Sunday, October 31, 2010

Michael O'Donnell - Blog Post #3

Name of Guidance: Guidance for Industry Part 11, Electronic Records; Electronic
Signatures — Scope and Application

Status of Guidance: Final

Link to Guidance: http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm072322.pdf

Release Date: 09/03/03

Target Audience: Health care industry and regulatory publishing professionals

Laws and Regulations Referenced:

  • 21 CFR Part 11, Electronic records and electronic signatures regulation
  • 21 CFR Part 58, Good Laboratory Practice for nonclinical laboratory studies regulation
  • 21 CFR Part 211, Good Manufacturing Practice regulation
  • 21 CFR Part 820, Quality system regulation


Summary:
Unlike the Food and Drug Administration’s (FDA) Guidances for Industry, “Providing Regulatory Submissions in Electronic Format - General Considerations," and “Providing Regulatory Submissions in Electronic Format - Receipt Date,” the subject of this 12-page guidance is specifically the scope of electronic records regulation of record-keeping practices and electronic signatures. The guidance emphasizes the need for being compliant with procedures for record copying, validation, record retention, and audit trail design in accordance with part 11 of Title 21 of the Code of Federal
Regulations (21 CFR part 11).

The FDA warns in this guidance that in fulfillment of 21 CFR part 11, companies should guard against unecessary measures in electronic submission development. The vast language of part 11 can lead a company into misinterpretation of the record-keeping process recommended for an electronic submission. If part 11 is incorrectly applied by a regulatory department to an electronic submission process, then the company could incur higher cost and lost time.

Recognizing that the subject matter of 21 CFR part 11 is too open to wide-ranging interpretation, this guidance aims to better focus what exactly this part 11 information means to the healthcare industry. Within the industry, the FDA is specifically targeting this guidance to the individuals responsible for publishing regulatory data to the FDA for review.

The key points identified by the guidance for regulatory professionals to consider:

    1. By correct identification of data, reduce the amount of records that are subject to 21 CFR part 11, so that the regulation can be more narrow in focus

    2. For records that are subject of part 11, the FDA can enforce the regulation at it’s own discretion, but there is a “grandfather clause,” in which older pre-1997 records systems included in a review have some degree of flexibility due to the limitation of the record systems from that era

    3. FDA enforces all predicate rules and requirements


Rationale:
With this guidance, the frequently referenced part 11 can be better understood. By narrowing the focus of 21 CFR part 11, The FDA is hoping to instill greater refinement of the overall process of electronic record handling, improved communication with regulatory professionals, and increased data integrity.

Resulting Recommendations:
Companies that are submitting regulatory documents to the FDA for review should follow this guidance to ensure a navigable audit trail and more accurate submission document record-keeping. This guidance should be utilized as a tool for better understanding of 21 CFR part 11 as it pertains to records, electronic signatures, records access and records control.

Impact:
Understanding what the FDA expects of handlers, controllers, or publishers of electronic regulatory documents is better understood with this guidance. Researching an audit trail of all phases of document development is easier. The accuracy of the signature process of parties responsible for different documentation in a regulatory submission is improved. The broad scope of 21 CFR part 11 is narrowed to a level that is more easier understood and better applicable at the regulatory professional level of implementation.

Saturday, October 30, 2010

Blog Post #3: Direct to Consumer Prescription Drug Advertisements

Rosalyn Finlayson

October 30, 2010

Name of Guidance:

Direct-to-Consumer Prescription Drug Advertisements: Presentation of the Major Statement in Television and Radio Advertisements in a Clear, Conspicuous, and Neutral Manner

Status of Guidance

Proposed Rule

Date of Guidance

March 29, 2010

Link to the Guidance

http://edocket.access.gpo.gov/2010/pdf/2010-6996.pdf

Target audience

Manufacturers of prescription drugs, advertising agencies and the general public.

Laws An Regulations Referenced

21 USC 352 Section 502n: stipulations for “true statements” in advertising

21 CFR 202.1: describes requirements for print and broadcast advertising

58 FR 42364: outlines “clear and conspicuous” disclosure in advertising

63 FR 24996: outlines “clear and conspicuous” disclosure in audio advertisements

Public Law 102-556: Telephone Disclosure & Dispute Resolution Act of 1992

Public Law 110-85 Section 901: Food Drug Administration Amendments Act of 2007

Public Law 106-102: Gramm Leach Bliley Act

Summary

This proposed rule is a follow up to the Food & Drug Administrations May 2009 guideline for Presenting Risk Information in Prescription Drug and Medical Device Promotion. Under this proposed rule the authority of the FDA would expand from making recommendations to having legal authority to enforce compliance. The proposed rule clarifies terminology of the following terms; neutral, major statement, and clear and conspicuous nature. Discussed in this proposal are the Eastern Research Groups findings of peer-reviewed literature on direct to consumer advertising published from 2004 through 2008. The proposed rule also discusses the FDA’s decision to adhere to standards that are consistent with previous regulations adopted by a number of other federal agencies. The proposed effective date of this particular rule will be 90 days after publication in the Federal Register. The FDA solicits public comment on several segments of this proposed rule.

Rationale

This proposed rule was developed because of the growing body of research indicating that there is an influence of direct to consumer advertising on public health. The FDA has taken into consideration the research findings of the Eastern Research Group and revised some of the recommendations contained in the May 2009 guideline Presenting Risk Information in Prescription Drug and Medical Device Promotion. The FDA has revised its previous recommendations in order to be aligned with the disclosure regulations and standards presented by other federal agencies. This proposed rule could potentially have a positive effect on the health outcomes of the general public through better communication of risk information in prescription drug television and radio advertisements.

Resulting Recommendations

Based on the regulations of a number of other federal agencies and the findings presented by the Eastern Research Group the FDA is presenting the following regulations in this proposed rule.

All risk factors, major side effects and any contraindications of prescriptions drugs must be presented in a neutral, clear and conspicuous manner regardless of how the benefit is presented.

Communication of risk information in major statements in consumer-directed prescription drug advertisements must be presented so that the consumer receives a fair and accurate impression of the drug being promoted.

The major statement in television advertisements must be included in both the audio and visual parts of the presentation.

Advertisements for prescription drugs must follow FTC regulations for the ease of comprehension of the language as well as the formatting and location of textual information in the disclosure.

Advertisements for prescription drugs must follow FTC regulations for the audio components of advertisements such as pacing, volume and quality of speech.

Impact

The implementation of this proposed rule would further regulate the manner in which manufacturers of prescription drugs would be able to present drug information in directly to consumer advertisements. This proposed rule is designed to ensure information about a prescription drug is clearly presented in advertisements targeting the general public.

Thursday, October 28, 2010

Blog #3 Integrated Summaries of Effectiveness and Safety: Location Within the Common Technical document

Theresa Seiverd
BW706-Blog 3

Name: Integrated Summaries of Effectiveness and Safety: Location Within the Common Technical document

Status: Final Guidance

Release Date: April 2009

Link to Guidance:
http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM136174.pdf

Target Audience: Industry, specifically clinicians, biostatisticians, and writers who work together to write the Integrated Summary of Efficacy (ISE) and Integrated Summary of Safety

Laws and regulations referenced:
21 CFR 314.50(c)(2)(viii)
21 CFR 314.50(d)(5)(v)
21 CFR 314.50(d)(5)(vi)(a)

Summary: There has been confusion on the part of applicants submitting a new drug application (NDA) or a biologic license application (BLA) in omitting the ISE and ISS documents from Module 5 of the Common Technical Document (CTD) because of their similarity to the Summary of Clinical Efficacy (SCE) and Summary of Clinical Safety (SCS) that are located in Module 2 of the CTD. Many applicants have argued that it is redundant to submit a separate ISE and ISS because of the similarity to the SCE and SCS. Consequently, the FDA had observed an increase in the number of NDAs without a fully comprehensive ISE and ISS that is required by US regulations.

This guidance was intended to clarify the location regarding the ISE and ISS documents in the CTD and the differences of intent between the ISE/ SCE and ISS/SCS.

Rationale: The ISE and ISS are documents that are unique to the United States. A common assumption is that the documents are summary documents that provide an overview of efficacy and safety data. However, the ISE and ISS provide a detailed integrated analysis with more tables, figures, and listings that are far more extensive and may number up to a 1000 pages. Unlike the SCE, the ISE will often explore similarities and differences among an extensive set of baseline characteristics to determine whether these factors influence outcomes. Similarly, the ISS will analyze data across multiple studies with a more in depth analysis to identify safety signals that the overall safety database may not identify. 1 Therefore, it is essential that these documents are not omitted from the CTD and have a separate location from the clinical summaries section located in Module 2 of the CTD.

Resulting recommendation: Due to the extensive length of the ISE and ISS compared to the SCE and SCS documents, the appropriate location in the CTD is Module 5, or more specifically Section 5.3.5.3, “Reports of Data from More than One Study”. The guidance does provide examples of when it is acceptable to use the same information in the ISE or ISS in the text of the SCE and SCE for both modules, or how to appropriately cross reference between Module 2 and Module 5. For instance, the text summary must be listed in both modules even though it may be the same, but it is okay to just include the tables and appendices in Module 5 and reference this information in Module 2. It is not acceptable, to include all information for the integrated summaries (ie, SCE and SCS) in Module 2 and submit nothing in Module 5 of the CTD.

Impact: This guidance has provided a map for Applicants to develop a strategy for creating the ISE and ISS documents in Module 5 that can be readily condensed and/or mapped to produce the SCE and SCS documents that meet the US requirement for the clinical summary that is located in Module 2 of the CTD. By developing a strategy to produce the larger more comprehensive documents first, applicants can move towards producing better quality and consistency among documents at a much faster rate. This will have the added benefit of facilitating FDA’s review since the information between Module 2 and Module 5 is similar. FDA may choose to review both these sections together instead of taking a sequential approach in reviewing each module in order by number. It would make sense to review these modules in tandem to get a better comprehensive understanding of the efficacy and safety for the drug under review.
____________________________
1 Swartz D., Umen M., Nomides K., and Vanderhoof M., Understanding the differences and effectively transitioning between the US Integrated Summaries of Effectiveness and Safety (ISE/ISS) and the CTD Summaries of Clinical Efficacy and Safety (SCE/SCS). Drug Information Journal, Volume 44, pp 641-648, 2010.

Sunday, October 17, 2010

FDA GUIDANCE BLOG #3
Laura Salomon

Name of Guidance:
Guidance for Industry - Submitting and Reviewing Complete Responses to Clinical Holds

Status of Guidance/Release Date:
Submitting and Reviewing Complete Responses to Clinical Holds is a final guidance that was issued in October, 2000.

Link to the Guidance:
The guidance can be found at the following location:
http://www.fda.gov/RegulatoryInformation/Guidances/ucm127537.htm

Target Audience:
This guidance is targeted to sponsors who submit Investigational New Drug (IND) applications to the FDA and are subsequently notified by the Agency that a clinical hold has been issued.

Laws and Regulations Referenced:
63 FR 68687 Federal Register of December 14, 1998

21 CFR 312.42 Clinical Holds and Requests for Modification

Food Drug and Cosmetic Act, Section 505(i)(3) – states the Agency will respond within 30 calendar days to a sponsor’s request that a clinical hold be removed from an investigation

Prescription Drug User Fee Act of 1992 (PDUFA) – allows the Agency to collect fees from companies that manufacture certain human drug and biological products

Summary:
After submitting an IND to the Agency, a sponsor may be notified that a clinical hold has been issued on the application. If a clinical hold is issued, the sponsor must suspend an ongoing clinical investigation and/or delay any proposed investigations, and studies cannot resume until the sponsor has received word from the Agency. A letter detailing reasons for the clinical hold must be sent to the sponsor within 30 days of the initial notification; however, the sponsor can respond to this Clinical Hold Letter, and the Agency has committed to evaluate this response make a final decision within 30 days. This guidance describes procedures for sponsors to submit responses to clinical holds correctly to ensure the Agency evaluates these responses within the established 30-day period.

According to this guidance, there are several steps sponsors can take to make sure their Complete Response, detailing rectification of issues resulting in clinical hold, is addressed by the FDA accordingly:

  • Complete Responses should not address issues other than those related to clinical hold.
  • Cover letters should include at the top in bold letters “CLINICAL HOLD COMPLETE RESPONSE”.
  • Responses should be sent in triplicate, and cover letters can be faxed to FDA contacts.

When the FDA receives copies of the response, the 30-day clock for review begins. Amendments providing additional information will not extend this time period; the Agency will either stop the clock or begin another 30-day period based on the receipt date of the amendment. Furthermore, if the FDA believes the response is incomplete (i.e. does not address all clinical hold issues), the 30-day clock will not begin.

Rationale:
This guidance is a communication from the FDA to sponsors of its commitment to respond to Complete Responses within 30 days of receipt. This timeline was originally endorsed by the Secretary of Health and Human Services in 1998 when IND regulations were amended to reflect this new policy in the Federal Register of December 14. The original goal of a 75% timely response rate was increased to 90% between 1999 and 2002. To ensure that the FDA continues to meet its standards, this guidance describes steps sponsors can take to ensure their responses are appropriately handled so that the 30-day clock can begin.

Resulting Recommendations:
The FDA recommends that sponsors who receive notification of clinical hold follow the instructions in this guidance. Doing so helps to ensure that the 30-day period begins once a Complete Response is received; this process allows the FDA to assess if a clinical hold can be lifted in a timely manner.

Impact:
The FDA’s commitment towards standardization, prompt review, and clear communication with applicants is evident in Submitting and Reviewing Complete Responses to Clinical Holds. As a result of the Agency’s commitment and clear instructions to sponsors for responding to clinical holds, IND studies should be able to resume within a month as long as sponsor responses are viewed as complete. This timeline helps sponsors to prepare accordingly for resuming their studies and continue investigating a drug product in trial.

A main goal in drug development and getting a product to market is doing so quickly and correctly. It is, therefore, in a sponsor’s best interest to ensure that responses to clinical hold are handled appropriately so that time isn’t inadvertently wasted in correcting errors.

Monday, October 11, 2010

Michael O'Donnell Blog Post #2

Name of Guidance: International Conference on Harmonization (ICH) - Guidance for Industry: Granularity Document Annex to M4: Organization of the CTD

Status: Final within ICH, but at time of update was still pending inclusion in future revised FDA M4 guidance

Release Date: October 2005

Link: http://www.fda.gov/RegulatoryInformation/Guidances/ucm129555.htm

Target Audience: Regulatory operations departments in pharmaceutical companies, or individual preparers of Common Technical Document (CTD) -based submissions to the US Food and Drug Administration (FDA)

Laws and Regulations Referenced: None

Guidances referenced: ICH M4 Guidance

Summary: This guidance is an annex to a previous International Conference on Harmonisation (ICH) guidance on CTD assembly and organization, and is included among FDA guidances due to the FDA being an ICH participant. The fundamental organization of the CTD used in regulatory submissions is explained, with a strong emphasis on the concept of document granularity. The guidance answers definitively these two basic questions:

  1. What is officially considered a document in a paper submission?

  2. What is a document in an electronic or digital submission?

Rationale: The annex became necessary to provide answers to numerous questions raised by the target audience of the original M4 guidance. By further explaining granularity, and how this concept contributes to pagination and table of contents structure, documents from Module 2 through Module 5 have a road-map for the complete understanding of internationally compliant document assembly.

Resulting Recommendations: The ICH recommends that this guidance be utilized by its participating regulatory agencies: the FDA, the European Medicines Agency (EMEA), and Japan's Ministry of Health, Labour, and Welfare (MHLW).

Impact: This guidance explicates what constitutes a CTD document format, as well as an electronic Common Technical Document (eCTD) format. Explained in detail are the management of multiple documents within the same module, resulting pagination, format, and table of contents issues encountered during CTD and eCTD submissions. Embracing document granularity in a CTD format as the accepted international standard allows for greater global sharing of pharmaceutical information. Perhaps of equal importance, when this standard is adhered to properly, is that submission review turnaround time is reduced significantly.





Guidance for Industry: Safety Testing of Drug Metabolites

Name of Guidance: Guidance for Industry: Safety Testing of Drug Metabolites

Status of Guidance: Final

Release Date: 14 February 2010

Link: http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm079266.pdf

Target Audience: Individuals/companies developing non-clinical safety evaluation plans for new chemical drug products.

Laws and Regulations Referenced: Good Laboratory Practices (21 CFR part 58). GLPs are the general regulations that apply to the conduct of all nonclinical studies to be submitted for evaluation by the FDA. GLPs set standards for operations, animal care and use, documentation, key personell, and quality assurance.

Summary: Metobolic profiles should be identified prior to initiation of human clinical trials using human and animal in vitro models and in vivo methods early in the development process. Significant differences between human and animal in vitro and in vivo metabolite investigations can signal the need for safety evaluations of just the metabolite. Analytical methods for synthesizing and qualifying the metabolite for direct administration to animals and bioanalytical methods for measuring systemic exopsure must be developed.

When it is determined that additional metabolite safety ecaluations are needed, standard nonclincal safety evalutaions should be performed. General toxicity studies should be performed at multiples of human exposure to the metabolite in order to collect relavent toxicokinetic data. Standard genotoxitiy tests that evalutate mutations and chromosamal aberrations should also be performed. A complete genotoxocity battery is only necessary if positive results are obtaine from one or both of the intitial tests. Developmental toxicity studies should be performed in at least one species if the drug is intended for use in women of child bearing potential. Carcinogenicity studies should be performed when the parent drug is intended for long term (6+ months) or intermitant long term use.

Rationale: The standard approach to nonclinical drug safety testing compares systemic exposure of investigational drug products in animal models to exposure in humans to assess the risks applicable to humans. Due to differences in drug metabolism between species, metabolites of drug products can be formed in humans that do not form in animals. When metabolites of the drug product are formed only in humans or in significantly larger proportions in humans than in animals, safety assessments in animals must be performed to assess potential risks of the metabolite in humans. Further safety assessments of metabolites are not necessary if the metabolite was formed in at least one species during assessments of the parent drug at greater than 10% of parent systemic exposure.

Resulting Recommendations: The timing and extent of the nonclinical safety evaluations of dispropportionate drug metabolites should be determined as early in the drug development process as possible. Careful consideration of the exposure to drug metabolites in several species should be made when determining additional safety evaluations. If the indication of the parent drug is for a life threatening condition with limited or no other treatment options, the extent of metabolite testing may be reduced with adequate consideration by review agencies.

Impact: The formation of drug metabolites can complicate the nonclinical safety evaluation requirements for the parent drug. For parent drugs with more than one disproportionate metabolite, additional testing can be costly and create significant developmental delays, which is why early determination of metabolic profiles is important for designing a complete development plan. Safety testing of metabolites is necessary determine toxicities and pharmacology.

Sunday, October 10, 2010

Jeff Moore Blog Post 2

Name of Guidance
Guidance for Industry and Investigators: Safety Reporting Requirements for INDs and BA/BE Studies

Status of Guidance
Draft

Release Date
September 2010

Link to the Guidance
http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM227351.pdf

Target Audience
Sponsors and investigators conducting investigational new drug application (IND) studies or bioavailability/bioequivalence (BA/BE) studies.

Laws and Regulations Referenced (and what each Law states in relevant part)
• 21 CFR 312.32(c)(1)(i)(A) and (B)—requires sponsors investigating a drug under an IND to submit a written safety report notifying the FDA and all participating investigators of any adverse event (AE) that is serious, unexpected, and “associated with the use of the drug,” and any finding from animal testing that suggests a significant risk for humans.
• 21 CFR 312.32(a)—defines the phrase “associated with the use of the drug” as meaning “there is a reasonable possibility that the experience may have been caused by the drug.”
• 21 CFR 312.53(c)(1)(vii), 312.66, and 56.108(b)(1)—require investigators to report to the investigational review board (IRB) all unanticipated problems involving risk to subjects.
• 21 CFR 312.32(b)—requires sponsors to review all information relevant to the safety of the drug.

Other Guidances Referenced
• Guidance for Clinical Investigators, Sponsors, and IRBs: Adverse Event Reporting to IRBs—Improving Human Subject Protection (http://www.fda.gov/downloads/Drugs/ GuidanceComplianceRegulatoryInformation/Guidances/ucm079753.pdf)
• ICH E2A Guideline for Industry, Clinical Safety Data Management: Definitions and Standards for Expedited Reporting (http://www.fda.gov/downloads/Drugs/ GuidanceComplianceRegulatoryInformation/Guidances/ucm073087.pdf)

Summary
This guidance provides FDA recommendations regarding when and how to submit a safety report, defines terms used in safety reports, and provides clarification regarding other safety reporting issues.

Rationale
On September 29, 2010, FDA published a final rule changing IND safety reporting requirements (21 CFR part 312) and adding safety reporting requirements for BA and BE studies (21 CFR part 320).
The changes for IND safety reports were implemented because sponsors too frequently submitted safety reports for serious AEs that were unlikely to be related to the investigational drug. Although such reports consume resources of FDA, investigators, and IRBs, they provide no meaningful information about a drug’s safety. The final rule revises definitions for safety reporting and clarifies when and how to submit IND safety reports.
Certain in vivo BA and BE studies were previously exempted from IND safety report requirements. However, because the FDA has received important safety information from such studies, the final rule contains new safety reporting requirements for previously-exempt BA and BE studies.

Resulting Recommendations
The final rule provides definitions for terms that are meant to be clear, consistent, and harmonious with international terms. The definitions are as follows:
Adverse event means any untoward medical occurrence associated with the use of a drug in humans, whether or not considered drug related.
Adverse reaction means any adverse event caused by a drug.
Suspected adverse reaction means any adverse event for which there is a reasonable possibility that the drug caused the adverse event. A suspected adverse reaction implies a lesser degree of certainty about causality than adverse reaction.
Unexpected refers to an adverse event, adverse reaction, or suspected adverse reaction that is not listed in the investigator brochure or is not listed at the observed specificity or severity; or, if an investigator brochure is not available, is not consistent with the risk information described in the investigational plan or the current application.
Serious refers to an adverse event, adverse reaction, or suspected adverse reaction that, in the view of either the investigator or sponsor, results in any of the following: death, a life-threatening adverse event, inpatient hospitalization or prolongation of existing hospitalization, a persistent or significant incapacity or substantial disruption of the ability to conduct normal life functions or a congenital anomaly/birth defect.
Events that require medical or surgical intervention to prevent an outcome listed above may also be considered serious.
Life-threatening refers to an adverse event, adverse reaction, or suspected adverse reaction that, in the view of either the investigator or sponsor, places the subject at immediate risk of death.
According to the final rule, it is the responsibility of the sponsor to review all new safety information relevant to the drug, whether from clinical or epidemiological investigations, animal or in vitro studies, reports in the scientific literature, unpublished scientific papers, foreign regulatory authorities, or foreign commercial marketing experience. Literature searches should be conducted at least annually.
If an IND safety report is required, the sponsor must submit it to FDA and all participating investigators no later than 15 calendar days after receiving the safety information. (Fatal or life-threatening adverse events must be reported no later than 7 calendar days after becoming aware of the occurrence.) In each IND safety report, the sponsor must analyze the significance of the suspected adverse reaction in light of any previously-submitted safety reports. Sponsors should periodically review their entire safety database, not only for IND safety reporting, but also to update investigator brochures.
An IND safety report is required when any of the following occurs:
Serious and unexpected suspected adverse reaction. The event must be a suspected adverse reaction (as defined above) that is both serious and unexpected. Otherwise, it should not be submitted as an IND safety report.
Findings from other sources. This includes findings from other studies, or any findings from animal or in vitro testing, that suggest a significant risk to humans. A finding that suggests a significant risk requires a protocol amendment to be submitted as well as an IND safety report.
Increased occurrence of serious suspected adverse reactions. A clinically important increase in the rate of a serious suspected adverse reaction over that listed in the protocol or investigator brochure must be reported.
Other IND safety reporting issues include alternative reporting arrangements (must be agreed to in advance of the investigational study), investigator brochures (a serious suspected adverse reaction is considered unexpected if not listed in the investigator brochure; until the investigator brochure is updated, all subsequent, similar, serious suspected adverse reactions must be submitted as IND safety reports), and unblinding (if the blind was broken and the subject was receiving placebo, the event should not be reported in an IND safety report), investigator reporting (except for study endpoints, the investigator must immediately report all serious adverse events, drug-related or not, to the sponsor), and investigations of marketed drugs (the only reports that must be submitted as IND safety reports for a US-marketed or US-approved drug are those arising from a study conducted under the IND; all others should be reported per the relevant postmarket safety reporting requirements).
For IND safety reports for individual cases, a sponsor should use FDA Form 3500A. Foreign suspected adverse reaction reports may be submitted on a CIOMS I Form instead. A narrative format must be used for reports of overall findings or pooled analyses. The report must be submitted to the CDER (or CBER) review division responsible for review of the IND.
Persons conducting a BA or BE study must notify FDA and all participating investigators of any serious adverse event, drug-related or not, observed during conduct of the study, no later than 15 calendar days after becoming aware of its occurrence. This includes BA and BE studies exempt from the IND requirements under part 312 that are conducted in the US, but not those conducted outside the US. Serious adverse events observed in the investigational drug group and in the approved drug group must be reported. Fatal or life-threatening adverse events must be reported no later than 7 calendar days after becoming aware of the occurrence.

Impact
FDA’s impact analysis has determined the final rule is not a significant regulatory action and imposes a minimal burden on small entities (less than 0.2 percent of average shipment value for entities with less than 10 employees).1 Implementation of the final rule, and publication of this guidance, is expected to provide clarity for sponsors regarding IND safety report submissions, thus reducing the over- or underreporting of pertinent safety information.

________________________________________
1. Investigational New Drug Safety Reporting Requirements for Human Drug and Biological Products and Safety Reporting Requirements for Bioavailability and Bioequivalence Studies in Humans. Fed Regist. 2010;75(188):59935-59963. To be codified at 21 CFR §312 and §320. Available at: http://www.gpo.gov/fdsys/pkg/FR-2010-09-29/pdf/2010-24296.pdf. Accessed on October 10, 2010.

Michelle Keyvani Blog #2

Name of Guidance:
Financial Relationships and Interests in Research Involving Human Subjects: Guidance for Human Subject Protection

Status of Guidance:
Final Guidance dated May 5, 2004
http://www.hhs.gov/ohrp/humansubjects/finreltn/fguid.pdf

Target Audience:
Investigators, IRB members and staffs, institutions engaged in human subjects research and their officials, and other interested members of the research community

Laws and Regulations Referenced:
45 CFR part 46 - require that institutions performing HHS conducted or supported non-exempt research involving human subjects have the research reviewed and approved by an IRB whose goal is to help ensure that the rights and welfare of human subjects are protected.

21 CFR parts 50, 56 - require that FDA regulated research involving human subjects is reviewed and approved by such an IRB.

45 CFR 46.111(a)(1), 21 CFR 56.111(a)(1) - risks to subjects are minimized

45 CFR 46.111(a)(2), 21 CFR 56.111(a)(2) risks to subjects are reasonable in relation to anticipated benefits, if any, to subjects

45 CFR 46.111(a)(3), 21 CFR 56.111(a)(3) - selection of subjects is equitable

45 CFR 46.111(a)(4), 21 CFR 56.111(a)(4) - informed consent will be sought from each prospective subject

45 CFR 46.116, 21 CFR 50.20 - the possibility of coercion or undue influence is minimized

45 CFR 46.109(b), 21 CFR 56.109(b) - require that additional information be given to subjects "when in the IRB's judgment the information would meaningfully add to protection of the rights and welfare of subjects"

45 CFR 46.124 - the funding agency may impose additional conditions as necessary for the protection of human subjects

45 CFR 46.107(e), 21CFR 56.107 - requires that "no IRB may have a member participate in the IRB's initial or continuing review of any project in which the member has a conflicting interest, except to provide information requested by the IRB"

Summary
Institutions and individuals involved in human subjects research may establish financial relationships for research projects, such as payments for services, equity interests, or intellectual property rights. A financial interest related to a research study may be a conflicting financial interest and affect the rights and welfare of human subjects. This guidance provides some possible approaches to consider in assuring that human subjects are adequately protected.

Rationale
Concerns have grown that financial conflicts of interest in research may affect the rights and welfare of human subjects in research. Relationships between government, academia, industry and others often legitimately include financial relationships and may not cause conflicts of interests. However, when financial interests affect the rights and welfare of human subjects in research IRBs, institutions, and investigators need to consider what actions regarding financial interests may be necessary to protect those subjects.

Resulting Recommendations
In establishing and implementing methods to protect the rights and welfare of human subjects from conflicts of interest created by financial relationships of parties involved in research, the Department recommends that IRBs, institutions engaged in research, and investigators consider the following questions:
  • Does the research involve financial relationships that could create potential or actual conflicts of interest? a) How is the research supported or financed? b) Where and by whom was the study designed? c) Where and by whom will the resulting data be analyzed?
  • What interests are created by the financial relationships involved in the situation? a)Do individuals or institutions receive any compensation that may be affected by the study outcome? b) Do individuals or institutions involved in the research: have nay proprietary interests in the product, including patents, trademarks, copyrights, or licensing agreements? have an equity interest in the research sponsor and, if so, is the sponsor a publicly held company or non-publicly held company? receive significant payments of other sorts? (e.g., grants, compensation in the form of equipment, retainers for ongoing consultation, or honoraria), receive payment per participant or incentive payments, and are those payments reasonable?
  • Given the financial relationships involved, is the institution an appropriate site for the research?
  • How should financial relationships that potentially create a conflict of interest be managed?
  • Would the rights and welfare of human subjects be better protected by any or a combination of the following: a) reduction of the financial interest? b) disclosure of the financial interest to prospective subjects? c) separation of responsibilities for financial decisions and research decisions? d) additional oversight or monitoring of the research? e) an independent data and safety monitoring committee or similar monitoring body? f) modification of role(s) of particular research staff or changes in location for certain research activities, e.g., a change of the person who seeks consent, or a change of investigator? g) elimination of the financial interest?

Institutions - Actions to consider:

  • Establishing the independence of institutional responsibility for research activities from the management of the institution's financial interests.
  • Establishing conflict of interest committees (COICs) or identifying other bodies or persons and procedures to a) deal with individuals' or institutional financial interests in research or verify the absence of such interests and b) address institutional financial interests in research
  • Establishing criteria to determine what constitutes an institutional conflict of interest, including identifying leadership positions for which the individual's financial interests are such that they may need to be treated as institutional financial interests.
  • Establishing clear channels of communication between COICs and IRBs.
  • Establishing policies on providing information, recommendations, or findings from COIC deliberations to IRBS.
  • Establishing measures to foster the independence of IRBs and COICs.
  • Determining whether particular individuals should report financial interests to the COIC. These individuals could include IRB members and staff and appropriate officials of the institutions, along with investigators, among those who report financial interests to COICs.
  • Establishing procedures for disclosure of institutional financial relationships to COICs.
  • Providing training to appropriate individuals regarding financial interest requirements.
  • Using independent organizations to hold or administer the institution's financial interest.
  • Including individuals from outside the institution in the review and oversight of financial interests in research.
  • Establishing policies regarding the types of financial relationships that may be held by parties involved in the research and circumstances under which those financial relationships and interests may or may not be held.

IRB Operations - Policies and procedures to consider:

  • Reminding members of conflict of interest policies at each meeting and documenting any actions taken regarding IRB member conflicts of interest related to particular protocols.
  • Developing educational materials for IRB members to ensure their awareness of federal regulations and institutional policies regarding financial relationships and interests in human subjects research.

IRB Review - Actions to consider:

  • Determining whether methods used for management of financial interests of parties involved in the research adequately protect the rights and welfare of human subjects.
  • Determining whether other actions are necessary to minimize risks to subjects.
  • Determining the kind, amount, and level of detail of information to be provided to research subjects regarding the source of funding, funding arrangements, financial interests of parties involved in the research, and any financial interest management techniques applied.

Investigators - Actions to consider:

  • Including information in the informed consent document, such as a) the source of funding and funding arrangements for the conduct and review of research, or b) information about a financial arrangement of an institution or an investigator and how it is being managed.
  • Using special measures to modify the informed consent process when a potential or actual financial conflict exists, such as a) having another individual who does not have a potential or actual conflict of interest involved in the consent process, especially when a potential or actual conflict of interest could influence the tone, presentation, or type of information presented during the consent process.
  • Using independent monitoring of the research.

Monday, October 4, 2010

Blog Post #2: Good Reprint Practices for the Distribution of Medical Journal Articles and Medical or Scientific Reference Publications on Unapproved

Rosalyn Finlayson

October 4, 2010

Name of Guidance:

Good Reprint Practices for the Distribution of Medical Journal Articles and Medical or Scientific Reference Publications on Unapproved New Uses of Approved Drugs and Approved or Cleared Medical Devices

Status of Guidance

Guidance for industry

Date of Guidance

January 2009

Link to the Guidance

http://www.fda.gov/oc/op/goodreprint.html

Target audience

Manufacturers of drugs and medical devices, health care professionals, and editors of medical journals and scientific or medical reference publications

Laws And Regulations Referenced

21 U.S.C. 355 Federal Food, Drug and Cosmetic Act (FD&C Act)

42 U.S.C. 351 Public Health Service Act

Section 410 Food and Drug Administration Modernization Act (FDAMA)

Summary

This guidance provides a brief history of Section 401 of the Food and Drug Administration Modernization Act (FDAMA) and the Federal Food, Drug and Cosmetic (FD&C) Act. Described in FDAMA Section 410 and the FD&C Acts are the conditions under which manufacturers of drugs and medical devices may disseminate information in journal articles and reference publications regarding the unapproved uses of approved drugs and approved or cleared medical devices.

Although FDAMA Section 401 ceased to be effective on September 30, 2006, the FDA still retains legal authority over the distribution of information about unapproved uses. The FDA evaluates whether or not the distribution of medical and scientific information in journals and reference publications is actually a promotion for an unapproved “new use” for approved drugs and approved or cleared medical devices. The FDA determines whether or not such an activity would cause a particular product to be in violation of the FDC act.

In this guidance the FDA provides its current view and recommendations for “Good Reprint Practices” for journals and reference publications that present information about unapproved uses of approved drugs and approved or cleared medical devices.

Rationale

Articles in medical journals and reference publications often publish information discussing the safety or effectiveness of using approved drugs and approved or cleared medical devices for an unapproved use. Unapproved uses are any use of the drug or medical device that is not included in the products labeling or stated as an intended use for the product. Of concern to the FDA is having an approved drug or medical device marketed for an unapproved use. The FDA will classify approved drugs and approved or cleared medical devices as an unapproved new drug or medical device with respect to the new use. If an approved drug or medical device is marketed for an unapproved use that particular drug or medical device is considered misbranded.

The FDA recognizes that it is in the best interest of public health for health care professionals to receive information from medical journal articles and reference publications about unapproved uses that is truthful and not misleading. In addition the FDA acknowledges the value of having new uses for previously approved products and encourages manufacturers of such products to seek approvals and clearances for new uses.

The FDA recommendations in this guidance also serve the purpose of providing manufacturers of drugs and medical devices with guidelines to follow in order to avoid engaging in conduct that unlawfully promotes an unapproved use of a drug or medical device.

Resulting Recommendations

The FDA recommends compliance with the following principles of “Good Reprint Practices” for publications distributing scientific and medical information.

Journal Articles

  • Scientific or medical journal articles that are distributed should be published by an organization that has an editorial board. It is recommended that the editorial board of a journal use experts with demonstrated expertise in the subject under review. Editorial boards need to be independent of the organization publishing the journal in order to be able to objectively review, select, reject or provide commentary about proposed articles.
  • Recommendations for journal articles state that articles need to be peer-reviewed. Published articles should be in accordance with the peer-review procedures of the organization publishing the journal. The articles published cannot be in the form of a manufacturer funded special supplement to the publication.

Reference Publications

  • The primary distributor of a reference publication should not be the manufacturer of the drug or medical device. Distribution of reference publications should be made available to bookstores or other independent distribution channels that sell medical textbooks or periodicals.
  • A reference publication should not be written, edited, excerpted or published specifically for or at the request of a drug or device manufacturer. The editor of the reference publication should not have any type of financial connection to a manufacturer of drugs or medical devices. Manufacturers of drugs or medical devices of should not have any type of significant influence over a reference publication.

Information

  • Information in journals and reference publications should address well-controlled, scientifically sound clinical investigations of acceptable quality. Experts experienced in pharmacokinetic or pharmacodynamic studies and meta-analyses should appraise the clinical investigations presented in journals and reference publications.
  • A manufacturer that has been informed that a clinical investigation of their product was deemed by the FDA to be an inadequate and not well controlled study should not allow that information to be disseminated in journals or reference publications.
  • Journal and reference publication information must not be false or misleading.
  • Conclusions drawn from a clinical investigation should not be characterized as the only definitive credible evidence if there are existing studies with contradictory conclusions.
  • All information presented must not pose a significant risk to public health if the presented information is followed.

The F.D.A. guidance states that letters to the editor, abstracts, phase 1 reports of trials in healthy subjects, and reference publications that contain little or no substantive discussion of the relevant investigation or data would not be considered consistent with “Good Reprint Practices”.

Impact

The recommendations in this guidance serve as a safe guard to ensure that accurate, truthful and non-misleading information about unapproved uses for approved drugs and medical devices is disseminated to health care professionals and other entities concerned with public health. Manufacturers who follow the recommendations in this guidance will not face enforcement action for attempting to establish and promote their product for an unapproved use.