throbber
HHS Public Access
`Author manuscript
`J Control Release. Author manuscript; available in PMC 2016 December 10.
`Published in final edited form as:
`J Control Release. 2015 December 10; 219: 644–651. doi:10.1016/j.jconrel.2015.09.052.
`
`In Vitro-In Vivo Correlation for Complex Non-Oral Drug
`Products: Where Do We Stand?
`
`Jie Shen and Diane J. Burgess*
`University of Connecticut, School of Pharmacy, Storrs, CT 06269
`
`Abstract
`In vitro -in vivo correlation (IVIVC) is a predictive mathematical model describing the
`relationship between an in vitro property and a relevant in vivo response of drug products. Since
`the U.S. Food and Drug Administration (FDA) published a regulatory guidance on the
`development, evaluation, and applications of IVIVC for extended release (ER) oral dosage forms
`in 1997, IVIVC has been one of the most important issues in the field of pharmaceutics. However,
`even with the aid of the FDA IVIVC Guidance, only very limited Abbreviated New Drug
`Application (ANDA) submission for ER oral drug products included adequate IVIVC data to
`enable the completion of bioequivalence (BE) review within first review cycle. Establishing an
`IVIVC for non-oral dosage forms has remained extremely challenging due to their complex nature
`and the lack of in vitro release methods that are capable of mimicking in vivo drug release
`conditions. This review presents a general overview of recent advances in the development of
`IVIVC for complex non-oral dosage forms (such as parenteral polymeric microspheres/implants,
`and transdermal formulations), and briefly summarizes the knowledge gained over the past two
`decades. Lastly this review discusses possible directions for future development of IVIVC for
`complex non-oral dosage forms.
`
`Graphical abstract
`
`*Address for Correspondence: Diane J. Burgess, Department of Pharmaceutical Sciences, School of Pharmacy, University of
`Connecticut, 69 North Eagleville Road U3092, Storrs, CT 06269-3092, USA. d.burgess@uconn.edu.
`Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
`customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of
`the resulting proof before it is published in its final citable form. Please note that during the production process errors may be
`discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Page 1
`
`SHIRE EX. 2077
`KVK v. SHIRE
`IPR2018-00290
`
`

`

`Shen and Burgess
`
`Page 2
`
`Keywords
`In vitro-in vivo correlation (IVIVC); non-oral; parenteral; prediction; in vitro release testing;
`modeling
`
`1. Introduction
`In vitro-in vivo correlation (IVIVC) is defined by the U.S. Food and Drug Administration
`(FDA) as “a predictive mathematical model describing the relationship between an in vitro
`property of a dosage form and a relevant in vivo response” [1]. Generally the in vitro
`property is the rate or extent of drug dissolution or release, while the in vivo response is the
`plasma drug concentration or amount absorbed. In the case of non-oral drug products (e.g.
`transdermal and ophthalmic dosage forms), an in vitro property could be in vitro drug
`permeation across the membrane of interest, while an in vivo property could be in vivo drug
`permeation. The history of IVIVC can be traced back to as early as 1950s, when
`pharmaceutical scientists attempted to correlate in vitro drug dissolution profiles of oral
`formulations with their respective in vivo pharmacokinetic profiles by means of
`mathematical modeling [2, 3]. In 1997, the U.S. FDA published a regulatory guidance
`related to the development, evaluation, and applications of IVIVC for extended release oral
`dosage forms. Since then, the establishment and application of IVIVC has increasingly
`gained more significance in the field of pharmaceutics. Generally, IVIVC can be categorized
`into five different levels: Levels A, B, C, D, and multiple Level C (Figure 1).
`•
`Level A represents a point-to-point relationship between in vitro and in vivo
`profiles. Generally the correlations are linear. However, non-linear correlations are
`also acceptable [4]. A Level A correlation is considered the most informative and is
`recommended by the U.S. FDA. It is also the only level of IVIVC that can be used
`to obtain biowaiver.
`Level B correlation utilizes the principles of statistical moment analysis. A mean in
`vitro dissolution time (MDTin vitro) is compared to either a mean in vivo residence
`(MRTin vivo) or dissolution time (MDTin vivo). Similar to a Level A IVIVC, a Level
`B correlation compares all in vitro and in vivo data available. However, since
`various in vivo release profiles may result in the same MRTin vivo or MDTin vivo, a
`Level B correlation is not considered to be a point-to-point correlation, and does
`not necessarily reflect the actual in vivo plasma profile and hence may lack
`sufficient predictability.
`Level C correlation establishes a single point relationship between a dissolution
`parameter (e.g. the time required for 50% dissolution, T50%) and a pharmacokinetic
`parameter such as Cmax, Tmax or AUC. Since it is based on a single point analysis,
`it is does not reflect the complete shape of the plasma concentration time curve,
`which is critical to define in vivo performance of a drug product. Accordingly, a
`Level C IVIVC is limited in predicting in vivo drug performance. Nevertheless,
`Level C correlations may be useful in the early stages of formulation development
`when pilot formulations are being selected.
`
`•
`
`•
`
`J Control Release. Author manuscript; available in PMC 2016 December 10.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Page 2
`
`

`

`Shen and Burgess
`
`Page 3
`
`•
`
`• Multiple Level C correlation relates multiple dissolution time points to one or more
`pharmacokinetic parameter(s) (e.g. Cmax, Tmax or AUC). A multiple Level C
`correlation should be based on at least three dissolution time points covering the
`early, middle, and late stages of the dissolution profile. A multiple Level C
`correlation can be as useful as a Level A correlation. However, if a multiple Level
`C correlation is obtainable, then the development of a Level A correlation should
`also be feasible and is more preferable.
`Level D correlation is a rank order correlation comparing in vitro and in vivo
`release profiles. A level D correlation is only qualitative and is not adopted in the
`U.S. FDA IVIVC Guidance.
`A meaningful IVIVC can be used to guide formulation and/or process development changes
`in the various stages of drug product development. In addition, an IVIVC can be used to
`support and/or validate the use of an in vitro dissolution method and can help set clinically
`relevant dissolution specifications to ensure product quality [5]. Most importantly, when a
`Level A IVIVC is established and validated, the in vitro release method can be used as a
`surrogate for bioequivalence studies when pre-approval and post-approval changes are
`required (e.g. formulation composition, as well as manufacturing process, equipment and
`site) [6–8]. Through the successful development and application of a meaningful IVIVC, the
`in vivo performance may be accurately predicted from the in vitro performance of drug
`products and therefore, human or animal studies can be minimized and the regulatory
`burden can be reduced [9, 10].
`
`Despite the publication of the FDA IVIVC guidance on ER oral dosage forms nearly two
`decades ago, only 14 ANDA submissions had IVIVC data, most of which were deficient and
`thereby, not acceptable [11]. Compared to the ER oral dosage forms, the establishment of an
`IVIVC for non-oral drug products (e.g. parenteral microspheres and implants, as well as
`transdermal and ophthalmic products) has been even more challenging due to their complex
`characteristics as well as the lack of standardized, compendial in vitro release testing
`methods [10]. In recent years, there has been significant interest within the pharmaceutical
`industry, academia, and regulatory agencies in developing suitable in vitro release testing
`methods as well as establishing IVIVCs for complex non-oral drug products. Notably, the
`U.S. FDA has funded over 20 research grants to advance in vitro equivalence methods for
`complex non-oral drug products and drug-device combinations in the past two years.
`Through collective and collaborative efforts in the field of pharmaceutics and drug delivery,
`some “ground-breaking” progress has been achieved. This review highlights recent advances
`in the development of IVIVC for complex non-oral dosage forms and briefly summarizes
`the knowledge gained over the past two decades. Lastly this review discusses possible
`directions for future development of IVIVC for these complex dosage forms.
`
`2. Current State-of-the-Art
`To date, there is no regulatory IVIVC guidance available for complex non-oral drug
`products. The same principles of developing IVIVC for ER oral dosage forms as detailed in
`the FDA IVIVC Guidance have been applied to develop IVIVC for various complex non-
`
`J Control Release. Author manuscript; available in PMC 2016 December 10.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Page 3
`
`

`

`Shen and Burgess
`
`Page 4
`
`oral dosage forms such as parenteral polymeric microspheres and implants [12–17],
`transdermal patches/gels [18, 19], as well as ocular inserts [20].
`
`2.1. Approaches to develop IVIVCs
`A Level A IVIVC is generally considered the highest level of correlation and is desirable
`from a regulatory point of view. Typically, developing a Level A IVIVC involves the
`following procedures (Figure 2): 1) obtaining formulations (preferably, three or more) with
`different release rates (e.g. slow, medium, and fast) or using one formulation if its in vitro
`dissolution is independent of dissolution testing conditions (e.g. pH, media, and agitation);
`2) obtaining in vivo plasma concentration profiles or in vivo dissolution profiles of the
`selected formulations; 3) estimating in vivo absorption or dissolution time course of each
`formulation using an appropriate deconvolution technique (e.g. model-dependent, and
`model-independent numerical) (Table 1); 4) establishing a correlation/relationship between
`the estimated fraction in vivo release/absorption and the faction in vitro release, using a
`linear (preferably) or non-linear model (e.g. Sigmoid, Hixon-Crowell, Weibull, Higuchi, and
`Logistic) [21]; and 5) evaluating the predictability of the developed IVIVC internally and/or
`externally. Based on the FDA IVIVC Guidance, an average percentage prediction error (%,
`PE) of 10% or less for pharmacokinetic parameters of interest (e.g. Cmax or AUC)
`establishes the predictability of a developed IVIVC. When developing a Level A IVIVC,
`there may be disparity between deconvoluted in vivo and in vitro dissolution profiles due to
`the intrinsic difference between in vitro and in vivo dissolution conditions. Accordingly,
`time shifting/scaling may be utilized to allow the deconvoluted in vivo data to be on the
`same time scale as the in vitro dissolution data, which in turn makes it possible to establish a
`correlation/relationship between in vitro and in vivo release data.
`
`Although a Level A IVIVC is most informative and recommended by the U.S. FDA, other
`levels of IVIVC (e.g. multiple Level C, and Level B) can be helpful to assure product
`quality, and to assist in formulation development. When developing a Level B IVIVC, at
`least three formulations are required. Based on the principles of statistical moment analysis,
`a mean residence time (MRTin vivo), mean absorption time (MATin vivo), or mean in vivo
`dissolution time (MDTin vivo) is calculated and related to a mean in vitro dissolution time
`(MDTin vitro) (Figure 3A). All parameters determined are model-independent. In the case of
`developing a multiple Level C correlation, one or more pharmacokinetic parameters (e.g.
`Cmax, Tmax or AUC) are correlated with at least three dissolution time points covering the
`early, middle, and late stages of the dissolution profile. Based on the U.S. FDA IVIVC
`Guidance, the recommendations for assessing the predictability of Level C correlations
`depend on the type of application for which the correlation is to be used. The methods and
`criteria for assessing the predictability are the same as that for Level A correlations
`described above.
`
`The development of IVIVCs for non-oral drug products is a complicated process, due to not
`only their complex characteristics (e.g. multi-phasic release) but also the lack of suitable in
`vitro release testing methods. Despite that extensive efforts have been devoted in this area,
`there are only a few literature reports on the establishment of IVIVCs for these drug
`products based on multiple formulations, albeit with different in vitro release testing
`
`J Control Release. Author manuscript; available in PMC 2016 December 10.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Page 4
`
`

`

`Shen and Burgess
`
`Page 5
`
`methods such as USP apparatus 4 methods [15, 16], dialysis membrane methods [14, 22],
`and Franz diffusion cells [18–20] (Table 2).
`
`2.2. IVIVCs for parenteral polymeric microspheres/implants
`Parenteral polymeric microspheres/implants, particularly poly(lactic-co-glycolic acid)
`(PLGA)/poly(lactic acid) (PLA)-based microsphere/implant drug products have been one of
`the most successful complex non-oral polymeric drug products on the market. The PLGA/
`PLGA-based microsphere/implant drug products are biodegradable, biocompatible, and
`possess the capability of delivering a variety of therapeutics (e.g. small molecules and
`biologics) in a controlled manner over periods of days to several months [33–36]. These ER
`parenteral drug products normally contain substantial amounts of potent therapeutics.
`Therefore, it is critical to assure consistent product performance and safety through in vitro
`quality control tools such as discriminatory in vitro release testing methods, as well as
`reliable IVIVCs or in vitro-in vivo relationship (IVIVR) in the event that an IVIVC is not
`feasible. Over the past two decades, the development of IVIVCs for polymeric
`microspheres/implants has received the most attention, as a result of which considerable
`progress has been achieved (Table 2). However, most reported literature are “proof-of-
`concept” research that only demonstrated the possibility of developing point-to-point linear
`correlations or Level B correlations based on one formulation. Encouragingly, Level A
`IVIVCs established using two or more microsphere formulations with different release
`characteristics have recently be presented [14, 16, 22, 23]. It should be noted that multiple
`formulations with different release characteristics are essential to develop a reliable IVIVC.
`
`One of the most challenging aspects of developing IVIVCs for complex microsphere/
`implant drug products is to design in vitro release studies in such a way that the in vivo
`behavior of these products is reflected as much as possible. PLGA/PLA-based polymeric
`microspheres/implants are normally administrated into subcutaneous or muscular tissues or
`directly injected into local areas (e.g. knee joints). Following injection/implantation,
`therapeutics are slowly released from microspheres/implants into the tissue fluids via
`complex release mechanisms (e.g. diffusion, polymer erosion or a combination thereof) [37,
`38], and are subsequently transported into the systemic blood circulation system via
`diffusion and/or convective processes [39–41]. Due to the lack of compendial in vitro
`release methods, various in vitro release methods (e.g. sample-and-separate [23, 26, 27],
`membrane dialysis [14, 22], and flow through [15, 16]) have been utilized to determine in
`vitro drug release characteristics and to develop IVIVCs. Although it is feasible to develop
`IVIVCs for parenteral microspheres/implants based on a simple sample-and-separate
`method [17, 23–25], there are limitations associated with this method such as poor
`hydrodynamic conditions, loss of product (e.g. microspheres) during sampling as well as
`inability to mimic different in vivo drug release conditions. For example, the presence of the
`in vivo boundary layers as well as the small interstitial fluid volume available for drug
`release at the administration sites. It has been reported that the correlation/relationship
`between the in vitro and in vivo data of huperzine microspheres was sensitive to the route of
`administration. Additionally, the sample-and-separate method appeared to better reflect drug
`release from PLGA microspheres in muscular tissues compared to that in subcutaneous
`tissues, thus a better correlation was obtained for the intramuscular route [23]. Compared to
`
`J Control Release. Author manuscript; available in PMC 2016 December 10.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Page 5
`
`

`

`Shen and Burgess
`
`Page 6
`
`the sample-and-separate method, membrane dialysis and flow though (USP apparatus 4)
`methods have more complex apparatus setup. However, they appear to be more capable of
`mimicking in vivo drug release conditions of parenteral microspheres/implants (e.g.
`microspheres/implants are exposed to a limited volume of release media at a time).
`Accordingly, these two release methods may be more suitable to develop a Level A IVIVC
`for parenteral microspheres/implants. A Level A IVIVC based on three olanzapine
`microsphere formulations with different release rates (i.e. fast, medium, and slow) has been
`recently reported using a membrane dialysis method [13]. In our ongoing research, we have
`successfully developed a Level A IVIVC using different combinations of three risperidone
`microsphere formulations that are compositionally equivalent but prepared using different
`manufacturing processes. Notably, both external predictability and robustness (IVIVCs not
`affected by the formulation combinations) of the developed IVIVC have been validated.
`Compared to the sample-and-separate method, the USP apparatus 4 method demonstrated
`not only better discrimination against compositionally equivalent microsphere formulations
`with manufacturing differences, but also better predictability of the in vivo PK profiles
`obtained from the animal study using a rabbit model. More importantly, the USP apparatus 4
`method demonstrated better predictability of the in vivo initial burst release phase, whereas
`the sample-and-separate method was not able to discriminate the initial burst release phase.
`Reliable detection of the in vitro initial burst can be important, especially in regulatory
`applications when two formulations are being evaluated for equivalence.
`
`To help understand drug release mechanisms and guide the establishment of IVIVCs for
`polymeric microspheres/implants, different mathematical models (e.g. Higuchi, and
`Weibull) have often been used (Table 3). These mathematical models assume zero
`dissolution at time zero and complete dissolution at sufficient time t. By introducing a
`parameter that represents the degree of dissolution, these mathematical models can easily be
`modified to account for incomplete dissolution (if any). For example, a Level A IVIVC was
`established for buserelin implants, from which drug release can be described using the
`Higuchi model [17]. Interestingly, when buserelin release from implants was governed by a
`combination of diffusion, dissolution as well as erosion, a Level B IVIVC appeared more
`suitable [17].
`
`Another challenging aspect of developing IVIVCs for complex microspheres/implants is
`how to deconvolute in vivo data and correlate that with in vitro release data. Due to their
`complex release characteristics (e.g. bi- or tri-phasic release profiles), it may be very
`difficult to correlate deconvoluted in vivo data with multi-phasic in vitro release data using a
`simple mathematical model. In some cases, it may not be possible to predict the initial in
`vivo drug release based on the in vitro release data from the burst release phase, since the
`rate-limiting step for the initial in vivo drug availability may actually be drug permeation
`across the tissue barriers. Although an IVIVC may still be developed using post-burst in vivo
`and in vitro release data, the “post-burst” IVIVC may have limited prediction capability
`[26]. Furthermore, pharmacokinetic parameters of testing dosage forms are normally needed
`when using the deconvolution approach. For drugs that fit into a two-compartment model,
`an extra in vivo study (intravenous administration) is required in order to perform
`deconvolution. Accordingly, in order to simplify the development of IVIVCs and avoid the
`
`J Control Release. Author manuscript; available in PMC 2016 December 10.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Page 6
`
`

`

`Shen and Burgess
`
`Page 7
`
`use of pharmacokinetic parameters, other approaches (e.g. fractional AUC) have been
`reported [13, 14, 23, 29]. The factional AUC is determined by dividing cumulative AUC at
`time “t” with cumulative AUC0-t and plotting along with the percent drug release in vitro. In
`a manner similar to the deconvolution approach, the in vitro and in vivo drug release data are
`then compared. It has been demonstrated that in vivo profiles of PLGA microspheres
`obtained using both the Wagner-Nelson and fractional AUC methods were nearly
`superimposable, suggesting that fractional AUC could be used as an alternative to the
`commonly used Wagner-Nelson method [14, 22]. However, it should be noted that these
`methods are not adopted in the FDA IVIVC Guidance and therefore, may not be useful for
`regulatory purposes. In addition to comparing the drug absorption percentage with the
`amount of drug released at different time points, the time for 0% to 90% absorbed in vivo
`with the time for releasing the same amounts of the drug in vitro (Levy plot, Figure 3B) has
`been compared and good linear correlation was shown [17, 30].
`
`Considering that real-time in vitro release testing of parenteral microspheres/implants
`normally requires extended periods of time, it may be necessary to develop IVIVCs based
`on discriminatory accelerated in vitro release tests. To this end, a relationship between
`accelerated and real-time in vitro release data should be established such that the accelerated
`in vitro test could maintain “bio-relevance”. Ideally, drug release from real-time and
`accelerated tests should follow the same release mechanism with a one-to-one correlation
`between the release profiles [42]. However, it is possible that the drug mechanism(s) may
`change since accelerated release tests are typically performed under extreme conditions (e.g.
`high temperatures, extreme pH conditions) [43]. Nevertheless, developing an IVIVC based
`on accelerated testing may still be feasible as long as all microsphere/implant formulations
`experience similar changes and their release characteristics can be differentiated. The
`possibility of developing an IVIVC based on accelerated testing for PLGA microspheres has
`recently been demonstrated using commercial Risperdal® Consta® [15]. Despite that the
`accelerated in vitro release profiles obtained at elevated temperatures (50°C and 54.5°C) did
`not show a good linear correlation with the real-time in vitro release profile, a one-to-one
`linear correlation between the accelerated in vitro data and the time scaled in vivo data of
`Risperdal® Consta® was shown [15].
`
`Physiologic responses (e.g. foreign body response) to biomaterials is another important issue
`that must be considered when developing a reliable IVIVC as they may result in polymer
`degradation mechanism changes in vivo. For instance, acidic PLGA degradation products
`may accumulate at the local sites and hence lower the pH in the interstitial space
`immediately surrounding the microspheres. This may result in a change in the degradation
`mechanism of PLGA microspheres from bulk erosion to surface erosion, thus accelerating
`polymer degradation with subsequent increased drug release in vivo [16, 44]. On the other
`hand, chronic inflammation in response to the presence of microspheres in the interstitial
`site, and fibrosis may form and isolate microspheres, thus slowing down drug absorption/
`drug release in vivo [45]. The formation of fibrosis was speculated to be responsible for the
`slower in vivo release/absorption of Risperdal® Consta® 30 days following administration to
`humans [15]. Interestingly, it was noted in our recent research that Risperdal® Consta®
`demonstrated faster drug release in rabbits compared to in vitro real time release. This
`
`J Control Release. Author manuscript; available in PMC 2016 December 10.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Page 7
`
`

`

`Shen and Burgess
`
`Page 8
`
`suggests that differences in drug absorption and drug release between animals and humans
`must be taken into consideration, and an IVIVC study based on animal data may not be fully
`extrapolated to humans.
`
`2.3. IVIVCs for transdermal dosage forms
`Transdermal drug delivery systems (TDDS) are one of the first generation controlled release
`drug products that appeared on the market [47, 48]. Since the first transdermal drug product
`(Transdermal Scop®) was approved by the U.S. FDA in 1979, there have been different
`generations of transdermal drugs products (e.g. passive transdermal patches, and
`iontophoretic transdermal devices) developed and commercialized [48, 49]. Unfortunately,
`unlike parenteral microspheres/implants, the development of IVIVCs for TDDS has not yet
`been given substantial attention. To date, there are only very few literature reports on
`IVIVCs for TDDS (Table 2).
`
`Transdermal drug delivery involves a few consecutive steps: i) drug release from the
`formulation; ii) drug penetration/diffusion into/through skin; and iii) drug arriving at the site
`of action to trigger a pharmacological response. In most cases, drug penetration/diffusion
`through the skin is the rate-limiting step, which can be described using different
`mathematical models (Table 4). The development of IVIVC for TDDS is somewhat
`different compared to that of parenteral microspheres/implants. One of challenges to
`develop an IVIVC for TDDS is to mimic the process of drug permeation across human skin
`as much as possible. Various in vitro dissolution methods (e.g. apparatus 5 (paddle over disk
`method), apparatus 6 (rotating cylinder method), and apparatus 7 (reciprocating holder
`method)) have been recommended as quality control tools for transdermal/topical drug
`products (such as transdermal patches and films) [50]. However, these dissolution methods
`may not reflect complex mechanism(s) of drug permeation/diffusion across skin.
`Accordingly, instead of in vitro dissolution, in vitro skin permeation is more often used for
`the development of IVIVCs for TDDS [18, 19].
`
`Franz diffusion cells are the most widely used apparatus for determining in vitro drug
`permeation of TDDS and for the development of IVIVCs. Franz diffusion cells consist of a
`donor compartment and receptor compartment with the membrane of interest (e.g. excised
`human or animal skin) mounted as a barrier between the two compartments. Due to the
`inherent variability in absorption between individuals and between anatomical sites (e.g.
`abdominal vs forearm skin), it is important to control for skin source and viability, as well as
`to evaluate in vitro permeability across skin from several donors in cases where in vitro skin
`permeation data is correlated [51]. Excised human skin has been demonstrated to be the
`most appropriate in vitro skin model that may potentially be used as a surrogate for more
`costly and time-consuming in vivo bioequivalence studies [52]. Other skin models (such as
`excised porcine skin [19] and rat skin [32]) have also been used for in vitro drug permeation
`testing, and for the development of IVIVCs.
`
`Generally IVIVCs for transdermal drug products could be categorized into the same levels
`as those described in the FDA IVIVC Guidance for extended release oral dosage forms (i.e.
`Levels A, B, C, and multiple C). However, since in vitro dissolution is not commonly used
`for the development of IVIVCs, Level B (which uses mean dissolution time) and Level C
`
`J Control Release. Author manuscript; available in PMC 2016 December 10.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Page 8
`
`

`

`Shen and Burgess
`
`Page 9
`
`IVIVCs may not be applicable for TDDS [51]. A Level A IVIVC that correlates in vitro
`drug permeation across skin with in vivo drug permeation or absorption is more desirable.
`Similar to parenteral microspheres/implants, different deconvolution techniques (Table 1)
`can be utilized to deconvolute in vivo data and correlate with in vitro drug permeation data.
`In some cases, the steady state flux in vitro is extrapolated to determine the in vivo plasma
`concentration using pharmacokinetic modeling [53]. Most recently, a Level A IVIVC (a
`second order polynomial correlation) between the in vitro permeation percent and the
`deconvoluted in vivo absorption percent was established using three estradiol TDDS [18]. In
`this study, the Wagner-Nelson method was used to deconvolute human pharmacokinetic
`data obtained from the literature. The developed Level A IVIVC was validated internally
`and externally and showed less than 15% PE for both Cmax and AUC.
`
`In order to facilitate the development of a good in vitro predictive model for TDDS, some
`improvements on current in vitro testing apparatus and in vivo analytical techniques have
`been reported. An inherent problem with Franz diffusion cells is the lack of
`microvasculature, which is present in the in vivo environment and helps in rapid clearance of
`the drug. To overcome this, flow-through diffusion cells have been designed in such a way
`that the receptor buffer is continuously removed to help maintain the sink conditions in vitro
`[54], which may be more relevant to the actual in vivo conditions and hence, could
`potentially benefit the development of IVIVCs for TDDS. In addition, new technologies
`(e.g. microdialysis and Confocal Raman Spectroscopy (CRS)) have been implemented to
`obtain in vivo drug adsorption data in a continuous fashion [31, 32]. For example, the real-
`time drug disposition in skin was monitored using CRS and correlated with in vitro human
`skin permeation data obtained using Franz diffusion cells. In this “proof-of-concept” study,
`a good correlation was obtained between the in vitro flux of niacinamide and signal intensity
`of niacinamide permeated into the mid ventral forearm at 4 µm in vivo after a 30 min
`application [31].
`
`3. Future perspectives
`Despite the encouraging progress that has been made over the past two decades, the
`development and application of IVIVCs for complex non-oral dosage forms still remains at
`an infant stage. At present, there is sparse or no literature reports on IVIVC for most
`complex non-oral dosage forms (e.g. parenteral nanoparticulate systems, and ophthalmic
`dosage forms). With the ongoing commercialization of novel and generic complex non-oral
`drug products, it is essential to initiate the development of IVIVCs to help assure the product
`performance and safety, as well as to assist in product development in a timely and cost-
`effective fashion.
`
`One of the biggest hurdles yet to conquer in developing IVIVCs for complex non-oral drug
`products is the dearth of bio-relevant in vitro dissolution methods that are capable of
`reflecting the complex and dynamic in vivo environment these dosage forms are
`encountered. This is particularly the case for nanoparticulate systems such as liposomal
`products, as well as ophthalmic drug products. Liposomal formulations have often showed a
`poor correlation between their in vitro and in vivo performance, due to their possible
`multiple fates in the blood circulation [55]. For example, “stealth” liposomal drug products
`
`J Control Release. Author manuscript; available in PMC 2016 December 10.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Page 9
`
`

`

`Shen and Burgess
`
`Page 10
`
`are designed to be stable with no (or very little) drug release prior to reaching their target
`organs and cells (e.g. Doxil®), while other liposomal drug products are designed to provide
`

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