CMC Review in the 21st Century - PowerPoint PPT Presentation

1 / 30
About This Presentation
Title:

CMC Review in the 21st Century

Description:

Absence of critical information on pharmaceutical development prevents full ... Focuses resources on critical issues ... Peer/critical review of CMC evaluation ... – PowerPoint PPT presentation

Number of Views:55
Avg rating:3.0/5.0
Slides: 31
Provided by: nasr7
Learn more at: https://www.aaps.org
Category:
Tags: 21st | cmc | century | review

less

Transcript and Presenter's Notes

Title: CMC Review in the 21st Century


1
CMC Review in the 21st Century
  • Moheb M. Nasr, Ph.D.
  • Office of New Drug Chemistry (ONDC)
  • OPS, CDER, FDA
  • NASRM_at_CDER.FDA.GOV
  • AAPS 39th Annual Pharmaceutical Technologies
    Conference at Arden House
  • January 30, 2004

2
CMC Review in the 21st Century
  • Office of New Drug Chemistry (ONDC) Current
    Review Practices
  • Current Challenges
  • Desired State
  • Recent Initiatives
  • Risk Based CMC Review Initiative
  • Process Analytical Technology (PAT)
  • Product Quality System for the 21st Century
  • Continuous Marketing Application (CMA)
  • Moving Forward Future Directions

3
Office of New Drug Chemistry (ONDC)
  • The mission of the Office of New Drug Chemistry
    (ONDC) is to evaluate the chemistry,
    manufacturing, and controls (CMC) portion of new
    drug applications (IND and NDAs), utilizing risk
    management and current scientific principles.
    The CMC review is a key element in the
    establishment of critical quality attributes,
    which are the basis of the control of drug
    quality related to drug safety and efficacy.
    ONDC initiates and participates in guidance
    development, participates in internal and
    external meetings, presents Agency policy
    regarding CMC issues to outside audiences, and
    collaborates in research, both internally and
    externally.

4
(No Transcript)
5
CMC Review
  • To assure the identity, purity, quality, and
    strength/potency, as related to the safety and
    efficacy of new drugs throughout their life
    cycle
  • IND NDA Post Approval
  • ,

6
ONDC WORKLOAD
  • Completed CMC Reviews in FY 2003
  • 159 NDAs (103 original and 56 resubmitted)
  • 342 Commercial INDs
  • 507 Research INDs
  • 1858 CMC Supplements (820 PAS), not including
    efficacy and labeling
  • 1132 Annual reports

7
CMC Review Practices at ONDC
  • Highest Quality Review conducted by competent and
    dedicated staff
  • Evaluation of ALL CMC information in submitted
    applications
  • Absolute adherence to FDA and ICH guidances
  • Tight specification to assure consistency of
    manufacturing processes
  • Attempts made to provide regulatory relief to
    industry resulting, at times, in an increased
    workload of ONDC reviewers
  • Late and voluminous CMC submissions often leading
    to delayed CMC review
  • Regulatory decisions made based on submitted data
    and individuals experience with products and
    processes
  • Absence of critical information on pharmaceutical
    development prevents full utilization of
    risk-based assessment in CMC review

8
Current Challenges
  • Product Quality System for the 21st Century A
    new Paradigm
  • First Cycle Review
  • Review and Inspection Boundaries and Roles
  • Work Load
  • Consistency among 19 Chemistry Teams across CDER
    clinical divisions
  • Guidance and policy development
  • Lack of expertise in some critical CMC areas
  • Novel dosage forms and combination drug products
  • New technologies

9
Pharmaceutical Quality
  • Quality A product or service that meets/exceeds
    customers needs
  • Old Paradigm
  • Establishment and enforcement of public/private
    standards to assure the integrity of interstate
    commerce (adulteration, counterfeit, etc.)
  • Relevance to safety and efficacy?
  • New Paradigm (JW, September 17, 2003)
  • FDA stands in for the customer and establishes
    enforces quality standards in the realm of
    clinical performance (ultimate metric!)

10
Pharmaceutical QualityThe New Paradigm (JW)
  • Clinical performance delivery of efficacy and
    safety as described in the label and derived from
    the clinical trials
  • Fitness for use is a surrogate for clinical
    performance
  • A product that is fit for use meets established
    quality attributes (Purity, Potency/strength,
    Identity, Bioavailability/delivery,
    Labeling/packaging, Physical performance, etc.)
  • Products must be made in compliance with cGMP

11
Pharmaceutical QualityThe New Paradigm (JW)
  • Need to examine and evaluate linkages between
  • Quality attributes and clinical performance?
  • Values/specifications and safety and
    effectiveness?
  • cGMP compliance/inspection and safety and
    efficacy?
  • Ultimate Goal The availability of high quality
    of safe and effective drugs to the patient at
    reasonable cost

12
The Desired State
  • Product quality and performance are ensured
    through the design of effective and efficient
    manufacturing processes
  • Product and process specifications are based on a
    mechanistic understanding of how formulation and
    process factors affect product quality and
    performance
  • Continuous quality assurance and process
    improvement
  • Relevant regulatory policies and procedures are
    tailored to reflect current level of scientific
    knowledge
  • Risk-based regulatory approaches recognize
  • the level of scientific understanding of how
    formulation and manufacturing process factors
    affect product quality and performance
  • the capability of process control strategies to
    prevent or mitigate the risk of producing a poor
    quality product

13
Risk Based CMC Review
  • Benefits of a Risk Based System
  • Patients
  • Increased availability
  • Faster approval of new products
  • Continue to receive quality products
  • FDA
  • More product and process knowledge shared by
    industry
  • More efficient resource allocation for review and
    inspection
  • Increased trust and understanding of industry
    decision making
  • FDA/PQRI Workshop, April 2003, Washington, D.C.

14
Risk Based CMC Review
  • Benefits of a Risk Based System
  • Industry
  • Fewer, more efficient, science based inspections
    resulting in increased consistency
  • Faster, more consistent reviews
  • Potential for reduced regulatory burden
  • Manage changes and nonconformance with less FDA
    oversight
  • Focuses resources on critical issues
  • Flexibility to focus on what should be done, not
    what can be done
  • Improves communication with FDA
  • FDA/PQRI Workshop, April 2003, Washington, D.C.

15
Risk Based CMC Review Initiative
  • Goal To provide regulatory relief by
    incorporating science based risk assessment to
    CMC review
  • Current Risk-Based CMC Initiative
  • Evolved over the last few years
  • Multi-Tiered (establish attributes, propose and
    finalize a drug list before determining
    eligibility for regulatory relief)
  • Product Specific
  • Synthetic Drug Substances only!
  • Characterization achieved by traditional
    analytical techniques
  • IR oral solids, oral solutions, non-sterile
    topical solutions and sterile solutions of simple
    salts
  • A more progressive and expanded Approach will
    focus more on Process Understanding

16
Process Analytical Technology (PAT)
  • PAT Initiative only a part of the broader
    Product Quality System for the 21st Century
    Initiative
  • PAT is an example of science and risk-based
    system approaches to product quality assurance
  • PAT provides an opportunity to move from the
    current testing to document quality paradigm to
    a Continuous Quality Assurance paradigm that
    can improve our ability to ensure quality is
    built-in or is by design - ultimate
    realization of the true spirit of cGMP!
  • Greater insight and understating of manufacturing
    processes
  • At/On/In-line measurement of performance
    attributes
  • Real-time or rapid feedback controls (focus on
    prevention)
  • Potential for significant reduction in production
    and development cycle time
  • Minimize risks of poor product quality and reduce
    regulatory concerns

17
PAT Guidance - An Integrated Systems Approach
  • The draft PAT guidance embraces an integrated
    systems approach to CMC review and cGMP
    inspections
  • Companies will also need to coordinate product
    development, manufacturing, quality assurance,
    and information/knowledge management functions in
    an integrated manner.

18
Process Analytical Technology (PAT)
Regulatory Framework
  • Implementation of PAT is not a requirement
  • Research exemption
  • Continuous improvement without the fear of being
    considered non-compliant
  • Regulatory support and flexibility during
    development implementation
  • Eliminate the fear of delayed approval
  • Dispute avoidance/resolution
  • Science Risk based regulatory approach
  • Low risk categorization based on a higher level
    of process understanding

19
A Product Quality System for the 21st Century
  • Initiative focus on product quality and not just
    cGMP
  • Major changes in pharmaceutical business over the
    years
  • Greater dependence on medicine in health care
  • Advances in pharmaceutical and manufacturing
    sciences
  • Applications of biotechnology
  • Globalization of industry
  • Some regulatory adjustments over the years
  • 1978 cGMP Revision
  • CDER-ORA agreement in early 90s
  • SUPAC, BACPAC, FDAMA in late 90s
  • Team Biologics

20
A Product Quality System for the 21st Century -
Fundamental Principles
  • Risk Management (priorities/resource allocation
    and setting regulatory requirements)
  • Science-based regulatory approaches (conduct
    scientific risk assessment and facilitate
    technological advances)
  • Strong public health focus
  • International cooperation
  • Assessment and implementation of appropriate
    quality management systems
  • Integrated product quality regulatory practice
    (review and inspection processes)

21
NEW DRUG DEVELOPMENT
Discovery/ Preclinical

IND Process
NDA Review
I
HISTORICAL
II
P O S T A P P R O V A L
III


I

CURRENT
II
Dt
III
I


Dt
FUTURE GOAL
II
III
INDUSTRY TIME


FDA TIME
22
Continuous Marketing Application (CMA)
  • Pilot Programs to test whether providing early
    review of selected applications and additional
    feedback and advice to sponsors can further
    shorten drug development and review times
  • Enhance interaction between FDA and applicants
  • Only Fast Track (FT) products are eligible
  • Serious or life-threatening
  • Potential to address unmet medical need

23
Continuous Marketing Application (CMA)
  • Pilot 1 - Reviewable Units (RU) for FT under
    PDUFA
  • Review a limited number of presubmitted portions
    (RU)
  • Issue a Discipline Review Letter (DRL) for each
    RU within 6 months of receipt
  • Implementation October 1, 2003
  • CMC
  • Prefer the complete technical section
  • Drug substance RU may be submitted
  • Rarely, drug/biologic product RU may be submitted

24
Continuous Marketing Application (CMA)
  • Pilot 2 - Scientific Feedback and Interaction
    During Development of FT Products under PDUFA
  • More frequent scientific interaction
  • Limited to no more than one FT product per review
    division
  • Implementation October 6, 2003
  • Pilot 2 Applicants should describe
  • Past and projected milestone in drug development
  • Value of frequent communication
  • Potential for improvements in the efficiency of
    drug development
  • Termination
  • Marketing application submitted
  • Applicant withdraws from the pilot
  • FDA terminates

25
Problems and Issues in Current Paradigm
  • For FDA
  • Resource intensive
  • Expensive and time-consuming litigation and legal
    actions
  • Dealing with recalls and drug shortages
  • For Industry
  • Discourage continuous improvement
  • Regulatory burden, not a value added activity
  • Consequences of non-compliance
  • For Public
  • High cost of drugs
  • Hostility, at times, towards industry and
    regulators

26
Moving Forward- Future Directions
  • ONDC will develop new strategies to recruit,
    hire and train CMC reviewers with expertise in
    drug discovery, analytical chemistry,
    pharmaceutical development/formulation and
    pharmaceutical engineering
  • We will build a strong and independent scientific
    organization to better serve the public and all
    internal (OND, OC, OGD) and external (industry,
    scientific organizations and academic institutes)
    stakeholders
  • We will reengineer CMC review process at CDER
  • To address problems identified by FDA, Public and
    Industry
  • To meet expectations of the new paradigm and to
    achieve the desired state
  • To establish a modern quality system with
    appropriate metrics to measure quality of CMC
    review and performance

27
Moving Forward- Future Directions
  • ONDC will provide better work environment and job
    satisfaction to CMC reviewers
  • The new ONDC organization will be structured to
    facilitate the implementation of the new review
    processes
  • Consider establishing an FDA CMC Scientific
    Advisory Board
  • Provide consultation
  • Oversee regulatory research program
  • Restructure and modernize ONDC training program
  • Develop a seminar series

28
Moving Forward- Future Directions
  • CMC specification to be based on
  • Clinical Relevance
  • Process capabilities
  • Risk-based assessment
  • Knowledge gained from Pharmaceutical Development
    Reports
  • Better utilization of modern statistical
    methodologies
  • Review practices based on good scientific
    principles (GSP)
  • Increased emphasis on manufacturing science
  • Peer/critical review of CMC evaluation by FDA
    scientists and clinicians
  • Better integration of review and inspection

29
Moving Forward- Future Directions
  • Regulatory relief based on
  • Process understanding and control (pharmaceutical
    development reports)
  • Quality systems through out manufacturing
    processes
  • Continuous improvement
  • pharmaceutical Development Reports may
    facilitate
  • Meeting first cycle review goal
  • Science based specifications
  • Risk-based GMP inspection

30
Acknowledgments
  • Janet Woodcock (CDER)
  • Helen Winkle (OPS)
  • Ajaz Hussain (OPS)
  • Chi-Wan Chen (ONDC)
  • Chuck Hoiberg (ONDC, Pfizer)
Write a Comment
User Comments (0)
About PowerShow.com