Saturday, March 31, 2012

Q8 Pharmaceutical Development


 Q8 Pharmaceutical Development

1. Introduction

1.1 Objective of the Guideline

This guideline describes the suggested contents for the 3.2.P.2 Pharmaceutical Development section of a regulatory submission in the ICH M4 Common Technical Document (CTD) format. 

The Pharmaceutical Development section provides an opportunity to present the knowledge gained through the application of scientific approaches, and risk management*, to the development of a product and its manufacturing process. It is first produced for the original marketing application and can be updated to support new knowledge gained over the lifecycle* of a product.  The guideline also indicates areas where the provision of greater understanding of pharmaceutical and manufacturing sciences can create a basis for flexible regulatory approaches. The Pharmaceutical Development section is intended to provide a more comprehensive understanding of the product and manufacturing process for reviewers and inspectors.

1.2 Background

During the July 2003 ICH meeting in Brussels, agreement was reached on a common vision and approach for developing an international plan for a harmonized pharmaceutical quality system that would be applicable across the life cycle of a product. This plan emphasizes an integrated approach to review (assessment) and inspection based on scientific risk management. Several actions were outlined to implement this vision. An expert-working group (EWG) was established to develop guidance for pharmaceutical development, which will cover the lifecycle of a product.

1.3 Scope

This guideline is intended to provide guidance on the contents of Section 3.2.P.2 (Pharmaceutical Development) for drug products as defined in the scope of Module 3 of the Common Technical Document (ICH topic M4). The guideline does not apply to contents of submissions for drug products during the clinical research stages of drug development. However the principles in this guideline are important to consider during these stages.  This guideline might also be appropriate for other types of products.  To determine the applicability of this guideline for a particular type of product, applicants should consult with the appropriate regulatory authorities.

2. Pharmaceutical Development

The aim of pharmaceutical development is to design a quality* product and the manufacturing process to deliver the product in a reproducible manner. The information and knowledge gained from pharmaceutical development studies provide scientific understanding to support the establishing of specifications and manufacturing controls. 

Information from pharmaceutical development studies is a basis for risk management. It is important to recognize that quality cannot be tested into products; i.e., quality should be built in by design. Changes in formulation and manufacturing processes during development should be looked upon as opportunities to gain additional knowledge and further support establishment of the design space. Inclusion of knowledge gained from experiments giving negative results also can be useful in supporting the selected product and its manufacturing process. 

The Pharmaceutical Development section should describe the knowledge that establishes that the type of dosage form selected and the formulation proposed are satisfactory for the purpose specified in the application.  This section should include sufficient information in each part to provide an understanding of the development of the drug product and its manufacturing process. Summary tables and graphs are encouraged.

At a minimum, those aspects of drug substances, excipients, and manufacturing processes that are critical and that present a significant risk* to product quality, and therefore should be monitored or otherwise controlled, should be identified and discussed.  These critical formulation attributes and process parameters are generally identified through an assessment of the extent to which their variation can have impact on the quality of the drug product.

In addition, the applicant can choose to conduct other pharmaceutical development studies that can lead to an enhanced knowledge of product performance over a wider range of material attributes, processing options and process parameters. Inclusion of this additional information in this section provides an opportunity to demonstrate a higher degree of understanding of manufacturing processes and process controls.  This scientific understanding establishes the design space. In these situations, opportunities exist to develop more flexible regulatory approaches, for example, to facilitate:

·         risk based regulatory decisions (reviews and inspections);
·         manufacturing process improvements, within the approved design space described in the    dossier, without further regulatory review;
·          “real time” quality control, leading  to a reduction of end-product release testing.

To realise this flexibility, the applicant should demonstrate an enhanced knowledge of product performance over a range of material attributes (e.g. particle size distribution, moisture content, flow properties), processing options and process parameters. This knowledge can be gained by, for example, application of formal experimental designs* or PAT*. Appropriate use of risk management principles can be helpful in prioritising the additional pharmaceutical development studies to collect such knowledge.

The design and conduct of the pharmaceutical development studies should be consistent with their intended scientific purpose and the stage of the development of the product.  It should be recognized that the level of knowledge gained, and not the volume of data, provides the basis for science-based submissions and their regulatory evaluation.

2.1 Components of the Drug Product

2.1.1 Drug Substance

The physicochemical and biological properties of the drug substance that can influence the performance of the drug product and its manufacturability, or were specifically designed into the drug substance (e.g., crystal engineering), should be identified and discussed.  Examples of physicochemical and biological properties that might need to be examined include solubility, water content, particle size, crystal properties, biological activity, and permeability.  These properties could be inter-related and might need to be considered in combination.  Some of these properties can change with time and might be supplier dependent.


To evaluate the potential effect of drug substance physicochemical properties on the performance of the drug product, studies on drug product might be warranted.  For example, the ICH Q6A Specifications: Test Procedures and Acceptance Criteria for New Drug Substances and New Drug Products: Chemical Substances describes some of the circumstances in which drug product studies are recommended (e.g., Decision Tree #3 and #4 (Part 2)). The knowledge gained from the studies investigating the potential effect of drug substance properties on drug product performance can be used, as appropriate, to justify elements of the drug substance specification (3.2.S.4.5).

The compatibility of the drug substance with excipients listed in 3.2.P.1 should be discussed.  For products that contain more than one drug substance, the compatibility of the drug substances with each other should also be discussed.

2.1.2 Excipients

The excipients chosen, their concentration, and the characteristics that can influence the drug product performance (e.g., stability, bioavailability) or manufacturability should be discussed relative to the respective function of each excipient. Compatibility of excipients with other excipients, where relevant (for example combination of preservatives in a dual preservative system), should be established. The ability of excipients (e.g., antioxidants, penetration enhancers, disintegrants, release controlling agents) to provide their intended functionality, and to perform throughout the intended drug product shelf life, should also be demonstrated. The information on excipient performance can be used, as appropriate, to justify the choice and quality attributes of the excipient, and to support the justification of the drug product specification (3.2.P.5.6).

Information to support the safety of excipients, when appropriate, should be cross-referenced (3.2.P.4.6).

 2.2 Drug Product

 2.2.1 Formulation Development

 A summary should be provided describing the development of the formulation, including identification of those attributes that are critical to the quality of the drug product, taking into consideration intended usage and route of administration.

The summary should highlight the evolution of the formulation design from initial concept up to the final design. This summary should also take into consideration the choice of drug product components, (e.g. the properties of the drug substance, excipients, container closure system, any relevant dosing device) the manufacturing process, and, if appropriate, experiences gained from the development of similar drug product(s).

Information from formal experimental designs can be useful in identifying critical or interacting variables that might be important to ensure the quality of the drug product.  Any excipient ranges included in the batch formula (3.2.P.3.2) should be justified in this section of the application: this justification can often be based on the experience gained during the development of the formulation and manufacturing process. 

A summary of all formulations used in clinical safety and efficacy, bioavailability, or bioequivalence studies should be provided. Any changes between the proposed commercial formulation and those formulations used in pivotal clinical batches and primary stability batches should be clearly described and the rationale for the changes provided.

Information from comparative in vitro studies (e.g., dissolution), or comparative in vivo studies (e.g., bioequivalence), that links clinical formulations to the proposed commercial formulation described in 3.2.P.1 should be summarized and a cross-reference to the studies (with study numbers) should be provided. Where attempts have been made to establish an in vitro/in vivo correlation the results of those studies, and a cross-reference to the studies (with study numbers), should be provided in this section. A successful correlation can assist in the selection of appropriate dissolution acceptance criteria, and can potentially reduce the need for further bioequivalence studies following changes to the product or its manufacturing process.

Any special design features of the drug product (e.g., tablet score line, overfill, anti-counterfeiting measure) should be identified and a rationale provided for their use. Information to support the appropriateness of such features should be provided.

2.2.2 Overages

The use of overages of drug substance(s) in drug products is discouraged.

An overage is a fixed amount of the drug substance added to the formulation in excess of the label claim. Any overages in the manufacture of the drug product, whether they appear in the final formulated product or not, should be justified considering the safety and efficacy of the product. Information should be provided on the 1) amount of overage, 2) reason for the overage, (e.g., to compensate for expected and documented manufacturing losses), and 3) justification for the amount of overage.  The overage should be included in the amount of drug substance listed in the representative batch formula (3.2.P.3.2).

2.2.3 Physicochemical and Biological Properties

 The physicochemical and biological properties relevant to the performance or manufacturability of the drug product should be identified and discussed. These could include formulation attributes such as pH, osmolarity, ionic strength, lipophilicity, dissolution, redispersion, reconstitution, particle size distribution, particle shape, aggregation, polymorphism, rheological properties, globule size of emulsions, biological activity or potency, and/or immunological activity. Physiological implications of formulation attributes such as pH should also be addressed.  The discussion should cross-reference any relevant stability data in 3.2.P.8.3.

A summary of the development studies that were carried out to investigate the potential impacts of the physicochemical and biological properties of the drug product and the appropriateness of the drug product acceptance criteria should be reported in this section of the application (3.2.P.2.2.3). These studies could include, for example, the development of a dissolution or drug release test, or the development of a test for respirable fraction of an inhaled product, where appropriate. Physiological implications of drug substance and formulation attributes should be addressed.  For example, information could be provided from studies to investigate whether acceptance criteria for polymorphism should be included in the drug product specification. Similarly, information to support the robustness of the formulation and manufacturing process with respect to the selection of dissolution versus disintegration testing, or other means to assure drug release, could be provided in this section. See also ICH Q6A Specifications: Test Procedures And Acceptance Criteria For New Drug Substances And New Drug Products: Chemical Substances; Decision Tree #4 (Part 3) and Decision Tree #7 (Part 1).

2.3 Manufacturing Process Development

The selection, the control, and any optimisation of the manufacturing process described in 3.2.P.3.3 (i.e., intended for commercial production batches) should be explained. It is important to consider the critical formulation attributes, together with the available manufacturing process options (e.g., dry granulation vs. wet granulation, terminal sterilisation vs. aseptic processing), in order to address the selection of the manufacturing process and confirm the appropriateness of the components (i.e., excipients). Appropriateness of the equipment used for the intended products should be discussed. Process development studies should provide the basis for process optimisation, process validation and process control requirements. Where appropriate, such studies should address microbiological as well as physical and chemical attributes. The knowledge gained from process development studies can be used, as appropriate, to justify the drug product specification (3.2.P.5.6).  An assessment of the ability of the process to reliably produce a product of the intended quality (e.g., the performance of the manufacturing process under different operating conditions, at different scales, or with different equipment) should be provided.

The manufacturing process development programme should identify the critical process parameters that should be monitored or controlled (e.g., granulation end point) to ensure that the product is of the desired quality.  

For those products intended to be sterile an appropriate method of sterilization for the drug product and primary packaging material should be chosen and the choice justified.

Significant differences between the manufacturing processes used to produce the clinical safety and efficacy, bioavailability, bioequivalence, or primary stability batches and the process described in 3.2.P.3.3 should be discussed. The discussion should summarise the influence of the differences on the performance and manufacturability of the product.  The information should be presented in a way that facilitates comparison of the processes and the corresponding batch analyses information (3.2.P.5.4). The information should include, for example,  (1) the identity (e.g., batch number) and use of the batches produced using the specified equipment (e.g., bioequivalence study batch number), (2) the manufacturing site, (3) the batch size, and (4) any significant equipment differences (e.g., different design, operating principle, size).

In order to provide flexibility for future process optimisation, when describing the development of the manufacturing process, it is useful to describe any measurement systems that allow monitoring of critical attributes or process end-points. Collection of process monitoring data during the development of the manufacturing process can provide useful information to enhance process understanding. The process controls that provide process adjustment capabilities to ensure control of all critical attributes should be described.  These provide a means for a risk control strategy.

An assessment of process robustness can be useful in risk assessment and risk reduction*, to support future manufacturing and process optimisation, especially in conjunction with the use of structured risk management tools.

2.4 Container Closure System

The choice and rationale for selection of the container closure system(s) for the commercial product(s) (described in 3.2.P.7) should be discussed. Consideration should be given to the intended use of the drug product and the suitability of the container closure system for storage and transportation (shipping), including the storage and shipping container for bulk drug product, where appropriate.

The choice of materials for primary packaging should be justified. The discussion should describe studies performed to demonstrate the integrity of the container and closure. A possible interaction between product and container(s) or label should be considered. This applies also to admixture or dilution of products prior to administration e.g. product added to large volume infusion containers.

The choice of primary packaging materials should consider, e.g., choice of materials, protection from moisture and light, compatibility of the materials of construction with the dosage form (including sorption to container and leaching), and safety of materials of construction.

If a dosing device is used (e.g., dropper pipette, pen injection device), it is important to demonstrate that a reproducible and accurate dose of the product is delivered under testing conditions which, as far as possible, simulate the use of the product.

2.5 Microbiological Attributes

Where appropriate, the microbiological attributes of the drug product should be discussed in this section (3.2.P.2.5). The discussion should include, for example:

·         The rationale for performing or not performing microbial limits testing for nonsterile drug products, (e.g., Decision Tree #8 in ICH Q6A Specifications: Test Procedures and Acceptance Criteria for New Drug Substances and New Drug Products: Chemical Substances)

·         The selection and effectiveness of preservative systems in products containing antimicrobial preservative or the antimicrobial effectiveness of products that are inherently antimicrobial

·         For sterile products, the integrity of the container closure system as it relates to preventing microbial contamination.

Although chemical testing for preservative content is the attribute normally included in the drug product specification, antimicrobial preservative effectiveness should be demonstrated during development. The lowest specified concentration of antimicrobial preservative should be demonstrated to be effective in controlling microorganisms by using an antimicrobial preservative effectiveness test.

2.6 Compatibility

The compatibility of the drug product with reconstitution diluent(s) or dosage devices (e.g., precipitation of drug substance in solution, sorption on injection vessels, stability) should be addressed to provide appropriate and supportive information for the labelling. This information should cover the recommended in-use shelf life, at the recommended storage temperature and at the likely extremes of concentration. Where the label recommends dilution or mixing of solid dose forms (for example with drinks) prior to administration, appropriate compatibility studies should be described.

3. Glossary

Design Space: the design space is the established range of process parameters that has been demonstrated to provide assurance of quality. In some cases design space can also be applicable to formulation attributes. Working within the design space is not generally considered as a change of the approved ranges for process parameters and formulation attributes. Movement out of the design space is considered to be a change and would normally initiate a regulatory post approval change process.

Formal Experimental Design: a structured, organized method for determining the relationship between factors (Xs) affecting a process and the output of that process (Y). Also known as “Design of Experiments”.

Lifecycle: all phases in the life of a product from the initial development through pre- and post-approval until the product’s discontinuation.

PAT:  Process Analytical Technologies - a system for designing, analyzing, and controlling manufacturing through timely measurements (i.e., during processing) of critical quality and performance attributes of raw and in-process materials and processes with the goal of assuring final product quality. 

Quality: degree to which a set of inherent properties of a product, system or process fulfils requirements

Risk: the combination of the probability of occurrence of harm and the severity of that harm (from ISO/IEC Guide 51)

Risk Management: systematic application of quality management policies, procedures, and practices to the tasks of assessing, controlling and communicating risk.

Risk Reduction: actions taken to lessen the probability of occurrence of harm and the severity of that harm

 

Tuesday, March 27, 2012

IND APPLICATION TEMPLATE: CMC section

  IND APPLICATION TEMPLATE:
 CHEMISTRY, MANUFACTURING AND CONTROL INFORMATION


D.   Chemistry, Manufacturing and Control Information 1.   Introduction:
 1.1  Potential human risk(s) Indicate if 1) the chemistry of either the investigational drug substance (i.e., the active ingredient) or the investigational drug product, or 2) the manufacturing process for either the investigational drug substance or investigational drug product, presents any signals of potential human risk.  If so, discuss the signals of potential risks; to include a description of the steps proposed to monitor for such risk(s), or the reason(s) why the signals should be dismissed. 1.2  Chemistry and manufacturing: Pre-clinical versus clinical studies Describe any chemistry or manufacturing differences between the investigational drug product proposed for clinical research study use versus the investigational drug product that was used in the pre-clinical (i.e, animal) toxicology studies that formed the basis that it was safe to proceed with the clinical research study.  If there are no differences in the investigational drug product, this should be specifically stated.  If there are differences in the investigational drug product, discuss how these differences may affect the safety profile of the investigational drug product. 2.  Investigational Drug Substance: 2.1   Description of investigational drug substance
 Provide a description of the investigational drug substance (i.e., the active drug substance); including its physical, chemical (e.g., chemical name and/or structure), or biological characteristics. 2.2  Manufacturer of the investigational drug substance Provide the name and full address of the manufacturer of the investigational drug substance. Specify if the manufacture of the investigational drug substance was or was not (will or will not be) performed in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards at 21 CFR Part 211.  If so, include with the manufacturer’s address, its FDA Drug Establishment Registration Number. 2.3   Method of preparation of the investigational  drug substance
 For an investigational drug substance manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Provide a detailed flow diagram of the process for manufacture of the investigational drug substance; and incorporate a list of the components (i.e., reagents, solvents, excipients, catalysts, etc.) used in the manufacturing process.  (Note: Step-by-step procedures for, and a listing of the specifications and quantities of the components used in, the manufacture of the investigational drug substance are ordinarily not required to be included in the IND application for the initial stage of drug development; as it is assumed that the manufacturer will establish such procedures and specifications consistent with cGMP compliance.) Alternately, if the cGMP manufacturer of the investigational drug substance has submitted, or will submit, a respective Drug Master File to the FDA (or if the manufacturer has in place a FDA-accepted IND for the investigational drug substance/product), incorporate (i.e., into a referenced appendix of the IND application) a letter or written notification from the manufacturer authorizing the FDA to access its Drug Master File (or IND) in support of the chemistry, manufacturing and control information required to be submitted under this IND application. If the investigational drug product is approved by the FDA for commercial marketing, specify such and incorporate (i.e., into a referenced appendix of the IND application) the corresponding FDA-approved product labeling (i.e., product insert). For an investigational drug substance not  manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Provide a flow diagram of the process for manufacture of the investigational drug substance. Incorporate Drug Master Card(s) listing the components (i.e., reagents, solvents, excipients, catalysts, etc.) used currently in the manufacture of the investigational drug substance; to include, for each listed agent, its manufacturer or source, product specifications, and quantity used in the manufacture.  (See Example Drug Master Card.1)
 (Note: the Drug Master Card(s) may provide for alternative manufacturers of inactive components used in the manufacture of the investigational drug substance.)(Note: for novel components [i.e., non-commercially available components manufactured specifically for use in the preparation of the investigational drug substance] additional manufacturing information may need to be provided.(See Example Drug Master Card.2)
 Incorporate Drug Master Card(s) listing the step-by-step procedures used currently for the manufacture of the investigational drug substance.  (See Example Drug Master Card.1) 2.4   Acceptable limits and analytical methods use to assure the identity, strength, quality and purity of the investigational drug substance
 For an investigational drug substance manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Specify the acceptable limits of identity, strength, quality and purity that will form the basis for the release/acceptance of the investigational drug substance for its use in the preparation of the investigational drug product. Provide a brief description of the various test (i.e., analytical) methods that are (will be) used to establish that the investigational drug substance meets the defined, acceptable limits of identity, strength, quality and purity.  (Note: Validation procedures/data for the test (i.e., analytical) methods are ordinarily not required to be included in the IND application for the initial stage of drug development; as it is assumed that the manufacturer will perform such validation procedures consistent with cGMP compliance.) Provide copies of analytical data verifying that the manufacturing process results in an investigational drug substance that meets or exceeds the specified, acceptable limits of identity, strength, quality and purity. Submit a copy of the “certificate of analysis” that will form the basis for release/acceptance of the investigational drug substance for its use in the preparation of the investigational drug product.  Alternately, if the cGMP manufacturer of the investigational drug substance has submitted, or will submit, a respective Drug Master File to the FDA (or if the manufacturer has in place a FDA-accepted IND for the investigational drug substance/product), incorporate (i.e., into a referenced appendix of the IND application) a letter or written notification from the manufacturer authorizing the FDA to access its Drug Master File (or IND) in support of the chemistry, manufacturing, and control information required to be submitted under this IND application. Submit a copy of the “certificate of analysis” that will form the basis for release/acceptance of the investigational drug substance for its use in the preparation of the investigational drug product. Alternately, if the investigational drug product is approved by the FDA for commercial marketing, specify such and incorporate (i.e., into a referenced appendix of the IND application) the corresponding FDA-approved product labeling.  For an investigational drug substance not  manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Specify the acceptable limits of identity, strength, quality and purity that will form the basis for the release/acceptance of the investigational drug substance for its use in the preparation of the investigational drug product. Provide a description of the various test (i.e., analytical) methods that will be used to establish that the investigational drug substance meets the defined, acceptable limits of identity, strength, quality and purity.  Summarize the procedures that will be used to verify the correct operation of the respective analytical equipment. Provide copies of analytical data verifying that the manufacturing process results in an investigational drug substance that meets or exceeds the specified, acceptable limits of identity, strength, quality and purity. Submit a copy of the Drug Master Card(s) outlining the acceptable limits of identity, strength, quality and purity and corresponding test (i.e., analytical) methods that will form the basis for release/acceptance of the investigational drug substance for its use in the preparation of the investigational drug product.  (See Example Drug Master Card.1) 2.5   Information to support the stability of the investigational drug substance
     For an investigational drug substance manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Specify the expiry period assigned currently to the investigational drug substance.  (Note: If stability of the investigational drug substance is continuing to be studied for the purpose of extending the current assigned expiry period, indicate such and state that the assigned expiry period may be revised based on the results of the ongoing stability study.) Provide a brief description of the stability study and the test (i.e., analytical) methods that were used to monitor the stability of the investigational drug substance.  (Note: The stability study should involve storage of the investigational drug substance in the final container/closure system specified in its manufacturing process and under environmental conditions specified in the product labeling.  The extent of the test methods used to monitor the stability of the investigational drug substance should be sufficient to verify that the specified, acceptable limits of identity, strength, quality and purity are retained throughout the assigned expiry period.) (Note: There is no need to submit the actual stability study protocol.) Summarize, in tabular or graphical format, the data supporting the expiry period assigned currently to the investigational drug substance.  (Note: There is no need to submit detailed stability data.) Alternately, if the cGMP manufacturer of the investigational drug substance has submitted, or will submit, a respective Drug Master File to the FDA (or if the manufacturer has in place a FDA-accepted IND for the investigational drug substance/product), incorporate (i.e., into a referenced appendix of the IND application) a letter or written notification from the manufacturer authorizing the FDA to access its Drug Master File (or IND or NDA) in support of the chemistry, manufacturing, and control information required to be submitted under this IND application. Alternately, if the investigational drug product is approved by the FDA for commercial marketing, specify such and incorporate (i.e., into a referenced appendix of the IND application) the corresponding FDA-approved product labeling (i.e., product insert). For an investigational drug substance not manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Specify the expiry period assigned currently to the investigational drug substance.  (Note: If stability of the investigational drug substance is continuing to be studied for the purpose of extending the current assigned expiry period, indicate such and state that the assigned expiry period may be revised based on the results of the ongoing stability study.) Provide a brief description of the stability study and the test (i.e., analytical) methods that were used to monitor the stability of the investigational drug substance.  (Note: The stability study should involve storage of the investigational drug substance in the final container/closure system specified in its manufacturing process and under environmental conditions specified in the product labeling.  The extent of the test methods used to monitor the stability of the investigational drug substance should be sufficient to verify that the specified, acceptable limits of identity, strength, quality and purity are retained throughout the assigned expiry period.) (Note: There is no need to submit the actual stability study protocol.) Summarize, in tabular or graphical format, the data supporting the expiry period assigned currently to the drug substance.  (Note: There is no need to submit detailed stability data.) 3.   Investigational drug product:
         3.1  Description of the investigational drug product
 Provide a description of the of the investigational drug product (i.e., the final formulation of the investigational drug that will be used in clinical research studies) 3.2  Manufacturer of the investigational drug product Provide the name and full address of the manufacturer of the investigational drug product. Specify if the manufacture of the investigational drug product was or was not (will or will not be) performed in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards.  If so, include with the manufacturer’s address, its FDA Drug Establishment Registration Number. 3.3 Components used in the manufacture of the investigational drug product For an investigational drug product manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Provide a list of all components used in the manufacture of the investigational drug product; including both those components that are intended to appear in the final product and those components that are not intended to appear in the final product (i.e., but which were used in the manufacturing process).  For each of the inactive components, include a reference to its quality specification (e.g., USP, NF, ACS) (Note: this list may include reasonable alternatives for inactive components used in the manufacture of the investigational drug product.) Alternately, if the cGMP manufacturer of the investigational drug product has submitted, or will submit, a respective Drug Master File to the FDA (or if the manufacturer has in place a FDA-accepted IND for the investigational drug substance/product), incorporate (i.e., into a referenced appendix of the IND application) a letter or written notification from the manufacturer authorizing the FDA to access its Drug Master File (or IND) in support of the chemistry, manufacturing and control information required to be submitted under this IND application. Alternately, if the investigational drug product is approved by the FDA for commercial marketing, specify such and incorporate (i.e., into a referenced appendix of the IND application) the corresponding FDA-approved product labeling (i.e., product insert). For an investigational drug product not manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Incorporate Drug Master Card(s) listing all of the components (including the investigational drug substance) used currently in the manufacture of the investigational drug product; to include, for each listed component, its manufacturer, quality specification (e.g., USP, NF, ACS), and quantity used in the preparation.  (See Example Drug Master Card.1)
 (Note: the Drug Master Card(s) may provide for alternative manufacturers of inactive components used in the manufacture of the investigational drug product.)(Note: for novel components [i.e., non-commercially available components manufactured specifically for use in the preparation of the investigational drug product] additional manufacturing information may need to be provided.(See Example Drug Master Card.2)
 Describe the procedures (e.g., inspection of the “certificate of analysis” provided by the manufacturer, defined identity test(s)) that will be used to verify that the investigational drug substance meets acceptable standards of identity, strength, quality and purity for use in the manufacture of the investigational drug product. Describe the procedures (e.g., inspection of the product label and/or the “certificate of analysis” provided by the manufacturer) that will be used to verify that the inactive components used in the manufacture of the investigational drug product meet or exceed defined quality specifications. 3.4  Quantitative composition of the investigational drug product For an investigational drug product manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Summarize the quantitative composition of the investigational drug product.  I.e., address the quantity of the investigational drug substance and the quantities of components that are intended to appear in the investigational drug product.  (Note: For components used in the manufacture of the investigational drug product, but which are not intended to appear in the final product, describe the manufacturing processes or procedures that result in the elimination of these components.) Alternately, if the cGMP manufacturer of the investigational drug product has submitted, or will submit, a respective Drug Master File to the FDA (or if the manufacturer has in place a FDA-accepted IND for the investigational drug product), incorporate a letter or written notification from the manufacturer authorizing the FDA to access its Drug Master File (or IND) in support of the chemistry, manufacturing and control information required to be submitted under this IND application. Alternately, if the investigational drug product is approved by the FDA for commercial marketing, specify such and incorporate (i.e., into a referenced appendix of the IND application) the corresponding FDA-approved product labeling (i.e., product insert). For an investigational drug product not manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Summarize the quantitative composition of the investigational drug product.  I.e., address the quantity of the investigational drug substance and the quantities of components that are intended to appear in the investigational drug product. (Note:  For components used in the manufacture of the investigational drug product, but which are not intended to appear in the final product, describe the manufacturing processes or procedures that result in the elimination of these components.) 3.5  Method of manufacture and final packaging of the investigational drug product For an investigational drug product manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Provide a detailed flow diagram of the process for manufacture and packaging of the investigational drug product.  (Note: For sterile investigational drug products, the flow diagram should address the sterilization process.) (Note: Step-by-step procedures used for the manufacture and packaging of the investigational drug product are ordinarily not required to be included in the IND application for the initial stage of drug development; as it is assumed that the manufacturer will establish such procedures consistent with cGMP compliance.) Alternately, if the cGMP manufacturer of the investigational drug product has submitted, or will submit, a respective Drug Master File to the FDA (or if the manufacturer has in place a FDA-accepted for the investigational drug product), incorporate a letter or written notification from the manufacturer authorizing the FDA to access its Drug Master File (or IND) in support of the chemistry, manufacturing and control information required to be submitted under this IND application. Alternately, if the investigational drug product is approved by the FDA for commercial marketing, specify such and incorporate (i.e., into a referenced appendix of the IND application) the corresponding FDA-approved product labeling (i.e., product insert). For an investigational drug product not manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Incorporate Drug Master Card(s) listing the step-by-step procedures used currently for the preparation and packaging of the investigational drug product; to include, for sterile investigational drug  products, the procedures for sterilization of the product.  (See Example Drug Master Card.1)
 3.6  Acceptable limits and analytical methods use to assure the identity, strength, quality and purity of the investigational drug product For an investigational drug product manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Specify the acceptable limits of identity, strength, quality and purity that will form the basis for the release of the investigational drug product for human use. Provide a brief description of the various test (i.e., analytical) methods that will be used to establish that the investigational drug product meets the defined, acceptable limits of identity, strength, quality and purity. (Note: The test [i.e., analytical] methods that should be submitted will vary according the dosage form of the investigational drug product.  For example, for sterile products, the sterility and bacterial endotoxin (i.e., pyrogen) test methods should be submitted.) (Note: Validation procedures/data for the test [i.e., analytical] methods are ordinarily not required to be included in the IND application for the initial stage of drug development; as it is assumed that the manufacturer will perform such validation procedures consistent with cGMP compliance.) Provide copies of analytical data verifying that the manufacturing process results in an investigational drug product that meets or exceeds the specified, acceptable limits of identity, strength, quality and purity. Alternately, if the cGMP manufacturer of the investigational drug product has submitted, or will submit, a respective Drug Master File to the FDA (or if the manufacturer has in place a FDA-accepted IND for the investigational drug product), incorporate (i.e., into a referenced appendix of the IND application) a letter or written notification from the manufacturer authorizing the FDA to access its Drug Master File (or IND) in support of the chemistry, manufacturing, and control information required to be submitted under this IND application. Alternately, if the investigational drug product is approved by the FDA for commercial marketing, specify such and incorporate (i.e., into a referenced appendix of the IND application) the corresponding FDA-approved product labeling. Submit a copy of the “certificate of analysis” that will form the basis for release/acceptance of the investigational drug product for use in the clinical research study. For an investigational drug product not  manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Specify the acceptable limits of identity, strength, quality and purity that will form the basis for the release of the investigational drug product for use in the clinical research study. Provide a description of the various test (i.e., analytical) methods that will be used to establish that the investigational drug product meets the defined, acceptable limits of identity, strength, quality and purity.  Summarize the procedures that will be used to verify the correct operation of the respective analytical equipment. (Note: The test (i.e., analytical) methods that should be submitted will vary according the dosage form of the investigational drug product.  For example, for sterile products, the sterility and bacterial endotoxin (i.e., pyrogen) test methods should be submitted.) Provide copies of analytical data verifying that the manufacturing process results in an investigational drug product that meets or exceeds the specified, acceptable limits of identity, strength, quality and purity. Submit a copy of the Drug Master Card(s) outlining the acceptable limits of identity, strength, quality and purity and corresponding test (i.e., analytical) methods that will form the basis for release of the investigational drug product for human use.  (See Example Drug Master Card.1)
 3.7  Information to support the stability of the investigational drug product For an investigational drug substance manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Specify the expiry period assigned currently to the investigational drug product.  (Note: If stability of the investigational drug substance is continuing to be studied for the purpose of extending the current assigned expiry period, indicate such and state that the assigned expiry period may be revised based on the results of the ongoing stability study.) Provide a brief description of the stability study and the test (i.e., analytical) methods that were used to monitor the stability of the investigational drug product.  (Note: The stability study should involve storage of the investigational drug product in the final container/closure system specified in its manufacturing process and under environmental conditions specified in the product labeling.  The extent of the test methods used to monitor the stability of the investigational drug product should be sufficient to verify that the specified, acceptable limits of identity, strength, quality and purity are retained throughout the assigned expiry period.) (Note: There is no need to submit the actual stability study protocol.) Summarize, in tabular or graphical format, the data supporting the expiry period assigned currently to the investigational drug product.  (Note: There is no need to submit detailed stability data.) Alternately, if the cGMP manufacturer of the investigational drug product has submitted, or will submit, a respective Drug Master File to the FDA (or if the manufacturer has in place a FDA-accepted IND for the investigational drug substance/product), incorporate (i.e., into a referenced appendix of the IND application) a letter or written notification from the manufacturer authorizing the FDA to access its Drug Master File (or IND or NDA) in support of the chemistry, manufacturing, and control information required to be submitted under this IND application. Alternately, if the investigational drug product is approved by the FDA for commercial marketing, specify such and incorporate (i.e., into a referenced appendix of the IND application) the corresponding FDA-approved product labeling (i.e., product insert). For an investigational drug substance not manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Specify the expiry period assigned currently to the investigational drug product.  (Note: If stability of the investigational drug product is continuing to be studied for the purpose of extending the current assigned expiry period, indicate such and state that the assigned expiry period may be revised based on the results of the ongoing stability study.) Provide a brief description of the stability study and the test (i.e., analytical) methods that were used to monitor the stability of the investigational drug product.  (Note: The stability study should involve storage of the investigational drug product  in the final container/closure system specified in its manufacturing process and under environmental conditions specified in the product labeling.  The extent of the test methods used to monitor the stability of the investigational drug product should be sufficient to verify that the specified, acceptable limits of identity, strength, quality and purity are retained throughout the assigned expiry period.) (Note: There is no need to submit the actual stability study protocol.) Summarize, in tabular or graphical format, the data supporting the expiry period assigned currently to the investigational  drug product.
 (Note: There is no need to submit detailed stability data.) 4.  Investigational drug product labels and labeling For an investigational drug product manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Submit a mock-up or printed representation of the label that will be attached to the container of the investigational drug product. (Note: the product container label should contain, at a minimum, the name and address of the manufacturer; the identify of the investigational drug substance; the quantity of the investigational drug substance per unit (e.g.,capsule or tablet) or volume; the assigned batch or lot number; the specifications of “sterile” and “pyrogen-free” (if applicable); the conditions for appropriate storage of the product; the expiration date; and the statement, “Caution: New Drug – Limited by Federal (or United States) law to investigational use.”) Submit, if applicable, any additional labeling materials that will be provided to the investigators involved in clinical studies of the investigational drug product. (Note: The labeling of the investigational drug product shall not bear any statement that is false or misleading and shall not represent that the product is safe or effective for the purposes for which it is being investigated.) Alternately, if the cGMP manufacturer of the investigational drug product has submitted, or will submit, a respective Drug Master File to the FDA (or if the manufacturer has in place a FDA-accepted IND for the investigational drug product), the product container label information may be addressed by incorporating (i.e., into a referenced appendix of the IND application) a letter or written notification from the manufacturer authorizing the FDA to access its Drug Master File (or IND or NDA) in support of chemistry, manufacturing and control information required to be submitted under this IND application.) Alternately, if the investigational drug product is approved by the FDA for commercial marketing, specify such and incorporate (i.e., into a referenced appendix of the IND application) the corresponding FDA-approved product labeling (i.e., product insert). For an investigational drug product not manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Submit a mock-up or printed representation of the label that will be attached to the container of the investigational drug product. (Note: the product container label should contain, at a minimum, the name and address of the manufacturer; the identity of the investigational drug substance; the quantity of the investigational drug substance per unit (e.g.,capsule or tablet) or volume; the assigned batch or lot number; the specifications of “sterile” and “pyrogen-free” (if applicable); the conditions for appropriate storage of the product; the expiration date; and the statement, “Caution: New Drug – Limited by Federal (or United States) law to investigational use.”) Submit, if applicable, any additional labeling materials that will be provided to the investigators involved in clinical studies of the investigational drug product. (Note: The labeling of the investigational drug product shall not bear any statement that is false or misleading and shall not represent that the product is safe or effective for the purposes for which it is being investigated.) 5.  Placebo: composition, manufacture, and control information (Incorporate only if applicable)
 Provide a description of the placebo that will be used in the clinical research study (studies) including its physical, chemical (e.g., chemical identity and/or structure), or biological characteristics. Identify the manufacturer of the placebo product; to include the manufacturer’s full address.   Specify if the manufacture of the placebo product was or was not (will or will not be) performed in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards.  If so, include with the manufacturer’s address, its FDA Drug Establishment Registration Number. For a placebo product manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Provide a flow diagram of the process for manufacture and packaging of the placebo product; and incorporate a list of all of the components used in the manufacturing process.  (Note: For a sterile placebo product, the flow diagram should address the sterilization process.) Summarize the quantitative composition of the placebo product.  I.e., address the quantity of the placebo substance and the quantities of any components that are intended to appear in the placebo product.  For any components used in the manufacture of the placebo product, but which are not intended to appear in the final product, describe the manufacturing processes or procedures that result in the elimination of these components. Specify the acceptable limits of quality and purity of that will form the basis for the release of the placebo product for human use; and provide a description of the various test (i.e., analytical) methods that will be used to establish that the placebo product meets these defined limits. (Note: The test (i.e., analytical) methods that should be submitted will vary according the dosage form of the placebo product.  For example, for sterile placebo products, the sterility and bacterial endotoxin (i.e., pyrogen) test methods should be submitted.) Provide copies of analytical data verifying that the manufacturing process results in a placebo product that meets or exceeds the specified, acceptable limits of identity, strength, quality and purity. Submit a copy of the “certificate of analysis” that will form the basis for release of the placebo product human use. Provide a copy of the labeling that will be assigned to the placebo product. (Note: the placebo product container label should address, at a minimum, the name and address of the manufacturer; the identity of the placebo product; the assigned batch or lot number; the specifications of “sterile” and “pyrogen-free” (if applicable); the conditions for appropriate storage of the product ;and the expiration date.) Alternately, if the cGMP manufacturer of the placebo product has submitted, or will submit, a respective Drug Master File to the FDA (or if the manufacturer already has in place a FDA-accepted IND that addresses the placebo product), incorporate (i.e., within a referenced appendix to the IND application) a letter or written notification from the manufacturer authorizing the FDA to access its Drug Master File (or IND) in support of the chemistry, manufacturing, and control information for the placebo product. Submit a copy of the “certificate of analysis” that will form the basis for release of the placebo product human use. Alternately, if the placebo product is FDA-approved for commercial marketing, specify such.  No additional information is required For a placebo product not manufactured in full compliance with the FDA’s current Good Manufacturing Practice (cGMP) standards:
 Provide a flow diagram of the process for manufacture and packaging of the placebo product; and incorporate a list of the components (including quality specifications and quantities) used in the manufacturing process.  (Note: For a sterile placebo product, the flow diagram should address the sterilization process.) (Note: Alternately, incorporate Drug Master Card(s) that list the components [including quality specifications and quantities] and step-by-step procedures used in the preparation of the placebo product.) Summarize the quantitative composition of the placebo product.  I.e., address the quantity of the placebo substance and the quantities of any components that are intended to appear in the placebo product.  For any components used in the manufacture of the placebo product, but which are not intended to appear in the final product, describe the manufacturing processes or procedures that result in the elimination of these components. Specify the acceptable limits of quality and purity of that will form the basis for the release of the placebo product for human use; and provide a description of the various test (i.e., analytical) methods that will be used to establish that the placebo product meets these defined limits.   (Note: The test (i.e., analytical) methods that should be submitted will vary according the dosage form of the placebo product.  For example, for sterile placebo products, the sterility and bacterial endotoxin (i.e., pyrogen) test methods should be submitted.) Summarize the procedures that will be used to verify the correct operation of the respective analytical equipment.  Provide copies of analytical data verifying that the manufacturing process results in a placebo product that meets or exceeds the specified, acceptable limits of identity, strength, quality and purity.  Submit a copy of the “certificate of analysis” that will form the basis for release of the investigational drug product human use. (Note: Alternately, submit a copy of the Drug Master Card(s) outlining the acceptable limits of identity, strength, quality and purity and corresponding test (i.e., analytical) methods that will form the basis for release of the placebo product for human use.) Provide a copy of the labeling that will be assigned to the placebo product. (Note: the placebo product container label should address, at a minimum, the name and address of the manufacturer; the identity of the placebo product; the assigned batch or lot number; the specifications of “sterile” and “pyrogen-free” (if applicable); the conditions for appropriate storage of the product; and the expiration date.) 6.  Claim for categorical exclusion from or submission of an environmental assessment Incorporate the following statement, as written, unless there is a reason to believe that the distribution and use of the investigational drug product could have an environmental impact The submission of this IND application qualifies for a categorical exclusion from the requirement to submit an environmental assessment or an environmental impact statement in accordance with 21 CFR Sec. 25.31 (e).   To the Sponsor-Investigator’s knowledge, no extraordinary circumstances exist.           

Adapted from Guidance for Industry: Content and Format of Investigational New Drug Applications (INDs) for Phase 1 Studies of Drugs, Including Well-Characterized, Therapeutic , Biotechnology-Derived Products; Center for Drug Evaluation and Research (CDER) and Center for Biologics Evaluation and Research, U.S. Food and Drug Administration; November, 1995.