ArticlePDF Available

Abstract and Figures

Encapsulation into liposomes is a formulation strategy that can improve efficacy and reduce side effects of active pharmaceutical ingredients (APIs) that exhibit poor biodistribution or pharmacokinetics when administered alone. However, many APIs are unsuitable for liposomal formulations intended for parenteral administration due to their inherent physicochemical properties─lipid bilayer permeability and water-lipid equilibrium partitioning coefficient. Too high permeability results in premature leakage from liposomes, while too low permeability means the API is not able to pass across biological barriers. There are several options for solving this issue: (i) change of the lipid bilayer composition, (ii) addition of a permeability enhancer, or (iii) modification of the chemical structure of the API to design a prodrug. The latter approach was taken in the present work, and the effect of small changes in the molecular structure of the API on its permeation rate across a lipidic bilayer was systematically explored utilizing computer simulations. An in silico methodology for prodrug design based on the COSMOperm approach has been proposed and applied to four APIs (abiraterone, cytarabine, 5-fluorouracil, and paliperidone). It is shown that the addition of aliphatic hydrocarbon chains via ester or amide bonds can render the molecule more lipophilic and increase its permeability by approximately 1 order of magnitude for each 2 carbon atoms added, while the formation of fructose adducts can provide a more hydrophilic character to the molecule and reduce its lipid partitioning. While partitioning was found to depend only on the size and type of the added group, permeability was found to depend also on the added group location. Overall, it has been shown that both permeability and lipid partitioning coefficient can be systematically shifted into the desired liposome formulability window by appropriate group contributions to the parental drug. This can significantly increase the portfolio of APIs for which liposome or lipid nanoparticle formulations become feasible.
Content may be subject to copyright.
Computational Prodrug Design Methodology for Liposome
Formulability Enhancement of Small-Molecule APIs
Martin Balouch, Katerina Storchmannová, Frantisek Stepánek,*and Karel Berka*
Cite This: https://doi.org/10.1021/acs.molpharmaceut.2c01078
Read Online
ACCESS Metrics & More Article Recommendations
ABSTRACT: Encapsulation into liposomes is a formulation strategy that can improve ecacy and reduce side eects of active
pharmaceutical ingredients (APIs) that exhibit poor biodistribution or pharmacokinetics when administered alone. However, many
APIs are unsuitable for liposomal formulations intended for parenteral administration due to their inherent physicochemical
propertieslipid bilayer permeability and waterlipid equilibrium partitioning coecient. Too high permeability results in
premature leakage from liposomes, while too low permeability means the API is not able to pass across biological barriers. There are
several options for solving this issue: (i) change of the lipid bilayer composition, (ii) addition of a permeability enhancer, or (iii)
modification of the chemical structure of the API to design a prodrug. The latter approach was taken in the present work, and the
eect of small changes in the molecular structure of the API on its permeation rate across a lipidic bilayer was systematically explored
utilizing computer simulations. An in silico methodology for prodrug design based on the COSMOperm approach has been
proposed and applied to four APIs (abiraterone, cytarabine, 5-fluorouracil, and paliperidone). It is shown that the addition of
aliphatic hydrocarbon chains via ester or amide bonds can render the molecule more lipophilic and increase its permeability by
approximately 1 order of magnitude for each 2 carbon atoms added, while the formation of fructose adducts can provide a more
hydrophilic character to the molecule and reduce its lipid partitioning. While partitioning was found to depend only on the size and
type of the added group, permeability was found to depend also on the added group location. Overall, it has been shown that both
permeability and lipid partitioning coecient can be systematically shifted into the desired liposome formulability window by
appropriate group contributions to the parental drug. This can significantly increase the portfolio of APIs for which liposome or lipid
nanoparticle formulations become feasible.
KEYWORDS: lipid bilayer, permeability, partitioning coecient, prodrug, COSMOperm
1. INTRODUCTION
Since the first liposomal formulation of an active pharmaceut-
ical ingredient (API) was approved in 1995,
1
lipid nano-
formulations have rapidly developed, leading to the recent
rollout of mRNA vaccines.
2,3
The encapsulation of macro-
molecules such as antibodies or nucleic acids
4
within liposomal
structures is facilitated mainly by combining steric factors and
electrostatic interactions with charged lipids. Regarding small
molecules, formulability into liposomes for parenteral
application strongly depends on the lipid/water partitioning
and the permeation rate across the liposome bilayer, which is
unfavorable for many APIs. Only about 20 liposomal
formulations of small-molecule APIs are currently approved
in the EU and the U.S.A. Liposomal formulations in clinical
trials are often only previously used APIs in dierent
combinations or strengths
5
rather than new chemical entities.
Received: December 15, 2022
Revised: March 6, 2023
Accepted: March 9, 2023
Article
pubs.acs.org/molecularpharmaceutics
© XXXX The Authors. Published by
American Chemical Society A
https://doi.org/10.1021/acs.molpharmaceut.2c01078
Mol. Pharmaceutics XXXX, XXX, XXXXXX
Downloaded via PALACKY UNIV on March 24, 2023 at 10:55:05 (UTC).
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
The development of liposomal formulations of new APIs is
limited because many molecules in their native form are
inherently unsuitable for liposome encapsulation and release.
An APIs partitioning and permeation properties need to be
properly balanced to be suitable for liposome formulation
intended for parenteral administration. The permeability across
the liposome membrane should be suciently high to allow
the API to be released from the liposomes in the target tissue
or cell but suciently low to avoid premature spontaneous
leakage. The lipid/water partitioning coecient should be
suciently high to allow the API to overcome the energy
barrier represented by the lipid bilayer but suciently low to
prevent the API from being trapped in the membrane. The
phenomena of API partitioning and permeability were
investigated computationally and validated experimentally in
our recent publication.
6
Systematic rules for the formulability
of small-molecule APIs in liposomes have been proposed using
the liposome biochemical classification system (LBCS). LBCS
was designed as a two-dimensional (2D) diagram with
approximative areas for the partitioning and permeation
constants for each pair of API and lipid bilayer composition
that enable or prevent successful liposome formulation. The
meaning of these areas is explained in Figure 1.
To change the API/membrane behavior (i.e., the API
position in the LBCS parametric space), there are, in principle,
three options: (i) changing the membrane composition, (ii)
using a permeation enhancer, or (iii) modifying the structure
of the API. The membrane composition can be changed, e.g.,
by adding cholesterol or specific phospholipids to influence the
membrane phase transition temperature and lipid bilayer
structure. The key characteristics governing permeability
through membranes were published recently.
7
The bilayer
thickness, the lipid phase, and the sterol type were found to
play the main role. In the specific case of ceramide bilayers, the
length of the ceramide was found to play a crucial role as well.
8
Quantitatively, a change of lipid bilayer composition is
generally suitable for fine-tuning the permeation/partitioning
values but not for achieving profound, orders-of-magnitude
shifts within the LBCS diagram for a given API.
Permeation enhancers can achieve more dramatic shifts in
permeability, substances added to the APIlipid bilayer system
to modify the permeation rate
9,10
by various mechanisms such
as the disruption of phospholipid packing in the bilayer,
solubilization of the API,
11,12
or facilitating transport.
13
Permeation enhancers are mostly studied in the context of
skin permeation, where the lipidic membrane is easily
accessible. Also, there are studies into permeation enhancers
for the oral route of administration, e.g., for improving the oral
administration of peptides.
14
Examples of permeability
enhancers for oral peptide administration include citric acid,
sodium caprate, dodecyl-β-D-maltopyranoside, and others.
15
The third option for aecting permeation and partitioning
properties is to modify the molecular structure of the API itself,
i.e., to design a prodrug. In the pharmaceutical practice,
prodrugs are used for various reasons, such as to modify an
APIs solubility or dissolution rate. Examples include the
antipsychotic drug paliperidone, whose ester paliperidone
palmitate
16
controls the dissolution rate in an intramuscular
depot formulation. Another example is abiraterone, an
anticancer drug whose ester abiraterone acetate
17
is used for
improving solubility in the gastrointestinal tract. However,
there are no reports of designing prodrugs specifically to enable
liposomal formulations of small-molecule drugs. When
considering the prodrug design, there comes a question of
whether and how the API can be modified to obtain the
desired lipid/water partitioning and liposome bilayer perme-
ation behavior. Generally, the partition coecient and
permeation rate are considered to be strongly correlated
based on the MeyerOverton rule.
18
They correlate strongly
in the case of very simple molecules
19
(molar mass around and
under 50 Da), but in the case of more complex molecules, this
approximation works poorly.
20
Moreover, the combination of
prodrug synthesis and permeation enhancement is also
possible. The key principle is the modification of API and its
incorporation into the liposome.
21
This approach includes
diglyceride conjugate of doxorubicin
22
or cholesterol conjugate
of topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothe-
cin.
23
Therefore, the present work aims to propose a general
methodology for computational prodrug design by systemati-
cally investigating the relationship between API molecular
structure modification and its position in the LBSC diagram.
To this end, four APIs with dierent initial positions in the
LBSC diagram have been chosen (cytarabine, fluorouracil,
paliperidone, abiraterone) and systematically modified by
adding either lipophilic or hydrophilic groups of dierent
properties (e.g., length of acyl chain) or position. These
molecules represent general situations where an API is initially
unsuitable for liposomal formulation either because of too low
or too high permeability or due to an unsuitable partitioning
coecient, as shown in Figure 1. Apart from their position in
the LBCS diagram, the rationale for choosing these four APIs
is that they all provide opportunities for substituting functional
groups in their molecular structure without impacting the
pharmacophore responsible for their biological action. In the
Figure 1. Parametric space showing the sorting mechanism for LBCS
classification, where log Kis the membrane/water partition coecient
and log Perm is the permeation rate through the liposome membrane at
the designated temperature. Gray bin: APIs too permeable for
liposome formulation; blue bin: neutral APIs too permeable but
possible to move to the green bin if ionizable in liposome cavity by
pH; green bin: APIs suitable for liposome formulation and even for
thermally induced release; red bin: APIs suitable for liposome
formulation but not for thermally induced release; yellow bin:
lipophilic APIs suitable for entrapment in the liposome membrane.
Positions of the four APIs investigated in this work are marked by the
squares.
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article
https://doi.org/10.1021/acs.molpharmaceut.2c01078
Mol. Pharmaceutics XXXX, XXX, XXXXXX
B
case of paliperidone
16
and abiraterone,
17
the ability to form
ester prodrugs without negatively influencing bioactivity has
even been shown experimentally, and these ester prodrugs are
successfully marketed and clinically used. We aim to provide
more general guidelines for rational prodrug design by
systematically exploring the relationship between API molec-
ular structure modification and its position in the perme-
abilitypartitioning diagram.
2. MATERIALS AND METHODS
2.1. Preparation of Molecular Structures. Each
considered molecule was drawn in MarvinSketch 21.9
24
and
saved as a SMILES file. From the SMILES, the LigPrep and
MacroModel packages from Schrodinger Release 2017-2
software generated neutral conformers of all compounds in
vacuum using the OPLS_2005 force field.
25
These compounds
were cytarabine, its ester, amide, and alkyl chain analogues, as
described in Figure 2, and four APIs and their fructose adducts,
as shown in Figure 5. For each compound, a maximum of 10
conformers were selected based on MCMM/LMC2 con-
formation searching algorithm with Monte Carlo structure
selection with the MacroModel package from the Schrodinger
2017-2 software suite. The conformers were selected within 5
kcal/mol of the conformer with the lowest energy and RMSD
of at least 0.2 nm between individual conformers. A
subsequent DFT/B-P/cc-TZVP vacuum and COSMO water
optimization with fine grid option were carried out for each
conformer using Turbomole 6.3.
26
The COSMO files for each
conformer were obtained from this procedure and sub-
sequently used for partitioning and permeation calculation.
2.2. COSMOperm Permeability Calculation. For the
purpose of the present work, the membrane with a fixed
composition (DPPC/DPPG/cholesterol = 75:10:15 mol %) at
293 K was used as the permeation barrier. The lipid bilayer
structure was obtained by molecular dynamics (MD)
simulations, as described in detail in our recent work.
6
Briefly,
a membrane bilayer containing 128 preordered lipid molecules
was created by an in-house script. Slipids
27
parameters and the
TIP3P
28
water model were used for the simulation, and the
membrane was simulated at 293 K under periodic boundary
conditions for approx. 250 ns to ensure the thermodynamic
equilibrium of the membrane. The simulation was performed
using Gromacs 4.5.4. software.
29
From the MD simulation, 5
randomly chosen conformations from the last 10 ns were
chosen and saved as.pdb files. Using COSMOtherm X8
software and.cosmo files of all membrane components, the five
separate.mic files with a σ-profile representation of the
membrane sliced into 50 horizontal layers and its electron
density and charge of each layer were calculated using the
COSMO-RS approach.
30
For each combination of the lipid membrane (five snapshots
from the DPPC/DPPG/cholesterol membrane) and each
conformer of each calculated molecule (26 molecular variants
in total), the partition coecient between the membrane and
the water phase was calculated using the COSMOmic
31
approach, and the permeation rate was calculated using the
COSMOperm approach.
32
Briefly, the chemical potential of
the molecule in that part of the membrane was calculated using
the σ-profile of a permeating molecule and the σ-profile of a
specific layer of the membrane. Therefore, the energy profile
through the membrane can be obtained (an example of
calculated profiles for several cytarabine prodrugs is shown in
Figure 3). Then, the membrane/water partitioning coecient
can be calculated from the energy minima of the chemical
potential profile directly using the following equation
=Klog e G RT(min)/
where log Kis the lipid/water partition coecient, ΔG(min)
is the free energy change for permeating molecule between the
water environment and the energy minimum, Ris the universal
gas constant, and Tis the temperature in K. The permeation
rate across the liposome membrane was calculated using the
Diamond and Katz model for the steady-state flux of a solute
through the membrane
33
=
P D z K z
z
1 1
( ) ( )d
L
L
erm
where 2Lis the thickness of the membrane, D(z) is the
diusivity of the permeating molecule in the membrane layer,
and K(z) is the partition coecient of the permeating
Figure 2. Cytarabine molecule (A) and its ester (B), amide (C), and alkyl (D) prodrugs that were included in a systematic permeation study using
COSMOperm calculations. R1 = ethyl (CytO2), butyl (CytO4), hexyl (CytO6), octyl (CytO8), decyl (CytO10), dodecyl (CytO12), tetradecyl
(CytO14), and hexadecyl (CytO16); R2 = ethyl (CytN2), butyl (CytN4), hexyl (CytN6), octyl (CytN8), and decyl (CytN10); and R3 = ethyl
(CytC2), butyl (CytC4), hexyl (CytC6), octyl (CytC8), and decyl (CytC10).
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article
https://doi.org/10.1021/acs.molpharmaceut.2c01078
Mol. Pharmaceutics XXXX, XXX, XXXXXX
C
molecule in the membrane layer. Finally, the results for all
investigated molecular structures were incorporated into the
MolMeDB database.
34
3. RESULTS AND DISCUSSION
3.1. Molecule Selection and Case Study Strategy.
Recently,
6
the permeability and partitioning coecients of 56
APIs from the DrugBank database were calculated and sorted
into the LBCS diagram. Out of these, four APIs were identified
as particularly poorly permeating: allosamidin, azacitidine,
decitabine, and cytarabine. For the sake of case studies in the
present work, cytarabine has been selected. Cytarabine is an
antineoplastic agent used to treat acute myeloid leukemia,
acute lymphocytic leukemia, chronic myelogenous leukemia,
meningeal leukemia, and other meningeal neoplasms. Cytar-
abine is on the WHO list of essential medicines due to its
therapeutic significance and is usually administered by i.v.
infusion, intrathecal injection, or subcutaneous injection as a
solution, but liposomal formulations have also been proposed,
e.g., DepoCyte
35
(now discontinued) or Vyxeos liposomal.
36
There are numerous cytarabine prodrugs in development.
These comprise mostly cytarabine amino acids, cytarabine
phosphates, and cytarabine fatty acids,
37
also considered in this
work. Cytarabine amino acid prodrugs improve cytarabine
permeability,
38
and they have already achieved satisfactory
results in clinical trials.
39
Fatty acid conjugation to cytarabine
was done with various fatty acid chain lengths, even with some
non-common ones like 24-carbon long double acid chain.
40
When palmitic acid was attached, the octanol partition
coecient increased by 4 orders of magnitude.
41
As can be seen from the examples above, adding ester-
attached fatty acid is one of the common strategies to enhance
cytarabine permeability (and bioavailability). However, a
rational guideline for the choice of the fatty acid chain length
and its position seems to be lacking. Therefore, various lengths
of fatty acids were systematically investigated in this study. To
ensure the versatility of our approach, the fatty acids were
attached to two dierent cytarabine parts: nitrogen on the
benzene ring and oxygen on the fructose C5 carbon. The
temperature for the case study was chosen to be 293 K to
mimic the temperature of liposomal preparation during the
manufacture. To ensure sucient API retention in liposomes,
the API must not leak spontaneously out of the liposome at the
temperature at which it is manufactured. There can be
applications where also, at the body temperature, the API must
not permeate through liposomes instantaneously but slowly
over time. Though the absolute values of permeation rate will
dier with temperature, the trends and principles for
permeation modification drafted in this publication will stay
relevant and be modifiable for a specific situation.
3.2. Prodrug Design for Permeability Enhancement.
The permeability of the unmodified cytarabine molecule
obtained by COSMOperm calculation was log Perm =13.2,
which is approx. 45 orders of magnitude lower than the ideal
range for liposomal formulation. Hence, cytarabine has been
chosen for the present study as a representative candidate of a
poorly permeating substance to test the computational prodrug
design methodology. To improve the permeability of such a
poorly permeating molecule, prodrugs containing nonpolar
aliphatic hydrocarbon chains attached via a hydroxyl or amine
group have been systematically created in silico, and their
permeability and partitioning coecients have been calculated.
Cytarabine contains one amine group and three hydroxyl
groups (Figure 2A), which can be theoretically used for
Figure 3. Free energy profiles of cytarabine and its ester prodrugs
from acetyl to hexadecyl diering by two carbon lengths between
neighboring analogues.
Table 1. Calculated Permeability and Partitioning Coecients for Dierent Cytarabine Prodrugs through DPPC/DPPG/Chol
(75:10:15) Membrane at 293 K, Using the Mean of 5 Calculations through Randomly Chosen MD Snapshots from the Last 10
ns of Simulation
molecule log K(mol/mol) log Perm (cm/s) molecule log K(mol/mol) log Perm (cm/s)
cytarabine (Cyt) 0.68 ±0.17 13.19 ±0.04
substitution on OH group using esterification substitution on NH2group to make amides
CytO2 1.08 ±0.16 11.43 ±0.04 CytN2 1.15 ±0.26 11.33 ±0.03
CytO4 1.31 ±0.05 10.21 ±0.06 CytN4 1.35 ±0.04 10.18 ±0.04
CytO6 0.25 ±0.25 8.67 ±0.10 CytN6 1.37 ±0.04 8.85 ±0.04
CytO8 0.16 ±0.57 7.22 ±0.17 CytN8 0.00 ±0.29 7.42 ±0.05
CytO10 2.08 ±0.60 5.47 ±0.23 CytN10 1.37 ±0.36 5.77 ±0.06
CytO12 3.90 ±0.77 3.55 ±0.45 alkyl chain substitution on pyrimidine ring
CytO14 5.64 ±0.98 1.93 ±0.53 CytC2 0,92 ±0.11 11.65 ±0.05
CytO16 8.05 ±0.99 0.08 ±0.28 CytC4 0.27 ±0.13 10.42 ±0.08
CytC6 0.47 ±0.17 8.78 ±0.14
CytC8 1.91 ±0.22 7.75 ±0.21
CytC10 3.58 ±0.40 5.48 ±0.42
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article
https://doi.org/10.1021/acs.molpharmaceut.2c01078
Mol. Pharmaceutics XXXX, XXX, XXXXXX
D
prodrug preparation. The amine group and the pentose C5
hydroxyl group (Figures 2B,C) have been chosen due to their
opposite locations. To test the robustness of the computational
prodrug design approach, the alkylation of the pyrimidine ring
at position 5 next to the amine group (Figure 2D) has also
been considered. Esters with carboxylic acids containing 2, 4, 6,
8, 10, 12, 14, and 16 carbon atoms and amides with 2, 4, 6, 8,
and 10 carbon long carboxylic acid were created computa-
tionally as cytarabine prodrugs. Regarding the pyrimidine ring,
2, 4, 6, 8, and 10 carbons were added.
The COSMOperm method is based on calculating the
energy profile through the membrane. The energy profile of
cytarabine and its ester prodrugs calculated through one of the
randomly chosen snapshots from the last 10 ns of the MD
simulation of the membrane is shown in Figure 3. With
increasing acyl chain length attached to cytarabine, the energy
barrier within the membrane monotonically decreases,
resulting in higher permeability. Concurrently, the energy
minimum located around 20 Å from the membrane center
becomes more pronounced with increasing acyl chain length. A
lower minimum leads to a higher membrane/water partition-
ing coecient. The permeability and partitioning coecients
have been calculated as described in Section 2.2, and the
resulting values are summarized in Table 1.
The three prodrug families derived from cytarabine show
distinct trajectories in the LBCS parametric space of
partitioning and permeability coecient values (Figure 4). It
is clearly visible from this graphical representation that even a
substance that was initially far from the liposome formulability
window can be brought into the feasible range by appropriate
modification of its molecular structure. In the specific case of
cytarabine, its butyrate prodrugs seem to be the most
promising candidates for encapsulation into liposomes and
their thermal release. On the other hand, if the formulation
needs to be a liposome or lipid particle where the API is
supposed to remain dissolved in the membrane, prodrugs with
longer hydrocarbon chains (dodecylate, tetradecylate, hexade-
cylate, or higher) would be the appropriate choice in this case.
It is interesting to observe that while the permeability of the
N- and O- and C-substituted prodrugs seems to depend only
on the length of the added hydrocarbon chain, the water/
membrane partitioning coecient is also aected by the
position of the substitution. From the calculation results, the
acetylation of the hydroxyl and the amine group has led to an
almost identical increase in the permeation coecient (1.71
and 1.81 in log Perm, respectively), and the addition of each
further two carbons then resulted in an almost uniform
increase in log Perm (1.44 ±0.21 for acyls and 1.52 ±0.48 for
alkyls). The monotonous and nearly linear increase of log Perm
with the number of carbons added to the original molecular
structure continued until the end of the investigated range. A
more complex pattern could be seen in the partitioning
coecient. For the first 26 carbons added to the structure,
there was only a negligible eect on the values of log K,
followed by the onset of a sustained increase (approx. 1.9 in
log Kfor every two carbons).
This observation provides guidelines for the modification of
API behavior by the formation of a prodrug: it is possible to
increase permeability by several orders of magnitude, either
with or without a simultaneous increase in the partitioning
coecient, simply by choosing an appropriate length of the
carbon chain and its substitution position.
3.3. Prodrug Design for Permeability Reduction. The
case discussed above represented a situation where the API
permeability was initially too low, and the objective of prodrug
design was to improve the permeation. However, if the API
permeation rate is too high, such API cannot be successfully
encapsulated in liposomes due to premature leakage during
manufacturing and storage before administration. The
permeation rate through the membrane should be slowed
down to prevent excessive spontaneous leakage from lip-
osomes. This can be, in principle, achieved by adding polar
groups to the API structure. The choice of possible polar
groups is broad, the main limitation being that they should be
biocompatible and either not interfere with the pharmaco-
phore or be metabolizable. For the sake of the present work,
fructose has been chosen as an example of such a structure.
Four APIs with a dierent initial location in the LBCS space
(i.e., dierent initial permeability/partitioning combinations)
have been selected to demonstrate the computational prodrug
design for permeability reduction: anticancer drugs abirater-
one, cytarabine, and 5-fluorouracil and an antipsychotic drug
paliperidone. Their fructose adducts were formed computa-
tionally, and the eect on permeability has been investigated,
as described in Section 2.2. The structures are shown in Figure
5.
The free energy profiles of native drugs and their fructose
adducts, calculated by COSMOperm,
32
are shown in Figure 6.
All fructose adducts generally have an energy barrier higher by
approximately 5 kcal/mol than the corresponding free drugs.
Although the absolute increase of the energy barrier in the
middle of the membrane (position 0 Å in Figure 6) due to
fructose addition was identical for all four investigated APIs,
the consequence on permeability was not the same. For the
two pyrimidine derivates (cytarabine and 5-fluorouracil) that
already had a high energy barrier before fructose addition (19
and 11 kcal/mol, respectively) and no or small energy
minimum within the membrane, the fructose addition
increased the overall energy barrier. Thus, the permeability
of both molecules has decreased significantly (by approx. 4
orders of magnitude), as can be seen in Table 2.
Figure 4. Cytarabine and its prodrugs in LBCS parametric space. The
squares, circles, and triangles represent dierent locations of
cytarabine substitution as indicated; the numbers represent the
length of the hydrocarbon chain (acyls for O- and N-positions, alkyls
for C-position).
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article
https://doi.org/10.1021/acs.molpharmaceut.2c01078
Mol. Pharmaceutics XXXX, XXX, XXXXXX
E
On the other hand, when considering the two already rapidly
permeating molecules (abiraterone and paliperidone), a
dierent pattern was found. Their energy in the membrane
center increased by approximately 5 kcal/mol, as in the case of
cytarabine and 5-fluorouracil. However, the fructose addition
also aected the position and magnitude of an energy
minimum within the membrane (at approx. 1520 Å from
the membrane center, as shown in Figure 6). In the case of
paliperidone, the minimum was higher by 3 kcal/mol.
Therefore, there was still an additional energy barrier which
decreased permeability by about 2 orders of magnitude.
However, abiraterone showed an increase of the energy
minimum by the same amount as in the middle of the
membrane, and consequently, the predicted permeation rate of
abiraterone and its fructose adduct did not dier significantly.
When plotting the positions of the original drugs and their
fructose prodrugs in the LBCS parametric space (Figure 7),
the combined eect of fructose addition on permeability and
partitioning coecient can be visualized. The expected
permeability reduction due to the addition of a polar substance
(fructose) resulted in a shift along the log Perm axis, which was
rather uneven depending on the starting structure. As
Figure 5. Structures of fructose adducts with cytarabine (A), abiraterone (B), 5-fluorouracil (C), and paliperidone (D).
Figure 6. Calculated free energy profiles of the selected molecules and
their fructose prodrugs within a lipid bilayer (Abi = abiraterone, FU =
5-fluorouracil, Pali = paliperidone, Cyt = cytarabine, X-fru = fructose
prodrug of compound X).
Table 2. Calculated Partition and Permeation Coecients
for Dierent APIs and Their Fructose Prodrugs through a
DPPC/DPPG/Chol (75:10:15) Membrane at 293 K, Using
5 Calculations through Randomly Chosen MD Snapshots
from the Last 10 ns of Simulation
molecule log K(mol/mol) log Perm (cm/s)
cytarabine (Cyt) 0.68 ±0.17 13.19 ±0.04
Cyt-fru 1.35 ±0.05 17.31 ±0.04
fluorouracil (FU) 0.69 ±0.11 7.28 ±0.04
FU-fru 1.13 ±0.06 11.21 ±0.04
abiraterone (Abi) 5.95 ±0.98 4.59 ±0.79
Abi-fru 2.70 ±0.72 4.00 ±0.61
paliperidone (Pali) 2.95 ±0.43 4.24 ±0.58
Pali-fru 0.63 ±0.38 6.59 ±0.97
Figure 7. Selected drugs and their corresponding fructose prodrugs in
LBCS parametric space as defined in Figure 1. Black squares with P/
A/U/Crepresent the position of the studied drug in LBCS. Each
molecule representing a square leads an arrow to the square, with f
inside representing the corresponding fructose adduct.
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article
https://doi.org/10.1021/acs.molpharmaceut.2c01078
Mol. Pharmaceutics XXXX, XXX, XXXXXX
F
discussed above, cytarabine and 5-fluorouracil show the most
significant permeability reduction after fructose addition (4
orders of magnitude), the permeability reduction of paliper-
idone was intermediate (about 2 orders of magnitude), and the
permeability of abiraterone was practically unaected by
fructose addition. Thus, fructose addition would appear to
be sucient for moving 5-fluorouracil into a feasible region in
the LBCS space, whereas, for abiraterone and paliperidone, this
might not be the case. Regarding the partitioning coecient, a
systematic shift to lower log Kvalues can be seen for all four
substances, but the extent to which this happened was again
uneven. In general, the more hydrophobic the original
substance, the more pronounced the eect of fructose addition
due to the increased polarity of the adduct. Thus, for
abiraterone and paliperidone, the addition of fructose resulted
in a reduction of log Kby 3.2 and 2.3, respectively, whereas for
5-fluorouracil and cytarabine, the log Kreduction was a more
moderate 1.8 and 1.3, respectively (Figure 7 and Table 2).
In summary, these results illustrate that the attachment of
fructose to the drug molecule can be an eective strategy to
change its liposomal formulation suitability. Depending on the
position of the initial molecule in the LBCS space, fructose
addition can change either permeability or partitioning
coecient alone or both parameters simultaneously, improving
the liposome formulability of the prodrug.
4. CONCLUSIONS
The ability of small-molecule APIs to be successfully
formulated into liposomal carriers intended for parenteral
administration depends on the combination of their water/
lipid equilibrium partitioning coecient and permeability
across the lipid bilayer. As only a few APIs in their native
form fall into the ideal formulability window, the possibilities
for permeability and partitioning coecient modification by a
systematic change of the API molecular structure have been
investigated in this work. We have investigated the feasibility of
moving the position of an API in the LBCS formulation
diagram in either direction, i.e., permeability enhancement or
permeability suppression using a computational approach
based on COSMOperm. We have created a systematic line
of prodrugs derived from a poorly permeating API
(cytarabine) to study the eects on permeability enhancement
by calculating partitioning and permeation coecients. These
prodrugs were esters and amides of cytarabine with various
carbon chain lengths. Various carbon chains were also added to
compare these results with simple carbon chain addition. A
linear dependence of permeability coecients on the length of
a hydrocarbon chain added to the molecule was found,
independently of the substitution location. However, the
dependence of the membrane partitioning coecient on the
substitution location and the hydrocarbon chain length was not
monotonic, especially for shorter carboxyls. Based on the
computational results, it was possible to identify potential
cytarabine prodrugs that should exhibit enhanced permeability
and yet avoid overarching membrane partitioning that would
block the drugs availability to the aqueous phase. This means
that using our approach, on-demand permeability can be
reached by the addition of a proper length of carboxylic acid to
form a prodrug.
Prodrug design strategies for permeability reduction have
been investigated as well. From the various possibilities of
polar molecules that can be added to the API to form a
prodrug, fructose was chosen as an example of a very polar and
biocompatible molecule. The eect of adding fructose as a
representative polar structure to four APIs with the dierent
initial positions in the LBCS diagram (cytarabine, abiraterone,
5-fluorouracil, and paliperidone) was explored. It was found
that although fructose addition resulted in a nearly identical
increase of the free energy barrier represented by the lipid
bilayer, the eect on permeation rate was strongly API-specific.
Three scenarios were identified depending on the initial API:
(i) reduction of permeability only, (ii) reduction of
partitioning coecient only, and (iii) simultaneous reduction
of both permeability and partitioning coecient. Therefore,
the addition of fructose to API into prodrug formation does
not universally lead to permeation reduction. Rather, it is
dependent on the API structure and the initial position of the
substance in the LBCS diagram.
In summary, a computational methodology for virtual
prodrug design has been developed. It has been demonstrated
that by systematically modifying the molecular structure of the
original API, it is possible to change permeability and
partitioning coecient either simultaneously or individually
and thus move the position of the API in the LBCS diagram in
the desired direction. Hence, an API with initially unfavorable
properties (either too high or too low permeability) can be
converted into a prodrug more suitable for a liposomal
formulation. Of course, the computational selection of
potential prodrug candidates is only the first step, which
must be followed by the actual synthesis of the proposed
structures, their physicochemical characterization, and phar-
macological evaluation.
AUTHOR INFORMATION
Corresponding Authors
Frantis
ek S
te
pánek Department of Chemical Engineering,
University of Chemistry and Technology, Prague, 166 28
Prague 6, Czech Republic; orcid.org/0000-0001-9288-
4568; Email: frantisek.stepanek@vscht.cz
Karel Berka Department of Physical Chemistry, Faculty of
Science, PalackyUniversity Olomouc, 771 46 Olomouc,
Czech Republic; orcid.org/0000-0001-9472-2589;
Email: karel.berka@upol.cz
Authors
Martin Balouch Department of Chemical Engineering,
University of Chemistry and Technology, Prague, 166 28
Prague 6, Czech Republic
Kater
ina Storchmannová Department of Physical
Chemistry, Faculty of Science, PalackyUniversity Olomouc,
771 46 Olomouc, Czech Republic
Complete contact information is available at:
https://pubs.acs.org/10.1021/acs.molpharmaceut.2c01078
Author Contributions
M.B. and K.S.: methodology, investigation, data analysis, and
manuscript writing. F.S.: conceptualization, methodology,
supervision, and manuscript writing. K.B.: methodology,
supervision, data analysis, and manuscript writing.
Notes
The authors declare no competing financial interest.
ACKNOWLEDGMENTS
F.S. would like to acknowledge support from the Czech
Science Foundation (Project No. 19-26127X). M.B. would like
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article
https://doi.org/10.1021/acs.molpharmaceut.2c01078
Mol. Pharmaceutics XXXX, XXX, XXXXXX
G
to acknowledge support from the Pharmaceutical Applied
Research Center (PARC). K.S. and K.B. acknowledge support
from Palacky University Olomouc Project
IGA_PrF_2023_018 and FunGIM project DSGC-2021-0060
within OP VVV CZ.02.2.69/0.0/0.0/19_073/0016713. K.S.
and K.B. acknowledge support from the ELIXIR-CZ infra-
structure (Project LM2023055). Open Access funding
provided by the CzechELib project.
REFERENCES
(1) Barenholz, Y. C. Doxil - The first FDA-approved nano-drug:
Lessons learned. J. Controlled Release 2012,160, 117134.
(2) Mulligan, M. J.; Lyke, K. E.; Kitchin, N.; Absalon, J.; Gurtman,
A.; Lockhart, S.; Neuzil, K.; Raabe, V.; Bailey, R.; Swanson, K. A.; Li,
P.; Koury, K.; Kalina, W.; Cooper, D.; Fontes-Garfias, C.; Shi, P.-Y.;
Tureci, O
.; Tompkins, K. R.; Walsh, E. E.; Frenck, R.; Falsey, A. R.;
Dormitzer, P. R.; Gruber, W. C.; Sahin, U.; Jansen, K. U. Phase I/II
study of COVID-19 RNA vaccine BNT162b1 in adults. Nature 2020,
586, 589593.
(3) Jackson, L. A.; Anderson, E. J.; Rouphael, N. G.; Roberts, P. C.;
Makhene, M.; Coler, R. N.; McCullough, M. P.; Chappell, J. D.;
Denison, M. R.; Stevens, L. J.; Pruijssers, A. J.; McDermott, A.; Flach,
B.; Doria-Rose, N. A.; Corbett, K. S.; Morabito, K. M.; O’Dell, S.;
Schmidt, S. D.; Swanson, P. A.; Padilla, M.; Mascola, J. R.; Neuzil, K.
M.; Bennett, H.; Sun, W.; Peters, E.; Makowski, M.; Albert, J.; Cross,
K.; Buchanan, W.; Pikaart-Tautges, R.; Ledgerwood, J. E.; Graham, B.
S.; Beigel, J. H. An mRNA Vaccine against SARS-CoV-2 - Preliminary
Report. N. Engl. J. Med. 2020,383, 19201931.
(4) Filipczak, N.; Pan, J.; Yalamarty, S. S. K.; Torchilin, V. P. Recent
advancements in liposome technology. Adv. Drug Delivery Rev. 2020,
156, 422.
(5) Crommelin, D. J. A.; van Hoogevest, P.; Storm, G. The role of
liposomes in clinical nanomedicine development. What now? Now
what? J. Controlled Release 2020,318, 256263.
(6) Balouch, M.; Srejber, M.; Soltys, M.; Janská, P.; Stepánek, F.;
Berka, K. In silico screening of drug candidates for thermoresponsive
liposome formulations. Mol. Syst. Des. Eng. 2021,6, 368380.
(7) Frallicciardi, J.; Melcr, J.; Siginou, P.; Marrink, S. J.; Poolman, B.
Membrane thickness, lipid phase and sterol type are determining
factors in the permeability of membranes to small solutes. Nat.
Commun. 2022,13, No. 1605.
(8) Paloncyová, M.; Vávrová, K.; Sovová, Z.; DeVane, R.; Otyepka,
M.; Berka, K. Structural Changes in Ceramide Bilayers Rationalize
Increased Permeation through Stratum Corneum Models with
Shorter Acyl Tails. J. Phys. Chem. B 2015,119, 98119819.
(9) Karande, P.; Mitragotri, S. Enhancement of transdermal drug
delivery via synergistic action of chemicals. Biochim. Biophys. Acta,
Biomembr. 2009,1788, 23622373.
(10) Chen, Y.; Quan, P.; Liu, X.; Wang, M.; Fang, L. Novel chemical
permeation enhancers for transdermal drug delivery. Asian J. Pharm.
Sci. 2014,9, 5164.
(11) Gupta, R.; Dwadasi, B. S.; Rai, B.; Mitragotri, S. Effect of
Chemical Permeation Enhancers on Skin Permeability: In silico
screening using Molecular Dynamics simulations. Sci. Rep. 2019,9,
No. 1456.
(12) Vovesná, A.; Zhigunov, A.; Balouch, M.; Zbytovská, J.
Ceramide liposomes for skin barrier recovery: A novel formulation
based on natural skin lipids. Int. J. Pharm. 2021,596, No. 120264.
(13) Fathi-Azarbayjani, A.; Ng, K. X.; Chan, Y. W.; Chan, S. Y. Lipid
Vesicles for the Skin Delivery of Diclofenac: Cerosomes vs. Other
Lipid Suspensions. Adv. Pharm. Bull. 2015,5, 2533.
(14) Berg, S.; Edlund, H.; Goundry, W. R. F.; Bergström, C. A. S.;
Davies, N. M. Considerations in the developability of peptides for oral
administration when formulated together with transient permeation
enhancers. Int. J. Pharm. 2022,628, No. 122238.
(15) Maher, S.; Mrsny, R. J.; Brayden, D. J. Intestinal permeation
enhancers for oral peptide delivery. Adv. Drug Delivery Rev. 2016,106,
277319.
(16) Nasrallah, H. A.; Gopal, S.; Gassmann-Mayer, C.; Quiroz, J. A.;
Lim, P.; Eerdekens, M.; Yuen, E.; Hough, D. A Controlled, Evidence-
Based Trial of Paliperidone Palmitate, A Long-Acting Injectable
Antipsychotic, in Schizophrenia. Neuropsychopharmacology 2010,35,
20722082.
(17) Fizazi, K.; Scher, H. I.; Molina, A.; Logothetis, C. J.; Chi, K. N.;
Jones, R. J.; Staffurth, J. N.; North, S.; Vogelzang, N. J.; Saad, F.;
Mainwaring, P.; Harland, S.; Goodman, O. B.; Sternberg, C. N.; Li, J.
H.; Kheoh, T.; Haqq, C. M.; de Bono, J. S. Abiraterone acetate for
treatment of metastatic castration-resistant prostate cancer: final
overall survival analysis of the COU-AA-301 randomised, double-
blind, placebo-controlled phase 3 study. Lancet Oncol. 2012,13, 983
992.
(18) Awoonor-Williams, E.; Rowley, C. N. Molecular simulation of
nonfacilitated membrane permeation. Biochim. Biophys. Acta, Bio-
membr. 2016,1858, 16721687.
(19) Walter, A.; Gutknecht, J. Permeability of small nonelectrolytes
through lipid bilayer membranes. J. Membr. Biol. 1986,90, 207217.
(20) Bittermann, K.; Goss, K.-U. Predicting apparent passive
permeability of Caco-2 and MDCK cell-monolayers: A mechanistic
model. PLoS One 2017,12, No. e0190319.
(21) Shah, V. M.; Nguyen, D. X.; Al Fatease, A.; Patel, P.; Cote, B.;
Woo, Y.; Gheewala, R.; Pham, Y.; Huynh, M. G.; Gannett, C.; Rao, D.
A.; Alani, A. W. G. Liposomal formulation of hypoxia activated
prodrug for the treatment of ovarian cancer. J. Controlled Release
2018,291, 169183.
(22) Kuznetsova, N. R.; Svirshchevskaya, E. V.; Skripnik, I. V.;
Zarudnaya, E. N.; Benke, A. N.; Gaenko, G. P.; Molotkovskii, Y. G.;
Vodovozova, E. L. Interaction of liposomes bearing a lipophilic
doxorubicin prodrug with tumor cells. Biochem. (Moscow), Suppl. Ser.
2013,7, 1220.
(23) Shi, L.; Wu, X.; Li, T.; Wu, Y.; Song, L.; Zhang, W.; Yin, L.;
Wu, Y.; Han, W.; Yang, Y. An esterase-activatable prodrug formulated
liposome strategy: potentiating the anticancer therapeutic efficacy and
drug safety. Nanoscale Adv. 2022,4, 952966.
(24) MarvinSketch, 21.9; ChemAxon Ltd: Budapest, Hungary, 2021.
(25) Banks, J. L.; Beard, H. S.; Cao, Y.; Cho, A. E.; Damm, W.;
Farid, R.; Felts, A. K.; Halgren, T. A.; Mainz, D. T.; Maple, J. R.;
Murphy, R.; Philipp, D. M.; Repasky, M. P.; Zhang, L. Y.; Berne, B. J.;
Friesner, R. A.; Gallicchio, E.; Levy, R. M. Integrated Modeling
Program, Applied Chemical Theory (IMPACT). J. Comput. Chem.
2005,26, 17521780.
(26) TURBOMOLE 6.3, a development of University of Karlsruhe and
Forschungszentrum Karlsruhe GmbH, 1989-2007; TURBOMOLE
GmbH, 2011.
(27) Jämbeck, J. P. M.; Lyubartsev, A. P. Derivation and Systematic
Validation of a Refined All-Atom Force Field for Phosphatidylcholine
Lipids. J. Phys. Chem. B 2012,116, 31643179.
(28) Horn, H. W.; Swope, W. C.; Pitera, J. W.; Madura, J. D.; Dick,
T. J.; Hura, G. L.; Head-Gordon, T. Development of an improved
four-site water model for biomolecular simulations: TIP4P-Ew. J.
Chem. Phys. 2004,120, 96659678.
(29) Hess, B.; Kutzner, C.; van der Spoel, D.; Lindahl, E.
GROMACS 4: Algorithms for Highly Efficient, Load-Balanced, and
Scalable Molecular Simulation. J. Chem. Theory Comput. 2008,4,
435447.
(30) Klamt, A.; Jonas, V.; Burger, T.; Lohrenz, J. C. W. Refinement
and Parametrization of COSMO-RS. J. Phys. Chem. A 1998,102,
50745085.
(31) Klamt, A.; Huniar, U.; Spycher, S.; Keldenich, J. COSMOmic:
A Mechanistic Approach to the Calculation of MembraneWater
Partition Coefficients and Internal Distributions within Membranes
and Micelles. J. Phys. Chem. B 2008,112, 1214812157.
(32) Schwöbel, J. A. H.; Ebert, A.; Bittermann, K.; Huniar, U.; Goss,
K.-U.; Klamt, A. COSMOperm: Mechanistic Prediction of Passive
Membrane Permeability for Neutral Compounds and Ions and Its pH
Dependence. J. Phys. Chem. B 2020,124, 33433354.
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article
https://doi.org/10.1021/acs.molpharmaceut.2c01078
Mol. Pharmaceutics XXXX, XXX, XXXXXX
H
(33) Diamond, J. M.; Katz, Y. Interpretation of nonelectrolyte
partition coefficients between dimyristoyl lecithin and water. J.
Membr. Biol. 1974,17, 121154.
(34) Juracka, J.; Srejber, M.; Melíková, M.; Bazgier, V.; Berka, K.
MolMeDB: Molecules on Membranes Database. Database 2019,
2019, No. baz078.
(35) Domínguez, A. R.; Hidalgo, D. O.; Garrido, R. V.; Sánchez, E.
T. Liposomal cytarabine (DepoCyte) for the treatment of neoplastic
meningitis. Clin. Transl. Oncol. 2005,7, 232238.
(36) Tzogani, K.; Penttilä, K.; Lapveteläinen, T.; Hemmings, R.;
Koenig, J.; Freire, J.; Márcia, S.; Cole, S.; Coppola, P.; Flores, B.;
Barbachano, Y.; Roige, S. D.; Pignatti, F. EMA Review of
Daunorubicin and Cytarabine Encapsulated in Liposomes (Vyxeos,
CPX-351) for the Treatment of Adults with Newly Diagnosed,
Therapy-Related Acute Myeloid Leukemia or Acute Myeloid
Leukemia with Myelodysplasia-Related Changes. Oncologist 2020,
25, e1414e1420.
(37) Chhikara, B. S.; Parang, K. Development of cytarabine prodrugs
and delivery systems for leukemia treatment. Expert Opin. Drug
Delivery 2010,7, 13991414.
(38) Sun, Y.; Sun, J.; Shi, S.; Jing, Y.; Yin, S.; Chen, Y.; Li, G.; Xu, Y.;
He, Z. Synthesis, Transport and Pharmacokinetics of 5-Amino Acid
Ester Prodrugs of 1-β-d-Arabinofuranosylcytosine. Mol. Pharmaceutics
2009,6, 315325.
(39) Tessler, S.; Mishalian, I.; Peri-Naor, R.; Gengrinovitch, S.;
Mayer, R.; Yakar, R. B.; Peled, A.; Flaishon, L. BST-236, a Novel
Cytarabine Prodrug, Is Safer and As Effective As Cytarabine in In
Vivo Leukemia Models. Blood 2018,132, 1451.
(40) Liu, R.; Zhang, J.; Zhang, D.; Wang, K.; Luan, Y. Self-
assembling nanoparticles based on cytarabine prodrug for enhanced
leukemia treatment. J. Mol. Liq. 2018,251, 178184.
(41) Zhang, J.; Zhang, D.; Hu, X.; Liu, R.; Li, Z.; Luan, Y. Rational
design of a new cytarabine-based prodrug for highly efficient oral
delivery of cytarabine. RSC Adv. 2018,8, 1310313111.
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article
https://doi.org/10.1021/acs.molpharmaceut.2c01078
Mol. Pharmaceutics XXXX, XXX, XXXXXX
I
Recommended by ACS
Theoretical Investigations on the Plasmon-Mediated
Dissociation of Small Molecules in the Presence of Silver
Atomic Wires
Olivia A. Hull and Christine M. Aikens
MARCH 02, 2023
THE JOURNAL OF PHYSICAL CHEMISTRY A READ
Formulation, Statistical Optimization, and
In Vivo
Pharmacodynamics of
Cydonia oblonga
Mucilage/Alginate
Mucoadhesive Microspheres for the Delivery of Metformi...
Sobia Noreen, Shazia Noureen,
et al.
FEBRUARY 05, 2023
ACS OMEGA READ
Molecular Dynamics and Diffusion in Amorphous Solid
Dispersions Containing Imidacloprid
Ali Mansuri, Markus Thommes,
et al.
MARCH 17, 2023
MOLECULAR PHARMACEUTICS READ
Structural Modification and Pharmacological Evaluation of
Substituted Quinoline-5,8-diones as Potent NSD2 Inhibitors
Hairong Tang, Ao Zhang,
et al.
JANUARY 15, 2023
JOURNAL OF MEDICINAL CHEMISTRY READ
Get More Suggestions >
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Cell membranes provide a selective semi-permeable barrier to the passive transport of molecules. This property differs greatly between organisms. While the cytoplasmic membrane of bacterial cells is highly permeable for weak acids and glycerol, yeasts can maintain large concentration gradients. Here we show that such differences can arise from the physical state of the plasma membrane. By combining stopped-flow kinetic measurements with molecular dynamics simulations, we performed a systematic analysis of the permeability of a variety of small molecules through synthetic membranes of different lipid composition to obtain detailed molecular insight into the permeation mechanisms. While membrane thickness is an important parameter for the permeability through fluid membranes, the largest differences occur when the membranes transit from the liquid-disordered to liquid-ordered and/or to gel state, which is in agreement with previous work on passive diffusion of water. By comparing our results with in vivo measurements from yeast, we conclude that the yeast membrane exists in a highly ordered and rigid state, which is comparable to synthetic saturated DPPC-sterol membranes.
Article
Full-text available
Liposomal nanomedicine represents a common and versatile carrier for the delivery of both lipophilic and hydrophilic drugs. However, the direct formulation of many chemotherapeutics into a liposomal system remains an enormous challenge. Using the topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN38) as a model drug, we combined lipophilic prodrug construction with subsequent integration into an exogenous liposomal scaffold to assemble a prodrug-formulated liposome for systemic administration. Reconstructing SN38 with lipid cholesterol via the esterase-activatable bond endows the resulting prodrug with elevated miscibility with liposomal compositions and esterase-responsive drug release in cancerous cells. The systemic administration of the prodrug-based nanoassemblies (Chol-SN38@LP) exhibited preferential accumulation of therapeutic payloads in tumor lesions. Compared to the SN38 clinical counterpart irinotecan, our prodrug-based nanoassemblies with adaptive features showed elevated therapeutic efficacy (∼1.5 times increase of tumor inhibition) in a preclinical A549 lung carcinoma cell-derived mouse model and improved drug tolerability (i.e., alleviated bloody diarrhea and liver damage) in multiple mice models. These results may be ascribed to extended systemic circulation and preferential tumor accumulation of our nanodrugs. Hence, our findings demonstrate that rational engineering of therapeutic nanomedicine is a promising approach for effective and safe delivery of antitumor chemotherapeutics, especially to rescue drug candidates that have failed in clinical trials owing to poor PK properties or severe toxicity in patients.
Article
Full-text available
Liposomal formulations can be advantageous in a number of scenarios such as targeted delivery to reduce the systemic toxicity of highly potent Active Pharmaceutical Ingredients (APIs), to increase drug bioavailability by prolonging systemic circulation, to protect labile APIs from degradation in the gastrointestinal tract, or to improve skin permeation in dermal delivery. However, not all APIs are suitable for encapsulation in liposomes. Some of the issues are too high permeability of the API across the lipid bilayer, which may lead to premature leakage, too low permeability, which may hinder the drug release process, or too strong membrane affinity, which may reduce the overall efficacy of drug release from liposomes. Since the most reliable way to test API encapsulation and release from liposomes so far has been experimental, an in silico model capable of predicting API transport across the lipid bilayer might accelerate formulation development. In this work, we demonstrate a new in silico approach to compute the temperature dependent permeability of a set of compounds across the bilayer of virtual liposomes constructed by molecular dynamics simulation. To validate this approach, we have conducted a series of experiments confirming the model predictions using a homologous series of fluorescent dyes. Based on the performance of individual molecules, we have defined a set of selection criteria for identifying compatible APIs for stable encapsulation and thermally controlled release from liposomes. To further demonstrate the in silico-based methodology, we have screened the DrugBank database, identified potent drugs suitable for liposome encapsulation and successfully carried out the loading and thermal release of one of them - an antimicrobial compound cycloserine.
Article
Full-text available
In March 2020, the World Health Organization (WHO) declared a pandemic of coronavirus disease 2019 (COVID-19), due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)¹. With rapidly accumulating cases and deaths reported globally², a vaccine is urgently needed. We report the available safety, tolerability, and immunogenicity data from an ongoing placebo-controlled, observer-blinded dose escalation study among 45 healthy adults, 18 to 55 years of age, randomized to receive 2 doses, separated by 21 days, of 10 µg, 30 µg, or 100 µg of BNT162b1, a lipid nanoparticle-formulated, nucleoside-modified mRNA vaccine that encodes trimerized SARS-CoV-2 spike glycoprotein receptor-binding domain (RBD). Local reactions and systemic events were dose-dependent, generally mild to moderate, and transient. A second vaccination with 100 µg was not administered due to increased reactogenicity and a lack of meaningfully increased immunogenicity after a single dose compared to the 30 μg dose. RBD-binding IgG concentrations and SARS-CoV-2 neutralizing titers in sera increased with dose level and after a second dose. Geometric mean neutralizing titers reached 1.9- to 4.6-fold that of a panel of COVID-19 convalescent human sera at least 14 days after a positive SARS-CoV-2 PCR. These results support further evaluation of this mRNA vaccine candidate. (ClinicalTrials.gov identifier: NCT04368728).
Article
This paper reviews many of the properties of a peptide that need to be considered prior to development as an oral dosage form when co-formulated with a permeation enhancer to improve oral bioavailability, including the importance and implications of peptide half-life on variability in pharmacokinetic profiles. Clinical considerations in terms of food and drug-drug interactions are also discussed. The paper further gives a brief overview how permeation enhancers overcome barriers that limit oral absorption of peptides and thereby improve their oral bioavailability, albeit bioavailabilities are still low single digit and variability is high.
Article
Diseases related to a disrupted skin barrier are accompanied by lower levels of ceramides in the stratum corneum (SC) lipid matrix. Delivering ceramides directly into damaged skin is a viable alternative to conventional corticosteroids, but is hindered by their low skin bioavailability and limited nanoformulation ability. Here, we developed stable liposomal systems containing ceramides and other SC lipids, and tested their effectiveness in skin barrier repair. Lipid film hydration and high-pressure homogenization were used to prepare different types of liposomes. To determine the stability, the particle size and polydispersity index were measured. The optimal systems were found to include ceramide 3 and 6, cholesterol and stearic acid, with 10% urea in phosphate-buffered saline as the aqueous phase. The ability of the system to repair chemically-damaged porcine skin was tested. While treatment by a standard lipid suspension reduced the passage of a model permeant only to a limited extent, drug flux through the liposomally-treated skin was much closer to permeation through intact skin. The non-homogenised liposomes were more effective than their homogenised version. These findings were also confirmed by FTIR measurements. This suggests that our approach to liposomal development has considerable potential for the repair of a disrupted skin barrier.
Article
Background The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in late 2019 and spread globally, prompting an international effort to accelerate development of a vaccine. The candidate vaccine mRNA-1273 encodes the stabilized prefusion SARS-CoV-2 spike protein. Methods We conducted a phase 1, dose-escalation, open-label trial including 45 healthy adults, 18 to 55 years of age, who received two vaccinations, 28 days apart, with mRNA-1273 in a dose of 25 μg, 100 μg, or 250 μg. There were 15 participants in each dose group. Results After the first vaccination, antibody responses were higher with higher dose (day 29 enzyme-linked immunosorbent assay anti–S-2P antibody geometric mean titer [GMT], 40,227 in the 25-μg group, 109,209 in the 100-μg group, and 213,526 in the 250-μg group). After the second vaccination, the titers increased (day 57 GMT, 299,751, 782,719, and 1,192,154, respectively). After the second vaccination, serum-neutralizing activity was detected by two methods in all participants evaluated, with values generally similar to those in the upper half of the distribution of a panel of control convalescent serum specimens. Solicited adverse events that occurred in more than half the participants included fatigue, chills, headache, myalgia, and pain at the injection site. Systemic adverse events were more common after the second vaccination, particularly with the highest dose, and three participants (21%) in the 250-μg dose group reported one or more severe adverse events. Conclusions The mRNA-1273 vaccine induced anti–SARS-CoV-2 immune responses in all participants, and no trial-limiting safety concerns were identified. These findings support further development of this vaccine. (Funded by the National Institute of Allergy and Infectious Diseases and others; mRNA-1273 ClinicalTrials.gov number, NCT04283461).
Article
The liposomes have continued to be well-recognized as an important nano-sized drug delivery system with attractive properties, such a characteristic bilayer structure assembling the cellular membrane, easy-to-prepare and high bio-compatibility. Extensive effort has been devoted to the development of liposome-based drug delivery systems during the past few decades. Many drug candidates have been encapsulated in liposomes and investigated for reduced toxicity and extended duration of therapeutic effect. The liposomal encapsulation of hydrophilic and hydrophobic small molecule therapeutics as well as other large molecule biologics have been established among different academic and industrial research groups. To date, there has been an increasing number of FDA-approved liposomal-based therapeutics together with more and more undergoing clinical trials, which involve a wide range of applications in anticancer, antibacterial, and antiviral therapies. In order to meet the continuing demand for new drugs in clinics, more recent advancements have been investigated for optimizing liposomal-based drug delivery system with more reproducible preparation technique and a broadened application to novel modalities, including nucleic acid therapies, CRISPR/Cas9 therapies and immunotherapies. This review focuses on the recent liposome’ preparation techniques, the excipients of liposomal formulations used in various novel studies and the routes of administration used to deliver liposomes to targeted areas of disease. It aims to update the research in liposomal delivery and highlights future nanotechnological approaches.
Article
On June 28, 2018, the Committee for Medicinal Products for Human Use adopted a positive opinion, recommending the granting of a marketing authorization for the medicinal product Vyxeos, intended for the treatment of acute myeloid leukemia (AML). Vyxeos was designated as an orphan medicinal product on January 11, 2012. The applicant for this medicinal product was Jazz Pharmaceuticals Ireland Limited. Vyxeos is a liposomal formulation of a fixed combination of daunorubicin and cytarabine, antineoplastic agents that inhibit topoisomerase II activity and also cause DNA damage. The strength of Vyxeos is 5 units/mL, where 1 unit equals 1.0 mg cytarabine plus 0.44 mg daunorubicin. The marketing authorization holder Jazz Pharmaceuticals had found that this was an optimal ratio for the efficacy of the product. Study CLTR0310-301, a phase III, multicenter, randomized, trial of Vyxeos (daunorubicin-cytarabine) liposome injection versus standard 3+7 daunorubicin and cytarabine in patients aged 60–75 years with untreated high-risk (secondary) AML, showed a statistically significant difference between the two groups in overall survival (OS) with a median OS of 9.56 months in the daunorubicin-cytarabine arm compared with 5.95 months for standard chemotherapy (hazard ratio, 0.69; 95% confidence interval, 0.52–0.90; one-sided p = .003). The most common side effects were hypersensitivity including rash, febrile neutropenia, edema, diarrhea/colitis, mucositis, fatigue, musculoskeletal pain, abdominal pain, decreased appetite, cough, headache, chills, arrhythmia, pyrexia, sleep disorders, and hypotension. Implications for Practice Vyxeos has demonstrated a clinically significant improvement in overall survival compared with the standard of care 7+3 in the proposed population of patients with newly diagnosed acute myeloid leukemia (AML) with myelodysplasia-related changes and therapy-related AML. This is remarkable given the very poor prognosis of these patients and their unmet medical need. Secondary endpoints support the primary outcome, in particular an increased rate of hematopoietic stem cell transplantation, which is potentially the only curative treatment in AML.
Article
We present the new and entirely mechanistic COSMOperm method to predict passive membrane permeabilities for neutral compounds, as well as anions and cations. The COSMOperm approach is based on compound specific free energy profiles within a biomembrane of interest from COSMO-RS (Conductor-like Screening Model for Realistic Solvation) calculations. These are combined with membrane layer specific diffusion coefficients, for example, in the water phase, the polar head groups and the alkyl tails of biochemical phospholipid bilayers. COSMO-RS utilizes first-principle quantum chemical structures and physically sound intermolecular interactions (electrostatic, hydrogen bond and van der Waals). For this reason, it is unbiased towards different application scenarios, such as cosmetics, industrial chemical or pharmaceutical industries. A fully predictive calculation of passive permeation through phospholipid bilayer membranes results in a performance of r2 = 0.92; rmsd = 0.90 log10 units for neutral compounds and anions, as compared to gold standard black lipid membrane (BLM) experiments. It will be demonstrated that new membrane types can be generated by the related COSMOplex method and directly used for permeability studies by COSMOperm.