SlideShare a Scribd company logo
2014.09.30. Bioavailability Enhancement Webinar Series: Optimizing Technology Choice to
Enhance Bioavailability
Capsugel (Bend Research)
Abstract:
An increasing number of active compounds in pipelines today have properties that require
functional formulation to enable exposure and efficacy. Despite many new technology choices, it
is often difficult to match the right drug-delivery technology to a given molecule and problem
statement. This problem is exacerbated by the need to save time and valuable drug in early
development. This webinar describes an efficient strategy for mating enabling drug-delivery
technologies with problem statements based on challenging compound properties and product
concepts, building on an understanding of gut physiology, key molecule physicochemical
properties, and the target product profile.
Company Summary:
Capsugel Dosage Form Solutions designs, develops and manufactures innovative dosage forms
addressing bioavailability and other pressing product development challenges, including
bioavailability enhancement, modified release, abuse deterrence, biotherapeutic processing, and
inhalation formulation.
Speakers Bio:
Dr. David Vodak
Vice PresidentBend Research Moderator
Dr. Vodak's areas of expertise are research and development of novel pharmaceutical drug-
delivery systems. Dr. Vodak holds a PhD in materials chemistry from the University of
Michigan and a B.A. in chemistry from Willamette University.
Dr. David Lyon
Senior Vice PresidentBend Research
Dr. Lyon is the Senior Vice President at Bend Research. He leads development activities for new
technologies, oversees the development of predictive biomodels, and provides technical
leadership to the research groups for new and applied technologies.
Science-Based Technology Selection
And Formulation Development
For Oral Bioavailability Enhancement
Capsugel Dosage Form Solutions
White Paper
Authors: Hywel Williams, Michael Morgen, Eduardo Jule, Jan Vertommen,
Hassan Benameur, Dwayne Friesen and David Vodak
Capsugel Dosage Form Solutions White Paper August 2014
Page 2
Science-Based Technology Selection and Formulation
Development For Oral Bioavailability Enhancement
Hywel Williams, Michael Morgen, Eduardo Jule, Jan Vertommen, Hassan Benameur, Dwayne Friesen and David Vodak
Capsugel
Introduction
The increasing fraction of poorly water-soluble
compounds in pharmaceutical discovery is leading
to significant growth in the use of enabling
technologies to improve oral drug absorption and
bioavailability (BA). Commonly used technologies
in this area have been extensively reviewed (1)
and include salt selection, cocrystals, amorphous
solid dispersions, particle size reduction,
cyclodextrins, amorphous/lipid micro- and
nanoparticulates, adsorbents and lipid-based
technologies. Many of these technologies have
been shown to enhance drug BA, with most
commercial products utilizing solid amorphous
dispersion, nanocrystalline drug or lipid-based
technologies. Examples include Neoral®
(cyclosporine, Abbott), a lipid-based liquid-filled
capsule; Incivek® (teleprevir, Vertex), an
amorphous drug dispersion produced by spray
drying; Kaletra® (lopinavir and ritonavir, Abbott)
an amorphous drug dispersion produced by hot-
melt extrusion (HME); and Emend® (aprepitant,
Merck), a nanocrystal-containing tablet.
Increasing use of such enabling technologies will
be driven by the need to deliver the estimated
40% to 70% of the NCE pipeline candidates that
are poorly water-soluble. Enabling technologies
are also widely explored in the 505(b)(2) product
pathway to reformulate existing products on the
market into products that are better performing
(e.g., “super generics”) or during the product
patent life through life-cycle management
approaches. Drivers toward the 505(b)(2)
regulatory pathway include faster time to market,
lower development costs by avoiding certain costly
and repetitive preclinical and clinical trials, and 3
to 5 years of market exclusivity dependent upon
the extent of change to the previously approved
drug. One example of a marketed 505(b)(2)
product is Absorica™ (Ranbaxy), a hard capsule
product containing a lipid formulation. This
product, provides higher drug absorption in the
fasted state than the original Roaccutane®/
Accutane® (Roche) softgel product, thus offering
patients the potential to benefit from acne
treatment independently from meals (2) and
granting Ranbaxy the aforementioned benefits of
a 3-year period of market exclusivity.
Due to the wide applicability of enabling
technologies to NCEs and off-patent drugs, the BA
enhancement landscape is innovative, dynamic
and diverse. Indeed, the formulation of poorly
water-soluble drugs is a key focus for many
contract research/development/manufacturing
organizations (CROs/CDMOs), supporting drug
Capsugel Dosage Form Solutions White Paper August 2014
Page 3
development work with one or more BA-enhancing
technology approaches to advance such drug
candidates. A much smaller number of companies
have both a broad range of technologies and the
capacity to implement and scale them from
design and development to commercial scale
production. Having the ability to understand and
provide multiple technology or formulation
approaches under one roof is extremely
advantageous, since the need to partner with
multiple companies during a drug development
program results in higher costs, significant
program delays and inefficiencies and increased
risk in the development process.
Optimal application of enabling technologies is
based on key principles, including the following.
• The diverse needs of all drug compounds
currently in development across and within
pharmaceutical companies cannot be
addressed by a single enabling technology.
• Development success is more probable if a
technology is appropriately matched to the
compound properties and product needs early
in the development process.
• In many cases, more than one technology can
be utilized successfully and commercial
considerations such as desired dosage format
can play a decisive role.
Using a technology ill-suited to a compound or
problem statement often results in development
delays, additional costs or even failure, due to
poor manufacturability, stability, performance, or
shortcomings in some other aspect of the target
product profile. Appropriate application of
technology is therefore critical to achieve success
for development projects where achieving
adequate oral absorption is required. Effective
application of technology for enabled formulations
can remain elusive, since it relies on many inputs,
not the least of which is expertise with a range of
alternative or complementary technologies,
involving a clear understanding of the
fundamental science governing the mechanisms
of drug solubilization, absorption and metabolic
fate.
The purpose of this article is therefore to highlight
key physicochemical and biological obstacles to
drug exposure following oral administration and
how effective use of formulation technology relies
on an understanding of the drivers to oral BA. We
will then discuss the formulation development
tools that have been developed from a deep
investigation of key technologies and leveraging
experience of hundreds of BA-enhancement
projects.
Physicochemical Obstacles to Oral BA
Physicochemical obstacles to oral drug BA include
low aqueous solubility (a thermodynamic and
form-dependent property) and a slow rate of
dissolution (a kinetic property). The drug
concentration gradient from the intestinal lumen
across the unstirred mucus layer and into the
intestinal wall is the driving force for passive
absorption of drugs. Low aqueous solubility of a
drug can therefore limit this gradient and result in
low absorption from the intestine. A slow rate of
dissolution can also limit absorption, particularly
where the solubility of the drug form is sufficiently
Capsugel Dosage Form Solutions White Paper August 2014
Page 4
low that it is necessary to maintain the
concentration of drug near its solubility limit in
order for drug absorption to be complete over the
limited time that the drug transits the GI tract.
Low drug solubility is a property common to drugs
that are in Class II and IV of the
Biopharmaceutical Classification System (BCS).
Factors underpinning the property of low solubility
are well described (3) and include:
• A high crystal lattice energy (which generally
increases with increasing melting temperature
of a compound) and results in low solubility in
essentially all solvents, sometimes referred to
as “brick dust”;
• a low energy of aqueous solvation (which
generally decreases with increasing Log P
value of a compound, i.e., lipophilicity), often
referred to as “greaseball” compounds; and
• a combination of both, where the impact of a
high crystal energy on solubility is exacerbated
by a low solvation energy.
Enabling technologies increase solubility and
dissolution rate by reducing the drug lattice
energy, increasing drug surface area, or
increasing the energy of solvation. For example,
lipids, surfactants and cosolvents increase the
volume and character of hydrophobic micro-
phases of GI fluids, such as vesicles and micelles.
Many low solubility compounds have favorable
intermolecular interactions with such hydrophobic
colloids, leading to increased drug solubilization.
Nanocrystals enhance the dissolution rate by
increasing the drug surface area and may
increase drug solubility if particles are very small
(~<100 nm) and/or show change in crystalline
structure, particularly at the crystal surface. Spray
drying and HME solid dispersions increase
apparent drug solubility and, therefore, dissolution
rate by molecularly dispersing a high energy
amorphous form in a matrix material (4). On the
other hand, lipid-based technologies are effective
in augmenting drug solubility as dispersed and
digested lipid components mix with endogenous
bile salts and phospholipids to form a range of
colloidal species such that the dissolving “solvent”
is more favorable to the drug (i.e., “like dissolves
like”) (5).
In many cases, technology approaches have the
capacity to increase drug solubility through both
solid-state and solvation effects. For example, the
introduction of a counterion or conformer in salts
and cocrystals, respectively, can increase
solubility in two ways: first, by altering both the
solid-state energy through changes in molecular
packing in the crystal; and second, by increasing
the solvation energy by changing the nature of the
local solvent, i.e., by changing pH in the case of a
salt counterion, or by changing the drug to the
ionized form (1). In addition, solid dispersions that
use amphiphilic polymers such as hydroxypropyl
methylcellulose acetate succinate (HPMCAS) (6)
or nonionic surfactants (7) may also affect
solvation. Finally, predissolving a drug within a
lipid-based formulation can eliminate the solid-
state obstacles to solubility and dissolution and, if
properly formulated, will maintain the compound
in solution throughout the GI tract (albeit, with a
high proportion of the drug solubilized in a
Capsugel Dosage Form Solutions White Paper August 2014
Page 5
colloidal state rather than in the aqueous phase of
the GI fluid).
Figure 1 matches compound solubility/
dissolution obstacles to formulation technology,
which forms the foundation of a science-based
technology selection process. Where low solubility
stems primarily from a high crystal lattice energy,
solubility will benefit most from a reduction in
solid-state interactions (e.g., solid dispersions)
while those compounds that show limited affinity
for aqueous solvents would benefit most from
approaches that enrich the GI environment with
exogenous solubilizers (e.g., lipid-based
formulations). This relatively simple differentiation
based on the physicochemical properties of the
drug, while well recognized, is often overlooked in
utilizing what is known, available and, in some
cases, proprietary. As discussed throughout this
article, technology selection and formulation
development based on scientific understanding of
mechanistic barriers to absorption is likely to
result in more rapid and successful development
with reduced costs.
Biological Obstacles to BA
In some cases, it is necessary to overcome not
only physicochemical obstacles to absorption, but
also biological barriers, which include (8):
• Efflux of absorbed drug back into the
intestinal lumen (often P-gp or BCRP
transporter mediated);
Figure 1: Simplified diagram illustrating the principal mechanisms by which various enabling technologies increase
drug solubility/dissolution rate to lead to improved oral BA. See the supporting text for a more detailed description of
some of these enabling technologies.
Capsugel Dosage Form Solutions White Paper August 2014
Page 6
• presystemic drug metabolism in the intestine
(principally via cytochrome P450 enzymes);
and
• extensive hepatic first-pass drug metabolism.
A good example of high drug absorption
accompanied by low BA is that of testosterone.
(<25 µg/ml), testosterone is well absorbed from
the intestine, but shows extremely low BA due to
extensive first-pass metabolism (9). Thus,
formulation work to alter drug physicochemical
properties to improve intestinal absorption would
be ineffective to improve BA in this case. Certain
enabling technologies have the capacity to
attenuate these biological obstacles to drug BA,
particularly by reducing efflux and metabolism in
the intestine. Indeed, fatty acids and nonionic
surfactants (typically polyethoxylated esters/
ethers of oils/fatty acids) commonly used in lipid-
based technologies have frequently been shown
to inhibit P-gp and BCRP efflux transporters in
intestinal cell models (10) or increase
transcellular permeability (11), with evidence that
these effects may also lead to higher in vivo
exposure (12). These same excipients are also
increasingly implicated in the inhibition of a
variety of cytochrome P450 enzymes, which have
the potential to metabolize drug in the intestinal
wall (13, 14).
For highly lipophilic compounds, lipid-based
formulations can also increase the fraction of drug
that enters the lymphatic system, avoiding hepatic
metabolic pathways (15, 16). For example, the
undecanoate ester of testosterone exhibits much
lower aqueous solubility than the native form
(<1 ng/ml cf. ~25 µg/ml) yet demonstrates higher
oral BA due a greater lipophilicity and a greater
propensity to enter the systemic circulation via the
lymph, particularly when formulated as a lipid
solution (Andriol Testocaps®) (17). Indeed, lipidic
excipients have repeatedly been shown to
increase the BA of highly lipophilic drugs — i.e.,
those with Log D values >5 and solubility in long-
chain triglyceride >50 mg/g) via the lymphatic
system [reviewed in (18)].
Lipid-based formulations therefore have the
capacity to address both physicochemical and
biological obstacles to achieving satisfactory drug
exposure. This highlights the value of
understanding the key determinants of low oral BA
of a compound of interest and selecting an
appropriate technology that overcomes the rate-
limiting barrier.
Beyond Physicochemical and
Biopharmaceutical Properties
Besides the physicochemical and biopharm-
aceutical properties of a compound, there are a
number of other considerations that may impact
technology selection and formulation
development for a particular application, including
target dose, preferred final dosage form and size,
frequency of administration, specific storage
and/or packaging requirements, excipient
acceptance and potential intellectual property
rights. These factors may play an important part in
the technology selection process. These
constraints can often be identified prior to the
initiation of development work and therefore
Capsugel Dosage Form Solutions White Paper August 2014
Page 7
reduce the risk of pursuing certain approaches
that are later deemed to be unsuitable.
Technology Selection in BA
Enhancement
Capsugel Dosage Form Solutions offers
development capabilities (GMP/non-GMP) in
amorphous spray-dried dispersions (SDDs), HME,
nanocrystals, liquid/semi-solid filled capsules and
lipid multiparticulates. Each of these enabling
technologies has a proven capacity for increasing
drug absorption and BA via several different
mechanisms, which have been deeply
investigated and form the basis of our drug
development capability. Collectively, the utility of
these respective technologies covers a broad
space in terms of drug properties and target
performance. Access to such a broad range of
complementary technologies and capabilities is
critical for optimal drug development, enabling
flexibility in selecting an optimal technology
platform for a particular compound.
The process for developing formulations based on
appropriate technologies is governed by multiple
inputs (Figure 2) to ensure that an informed
decision is made for each new compound and
associated target product profile. Ensuring that a
particular technology is well matched to a drug
compound ensures rapid and efficient feasibility
assessment, better performance in vivo of early
concept formulations and ultimate success in
reaching the target product profile.
As evident from Figure 2, this selection of
approaches takes into consideration compound
qualification and the overall product needs, which
Physicochemical
Properties
Biopharmaceutical
Properties
Global Market/Regulatory
Experience
Final Dosage Form Technology Mapping
Feasibility/Performance
Boundaries
Barriers to Absorption
Compound and
Formulation Properties
Product Needs
Compound
Qualification
Absorption
models
Past Project
Databases
Technology
Solution
Figure 2: Schematic summarizing
the various inputs required for
optimal enabling technology
selection.
Capsugel Dosage Form Solutions White Paper August 2014
Page 8
in turn necessitates a thorough dialogue and
teamwork with the customer. “Product needs”
that require consideration include the target dose
and client expectations concerning the final
dosage form size, shape, appearance and
packaging. Detailed target product understanding
based on extensive client discussions is critical to
technology selection, and preclinical and early
clinical development, since they may affect critical
elements of ultimate success, such as
compliance. Within Capsugel, such discussions
are greatly supported by experience developing
formulations in the US, Europe and Asia, across
which there may be significant variation in both
regulatory requirements and patient preferences.
Technology selection and formulation
development should also draw upon compound-
specific elements in the “Compound Qualification”
input, that is, a consideration of all drug
physicochemical and biological properties that
may constitute obstacles to drug BA and those
properties that experience has taught are
essential to feasibility and scale-up of robust SDD,
HME, nanocrystal and lipid-based technologies.
Again, essential to the collection of these
properties is an effective dialogue for exchange of
information. If needed, in silico tools may be used
to predict how certain compound properties such
as Log P, solubility and compound ionization are
expected to impact performance (though
experimental measurements are always
preferred).
From a deep fundamental understanding of
enabling technologies and past development
work, two additional tools are employed in the
formulation development process — internal
predictive physiological-based pharmacokinetic
(PBPK) models and technology maps. Firstly, we
use PBPK models based on mass transport under
physiologically relevant conditions to support
formulation development. These models are often
useful in predicting pharmacokinetic (and,
potentially, pharmacodynamic) performance
based on compound and formulation properties
(19). Originally developed for our SDD capabilities
but translatable to other enabling technologies,
these models are based on the assumption that
the time-concentration profile of all drug species —
dissolved free drug, drug in natural or formulation-
derived micelles and various undissolved but
“high-energy” particulates — drive the extent of
intestinal absorption of a poorly water-soluble
drug. Although these models were developed
primarily for solid dosage forms (SDDs,
amorphous or crystalline nanoparticles, or salt
forms), we are in the process of adapting these
models to account for the performance of lipid-
based formulations — including the incorporation
of important attributes such as the impact of
formulation dispersion, digestion, supersaturation
and overall capacity to increase dissolved drug
above its equilibrium level in lipid colloids and in
free solution.
Secondly, a retrospective analysis of our past
development projects had been used to produce
technology maps that relate key physicochemical
drug properties to oral absorption. The maps are
based on our extensive formulation experience,
including evaluation of >1000 compounds in vitro,
Capsugel Dosage Form Solutions White Paper August 2014
Page 9
>500 compounds in preclinical in vivo studies and
>100 compounds in clinical studies using SDD
technology. The graph of Figure 3 is an example
of a technology map, in which data points denote
compounds that have been successfully
developed over the past few years. In this graph,
compound solubility in aqueous media (lowest
energy crystalline form; no micelles in the media)
is plotted with respect to Log P.
The solid diagonal line in this map traces the
maximal solubility (Smax) of the lowest-energy,
neutral form of the compound, calculated via a
modified general solubility equation (Smax (mg/ml)
= 1000 * 10(-LogP)) that assumes that compound
solid-state interactions are negligible (that is, the
compound is a liquid at ambient temperature).
Figure 3: Graphic plotting compound aqueous solubility with respect to Log P for a range of compounds previously
developed into SDDs (squares) or lipid formulations (circles) at Capsugel, with subsequent overlay of the optimal
space(s) for nanocrystal, amorphous (including SDDs and HME) and lipid-based technologies at a standardized 100 mg
dose per dosage unit. This visualization provides a simplistic 2D insight into how drug physicochemical properties can
affect the feasibility and performance of various enabling technologies, but should not be viewed in isolation because it
does not consider other important properties such as biological obstacles to drug exposure.
0 1 2 3 4 5 6 7 8 9 10
1000000
10000
100
0.0001
0.01
1.0
0.000001
CrystallineSolubility
(neutralform)(µg/ml)
Log P
Vertical Distance from
Diagonal Proportional to Tm
Bulk crystals
well absorbed
Assumes
100-mg active dose
Challenging space, few compounds exist.
Expect enabling technologies to only
moderately enhance absorption
Dissolution limited
absorption
Solubility limited
absorption
Concept Technology Map Based on Compound
Physicochemical Properties at a Fixed Dose
Capsugel Dosage Form Solutions White Paper August 2014
Page 10
Decreasing aqueous solubility at a constant Log P
value therefore is driven primarily by an increase
in the overall solid-state interactions, which is
directly proportional to compound melting
temperature (Tm). Thus, in general, the further a
compound falls below the diagonal line, the higher
its Tm value. In the upper region of this map,
crystalline solubility is sufficient that high BA of a
100 mg dose can be achieved using simple,
nonenabling formulations. With increasing Log P
and/or increasing Tm, however, the decrease in
solubility creates the need for enabling
technologies to maintain good in vivo
performance. Particle size reduction technologies
(i.e., micronization, nanocrystals) can offer
acceptable BA at a 100 mg dose when solubility
falls below 1 mg/ml, by overcoming instances
where the dissolution rate of unprocessed drug is
too slow to maintain the drug concentration at its
equilibrium level in the intestine. As the solubility
decreases further, the utility of such technologies
diminish as solubility reaches the point at which
absorption is inadequate even if high (even
instantaneous) dissolution rates are achieved. At
these low solubilities, it is necessary to utilize
technologies that improve drug concentration in
the GI lumen above its equilibrium solubility
and/or drug transport across the unstirred water
layer via sub-micron colloids. Amorphous solid
dispersions (including SDD and HME) are highly
effective at raising the concentration of dissolved
drug above its equilibrium solubility across a
broad range of Log P values (~0 to 6). For
compounds with high lipophilicity (i.e., Log P
>~6), additional excipients provided by lipid
technologies are necessary to solubilize and
enhance transport of the compound through the
(unstirred) aqueous boundary layer — a process
that can be slow and often limit absorption for
lipophilic drugs. Lipid technologies also cover a
broad Log P range of ~3 to 10, hence there is
overlap region of amorphous and lipid approaches
between Log P 3 and 6 values, with progressively
greater applicability of lipid approaches with
increasing compound lipophilicity. Notably, the
optimal utility of lipid technologies in Figure 3
corresponds to the space below the solid diagonal
line (where Tm is effectively at ambient
temperature or less), reflecting the fact that
compound solubility in oil will decrease with
increasing Tm. Indeed, lipid formulations have
proven utility in delivering low to moderate melting
compounds (e.g., oily compounds to Tm <200°C),
but development of lipid solutions becomes
challenging with high melting compounds unless
the compound dose per dosage unit is low (i.e.,
<50 mg). In such cases lipid suspensions are a
viable option to improve BA when lipidic excipients
are still needed. Similarly for solid dispersions, a
high Tm can be limiting to feasibility, for example,
by requiring the use of higher process
temperatures in HME, which increases the risk of
compound and/or excipient degradation. For
SDDs, a high Tm can be limit solubility in
commonly used organic spray solvents resulting in
an inefficient, low throughput process. In order to
efficiently process such high Tm compounds, a
high-temperature spray dry process (“hot
process”) has been developed (20). In this
process, the drug suspension is heated to high
Capsugel Dosage Form Solutions White Paper August 2014
Page 11
temperatures—often well above the ambient-
pressure boiling point of the solvent — to dissolve
the drug immediately before it is introduced into
the spray dryer.
Table 1 lists specific compounds that exemplify
the relationship between drug physicochemical
properties and the enabling capacity of
amorphous and lipid-based technologies.
Compounds 1 through 9 utilized either
nanocrystal or amorphous dispersion technology,
while Compounds 10 through 18 utilized lipid-
based technology, all for the purpose of BA
enhancement. The developed formulations have
been subsequently assessed as optimal to sub-
optimal based on their location on the technology
map in Figure 3 (i.e., the physicochemical
properties of the compound). In some cases, more
than one technology was utilized for comparative
purposes.
Nanocrystal and Amorphous Dispersions
Compounds 1 through 6 were all successfully
formulated as amorphous SDDs and all six
provided targeted exposure when dosed in the
clinic. The Log P values for these compounds
ranged from about 2 to about 10. Aqueous
solubility of the neutral crystalline form ranged
from less than 0.01 μg/ml to ~100 μg/ml and the
Tm ranged from ~80°C to about 240°C. It is clear
from this broad range of properties that SDDs can
be successfully formulated for compounds having
a broad range of properties. Compound 6 was
particularly challenging to formulate due to its very
high Tm and strong tendency to recrystallize from
amorphous or solution states. Despite this, low
(10% w/w) active loading SDDs were developed
that stabilized the amorphous form and
performed well in vivo. Additionally, solid
nanocrystalline dispersions with higher active
loadings were developed that performed as well or
better than the SDD. Similarly, Compounds 7 and
8 also had a strong tendency to crystallize. In the
case of Compound 7, the nanocrystalline
formulations that did not generate highly
supersaturated solutions upon dissolution
performed the best in vivo. In the case of
Compound 8, an acid-soluble base, using a
nonenteric dispersion polymer, PVP/VA, made via
HME promoted gastric dissolution and, though it
precipitated rapidly at intestinal pH in vitro, it
nonetheless performed the best in vivo.
Finally, Compound 9, a high Log P liquid (Tm
<20°C) that would not normally be considered
ideal for solid dispersions, was formulated as an
amorphous dispersion adsorbed to a high-surface-
area silicon dioxide carrier. This formulation
provided very rapid dissolution of the compound
and, in the clinic, resulted in near complete
absorption at doses up to greater than 1 gram.
Lipid-Based Formulations
Compounds 10 to 18 in Table 1 cover a broad
range of Log P values (i.e., between 3 and 7),
though all showed enhanced BA when formulated
with lipids, compared to that obtained with dosage
forms based on crystalline drug. Compounds 10
through 16 were good candidates for lipid
formulation technology based on physicochemical
properties, and robust-performing (both in vitro
Capsugel Dosage Form Solutions White Paper August 2014
Page 12
Table 1: Selected physicochemical properties of 18 past compounds in relation to the performance of the developed formulation. Cells are color coded based on
suitability for the respective technology based on the physicochemical properties shown (green = optimal, orange = moderate, red = nonoptimal) **These
compounds had proven biological barriers to BA, namely susceptibility to P-gp efflux
Compound #
Melting
Temperature (°C)
Log P/
Log D
Aq. Solubility
(µg/ml)
Technology / Formulation
In Vivo Performance
(clinical data unless stated otherwise)
1 80 – 100 6 – 7 0.01 – 0.1 HPMCAS SDD
6-fold increase in fasted exposure compared to softgel reference. Crystalline
exposure in animals near zero
2 90 – 100 ~3 50 – 100 HPMCAS SDD 6-fold increase in fasted exposure compared tocrystalline @ 300 mg dose
3 150 – 170 ~4 1 – 5 HPMCAS SDD 25% increase in AUC, 50% reduction in Tmax
4 Tg = 80 ~8 0.01 – 0.001 HPMC SDD Near complete absorption at therapeutic dose
5 ~250 ~1.5 – 2 ~10 SDD Large enhancement versus bulk crystals in dogs
6 210 – 230 4 – 5 0.1 – 0.5 HPMCAS SDD/nanocrystal
Both well absorbed; limiting recrystallization following dissolution the
challenge
7 150 – 160 4 – 5 ~1 SDD granules & nanocrystals
All formulations had improved in vivo absorption in dogs relative to bulk;
nanocrystal suspension performed best
8 200 – 220 ~3 ~5 PVP/VA HME dispersion
PVP/VA HME dispersion (particles <10 micron) fully dissolved in gastric;
performed better than HPMCAS dispersions in dogs
9 <20 9 – 10 <0.01 Amorphous dispersion adsorbed to SiO2 Near complete absorption up to doses >1 gram
10 ~150 ~5 ~4 Self-emulsifying lipid solution
4-fold increase in AUC and 7-fold increase in Cmax compared to reference
tablet dosage form in dogs
11 nd 3 – 5 <1 Self-emulsifying lipid solution
>3-fold increase in fasted exposure compared to powder-based dosage form
in dogs
12 ~140 >5 ~5 Self-emulsifying lipid solution
>2-fold increase in fasted exposure compared to reference tablet dosage
form in dogs
13 ~90 >5 <1 Self-emulsifying lipid solution Increase in fasted exposure compared to reference dosage form in dogs
14 nd >5 ~5 Self-emulsifying lipid solution
Significant increase in fasted exposure compared to powder-based dosage
form in dogs
15 ~160 3 – 5 <1 Self-emulsifying lipid solution
Significant increase in exposure compared to reference powder-based
dosage form in dogs
16 160 – 190°C 5 – 7 <1 Self-emulsifying lipid solution Offering good oral exposure in monkeys and in clinical trials
17 150 – 220°C 2 – 3 10 Oil/surfactant self-emulsifying lipid solutions**
>2-fold increase in exposure compared to an aqueous suspension in dogs.
Lipid formulation AUC at 30 mg compound higher than 300 mg compound
as a powder in capsule
18 Nd 2 – 3 <10 Self-emulsifying lipid suspension** 2-fold increase in fasted exposure compared to powder in capsule
Capsugel Dosage Form Solutions White Paper August 2014
Page 13
and in vivo) self-emulsifying lipid solutions were
developed in each case. Compound 17 exhibited
both physicochemical (i.e., low solubility) and
biological (i.e., P-gp efflux, CYP P450-mediated
intestinal metabolism) obstacles to exposure.
Several oil/surfactant two-component self-
emulsifying formulations incorporating excipients
with capacity to impact these biological barriers
were subsequently designed, developed and later
characterized in a series of in vitro tests. From
these tests, lead formulations were identified that
were effective in solubilizing the compound as the
formulation was dispersed and digested in
simulated gastric/intestinal conditions. In fasted
dogs, the lead lipid formulations provided over a
2-fold increase in exposure relative to an aqueous
suspension and gave a higher exposure at a 30
mg compound dose than that of a powder-in-
capsule formulation at a 300 mg dose. The
physicochemical properties of Compound 18 were
such that it was not possible to completely
dissolve the target dose in the lipid vehicle. A lipid
suspension, however, was developed and later
showed better performance than a powder-in-
capsule formulation in the clinic due, in part, to
the formulation addressing biological barriers to
absorption (i.e., efflux, metabolism).
Graphs similar to that in Figure 3 have been
created using the Tm or Tm/Tg (glass transition
temperature) ratio (for SDDs) versus Log P, similar
to the reference map depicted in Figure 3 for
crystalline solubility versus Log P. Such technology
maps assist experienced formulators in the
selection of the appropriate enabling technology
when the physicochemical properties of a drug are
the critical factor impacting oral absorption. Such
two-dimension maps are not the sole predictor of
the ultimate formulation or commercial success,
since there are not just two factors but many
parameters that mechanistically affect BA. For
example, the cyclic peptide cyclosporine (Log P
2.9: water solubility ~7 µg/ml) is available as a
commercial lipid formulation (Neoral®) at 25 and
100 mg doses. According to our crystalline
solubility versus Log P technology map (Figure 3),
cyclosporine would not be considered an ideal
candidate for a lipid formulation. Thus, while
conceptual maps are powerful references to the
experienced formulator, many considerations can
come into play, requiring the use of
complementary tests and analysis to optimally
formulate compounds.
By utilizing predictive PBPK and mapping,
formulators can focus initial experiments on the
technology that is most likely to be optimal – an
approach much more efficient than parallel
empirical formulation screening, since it can
minimize compound usage, accelerate
formulation development and, ultimately, increase
the chance of technical and commercial success.
Conclusions and Future Work
The companies that comprise Capsugel’s Dosage
Form Solutions (DFS) — legacy Capsugel, Encap
Drug Delivery and Bend Research — have been at
the respective forefronts of amorphous
dispersion, nanocrystal technology and lipid-based
formulation, expanding these technologies’
application and range in overcoming drug
physicochemical properties and biological
Capsugel Dosage Form Solutions White Paper August 2014
Page 14
interactions that negatively impact oral BA. The
fundamental understanding derived from this
collective investment across the key enabling
technologies has facilitated advances in science-
based technology guidance and formulation
development selection for BA enhancement. Our
development process, which relies on a series of
inputs ranging from product needs, drug
properties, past project experience, conceptual
technology maps and absorption modeling, has
been summarized in this article.
The advantages to this mechanistic science-based
process have also been discussed and can be
contrasted to instances when a drug has been
progressed down a specific technology path, or
parallel paths, where drug properties and product
needs stretch that technology’s range. This
approach is common in the industry where a
pharmaceutical company, CRO, or CDMO has
strong expertise and experience in a specific
technology. Based on past experiences, however,
this strategy is likely to be sub-optimal or
unsuccessful either early during initial feasibility
assessment or later on during development. More
empirical approaches that focus on “screening”
various technologies are also considered
suboptimal. In addition to delaying development
and requiring what may be a substantial amount
of compound to effectively evaluate several
approaches, the risk in this screening approach is
that a compound fails to perform across all
technologies (i.e., the compound is considered
“undruggable”). In many cases, however, this lack
of success may stem from inappropriate or sub-
optimal formulation design and development
rather than fundamental technology limitations.
Access to the range of key technologies,
fundamental scientific understanding of each
technology’s application and limitations and
extensive experience across the technology
options are considered key in ensuring that an
optimized, fit-for-purpose dosage form is rapidly
identified and developed. It is also important to
note that the Capsugel’s approach to formulation
work relies on compound properties that are often
already known (or otherwise measurable in silico)
but require the in-depth understanding of the
technology constraints in relation to product
needs.
We continue to expand our fundamental
understanding and our absorption models and
technology maps are routinely updated and
refined through data and experience gained from
an expanding product development pipeline of
NCE’s and existing drugs. Capsugel is currently
performing a deeper scientific analysis of all our
development projects to establish better
relationships between drug properties and
development success using SDD, HME,
nanocrystal and lipid-based technologies. An
initiative has been launched to further validate
our technology selection/formulation develop-
ment strategy: compounds are being progressed
through our formulation development process,
and SDD and lipid-based technologies will be
tested in vitro and in vivo using both technologies
for head-to-head feasibility and performance
comparisons. A particular focus will be on
compounds lying in areas of the maps between
“adjacent” technologies, for which we will also
Capsugel Dosage Form Solutions White Paper August 2014
Page 15
evaluate multiple enabling technologies to refine
maps and models by identifying properties that
are the best indicators of development success
(performance, stability, manufacturability) for
specific technologies/formulations.
Reference List
1. H.D. Williams, N.L. Trevaskis, S.A. Charman,
R.M. Shanker, W.N. Charman, C.W. Pouton,
and C.J. Porter. Strategies to address low drug
solubility in discovery and development.
Pharmacological Reviews. 65:315-499
(2013).
2. G.F. Webster, J.J. Leyden, and J.A. Gross.
Comparative pharmacokinetic profiles of a
novel isotretinoin formulation (isotretinoin-
Lidose) and the innovator isotretinoin
formulation: A randomized, 4-treatment,
crossover study. Journal of the American
Academy of Dermatology. 69:762-767 (2013).
3. D. Grantand T. Higuchi. Solubility behavior of
organic compounds, Wiley-Interscience, New
York, 1990.
4. C. Leunerand J. Dressman. Improving drug
solubility for oral delivery using solid
dispersions. European journal of
Pharmaceutics and Biopharmaceutics. 50:47-
60 (2000).
5. C.J.H. Porter, N.L. Trevaskis, and W.N.
Charman. Lipids and lipid-based formulations:
optimizing the oral delivery of lipophilic drugs.
Nature Reviews Drug Discovery. 6:231-248
(2007).
6. D.T. Friesen, R. Shanker, M. Crew, D.T.
Smithey, W. Curatolo, and J. Nightingale.
Hydroxypropyl methylcellulose acetate
succinate-based spray-dried dispersions: an
overview. Molecular Pharmaceutics. 5:1003-
1019 (2008).
7. E. Sjökvist, C. Nyström, M. Aldén, and N.
Caram-Lelham. Physicochemical aspects of
drug release. XIV. The effects of some ionic
and non-ionic surfactants on properties of a
sparingly soluble drug in solid dispersions.
International journal of pharmaceutics.
79:123-133 (1992).
8. C.-Y. Wuand L.Z. Benet. Predicting drug
disposition via application of BCS:
transport/absorption/elimination interplay
and development of a biopharmaceutics drug
disposition classification system. Pharm Res.
22:11-23 (2005).
9. U. Täuber, K. Schröder, B. Düsterberg, and H.
Matthes. Absolute bioavailability of
testosterone after oral administration of
testosterone-undecanoate and testosterone.
European journal of drug metabolism and
pharmacokinetics. 11:145-149 (1986).
10. T. Yamagata, H. Kusuhara, M. Morishita, K.
Takayama, H. Benameur, and Y. Sugiyama.
Improvement of the oral drug absorption of
topotecan through the inhibition of intestinal
xenobiotic efflux transporter, breast cancer
resistance protein, by excipients. Drug
Metabolism and Disposition. 35:1142-1148
(2007).
11. B.J. Aungst. Absorption enhancers:
applications and advances. The AAPS journal.
14:10-18 (2012).
12. M. Martin-Facklam, J. Burhenne, R. Ding, R.
Fricker, G. Mikus, I. Walter-Sack, and W.E.
Capsugel Dosage Form Solutions White Paper August 2014
Page 16
Haefeli. Dose-dependent increase of
saquinavir bioavailability by the pharmaceutic
aid cremophor EL. British Journal of Clinical
Pharmacology. 53:576-581 (2002).
13. A. Christiansen, T. Backensfeld, K. Denner,
and W. Weitschies. Effects of non-ionic
surfactants on cytochrome P450-mediated
metabolism in vitro. European Journal of
Pharmaceutics and Biopharmaceutics.
78:166-172 (2011).
14. R.J. Mountfield, S. Senepin, M. Schleimer, I.
Walter, and B. Bittner. Potential inhibitory
effects of formulation ingredients on intestinal
cytochrome P450. International Journal of
Pharmaceutics. 211:89-92 (2000).
15. T. Noguchi, W.N.A. Charman, and V.J. Stella.
The effect of drug lipophilicity and lipid
vehicles on the lymphatic absorption of
various testosterone esters. International
Journal of Pharmaceutics. 24:173-184
(1985).
16. D.M. Shackleford, W.A. Faassen, N. Houwing,
H. Lass, G.A. Edwards, C.J.H. Porter, and W.N.
Charman. Contribution of lymphatically
transported testosterone undecanoate to the
systemic exposure of testosterone after oral
administration of two andriol formulations in
conscious lymph duct-cannulated dogs.
Journal of Pharmacology and Experimental
Therapeutics. 306:925-933 (2003).
17. E. Nieschlag, J. Mauss, A. Coert, and P.
Kićović. Plasma androgen levels in men after
oral administration of testosterone or
testosterone undecanoate. Acta
Endocrinologica. 79:366-374 (1975).
18. N.L. Trevaskis, W.N. Charman, and C.J.H.
Porter. Lipid-based delivery systems and
intestinal lymphatic drug transport: A
mechanistic update. Advanced Drug Delivery
Reviews. 60:702-716 (2008).
19. Bend Research Inc., Enhanced drug discovery:
early adoption of delivery technologies by
bend research clients enables and
accelerates drug discovery
http://www.bendresearch.com/sites/default/
files/TB_Enhanced%20Drug%20Discovery.pdf
2013.
20. D.T. Friesen, D.D. Newbold, J.M. Baumann,
D.B. Dubose, and D.L. Millard. Spray-drying
process, WO 2010 111132 A3.

More Related Content

What's hot

The role and added value of proper formulation development
The role and added value of proper formulation development The role and added value of proper formulation development
The role and added value of proper formulation development
Mohamad Haitham Ayad
 
Formulation design, optimization and evaluation of domperidone maleate gastro...
Formulation design, optimization and evaluation of domperidone maleate gastro...Formulation design, optimization and evaluation of domperidone maleate gastro...
Formulation design, optimization and evaluation of domperidone maleate gastro...
Dr. Raghavendra Kumar Gunda
 
The Viscosity Reduction Platform: Enabling subcutaneous (subQ) delivery
The Viscosity Reduction Platform: Enabling subcutaneous (subQ) deliveryThe Viscosity Reduction Platform: Enabling subcutaneous (subQ) delivery
The Viscosity Reduction Platform: Enabling subcutaneous (subQ) delivery
Merck Life Sciences
 
Design Formulation and Evaluation of Ranitidine HCl Gastro Retentive Floating...
Design Formulation and Evaluation of Ranitidine HCl Gastro Retentive Floating...Design Formulation and Evaluation of Ranitidine HCl Gastro Retentive Floating...
Design Formulation and Evaluation of Ranitidine HCl Gastro Retentive Floating...
Dr. Raghavendra Kumar Gunda
 
SEMINAR ON COPROCESSED EXCIPIENTS 1
SEMINAR ON COPROCESSED EXCIPIENTS 1SEMINAR ON COPROCESSED EXCIPIENTS 1
SEMINAR ON COPROCESSED EXCIPIENTS 1NITIN KANWALE
 
Review on Recent challenges and advances in ophthalmic drug delivery
Review on Recent challenges and advances in ophthalmic drug delivery Review on Recent challenges and advances in ophthalmic drug delivery
Review on Recent challenges and advances in ophthalmic drug delivery
Gamachu Diba
 
Solid Lipid Nanoparticles: A Strategy to Improve Oral Delivery of the Biophar...
Solid Lipid Nanoparticles: A Strategy to Improve Oral Delivery of the Biophar...Solid Lipid Nanoparticles: A Strategy to Improve Oral Delivery of the Biophar...
Solid Lipid Nanoparticles: A Strategy to Improve Oral Delivery of the Biophar...
BRNSS Publication Hub
 
Dissolution -final
Dissolution  -finalDissolution  -final
Dissolution -final
Pushpendu Gaurav
 
Lionberger gpha-2015
Lionberger gpha-2015Lionberger gpha-2015
Lionberger gpha-2015
Georgi Daskalov
 
Long acting injectable microparticle formulation - a new dimension for peptid...
Long acting injectable microparticle formulation - a new dimension for peptid...Long acting injectable microparticle formulation - a new dimension for peptid...
Long acting injectable microparticle formulation - a new dimension for peptid...
Merck Life Sciences
 
SMi Group's Lyophilization USA 2018
SMi Group's Lyophilization USA 2018SMi Group's Lyophilization USA 2018
SMi Group's Lyophilization USA 2018
Dale Butler
 
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating Tablets
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating TabletsDesign, Formulation and Evaluation of Atenolol Gastro Retentive Floating Tablets
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating Tablets
Dr. Raghavendra Kumar Gunda
 
Polymer based drug delivery systems for parenteral controlled release: from s...
Polymer based drug delivery systems for parenteral controlled release: from s...Polymer based drug delivery systems for parenteral controlled release: from s...
Polymer based drug delivery systems for parenteral controlled release: from s...
Merck Life Sciences
 
Drug Formulations
Drug FormulationsDrug Formulations
Drug Formulations
Cognibrain Healthcare
 

What's hot (16)

The role and added value of proper formulation development
The role and added value of proper formulation development The role and added value of proper formulation development
The role and added value of proper formulation development
 
Formulation design, optimization and evaluation of domperidone maleate gastro...
Formulation design, optimization and evaluation of domperidone maleate gastro...Formulation design, optimization and evaluation of domperidone maleate gastro...
Formulation design, optimization and evaluation of domperidone maleate gastro...
 
The Viscosity Reduction Platform: Enabling subcutaneous (subQ) delivery
The Viscosity Reduction Platform: Enabling subcutaneous (subQ) deliveryThe Viscosity Reduction Platform: Enabling subcutaneous (subQ) delivery
The Viscosity Reduction Platform: Enabling subcutaneous (subQ) delivery
 
Design Formulation and Evaluation of Ranitidine HCl Gastro Retentive Floating...
Design Formulation and Evaluation of Ranitidine HCl Gastro Retentive Floating...Design Formulation and Evaluation of Ranitidine HCl Gastro Retentive Floating...
Design Formulation and Evaluation of Ranitidine HCl Gastro Retentive Floating...
 
SEMINAR ON COPROCESSED EXCIPIENTS 1
SEMINAR ON COPROCESSED EXCIPIENTS 1SEMINAR ON COPROCESSED EXCIPIENTS 1
SEMINAR ON COPROCESSED EXCIPIENTS 1
 
Review on Recent challenges and advances in ophthalmic drug delivery
Review on Recent challenges and advances in ophthalmic drug delivery Review on Recent challenges and advances in ophthalmic drug delivery
Review on Recent challenges and advances in ophthalmic drug delivery
 
Solid Lipid Nanoparticles: A Strategy to Improve Oral Delivery of the Biophar...
Solid Lipid Nanoparticles: A Strategy to Improve Oral Delivery of the Biophar...Solid Lipid Nanoparticles: A Strategy to Improve Oral Delivery of the Biophar...
Solid Lipid Nanoparticles: A Strategy to Improve Oral Delivery of the Biophar...
 
Aid.2013.0099
Aid.2013.0099Aid.2013.0099
Aid.2013.0099
 
Dissolution -final
Dissolution  -finalDissolution  -final
Dissolution -final
 
Lionberger gpha-2015
Lionberger gpha-2015Lionberger gpha-2015
Lionberger gpha-2015
 
srikanth final presentation
srikanth final presentationsrikanth final presentation
srikanth final presentation
 
Long acting injectable microparticle formulation - a new dimension for peptid...
Long acting injectable microparticle formulation - a new dimension for peptid...Long acting injectable microparticle formulation - a new dimension for peptid...
Long acting injectable microparticle formulation - a new dimension for peptid...
 
SMi Group's Lyophilization USA 2018
SMi Group's Lyophilization USA 2018SMi Group's Lyophilization USA 2018
SMi Group's Lyophilization USA 2018
 
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating Tablets
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating TabletsDesign, Formulation and Evaluation of Atenolol Gastro Retentive Floating Tablets
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating Tablets
 
Polymer based drug delivery systems for parenteral controlled release: from s...
Polymer based drug delivery systems for parenteral controlled release: from s...Polymer based drug delivery systems for parenteral controlled release: from s...
Polymer based drug delivery systems for parenteral controlled release: from s...
 
Drug Formulations
Drug FormulationsDrug Formulations
Drug Formulations
 

Similar to 2014.09.30. Bioavailability Enhancement Webinar Series: Optimizing Technology Choice to Enhance Bioavailability

A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...Dhaval shah
 
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...Dhaval shah
 
Bioavailability and bioequivalence study
Bioavailability and bioequivalence studyBioavailability and bioequivalence study
Bioavailability and bioequivalence study
Mcpl Moshi
 
Bioavailability and Bioequivalence study
Bioavailability and Bioequivalence studyBioavailability and Bioequivalence study
Bioavailability and Bioequivalence study
Mcpl Moshi
 
Solubility and Solubility Enhancement Techniques: A Comprehensive Review
Solubility and Solubility Enhancement Techniques: A Comprehensive ReviewSolubility and Solubility Enhancement Techniques: A Comprehensive Review
Solubility and Solubility Enhancement Techniques: A Comprehensive Review
BRNSSPublicationHubI
 
SUMIQUIM - Lubrizol, Guide to Why Mucoadhesion Matters
SUMIQUIM - Lubrizol, Guide to Why Mucoadhesion MattersSUMIQUIM - Lubrizol, Guide to Why Mucoadhesion Matters
SUMIQUIM - Lubrizol, Guide to Why Mucoadhesion Matters
José Luis García O.
 
Oral thin film
Oral thin filmOral thin film
Oral thin film
KH. Hussan Reza
 
Biodegradable polymers as drug carriers
Biodegradable polymers as drug carriers Biodegradable polymers as drug carriers
Biodegradable polymers as drug carriers
MsK for drug correlation
 
Formulation design for poorly water-soluble drugs based on biopharmaceutics c...
Formulation design for poorly water-soluble drugs based on biopharmaceuticsc...Formulation design for poorly water-soluble drugs based on biopharmaceuticsc...
Formulation design for poorly water-soluble drugs based on biopharmaceutics c...
siddy-07
 
wp3252en-ms.pdfmmmmgxfgbio solubility be
wp3252en-ms.pdfmmmmgxfgbio solubility bewp3252en-ms.pdfmmmmgxfgbio solubility be
wp3252en-ms.pdfmmmmgxfgbio solubility be
TearDrops2
 
The Integrated Early Drug Development Platform White Paper
The Integrated Early Drug Development Platform White PaperThe Integrated Early Drug Development Platform White Paper
The Integrated Early Drug Development Platform White Paper
Covance
 
Vol. 4, Issue 8, August 2016, PharmaTutor, Paper-2
Vol. 4, Issue 8, August 2016, PharmaTutor, Paper-2Vol. 4, Issue 8, August 2016, PharmaTutor, Paper-2
Vol. 4, Issue 8, August 2016, PharmaTutor, Paper-2KH. Hussan Reza
 
2.Sagar Goda Biological classification system (BCS); its significance on diss...
2.Sagar Goda Biological classification system (BCS); its significance on diss...2.Sagar Goda Biological classification system (BCS); its significance on diss...
2.Sagar Goda Biological classification system (BCS); its significance on diss...
Sagar Goda
 
CHI's Biologics Formulation & Delivery Summit 2014
CHI's Biologics Formulation & Delivery Summit 2014CHI's Biologics Formulation & Delivery Summit 2014
CHI's Biologics Formulation & Delivery Summit 2014
James Prudhomme
 
eye of the storm
eye of the stormeye of the storm
eye of the stormJan Malek
 
02_IJPBA_1938_21.pdf
02_IJPBA_1938_21.pdf02_IJPBA_1938_21.pdf
02_IJPBA_1938_21.pdf
BRNSS Publication Hub
 
Formulation Development and Evaluation of Carbamazepine Fast Dissolving Tablets
Formulation Development and Evaluation of Carbamazepine Fast Dissolving TabletsFormulation Development and Evaluation of Carbamazepine Fast Dissolving Tablets
Formulation Development and Evaluation of Carbamazepine Fast Dissolving Tablets
Dr. Raghavendra Kumar Gunda
 

Similar to 2014.09.30. Bioavailability Enhancement Webinar Series: Optimizing Technology Choice to Enhance Bioavailability (20)

A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
 
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
 
Bioavailability and bioequivalence study
Bioavailability and bioequivalence studyBioavailability and bioequivalence study
Bioavailability and bioequivalence study
 
Bioavailability and Bioequivalence study
Bioavailability and Bioequivalence studyBioavailability and Bioequivalence study
Bioavailability and Bioequivalence study
 
Stabicon Business Portfolio
Stabicon Business PortfolioStabicon Business Portfolio
Stabicon Business Portfolio
 
Solubility and Solubility Enhancement Techniques: A Comprehensive Review
Solubility and Solubility Enhancement Techniques: A Comprehensive ReviewSolubility and Solubility Enhancement Techniques: A Comprehensive Review
Solubility and Solubility Enhancement Techniques: A Comprehensive Review
 
SUMIQUIM - Lubrizol, Guide to Why Mucoadhesion Matters
SUMIQUIM - Lubrizol, Guide to Why Mucoadhesion MattersSUMIQUIM - Lubrizol, Guide to Why Mucoadhesion Matters
SUMIQUIM - Lubrizol, Guide to Why Mucoadhesion Matters
 
Oral thin film
Oral thin filmOral thin film
Oral thin film
 
Biodegradable polymers as drug carriers
Biodegradable polymers as drug carriers Biodegradable polymers as drug carriers
Biodegradable polymers as drug carriers
 
Formulation design for poorly water-soluble drugs based on biopharmaceutics c...
Formulation design for poorly water-soluble drugs based on biopharmaceuticsc...Formulation design for poorly water-soluble drugs based on biopharmaceuticsc...
Formulation design for poorly water-soluble drugs based on biopharmaceutics c...
 
wp3252en-ms.pdfmmmmgxfgbio solubility be
wp3252en-ms.pdfmmmmgxfgbio solubility bewp3252en-ms.pdfmmmmgxfgbio solubility be
wp3252en-ms.pdfmmmmgxfgbio solubility be
 
The Integrated Early Drug Development Platform White Paper
The Integrated Early Drug Development Platform White PaperThe Integrated Early Drug Development Platform White Paper
The Integrated Early Drug Development Platform White Paper
 
Vol. 4, Issue 8, August 2016, PharmaTutor, Paper-2
Vol. 4, Issue 8, August 2016, PharmaTutor, Paper-2Vol. 4, Issue 8, August 2016, PharmaTutor, Paper-2
Vol. 4, Issue 8, August 2016, PharmaTutor, Paper-2
 
2.Sagar Goda Biological classification system (BCS); its significance on diss...
2.Sagar Goda Biological classification system (BCS); its significance on diss...2.Sagar Goda Biological classification system (BCS); its significance on diss...
2.Sagar Goda Biological classification system (BCS); its significance on diss...
 
CHI's Biologics Formulation & Delivery Summit 2014
CHI's Biologics Formulation & Delivery Summit 2014CHI's Biologics Formulation & Delivery Summit 2014
CHI's Biologics Formulation & Delivery Summit 2014
 
eye of the storm
eye of the stormeye of the storm
eye of the storm
 
Bcs
BcsBcs
Bcs
 
02_IJPBA_1938_21.pdf
02_IJPBA_1938_21.pdf02_IJPBA_1938_21.pdf
02_IJPBA_1938_21.pdf
 
Formulation Development and Evaluation of Carbamazepine Fast Dissolving Tablets
Formulation Development and Evaluation of Carbamazepine Fast Dissolving TabletsFormulation Development and Evaluation of Carbamazepine Fast Dissolving Tablets
Formulation Development and Evaluation of Carbamazepine Fast Dissolving Tablets
 
04_AJP_460_15_OA_20151231_V1
04_AJP_460_15_OA_20151231_V104_AJP_460_15_OA_20151231_V1
04_AJP_460_15_OA_20151231_V1
 

More from Valentyn Mohylyuk

202312.07-08. Conference - Booklet.pdf
202312.07-08. Conference - Booklet.pdf202312.07-08. Conference - Booklet.pdf
202312.07-08. Conference - Booklet.pdf
Valentyn Mohylyuk
 
IVIVC for Extended-Release Hydrophilic Matrix Tablets in Consideration of Bio...
IVIVC for Extended-Release Hydrophilic Matrix Tablets in Consideration of Bio...IVIVC for Extended-Release Hydrophilic Matrix Tablets in Consideration of Bio...
IVIVC for Extended-Release Hydrophilic Matrix Tablets in Consideration of Bio...
Valentyn Mohylyuk
 
Wurster Fluidised Bed Coating of Microparticles: Towards Scalable Production ...
Wurster Fluidised Bed Coating of Microparticles: Towards Scalable Production ...Wurster Fluidised Bed Coating of Microparticles: Towards Scalable Production ...
Wurster Fluidised Bed Coating of Microparticles: Towards Scalable Production ...
Valentyn Mohylyuk
 
2017.02. droplet size
2017.02. droplet size2017.02. droplet size
2017.02. droplet size
Valentyn Mohylyuk
 
IGAsorp: dynamic vapor sorption analyzer
IGAsorp: dynamic vapor sorption analyzerIGAsorp: dynamic vapor sorption analyzer
IGAsorp: dynamic vapor sorption analyzer
Valentyn Mohylyuk
 
Distek Opt-Diss 410 in-situ fiber optic UV system
Distek Opt-Diss 410 in-situ fiber optic UV systemDistek Opt-Diss 410 in-situ fiber optic UV system
Distek Opt-Diss 410 in-situ fiber optic UV system
Valentyn Mohylyuk
 
PATVIS APA: visual inspection system for automated particle analysis
PATVIS APA: visual inspection system for automated particle analysisPATVIS APA: visual inspection system for automated particle analysis
PATVIS APA: visual inspection system for automated particle analysis
Valentyn Mohylyuk
 
NETZSCH Nevio Instrument Series for THERMAL ANALYSIS
NETZSCH Nevio Instrument Series for  THERMAL ANALYSISNETZSCH Nevio Instrument Series for  THERMAL ANALYSIS
NETZSCH Nevio Instrument Series for THERMAL ANALYSIS
Valentyn Mohylyuk
 
2018.10.05. aaps. particles investigation
2018.10.05. aaps. particles investigation2018.10.05. aaps. particles investigation
2018.10.05. aaps. particles investigation
Valentyn Mohylyuk
 
2018.10.03. aaps sr supension
2018.10.03. aaps sr supension2018.10.03. aaps sr supension
2018.10.03. aaps sr supension
Valentyn Mohylyuk
 
2018.09.07. APS@FIP poster "Development of multi-dose oral sustained release ...
2018.09.07. APS@FIP poster "Development of multi-dose oral sustained release ...2018.09.07. APS@FIP poster "Development of multi-dose oral sustained release ...
2018.09.07. APS@FIP poster "Development of multi-dose oral sustained release ...
Valentyn Mohylyuk
 
2018.09.07. Development of multi-dose oral sustained release suspensions for ...
2018.09.07. Development of multi-dose oral sustained release suspensions for ...2018.09.07. Development of multi-dose oral sustained release suspensions for ...
2018.09.07. Development of multi-dose oral sustained release suspensions for ...
Valentyn Mohylyuk
 
2017.10.18. Обзор мероприятия: Индустрия 4.0: Тенденции в области фармацевтич...
2017.10.18. Обзор мероприятия: Индустрия 4.0: Тенденции в области фармацевтич...2017.10.18. Обзор мероприятия: Индустрия 4.0: Тенденции в области фармацевтич...
2017.10.18. Обзор мероприятия: Индустрия 4.0: Тенденции в области фармацевтич...
Valentyn Mohylyuk
 
2017.11.07-08. Medicines for older adults:
2017.11.07-08. Medicines for older adults: 2017.11.07-08. Medicines for older adults:
2017.11.07-08. Medicines for older adults:
Valentyn Mohylyuk
 
2017.06.13 14. 3rd tech seminar from shin etsu (bradford england)
2017.06.13 14. 3rd tech seminar from shin etsu (bradford england)2017.06.13 14. 3rd tech seminar from shin etsu (bradford england)
2017.06.13 14. 3rd tech seminar from shin etsu (bradford england)
Valentyn Mohylyuk
 
2016.02.17. Extract from PhD Dissertation (Mohylyuk Valentyn)
2016.02.17. Extract from PhD Dissertation (Mohylyuk Valentyn)2016.02.17. Extract from PhD Dissertation (Mohylyuk Valentyn)
2016.02.17. Extract from PhD Dissertation (Mohylyuk Valentyn)
Valentyn Mohylyuk
 
11 International Controlled Release Seminar
11 International Controlled Release Seminar11 International Controlled Release Seminar
11 International Controlled Release Seminar
Valentyn Mohylyuk
 
2017. Micropellets coated with Kollicoat Smartseal 30 D (Full version)
2017. Micropellets coated with Kollicoat  Smartseal 30 D (Full version)2017. Micropellets coated with Kollicoat  Smartseal 30 D (Full version)
2017. Micropellets coated with Kollicoat Smartseal 30 D (Full version)
Valentyn Mohylyuk
 
2016.10.19. конференция
2016.10.19. конференция2016.10.19. конференция
2016.10.19. конференция
Valentyn Mohylyuk
 
2016. Dosage Form Optimization: Technology to Advance the Patient-Centric Dru...
2016. Dosage Form Optimization: Technology to Advance the Patient-Centric Dru...2016. Dosage Form Optimization: Technology to Advance the Patient-Centric Dru...
2016. Dosage Form Optimization: Technology to Advance the Patient-Centric Dru...
Valentyn Mohylyuk
 

More from Valentyn Mohylyuk (20)

202312.07-08. Conference - Booklet.pdf
202312.07-08. Conference - Booklet.pdf202312.07-08. Conference - Booklet.pdf
202312.07-08. Conference - Booklet.pdf
 
IVIVC for Extended-Release Hydrophilic Matrix Tablets in Consideration of Bio...
IVIVC for Extended-Release Hydrophilic Matrix Tablets in Consideration of Bio...IVIVC for Extended-Release Hydrophilic Matrix Tablets in Consideration of Bio...
IVIVC for Extended-Release Hydrophilic Matrix Tablets in Consideration of Bio...
 
Wurster Fluidised Bed Coating of Microparticles: Towards Scalable Production ...
Wurster Fluidised Bed Coating of Microparticles: Towards Scalable Production ...Wurster Fluidised Bed Coating of Microparticles: Towards Scalable Production ...
Wurster Fluidised Bed Coating of Microparticles: Towards Scalable Production ...
 
2017.02. droplet size
2017.02. droplet size2017.02. droplet size
2017.02. droplet size
 
IGAsorp: dynamic vapor sorption analyzer
IGAsorp: dynamic vapor sorption analyzerIGAsorp: dynamic vapor sorption analyzer
IGAsorp: dynamic vapor sorption analyzer
 
Distek Opt-Diss 410 in-situ fiber optic UV system
Distek Opt-Diss 410 in-situ fiber optic UV systemDistek Opt-Diss 410 in-situ fiber optic UV system
Distek Opt-Diss 410 in-situ fiber optic UV system
 
PATVIS APA: visual inspection system for automated particle analysis
PATVIS APA: visual inspection system for automated particle analysisPATVIS APA: visual inspection system for automated particle analysis
PATVIS APA: visual inspection system for automated particle analysis
 
NETZSCH Nevio Instrument Series for THERMAL ANALYSIS
NETZSCH Nevio Instrument Series for  THERMAL ANALYSISNETZSCH Nevio Instrument Series for  THERMAL ANALYSIS
NETZSCH Nevio Instrument Series for THERMAL ANALYSIS
 
2018.10.05. aaps. particles investigation
2018.10.05. aaps. particles investigation2018.10.05. aaps. particles investigation
2018.10.05. aaps. particles investigation
 
2018.10.03. aaps sr supension
2018.10.03. aaps sr supension2018.10.03. aaps sr supension
2018.10.03. aaps sr supension
 
2018.09.07. APS@FIP poster "Development of multi-dose oral sustained release ...
2018.09.07. APS@FIP poster "Development of multi-dose oral sustained release ...2018.09.07. APS@FIP poster "Development of multi-dose oral sustained release ...
2018.09.07. APS@FIP poster "Development of multi-dose oral sustained release ...
 
2018.09.07. Development of multi-dose oral sustained release suspensions for ...
2018.09.07. Development of multi-dose oral sustained release suspensions for ...2018.09.07. Development of multi-dose oral sustained release suspensions for ...
2018.09.07. Development of multi-dose oral sustained release suspensions for ...
 
2017.10.18. Обзор мероприятия: Индустрия 4.0: Тенденции в области фармацевтич...
2017.10.18. Обзор мероприятия: Индустрия 4.0: Тенденции в области фармацевтич...2017.10.18. Обзор мероприятия: Индустрия 4.0: Тенденции в области фармацевтич...
2017.10.18. Обзор мероприятия: Индустрия 4.0: Тенденции в области фармацевтич...
 
2017.11.07-08. Medicines for older adults:
2017.11.07-08. Medicines for older adults: 2017.11.07-08. Medicines for older adults:
2017.11.07-08. Medicines for older adults:
 
2017.06.13 14. 3rd tech seminar from shin etsu (bradford england)
2017.06.13 14. 3rd tech seminar from shin etsu (bradford england)2017.06.13 14. 3rd tech seminar from shin etsu (bradford england)
2017.06.13 14. 3rd tech seminar from shin etsu (bradford england)
 
2016.02.17. Extract from PhD Dissertation (Mohylyuk Valentyn)
2016.02.17. Extract from PhD Dissertation (Mohylyuk Valentyn)2016.02.17. Extract from PhD Dissertation (Mohylyuk Valentyn)
2016.02.17. Extract from PhD Dissertation (Mohylyuk Valentyn)
 
11 International Controlled Release Seminar
11 International Controlled Release Seminar11 International Controlled Release Seminar
11 International Controlled Release Seminar
 
2017. Micropellets coated with Kollicoat Smartseal 30 D (Full version)
2017. Micropellets coated with Kollicoat  Smartseal 30 D (Full version)2017. Micropellets coated with Kollicoat  Smartseal 30 D (Full version)
2017. Micropellets coated with Kollicoat Smartseal 30 D (Full version)
 
2016.10.19. конференция
2016.10.19. конференция2016.10.19. конференция
2016.10.19. конференция
 
2016. Dosage Form Optimization: Technology to Advance the Patient-Centric Dru...
2016. Dosage Form Optimization: Technology to Advance the Patient-Centric Dru...2016. Dosage Form Optimization: Technology to Advance the Patient-Centric Dru...
2016. Dosage Form Optimization: Technology to Advance the Patient-Centric Dru...
 

Recently uploaded

MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdfMANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
Jim Jacob Roy
 
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptxMaxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Dr. Rabia Inam Gandapore
 
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #GirlsFor Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
Savita Shen $i11
 
New Drug Discovery and Development .....
New Drug Discovery and Development .....New Drug Discovery and Development .....
New Drug Discovery and Development .....
NEHA GUPTA
 
heat stroke and heat exhaustion in children
heat stroke and heat exhaustion in childrenheat stroke and heat exhaustion in children
heat stroke and heat exhaustion in children
SumeraAhmad5
 
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
Oleg Kshivets
 
Ophthalmology Clinical Tests for OSCE exam
Ophthalmology Clinical Tests for OSCE examOphthalmology Clinical Tests for OSCE exam
Ophthalmology Clinical Tests for OSCE exam
KafrELShiekh University
 
Light House Retreats: Plant Medicine Retreat Europe
Light House Retreats: Plant Medicine Retreat EuropeLight House Retreats: Plant Medicine Retreat Europe
Light House Retreats: Plant Medicine Retreat Europe
Lighthouse Retreat
 
How STIs Influence the Development of Pelvic Inflammatory Disease.pptx
How STIs Influence the Development of Pelvic Inflammatory Disease.pptxHow STIs Influence the Development of Pelvic Inflammatory Disease.pptx
How STIs Influence the Development of Pelvic Inflammatory Disease.pptx
FFragrant
 
Evaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animalsEvaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animals
Shweta
 
Physiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdfPhysiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdf
MedicoseAcademics
 
Sex determination from mandible pelvis and skull
Sex determination from mandible pelvis and skullSex determination from mandible pelvis and skull
Sex determination from mandible pelvis and skull
ShashankRoodkee
 
Flu Vaccine Alert in Bangalore Karnataka
Flu Vaccine Alert in Bangalore KarnatakaFlu Vaccine Alert in Bangalore Karnataka
Flu Vaccine Alert in Bangalore Karnataka
addon Scans
 
Cervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptxCervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptx
Dr. Rabia Inam Gandapore
 
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
Savita Shen $i11
 
basicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdfbasicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdf
aljamhori teaching hospital
 
Pictures of Superficial & Deep Fascia.ppt.pdf
Pictures of Superficial & Deep Fascia.ppt.pdfPictures of Superficial & Deep Fascia.ppt.pdf
Pictures of Superficial & Deep Fascia.ppt.pdf
Dr. Rabia Inam Gandapore
 
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdfAlcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Dr Jeenal Mistry
 
Physiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of TastePhysiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of Taste
MedicoseAcademics
 
Pharma Pcd Franchise in Jharkhand - Yodley Lifesciences
Pharma Pcd Franchise in Jharkhand - Yodley LifesciencesPharma Pcd Franchise in Jharkhand - Yodley Lifesciences
Pharma Pcd Franchise in Jharkhand - Yodley Lifesciences
Yodley Lifesciences
 

Recently uploaded (20)

MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdfMANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
 
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptxMaxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
 
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #GirlsFor Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
 
New Drug Discovery and Development .....
New Drug Discovery and Development .....New Drug Discovery and Development .....
New Drug Discovery and Development .....
 
heat stroke and heat exhaustion in children
heat stroke and heat exhaustion in childrenheat stroke and heat exhaustion in children
heat stroke and heat exhaustion in children
 
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
 
Ophthalmology Clinical Tests for OSCE exam
Ophthalmology Clinical Tests for OSCE examOphthalmology Clinical Tests for OSCE exam
Ophthalmology Clinical Tests for OSCE exam
 
Light House Retreats: Plant Medicine Retreat Europe
Light House Retreats: Plant Medicine Retreat EuropeLight House Retreats: Plant Medicine Retreat Europe
Light House Retreats: Plant Medicine Retreat Europe
 
How STIs Influence the Development of Pelvic Inflammatory Disease.pptx
How STIs Influence the Development of Pelvic Inflammatory Disease.pptxHow STIs Influence the Development of Pelvic Inflammatory Disease.pptx
How STIs Influence the Development of Pelvic Inflammatory Disease.pptx
 
Evaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animalsEvaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animals
 
Physiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdfPhysiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdf
 
Sex determination from mandible pelvis and skull
Sex determination from mandible pelvis and skullSex determination from mandible pelvis and skull
Sex determination from mandible pelvis and skull
 
Flu Vaccine Alert in Bangalore Karnataka
Flu Vaccine Alert in Bangalore KarnatakaFlu Vaccine Alert in Bangalore Karnataka
Flu Vaccine Alert in Bangalore Karnataka
 
Cervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptxCervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptx
 
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
 
basicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdfbasicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdf
 
Pictures of Superficial & Deep Fascia.ppt.pdf
Pictures of Superficial & Deep Fascia.ppt.pdfPictures of Superficial & Deep Fascia.ppt.pdf
Pictures of Superficial & Deep Fascia.ppt.pdf
 
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdfAlcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
 
Physiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of TastePhysiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of Taste
 
Pharma Pcd Franchise in Jharkhand - Yodley Lifesciences
Pharma Pcd Franchise in Jharkhand - Yodley LifesciencesPharma Pcd Franchise in Jharkhand - Yodley Lifesciences
Pharma Pcd Franchise in Jharkhand - Yodley Lifesciences
 

2014.09.30. Bioavailability Enhancement Webinar Series: Optimizing Technology Choice to Enhance Bioavailability

  • 1. 2014.09.30. Bioavailability Enhancement Webinar Series: Optimizing Technology Choice to Enhance Bioavailability Capsugel (Bend Research) Abstract: An increasing number of active compounds in pipelines today have properties that require functional formulation to enable exposure and efficacy. Despite many new technology choices, it is often difficult to match the right drug-delivery technology to a given molecule and problem statement. This problem is exacerbated by the need to save time and valuable drug in early development. This webinar describes an efficient strategy for mating enabling drug-delivery technologies with problem statements based on challenging compound properties and product concepts, building on an understanding of gut physiology, key molecule physicochemical properties, and the target product profile. Company Summary: Capsugel Dosage Form Solutions designs, develops and manufactures innovative dosage forms addressing bioavailability and other pressing product development challenges, including bioavailability enhancement, modified release, abuse deterrence, biotherapeutic processing, and inhalation formulation. Speakers Bio: Dr. David Vodak Vice PresidentBend Research Moderator Dr. Vodak's areas of expertise are research and development of novel pharmaceutical drug- delivery systems. Dr. Vodak holds a PhD in materials chemistry from the University of Michigan and a B.A. in chemistry from Willamette University. Dr. David Lyon Senior Vice PresidentBend Research Dr. Lyon is the Senior Vice President at Bend Research. He leads development activities for new technologies, oversees the development of predictive biomodels, and provides technical leadership to the research groups for new and applied technologies.
  • 2.
  • 3.
  • 4.
  • 5.
  • 6.
  • 7.
  • 8.
  • 9.
  • 10.
  • 11.
  • 12.
  • 13.
  • 14.
  • 15.
  • 16.
  • 17.
  • 18.
  • 19.
  • 20.
  • 21.
  • 22.
  • 23.
  • 24.
  • 25.
  • 26.
  • 27.
  • 28.
  • 29.
  • 30.
  • 31.
  • 32.
  • 33.
  • 34.
  • 35.
  • 36.
  • 37.
  • 38.
  • 39.
  • 40.
  • 41.
  • 42.
  • 43.
  • 44.
  • 45. Science-Based Technology Selection And Formulation Development For Oral Bioavailability Enhancement Capsugel Dosage Form Solutions White Paper Authors: Hywel Williams, Michael Morgen, Eduardo Jule, Jan Vertommen, Hassan Benameur, Dwayne Friesen and David Vodak
  • 46. Capsugel Dosage Form Solutions White Paper August 2014 Page 2 Science-Based Technology Selection and Formulation Development For Oral Bioavailability Enhancement Hywel Williams, Michael Morgen, Eduardo Jule, Jan Vertommen, Hassan Benameur, Dwayne Friesen and David Vodak Capsugel Introduction The increasing fraction of poorly water-soluble compounds in pharmaceutical discovery is leading to significant growth in the use of enabling technologies to improve oral drug absorption and bioavailability (BA). Commonly used technologies in this area have been extensively reviewed (1) and include salt selection, cocrystals, amorphous solid dispersions, particle size reduction, cyclodextrins, amorphous/lipid micro- and nanoparticulates, adsorbents and lipid-based technologies. Many of these technologies have been shown to enhance drug BA, with most commercial products utilizing solid amorphous dispersion, nanocrystalline drug or lipid-based technologies. Examples include Neoral® (cyclosporine, Abbott), a lipid-based liquid-filled capsule; Incivek® (teleprevir, Vertex), an amorphous drug dispersion produced by spray drying; Kaletra® (lopinavir and ritonavir, Abbott) an amorphous drug dispersion produced by hot- melt extrusion (HME); and Emend® (aprepitant, Merck), a nanocrystal-containing tablet. Increasing use of such enabling technologies will be driven by the need to deliver the estimated 40% to 70% of the NCE pipeline candidates that are poorly water-soluble. Enabling technologies are also widely explored in the 505(b)(2) product pathway to reformulate existing products on the market into products that are better performing (e.g., “super generics”) or during the product patent life through life-cycle management approaches. Drivers toward the 505(b)(2) regulatory pathway include faster time to market, lower development costs by avoiding certain costly and repetitive preclinical and clinical trials, and 3 to 5 years of market exclusivity dependent upon the extent of change to the previously approved drug. One example of a marketed 505(b)(2) product is Absorica™ (Ranbaxy), a hard capsule product containing a lipid formulation. This product, provides higher drug absorption in the fasted state than the original Roaccutane®/ Accutane® (Roche) softgel product, thus offering patients the potential to benefit from acne treatment independently from meals (2) and granting Ranbaxy the aforementioned benefits of a 3-year period of market exclusivity. Due to the wide applicability of enabling technologies to NCEs and off-patent drugs, the BA enhancement landscape is innovative, dynamic and diverse. Indeed, the formulation of poorly water-soluble drugs is a key focus for many contract research/development/manufacturing organizations (CROs/CDMOs), supporting drug
  • 47. Capsugel Dosage Form Solutions White Paper August 2014 Page 3 development work with one or more BA-enhancing technology approaches to advance such drug candidates. A much smaller number of companies have both a broad range of technologies and the capacity to implement and scale them from design and development to commercial scale production. Having the ability to understand and provide multiple technology or formulation approaches under one roof is extremely advantageous, since the need to partner with multiple companies during a drug development program results in higher costs, significant program delays and inefficiencies and increased risk in the development process. Optimal application of enabling technologies is based on key principles, including the following. • The diverse needs of all drug compounds currently in development across and within pharmaceutical companies cannot be addressed by a single enabling technology. • Development success is more probable if a technology is appropriately matched to the compound properties and product needs early in the development process. • In many cases, more than one technology can be utilized successfully and commercial considerations such as desired dosage format can play a decisive role. Using a technology ill-suited to a compound or problem statement often results in development delays, additional costs or even failure, due to poor manufacturability, stability, performance, or shortcomings in some other aspect of the target product profile. Appropriate application of technology is therefore critical to achieve success for development projects where achieving adequate oral absorption is required. Effective application of technology for enabled formulations can remain elusive, since it relies on many inputs, not the least of which is expertise with a range of alternative or complementary technologies, involving a clear understanding of the fundamental science governing the mechanisms of drug solubilization, absorption and metabolic fate. The purpose of this article is therefore to highlight key physicochemical and biological obstacles to drug exposure following oral administration and how effective use of formulation technology relies on an understanding of the drivers to oral BA. We will then discuss the formulation development tools that have been developed from a deep investigation of key technologies and leveraging experience of hundreds of BA-enhancement projects. Physicochemical Obstacles to Oral BA Physicochemical obstacles to oral drug BA include low aqueous solubility (a thermodynamic and form-dependent property) and a slow rate of dissolution (a kinetic property). The drug concentration gradient from the intestinal lumen across the unstirred mucus layer and into the intestinal wall is the driving force for passive absorption of drugs. Low aqueous solubility of a drug can therefore limit this gradient and result in low absorption from the intestine. A slow rate of dissolution can also limit absorption, particularly where the solubility of the drug form is sufficiently
  • 48. Capsugel Dosage Form Solutions White Paper August 2014 Page 4 low that it is necessary to maintain the concentration of drug near its solubility limit in order for drug absorption to be complete over the limited time that the drug transits the GI tract. Low drug solubility is a property common to drugs that are in Class II and IV of the Biopharmaceutical Classification System (BCS). Factors underpinning the property of low solubility are well described (3) and include: • A high crystal lattice energy (which generally increases with increasing melting temperature of a compound) and results in low solubility in essentially all solvents, sometimes referred to as “brick dust”; • a low energy of aqueous solvation (which generally decreases with increasing Log P value of a compound, i.e., lipophilicity), often referred to as “greaseball” compounds; and • a combination of both, where the impact of a high crystal energy on solubility is exacerbated by a low solvation energy. Enabling technologies increase solubility and dissolution rate by reducing the drug lattice energy, increasing drug surface area, or increasing the energy of solvation. For example, lipids, surfactants and cosolvents increase the volume and character of hydrophobic micro- phases of GI fluids, such as vesicles and micelles. Many low solubility compounds have favorable intermolecular interactions with such hydrophobic colloids, leading to increased drug solubilization. Nanocrystals enhance the dissolution rate by increasing the drug surface area and may increase drug solubility if particles are very small (~<100 nm) and/or show change in crystalline structure, particularly at the crystal surface. Spray drying and HME solid dispersions increase apparent drug solubility and, therefore, dissolution rate by molecularly dispersing a high energy amorphous form in a matrix material (4). On the other hand, lipid-based technologies are effective in augmenting drug solubility as dispersed and digested lipid components mix with endogenous bile salts and phospholipids to form a range of colloidal species such that the dissolving “solvent” is more favorable to the drug (i.e., “like dissolves like”) (5). In many cases, technology approaches have the capacity to increase drug solubility through both solid-state and solvation effects. For example, the introduction of a counterion or conformer in salts and cocrystals, respectively, can increase solubility in two ways: first, by altering both the solid-state energy through changes in molecular packing in the crystal; and second, by increasing the solvation energy by changing the nature of the local solvent, i.e., by changing pH in the case of a salt counterion, or by changing the drug to the ionized form (1). In addition, solid dispersions that use amphiphilic polymers such as hydroxypropyl methylcellulose acetate succinate (HPMCAS) (6) or nonionic surfactants (7) may also affect solvation. Finally, predissolving a drug within a lipid-based formulation can eliminate the solid- state obstacles to solubility and dissolution and, if properly formulated, will maintain the compound in solution throughout the GI tract (albeit, with a high proportion of the drug solubilized in a
  • 49. Capsugel Dosage Form Solutions White Paper August 2014 Page 5 colloidal state rather than in the aqueous phase of the GI fluid). Figure 1 matches compound solubility/ dissolution obstacles to formulation technology, which forms the foundation of a science-based technology selection process. Where low solubility stems primarily from a high crystal lattice energy, solubility will benefit most from a reduction in solid-state interactions (e.g., solid dispersions) while those compounds that show limited affinity for aqueous solvents would benefit most from approaches that enrich the GI environment with exogenous solubilizers (e.g., lipid-based formulations). This relatively simple differentiation based on the physicochemical properties of the drug, while well recognized, is often overlooked in utilizing what is known, available and, in some cases, proprietary. As discussed throughout this article, technology selection and formulation development based on scientific understanding of mechanistic barriers to absorption is likely to result in more rapid and successful development with reduced costs. Biological Obstacles to BA In some cases, it is necessary to overcome not only physicochemical obstacles to absorption, but also biological barriers, which include (8): • Efflux of absorbed drug back into the intestinal lumen (often P-gp or BCRP transporter mediated); Figure 1: Simplified diagram illustrating the principal mechanisms by which various enabling technologies increase drug solubility/dissolution rate to lead to improved oral BA. See the supporting text for a more detailed description of some of these enabling technologies.
  • 50. Capsugel Dosage Form Solutions White Paper August 2014 Page 6 • presystemic drug metabolism in the intestine (principally via cytochrome P450 enzymes); and • extensive hepatic first-pass drug metabolism. A good example of high drug absorption accompanied by low BA is that of testosterone. (<25 µg/ml), testosterone is well absorbed from the intestine, but shows extremely low BA due to extensive first-pass metabolism (9). Thus, formulation work to alter drug physicochemical properties to improve intestinal absorption would be ineffective to improve BA in this case. Certain enabling technologies have the capacity to attenuate these biological obstacles to drug BA, particularly by reducing efflux and metabolism in the intestine. Indeed, fatty acids and nonionic surfactants (typically polyethoxylated esters/ ethers of oils/fatty acids) commonly used in lipid- based technologies have frequently been shown to inhibit P-gp and BCRP efflux transporters in intestinal cell models (10) or increase transcellular permeability (11), with evidence that these effects may also lead to higher in vivo exposure (12). These same excipients are also increasingly implicated in the inhibition of a variety of cytochrome P450 enzymes, which have the potential to metabolize drug in the intestinal wall (13, 14). For highly lipophilic compounds, lipid-based formulations can also increase the fraction of drug that enters the lymphatic system, avoiding hepatic metabolic pathways (15, 16). For example, the undecanoate ester of testosterone exhibits much lower aqueous solubility than the native form (<1 ng/ml cf. ~25 µg/ml) yet demonstrates higher oral BA due a greater lipophilicity and a greater propensity to enter the systemic circulation via the lymph, particularly when formulated as a lipid solution (Andriol Testocaps®) (17). Indeed, lipidic excipients have repeatedly been shown to increase the BA of highly lipophilic drugs — i.e., those with Log D values >5 and solubility in long- chain triglyceride >50 mg/g) via the lymphatic system [reviewed in (18)]. Lipid-based formulations therefore have the capacity to address both physicochemical and biological obstacles to achieving satisfactory drug exposure. This highlights the value of understanding the key determinants of low oral BA of a compound of interest and selecting an appropriate technology that overcomes the rate- limiting barrier. Beyond Physicochemical and Biopharmaceutical Properties Besides the physicochemical and biopharm- aceutical properties of a compound, there are a number of other considerations that may impact technology selection and formulation development for a particular application, including target dose, preferred final dosage form and size, frequency of administration, specific storage and/or packaging requirements, excipient acceptance and potential intellectual property rights. These factors may play an important part in the technology selection process. These constraints can often be identified prior to the initiation of development work and therefore
  • 51. Capsugel Dosage Form Solutions White Paper August 2014 Page 7 reduce the risk of pursuing certain approaches that are later deemed to be unsuitable. Technology Selection in BA Enhancement Capsugel Dosage Form Solutions offers development capabilities (GMP/non-GMP) in amorphous spray-dried dispersions (SDDs), HME, nanocrystals, liquid/semi-solid filled capsules and lipid multiparticulates. Each of these enabling technologies has a proven capacity for increasing drug absorption and BA via several different mechanisms, which have been deeply investigated and form the basis of our drug development capability. Collectively, the utility of these respective technologies covers a broad space in terms of drug properties and target performance. Access to such a broad range of complementary technologies and capabilities is critical for optimal drug development, enabling flexibility in selecting an optimal technology platform for a particular compound. The process for developing formulations based on appropriate technologies is governed by multiple inputs (Figure 2) to ensure that an informed decision is made for each new compound and associated target product profile. Ensuring that a particular technology is well matched to a drug compound ensures rapid and efficient feasibility assessment, better performance in vivo of early concept formulations and ultimate success in reaching the target product profile. As evident from Figure 2, this selection of approaches takes into consideration compound qualification and the overall product needs, which Physicochemical Properties Biopharmaceutical Properties Global Market/Regulatory Experience Final Dosage Form Technology Mapping Feasibility/Performance Boundaries Barriers to Absorption Compound and Formulation Properties Product Needs Compound Qualification Absorption models Past Project Databases Technology Solution Figure 2: Schematic summarizing the various inputs required for optimal enabling technology selection.
  • 52. Capsugel Dosage Form Solutions White Paper August 2014 Page 8 in turn necessitates a thorough dialogue and teamwork with the customer. “Product needs” that require consideration include the target dose and client expectations concerning the final dosage form size, shape, appearance and packaging. Detailed target product understanding based on extensive client discussions is critical to technology selection, and preclinical and early clinical development, since they may affect critical elements of ultimate success, such as compliance. Within Capsugel, such discussions are greatly supported by experience developing formulations in the US, Europe and Asia, across which there may be significant variation in both regulatory requirements and patient preferences. Technology selection and formulation development should also draw upon compound- specific elements in the “Compound Qualification” input, that is, a consideration of all drug physicochemical and biological properties that may constitute obstacles to drug BA and those properties that experience has taught are essential to feasibility and scale-up of robust SDD, HME, nanocrystal and lipid-based technologies. Again, essential to the collection of these properties is an effective dialogue for exchange of information. If needed, in silico tools may be used to predict how certain compound properties such as Log P, solubility and compound ionization are expected to impact performance (though experimental measurements are always preferred). From a deep fundamental understanding of enabling technologies and past development work, two additional tools are employed in the formulation development process — internal predictive physiological-based pharmacokinetic (PBPK) models and technology maps. Firstly, we use PBPK models based on mass transport under physiologically relevant conditions to support formulation development. These models are often useful in predicting pharmacokinetic (and, potentially, pharmacodynamic) performance based on compound and formulation properties (19). Originally developed for our SDD capabilities but translatable to other enabling technologies, these models are based on the assumption that the time-concentration profile of all drug species — dissolved free drug, drug in natural or formulation- derived micelles and various undissolved but “high-energy” particulates — drive the extent of intestinal absorption of a poorly water-soluble drug. Although these models were developed primarily for solid dosage forms (SDDs, amorphous or crystalline nanoparticles, or salt forms), we are in the process of adapting these models to account for the performance of lipid- based formulations — including the incorporation of important attributes such as the impact of formulation dispersion, digestion, supersaturation and overall capacity to increase dissolved drug above its equilibrium level in lipid colloids and in free solution. Secondly, a retrospective analysis of our past development projects had been used to produce technology maps that relate key physicochemical drug properties to oral absorption. The maps are based on our extensive formulation experience, including evaluation of >1000 compounds in vitro,
  • 53. Capsugel Dosage Form Solutions White Paper August 2014 Page 9 >500 compounds in preclinical in vivo studies and >100 compounds in clinical studies using SDD technology. The graph of Figure 3 is an example of a technology map, in which data points denote compounds that have been successfully developed over the past few years. In this graph, compound solubility in aqueous media (lowest energy crystalline form; no micelles in the media) is plotted with respect to Log P. The solid diagonal line in this map traces the maximal solubility (Smax) of the lowest-energy, neutral form of the compound, calculated via a modified general solubility equation (Smax (mg/ml) = 1000 * 10(-LogP)) that assumes that compound solid-state interactions are negligible (that is, the compound is a liquid at ambient temperature). Figure 3: Graphic plotting compound aqueous solubility with respect to Log P for a range of compounds previously developed into SDDs (squares) or lipid formulations (circles) at Capsugel, with subsequent overlay of the optimal space(s) for nanocrystal, amorphous (including SDDs and HME) and lipid-based technologies at a standardized 100 mg dose per dosage unit. This visualization provides a simplistic 2D insight into how drug physicochemical properties can affect the feasibility and performance of various enabling technologies, but should not be viewed in isolation because it does not consider other important properties such as biological obstacles to drug exposure. 0 1 2 3 4 5 6 7 8 9 10 1000000 10000 100 0.0001 0.01 1.0 0.000001 CrystallineSolubility (neutralform)(µg/ml) Log P Vertical Distance from Diagonal Proportional to Tm Bulk crystals well absorbed Assumes 100-mg active dose Challenging space, few compounds exist. Expect enabling technologies to only moderately enhance absorption Dissolution limited absorption Solubility limited absorption Concept Technology Map Based on Compound Physicochemical Properties at a Fixed Dose
  • 54. Capsugel Dosage Form Solutions White Paper August 2014 Page 10 Decreasing aqueous solubility at a constant Log P value therefore is driven primarily by an increase in the overall solid-state interactions, which is directly proportional to compound melting temperature (Tm). Thus, in general, the further a compound falls below the diagonal line, the higher its Tm value. In the upper region of this map, crystalline solubility is sufficient that high BA of a 100 mg dose can be achieved using simple, nonenabling formulations. With increasing Log P and/or increasing Tm, however, the decrease in solubility creates the need for enabling technologies to maintain good in vivo performance. Particle size reduction technologies (i.e., micronization, nanocrystals) can offer acceptable BA at a 100 mg dose when solubility falls below 1 mg/ml, by overcoming instances where the dissolution rate of unprocessed drug is too slow to maintain the drug concentration at its equilibrium level in the intestine. As the solubility decreases further, the utility of such technologies diminish as solubility reaches the point at which absorption is inadequate even if high (even instantaneous) dissolution rates are achieved. At these low solubilities, it is necessary to utilize technologies that improve drug concentration in the GI lumen above its equilibrium solubility and/or drug transport across the unstirred water layer via sub-micron colloids. Amorphous solid dispersions (including SDD and HME) are highly effective at raising the concentration of dissolved drug above its equilibrium solubility across a broad range of Log P values (~0 to 6). For compounds with high lipophilicity (i.e., Log P >~6), additional excipients provided by lipid technologies are necessary to solubilize and enhance transport of the compound through the (unstirred) aqueous boundary layer — a process that can be slow and often limit absorption for lipophilic drugs. Lipid technologies also cover a broad Log P range of ~3 to 10, hence there is overlap region of amorphous and lipid approaches between Log P 3 and 6 values, with progressively greater applicability of lipid approaches with increasing compound lipophilicity. Notably, the optimal utility of lipid technologies in Figure 3 corresponds to the space below the solid diagonal line (where Tm is effectively at ambient temperature or less), reflecting the fact that compound solubility in oil will decrease with increasing Tm. Indeed, lipid formulations have proven utility in delivering low to moderate melting compounds (e.g., oily compounds to Tm <200°C), but development of lipid solutions becomes challenging with high melting compounds unless the compound dose per dosage unit is low (i.e., <50 mg). In such cases lipid suspensions are a viable option to improve BA when lipidic excipients are still needed. Similarly for solid dispersions, a high Tm can be limiting to feasibility, for example, by requiring the use of higher process temperatures in HME, which increases the risk of compound and/or excipient degradation. For SDDs, a high Tm can be limit solubility in commonly used organic spray solvents resulting in an inefficient, low throughput process. In order to efficiently process such high Tm compounds, a high-temperature spray dry process (“hot process”) has been developed (20). In this process, the drug suspension is heated to high
  • 55. Capsugel Dosage Form Solutions White Paper August 2014 Page 11 temperatures—often well above the ambient- pressure boiling point of the solvent — to dissolve the drug immediately before it is introduced into the spray dryer. Table 1 lists specific compounds that exemplify the relationship between drug physicochemical properties and the enabling capacity of amorphous and lipid-based technologies. Compounds 1 through 9 utilized either nanocrystal or amorphous dispersion technology, while Compounds 10 through 18 utilized lipid- based technology, all for the purpose of BA enhancement. The developed formulations have been subsequently assessed as optimal to sub- optimal based on their location on the technology map in Figure 3 (i.e., the physicochemical properties of the compound). In some cases, more than one technology was utilized for comparative purposes. Nanocrystal and Amorphous Dispersions Compounds 1 through 6 were all successfully formulated as amorphous SDDs and all six provided targeted exposure when dosed in the clinic. The Log P values for these compounds ranged from about 2 to about 10. Aqueous solubility of the neutral crystalline form ranged from less than 0.01 μg/ml to ~100 μg/ml and the Tm ranged from ~80°C to about 240°C. It is clear from this broad range of properties that SDDs can be successfully formulated for compounds having a broad range of properties. Compound 6 was particularly challenging to formulate due to its very high Tm and strong tendency to recrystallize from amorphous or solution states. Despite this, low (10% w/w) active loading SDDs were developed that stabilized the amorphous form and performed well in vivo. Additionally, solid nanocrystalline dispersions with higher active loadings were developed that performed as well or better than the SDD. Similarly, Compounds 7 and 8 also had a strong tendency to crystallize. In the case of Compound 7, the nanocrystalline formulations that did not generate highly supersaturated solutions upon dissolution performed the best in vivo. In the case of Compound 8, an acid-soluble base, using a nonenteric dispersion polymer, PVP/VA, made via HME promoted gastric dissolution and, though it precipitated rapidly at intestinal pH in vitro, it nonetheless performed the best in vivo. Finally, Compound 9, a high Log P liquid (Tm <20°C) that would not normally be considered ideal for solid dispersions, was formulated as an amorphous dispersion adsorbed to a high-surface- area silicon dioxide carrier. This formulation provided very rapid dissolution of the compound and, in the clinic, resulted in near complete absorption at doses up to greater than 1 gram. Lipid-Based Formulations Compounds 10 to 18 in Table 1 cover a broad range of Log P values (i.e., between 3 and 7), though all showed enhanced BA when formulated with lipids, compared to that obtained with dosage forms based on crystalline drug. Compounds 10 through 16 were good candidates for lipid formulation technology based on physicochemical properties, and robust-performing (both in vitro
  • 56. Capsugel Dosage Form Solutions White Paper August 2014 Page 12 Table 1: Selected physicochemical properties of 18 past compounds in relation to the performance of the developed formulation. Cells are color coded based on suitability for the respective technology based on the physicochemical properties shown (green = optimal, orange = moderate, red = nonoptimal) **These compounds had proven biological barriers to BA, namely susceptibility to P-gp efflux Compound # Melting Temperature (°C) Log P/ Log D Aq. Solubility (µg/ml) Technology / Formulation In Vivo Performance (clinical data unless stated otherwise) 1 80 – 100 6 – 7 0.01 – 0.1 HPMCAS SDD 6-fold increase in fasted exposure compared to softgel reference. Crystalline exposure in animals near zero 2 90 – 100 ~3 50 – 100 HPMCAS SDD 6-fold increase in fasted exposure compared tocrystalline @ 300 mg dose 3 150 – 170 ~4 1 – 5 HPMCAS SDD 25% increase in AUC, 50% reduction in Tmax 4 Tg = 80 ~8 0.01 – 0.001 HPMC SDD Near complete absorption at therapeutic dose 5 ~250 ~1.5 – 2 ~10 SDD Large enhancement versus bulk crystals in dogs 6 210 – 230 4 – 5 0.1 – 0.5 HPMCAS SDD/nanocrystal Both well absorbed; limiting recrystallization following dissolution the challenge 7 150 – 160 4 – 5 ~1 SDD granules & nanocrystals All formulations had improved in vivo absorption in dogs relative to bulk; nanocrystal suspension performed best 8 200 – 220 ~3 ~5 PVP/VA HME dispersion PVP/VA HME dispersion (particles <10 micron) fully dissolved in gastric; performed better than HPMCAS dispersions in dogs 9 <20 9 – 10 <0.01 Amorphous dispersion adsorbed to SiO2 Near complete absorption up to doses >1 gram 10 ~150 ~5 ~4 Self-emulsifying lipid solution 4-fold increase in AUC and 7-fold increase in Cmax compared to reference tablet dosage form in dogs 11 nd 3 – 5 <1 Self-emulsifying lipid solution >3-fold increase in fasted exposure compared to powder-based dosage form in dogs 12 ~140 >5 ~5 Self-emulsifying lipid solution >2-fold increase in fasted exposure compared to reference tablet dosage form in dogs 13 ~90 >5 <1 Self-emulsifying lipid solution Increase in fasted exposure compared to reference dosage form in dogs 14 nd >5 ~5 Self-emulsifying lipid solution Significant increase in fasted exposure compared to powder-based dosage form in dogs 15 ~160 3 – 5 <1 Self-emulsifying lipid solution Significant increase in exposure compared to reference powder-based dosage form in dogs 16 160 – 190°C 5 – 7 <1 Self-emulsifying lipid solution Offering good oral exposure in monkeys and in clinical trials 17 150 – 220°C 2 – 3 10 Oil/surfactant self-emulsifying lipid solutions** >2-fold increase in exposure compared to an aqueous suspension in dogs. Lipid formulation AUC at 30 mg compound higher than 300 mg compound as a powder in capsule 18 Nd 2 – 3 <10 Self-emulsifying lipid suspension** 2-fold increase in fasted exposure compared to powder in capsule
  • 57. Capsugel Dosage Form Solutions White Paper August 2014 Page 13 and in vivo) self-emulsifying lipid solutions were developed in each case. Compound 17 exhibited both physicochemical (i.e., low solubility) and biological (i.e., P-gp efflux, CYP P450-mediated intestinal metabolism) obstacles to exposure. Several oil/surfactant two-component self- emulsifying formulations incorporating excipients with capacity to impact these biological barriers were subsequently designed, developed and later characterized in a series of in vitro tests. From these tests, lead formulations were identified that were effective in solubilizing the compound as the formulation was dispersed and digested in simulated gastric/intestinal conditions. In fasted dogs, the lead lipid formulations provided over a 2-fold increase in exposure relative to an aqueous suspension and gave a higher exposure at a 30 mg compound dose than that of a powder-in- capsule formulation at a 300 mg dose. The physicochemical properties of Compound 18 were such that it was not possible to completely dissolve the target dose in the lipid vehicle. A lipid suspension, however, was developed and later showed better performance than a powder-in- capsule formulation in the clinic due, in part, to the formulation addressing biological barriers to absorption (i.e., efflux, metabolism). Graphs similar to that in Figure 3 have been created using the Tm or Tm/Tg (glass transition temperature) ratio (for SDDs) versus Log P, similar to the reference map depicted in Figure 3 for crystalline solubility versus Log P. Such technology maps assist experienced formulators in the selection of the appropriate enabling technology when the physicochemical properties of a drug are the critical factor impacting oral absorption. Such two-dimension maps are not the sole predictor of the ultimate formulation or commercial success, since there are not just two factors but many parameters that mechanistically affect BA. For example, the cyclic peptide cyclosporine (Log P 2.9: water solubility ~7 µg/ml) is available as a commercial lipid formulation (Neoral®) at 25 and 100 mg doses. According to our crystalline solubility versus Log P technology map (Figure 3), cyclosporine would not be considered an ideal candidate for a lipid formulation. Thus, while conceptual maps are powerful references to the experienced formulator, many considerations can come into play, requiring the use of complementary tests and analysis to optimally formulate compounds. By utilizing predictive PBPK and mapping, formulators can focus initial experiments on the technology that is most likely to be optimal – an approach much more efficient than parallel empirical formulation screening, since it can minimize compound usage, accelerate formulation development and, ultimately, increase the chance of technical and commercial success. Conclusions and Future Work The companies that comprise Capsugel’s Dosage Form Solutions (DFS) — legacy Capsugel, Encap Drug Delivery and Bend Research — have been at the respective forefronts of amorphous dispersion, nanocrystal technology and lipid-based formulation, expanding these technologies’ application and range in overcoming drug physicochemical properties and biological
  • 58. Capsugel Dosage Form Solutions White Paper August 2014 Page 14 interactions that negatively impact oral BA. The fundamental understanding derived from this collective investment across the key enabling technologies has facilitated advances in science- based technology guidance and formulation development selection for BA enhancement. Our development process, which relies on a series of inputs ranging from product needs, drug properties, past project experience, conceptual technology maps and absorption modeling, has been summarized in this article. The advantages to this mechanistic science-based process have also been discussed and can be contrasted to instances when a drug has been progressed down a specific technology path, or parallel paths, where drug properties and product needs stretch that technology’s range. This approach is common in the industry where a pharmaceutical company, CRO, or CDMO has strong expertise and experience in a specific technology. Based on past experiences, however, this strategy is likely to be sub-optimal or unsuccessful either early during initial feasibility assessment or later on during development. More empirical approaches that focus on “screening” various technologies are also considered suboptimal. In addition to delaying development and requiring what may be a substantial amount of compound to effectively evaluate several approaches, the risk in this screening approach is that a compound fails to perform across all technologies (i.e., the compound is considered “undruggable”). In many cases, however, this lack of success may stem from inappropriate or sub- optimal formulation design and development rather than fundamental technology limitations. Access to the range of key technologies, fundamental scientific understanding of each technology’s application and limitations and extensive experience across the technology options are considered key in ensuring that an optimized, fit-for-purpose dosage form is rapidly identified and developed. It is also important to note that the Capsugel’s approach to formulation work relies on compound properties that are often already known (or otherwise measurable in silico) but require the in-depth understanding of the technology constraints in relation to product needs. We continue to expand our fundamental understanding and our absorption models and technology maps are routinely updated and refined through data and experience gained from an expanding product development pipeline of NCE’s and existing drugs. Capsugel is currently performing a deeper scientific analysis of all our development projects to establish better relationships between drug properties and development success using SDD, HME, nanocrystal and lipid-based technologies. An initiative has been launched to further validate our technology selection/formulation develop- ment strategy: compounds are being progressed through our formulation development process, and SDD and lipid-based technologies will be tested in vitro and in vivo using both technologies for head-to-head feasibility and performance comparisons. A particular focus will be on compounds lying in areas of the maps between “adjacent” technologies, for which we will also
  • 59. Capsugel Dosage Form Solutions White Paper August 2014 Page 15 evaluate multiple enabling technologies to refine maps and models by identifying properties that are the best indicators of development success (performance, stability, manufacturability) for specific technologies/formulations. Reference List 1. H.D. Williams, N.L. Trevaskis, S.A. Charman, R.M. Shanker, W.N. Charman, C.W. Pouton, and C.J. Porter. Strategies to address low drug solubility in discovery and development. Pharmacological Reviews. 65:315-499 (2013). 2. G.F. Webster, J.J. Leyden, and J.A. Gross. Comparative pharmacokinetic profiles of a novel isotretinoin formulation (isotretinoin- Lidose) and the innovator isotretinoin formulation: A randomized, 4-treatment, crossover study. Journal of the American Academy of Dermatology. 69:762-767 (2013). 3. D. Grantand T. Higuchi. Solubility behavior of organic compounds, Wiley-Interscience, New York, 1990. 4. C. Leunerand J. Dressman. Improving drug solubility for oral delivery using solid dispersions. European journal of Pharmaceutics and Biopharmaceutics. 50:47- 60 (2000). 5. C.J.H. Porter, N.L. Trevaskis, and W.N. Charman. Lipids and lipid-based formulations: optimizing the oral delivery of lipophilic drugs. Nature Reviews Drug Discovery. 6:231-248 (2007). 6. D.T. Friesen, R. Shanker, M. Crew, D.T. Smithey, W. Curatolo, and J. Nightingale. Hydroxypropyl methylcellulose acetate succinate-based spray-dried dispersions: an overview. Molecular Pharmaceutics. 5:1003- 1019 (2008). 7. E. Sjökvist, C. Nyström, M. Aldén, and N. Caram-Lelham. Physicochemical aspects of drug release. XIV. The effects of some ionic and non-ionic surfactants on properties of a sparingly soluble drug in solid dispersions. International journal of pharmaceutics. 79:123-133 (1992). 8. C.-Y. Wuand L.Z. Benet. Predicting drug disposition via application of BCS: transport/absorption/elimination interplay and development of a biopharmaceutics drug disposition classification system. Pharm Res. 22:11-23 (2005). 9. U. Täuber, K. Schröder, B. Düsterberg, and H. Matthes. Absolute bioavailability of testosterone after oral administration of testosterone-undecanoate and testosterone. European journal of drug metabolism and pharmacokinetics. 11:145-149 (1986). 10. T. Yamagata, H. Kusuhara, M. Morishita, K. Takayama, H. Benameur, and Y. Sugiyama. Improvement of the oral drug absorption of topotecan through the inhibition of intestinal xenobiotic efflux transporter, breast cancer resistance protein, by excipients. Drug Metabolism and Disposition. 35:1142-1148 (2007). 11. B.J. Aungst. Absorption enhancers: applications and advances. The AAPS journal. 14:10-18 (2012). 12. M. Martin-Facklam, J. Burhenne, R. Ding, R. Fricker, G. Mikus, I. Walter-Sack, and W.E.
  • 60. Capsugel Dosage Form Solutions White Paper August 2014 Page 16 Haefeli. Dose-dependent increase of saquinavir bioavailability by the pharmaceutic aid cremophor EL. British Journal of Clinical Pharmacology. 53:576-581 (2002). 13. A. Christiansen, T. Backensfeld, K. Denner, and W. Weitschies. Effects of non-ionic surfactants on cytochrome P450-mediated metabolism in vitro. European Journal of Pharmaceutics and Biopharmaceutics. 78:166-172 (2011). 14. R.J. Mountfield, S. Senepin, M. Schleimer, I. Walter, and B. Bittner. Potential inhibitory effects of formulation ingredients on intestinal cytochrome P450. International Journal of Pharmaceutics. 211:89-92 (2000). 15. T. Noguchi, W.N.A. Charman, and V.J. Stella. The effect of drug lipophilicity and lipid vehicles on the lymphatic absorption of various testosterone esters. International Journal of Pharmaceutics. 24:173-184 (1985). 16. D.M. Shackleford, W.A. Faassen, N. Houwing, H. Lass, G.A. Edwards, C.J.H. Porter, and W.N. Charman. Contribution of lymphatically transported testosterone undecanoate to the systemic exposure of testosterone after oral administration of two andriol formulations in conscious lymph duct-cannulated dogs. Journal of Pharmacology and Experimental Therapeutics. 306:925-933 (2003). 17. E. Nieschlag, J. Mauss, A. Coert, and P. Kićović. Plasma androgen levels in men after oral administration of testosterone or testosterone undecanoate. Acta Endocrinologica. 79:366-374 (1975). 18. N.L. Trevaskis, W.N. Charman, and C.J.H. Porter. Lipid-based delivery systems and intestinal lymphatic drug transport: A mechanistic update. Advanced Drug Delivery Reviews. 60:702-716 (2008). 19. Bend Research Inc., Enhanced drug discovery: early adoption of delivery technologies by bend research clients enables and accelerates drug discovery http://www.bendresearch.com/sites/default/ files/TB_Enhanced%20Drug%20Discovery.pdf 2013. 20. D.T. Friesen, D.D. Newbold, J.M. Baumann, D.B. Dubose, and D.L. Millard. Spray-drying process, WO 2010 111132 A3.