throbber
AAPS PharmSci 2002; 4 (2) article 7 (http://www.aapspharmsci.org).
`Assuring Quality and Performance of Sustained and Controlled Release
`Parenterals: Workshop Report
`Submitted: March 17, 2002; Published: April 17, 2002
`
`Diane J. Burgess1, Ajaz S. Hussain2, Thomas S. Ingallinera 3 and Mei-Ling Chen2
`
`1Department of Pharmaceutics, University of Connecticut, 372 Fairfield Road, Storrs, CT 06269
`
`2Office of Pharmaceutical Science, Center for Drug Evaluation and Research, Food and Drug Administration, 1451
`Rockville Pike, HFD-350, Rockville, MD 20857
`
`3InfiMed Therapeutics, Inc., 8 Lantern Rd Framingham, MA 01702
`
`* This report may contain statements of opinion that are those of the author(s) and do not necessarily reflect the opinions of AAPS or its members, the
`Food and Drug Administration, or the United States Pharmacopoeia.
`
`PLANNING COMMITTEE
`
`Diane J. Burgess, Ph.D., University of Connecticut, Co-
`Chair
`Ajaz S. Hussain, Ph.D., Food and Drug Administration,
`Co-Chair
`Thomas S. Ingallinera, Ph.D., InfiMed Therapeutics, Inc.,
`Co-Chair
`Mei-Ling Chen, Ph.D., Food and Drug Administration
`Yuan-yuan Chiu, Ph.D., Food and Drug Administration
`Larry A. Gatlin, Ph.D., Biogen, Inc.
`Barbara B. Hubert, United States Pharmacopeia
`Funmi Johnson, Ph.D., Alkermes, Inc.
`Marilyn Martinez, Ph.D., Food and Drug Administration
`Eyal S. Ron, Ph.D., eNOS Pharmaceuticals, Inc.
`Arthur J. Tipton, Ph.D., Southern BioSystems, Inc.
`Lawrence Yu, Ph.D., Food and Drug Administration
`Roger L. Williams, M.D., United States Pharmacopeia
`Melvin H. Weinswig, Ph.D., University of Wisconsin-
`Madison, Continuing Education
`
`SPEAKERS AND DISCUSSION LEADERS
`
`Diane J. Burgess, Ph.D., University of Connecticut, Co-
`Chair
`Ajaz S. Hussain, Ph.D., Food and Drug Administration,
`Co-Chair
`Thomas S. Ingallinera, Ph.D., InfiMed Therapeutics, Inc.,
`Co-Chair
`Hae-Young Ahn, Ph.D., Food and Drug Administration
`Mei-Ling Chen, Ph.D., Food and Drug Administration
`Yuan-yuan Chiu, Ph.D., Food and Drug Administration
`Todd Darrington, Ph.D., Pfizer, Inc.
`Eric Duffy, Ph.D., Food and Drug Administration
`Thomas H. Ferguson, Ph.D., Eli Lilly and Company
`Larry A. Gatlin, Ph.D., Biogen, Inc.
`
`Correspondence to:
`Diane Burgess
`Telephone: 860-486-3760
`Facsimile: 860-486-4998
`E-mail: diane.burgess@uconn.edu
`
`Anthony Huang, Ph.D., Alza Corporation
`Barbara B. Hubert, United States Pharmacopeia
`Andrew Janoff, Ph.D., The Liposome Company
`Funmi Johnson, Ph.D., Alkermes, Inc.
`Kofi Kumi, Ph.D., Food and Drug Administration
`Johnny Lau, Ph.D., Food and Drug Administration
`Patrick Marroum, Ph.D., Food and Drug Administration
`Frank Martin, Ph.D., Alza Corporation
`Marilyn Martinez, Ph.D., Food and Drug Administration
`Mehul Mehta, Ph.D., Food and Drug Administration
`Chien-Hua Niu, Ph.D., Food and Drug Administration
`Kanaiyalal R. Patel, Ph.D., Monsanto Company
`Atiqur Rahman, Ph.D., Food and Drug Administration
`Moo-Jhong Rhee, Ph.D., Food and Drug Administration
`Eyal S. Ron, Ph.D., eNOS Pharmaceuticals, Inc.
`Arthur J. Tipton, Ph.D., Southern BioSystems, Inc.
`David Young, Pharm.D., Ph.D., GloboMax USA
`Lawrence Yu, Ph.D., Food and Drug Administration
`Roger L. Williams, M.D., United States Pharmacopeia
`Helen Winkle, Food and Drug Administration
`Sui-Ming Wong, Ph.D., Novartis Pharmaceutical
`Corporation
`Jeremy C. Wright, Ph.D., Alza Corporation
`Liang Zhou, Ph.D., Food and Drug Administration
`
`ABSTRACT
`
`This is a summary report of the American Association of
`Pharmaceutical Scientists,
`the Food and Drug
`Administration and the United States Pharmacopoeia co-
`sponsored workshop on
`"Assuring Quality and
`Performance of Sustained and Controlled Release
`Parenterals." Experts from the pharmaceutical industry,
`the regulatory authorities and academia participated in
`this workshop to review, discuss and debate formulation,
`processing and manufacture of sustained and controlled
`release parenterals and
`identify critical process
`parameters and their control. Areas were identified
`where research is needed in order to understand the
`performance of these drug delivery systems and to
`assist
`in
`the development of appropriate
`testing
`procedures. Recommendations were made for future
`workshops, meetings and working groups in this area.
`ALL 2200 AAA
`PROLLENIUM V. ALLERGAN
`IPR2019-01505, et al.
`
`1
`
`

`

`AAPS PharmSci 2002; 4 (2) article 7 (http://www.aapspharmsci.org).
`
`
`
`INTRODUCTION
`
`This report summarizes the outcome of the workshop on
`"Assuring Quality and Performance of Sustained and
`Controlled Release Parenterals," which was held in April
`2001 in Washington, DC. This workshop was sponsored
`by
`the American Association of Pharmaceutical
`Scientists (AAPS), the Food and Drug Administration
`(FDA) and the United States Pharmacopoeia (USP). The
`overall goal of this workshop was to identify future
`directions for regulatory activity and public standards in
`the rapidly emerging area of controlled release (CR)
`parenteral products. Presentations focused on dispersed
`systems
`(microspheres,
`liposomes,
`gels
`and
`suspensions) as well as implants of small molecule and
`protein/peptide therapeutics for human and animal use.
`The objectives of the workshop were to:
`
`•
`
`•
`
`•
`
`•
`
`•
`
`•
`
`and
`processing
`formulation,
`Review
`manufacture of CR parenterals. Identify and
`discuss critical process parameters and their
`control.
`
`Identify new and emerging methods of in vitro
`release testing for CR parenterals and their
`ability to predict product performance.
`
`Discuss accelerated stability and in vitro release
`testing methods for CR parenterals.
`
`Discuss bioavailability, bioequivalence and
`pharmaceutical equivalence for CR parenterals.
`
`for
`the opportunity
`Explore
`correlation of CR parenterals.
`
`in vitro-in vivo
`
`Identify future directions for regulatory activity
`and public standards in this area.
`
`the
`from
`together experts
`This workshop brought
`pharmaceutical industry, the regulatory authorities and
`academia to discuss and debate issues pertaining to
`assuring the quality and performance of sustained and
`controlled
`release parenterals. The workshop was
`divided into formal presentations in the morning and
`parallel breakout discussion sessions in the afternoon.
`The breakout sessions served
`to
`identify
`future
`directions for regulatory activity and public standards in
`this rapidly emerging area. At the close of each breakout
`session
`the moderators were asked
`to prepare a
`summary of the key points discussed in their session.
`This report represents a compilation of these summaries
`together with background information explaining the
`need for regulatory activity in this area. Since many of
`the same concerns and issues were raised in different
`parallel sessions, this report is not divided by the
`breakout sessions, but rather by
`the key
`issues
`discussed.
`
`
`
`formulation,
`the workshop,
`first day of
`the
`On
`development and manufacture of the different products
`were reviewed and critical process parameters were
`identified. The breakout sessions focused on chemistry,
`manufacturing, and control issues and were divided by
`product (liposomes, microspheres, gels, suspensions
`and
`implants). The
`second day
`centered on
`biopharmaceutics issues, including physiology of the
`parenteral routes, bioavailability and bioequivalence, in
`vitro release testing and the possibility of in vitro-in vivo
`correlation.
`
`BACKGROUND
`
`Controlled release drug delivery systems are used to
`improve the therapeutic response by providing blood
`levels that are more consistent and stable compared to
`immediate release dosage forms. They can result in a
`reduction
`in adverse reactions since
`less drug
`is
`required and since the drug may be targeted to the site
`in vivo avoiding high systemic levels. As a consequence
`of targeted and controlled release, patient compliance
`may be improved due to lower dosing frequencies and
`simpler dosing regimens. With
`targeting and more
`sustained, predictable levels, efficacy may also be
`enhanced. CR parenteral drug delivery systems include:
`suspensions,
`liposomes, microspheres, gels and
`implants. Tiny microspheres and
`larger
`implantable
`devices can be used to modify release over periods of
`months to years. Suspensions, liposomes and gels may
`not achieve quite as long durations of action; however,
`they can be localized at the site of action in vivo and
`liposomes may achieve
`targeted delivery both by
`passive and active means
`following
`intravenous
`administration. These delivery systems are becoming
`increasingly utilized by the pharmaceutical industry to
`deliver drugs for treatment or prevention of a variety of
`diseases.
`
`Not all drugs are candidates for controlled delivery via
`the parenteral route. The candidate drug should be
`potent with known toxicity and pharmacokinetic profiles.
`A CR parenteral dosage form is usually selected when
`there are problems associated with oral delivery (e.g.
`gastric irritation, first pass effects or poor absorption)
`and a need for extended release or targeted delivery
`(e.g. rapid clearance). Both systemic and localized
`delivery can be achieved using CR parenterals. In
`addition,
`the drug must be compatible with
`the
`manufacturing process, which may be fairly harsh for
`some of
`these products. Examples of disease
`applications for CR parenteral delivery include: fertility,
`hormone therapy, protein therapy, infections (antibiotics
`and antifungals), cancer therapy, orthopedic surgery and
`post-operative
`pain,
`chronic
`pain,
`CNS
`vaccination/immunization,
`disorders,
`and
`immunosupression. Approved CR parenteral products
`are listed in Table 1.
`
`
`
`2
`
`

`

`AAPS PharmSci 2002; 4 (2) article 7 (http://www.aapspharmsci.org).
`
`Table 1 - Approved CR Parenteral Products
`
`?
`
`Trade Name Active Ingredient
`
`Approval
`Date
`
`?
`
`Suspension Products
`
`Depo-Medrol Methylprednisolone
`
`pre-1982
`
`Depo-Provera Medoxyprogesterone pre-1982
`
`Celestone
`Soluspan
`
`Insulin
`
`?
`
`Betamethasone
`
`pre-1982
`
`Lente
`NPH
`
`Unltralente
`
`pre-1962
`
`Microsphere Products
`
`Lupron Depot Leuprolide
`
`Sandostatin
`LAR
`
`Octreotide
`
`Nutropin Depot Somatropin
`
`Trelstar Depot Triptorelin
`
`?
`
`Liposome Products
`
`Doxil
`
`Daunorubicin
`
`Daunoxome
`
`Daunorubicin
`
`Ambisome
`
`Amphotericin B
`
`Depocyt
`
`Cytarabine
`
`?
`Lipid Complex Products
`
`Ambelcet
`
`Amphotericin B
`
`Amphotec
`
`Amphotericin B
`
`Visudyne
`
`Verteporfin
`
`?
`Implant Products
`
`Norplant
`
`Levonorgestrel
`
`Gliadel
`
`Zoladex
`
`Viadur
`
`
`Carmustine
`
`Goserelin
`
`Leuprolide
`
`1989
`
`1998
`
`1999
`
`2000
`
`1995
`
`1996
`
`1997
`
`1999
`
`1995
`
`1997
`
`2000
`
`1990
`
`1996
`
`1998
`
`2000
`
`Although CR parenteral products are relatively low
`volume in sales compared to oral products, they offer
`significant and distinct therapeutic advantages for certain
`types of drugs and consequently their use is becoming
`more
`prevalent. CR
`parenterals
`are
`complex
`formulations and thereby present significant challenges
`in regulation and the development of standards. In
`addition, they are considered ?high risk? products since
`they are complex, are designed for prolonged and
`targeted release and, in the case of dispersed system
`CR parenterals, are almost impossible to remove from
`the body once administered. Consequently, there is a
`
`
`
`3
`
`pressing need to open a public dialog between industry,
`FDA and USP on how best to assure the quality and
`performance of these products. This workshop served to
`initiate this public dialog.
`
`Of paramount importance is to identify any gaps in our
`scientific understanding of CR parenteral products and
`determine regulatory policy issues that need to be
`addressed. Critical formulation and process variables for
`individual products must be identified in order to develop
`the necessary characterization studies that undergird the
`substance, excipient, and product specifications that
`allow batch release. Key
`issues discussed
`in
`this
`workshop include: in vitro drug release testing (need for
`quality assessment as well as in vivo relevance), the
`possibility of in vitro-in vivo correlation, stability testing to
`ensure that specifications are met during shelf-life, as
`well as in vivo stability, sterility assurance, sterility
`testing, foreign particulate matter, particle size analysis,
`bioavailability
`and
`bioequivalence
`assessments,
`qualification of new biopolymers, residual solvent levels,
`reconstitution of parenteral products, and nomenclature.
`
`issues and recommendations
`The major
`workshop are summarized below.
`
`from
`
`this
`
`IN VITRO RELEASE METHODS
`
`Because the issue of in vitro release testing was raised
`at many of the breakout sessions, attendees generally
`agreed that an immediate need for guidance in this area
`exists. This guidance should focus on regulatory and
`compendial approaches with respect
`to acceptable
`apparatus, media and sampling methods, test intervals,
`and total percent release. Attendees also requested
`guidance on the method development process for in vitro
`tests for quality control purposes as well as on how to
`ensure the in vivo relevance of these tests. A need for
`guidance on accelerated in vitro testing for routine
`quality control purposes was also expressed. The issue
`of in vitro - in vivo correlation was discussed.
`
`Although workshop attendees did not want a single
`approach to be set for in vitro release testing given the
`wide range of CR parenteral products, they noted a need
`for general guiding principles and encouraged research
`to ensure a scientific basis for the development of
`different tests, procedures (to include apparatus) and
`acceptance criteria. These general approaches could
`then be modified, as appropriate, for specific products.
`For example, a given product may have specific
`requirements with respect to media, sampling interval or
`temperature.
`
`Apparatus
`
`Current USP apparatus for in vitro release testing are
`designed for oral and transdermal products and may not
`be optimal for controlled release parenteral products.
`USP apparatus 1 (basket) and 2 (paddle) were designed
`
`

`

`AAPS PharmSci 2002; 4 (2) article 7 (http://www.aapspharmsci.org).
`
`for immediate- and modified-release oral formulations.
`USP apparatus 5 (paddle over disc), 6 (cylinder) and 7
`(reciprocating holder) were designed for the transdermal
`route. USP apparatus 3 (reciprocating cylinder) and 4
`(flow through cell) were designed for extended-release
`oral formulations. These latter two methods may be the
`most relevant to CR parenterals and may be suitable
`following appropriate modification. Alternative apparatus,
`such as small sample vials and vessels, with and without
`agitation, are currently used
`for CR parenterals.
`Problems that may be associated with these alternative
`apparatus include: lack of sink conditions and sample
`aggregation.
`
`Research is required to determine the scientific basis for
`the
`tests, procedures,
`including apparatus
`(e.g.,
`geometry and hydrodynamics), and acceptance criteria
`for CR parenterals. The apparatus and media used
`should take into account the release mechanism and the
`physical properties of
`the product (e.g. size and
`stability). In addition, in vitro release tests must also
`discriminate between
`the performance of different
`formulation variants and
`ideally should have bio-
`relevance.
`
`Method development
`
`Attendees considered the purpose of in vitro release
`testing since method design may vary according to the
`purpose of the test. Current uses of in vitro release
`testing include: 1) formulation development, to include
`assessment of dose-dumping and in vivo stability (e.g.,
`Stealth-type liposomes, which should remain stable
`without significant drug release until uptake at the target
`site in vivo); 2) quality control to support batch release,
`3) evaluation of the impact of manufacturing process
`changes on product performance, 4) substantiation of
`label claims; and 5) compendial testing.
`
`Although in vitro release testing of CR parenterals is
`primarily utilized for quality control purposes, many
`attendees agreed that in vitro release tests should be
`developed with
`regard
`to clinical outcomes
`(bio-
`relevance). The rationale for this understanding is that
`the ultimate purpose of quality control testing is to
`ensure the clinical performance, i.e., efficacy and safety
`of the product. In order to achieve in vivo relevance,
`physiological variables at the site need to be considered
`including: body temperature and metabolism (both can
`significantly affect blood
`flow), muscle pH, buffer
`capacity, vascularity, level of exercise, as well as volume
`and osmolarity of the products. Any tissue response,
`such as inflammation and/or fibrous encapsulation of the
`product may need to be considered. In vitro release
`methods should be designed based on in vivo release
`mechanisms. With this understanding, attendees noted
`the following general approaches for in vitro test method
`design: 1) identification of release media and conditions
`that result in reproducible release rates; 2) preparation of
`formulation variants that are expected to have different
`
`
`
`4
`
`biological profiles; 3) testing of formulation variants in
`vitro as well as in vivo; and 4) modification of in vitro
`release methods
`to allow discrimination between
`formulation variants that have different in vivo release
`profiles.
`
`relevance of sink
`the
`Attendees also discussed
`conditions in in vitro test design for CR parenterals,
`considering that sink conditions may not exist at a
`particular in vivo site. General agreement was that sink
`conditions should be used for in vitro testing for quality
`control purposes provided that the study design allowed
`for discrimination between formulation variants with
`different in vivo release profiles. However, participants
`argued that non-sink conditions may be necessary if the
`purpose of the in vitro test is to establish in vitro-in vivo
`correlation (IVIVC). Although IVIVC is not utilized at
`present for CR parenterals, with sufficient bio-relevance
`built into the in vitro tests to support an IVIVC it may
`allow subsequent waiver of in vivo studies (see the
`IVIVC section below).
`
`Attendees also considered other issues, including the
`percent total release required (e.g., 70%, 80%) and the
`value of physical/chemical properties in lieu of release
`data for some quality control purposes (e.g., for stable
`liposomal formulations that are designed for no release
`until uptake at the site).
`
`Development of IVIVC for CR parenterals
`
`for all CR
`IVIVC may not be possible
`Although
`parenteral products, many attendees agreed that this is
`an important area for research. The principles used in
`IVIVC of oral extended-release products may be applied
`to parenterals with appropriate modification, justified on
`a scientific basis. IVIVC modeling and measurements
`may be different for different types of products (e.g.
`targeted release versus extended release products).
`Similarly, in vitro release methods and media are likely
`to vary depending on the product and should be
`developed based on in vivo relevance. For example, in
`vitro cellular tests may be acceptable as long as they are
`reproducible and can be validated. Similarly, in vivo
`measurements may vary and may
`include plasma
`concentrations, efficacy/safety data, surrogate endpoint
`data, as well as tissue concentrations. Discussions
`stressed that both in vitro and in vivo measurements
`must be justified scientifically. In the case of some
`products, such as liposomes, it may be necessary to
`measure
`in vivo concentrations of both
`free and
`encapsulated drug. Models
`that represent multiple
`processes (e.g., physical and biological) should be
`considered, as appropriate.
`
`The use of animals was considered to be acceptable to
`prove that an in vitro release system is discriminating.
`However, the use of animal models was considered
`inappropriate to prove an IVIV C for regulatory purposes.
`Instead, bio-relevance should be developed using
`
`

`

`AAPS PharmSci 2002; 4 (2) article 7 (http://www.aapspharmsci.org).
`
`clinical data. Nevertheless, IVIVC modeling using animal
`data would be suitable for "proof of principle" for initial
`research purposes. Research in this area should be
`encouraged, possibly coordinated
`through Product
`Quality Research Initiative (PQRI).
`
`The issue of data variability with respect to IVIVC was
`discussed and the following potential solutions were
`suggested:
`
`•
`
`•
`
`•
`
`the number of dosage units or
`Increase
`individuals.
`
`Variability may be acceptable as long as its
`source can be estimated and a valid IVIVC is
`obtained.
`
`If the source and importance of the variability
`can be determined,
`it may be possible
`to
`minimize it.
`
`Attendees noted that tissue responses, such as fibrous
`encapsulation, may affect release in vivo and this needs
`to be considered in establishing an IVIVC. However,
`these types of tissue response may be difficult to
`simulate in vitro.
`
`Use of animal models in release testing
`
`In the development of in vitro release methods, animal
`data may be used to obtain tissue distribution and
`pharmacokinetic information. Plasma levels may not be
`the best measure of in vivo behavior for CR parenteral
`products intended for local delivery or targeted release,
`and therefore, discussion in some sessions centered on
`the use of animal models to investigate in vivo product
`performance. More extensive bio-data can be obtained
`using animal models, including tissue levels at the local
`site. Animal models were considered to be invaluable
`and serial tissue samples might be used to compare
`product performance before and after manufacturing
`changes for CR parenterals with tissue-specific delivery.
`Although data will be useful in initial development,
`ultimately human data must be used to establish an
`IVIVC.
`
`Selection of an appropriate animal model was discussed
`and it was suggested that comparative studies be
`performed between
`injection sites
`in humans and
`animals in order to establish interspecies differences in
`drug release. Larger animals such as sheep and dogs
`may be more representative of humans with regard to
`interspecies differences than would small laboratory
`animals. This may be particularly important with regard
`to issues such as injection volume. Since inter-subject
`variability significantly
`impacts
`in vivo data,
`inbred
`animals may be useful in identifying variables that affect
`the drug release and absorption processes. Extensive
`inter- and intra-subject variability may mask critical
`formulation and manufacturing variables unless very
`
`
`
`5
`
`large human populations are utilized. The identification
`of an appropriate animal model for CR parenteral
`products was recommended as a research project,
`possibly for investigation through PQRI. The initial step
`of
`this research project should be a retrospective
`literature review of parenteral bioavailability data to
`develop initial correlation predictions between humans
`and animals. This research study should
`include
`different animals as well as different sites and should
`attempt to establish correlations between human and
`animal data
`relating
`the
`findings
`to physiological
`parameters. Different dosage forms and drugs should be
`investigated to determine whether the results are drug-
`and/or dosage form-dependant.
`
`in
`Animal models could potentially be utilized
`pharmaceutical development. For SUPAC-type changes,
`attendees
`recommended
`that an animal-human
`correlation be established so that animal models can be
`used (along with
`in vitro specifications)
`in
`lieu of
`extensive post-approval human trials. To achieve this,
`out of specification batches would be used to test the
`sensitivity of the animal model. Tests should also
`examine the sensitivity of the animal model to changes
`in product performance when the duration of testing is
`truncated (e.g., 3-month release testing for a one-year
`release product).
`
`Concerns were raised with respect to animal lifespan as
`well as physiological and metabolic differences between
`species. Animal lifespan may be a concern for extended
`release dosage forms with unusually long durations of
`action. Metabolic differences were considered not to be
`of importance for formulation comparisons. However,
`such differences may be very significant if animal
`models were to be used as a surrogate for efficacy.
`Another potential problem area is antibody production
`when using human derived proteins. Since immuno-
`suppression may be a possibility, the impact of this on
`pharmacokinetic and pharmacodynamic
`responses
`needs to be considered.
`
`Accelerated in vitro release testing
`
`The need for accelerated release testing was discussed,
`particularly for extended release products. Accelerated
`release testing is desirable for routine quality control
`purposes. Attendees generally agreed that these tests
`should have relevance to "real time" in vitro release tests
`conducted under conditions that simulate the in vivo
`situation as closely as possible. "Real time" in vitro tests
`for the full product duration should be conducted during
`product development and are essential for validating
`accelerated release rate tests. Accelerated tests should
`be bio-relevant and the mechanism of drug release
`should not be altered in accelerated tests, rather it
`should only be speeded up. For example, in the case of
`PLGA microspheres that release drug primarily via
`polymer erosion the accelerated test should speed up
`the polymer erosion process.
`In
`the design of
`
`

`

`AAPS PharmSci 2002; 4 (2) article 7 (http://www.aapspharmsci.org).
`
`factors such as polymer
`test,
`in vitro
`accelerated
`transition and degradation
`temperature should be
`considered to avoid any change in the mechanisms of
`drug release.
`
`release
`"burst"
`initial
`the
`Attendees discussed
`associated with some CR parenterals. When a CR
`parenteral delivery system produces an initial burst
`release, accelerated release tests should be augmented
`by an initial "real time" study that allows adequate
`assessment of this burst. Specifications for accelerated
`release
`tests should be
`tied
`to safety and
`to
`manufacturing experience. For example, significant
`deviation from the expected result may indicate a
`manufacturing problem. Attendees expressed the view
`that mathematical modeling to predict long-term release
`from accelerated release is useful.
`
`Labeling requirements for release rates
`
`that
`Attendees were concerned about products
`displayed an initial rapid burst release followed by a
`second slower release rate. Total drug cont ent and in
`vivo release rate are label requirements for CR products.
`Agreement was reached that inclusion of the initial burst
`release on the label should be handled on a case-by-
`case basis. Provided the initial burst rate is supported by
`clinical safety/efficacy data and
`is covered by
`specifications, it may not need to be included. However,
`if there are safety implications, this rate must be
`included. Given that regulatory guidance is not available
`on how much burst release is acceptable, attendees
`note
`that
`this performance
`factor should also be
`assessed on a case-by-case basis, depending on the
`drug and the safety/efficacy implications. Attendees
`discussed eliminating burst release from products where
`it did not provide a clinical benefit, but agreed that this
`might be prohibitively expensive.
`
`STABILITY
`
`Attendees considered both shelf life and in vivo stability
`for CR parenterals.
`In addition
`to drug stability,
`attendees noted the importance of "inactive" ingredient
`stability and product stability for CR parenterals.
`
`Shelf life stability
`
`The initial shelf life stability of entrapped drug is a
`concern because manufacturing conditions for some CR
`parenterals (e.g. some microsphere products) may be
`harsh. In the case of protein drugs, documentation of
`lack of chemical breakdown only is insufficient because
`conformational changes may have occurred that can
`affect activity. Therefore, activity must be demonstrated
`as
`part
`of
`the
`stability
`testing
`utilizing
`a
`pharmacodynamic method. Stability testing of many of
`these products requires extraction of the drug. The
`method of extraction (e.g., solvent system) should be
`selected to avoid any potential alteration in drug stability.
`
`
`6
`
`life stability should be conducted at room
`Shelf
`temperature as occurs for other products.
`
`In vivo drug stability
`
`In vivo drug stability is an issue for controlled release
`parenterals, especially those intended for long term
`extended release. In the case of large implants, drug
`stability could be determined by analyzing the drug
`remaining in explanted systems. This method could only
`be feasible for dispersed system CR parenterals by
`utilizing an appropriate animal model where tissue
`samples could be excised. An alternative approach that
`might be acceptable would be an in vitro test that
`simulated
`in vivo conditions
`(e.g., 37? C and
`approximately 100% humidity). Attendees agreed that
`some in vivo drug degradation might be acceptable for
`extended release products, provided that the product
`was demonstrated to be safe and effective. A typical in
`vivo degradation profile should be established together
`with safety and efficacy data. Immunologic response
`may require assessment, since protein degradation may
`occur with prolonged in vivo residence time.
`
`In vivo product stability
`
`Attendees agreed that in vivo product stability is equally
`as
`important
`as
`drug
`stability
`since
`degradation/alteration of the product as a whole or
`unwanted tissue response to the product may affect
`performance and bioavailabilty. For example, fibrous
`encapsulation of an implant or microsphere product will
`reduce/eliminate blood
`flow and consequently affect
`drug
`release
`and
`bioavailability.
`Attendees
`recommended that in vivo evaluation of CR parenterals
`include evaluation of product stability and
`tissue
`response to the product as well as drug stability. This is
`another area where animal models may be useful.
`Further discussion of this issue is required and/or a
`research project should be
`initiated
`to determine
`appropriate animal models.
`
`In vivo integrity of targeted products
`
`Another in vivo stability issue is the integrity of products
`that are intended to remain stable without significant
`drug release until uptake at the target site. An example
`of this type of product is "Stealth-type" liposomes. There
`was no consensus as to how to determine in vivo
`integrity of such products. Attendees discussed a
`proposal to conduct a single-dose study over a sufficient
`time period, with measurement of both encapsulated and
`unencapsulated drug. However, there was no prevailing
`opinion as to what percentage/ratio of unencapsulated to
`encapsulated drug should remain in the circulation for a
`liposomal drug product to be considered stable.
`
`

`

`AAPS PharmSci 2002; 4 (2) article 7 (http://www.aapspharmsci.org).
`
`PARTICLE SIZE
`
`Attendees requested guidance/clarification on particle
`size specifications for dispersed system CR parenterals.
`Particle size may affect release rates of extended
`release products, such as microspheres. It may also
`affect targeting ability and reticuloendothelial system
`(RES) uptake of liposome products. Acceptable particle
`size ranges may vary
`for different CR parenteral
`systems. For example, a liposome system intended for
`targeted release, where
`targeting
`is particle size-
`dependent, may require more stringent particle size
`specifications
`than some other dispersed systems.
`Attendees noted the importance of a specification for
`particle size range as well as average particle size since
`a few large particles may have a significant effect on
`product performance as well as safety. Larger particles
`can
`cause
`capillary
`blockage when
`injected
`intravenously. Particle size may also affect syringability
`of the product. Attendees requested guidance on particle
`sizing instrumentation and techniques, particularly when
`more than one instrument is necessary to measure the
`entire size range. Concluding, attendees generally
`agreed that a workshop to address particle size issues
`would be useful.
`
`STERILIZATION, STERILITY ASSURANCE AND
`FOREIGN PARTICULATE MATTER
`
`Sterilization and sterility assurance
`
`CR parenterals are complex products usually containing
`polymers and/or lipids with glass transition temperatures
`below the temperature required for hea

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket