include MS-Word document for Module 2.3, cross-referenced
to the dossier by clearly indicating the title and section number of all the supporting
documents.
All sections and fields in the five (5) modules that would be applicable should be
completed.
It is understood that certain sections and fields may not apply and should be
indicated as such by reporting “not applicable” in the appropriate area with an
accompanying explanatory note.
To ensure easy access to Drugs, the FDA has developed and implemented alternative
Page 1 of 88
FDA/DRI/DER/GL-RAD/2013/06
1.2. OBJECTIVE
This revised guideline presents a common format for the preparation of an application
that will be submitted to the Food and Drugs Authority (FDA).
This revised guideline has been improved to assist in the following;
• Preparation of documentation for pharmaceutical products by providing clear
guidance on the format.
• Fully adopt the modular format of the Common Technical Document (CTD) as
developed by International Conference on Harmonization (ICH) as well as World
Health Organization (WHO) Guidelines on Submission of Documentation for
Prequalification of Multi-source Finished Pharmaceutical Products
• Provide guidance on the technical and other general data requirements.
• Reduce the time lines to compile applications for registration of medicines
• Give more details on the requirements for active pharmaceutical ingredients (API)
as well as finished pharmaceutical product (FPP).
Page 2 of 88
FDA/DRI/DER/GL-RAD/2013/06
1.3. SCOPE
This revised guideline is developed in pursuance of Section 118 of the Public Health Act,
2012, Act 851, these guidelines are hereby made to provide guidance to applicants on
the organization of information to be presented in registration applications for allopathic
drugs. Applicants are encouraged to familiarize themselves with this document and the
above law before completing the application form for registration of allopathic drugs.
2.0. GLOSSARY
In the context of this guideline, the following words/phrases are defined as follows.
Active Pharmaceutical Ingredient (API): A substance or compound that is intended to
be used in the manufacture of a pharmaceutical product as a therapeutically active
compound (ingredient).
API starting material: A raw material, intermediate, or an API that is used in the
production of an API and that is incorporated as a significant structural fragment into the
structure of the API. An API starting material can be an article of commerce, a material
purchased from one or more suppliers under contract or commercial agreement, or
produced in-house.
Allopathic drug: Any product or substance other than a medical device, which is to be
administered to one or more human beings or animals on its own, or as an ingredient in
the preparation of a substance, for a medicinal purpose.
Medicinal purpose: means treating or preventing a disease, diagnosing or ascertaining
the presence and extent of a physiological function, contraception, inducing anaesthesia,
altering normal physiologic function permanently or temporarily in any way in humans.
Applicant: The product owner or licence holder. Representatives of licence holders may
not hold themselves as applicants unless they own the product.
BCS highly soluble: An API for which the highest dose recommended or highest dose
strength available on the market as an oral solid dosage form is soluble in 250 ml or less
of aqueous media over the pH range of 1.2–6.8 at 37ºC
Page 3 of 88
FDA/DRI/DER/GL-RAD/2013/06
Page 4 of 88
FDA/DRI/DER/GL-RAD/2013/06
therapeutically equivalent and can therefore be used interchangeably with the innovator
or reference product. It is a pharmaceutical product usually intended to be
interchangeable with the innovator product, which is usually manufactured without a
license from the innovator company.
Innovator pharmaceutical product: Means a pharmaceutical product, which was first
registered (normally as a patented product) on the basis of documentation of efficacy,
safety and quality (according to the requirements at the time of registration).
Interchangeability: An interchangeable pharmaceutical product is one that is
therapeutically equivalent to an innovator (reference) product.
Label: Means any tag, brand, mark, pictorial or other descriptive matter, written, printed,
stenciled, marked, embossed or impressed on or attached to a container of any drug.
Manufacture (manufacturing): Means all operations of purchase of materials and
products, production, quality control, release, storage, shipment of finished products and
the related controls.
Manufacturer: Means a person or firm that is engaged in the manufacture of product(s).
New chemical entity: A chemical or biologically Active Pharmaceutical Ingredient (API)
that has not previously been registered as an ingredient of any pharmaceutical product.
New drug: means a generic copy of an innovator product:
i. That has not been previously registered as a pharmaceutical or biological product
in Ghana, or
ii. Which has been marketed in Ghana for a period of not less than ten (10) years or
any other period to be determined by the Authority from time to time, for public
health reasons
Pharmaceutical alternatives: Two or more medicinal products are said to be
pharmaceutical alternatives if they contain the same active ingredients, but which may
differ in salt, esters, dosage forms, strength and/ or route of administration.
Pharmaceutical equivalents: Products are pharmaceutical equivalents if they contain
the same amount of the same active substance(s) in the same dosage form; if they meet
the same or comparable standard; and if they are intended to be administered by the
same route.
Pilot-scale batch: A batch of an API or FPP manufactured by a procedure fully
representative of and simulating that to be applied to a full production-scale batch. For
example, for solid oral dosage forms, a pilot scale is generally, at a minimum, one-tenth
that of a full production scale or 100 000 tablets or capsules, whichever is the larger;
unless otherwise adequately justified
Page 5 of 88
FDA/DRI/DER/GL-RAD/2013/06
Primary batch: A batch of an API or FPP used in a stability study, from which stability
data are submitted in a registration application for the purpose of establishing a re-test
period or shelf-life.
Release specifications: Means the combination of physical, chemical, biological and
microbiological test requirements that determine whether a drug product is suitable for
release at the time of its manufacture.
Starting material: Means any substance of a defined quality used in the production of a
pharmaceutical product, but excluding packaging materials.
Shelf life specifications: Means the combination of physical, chemical, biological and
microbiological test requirements that an active ingredient must meet up to its retest date
or a drug product must meet during its shelf life.
Therapeutic equivalence: Two pharmaceutical products are therapeutically equivalent if
they are pharmaceutically equivalent and, after administration in the same molar dose,
their effects with respect to both efficacy and safety essentially the same, as determined
from appropriate bioequivalence, pharmacodynamic, clinical or in vitro studies.
Variation: Means a change to any aspect of a pharmaceutical product, including but not
limited to a change to formulation, method and site of manufacture, specifications for the
finished product and ingredients, container and container labelling and product
information.
Trade/Proprietary name means the (trade or brand) name which is unique to a particular
drug and by which it is generally identified. Refer to FDA Guidelines for labelling.
Page 6 of 88
FDA/DRI/DER/GL-RAD/2013/06
Dosage form of the product shall mean the form in which the drug is presented, e.g. eye
drops, emulsion, ointment, suppository, tablet, capsule, solution, suspension, injections.
Strength of the product shall be given per unit dosage form or per specified quantity: e.g.
mg per tablet, mg per capsule, mg per ml, mg per 5ml, mg per gram, etc.
Commercial presentation of the product shall mean the presentation of the product to be
registered i.e. list all pack sizes intended for marketing. eg 10 x 1tablets, 10 x 10tablets,
10 x 1 capsules, 200mls, 1ml vial etc.
The container/closure description should include all parts of the primary packaging
including desiccant, void filler or adsorbent cotton filler. Dimensions/volume/capacity may
be listed. Shape and colour of the bottle and the cap type (including plastic e.g. PP),
should be stated. E.g.: Blisters: colour and transparent/opaque, with number of units per
card and cards per box.]
[E.g. sealed LDPE bag, placed inside a round white HDPE bottle with plain PP screw cap
and aluminium tagger (packs of 100 Tablets & 1000 Tablets]
Description of the drug shall mean a full visual appearance of the drug as marketed
including colour, size, shape and other relevant features [e.g.: White coloured, biconvex
capsule-shaped film-coated tablet having a score on one side and “XL 5” debossed on
the other side.]
Page 7 of 88
FDA/DRI/DER/GL-RAD/2013/06
Specify clinical indication(s) which are supported by relevant information in Module 2 and
5 of the application dossiers.
1.2.12 Applicant
The name, physical address, telephone number, fax number, and e-mail address of the
applicant/license holder.
Where different activities of manufacture of a given product are carried out at different
manufacturing sites, the information on the following should be provided.
A copy of a valid manufacturing License shall be provided for each site. Only products
entirely manufactured at sites that are cGMP compliant by FDA shall be eligible for
registration.
Page 8 of 88
FDA/DRI/DER/GL-RAD/2013/06
Also attach a valid Certificate of Pharmaceutical Product from the country of origin as per
the WHO certification scheme and issued in the name of Ghana or with Ghana included
in the list of importing countries.
In the case of loan license manufacturing, the following should also be submitted;
Every applicant who is not resident in Ghana shall appoint one local representative who
must be a company incorporated in Ghana and authorized by FDA to import medicinal
products and must hold a wholesale dealers License.
1.2.17 Declaration
The applicant/license holder should indicate that the information submitted is true and
correct. Information on the name, position and signature of the applicant should be
provided. The application should be dated and stamped by the applicant.
Page 9 of 88
FDA/DRI/DER/GL-RAD/2013/06
1.3.1 Product information for Health Professionals (For All Products subject to Medical
Prescription)
The SmPC is an essential part of registration and cannot be altered without the prior
approval of FDA through a post approval variation.
Provide four (4) copies of the patient information leaflet (find information on PIL from
https://fdaghana.gov.gh/images/stories/pdfs/downloads/drugs%20guidelines/TEMPLATE%20PATIENT%20INFORM
ATION%20LEAFLET.pdf ) and any information intended for distribution with the product to the
patient. The patient information leaflet (PIL) should be in conformity with the SmPC. It
should be written in English, should be legible, indelible and comprehensible.
The PIL is an essential part of registration and cannot be altered without the prior approval
of FDA through a post approval variation.
Provide four (4) copies of the proposed outer and inner labels. It should be written in
English, should be legible, indelible and comprehensible.
The labelling is an essential part of registration and cannot be altered without the prior
approval of FDA through a post approval variation (find information on labelling template
from https://fdaghana.gov.gh/images/stories/pdfs/downloads/drugs%20guidelines/TEMPLATE%20LABELLING.pdf ).
1.4 Samples
Samples of the product and certificate of analysis of the FPP (s) and measuring devices
as per the FDA sample schedule (find information on sample schedule from
https://fdaghana.gov.gh/wp-content/uploads/2017/06/SAMPLE-SCHEDULE-FOR-REGISTRATION-AND-RE.pdf)
should be
provided for laboratory analysis and also to enable visual inspection of the product and
product package.
Page 10 of 88
FDA/DRI/DER/GL-RAD/2013/06
All products submitted for registration shall have at least 60% of its shelf life remaining.
This notwithstanding, products with a shelf life of less than 24 months shall have at least
80% of its shelf life remaining at the time of submission.
MODULE 2: CTD Summaries- Chemical, Pharmaceutical, Non-Clinical and Clinical
The Quality Overall Summary (QOS) is a summary that follows the scope and the outline
of Module 3. The QOS should not include detailed information, data or justification that
will be included in Module 3 or in other modules of the document. The QOS should include
sufficient information from each section to provide the Quality Evaluator with an overview
of Module 3. The QOS should also emphasise critical key parameters of the product and
provide, for instance, justification in cases where guideline were not followed. The QOS
should include a discussion of key issues that integrates information from sections in the
Quality Module and supporting information from other Modules, including cross-
referencing to volume and page number in other Modules.
The QOS normally should not exceed 40 pages of text, excluding tables and figures.
(excluding tables and figures).
Page 11 of 88
FDA/DRI/DER/GL-RAD/2013/06
For NCE:
For NCE:
The QOS should summarise the data on potential and actual impurities arising from the
synthesis, manufacture and/or degradation, and should summarise the basis for setting
the acceptance criteria for individual and total impurities. The QOS should also
summarise the impurity levels in batches of the drug substance used in the non-clinical
studies, in the clinical trials, and in typical batches manufactured by the proposed
commercial process. The QOS should state how the proposed impurity limits are
qualified.
Page 12 of 88
FDA/DRI/DER/GL-RAD/2013/06
A brief description and discussion of the information, from 3.2.S.6 should be included.
This section should include a summary of the studies undertaken (conditions, batches,
analytical procedures) and a brief discussion of the results and conclusions, the proposed
storage conditions, retest date or shelf-life, where relevant, as described in 3.2.S.7.1.
A tabulated summary of the stability results from 3.2.S.7.3, with graphical representation
where appropriate, should be provided.
A tabulated summary of the batch analyses provided under 3.2.P.5.4, with graphical
representation where appropriate should be included.
A tabulated summary of the stability results from 3.2.P.8.3, with graphical representation
where appropriate, should be included.
2.3. A APPENDICES
Page 14 of 88
FDA/DRI/DER/GL-RAD/2013/06
The primary purpose of the Nonclinical Written and Tabulated Summaries should be to
provide a comprehensive factual synopsis of the nonclinical data. The interpretation of
the data, the clinical relevance of the findings, cross-linking with the quality aspects of the
pharmaceutical product, and the implications of the nonclinical findings for the safe use
of the pharmaceutical product (i.e., as applicable to labelling) should be addressed in the
Overview.
The Nonclinical Overview should provide an integrated overall analysis of the information
in the Common Technical Document. In general, the Nonclinical Overview should not
exceed 30 pages.
General Aspects
The Nonclinical Overview should present an integrated and critical assessment of the
pharmacologic, pharmacokinetic, and toxicologic evaluation of the pharmaceutical
product. Where relevant guidelines on the conduct of studies exist, these should be taken
into consideration, and any deviation from this guideline should be discussed and justified.
The nonclinical testing strategy should be discussed and justified. There should be
comment on the GLP status of the studies submitted. Any association between nonclinical
findings and the quality characteristics of the human pharmaceutical, the results of clinical
trials, or effects seen with related products should be indicated, as appropriate.
An assessment of the impurities and degradants present in the drug substance and
product should be included along with what is known of their potential pharmacologic and
toxicologic effects.
This assessment should form part of the justification for proposed impurity limits in the
drug substance and product, and be appropriately cross-referenced to the quality
documentation. The implications of any differences in the chirality, chemical form, and
impurity profile between the compound used in the nonclinical studies and the product to
be marketed should be discussed.
Page 15 of 88
FDA/DRI/DER/GL-RAD/2013/06
Relevant scientific literature and the properties of related products should be taken into
account. If detailed references to published scientific literature are to be used in place of
studies conducted by the applicant, this should be supported by an appropriate
justification that reviews the design of the studies and any deviations from available
guideline. In addition, the availability of information on the quality of batches of drug
substance used in these referenced studies should be discussed.
The Nonclinical Overview should contain appropriate reference citations to the Tabulated
Summaries.
The onset, severity, and duration of the toxic effects, their dose-dependency and degree
of reversibility (or irreversibility), and species- or gender-related differences should be
evaluated and important features discussed, particularly with regard to:
Page 16 of 88
FDA/DRI/DER/GL-RAD/2013/06
Pharmacodynamics
toxic signs causes of death pathologic findings genotoxic activity - the chemical
structure of the compound, its mode of action, and its relationship to known genotoxic
compounds
carcinogenic potential in the context of the chemical structure of the compound, its
relationship to known carcinogens, its genotoxic potential, and the exposure data the
carcinogenic risk to humans - if epidemiologic data are available, they should be taken
into account
fertility, embryofetal development, pre-and post-natal toxicity studies in juvenile
animals the consequences of use before and during pregnancy, during lactation, and
during
paediatric development
local tolerance
other toxicity studies/ studies to clarify special problems
The evaluation of toxicology studies should be arranged in a logical order so that all
relevant data elucidating a certain effect / phenomenon are brought together.
Extrapolation of the data from animals to humans should be considered in relation to:
animal species used numbers of animals used routes of administration employed
dosages used
duration of treatment or of the study
systemic exposures in the toxicology species at no observed adverse effect levels and
at toxic doses, in relation to the exposures in humans at the maximum recommended
human dose. Tables or figures summarising this information are recommended.
the effect of the drug substance observed in nonclinical studies in relation to that
expected or observed in humans
The Integrated Overview and Conclusions should clearly define the characteristics of the
human pharmaceutical as demonstrated by the nonclinical studies and arrive at logical,
well-argued conclusions supporting the safety of the product for the intended clinical use.
Taking the pharmacology, pharmacokinetics, and toxicology results into account, the
implications of the nonclinical findings for the safe human use of the pharmaceutical
should be discussed (i.e., as applicable to labelling).
Page 17 of 88
FDA/DRI/DER/GL-RAD/2013/06
The Clinical Overview is intended to provide a critical analysis of the clinical data in the
Common Technical Document. The Clinical Overview will necessarily refer to application
data provided in the comprehensive Clinical Summary, the individual clinical study reports
(ICH E3), and other relevant reports; but it should primarily present the conclusions and
implications of those data, and should not recapitulate them. Specifically, the Clinical
Summary should provide a detailed factual summarisation of the clinical information in the
CTD, and the Clinical Overview should provide a succinct discussion and interpretation
of these findings together with any other relevant information (e.g., pertinent animal data
or product quality issues that may have clinical implications).
The Clinical Overview is primarily intended for use by regulatory agencies in the review
of the clinical section of a marketing application. It should also be a useful reference to
the overall clinical findings for regulatory agency staff involved in the review of other
sections of the marketing application. The Clinical Overview should present the strengths
and limitations of the development program and study results, analyse the benefits and
risks of the medicinal product in its intended use, and describe how the study results
support critical parts of the prescribing information.
The Clinical Overview should generally be a relatively short document (about 30 pages).
The length, however, will depend on the complexity of the application. The use of graphs
and concise tables in the body of the text is encouraged for brevity and to facilitate
understanding. It is not intended that material presented fully elsewhere be repeated in
Page 18 of 88
FDA/DRI/DER/GL-RAD/2013/06
Table of Contents
Introduction
The sequence and content of the Nonclinical Written Summary sections are described
below. It should be emphasised that no guideline can cover all eventualities, and common
sense and a clear focus on the needs of the regulatory authority assessor are the best
guides to constructing an acceptable document. Therefore, applicants can modify the
format if needed to provide the best possible presentation of the information, in order to
facilitate the understanding and evaluation of the results.
Page 19 of 88
FDA/DRI/DER/GL-RAD/2013/06
To allow authors flexibility in defining the optimal structure for the Written Summaries,
tables and figures should preferably be included within the text. Alternatively, they could
be grouped together at the end of each of the Nonclinical Written Summaries. Throughout
Page 20 of 88
FDA/DRI/DER/GL-RAD/2013/06
the text, reference citations to the Tabulated Summaries should be included, in the
following format: (Table X.X, Study/Report Number).
Although there is no formal limit to the length of the Nonclinical Written Summaries, it is
recommended that the total length of the three Nonclinical Written Summaries in general
not exceed 100-150 pages.
• Introduction
• Written Summary of Pharmacology
• Tabulated Summary of Pharmacology
• Written Summary of Pharmacokinetics
• Tabulated Summary of Pharmacokinetcs
• Written Summary of Toxicology
• Tabulated Summary of Toxicology
Refer ICH M4S_R2 for detailed discussion of content of the non- clinical written and
tabulated summaries.
The comparisons and analyses of results across studies provided in this document should
focus on factual observations. In contrast, the CTD Clinical Overview document should
provide critical analysis of the clinical study program and its results, including discussion
and interpretation of the clinical findings and discussion of the place of the test drug in the
armamentarium.
The length of the Clinical Summary will vary substantially according to the information to
be conveyed, but it is anticipated that (excluding attached tables) the Clinical Summary
will usually be in the range of 50 to 400 pages.
Page 21 of 88
FDA/DRI/DER/GL-RAD/2013/06
Table of Contents
Refer ICH M4E_R1 for detailed discussion of content of the clinical summary.
Page 22 of 88
FDA/DRI/DER/GL-RAD/2013/06
This module is intended to provide guidance on the format of a registration application for
drug substances and their corresponding drug products.
The information on the API can be submitted according to the following order of
preference:
• Provide the latest, valid European Certificate of Suitability (CEP) with all annexes.
• Provide a Drug Master File(s) [DMF(s)] submitted by the API manufacturer.
In addition, a written commitment should be included that the applicant will inform FDA in
the event that the CEP is withdrawn. It should also be acknowledged by the applicant that
withdrawal of the CEP will require additional consideration of the API data requirements
to support the dossier. The written commitment should accompany the copy of the CEP
in Module 1.
Along with the CEP the applicant should supply the following information in the dossier,
with data summarized in the QOS
Page 23 of 88
FDA/DRI/DER/GL-RAD/2013/06
• 3.2. S.4.2/3.2.S.4.3 Analytical procedures and validation – for any tests in addition
to those in the CEP and Ph. Eur monograph.
• 3.2. S.4.4 Batch analysis – results from two batches of at least pilot scale,
demonstrating compliance with the FPP manufacturer’s API specifications.
• 3.2. S.5 Reference standards or materials – information on the FPP manufacturer’s
reference standards.
• 3.2. S.6 Container-closure system – specifications including descriptions and
identification of primary packaging components.
• 3.2. S.7 Stability – exception: where the CEP specifies a re-test period that is the
same as or of longer duration than the re-test period proposed by the applicant.
Provide a Drug Master File(s) [DMF(s)] submitted by the API manufacturer. The DMF
should contain all the information listed under Section 3.2.S
For use of the DMF option, the API manufacturer should provide a signed declaration that
the synthesis and subsequent purification is conducted in accordance with what is
presented in the dossier.
For a drug product containing more than one drug substance, the information requested
for both options ―3.2.1 above should be provided in its entirety for each drug substance.
Information on the nomenclature of the drug substance should be provided. For example:
• Recommended International Nonproprietary Name (INN);
• Compendial name if relevant;
• Chemical name(s);
• Company or laboratory code;
• Other non-proprietary name(s), e.g., national name, United States Adopted Name
(USAN), Japanese Accepted Name (JAN); British Approved Name (BAN), and
• Chemical Abstracts Service (CAS) registry number.
Page 24 of 88
FDA/DRI/DER/GL-RAD/2013/06
The listed chemical names should be consistent with those appearing in scientific
literature and those appearing on the product labelling information (e.g. summary of
product characteristics, package leaflet (also known as patient information leaflet or PIL),
labelling).
This information should be consistent with that provided in section 3.2.S.1.1. For APIs
existing as salts, the molecular mass of the free base or acid should also be provided.
This information can be used in developing the specifications, in formulating FPPs and in
the testing for release and stability purposes.
The physical and chemical properties of the API should be discussed including the
physical description, solubilities in common solvents (e.g. water, alcohols,
dichloromethane, acetone), quantitative aqueous pH solubility profile (e.g. pH 1.2 to 6.8,
dose/solubility volume), polymorphism, pH and pKa values, UV absorption maxima and
molar absorptivity, melting point, refractive index (for a liquid), hygroscopicity, partition
coefficient, etc. This list is not intended to be exhaustive, but provides an indication as to
the type of information that could be included.
Some of the more relevant properties to be considered for APIs are discussed below in
greater detail.
Physical description
The description should include appearance, colour and physical state. Solid forms should
be identified as being crystalline or amorphous (see 3.2.S.3.1 for further information on
API solid forms).
Page 25 of 88
FDA/DRI/DER/GL-RAD/2013/06
The solubilities over the physiological pH range (pH 1.2 to 6.8) in several buffered media
should be provided in mg/ml. If this information is not readily available (e.g. literature
references), it should be generated in-house.
For solid oral dosage forms, the dose/solubility volume should be provided as determined
by:
* corresponding to the lowest solubility determined over the physiological pH range (pH
1.2 to 6.8) and temperature (37 ± 0.5°C).
As per the Biopharmaceutics Classification System (BCS), highly soluble (or highly water
soluble) APIs are those with a dose/solubility volume of less than or equal to 250 ml.
For example, compound A has as its lowest solubility at 37 ± 0.5°C, 1.0 mg/ml at pH 6.8
and is available in 100 mg, 200 mg and 400 mg strengths. This API would not be
considered a BCS highly soluble API as its dose/solubility volume is greater than 250 ml
(400 mg/1.0 mg/ml = 400 ml).
Polymorphism
As recommended in ICH‟s CTD-Q Questions and answers/location issues document the
following refers to where specific data should be located in the product dossier:
• The polymorphic form(s) present in the proposed API should be listed in Section
3.2. S.1.3
• If a polymorphic form is to be defined or limited (e.g. for APIs that are not BCS
highly soluble and/or where polymorphism has been identified as an issue), details
should be included in 3.2.S.4.1 through 3.2.S.4.5.
Page 26 of 88
FDA/DRI/DER/GL-RAD/2013/06
The name, address, and responsibility of each manufacturer, including contractors, and
each proposed production site or facility involved in manufacturing and testing should be
provided.
The facilities involved in the manufacturing, packaging, labelling, testing and storage of
the API should be listed. If certain companies are responsible only for specific steps (e.g.
milling of the API), this should be clearly indicated.
A valid manufacturing licence should be provided for the production of APIs. If available,
a certificate of GMP compliance should be provided in the dossier in Module 1.
Page 27 of 88
FDA/DRI/DER/GL-RAD/2013/06
Alternate processes should be explained and described with the same level of detail as
the primary process. Reprocessing steps should be identified and justified. Any data to
support this justification should be either referenced or filed in 3.2.S.2.5.
As discussed in ICH Q7 and WHO Technical Report Series, No. 957 Annex 2, the point
at which the API starting material is introduced into the manufacturing process is the
starting point of the application of GMP requirements. The API starting material itself
needs to be proposed and its choice justified by the manufacturer and accepted as such
by assessors. The API starting material should be proposed taking into account the
complexity of the molecule, the proximity of the API starting material to the final API, the
availability of the API starting material as a commercial chemical and the quality controls
placed upon the API starting material.
In situations where the API starting material is a complex molecule and only a minimal
number of synthetic steps from the final API, a further molecule called the starting material
for synthesis should be proposed and its choice justified by the applicant. The starting
material for synthesis defines the starting point in the manufacturing process for an API
to be described in an application. The applicant should propose and justify which
substances should be considered as starting materials for synthesis. See section
3.2.S.2.3 for further guidance. In the case where the precursor to the API is obtained from
fermentation, or is from plant or animal origin, such a molecule can be considered the API
starting material regardless of complexity.
Where there are multiple manufacturing sites for one API manufacturer, a comprehensive
list in tabular form should be provided comparing the processes at each site and
highlighting any differences.
Copies of the specifications for the materials used in the synthesis, extraction, isolation
and purification steps should be provided in the dossier, including starting materials,
Page 28 of 88
FDA/DRI/DER/GL-RAD/2013/06
The carry-over of impurities of the starting materials for synthesis into the final API should
be considered and discussed.
A letter of attestation should be provided confirming that the API and the starting materials
and reagents used to manufacture the API are without risk of transmitting agents of animal
spongiform encephalopathies.
Intermediates: Information on the quality and control of intermediates isolated during the
process should be provided.
The critical steps should be identified. These can be among others: steps where
significant impurities are removed or introduced, steps introducing an essential molecular
structural element such as a chiral centre or resulting in a major chemical transformation,
steps having an impact on solid-state properties and homogeneity of the API that may be
relevant for use in solid dosage forms.
Specifications for isolated intermediates should be provided and should include tests and
acceptance criteria for identity, purity and assay, where applicable.
A description and discussion should be provided of the significant changes made to the
manufacturing process and/or manufacturing site of the drug substance used in producing
nonclinical, clinical, scale-up, pilot, and, if available, production scale batches.
Reference should be made to the drug substance data provided in section 3.2.S.4.4.
Reference ICH Guideline: Q3A
Elucidation of structure
The dossier should include quality assurance (QA) certified and legible copies of the
spectra, peak assignments and a detailed interpretation of the data of the studies
performed to elucidate and/or confirm the structure of the API. The dossier should include
a list of the studies performed and a conclusion from the studies (e.g. if the results support
the proposed structure).
For APIs that are not described in an officially recognized pharmacopoeia, the studies
carried out to elucidate and/or confirm the chemical structure normally include elemental
analysis, infrared (IR), ultraviolet (UV), nuclear magnetic resonance (NMR) and mass
spectra (MS) studies. Other tests could include X-ray powder diffraction (XRPD) and
differential scanning calorimetry (DSC).
Isomerism/Stereochemistry
Page 30 of 88
FDA/DRI/DER/GL-RAD/2013/06
Where the potential for stereoisomerism exists, a discussion should be included of the
possible isomers that can result from the manufacturing process and the steps where
chirality was introduced. The identicality of the isomeric composition of the API to that of
the API in the comparator product should be established. Information on the physical and
chemical properties of the isomeric mixture or single enantiomer should be provided, as
appropriate.
The API specification should include a test to ensure isomeric identity and purity. The
potential for interconversion of the isomers in the isomeric mixture, or racemisation of the
single enantiomer should be discussed.
If, based on the structure of the API, there is not a potential for stereoisomerism, it is
sufficient to include a statement to this effect.
Polymorphism
Many APIs can exist in different physical forms in the solid state. Polymorphism is
characterized as the ability of an API to exist as two or more crystalline phases that have
different arrangements and/or conformations of the molecules in the crystal lattice.
Polymorphic forms of the same chemical compound differ in internal solid-state structure
and, therefore, may possess different chemical and physical properties, including
packing, thermodynamic, spectroscopic, kinetic, interfacial and mechanical properties.
These properties can have a direct impact on API processability, pharmaceutical product
manufacturability and product quality/performance, including stability, dissolution and
bioavailability. Unexpected appearance or disappearance of a polymorphic form may lead
to serious pharmaceutical consequences.
Applicants are expected to have adequate knowledge about the polymorphism of the APIs
used and/or produced. Information on polymorphism can come from the scientific
literature, patents, compendia or other references to determine if polymorphism is a
concern, e.g. for APIs that are not BCS highly soluble. In the absence of published data
Page 31 of 88
FDA/DRI/DER/GL-RAD/2013/06
for APIs that are not BSC highly soluble, polymorphic screening will be necessary to
determine if the API can exist in more than one crystalline form. Polymorphic screening
is generally accomplished via crystallization studies using different solvents and
conditions.
There are a number of methods that can be used to characterize the polymorphic forms
of an API. Demonstration of a nonequivalent structure by single crystal X-ray diffraction
is currently regarded as the definitive evidence of polymorphism. XRPD can also be used
to provide unequivocal proof of polymorphism. Other methods, including microscopy,
thermal analysis (e.g. DSC, thermal gravimetric analysis and hot-stage microscopy) and
spectroscopy (e.g. IR, Raman, solid-state nuclear magnetic resonance [ssNMR]) are
helpful to further characterize polymorphic forms. Where polymorphism is a concern, the
applicants/manufacturers of APIs should demonstrate that a suitable method, capable of
distinguishing different polymorphs, is available to them.
For APIs that are not BCS highly soluble contained in solid FPPs, or liquid FPPs
containing undissolved API, the particle size distribution of the material can have an effect
on the in vitro and/or in vivo behaviour of the FPP. Particle size distribution can also be
important in dosage form performance (e.g. delivery of inhalation products), achieving
uniformity of content in low-dose tablets (e.g. 2 mg or less), desired smoothness in
ophthalmic preparations and stability of suspensions.
If particle size distribution is an important parameter (e.g. as in the above cases), results
from an investigation of several batches of the API should be provided, including
characterization of the batch(es) used in the comparative bioavailability or biowaiver
studies. API specifications should include controls on the particle size distribution to
ensure consistency with the material in the batch(es) used in the comparative
bioavailability and biowaiver studies (e.g. limits for d10, d50 and d90). The criteria should
be established statistically based on the standard deviation of the test results from the
previously mentioned studies. The following is provided for illustrative purposes as
possible acceptance criteria for particle size distribution limits:
Page 32 of 88
FDA/DRI/DER/GL-RAD/2013/06
• d10 not more than (NMT) 10% of total volume less than X µm • d50 XX µm - XXX
µm
• d90 not less than (NLT) 90% of total volume less than XXXX µm.
Details on the principles for the control of impurities (e.g. reporting, identification and
qualification) are outlined in the ICH Q3A, Q3B and Q3C impurity guidelines. Additional
information to provide further guidance on some of the elements discussed in the ICH
guidelines is outlined below.
The ICH thresholds for reporting, identification (used to set the limit for individual unknown
impurities) and qualification are determined on the basis of potential exposure to the
impurity, e.g. by the maximum daily dose (MDD) of the API. For APIs available in multiple
dosage forms and strengths having different MDD values, it is imperative that the
thresholds and corresponding controls for each of the presentations be considered to
ensure that the risks posed by impurities have been addressed. This is normally achieved
by using the highest potential daily MDD, rather than the maintenance dose. For
parenteral products, the maximum hourly dose of the API should also be included.
It is acknowledged that APIs of semi-synthetic origin do not fall within the scope of the
ICH impurity guidelines. However, depending on the nature of the API and the extent of
the chemical modification steps, the principles on the control of impurities (e.g. reporting,
identification and qualification) could also be extended to APIs of semisynthetic origin. As
an illustrative example, an API whose precursor molecule was derived from a
fermentation process, or a natural product of plant or animal origin that has subsequently
undergone several chemical modification reactions generally would fall within this scope,
Page 33 of 88
FDA/DRI/DER/GL-RAD/2013/06
whereas an API whose sole chemical step was the formation of a salt from a fermentation
product generally would not fall within this scope. It is understood that there is some
latitude for these types of APIs.
Identification of impurities
It is recognized by the pharmacopoeias that APIs can be obtained from various sources
and thus can contain impurities not considered during the development of the monograph.
Furthermore, a change in the production or source may give rise to additional impurities
that are not adequately controlled by the official compendial monograph. As a result, each
product dossier is assessed independently to consider the potential impurities that may
arise from the proposed route(s) of synthesis.
For these reasons, the ICH limits for unspecified impurities (e.g. NMT
0.10% or 1.0 mg per day intake (whichever is lower) for APIs having a maximum daily
dose =2 g/day) are generally recommended, rather than the general limits for unspecified
impurities that may appear in the official compendial monograph that could potentially be
higher than the applicable ICH limit.
Qualification of impurities
The ICH impurity guidelines should be consulted for options on the qualification of
impurities. The limit specified for an identified impurity in an officially recognized
pharmacopoeia is generally considered to be qualified. The following is an additional
option for qualification of impurities in existing APIs:
The limit for an impurity present in an existing API can be accepted by comparing the
impurity results found in the existing API with those observed in an innovator product
using the same validated, stability-indicating analytical procedure (e.g. comparative HPLC
studies). If samples of the innovator product are not available, the impurity profile may
also be compared to a different prequalified FPP with the same route of administration
and similar characteristics (e.g. tablet versus capsule). It isrecommended that the studies
be conducted on comparable samples (e.g. age of samples) to obtain a meaningful
comparison of the impurity profiles.
A specified impurity present in the existing API is considered qualified if the amount of the
impurity in the existing API reflects the levels observed in the innovator or prequalified
FPP.
The basis for setting the acceptance criteria for the impurities should be provided. This is
established by considering the identification and qualification thresholds for APIrelated
impurities (e.g. starting materials, by-products, intermediates, chiral impurities or
degradation products) and the concentration limits for process-related impurities (e.g.
residual solvents) as per the applicable ICH guidelines (e.g. Q3A, Q3C).
The qualified level should be considered as the maximum allowable limit. However, limits
which are considerably wider than the actual manufacturing process capability are
generally discouraged. For this reason, the acceptance criteria are also set taking into
consideration the actual levels of impurities found in several batches of the API from each
manufacturer, including the levels found in the batches used for the comparative
bioavailability or biowaiver studies. When reporting the results of quantitative tests, the
actual numerical results should be provided rather than vague statements such as “within
limits” or “conforms”. In the cases where a large number of batches have been tested it is
acceptable to summarize the results of the total number of batches tested with a range of
analytical results.
If there are identified impurities specified in an official compendial monograph that are not
controlled by the proposed routine in-house analytical procedure, a justification for their
exclusion from routine analyses should be provided (e.g. “Impurities D, E and F listed in
the Ph.Int. monograph are not potential impurities from the proposed route of synthesis
used by manufacturer X”). If acceptable justification cannot be provided it should be
demonstrated that the routine in-house method is capable of separating and detecting the
impurities specified in the official compendial monograph at an acceptable level (e.g.
0.10%). If such a demonstration cannot be performed, a one-time study should be
conducted applying the pharmacopoeial method to several recent batches to demonstrate
the absence of the pharmacopoeial listed impurities.
ICH class II solvent(s) used prior to the last step of the manufacturing process may be
exempted from routine control in API specifications if suitable justification is provided.
Submission of results demonstrating less than 10% of the ICH Q3C limit (option I) of the
solvent(s) in three consecutive production-scale batches or six consecutive pilot-scale
batches of the API or a suitable intermediate would be considered acceptable justification.
The last step solvents used in the process should always be routinely controlled in the
final API.
For guidance on acceptable residual solvent limits, refer to ICH Q3C. The limit for residues
of triethylamine (TEA) is either 320 ppm on the basis of ICH Q3C option I or 3.2 mg/day
on the basis of permitted daily exposure (PDE).
The absence of known established highly toxic impurities (genotoxic) used in the process
or formed as a by-product should be discussed and suitable limits should be proposed.
The limits should be justified by appropriate reference to available guidances (e.g.
Page 35 of 88
FDA/DRI/DER/GL-RAD/2013/06
Copies of the API specifications, dated and signed by authorized personnel (e.g. the
person in charge of the quality control or quality assurance department) should be
provided in the dossier including specifications from each API manufacturer as well as
those of the FPP manufacturer.
Page 36 of 88
FDA/DRI/DER/GL-RAD/2013/06
The FPP manufacturer(s) API specification could be summarized in a tabular form under
the headings tests, acceptance criteria and analytical procedures (including types,
sources and versions for the methods).
In cases where there is more than one API manufacturer, the FPP manufacturer’s API
specifications should be one single compiled set of specifications that is identical for each
manufacturer. It is acceptable to lay down in the specification more than one acceptance
criterion and/or analytical method for a single parameter with the statement “for API from
manufacturer A” (e.g. in the case of residual solvents).
Any non-routine testing should be clearly identified as such and justified along with the
proposal on the frequency of non-routine testing.
The ICH Q6A guideline outlines recommendations for a number of universal and specific
tests and criteria for APIs.
The analytical procedures used for testing the API should be provided.
Copies of the in-house analytical procedures used to generate testing results provided in
the dossier, as well as those proposed for routine testing of the API by the FPP
manufacturer, should be provided.
Although HPLC is normally considered the method of choice for determining API-related
impurities, other chromatographic methods such as GC and TLC can also be used, if
appropriately validated. For determination of related substances, reference standards
should normally be available for each of the identified impurities, particularly those known
to be toxic and the concentration of the impurities should be quantitated against their own
reference standards.
Page 37 of 88
FDA/DRI/DER/GL-RAD/2013/06
In cases where the response factor is outside this range, it may still be acceptable to use
the API, provided a correction factor is applied. Data to support calculation of the
correction factor should be provided for an in-house method. Unspecified impurities may
be quantitated using a solution of the API as the reference standard at a concentration
corresponding to the limit established for individual unspecified impurities (e.g. 0.10%).
The system suitability tests (SSTs) represent an integral part of the method and are used
to ensure the adequate performance of the chosen chromatographic system. As a
minimum, HPLC and GC purity methods should include SSTs for resolution and
repeatability. For HPLC methods to control API-related impurities, this is typically done
using a solution of the API with a concentration corresponding to the limit for unspecified
impurities. Resolution of the two closest eluting peaks is generally recommended.
However, the choice of alternate peaks can be used if justified (e.g. choice of a toxic
impurity). In accordance with the Ph.Int. section on Methods of Analysis, the repeatability
test should include an acceptable number of replicate injections. HPLC assay methods
should include SSTs for repeatability and in addition either peak asymmetry, theoretical
plates or resolution. For TLC methods, the SSTs should verify the ability of the system to
separate and detect the analyte(s) (e.g. by applying a spot corresponding to the API at a
concentration corresponding to the limit of unspecified impurities).
Different sources of the same API or FPP can contain impurities and/or degradation
products that were not considered during the development of the monograph. Therefore
the monograph and compendial method should be demonstrated suitable to control the
impurity profile of the API from the intended source(s).
Page 38 of 88
FDA/DRI/DER/GL-RAD/2013/06
In general verification is not necessary for compendial API assay methods. However,
specificity of a specific compendial assay method should be demonstrated if there are
any potential impurities that are not specified in the compendial monograph. If an officially
recognized compendial method is used to control API-related impurities that are not
specified in the monograph, full validation of the method is expected with respect to those
impurities.
This data is used to establish the specifications and evaluate consistency in API quality.
Analytical results should be provided from at least two batches of at least pilot scale from
each proposed manufacturing site of the API and should include the batch(es) used in the
comparative bioavailability or biowaiver studies. A pilot-scale batch should be
manufactured by a procedure fully representative of and simulating that to be applied to
a full production- scale batch.
Copies of the certificates of analysis, both from the API manufacturer(s) and the FPP
manufacturer, should be provided for the profiled batches and any company responsible
for generating the test results should be identified. The FPP manufacturer’s test results
should be summarized in the dossier.
The discussion of results should focus on observations noted for the various tests, rather
than reporting comments such as “all tests meet specifications”. For quantitative tests
(e.g. individual and total impurity tests and assay tests), it should be ensured that actual
numerical results are provided rather than vague statements such as “within limits” or
“conforms”.
Page 39 of 88
FDA/DRI/DER/GL-RAD/2013/06
A discussion and justification should be provided for any incomplete analyses (e.g. results
not tested according to the proposed specification).
The justification for certain tests, analytical procedures and acceptance criteria may have
been discussed in other sections of the dossier (e.g. impurities, particle size distribution)
and does not need to be repeated here, although a cross-reference to their location should
be provided.
Information should be provided on the reference standard(s) used to generate data in the
dossier, as well as those to be used by the FPP manufacturer in routine API and FPP
testing.
The source(s) of the reference standards or materials used in the testing of the API should
be provided (e.g. those used for the identification, purity, and assay tests). These could
be classified as primary or secondary reference standards.
Otherwise, a primary standard may be a batch of the API that has been fully characterized
(e.g. by IR, UV, NMR, MS analyses). Further purification techniques may be needed to
render the material acceptable for use as a chemical reference standard. The purity
requirements for a chemical reference substance depend upon its intended use. A
Page 40 of 88
FDA/DRI/DER/GL-RAD/2013/06
chemical reference substance proposed for an identification test does not require
meticulous purification, since the presence of a small percentage of impurities in the
substance often has no noticeable effect on the test. On the other hand, chemical
reference substances that are to be used in assays should possess a high degree of
purity (such as 99.5% on the dried or water/solvent free basis). Absolute content of the
primary reference standard must be declared and should follow the scheme: 100% minus
organic impurities (quantitated by an assay procedure, e.g. HPLC, DSC, etc.) minus
inorganic impurities minus volatile impurities by loss on drying (or water content minus
residual solvents).
Reference standards should normally be established for specified impurities. Refer to 3.2.
S.4.2 for additional guidance.
A description of the container closure system(s) should be provided, including the identity
of materials of construction of each primary packaging component, and their
specifications.
The specifications should include description and identification (and critical dimensions
with drawings, where appropriate). Non-compendia methods (with validation) should be
included, where appropriate.
For non-functional secondary packaging components (e.g., those that do not provide
additional protection), only a brief description should be provided. For functional
secondary packaging components, additional information should be provided.
The suitability should be discussed with respect to, for example, choice of materials,
protection from moisture and light, compatibility of the materials of construction with the
drug substance, including sorption to container and leaching, and/or safety of materials
of construction.
Page 41 of 88
FDA/DRI/DER/GL-RAD/2013/06
The types of studies conducted, protocols used, and the results of the studies should be
summarized. The summary should include results, for example, from forced degradation
studies and stress conditions, as well as conclusions with respect to storage conditions
and retest date or shelf-life, as appropriate.
The WHO guideline Stability testing of active pharmaceutical ingredients and finished
pharmaceutical products should be consulted for recommendations on the core stability
data package required for product registration.
Stress testing of the API can help identify the likely degradation products, which can in
turn help establish the degradation pathways and the intrinsic stability of the molecule and
validate the stability indicating power of the analytical procedures used. The nature of the
stress testing will depend on the individual API and the type of FPP involved.
The objective of stress testing is not to completely degrade the API, but to cause
degradation to occur to a small extent, typically 10-30% loss of active by assay when
compared with non-degraded API. This target is chosen so that some degradation occurs,
but not enough to generate secondary products. For this reason, the conditions and
duration may need to be varied when the API is especially susceptible to a particular
stress factor. In the total absence of degradation products after 10 days, the API is
considered stable under the particular stress condition.
Photostability testing should be an integral part of stress testing. The standard conditions
are described in ICH Q1B. If “protect from light” is stated in one of the officially recognized
pharmacopoeia for the API, it is sufficient to state “protect from light” on labelling, in lieu
of photostability studies, when the container closure system is shown to be light
protective.
Available information on the stability of the API under accelerated and long-term
conditions should be provided, including information in the public domain or obtained from
scientific literature. The source of the information should be identified.
The required long-term storage conditions for APIs for the registration of the product is
either 30ºC±2ºC/65%±5%RH or 30ºC±2ºC/75%±5%RH. Studies covering the proposed
re-test period at the above mentioned long-term storage conditions will provide better
assurance of the stability of APIs at the conditions of the supply chain corresponding to
Page 42 of 88
FDA/DRI/DER/GL-RAD/2013/06
WHO Zone IVb. Alternative conditions should be supported with appropriate evidence,
which may include literature references or in-house studies, demonstrating that storage
at 30ºC is inappropriate for the API. For APIs intended for storage in a refrigerator and
those intended for storage in a freezer refer to the FDA stability guideline. APIs intended
for storage below -20°C should be treated on a case-by-case basis.
To establish the re-test period, data should be provided on not less than three batches of
at least pilot scale. The batches should be manufactured by the same synthesis route as
production batches and using a method of manufacture and procedure that simulates the
final process to be used for production batches. The stability testing programme should
be summarized and the results of stability testing should be summarized in the dossier
and in the tables in the dossier.
The information on the stability studies should include details such as storage conditions,
batch number, batch size, container closure system and completed (and proposed) test
intervals. The discussion of results should focus on observations noted for the various
tests, rather than reporting comments such as “all tests meet specifications”. Ranges of
analytical results where relevant and any trends that were observed should be included.
For quantitative tests (e.g. individual and total degradation product tests and assay tests),
it should be ensured that actual numerical results are provided rather than vague
statements such as “within limits” or “conforms”. Where
different from the methods described in S.4.2, descriptions and validation
of the methodology used in stability studies should be provided.
A storage statement should be established for display on the label based on the stability
evaluation of the API. The FDA stability guideline includes a number of recommended
storage statements that should be used, when supported by the stability studies.
A re-test period should be derived from the stability information and should be displayed
on the container label.
For APIs known to be labile (e.g. certain antibiotics), it is more appropriate to establish a
shelf-life rather than a re-test period
Page 43 of 88
FDA/DRI/DER/GL-RAD/2013/06
When available long term stability data on primary batches do not cover the proposed
retest period granted at the time of approval, a commitment should be made to continue
the stability studies post approval in order to firmly establish the re-test period.
Where the submission includes long-term stability data on three production batches
covering the proposed re-test period, a post approval commitment is considered
unnecessary.
The stability protocol for the commitment batches should be provided and should include,
but not be limited to, the following parameters:
• number of batch(es) and different batch sizes, if applicable;
• relevant physical, chemical, microbiological and biological test methods;
• acceptance criteria;
• reference to test methods;
• description of the container closure system(s);
• testing frequency;
• description of the conditions of storage (standardized conditions for long-term
testing as described in these guidelines and consistent with the API labelling,
should be used); and
• applicable parameters specific to the API.
Any differences in the stability protocols used for the primary batches and those
proposed for the commitment batches or ongoing batches should be scientifically
justified.
Results of the stability studies (e.g., forced degradation studies and stress conditions)
should be presented in an appropriate format such as tabular, graphical, or narrative.
Information on the analytical procedures used to generate the data and validation of these
procedures should be included.
The actual stability results used to support the proposed re-test period should be included
in the dossier. For quantitative tests (e.g. individual and total degradation product tests
and assay tests), it should be ensured that actual numerical results are provided rather
than vague statements such as “within limits” or “conforms”.
Page 44 of 88
FDA/DRI/DER/GL-RAD/2013/06
3.2. P.1 Description and Composition of the Drug Product (name, dosage form)
A description of the drug product and its composition should be provided. The information
provided should include, for example:
The description of the FPP should include the physical description, available strengths,
release mechanism (e.g. immediate, modified (delayed or extended)), as well as any
other distinguishable characteristics.
• Composition, i.e., list of all components of the dosage form, and their amount on
a per-unit basis (including overages, if any with justification) the function of the
components, and a reference to their quality standards (e.g., compendial
monographs or manufacturer’s specifications)
• The container closure used for the FPP (and accompanying reconstitution diluent,
if applicable) should be briefly described, with further details provided under
3.2.P.7 Container closure system.
Additionally, this section should identify and describe the formulation and process
attributes (critical parameters) that can influence batch reproducibility, product
performance and drug product quality. Supportive data and results from specific studies
Page 45 of 88
FDA/DRI/DER/GL-RAD/2013/06
Additional supportive data can be referenced to the relevant nonclinical or clinical sections
of the application.
These features should be discussed as part of the product development using the
principles of risk management over the entire lifecycle of the product (ref: ICH Q8). For a
discussion of additional pharmaceutical development issues specific to the development
of Fixed-Dose Combinations, reference should be made to WHO Technical Report
Series, No. 929, Annex 5, Section 6.3.2.
The compatibility of the drug substance with excipients listed in 3.2.P.1 should be
discussed. Additionally, key physicochemical characteristics (e.g., water content,
solubility, particle size distribution, polymorphic or solid state form) of the drug substance
that can influence the performance of the drug product should be discussed.
For fixed-dose combinations, the compatibility of drug substances with each other should
be discussed.
Page 46 of 88
FDA/DRI/DER/GL-RAD/2013/06
The choice of excipients listed in 3.2.P.1, their concentration, their characteristics that can
influence the drug product performance should be discussed relative to their respective
functions.
When choosing excipients, those with a compendial monograph are generally preferred.
Use of excipients in concentrations outside of established ranges is discouraged and
generally requires justification.
Ranges or alternates for excipients are normally not accepted, unless supported by
appropriate process validation data. Where relevant, compatibility study results (e.g.
compatibility of a primary or secondary amine API with lactose) should be included to
justify the choice of excipients. Specific details should be provided where necessary (E.g.
use of potato or corn starch).
Where antioxidants are included in the formulation, the effectiveness of the proposed
concentration of the antioxidant should be justified and verified by appropriate studies.
Antimicrobial preservatives are discussed in 3.2.P.2.5.
A brief summary describing the development of the drug product should be provided,
taking into consideration the proposed route of administration and usage. The differences
between clinical formulations and the formulation (i.e. composition) described in 3.2.P.1
should be discussed. Results from comparative in vitro studies (e.g., dissolution) or
comparative in vivo studies (e.g., bioequivalence) should be discussed when appropriate.
The requirements for bioequivalence studies should be taken into consideration example
when formulating multiple strengths and/or when the product is eligible for a biowaiver.
For product that have a functional score or break line, a study should be undertaken to
ensure the uniformity of dose in the tablet fragments. The data provided in the dossier
should include a description of the test method, individual values, mean and relative
standard deviation (RSD) of the results. Uniformity testing (i.e. content uniformity for split
portions containing less than 5 mg or less than 5% of the weight of the dosage unit portion,
or mass uniformity for other situations) should be performed on each split portion from a
minimum of 10 randomly selected whole tablets.
The splitting of the tablets should be performed in a manner that would be representative
of that used by the consumer (e.g. manually split by hand).
Page 47 of 88
FDA/DRI/DER/GL-RAD/2013/06
The uniformity test on split portions can be demonstrated on a one-time basis and does
not need to be added to the FPP specification(s). The tablet description in the
FPP specification and in the product information (e.g. summary of product characteristics,
labelling, package leaflet) should reflect the presence of a score.
Where relevant in the case of non-functional, labelling should state that the score line is
only to facilitate breaking for ease of swallowing and not to divide into equal doses.
The results of studies justifying the choice of in vitro dissolution or drug release conditions
(e.g. apparatus, rotation speed, medium) should be provided. Data should also be
submitted to demonstrate whether the method is sensitive to changes in manufacturing
processes and/or changes in grades and/or amounts of critical excipients and particle
size where relevant.
The dissolution method should be sensitive to any changes in the product that would
result in a change in one or more of the pharmacokinetic parameters. Use of a single point
test or a dissolution range should be justified based on the solubility and/or
biopharmaceutical classification of the API.
For extended-release FPPs, the testing conditions should be set to cover the entire time
period of expected release (e.g. at least three test intervals chosen for a 12-hour release
and additional test intervals for longer duration of release). One of the test points should
be at the early stage of drug release (e.g. within the first hour) to demonstrate absence of
dose dumping. At each test period, upper and lower limits should be set for individual
units. Generally, the acceptance range at each intermediate test point should not exceed
25% or ±12.5% of the targeted value. Dissolution results should be submitted for several
lots, including those lots used for pharmacokinetic and bioavailability or biowaiver studies.
Overages for the sole purpose of extending the shelf-life of the FPP are generally not
acceptable.
Parameters relevant to the performance of the drug product, such as pH, ionic strength,
dissolution, redispersion, reconstitution, particle size distribution, aggregation,
polymorphism, rheological properties, biological activity or potency, and/or immunological
activity, should be addressed.
Provide the formulation in tabular form for a typical batch and for an administration unit,
e.g. one tablet, 5 ml of oral solution, or the contents of an ampoule or bag of large volume
parenteral solution.
The rationale for choosing the particular pharmaceutical product (e.g. dosage form,
delivery system) should be provided. The scientific rationale for the choice of the
manufacturing, filling and packaging processes that can influence FPP quality and
performance should be explained (e.g. wet granulation using high shear granulator). API
stress study results may be included in the rationale. Any developmental work undertaken
to protect the FPP from deterioration should also be included (e.g. protection from light or
moisture).
Page 49 of 88
FDA/DRI/DER/GL-RAD/2013/06
The scientific rationale for the selection, optimization and scale-up of the manufacturing
process described in 3.2.P.3.3 should be explained, in particular the critical aspects (e.g.
rate of addition of granulating fluid, massing time, granulation end-point). A discussion of
the critical process parameters (CPP), controls and robustness with respect to the QTPP
and CQA of the product should be included.
Reference: ICH Q8
WHO Technical Report Series No. 970, 2012
Testing requirements to verify the suitability of the container closure system contact
material(s) depend on the dosage form and route of administration. The pharmacopoeias
provide standards that are required for packaging materials, including for example the
following:
Glass containers: USP <660>, Ph Eur 3.2.1
Plastic containers: Ph Eur 3.2.2, 3.2.2.1, USP <661>
Rubber/Elastomeric closures: USP <381>, Ph Eur 3.2.9
For solid oral dosage forms and solid APIs, compliance with regulations on food-contact
plastic materials, (for example (EU) No. 10/2011) can be considered acceptable.
The suitability of the container closure system used for the storage, transportation
(shipping) and use of any intermediate/in-process products (e.g. premixes, bulk FPP)
should also be discussed.
A device is required to be included with the container closure system for oral liquids or
solids (e.g. solutions, emulsions, suspensions and powders/granules for such), any time
the package provides for multiple doses.
In accordance with the Ph.Int. general chapter Liquid Preparations for Oral Use:
“Each dose from a multidose container is administered by means of a device suitable for
measuring the prescribed volume. The device is usually a spoon or a cup for volumes of
5 ml or multiples thereof, or an oral syringe for other volumes or, for oral drops, a suitable
dropper.”
Page 50 of 88
FDA/DRI/DER/GL-RAD/2013/06
Where appropriate, the microbiological attributes of the dosage form should be discussed,
including, for example, the rationale for not performing microbial limits testing for
nonsterile products and the selection and effectiveness of preservative systems in
products containing antimicrobial preservatives. For sterile products, the integrity of the
container closure system to prevent microbial contamination should be addressed.
The effectiveness of the agent should be justified and verified by appropriate studies (e.g.
USP or Ph.Eur. general chapters on antimicrobial preservatives) using a batch of the FPP.
If the lower limit for the proposed acceptance criterion for the assay of the preservative is
less than 90.0%, the effectiveness of the agent should be established with a batch of the
FPP containing a concentration of the antimicrobial preservative corresponding to the
lower proposed acceptance criteria.
As outlined in the WHO stability guideline (WHO Technical Report Series, No. 953, Annex
2, 2009), a single primary stability batch of the FPP should be tested for effectiveness of
the antimicrobial preservative (in addition to preservative content) at the proposed shelflife
for verification purposes, regardless of whether there is a difference between the release
and shelf-life acceptance criteria for preservative content.
3.2.P.2.6 Compatibility
Where a device is required for oral liquids or solids (e.g. solutions, emulsions,
suspensions and powders/granules for such reconstitution) that are intended to be
Page 51 of 88
FDA/DRI/DER/GL-RAD/2013/06
administered immediately after being added to the device, the compatibility studies
mentioned in the following paragraphs are not required.
Where the labelling does not specify the type of containers, compatibility (with respect to
parameters such as appearance, pH, assay, levels of individual and total degradation
products, subvisible particulate matter and extractables from the packaging components)
should be demonstrated in glass, PVC and polyolefin containers.
3.2.P.3 Manufacture
3.2.P.3.1 Manufacturer(s)
The name, address, and responsibility of each manufacturer, including contractors,
and each proposed production site or facility involved in manufacturing and testing
should be provided.
The facilities involved in the manufacturing, packaging, labelling and testing should be
listed.
If certain companies are responsible only for specific steps (e.g. manufacturing of an
intermediate), this should be clearly indicated.
The list of manufacturers/companies should specify the actual addresses of production or
manufacturing site(s) involved (including block(s) and unit(s)), rather than the
administrative offices.
For a mixture of an API with an excipient, the blending of the API with the excipient is
considered to be the first step in the manufacture of the final product and therefore the
mixture does not fall under the definition of an API. The only exceptions are in the cases
where the API cannot exist on its own. Similarly, for a mixture of APIs, the blending of the
APIs is considered to be the first step in the manufacture of the final product. Sites for
such manufacturing steps should be included in this section.
Page 52 of 88
FDA/DRI/DER/GL-RAD/2013/06
For each site where the major production step(s) are carried out, when applicable, attach
a WHO-type certificate of GMP issued by the competent authority in terms of the WHO
Certification Scheme on the Quality of Pharmaceutical Products Moving in International
Commerce (Module 1).
Justification for any differences to the product in the country or countries issuing
the WHO-type certificate(s)
When there are differences between the product for which this application is submitted
and that marketed in the country/countries which provided the WHO-type certificate(s),
provide data to support the applicability of the certificate(s) despite the differences.
Depending on the case, it may be necessary to provide validation data for differences in
site of manufacture, specifications, formulation, etc. Note that only minor differences are
likely to be acceptable. Differences in container labelling need not normally be justified.
A batch formula should be provided that includes a list of all components of the
dosage form to be used in the manufacturing process, their amounts on a per batch
basis,including overages, and a reference to their quality standards.
Provide a table in the application form to summarize the batch formula of the
FPP for each proposed commercial batch size and express the quantity of each
component on a per batch basis, including a statement of the total weight or measure of
the batch.
All components used in the manufacturing process should be included, including those
that may not be added to every batch (e.g. acid and alkali), those that may be removed
during processing (e.g. solvents) and any others (e.g. nitrogen, silicon for stoppers).
Page 53 of 88
FDA/DRI/DER/GL-RAD/2013/06
If the FPP is formulated using an active moiety, then the composition for the active
ingredient should be clearly indicated (e.g. “1 kg of active ingredient base = 1.075 kg
active ingredient hydrochloride”). All overages should be clearly indicated (e.g. “Contains
5 kg (corresponding to 2%) overage of the API to compensate for manufacturing losses”).
The components should be declared by their proper or common names, quality standards
(e.g. International Pharmacopoeia
(Ph.Int)., European Pharmacopoeia (Ph.Eur)., British Pharmacopoeia (BP), USP,
Japanese Pharmacopoeia (JP), In-House) and, if applicable, their grades (e.g.
“Microcrystalline Cellulose NF (PH 102)”) and special technical characteristics (e.g.
lyophilized, micronized, solubilised, emulsified).
Steps in the process should have the appropriate process parameters identified, such as
time, temperature, or pH. Associated numeric values can be presented as an expected
range. Numeric ranges for critical steps should be justified in Section 3.2.P.3.4. In certain
cases, environmental conditions (e.g., low humidity for an effervescent product) should
be stated.
The maximum holding time for bulk FPP prior to final packaging should be stated. The
holding time should be supported by the submission of stability data, if longer than 30
days.
For an aseptically processed FPP, sterile filtration of the bulk and filling into final
containers should preferably be continuous; any holding time should be justified.
For the manufacture of sterile products, the class (e.g. A, B, C etc.) of the areas should
be stated for each activity (e.g. compounding, filling, sealing etc), as well as the
sterilization parameters for equipment, container/closure, terminal sterilization etc.
Critical Steps: Tests and acceptance criteria should be provided (with justification,
including experimental data) performed at the critical steps identified in 3.2.P.3.3 of the
manufacturing process, to ensure that the process is controlled.
Intermediates: Information on the quality and control of intermediates isolated during the
process should be provided.
Examples of applicable in-process controls include:
• granulations: moisture (limits expressed as a range), blend uniformity (e.g. low
dose tablets), bulk and tapped densities, particle size distribution;
• solid oral products: average weight, weight variation, hardness, thickness, friability,
and disintegration checked periodically throughout compression, weight gain
during coating;
• semi-solids: viscosity, homogeneity, pH;
• transdermal dosage forms: assay of API-adhesive mixture, weight per area of
coated patch without backing;
• metered dose inhalers: fill weight/volume, leak testing, valve delivery;
• dry powder inhalers: assay of API-excipient blend, moisture, weight variation of
individually contained doses such as capsules or blisters;
• liquids: pH, specific gravity, clarity of solutions; and
• parenterals: appearance, clarity, fill volume/weight, pH, filter integrity tests,
particulate matter, leak testing of ampoules, pre-filtration and/or pre-sterilization
bioburden testing.
c) if the process validation studies have already been conducted (e.g. for sterile products),
a copy of the process validation report should be provided in the dossier in lieu of (a)
and (b) above.
Where ranges of batch sizes are proposed, it should be shown that variations in batch
size would not adversely alter the characteristics of the finished product. It is envisaged
that those parameters listed in the following validation scheme will need to be revalidated
once further scale-up is proposed after registration.
The process validation protocol should include inter alia the following:
• a reference to the current master production document;
• a discussion of the critical equipment;
• the process parameters that can affect the quality of the FPP (critical process
parameters (CPPs)) including challenge experiments and failure mode operation;
• details of the sampling: sampling points, stages of sampling, methods of sampling
and the sampling plans (including schematics of blender/storage bins for uniformity
testing of the final blend);
• the testing parameters/acceptance criteria including in-process and release
specifications and including comparative dissolution profiles of validation batches
against the batch(es) used in the bioavailability or biowaiver studies;
• the analytical procedures or a reference to appropriate section(s) of the dossier;
• the methods for recording/evaluating results; and • the proposed timeframe for
completion of the protocol.
3.2.P.4.1 Specifications
Page 56 of 88
FDA/DRI/DER/GL-RAD/2013/06
For excipients of natural origin, microbial limit testing should be included in the
specifications. Skip testing is acceptable if justified (submission of acceptable results of
five production batches).
For oils of plant origin (e.g. soy bean oil, peanut oil) the absence of aflatoxins or biocides
should be demonstrated.
The colours permitted for use are limited to those listed in the “Japanese pharmaceutical
excipients”, the EU “List of permitted food colours”, and the USFDA “Inactive ingredient
guide”. For proprietary mixtures, the supplier's product sheet with the qualitative
formulation should be submitted, in addition to the FPP manufacturer's specifications for
the product including identification testing.
For flavours the qualitative composition should be submitted, as well as a declaration that
the excipients comply with foodstuff regulations (e.g. USA or EU).
Information that is considered confidential may be submitted directly to the FDA by the
supplier with reference to the specific related product.
Page 57 of 88
FDA/DRI/DER/GL-RAD/2013/06
A discussion of the tests that are supplementary to those appearing in the officially
recognized compendial monograph as per the FDA officially recognised list of publications
should be provided.
The following excipients should be addressed in this section: gelatin, phosphates, stearic
acid, magnesium stearate and other stearates. If from plant origin a declaration to this
effect will suffice.
For these excipients from animal origin, a letter of attestation should be provided
confirming that the excipients used to manufacture the FPP are without risk of transmitting
agents of animal spongiform encephalopathies.
For excipient(s) used for the first time in an FPP or by a new route of administration, full
details of manufacture, characterisation, and controls, with cross references to supporting
safety data (nonclinical and/or clinical) should be provided according to the API and/or
FPP format. (Details in 3.2.A.3).
3.2.P.5.1 Specification(s)
Page 58 of 88
FDA/DRI/DER/GL-RAD/2013/06
tested according to the listed analytical procedures, will meet the listed acceptance
criteria. Specifications are critical quality standards that are proposed and justified by the
manufacturer and approved by regulatory authorities.”
A copy of the FPP specification(s) from the applicant (as well as the company responsible
for the batch release of the FPP, if different from the applicant), dated and signed by
authorized personnel (i.e. the person in charge of the quality control or quality assurance
department) should be provided in the dossier.
Two separate sets of specifications may be set out: after packaging of the FPP
(release) and at the end of shelf-life.
The specifications should be provided in a tabular form in the dossier and should include
the tests, acceptance criteria and analytical procedures. The analytical procedure should
include the types, sources and versions for the methods:
• the standard declared by the applicant could be as per the FDA officially recognised
list of publications or an in- House (manufacturer’s) standard;
• the specification reference number and version (e.g. revision number and/or date)
should be provided for version control purposes;
• for the analytical procedures, the type should indicate the kind of analytical
procedure used (e.g. visual, IR, UV, HPLC), the source refers to the origin of the
analytical procedure (e.g. Ph.Int., Ph.Eur., BP, USP, in-house) and the version (e.g.
code number/version/date) should be provided for version control purposes.
The following information provides guidance for specific tests that are not addressed by
ICH‟s Q6A guideline:
• fixed-dose combination FPPs (FDC-FPPs):
• analytical methods that can distinguish each API in the presence of the other API(s)
should be developed and validated,
• acceptance criteria for degradation products should be established with reference to
the API they are derived from. If an impurity results from a chemical reaction between
two or more APIs, its acceptance limits should in general be calculated with
reference to the worst case (the API with the smaller area under
the curve). Alternatively the content of such impurities could be calculated in
relation to their reference standards,
• a test and limit for content uniformity is required for each API present in the FPP at
less than 5 mg or less than 5% of the weight of the dosage unit,
• for the API(s) present at equal or greater than 5 mg and equal or greater than 5% of
the weight of the dosage unit, a test and limit for weight variation may be established
in lieu of content uniformity testing;
Page 59 of 88
FDA/DRI/DER/GL-RAD/2013/06
Inhalation and nasal products: consistency of delivered dose (throughout the use of the
product), particle or droplet size distribution profiles (comparable to the product used in in
vivo studies, where applicable) and if applicable for the dosage form, moisture content,
leak rate, microbial limits, preservative assay, sterility and weight loss;
Suppositories: uniformity of dosage units, melting point; and Transdermal dosage forms:
peal or shear force, mean weight per unit area, dissolution.
Unless there is appropriate justification, the acceptable limit for the API content of the FPP
in the release specifications is ± 5% of the label claim (i.e. 95.0-105.0%).
For products such as tablets, capsules and suppositories where a test for uniformity of
single dose preparations is required, a test and limit for content uniformity is required
when the API is present in the FPP at less than 5 mg or less than 5% of the weight of the
dosage unit. Otherwise, the test for mass uniformity may be applied.
Skip testing is acceptable for parameters such as identification of colouring materials and
microbial limits, when justified by the submission of acceptable supportive results for five
production batches. When skip testing justification has been accepted, the specifications
should include a footnote, stating at minimum the following skip testing requirements: at
minimum every tenth batch and at least one batch annually is tested. In addition, for
stability- indicating parameters such as microbial limits, testing will be performed at
release and shelf- life during stability studies.
Any differences between release and shelf-life tests and acceptance criteria should be
clearly indicated and justified. Note that such differences for parameters such as
dissolution are normally not accepted.
All analytical test procedures, including biological and microbiological methods where
relevant, must be described in sufficient detail to enable the procedures to be repeated if
necessary.
Page 60 of 88
FDA/DRI/DER/GL-RAD/2013/06
Information on the summary of analytical procedures and validation information used for
determination of assay, related substances and dissolution of the FPP should be provided
in a tabular form in 2.3.R.2.
Copies of the protocol and reports for method validation including representative
chromatograms/tracings for the in-house analytical procedures used during
pharmaceutical development (if used to support testing results provided in the dossier) as
well as those proposed for routine testing should be provided.
The monograph and compendial method(s) should be demonstrated suitable for the
control of the proposed FPP.
For officially recognized compendial as per the FDA officially recognised list of
publications for FPP assay methods, verification should include a demonstration of
specificity, accuracy and repeatability (method precision). If an officially recognized
compendial method is used to control related substances that are not specified in the
monograph, full validation of the method is expected with respect to those related
substances.
Page 61 of 88
FDA/DRI/DER/GL-RAD/2013/06
Information should include strength and batch number, batch size, date and site of
production and use (e.g. used in comparative bioavailability or biowaiver studies,
preclinical and clinical studies (if relevant), stability, pilot, scale-up and, if available,
production-scale batches) on relevant FPP batches used to establish the specification(s)
and evaluate consistency in manufacturing.
Analytical results tested by the company responsible for the batch release of the FPP
(generally, the applicant or the FPP manufacturer, if different from the applicant) should
be provided for not less than two batches of at least pilot scale.
The testing results should include the batch(es) used in the comparative bioavailability or
biowaiver studies. Copies of the certificates of analysis for these batches should be
provided in the dossier and the company responsible for generating the testing results
should be identified.
The discussion of results should focus on observations noted for the various tests, rather
than reporting comments such as “all tests meet specifications”. This should include
ranges of analytical results, where relevant. For quantitative tests (e.g. individual and total
impurity tests and assay tests), it should be ensured that actual numerical results are
provided rather than vague statements such as “within limits” or “conforms” (e.g. “levels
of degradation product A ranged from 0.2 to 0.4%”).
Dissolution results should be expressed at minimum as both the average and range of
individual results.
A discussion should be provided of all impurities that are potential degradation products
(including those among the impurities identified in 3.2.S.3.2 as well as potential
degradation products resulting from interaction of the API with other APIs (FDCs),
excipients or the container closure system) and FPP process-related impurities (e.g.
residual solvents in the manufacturing process for the FPP).
Page 62 of 88
FDA/DRI/DER/GL-RAD/2013/06
The justification for certain tests, analytical procedures and acceptance criteria (e.g.
degradation products, dissolution method development) may have been discussed in
other sections of the dossier and does not need to be repeated here, although a
crossreference to their location should be provided.
See Section 3.2.S.5 for information that should be provided on reference standards or
materials. Information should be provided on reference materials of FPP degradation
products, where not included in 3.2.S.5.
For non-functional secondary packaging components (e.g., those that neither provide
additional protection nor serve to deliver the product), only a brief description should be
provided. For functional secondary packaging components, additional information should
be provided.
Suitability information should be located in 3.2.P.2.
Page 63 of 88
FDA/DRI/DER/GL-RAD/2013/06
Descriptions, materials of construction and specifications (of the company responsible for
packaging the FPP, generally the FPP manufacturer) should be provided for the
packaging components that are:
• in direct contact with the dosage form (e.g. container, closure, liner, desiccant, filler);
• used for drug delivery (including the device(s) for multi-dose solutions, emulsions,
suspensions and powders/granules for such);
• used as a protective barrier to help ensure stability or sterility; and necessary to
ensure FPP quality during storage and shipping.
Primary packaging components are those that are in direct contact with the API or FPP.
The specifications for the primary packaging components should include a specific test
for identification (e.g. IR).
Information to establish the suitability (e.g. qualification) of the container closure system
should be discussed in Section 3.2.P.2. Comparative studies may be warranted for certain
changes in packaging components (e.g. comparative delivery study (droplet size) for a
change in manufacturer of dropper tips).
3.2.P.8 Stability
3.2.P.8.1 Stability Summary and Conclusions
The types of studies conducted, protocols used, and the results of the studies should be
summarised. The summary should include, for example, conclusions with respect to
storage conditions and shelf-life, and, if applicable, in-use storage conditions and shelf-
life.
The design of the formal stability studies for the finished product should be based on
knowledge of the behaviour and properties of the API and the dosage form. Describe the
methodology used during stability studies; if this is identical to methodology described
elsewhere in the data set, a cross-reference will suffice. If different methodology was
used, the test procedures applied to the stability tests on the finished product should be
validated or verified, and the accuracy as well as the precision (standard deviations)
should be recorded. Characterize the possible degradants identified by stress stability
testing during development pharmaceutics (compatibilities of the APIs with each other
Page 64 of 88
FDA/DRI/DER/GL-RAD/2013/06
and with the excipients as well as the effect of temperature on the rate of degradation
reactions). The tests for degradants should be validated to demonstrate that they are
specific to the FPP being examined and are of adequate sensitivity.
For long-term and accelerated stability studies, refer to FDA Guidelines on stability studies
Any in-use period and associated storage conditions should be justified with experimental
data, for example after opening, reconstitution and/or dilution of any sterile and/or
multidose products or after first opening of FPPs packed in bulk multidose containers (e.g.
bottles of
1000‟s). If applicable, the in-use period and storage conditions should be stated in the
product information.
The discussion of results should focus on observations noted for the various tests, rather
than reporting comments such as “all tests meet specifications”. This should include
ranges of analytical results and any trends that were observed. For quantitative tests (e.g.
individual and total degradation product tests and assay tests), it should be ensured that
actual numerical results are provided rather than vague statements such as “within limits”
or “conforms”. Dissolution results should be expressed at minimum as both the average
and range of individual results.
Reference document: ICHQ1A, Q1B, Q1C, Q1D, Q1E, Q3B, Q6A and Q6B WHO
Technical Report Series No. 970, 2012
When available long term stability data on primary batches do not cover the proposed
shelf-life granted at the time of approval, a commitment should be made to continue the
stability studies post approval in order to firmly establish the shelf-life.
Where the submission includes long-term stability data on three production batches
covering the proposed shelf-life, a post approval commitment is considered unnecessary.
If the submission includes data from stability studies on fewer than three production
batches, a commitment should be made to continue these studies through the proposed
Page 65 of 88
FDA/DRI/DER/GL-RAD/2013/06
shelf-life and to place additional production batches, to a total of at least three, on long
term stability studies through the proposed shelf-life.
If the submission does not include stability data on production batches, a commitment
should be made to place the first three production batches on long term stability studies
through the proposed shelf-life.
The stability protocol used for long-term studies for the stability commitment should be
the same as that for the primary batches, unless otherwise scientifically justified
The actual stability results/reports used to support the proposed shelf-life should be
provided in the dossier. For quantitative tests (e.g. individual and total degradation
product tests and assay tests), it should be ensured that actual numerical results are
provided rather than vague statements such as “within limits” or “conforms”. Dissolution
results should be expressed at minimum as both the average and range of individual
results.
Reference documents: ICH Q1A, Q1B, Q1C, Q1D, Q1E, Q2 and Q5C
3.2. A APPENDICES
Page 66 of 88
FDA/DRI/DER/GL-RAD/2013/06
For novel excipients: a dossier should be established containing the same data as
required for new active substances:
Page 67 of 88
FDA/DRI/DER/GL-RAD/2013/06
a) A strict definition of the excipient, its function and its conditions of use. If the
excipient is complex or is made of a mixture of compounds, the composition should be
specified in qualitative and quantitative terms.
b) For novel excipients and for excipients presented as a mixture of compounds the
following should be taken into consideration:
i. Any bibliographical data on the chemistry and on the toxicology and the field in which
the product is already used. ii. The provisions concerning additives in foodstuffs: any
criteria which are based on the toxicological data, with cross-references to these data.
The quality specifications which have been laid down in the directives are satisfactory
as long as the routine control tests used are validated. iii. The international
specifications (FAO/WHO/JECFA), and other publications such as the Food Chemical
Codex. iv. For medicinal products for cutaneous use, data on the starting material in
cosmetic products.
v. Data concerning the toxicology of the novel excipient should be presented according
to the dosage form and the route of administration of the medicinal product (if
applicable).
For solid oral dosage forms, pilot scale is generally, at a minimum, one-tenth that of full
production scale or 100 000 tablets or capsules, whichever is the larger. These batches
should be manufactured by a procedure fully representative of and simulating that to be
applied to a full production-scale batch.
Copies of the executed production documents for these batches should be submitted.
a) master formula;
b) dispensing, processing and packaging sections with relevant material and operational
details;
Page 68 of 88
FDA/DRI/DER/GL-RAD/2013/06
c) relevant calculations (e.g. if the amount of API is adjusted based on the assay results
or on the anhydrous basis);
d) identification of all equipment by, at minimum, type and working capacity (including
make, model and equipment number, where possible);
e) process parameters (e.g. mixing time, mixing speed, milling screen size, processing
temperature range, granulation end-point, tablet machine speed (expressed as target
and range));
f) list of in-process tests (e.g. appearance, pH, assay, blend uniformity, viscosity, particle
size distribution, Loss on Drying (LOD), weight variation, hardness, disintegration time,
weight gain during coating, leaker test, minimum fill, clarity, filter integrity checks) and
specifications; g) sampling plan with regard to the:
• steps where sampling should be done (e.g. drying, lubrication, compression),
• number of samples that should be tested (e.g. for blend uniformity testing of low
dose FPPs, blend drawn using a sampling thief from x positions in the blender),
• frequency of testing (e.g. weight variation every x minutes during compression or
capsule filling);
h) precautions necessary to ensure product quality (e.g. temperature and humidity
control, maximum holding times);
i) for sterile products, reference to SOPs in appropriate sections and a list of all
relevant SOPs at the end of the document; j) theoretical and actual yield; k)
compliance with the GMP requirements.
This guideline presents the organisation of the nonclinical reports in the applications that
will be submitted. This guideline is not intended to indicate what studies are required. It
merely indicates an appropriate format for the nonclinical data that have been acquired.
The appropriate location for individual-animal data is in the study report or as an appendix
to the study report.
Page 69 of 88
FDA/DRI/DER/GL-RAD/2013/06
4.2.1 Pharmacology
4.2.1.1 Primary Pharmacodynamics
4.2.1.2 Secondary Pharmacodynamics
4.2.1.3 Safety Pharmacology
4.2.1.4 Pharmacodynamic Drug Interactions
4.2.2 Pharmacokinetics
4.2.2.1 Analytical Methods and Validation Reports (if separate reports are available)
4.2.2.2 Absorption
4.2.2.3 Distribution
4.2.2.4 Metabolism
4.2.2.5 Excretion
4.2.2.6 Pharmacokinetic Drug Interactions (nonclinical)
4.2.2.7 Other Pharmacokinetic Studies
4.2.3 Toxicology
4.2.3.1 Single-Dose Toxicity (in order by species, by route)
4.2.3.2 Repeat-Dose Toxicity (in order by species, by route, by duration; including
supportive toxicokinetics evaluations)
4.2.3.3 Genotoxicity
4.2.3.3.1 In vitro
4.2.3.3.2 In vivo (including supportive toxicokinetics evaluations)
4.2.3.4 Carcinogenicity (including supportive toxicokinetics evaluations)
4.2.3.4.1 Long-term studies (in order by species; including range-finding studies that
cannot appropriately be included under repeat-dose toxicity or pharmacokinetics)
4.2.3.4.2 Short- or medium-term studies (including range-finding studies that cannot
appropriately be included under repeat-dose toxicity or pharmacokinetics)
4.2.3.4.3 Other studies
4.2.3.5 Reproductive and Developmental Toxicity (including range-finding studies and
supportive toxicokinetics evaluations) (If modified study designs are used, the following
sub-headings should be modified accordingly.) 4.2.3.5.1 Fertility and early embryonic
development
4.2.3.5.2 Embryo-fetal development
4.2.3.5.3 Prenatal and postnatal development, including maternal function
4.2.3.5.4 Studies in which the offspring (juvenile animals) are dosed and/or further
evaluated.
4.2.3.6 Local Tolerance
4.2.3.7 Other Toxicity Studies (if available)
4.2.3.7.1 Antigenicity
4.2.3.7.2 Immunotoxicity
4.2.3.7.3 Mechanistic studies (if not included elsewhere)
4.2.3.7.4 Dependence
Page 70 of 88
FDA/DRI/DER/GL-RAD/2013/06
4.2.3.7.5 Metabolites
4.2.3.7.6 Impurities
4.2.3.7.7 Other
References:
Non-Clinical Safety Studies For The Conduct Of Human Clinical Trials For
Pharmaceuticals (ICH M3[R2]) modification;
http://www.ema.europa.eu/pdfs/human/ich/028695en.pdf
EMEA: Non- Clinical Scientific Guidelines;
http://www.ema.europa.eu/htms/human/humanguidelines/nonclinical.htm CTD M4S
(R2) SAFETY; http://www.ich.org/LOB/media/MEDIA556.pdf
This module provides guidance on the organization of the study reports, other clinical
data, and references within an application for registration of a pharmaceutical product.
These elements should facilitate the preparation and review of a marketing application.
This module is not intended to indicate what studies are required for successful
registration.
It indicates an appropriate organization for the clinical study reports that are in the
application. This module recommends a specific organization for the placement of clinical
study reports and related information to simplify preparation and review of dossiers and
to ensure completeness. The placement of a report should be determined by the primary
objective of the study. Each study report should appear in only one section. Where there
are multiple objectives, the study should be cross-referenced in the various sections. An
explanation such as ―not applicable or ―no study conducted should be provided when
no report or information is available for a section or subsection.
Page 71 of 88
FDA/DRI/DER/GL-RAD/2013/06
sequence described in Section 5.1.3 below. Use of a different sequence should be noted
and explained in an introduction to the tabular listing.
• studies comparing the release and systemic availability of a drug substance from a solid
oral dosage form to the systemic availability of the drug substance given intravenously
or as an oral liquid dosage form
• dosage form proportionality studies, and
• food-effect studies.
Page 72 of 88
FDA/DRI/DER/GL-RAD/2013/06
These studies should characterise the drug‘s PK and provide information about the
absorption, distribution, metabolism, and excretion of a drug and any active metabolites
in healthy subjects and/or patients. Studies of mass balance and changes in PK related
to dose (e.g., determination of dose proportionality) or time (e.g., due to enzyme induction
or formation of antibodies) are of particular interest and should be included in Sections
5.1.3.3.1 and/or 5.1.3.3.2. Apart from describing mean PK in normal and patient
volunteers, PK studies should also describe the range of individual variability. In the ICH
E5 guideline on Ethnic Factors in the Acceptance of Foreign Data, factors that may result
in different responses to a drug in different populations are categorised as intrinsic ethnic
factors or extrinsic ethnic factors. In this document, these categories are referred to as
intrinsic factors and extrinsic factors, respectively. Additional studies can also assess
differences in systemic exposure as a result of changes in PK due to intrinsic (e.g., age,
gender, racial, weight, height, disease, genetic polymorphism, and organ dysfunction) and
extrinsic (e.g., drug-drug interactions, diet, smoking, and alcohol use) factors. Reports of
PK studies examining the influence of intrinsic and extrinsic factors on exposure should
be organised in Sections 5.1.3.3.3 and 5.1.3.3.4, respectively. In addition to standard
multiple-sample PK studies, population PK analyses based on sparse sampling during
clinical studies can also address questions about the contributions of intrinsic and
extrinsic factors to the variability in the dose-PKresponse relationship. Because the
methods used in population PK studies are substantially different from those used in
standard PK studies, these studies should be placed in Section 5.1.3.3.5.
Page 74 of 88
FDA/DRI/DER/GL-RAD/2013/06
Page 75 of 88
FDA/DRI/DER/GL-RAD/2013/06
The study reports should provide the level of detail appropriate to the study and its role in
the application. ICH E3 describes the contents of a full report for a study contributing
evidence pertinent to both safety and efficacy. Abbreviated reports can be provided for
some studies (see ICH E3 and individual guidance by region).
Within each section, studies should be categorized further, ordered by whether the study
report is complete or abbreviated (ICH E3), with completely reported studies presented
first. Published reports with limited or no further data available to the sponsor should be
placed last in this section.
In cases where the application includes multiple therapeutic indications, the reports
should be organized in a separate Section 5.1.3.5 for each indication. In such cases, if a
clinical efficacy study is relevant to only one of the indications included in the application,
it should be included in the appropriate Section 5.1.3.5; if a clinical efficacy study is
relevant to multiple indications, the study report should be included in the most
appropriate Section 5.1.3.5 and referenced as necessary in other Sections 5.1.3.5, e.g.,
Section 5.1.3.5A, Section 5.1.3.5B.
5.1.3.5.1 Study Reports of Controlled Clinical Studies Pertinent to the Claimed Indication
The controlled clinical study reports should be sequenced by type of control: Placebo
control (could include other control groups, such as an active comparator or other doses)
No-treatment control Dose-response (without placebo) Active control (without placebo)
External (Historical) control, regardless of the control treatment Within each control type,
where relevant to assessment of drug effect, studies should be organized by treatment
duration. Studies of indications other than the one proposed in the application, but that
provide support for efficacy in the proposed use, should be included in Section 5.1.3.5.1.
Where a pharmacodynamic study contributes to evidence of efficacy, it should be
included in Section 5.1.3.5.1. The sequence in which studies were conducted is not
considered pertinent to their presentation. Thus, placebo-controlled trials, whether early
or late, should be placed in Section 5.1.3.5.1. Controlled safety studies, including studies
in conditions that are not the subject of the application, should also be reported in Section
5.1.3.5.1.
Page 76 of 88
FDA/DRI/DER/GL-RAD/2013/06
Study reports of uncontrolled clinical studies (e.g., reports of open label safety studies)
should be included in Section 5.1.3.5.2. This includes studies in conditions that are not
the subject of the marketing application.
Page 77 of 88
FDA/DRI/DER/GL-RAD/2013/06
APPLICATIONS ONLY)
5.2.1.1 Bioavailability (BA) Study Reports BA studies in this section should include:
studies comparing the release and systemic availability of a drug substance from a solid
oral dosage form to the systemic availability of the drug substance given intravenously or
as an oral liquid dosage form dosage form proportionality studies, and
food-effect studies.
the drug product used in clinical studies supporting effectiveness and the to-
bemarketed drug product,
the drug product used in clinical studies supporting effectiveness and the drug product
used in stability batches, and
similar drug products from different manufacturers.
Page 78 of 88
FDA/DRI/DER/GL-RAD/2013/06
Bio-equivalence study report should contain at least the following items as described in
part 2 of the application form see Annex X:
Description of study design. The most appropriate study type is two-period, randomized,
crossover study. If other study types were used (e.g. parallel group design), these
should be justified by the applicant. In general, single-dose study with sufficiently long
period for blood samples collection is acceptable.
Data about preparations used: manufacturer, place of manufacture, batch number etc.
Reference preparation in bio-equivalence study should be innovator preparation from an
ICH associated countries or from WHO list of international comparator products if listed.
Page 79 of 88
FDA/DRI/DER/GL-RAD/2013/06
The 90% confidence interval for the AUC-ratio should lie within a bio-equivalence range
of 80-125%. In some specific cases of drugs with a narrow therapeutic range the
acceptance range may need to be tightened. The 90% confidence interval for the
Cmaxratio should lie within a bio-equivalence range of 80-125%. In some specific cases
of drugs with a narrow therapeutic range the acceptance range may need to be
tightened. In certain cases for drugs with an inherently high intra-subject variability, a
wider acceptance range (e.g., 75-133%) may be acceptable. The range used must be
defined prospectively and should be justified, taking into account safety and efficacy
considerations. Reference: EMEA, as described in "Guideline On The Investigation Of
Bioequivalence 2010", available at:
http://www.ema.europa.eu/pdfs/human/qwp/140198enrev1fin.pdf
Page 80 of 88
FDA/DRI/DER/GL-RAD/2013/06
Page 81 of 88
FDA/DRI/DER/GL-RAD/2013/06
To investigate batch to batch consistency of the products (test and reference) to be used
as basis for the selection of appropriate batches for the in vivo study.
ONLY)
Specific requirements for toxicity studies may vary from one drug to another depending
on the class of veterinary drug and the extent of its proposed use.
5.3.2.2 Microbiological Safety Studies
Page 83 of 88
FDA/DRI/DER/GL-RAD/2013/06
In this section of the Human Safety Requirements, information is provided regarding the
data requirements expected for demonstrating the microbiological safety of a drug
product. This section pertains to antimicrobial drug products as well as products
containing bacteria, for example, direct-fed microbial products.
REFERENCES
1. Guide to Care and Use of Experimental Animals, CCAC, 1993. (Website:
http://www.ccac.ca)
2. Target Animal Safety Guidelines for New Animal Drugs. Office of New Animal Drug
Evaluation. Centre for Veterinary Medicine, Food and Drug Administration, Rockville ,
MD 20855 , USA . 2001.
3. Uses of Antimicrobials in Food Producing Animals. Advisory Committee on Animal
Uses of Antimicrobials and Impact on Resistance and Human Health, 2002. (
http://www.hc-sc.gc.ca/dhp-mps/pubs/vet/amr-ram_final_report-rapport_06-
27_cppceng.php
4. http://www.hc-sc.gc.ca/dhp-mps/vet/legislation/guide-ld/vdd_nds_guide-eng.php#7
Page 84 of 88
FDA/DRI/DER/GL-RAD/2013/06
4. LANGUAGE
All applications and supporting documents shall be in English and legible. Where material
is not originally in English, a copy in the original language and a full translation should be
submitted, the accuracy of the translation is the responsibility of the applicant.
Authentication of the translation has to be done at the nearest Ghana Embassy or by the
National Drug Regulatory Authority of the country from where the document originates.
Reports submitted only in a language other than English will not be accepted.
5. DATA PRESENTATION
All information, data, tables, diagrams, attachments must be legible of font size 12 or
more. Pages must be presented in PDF readable format for Modules 2, 3,4 and 5 and a
MS-word version for the following;
• Module 1 and the following attachments to module 1
• SmPc, PIL
• QIS – refer to template of QIS
• Module 2.3- QOS
All pages shall be numbered sequentially with the format page numbered as page x of y
and have a table of contents indicating the sections and page numbers in the relevant
sections of the application form. Before submitting the completed form, check that you
have provided all requested information. Acronyms and abbreviations should be defined
the first time they are used in each part.
Dossiers should be arranged sequentially on the CD. The PDF data should be presented
in readable format on the CD. Dossiers could be submitted in volumes for the different
parts but shall be numbered serially (e.g. volume 1 of 2) for ease of reference.
References should be cited in accordance with the current edition of the Uniform
Requirements for Manuscripts Submitted to Biomedical Journals, International
Committee of Medical Journal Editors (ICMJE).
When direct reference is made to specifications, quality control procedures and test
methods in official compendia (FDA officially recognised list of publications), text books
or standard publications, reprints or authenticated copies of relevant pages shall be
enclosed. References to pharmacopoeias should be as per the current editions.
References should be provided for all in-house processes.
There shall be no cross reference of particulars or documentation between one product
and another.
Page 85 of 88
FDA/DRI/DER/GL-RAD/2013/06
7. SUBMISSION OF APPLICATION
Page 86 of 88
FDA/DRI/DER/GL-RAD/2013/06
2. Samples of the FPP as per FDA sample schedule (find information on sample
schedule from https://fdaghana.gov.gh/wp-content/uploads/2017/06/SAMPLE-SCHEDULE-FOR-REGISTRATIONAND-
RE.pdf ).
5. Non-refundable GMP inspection fee for facilities not yet inspected by the FDA (find
information on Approved fee schedule from
https://fdaghana.gov.gh/images/stories/pdfs/Quick%20links/FDA%20FEES%20SCHEDULE.pdf ).
5. Any other requirements that the FDA may determine from time to time.
8. PAYMENT OF FEES
Page 87 of 88
FDA/DRI/DER/GL-RAD/2013/06
The evaluation of applications is done on a first in first out (FIFO) basis unless the product
meets the expedited review process as set out in this guideline.
An application may be expedited if the product is for:
a. Public health programmes. These include Human Immune-deficiency Virus (HIV)/
Acquired Immune Deficiency Syndrome, Malaria, Tuberculosis, reproductive
health, neglected tropical diseases e.g. Buruli Ulcer, products on the Expanded
Programme of Immunization, and any other disease condition that may be
determined by the FDA from time to time.
b. Paediatric formulation.
c. Ministry of Health tender purposes only.
d. Post approval variation.
e. Renewal of registration.
f. WHO prequalified products
During evaluation, additional data and/or samples may be requested through a deferral
letter. Once a query has been issued to the applicant, the evaluation process stops until
FDA receives a written response to the query. Further processing of the application may
only be made if responses to queries issued in the same deferral letter contains all
outstanding information requested in one submission.
Failure to comply with this condition or if the queries have been reissued for a third time
and the applicant provides unsatisfactory responses, the application will be rejected. In
the event the responses to the queries are not submitted within twelve (12) months from
the date they were issued, it will be considered that the applicant has withdrawn the
application. Thereafter, registration of the product may only be considered upon
submission of a new application.
Page 88 of 88
FDA/DRI/DER/GL-RAD/2013/06
facility or use other means to verify whether the facility complies with cGMP Regulations
and/or guideline before a product is registered. No product shall be registered unless the
facility complies with cGMP.
The report of the cGMP inspection will form part of the registration process.
Label review, dossier evaluation, laboratory analysis and GMP status reports will be
presented before the Drug Registration Committee for review and making final decisions
for granting or rejecting registration of the product.
In the event, that there are safety, quality or efficacy issues to be resolved as per the
decision of the Committee, the application may be deferred pending resolution of the
issues. Should the applicant fail to provide the required data within twelve months, it will
be considered that the applicant has withdrawn the application. Thereafter, registration of
the product may only be considered upon submission of a new application.
The FDA will register the product in the event that data on safety, quality and efficacy is
deemed satisfactory by the Drug Registration Committee in accordance with Section 118
of the Public Health Act, 2012, Act 851. A certificate of registration shall be issued as per
Section 118, subsection 7(c) of the Public Health Act, 2012, Act 851. The registration shall
be valid for a period of three (3) years. In the event that the FDA suspends or cancels the
registration validity, the FDA will give reasons in writing.
The FDA makes the final decision on an application made under section 118 of Public
Health Act 2012 Act 851 for the registration and re-registration.
The FDA during the registration process can reject an application when it is not part of a
treatment regimen for a Programme under the Ministry of Health, for Safety or Quality
reasons.
An Applicant may appeal a decision made by the FDA as indicated in Section 119
subsection 6 of the Public Health Act 2012, Act 851 within sixty days after the date of the
notification of rejection.
The appeal representation shall be made in writing to the Authority addressed to:
Page 89 of 88
FDA/DRI/DER/GL-RAD/2013/06
Cantonments
Accra
On receipt of the intention to appeal, the FDA will subject the notice of appeal to its internal
appeal processes.
Where the FDA is satisfied with the representations submitted, the FDA may approve the
registration of the medicinal product or if the FDA is still not satisfied, it shall reject the
application. Decisions on outcome of appeals shall be communicated to applicants within
60 working days after receipt of an appeal.
11. TIMELINES
The following timelines will be implemented by the FDA in processing applications for
registration of products.
A new application will be processed within 6 months of receipt of the application. The
applicant will be required to provide any requested additional data within 12 months. In
case additional time is required, a formal request must be submitted to the FDA.
Page 90 of 88