Accepted Manuscript Lipophilic peptide character – What oral barriers fear the most
Ožbej Zupančič, Andreas Bernkop-Schnürch PII: DOI: Reference:
S0168-3659(17)30553-9 doi: 10.1016/j.jconrel.2017.04.038 COREL 8783
To appear in:
Journal of Controlled Release
Received date: Revised date: Accepted date:
19 February 2017 21 April 2017 25 April 2017
Please cite this article as: Ožbej Zupančič, Andreas Bernkop-Schnürch , Lipophilic peptide character – What oral barriers fear the most. The address for the corresponding author was captured as affiliation for all authors. Please check if appropriate. Corel(2017), doi: 10.1016/j.jconrel.2017.04.038
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT Lipophilic peptide character – what oral barriers fear the most Ožbej Zupančič, Andreas Bernkop-Schnürch* Review article Department of Pharmaceutical Technology, Institute of Pharmacy, Leopold-Franzens-
PT
University Innsbruck, Innrain 80/82, Center for Chemistry and Biomedicine, 6020 Innsbruck,
Corresponding author:
SC
*
RI
Austria / Europe
Department of Pharmaceutical Technology
Leopold-Franzens-University Innsbruck
NU
Institute of Pharmacy
MA
Innrain 80/82, Center for Chemistry and Biomedicine
PT E
Tel.: +43-512- 507-58601
D
6020 Innsbruck, Austria / Europe
Fax: +43-512-507- 58699
AC
CE
E-Mail:
[email protected]
ACCEPTED MANUSCRIPT Keywords: lipophilicity, hydrophobic ion pairing, REAL, cyclization, esterification, presystemic metabolism, intestinal permeation. Abstract Peptide therapeutics is currently one of the fastest growing markets worldwide and
PT
consequently convenient ways of administration for these drugs are highly on demand. In particular, oral dosage forms would be preferred. A relative large molecular weight and high
RI
hydrophilicity, however, result in comparatively very low oral bioavailability being in most
SC
cases below 1%. Lipid based formulations (LBF), in particular self-emulsifying drug delivery systems (SEDDS) and solid lipid nanoparticles (SLN) as well as liposomes are among the
NU
most promising tools for oral peptide delivery. Key to success in orally delivering peptides via
MA
LBF seems to be a sufficiently high lipophilic character of those therapeutic agents. Hence, different non-covalent and covalent peptide lipidization methods from drug delivery point of
D
view are presented. On the one hand, among non-covalent lipidization methods hydrophobic
PT E
ion pairing seems to be a promising way to sufficiently increase peptide lipophilicity providing high drug payloads in the lipid phase, a protective effect against presystemic metabolism via thiol-disulphide exchange reactions and proteolysis as well as an improved
CE
intestinal membrane permeability. On the other hand, covalent methods like conjugating fatty
AC
acids via amidation, esterification, reversible aqueous lipidization (REAL) and cyclization also show potential. The present review therefore describes those lipidization methods in detail and critically evaluates their contribution in successfully overcoming the oral barriers.
ACCEPTED MANUSCRIPT 1. Introduction Recent advances in biotechnology have led to rapid increase in synthesis and commercialization of numerous peptide drugs [1]. Currently more than 600 peptide drugs are in preclinical or clinical trials, giving the global peptide market a high growth potential [2,3]. On the one hand, peptides have advantages over conventional small molecule drugs including
PT
high potency, selectivity, excellent safety profile, tolerability and fewer side effects [1,4]. On
RI
the other hand, the majority of them has to be administered via injections being related with
SC
pain, discomfort and consequently low patient compliance [1]. Oral peptide delivery is indeed an ongoing challenge, which is evident from numerous published strategies over the past
NU
decades [3,5]. Comparing the data of so far available in vivo studies and clinical trials, delivering peptides via LBF such as (micro)emulsions including SEDDS [6–12], SLN [13–
MA
20] and liposomes [21–33] is likely one of the most promising strategies. Increasing intestinal membrane permeability, modulation of tight junctions and reducing proteolytic degradation
D
are just some of the benefits when utilizing lipid excipients in oral peptide delivery [3].
PT E
However, only few oral peptide formulations containing lipophilic auxiliary agents have up to date reached clinical trials or commercialization [34,35]. The only commercialized oral
CE
peptide LBF Sandimune Neoral® contains cyclosporine dissolved in lipid preconcentrate,
AC
forming an o/w emulsion under in vivo conditions, where the peptide remains incorporated in the lipid matrix of LBF. Following this example, this review will focus therefore just on one important aspect having a substantial impact on the success of oral peptide delivery strategies – namely the lipophilic character of the therapeutic peptide. 2. Lipidization 2.1 Hydrophilic character of peptides and its consequences
ACCEPTED MANUSCRIPT Peptide chemical structure has two fundamental disadvantages when it comes to oral administration – relative high molecular weight and high hydrophilicity [35–40]. The former restricts peptides to paracellular absorption, which comprises of a very small percentage of the total epithelial surface area [37,41–43]. The latter, on the other hand, prevents the therapeutic peptides from permeating the phospholipid bilayer of epithelial cell membranes as
PT
they are forced to break its hydrogen bonds with solvating water in order to interact with the
RI
lipid bilayer [37,43–45]. As a consequence of this low membrane permeability systemic
SC
uptake via the passive transcellular route is strongly limited. Thus, the key factor in increasing peptide lipophilicity is the reduction of its hydrogen bonding potential [44]. Furthermore,
NU
orally administered peptides containing disulphide bonds may interact in the intestine with endogenous glutathione and thiol substructures of food via thiol-disulphide exchange
MA
reactions leading to peptide conformational changes and subsequently their inactivation [46,47]. Moreover, an insufficient lipophilic character exposes peptides to harsh presystemic
D
metabolism by intestinally secreted trypsin, α-chymotrypsin, elastase, carboxypeptidases A
PT E
and B as well as membrane bound peptidases leading to their inactivation [48–51]. Additionally, polar functional groups of peptides lead to ionic interactions and hydrogen
CE
bonding with intestinal content and mucus contributing to a low oral bioavailability and high variability in drug uptake [44]. Indeed, the mucus layer covers the luminal surface of the
AC
gastrointestinal tract and is composed up to 95% of water as well as cross-linked and entangled mucin fibres, carbohydrates, DNA, lipids, salts, antibodies, bacteria and cellular debris [52]. As such, mucus acts as a filter for charged hydrophilic peptides either by size exclusion mechanism or ionic peptide-mucin interactions, resulting in poor peptide mucus permeation [53]. However, there are two opposing strategies to improve peptide diffusion through mucus – the mucoadhesive and the mucodiffusion principle [52]. Firstly, incorporating peptides in mucoadhesive polymers and their thiolated counterparts was shown
ACCEPTED MANUSCRIPT to facilitate peptide diffusion through mucus layer due to close contact of polymer matrix with absorption membrane, providing steep concentration gradient. Moreover, polymer matrix can protect peptides against proteolysis in GIT as well as offers controlled drug release [3,54]. On the contrary, mucodiffusive LBF with neutral or negatively charged surface show even greater potential [3]. The hydrophilic PEG surface corona of LBF enhances their mucus diffusion and
PT
also prevents their interaction with biological enzymes and mucin [3]. From oral peptide
RI
delivery point of view, it would be highly beneficial if LBF contain the peptide drug in its
SC
lipid matrix. Accordingly, a high lipophilic character of orally given peptides seems to be essential.
NU
2.2 How to measure lipophilicity
MA
Lipophilicity is defined as the affinity of a molecule or moiety for a lipophilic environment. Indeed, as from the drug delivery point of view it is advantageous to incorporate therapeutic
D
peptides in lipophilic carrier systems such as SEDDS, SLN and liposomes, the determination
PT E
of their solubility in lipophilic excipients used in these formulations is essential. Lipophilicity is commonly measured by peptide distribution behaviour in a biphasic system, which can be liquid-liquid or solid-liquid. On the one hand, the gold standard for direct liquid-liquid phase
CE
lipophilicity characterization of neutral substances is the octanol/water partitioning coefficient
AC
(log P) [55,56]. However, since most of peptides are ionized, distribution coefficient (log D) expressing the contribution of all neutral and ionized species at given pH, is utilized. For simplicity reasons, in the scope of this review, log P values of peptides are compared. Octanol is used due to its structural similarity with phospholipids in the cell membrane. However, octanol is also a known hydrogen bond donor/acceptor and therefore alternative solvents with different hydrogen bond properties such as heptane, chloroform, cyclohexane and propylene glycol dipelargonate have been proposed [57,58]. One the other hand, since chromatographic retention of substances in a reversed phase column and n-octanol/water partitioning are
ACCEPTED MANUSCRIPT energetically analogous, reversed phase chromatography is becoming the preferential indirect solid-liquid phase method for determining log P [56,59]. Octadecyl-bonded silica (C-18 RPHPLC) and immobilized artificial membranes mimicking the phospoholipid bilyer (IAM HPLC) are normally used as stationary phases. Here, a prolonged retention time of lipidized peptide is observed with respect to the native peptide [60–63]. From difference in retention
PT
times the representative lipophilicity factor, log kw, which is directly related to log P, can be
RI
calculated [56]. Noteworthy, peptide lipophilicity can be also determined by potentiometric
SC
titration, where the peptides are partitioned between liposomal membrane and water and the dissociation constants (pKa) are determined by adding high precision titrators [56]. In
NU
addition, fast and simple indirect methods to measure lipophilicity include thin layer chromatography (TLC) [56] and various computational methods [64].
MA
2.3 Covalent vs. non-covalent lipidization
D
Lipidization not only alters peptides lipophilicity, but it also may have a substantial impact on
PT E
its secondary structures, self-assembling properties and its ability to bind to target receptors. Moreover, a significant change in ADME properties including metabolic and plasma stability, membrane permeability and bioavailability can be expected [55]. In some cases, these
CE
changes may cause partial or total loss of the peptides biological activity [61,65,66]. From the
AC
commercial point of view, non-covalent lipidiziation is preferred. On the one hand, noncovalent lipidization offers a straightforward lipophilicity increase excluding intensive labour with multiple reactions and purification steps. Additionally, non-covalent interactions enable native peptide regeneration in vivo, restoring its secondary and tertiary structures and thus preserving its affinity to target receptors. Moreover, the peptide might not be regarded as a new active pharmaceutical ingredient (API) as it would definitely be the case for all covalently lipidized peptides. On the other hand, the shortcoming of such labile non-covalent bonds is the dissociation of peptide-complex in vivo before it reaches the desired target [67–
ACCEPTED MANUSCRIPT 69]. In such cases, reversible covalent lipidization methods, where the parent peptide is regenerated in vivo, is recommended. In the following chapters, different strategies to increase the lipophilic character of peptide drugs including non-covalent and covalent lipidization
CE
PT E
D
MA
NU
SC
RI
PT
approaches as illustrated in Figure 1 are described.
AC
Figure 1: Schematic presentation of non-covalent and covalent peptide lipidization methods. On the one hand, lipophilicity can be increased via ionic interactions between charged amino acids and oppositely charged surfactants. On the other hand, covalent methods utilize peptide primary amino groups, hydroxyl groups and disulphide bonds.
ACCEPTED MANUSCRIPT 3. Non-covalent lipidization Peptides can interact non-covalently with lipidization agents through hydrogen bonding, hydrophobic interactions, complexation with divalent metal ions and ionic interactions, forming water insoluble complexes. A typical example of hydrogen bonding and hydrophobic interactions are tannines. For instance, it was shown in the previous studies that protein-tannin
PT
complexes are stable at gastrointestinal pH and in the presence of proteolytic enzymes such as
RI
pepsin, trypsin, α- chymotrypsin, elastase and carboxypeptidase A and B as well as bile salts
SC
[70,71]. Furthermore, metal ions such as Zn2+, Cu2+ or Ni2+ are able to form coordinative complexes with peptides, which cause conformational changes and stabilize the peptide
NU
structure [72–74]. Oxytocin, for example, undergoes a conformational change when coordinated with Cu2+ or Zn2+. This causes the carbonyl oxygens of the ring to be directed
MA
towards the core reducing the peptides hydrogen bonding potential and rendering its surface more hydrophobic [73,75]. Also, Zn2+ is well known to coordinate with peptides, forming
D
water insoluble complexes. In previous studies, various water insoluble zinc-peptide
gonadotropin-releasing
PT E
complexes such as with insulin [76–78], hirudin [79,80], dalarein [81,82], buserelin [83], hormone
[81],
thyrotropin-releasing
hormone
[81],
CE
adenocorticothropic hormone [81] and thyroliberine [82] have been prepared. However, the
AC
most efficient approach for non-covalent peptide lipidization seems to be via ionic interactions, where peptide lipophilicity is increased by peptide net charge neutralization by anionic, amphoteric or cationic surfactants. 3.1 Interactions between peptides and surfactants Peptide-surfactant interactions are a complex process, dependent on peptides primary structure, pH, ionic strength, temperature, surfactant type (ionic, non-ionic) as well as on the surfactant to peptide molar ratio (S/P) [84]. Various well established analytical techniques to measure such interactions as presented in Table 1.
ACCEPTED MANUSCRIPT Table 1: Methods to measure peptide-surfactant interactions Technique
Description
Ref.
Morphology
Scanning Electron Microscopy X-ray Powder Diffraction
The morphological changes of unprocessed peptide, surfactants, physical peptide and surfactant mixture and PSC are compared
[85] [85] [86]
Peptide backbone and secondary structure
UV-circular dichroism Fourier transform infrared spectroscopy (FT-IR)
Near UV and far UV circular dichroism spectra are recorded in order to determine possible peptide backbone and secondary structure changes due to ionic interactions with surfactants. Spectra of peptide and PSC are compared to determine secondary structure of peptide in the PSC.
[84,85,87] [88]
Ionic interactions between peptide and surfactant
Fourier transform infrared spectroscopy (FT-IR)
DSC and FT-IR spectra of peptide, surfactant, physical mixture of peptide and surfactant and peptide-surfactant ion pair are compared.
Lipophilicity (noncovalent peptidesurfactant interactions Lipophilicity (covalent peptide-surfactant interactions)
RI
Stoichiometry of binding
Differential scanning calorimetry (DSC) Isothermal titration calorimetry
PT
Parameter
[85–87,89] [14,85]
[84,87]
Partitioning/distribution coefficient (log P/log D) (“shake flask method”)
Extraction of tested substance fromwater/1-octanol. Determination of concentation of the drug in aqueous and organic phase
[56,90,91]
Reverse phase highperformance liquid chromatography (RP-HPLC)
Partitioning of subsatances between aqueous mobile phase and C-18 RP-HPLC or IAM HPLC stationary phase. Lipophilicity is calculated from the retention time shift between the substance and its analogues
[56,92,93]
D
MA
NU
SC
Electrostatic binding of surfactants to peptides is an exothermic process whereas peptide unfolding is an endothermic process. By measuring the heat-flow the stoichiometry of binding could be determined
PT E
Indeed, this review primarily is focused on ionic interactions between peptides and surfactants as they result in significant lipophilicity increase of the peptide-surfactant complex (PSC).
CE
There are different concentration dependent mechanisms of surfactant binding on the peptide surface. Initially, below the critical micelle concentration (CMC), ionic surfactants bind non-
AC
cooperatively as monomers to the peptide native state [94]. Polar head groups of individual surfactants bind on oppositely charged side chains via electrostatic interactions, whereas alkyl chains interact through hydrophobic interactions with nearby hydrophobic patches [95,96]. The alkyl chain length has an impact on the degree of binding and stabilization of the complex [97]. With increasing surfactant amounts, the initial surfactant binding sites are saturated and the excess of surfactant molecules such as sodium dodecyl sulphate (SDS) may promote the peptide to unfold and aggregate. This leads to formation of clusters, which are stabilized by the association of peptide aggregates, leading to formation of shared micelles
ACCEPTED MANUSCRIPT [84,98]. Further addition of surfactant provides sufficient number of molecules for each peptide to form its own cluster. Above CMC, typically at 100 surfactant molecules per each peptide molecule, cooperative surfactant binding takes place, where interactions between peptides and micelles are predominant [84]. Such interactions are more common between peptides and non-ionic surfactants, where the micelles interact with the peptide hydrophobic
PT
surface [99–101]. Recently, non-ionic surfactants are being extensively investigated as the
RI
main component of niosomes, a cheap and more stable alternative for liposomes, where
SC
phospholipids are substituted with non-ionic surfactants [102]. For instance, physically stable niosomes with droplet size 95.7 nm and negative zeta potential containing peptides have been
NU
designed [103]. In addition, orally administered niosomes containing insulin-Tat complex showed significant increase in hypoglycemic activity in mice [104].
MA
3.2 Hydrophobic ion pairing
D
Many peptides contain at least one ionisable amino acid. At physiological pH, lysine, arginine
PT E
and histidine are positively charged, whereas glutamic and aspartic acid are negatively charged. By adjusting the pH value above their isoelectric point, peptides pose a negative net charge and vice versa, by decreasing pH below isoelectric point yields peptides of a positive
CE
net charge. Lipophilicity of such ionized peptides can be effectively increased by hydrophobic
AC
ion pairing [105–107]. Here, ionic interactions between charged peptides and oppositely charged surfactants lead to the formation of water insoluble PSC. In the scope of this review, surfactants are classified according to their charge in anionic, cationic and amphoteric. In Table 2 structures of representative surfactants, which successfully formed water-insoluble PSC with indicated peptides are presented. Table 2: Structures of representative surfactants, which successfully formed lipophilic complexes with indicated peptides. Type
Class
Structural formula
Name
Peptide
Ref.
ACCEPTED MANUSCRIPT Anionic
Fatty acids Sodium decanoate
Octreotide
[108]
Vancomycin
[88]
Desmopressin, leuprolide
[14,90]
[108–112]
Sodium oleate
Leuprolide, octreotide, salmon calcitonin, desmopressin
Sodium dodecylsulp hate
Desmopressin, octreotide, insulin, bivalirudin
[87,90,108,1 13–115,132]
Sodium laurate Sodium stearate
PT
Organo sulphates
RI
Sodium octadecylsul phate
MA
NU
Bile acids
AC
CE
PT E
D
Salicylatefatty acid conjugates
Alkyl phosphates
Phospholi pids
Amphote ric
Phospholi pids
Cationic
Fatty amines
Sodium docusate
Desmopressin,lanre otide, insulin, bivalirudin
[90,116,117, 132]
Sodium deoxycholat e
Octreotide, lanreotide
[108,116,118 ]
Lanreotide
[116]
Salmon calcitonin
[119,120]
SC
Organo sulphonate s
[90,116]
Desmopressin, lanreotide
Sodium taurocholate
Sodium N[8-(2hydroxybenz oyl)amino]c aprylate (SNAC)
[121,122]
8-(N-2hydroxy-5chlorobenzyl)aminocapryli c acid (5CNAC)
Sodium hexadecylph osphate
Thymopentin
[123]
DMPG
Salmon calcitonin Insulin bivalirudin
[112] [89] [132]
Phosphatidyl choline
Insulin
[85,86,124– 127]
Dodecylami ne chloride
Daptomycin, bivalirudin
[91,132]
ACCEPTED MANUSCRIPT Quarterly amines Distearyldim ethylammoni um bromide
Insulin
[85]
Most therapeutic peptides contain at least one cationic centre and could be therefore ion paired with anionic counter ions. Anionic surfactants such as fatty acids are preferred due to
PT
their ability to open tight junctions, binding to fatty acid binding proteins as well as solubilizing membranes and thus enhance drug permeation. In addition, fatty acids may also
RI
mask peptides from the gastrointestinal environment and eluding first pass effect by
SC
disguising the drug as a triglyceride as well as carry them across mucus as part of their
NU
absorption [128,129]. Moreover, fatty acids are generally considered as safe [130]. In contrast, ion pairing of anionic peptides with lipophilic cationic counter ions is more rarely
MA
used due to their potential toxicity [35]. Nonetheless, there are numerous reports about the use of fatty amines such as dodecylamine chloride [91,131,132], hexylamine [133] and quaternary
D
amines such as distearyldimethylammonium bromide [85], dodecyltrimethylamonium
PT E
chloride, cetylprydinium chloride, stearalkonium chloride and cetrimide [133]. In addition, cationization of bile salts via diethylamine spacer like for example Nα-deoxycholyl-L-lysyl-
CE
methylester [134] or deoxycholylethylamine [135] might be used as a safer alternative. Also, a likely promising alternative to cationic surfactants for the ionic complexation of peptides
AC
exhibiting an anionic net charge are amphoteric surfactants. On the one hand, they are still capable of ion pairing with anionic peptides and on the other hand most of them can be regarded as safe.
Hydrophobic ion pairing efficiency is a function of pH, which determines peptides net charge and ionization state of peptide and surfactant functional groups. However, it seems that the S/P also plays a prominent role in peptide complexation efficiency and the lipophilicity of the PSC. So far, SDS has been most commonly used as ion pairing agent. It was observed that an
ACCEPTED MANUSCRIPT equimolar ratio of SDS led to complexes of comparatively highest lipophilicity [113,136,137]. Similarly, practically quantitative complex formation of antibiotic peptide daptomycin was achieved at equimolar amounts of dodecylamine [91]. In contrast, as illustrated in Figure 2, the most efficient complex formation of desmopressin, which contains
MA
NU
SC
RI
PT
one cationic centre was at S/P of 1.8 [90].
D
Figure 2: Hydrophobic ion pairing of desmopressin acetate with sodium octadecyl sulphate
PT E
(○), sodium stearate (▲), sodium dodecyl sulphate (□) sodium oleate (●) and sodium docusate (■). After the addition of surfactants, the precipitated desmopressin complex was
CE
centrifuged and the amount of remaining peptide in supernatant determined by HPLC. Data are shown as mean ± SD (n =3).
AC
Furthermore, the optimum S/P ratio of 3.0 was observed when peptides with two cationic centres – lanreotide and leuprolide – were ion paired with sodium oleate and sodium deoxycholate [109,116]. An explanation for this phenomenon might be given by peptidesurfactant aggregation and mixed micelles formation as described above. In addition, when large excess of surfactant is added to PSC solution, its aqueous solubility is regained. Indeed, with increasing amounts of surfactant beyond the critical micellar concentration (CMC) micelles are formed, which most probably resolubilize the PSC. This assumption was
ACCEPTED MANUSCRIPT confirmed by performing the inverse experiment – adding increasing amounts of insulin to an excess of SDS. At the beginning of the experiment, where the amount of SDS was in great excess, no PSC was observed, indicating its immediate solubilization via micellization [87]. As mentioned earlier, PSC lipophilicity is also a function of S/P as illustrated in Figure 3. For instance, daptomycin log P was increased with dodecylamine chloride from - 5.0 at S/P 0:1 to
PT
+4.8 at S/P 4:1[91], whereas ion paring of lanreotide with sodium deoxyclolate increased its
RI
log P from -10.0 at S/P 0:1 to remarkable 9.0 at S/P 3:1[116]. In addition, the surfactant itself
SC
also has an impact on PSC lipophilicity. Desmopressin log P of -6.13, for instance, was increased to a positive value of 0.33 when using SDS and sodium docusate, but remained
CE
PT E
D
MA
NU
negative (log P = -1.80) when sodium oleate was used at S/P 1.8:1[90].
AC
Figure 3: (A) Log P (octanol/water) increase of daptomycin dodecylamine complex (●), lanreotide deoxycholate complex (■) and lanreotide docusat complex (□) as a function of surfactant : peptide molar ratio. (B) Log P (Capmul 907 P/water) increase of desmopressin oleate complex (●), desmopressin dodecyl sulphate complex (■) and desmopressin docusat complex (□) as a function of S/P. In relation to that, increased lipophilic nature of PSC enables its incorporation in the lipid matrix of LBF. Table 3 summarizes most representative examples, where hydrophobic ion
ACCEPTED MANUSCRIPT pairing resulted in successful peptide incorporation in LBF. For instance, 8.0% and 10.5% ion pair complexes, corresponding to 5.5% and 6.4% of daptomycin and lanreotide were dissolved in SEDDS formulations [91,116]. Moreover, ion pairing of thymopentin with hexadecylphosphate increased the apparent log P of the complex 280-fold with respect to bare peptide and subsequently the drug payload in SLN, which was increased from 1.7% to 5.2%,
PT
respectively [123]. In connection to that, ion pairing of vancomycin with lauric acid increased
RI
peptides log P from -1.41 to 1.37, yielding SLN with a 0.1% drug payload [88]. Also, PSC
SC
drug payloads of about 10% (w/w) can be achieved with high amounts of hydrophilic
NU
surfactants and co-solvents in LBF [138].
Table 3: LBS containing peptide-surfactant ion pair and their characteristics. Counter ion
LBF
Formulation
Desmopressin
Sodium docusate
SEDDS
40% Cremophor RH40 50% Capmul 907 P 10% Transcutol
Insulin
Dimyristoyl phosphatidylglycerol (DMPG)
SEDDS
Soyphosphatidylcholine
SEDDS
Leuprolide
Result
Ref.
Increased desmopressin docusate complex log P (Capmul 907 P/ water) from -6.30 to 0.33 SEDDS protected desmopressin against α– chymotrypsin and glutathione degradation
[90]
33% Labrafil 1944 47% Cremophor EL 20% Transcutol
1.13%
SEDDS protected insulin against in vitro trypsin and α – chymotrypsin degradation Sustained insulin release form SEDDS Mucus permeating SEDDS
[89]
35% Cremophor EL 32.5% ethyl oleate 32.5% ethanol
0.30%
The relative insulin oral bioavailability of SEDDS congaing insulin was 7-fold increased compared to oral insulin PBS solution
D
PT E
CE
Lanreotide
AC
β - Lactamase
Drug Payload 0.08%
MA
Peptide
Soyphosphatidylcholine
SEDDS
42% Lauroglycol FCC, 33% Cremophor EL 25% Transcutol
0.17%
The protein-phospholipid complex in SEDDS resulted in a relative bioavailability of 6.34%, which was 2.6-fold higher compared to protein in aqueous solution
Sodium deoxycholate
SEDDS
25% Capmul MCM 45% Mygliol 840 30% Cremophor EL
10.5%
25% Captex 8000 30% Capmul MCM 30% Kolliphor EL 15% Propylene glycol
9.7%
Increased lanreotide deoxycholate log P (octanol/ water) to 9.0 Sustained lanreotide release form SEDDS SEDDS protected lanreotide against in vitro glutathione and thiol-enriched casein,peptones thiol-disulphide exchange reactions
30% Ethyl oleate 30% Capmul MCM
0.4%
Sodium oleate
SEDDS
SEDDS protected leuprolide against in vitro trypsin and α –
[139]
[140] [141] [142]
[116]
[109].[110]
ACCEPTED MANUSCRIPT EP 30% Cremophor RH40 10% Ethanol 30% Captex355 30% Capmul MCM EP 30% Cremophor EL 10% Propylene glycol 33.3% Brij 97 33.3% Oleic acid 33.3% Ethanol
Dodecylamine hydrochloride
SEDDS
Vancomycin
Lauric acid
SLN
Thymopentin
Sodium hexadecyl phosphate
SLN
Leuprolide
Sodium stearate
Lipase stable SEDDS
57% olive oil 38%Tween 80 3% propylene glycol
2.0%
75.5% paraffin 25%, Brij 35 8.0% propylene glycol 1.5%, ethanol
2.0%
73.1 ± 8.1% of octreotidedeoxycholate complex was found in the octanol phase. Sustained release of octreotide from SEDDS for at least 24 h, Pancreatic lipase stable SEDDS
30% Dermofeel MCT 30% Capmul MCM 30% Cremophor EL 10% propylene glycol
[110]
[108]
Increased daptomycindodecylamine complex log P (octanol/ water) from -5.0 to 4.8 SEDDS protected daptomycin against in vitro α – chymotrypsin degradation Mucus permeating SEDDS
[91]
0.1%
Increased vancomycin log P from -1.41 to 1.37 and higher encapsulation of the drug in the lipid core of SLN
[88]
3.9% egg lecithin 9.7% sodium turodeoxycholate 4.0% butanol 5.9% stearic acid 1.5% sodium hexadecylphosphate
5.2%
Apparent log P increase by 280fold with respect to bare peptide and subsequently the drug payload in SLN
[123]
Stearic acid Liquid paraffin Span-80
-
Sustained drug release from SLN
[14]
MA
D
5% Compritol 888 ATO 3% Lutrol F68
PT E
CE
SLN
PT
0.4%
RI
SEDDS
[109] in vivo study in rats showed a 17.2-fold improved oral bioavailability of SEDDS containing leuprolide oleate compared to leuprolide acetate solution
8.0%
NU
Daptomycin
Sodium deoxycholate
0.4%
SC
Octreotide
chymotrypsin degradation
AC
4. Covalent lipidization
Most of peptides contain at least one primary amino, hydroxyl or carboxyl functional group, which can be converted to amides and esters. Additionally, disulphide bonds can also be manipulated to attach lipid moieties [143–147]. “The classical approach” to lipidize peptides is via amide bond formation between the primary amino group of the N-terminus or lysine subunits and a fatty acid [148–155]. Interestingly, coupling a fatty acid to tyrosine hydroxyl group via O-etherification was also reported [156]. Moreover, stabile ethers can also be
ACCEPTED MANUSCRIPT synthesized through S-ether formation. Such method was utilized in lipidizing salmon calcitonin with ε-maleimido lysine derivative of palmitic acid [62]. Worth mentioning is also a powerful iodoacetamide derivatization method, used mainly in analytical field [63,157]. Some lipophilic peptide derivatives have already been synthesized by this pathway and according to the increased lipophilicity of the synthesized derivatives as illustrated in Table 4,
PT
the iodoacetamide method might also find potential in oral peptide delivery.
Peptide sequence
Log kb
Log P calc.a
-12.75
0
-8.86
0.45
-8.28
0.55
-6.50
0.78
a
Log kb
-15.83
0
-8.45
0
-13.88
0.69
-4.56
1.30
-13.59
0.77
-3.98
1.30
-12.70
1.02
-2.20
1.30
0.92
-12.50
1.09
-1.80
1.32
1.22
-10.72
1.17
1.75
1.53
MA
D
CE
-2.54
Log P calc.a
PT E
-6.10
Log kb
NU
Log P calc.a
SC
substituent R
RI
Table 4: lipophilicity increase of the model peptides with iodoacetamide derivatization [63].
Calculated log P values with chemAxon software (www.chemicalize.org) Retention factor calculated from retention times of derivatives with respect to iodoacetamide derivatization
b
AC
4.1 Reversible aqueous lipidization (REAL) To keep the advantages of lipidization without taking the risk to lose the conjugates biological activity, various studies have utilized the REAL technology by forming in vivo labile disulphide bonds between model peptides and amphiphilic PEG-stearic acid conjugate 2-(2(2-(hexadecyloxy)ethoxy)ethoxy)ethyl 3-(2-(pyridin-2-yl)disulfanyl) propanoate or cysteinepalmitic acid conjugate N-palmitoyl-S-(pyridin-2-ylthio)cysteine. Such conjugates were shown to quantitatively regenerate the parent peptide in vivo. The common REAL synthetic pathway, where the peptide is lipidized in aqueous medium is presented in Figure 4. REAL
ACCEPTED MANUSCRIPT technology has already been successfully used on some model peptides and proteins containing disulphide bonds [61,62,93,158–160]. For instance, REAL lipidization of desmopressin (log P = -6.13) with various fatty acids yielded in increased RP-HPLC retention times and capacity factors of the lipidized derivatives. The most lipophilic palmitic acid derivatives with log P values of 5.65 and 5.32 were too lipophilic to be eluted from the In addition, in vivo results showed that REAL
PT
column with the chosen method [161].
RI
lipidization might be a promising tool in orally delivering peptide and protein drugs. Orally
SC
administered REAL lipidized calcitonin, for example, showed a 19-fold increased area under curve compared to unmodified peptide [92]. Nevertheless, one limitation of REAL technology
NU
is its restriction to peptide drugs containing disulphide bonds. However, most of therapeutically significant peptide drugs contain at least one primary amino group. Thus, an
MA
amine-reacting lipidizing reagent based on REAL concept, 3,4-bis(decylthiomethyl)-2,5furandione was used to reversibly modify Leu5-enkephalin. The lipidized conjugate showed a
D
significantly increased antinociception effect using a rodent inflammatory pain model. After
PT E
oral administration, the peak concentration and area under the curve of the conjugate were 4.4
AC
CE
and 21-fold higher compared to Leu5-enkephalin, respectively [162].
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
PT E
Figure 4: Common synthetic pathway in REAL lipidization. The water soluble lipidized derivatives are water soluble and converted to the parent drug under in vitro and in vivo
4.2 Cyclization
CE
conditions.
AC
Conventional linear peptides are charged under physiological conditions due to C- or Nterminal ends. As such, linear peptides show poor membrane permeability and high proteolytic instability compared to their cyclic counterparts [163,164]. In contrast, peptide cyclization was shown to alter peptides physicochemical properties such as charge, conformation, hydrophobicity and subsequently led to improved oral absorption [165–167]. Lipophilicity of such cyclic derivatives, commonly referred to as macrocycles [168], is most probably increased due to restricted conformational freedom, leading to formation of
ACCEPTED MANUSCRIPT intramolecular hydrogen bonds. Additionally, more lipophilic non-charged cyclic peptides can traverse the cell membrane by passive diffusion via transcellular pathway [169]. The poster child of cyclization technology is cyclosporine [163,168,170]. Although cyclization can be performed in different ways [171–173], the use of cyclization linkers as illustrated in Table 5 has shown to increase lipophilicity and Caco-2 monolayer permeation of cyclic peptides
RI
PT
compared to their linear counterparts.
SC
Table 5: Partitioning coefficient and Caco-2 monolayer permeation increase between linear
Linker
Structure
NU
encephalins and their cyclic counterparts.
Leu5-enkephalin
Papp x10-6 [cm/s] < 0.0031
log k'IAMa 0.17
Leu5-enkephalin cyclic prodrug
2.66 ± 0.12 ____
2.70 -
DADLE
0.086 ± 0.005
0.43
-1.30
3.16 ± 0.26
2.91
2.98
H-Try-Ala-GlyGly-Asp-AlaOH
< 0.17
-1.10
-5.20
H-Try-Ala-GlyGly-Asp-AlaOH cyclic prodrug
12.97 ± 1.48
-0.32
-2.24
Leu5-enkephalin
< 0.31
0.17
-1.86
Leu5-enkephalin cyclic prodrug
1.80
1.36
0.92
DADLE
0.78
0.43
-1.30
1.86
1.63
1.49
Leu5-enkephalin
< 0.0031
0.17
-1.86
Leu5-enkephalin cyclic prodrug
13.3 ± 0.426
3.32
3.94
DADLE
0.086 ± 0.005
0.43
-1.30
8.27 ± 0.51
3.32
4.51
Peptide
MA
Coumarinic acid
cyclic
D
DADLE prodrug
AC
CE
PT E
(Acyloxy)alkoxy
Phenylpropionic acid
DADLE prodrug
DADLE prodrug a
cyclic
cyclic
---
Log Pcalcb
Ref.
-1.86
[169]
2.41-- ---------------
[174,175]
[176]
Partitioning coefficient between 0.01 M phosphate buffer pH 7.4 and an immobilized artificial membrane column, composed of phosphatidylcholine analogues (IAM.PD.DD) b Partitioning coeficent calculated withChemAxon software (ww.chemicalize.org)
ACCEPTED MANUSCRIPT 4.3 Esterification with fatty acids A great advantage of forming peptide O- or S- esters with fatty acids is their weak enzymatic or chemical stability, regenerating the parent drug upon hydrolysis in vivo [177]. In previous studies, several prodrugs of desmopressin have been prepared by forming O-esters with tyrosine hydroxyl group using various fatty acids. The lipophilicity and the transport of ester
PT
prodrugs across Caco-2 monolayer was remarkably higher compared to desmopressin alone as
RI
shown in Table 6 [178]. Furthermore, O-esterification of tyrosine hydroxyl group with
NU
dalargin and dermorphin derivative [179–181].
SC
palmitic acid was shown to increase the oral bioavailability of leu5-enkephalin analogue,
MA
Table 6: Increased log P and Caco-2 permeation of desmopressin and its corresponding ester
D
prodrugs [178].
log Pa
log Pcalcb
Papp x 10-7 [cm/s]
Enhancement ratio
< -3.5
-6.20
5.15 ± 1.67
-
< -3
-5.43
9.44 ± 1.54
1.8
-2.25
-4.33
15.1 ± 1.1
2.9
-1.25
-4.10
9.77 ± 1.19
1.9
0.41
-3.21
8.50 ± 1.60
1.7
O-2-Ethylhexanoyl desmopressin
0.09
-3.11
ND
ND
O-isobutyloxycarbonyl desmopressin
-1.77
-4.27
5.99 + 0.57
PT E
Peptide Desmopressin O-propionyl desmopressin O-pivaloyl desmopressin
a
AC
O-octanoyl desmopressin
CE
O-hexanoyl desmopressin
1.2
0
experimental partitioning coefficient between octanol and aqueous buffer pH 7.40 and 21 C Partitioning coeficent calculated with ChemAxon software (ww.chemicalize.org)
b
5. Impact of lipidization on thiol-disulphide reactions Intestinal thiol compounds such as glutathione, cysteine, N-acetyl cysteine and homocysteine usually originate from food [182]. In addition, endogenous glutathione is present in most mucosal tissues as a part of antioxidant defence system [183,184]. Thus, peptides containing thiol or disulphide bonds can undergo chemical degradation via thiol/disulphide exchange
ACCEPTED MANUSCRIPT reactions with intestinal thiol compounds. To preserve the integrity of disulphide bonds, peptides containing disulphide bonds can be incorporated in LBF. Since the concept of so called “sulfhydryl barrier” is relatively fresh, only a handful of studies demonstrating protective effect of LBF against thiol/disulphide exchange reactions are available. For instance, two disulphide bond containing peptides, lanreotide and desmopressin were
PT
incorporated in different SEDDS via ion pairing and emulsified in reduced glutathione
RI
solution [90,116]. Figure 5 illustrates the protective effect of SEDDS containing PSC against
SC
glutathione degradation. The decelerated peptide degradation was observed by all SEDDS formulations except one, which contained 80% hydrophilic surfactants. It seemed that the
NU
increased amount of surfactants enabled diffusion of hydrophilic glutathione in the droplets, inactivating desmopressin [90]. Furthermore, an impact of potential thiol containing
MA
substances from protein dietary source on lanreotide inactivation was tested, where lanreotide was incubated in thiol enriched casein hydrolysed peptones. Similarly, the stability of
D
lanreotide in SEDDS was significantly higher compared to lanreotide in aqueous solution
PT E
[116]. On the other hand, the disulphide bonds may be protected by “preactivation” via REAL lipidization, where the peptide would remain protected against thiol compounds in
CE
gastrointestinal tract. The parent peptide would be regenerated under in vitro and in vivo
AC
conditions by reduction of labile disulphide bonds [160].
ACCEPTED MANUSCRIPT Figure 5: (A) Protective effect of two different SEDDS formulations containing lanreotide deoxycholate against thiol-disulphide exchange reactions with reduced glutathione (□; composed of 25% Capmul MCM, 30% Cremophor EL, 40% Myglyol 840 and ○, composed of 25% Capmul MCM, 30% Cremophor EL, 25% Captex 8000 and 15% propylene glycol) and thiol-enriched peptones (■,●) compared with blank lanreotide (▲). (B) Protective effect
PT
of SEDDS (○, 40% Cremophor RH40, 50% Capmul 907 P, 10% Transcutol) containing
RI
desmopressin docusate complex (○) against thiol-disulphide exchange reactions with reduced
SC
glutathione compared with blank desmopressin (▲).
NU
6. Impact of lipidization on peptide proteolytic stability
MA
The major barrier for orally administered peptides to overcome is the enzymatic barrier. The luminally secreted and brush border membrane enzymes, as well as the cytosolic enzymes of
D
the absorption membrane play prominent role in the peptide presystemic metabolism [48,49].
PT E
More specifically, trypsin, which cleaves basic amino acids arginine and lysine and αchymotrypsin, cleaving aromatic amino acids phenylalanine and tyrosine are among the most
CE
important pancreatic proteases responsible for rapid peptide degradation. Various in vitro studies have demonstrated that LBF can decrease the rate of proteolysis when the peptide is
AC
incorporated in LBF. Figure 6 depicts a typical degradation profile of leuprolide incorporated in SEDDS by trypsin and α-chymotrypsin [110]. Additionally, prolonged proteolytic stability of peptide drugs in SEDDS was also observed among desmopressin [90], daptomycin [91], dalargin [185], insulin [89]. Furthermore, modified SLN [17,186–191] and liposomes [32,192–194] containing peptide drugs also demonstrated protective effects against enzymatic degradation in the gastrointestinal tract. In addition, covalently lipidized peptides showed prolonged proteolytic stability when administered orally. For instance, a 12-fold increased half-time of REAL lipidized Leu5-enkephalin in mouse small intestinal mucosal homogenate
ACCEPTED MANUSCRIPT was observed with respect to native Leu5-enkephalin[162]. REAL, thioether and PEGylated thioether salmon calcitonin derivatives were shown to be significantly more stable in intestinal fluid with respect to native peptide [62,195]. Furthermore, fatty acid acylation of thyrotropin-releasing hormone, tetragastrin and insulin on the amino terminus improved the stability of N-terminus lipidized derivatives against intestinal enzymatic degradation
D
MA
NU
SC
RI
PT
[196,197].
PT E
Figure 6: Protective effect of SEDDS containing leuprolide oleate in three different formulations (□; 33% Brij 97, 33% oleic acid, 34% ethanol ■; 30% ethyl oleate, 30% Capmul
CE
MCM, 30% Cremophor RH40, 10% ethanol ●; 30% Captex 355, 30% Capmul MCM, 30% Cremophor EL, 10% propylene glycol) with respect to blank leuprolide (▲) against α-
AC
chymotrypsin (A) and trypsin (B). 7. Impact of lipidization on peptide intestinal permeation One of the crucial rate limiting factors in oral peptide delivery is the intestinal epithelium, which is comprised of a single monolayer of cells. A generally accepted in vitro model to assess intestinal drug absorption is the Caco-2 cell monolayer model [198]. Using the Caco-2 model, insulin and rhPTH1-34 in SEDDS showed up to 12-fold and 7-fold increased permeation with respect to peptide solutions, respectively [199–201]. Furthermore, SEDDS
ACCEPTED MANUSCRIPT containing β-lactamase showed a 24.9-fold increased diffusion of β-lactamase through MDCK monolayer compared with β-lactamase solution, respectively [142]. In relation to that, SLN decorated with cationic cell penetrating peptide octaarginine containing insulin increased the Caco-2 permeability by up to 18.44 times [16]. The use of such cationic cell penetrating peptides is indeed a remarkable way to boost LBF cellular uptake. For instance, SEDDS
PT
loaded with covalently lipidized HIV-1 Tat-protein 49-57 with oleoyl chloride showed 1.7-
RI
fold improved transfection efficiency for pDNA compared to Lipofectin [202]. However,
SC
covalent peptide lipidization without using cell penetrating peptides also significantly improved peptide Caco-2 permeability. For instance, Caco-2 permeability of REAL lipidized
NU
Bowmann-Birk protease inhibitor and Leu5-enkephalin were increased 140-fold and 131-fold compared to native peptides [66,162]. In addition, fatty acid acylation of tetragastrin on the
MA
amino termini improved the amount of acetyl, butyl and hexyl tetragatrin transported for 13fold, 19-fold and 18-fold, respectively [203]. Also, S-ether of salmon calcitonin with ε-
D
maleimido lysine derivative of palmitic acid showed a 2-fold higher cellular uptake than
PT E
native peptide in Caco-2 model. Additionally, oral administration of conjugate suppressed plasma calcium levels (60% of baseline) for up to 10 h [60,62].
CE
8. Oral delivery lipid-based formulations for peptides
AC
The most commonly investigated LBF used in oral peptide delivery are SEDDS, SLN and liposomes. All these LBF show common characteristics favourable for oral peptide delivery such as controlled drug release properties, biocompatibility, protective effect against proteolysis and enhanced absorption though intestinal epithelium [3,34]. However, they differ in some significant features as illustrated in Table 7. In order to understand the parameters influencing the choice of LBF for effective oral delivery, their physico-chemical features and limitations should be taken into consideration.
ACCEPTED MANUSCRIPT Table 7: LBF used in oral peptide delivery and their properties [3,34].
Description
Composition
SEDDS
Isotropic mixture of lipids, surfactants and co-solvents, spontaneously forming o/w emulsion upon mixing with water.
Middle chain mono-, di- and triglycerides
Advantages
Disadvantages
Simple preparation procedure
Poor in vitro - in vivo correlation
W/O/W double emulsion
Spontaneous selfemulsification
Limited peptide drug payload
Melt dispersion
Thermodynamically stable
Dilution effect in vivo may cause phase inversion and drug leakage
Hydrophobic surfactants (HLB < 12)
RI
Mucus permeability
Hydrophilic surfactants (HLB > 12)
Could be filled in soft/hard gelatine capsules
SC
Peptides dissolved in the oily preconcentrate
Long chain and medium chain fatty acids
Peptide entrapment methods Hydrophobic ion pairing
Co-solvents Long-chain triglycerides
W/O/W double emulsion
Partial glycerides
Hydrophobic ion pairing
Fatty acids
Reverse micelles
Waxes
Melt dispersion
Phospholipids
Limited peptide payload in the lipid core
Ease of large scale production – facile and liable scale up
Unpredictable gelation tendency of SLN dispersions
Low toxicity due to omitting organic solvents
Aqueous core with one or more amphiphilic bilayers made of phospholipids and cholesterol.
Phospholipids (dipalmytoyl phosphatidylcholine, distearoyl phosphatidylcholine ,dicetyl phosphate)
Hydrophilic peptides are incorporated in the inner aqueous core, lipophilic peptides in the lipid bilyer, amphiphilic peptides are partitioned between both phased
Cholesterol
AC
Liposomes
CE
PT E
Steroids
Reduced drug leakage due to solid core
D
Peptides are dissolved or physically dispersed in the solid core.
NU
Solid state at room and body temperature, lipid core with emulsifier layer
MA
SLN
PT
LBF
Saturated and unsaturated fatty acids
Film hydration or reverse-phase evaporation, followed by particle size reduction via sonication, extrusion, high pressure homogenization Detergent dialysis Solvent injection method
Restructuring process can lead to potential drug expulsion and the inability to achieve a prolonged release of the encapsulated drug
High biocompatibility
Low stability in the GIT
Weak immunogenicity and low intrinsic toxicity (natural phospholipids)
High production costs
High specificity for localized delivery
Limited drug payload capacity of the aqueous core and bilayer Leakage of the incorporated peptide drug Sterilization difficult due to phospholipid sensitivity to radiation and heat Difficult batch-tobatch
ACCEPTED MANUSCRIPT
9. Lipid-based formulations as oral vaccine delivery systems
PT
reproducibility and large scale manufacturing
RI
As many antigens are peptides, they are exposed to same barriers as »classic peptides« as
SC
presented above. To illicit an immune response and achieve mucosal immunization, orally administered antigens must reach M-cells, which represent only about 1% of intestinal
NU
epithelial cells and lie below the level of villous tips [204]. Thus, repeated high doses of
MA
antigens must be applied to achieve an immune response. However, such methods usually do not induce systemic immunity, but may lead only to a short-lived secretion of antigen-specific
D
IgA and the development of systemic tolerance. In addition, high doses of antigens may cause
PT E
severe allergic reactions and toxic effects [204,205]. However, effective immunization with low doses of antigens is feasible by co-administration of adjuvants, which enhance the immune response. Certain LBF have shown to be versatile adjuvants with an excellent safety
CE
profile. In addition, there are already some examples of marketed vaccines containing LBF as
AC
adjuvants such as emulsions and liposomes as well as immune-stimulating complexes (ISCOMs) [206,207]. Emulsions usually contain squalene oil as adjuvants, whereas ISCOMs resemble liposomes, being composed of cholesterol, phospholipids and saponine adjuvants, in particular Quil-A or QS-21 [206]. Although limited data on oral delivery of peptide vaccine are available [208,209], peptide lipidization might be a promising approach in achieving mucosal and systemic immunity. For instance, covalent lipidization of a peptide antigen derived from the E7 oncoprotein of human papillomavirus type 16 on with palmitic acid resulted in significantly increased antigen entrapment efficiency in liposomes, from initial
ACCEPTED MANUSCRIPT 27.1% to 94.1%, respectively [210]. Hence, incorporation of peptides in such LBF would
PT
provide the added value of immune response.
10. Concluding remarks
RI
Oral peptide delivery is still an ongoing challenge due to several physiological barriers
SC
including presystemic metabolism by thiol-disulphide exchange reactions and proteolysis as well as poor intestinal membrane permeability. However, it seems the key to successfully
NU
overcome these barriers lies in sufficient peptide lipophilicity. In this review, systematically
MA
structured ways to increase peptide lipophilicity were presented. From drug delivery point of view, the lipidization via non-covalent hydrophobic ion pairing and incorporation in LBF
D
seems to bare the highest potential due to its reversibility, simplicity and efficiency. In
PT E
addition, covalent methods also offer efficient ways in protecting peptides against presystemic metabolism and increasing intestinal permeability. Moreover, a simultaneous combination of
AC
the lipid core.
CE
two or more different lipidization strategies may lead to a higher peptide or protein payload in
11. References
[1] Wang J, Yadav V, Smart AL, Tajiri S, Basit AW. Toward oral delivery of biopharmaceuticals: an assessment of the gastrointestinal stability of 17 peptide drugs. Molecular Pharmaceutics 2015;12:966–73. [2] Kaspar AA, Reichert JM. Future directions for peptide therapeutics development. Drug Discovery Today 2013;18:807–17. [3] Niu Z, Conejos-Sánchez I, Griffin BT, O’Driscoll CM, Alonso MJ. Lipid-based nanocarriers for oral peptide delivery. Advanced Drug Delivery Reviews 2016; 106,:337-354. [4] Fosgerau K, Hoffmann T. Peptide therapeutics: current status and future directions. Drug Discovery Today 2015;20:122–8.
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[5] Griffin BT, O'Driscoll CM. Opportunities and challenges for oral delivery of hydrophobic versus hydrophilic peptide and protein-like drugs using lipid-based technologies. Therapeutic Delivery 2011;2:1633–53. [6] Sharma G, Wilson K, van der Walle CF, Sattar N, Petrie JR, Kumar MR. Microemulsions for oral delivery of insulin: design, development and evaluation in streptozotocin induced diabetic rats. European Journal of Pharmaceutics and Biopharmaceutics 2010;76:159–69. [7] Elsayed A, Al Remawi M, Qinna N, Farouk A, Badwan A. Formulation and characterization of an oily-based system for oral delivery of insulin. European Journal of Pharmaceutics and Biopharmaceutics 2009;73:269–79. [8] Onuki Y, Morishita M, Takayama K. Formulation optimization of water-in-oil-water multiple emulsion for intestinal insulin delivery. Journal of Controlled Release 2004;97:91–9. [9] Dogru ST, Calis S, Öner F. Oral multiple w/o/w emulsion formulation of a peptide salmon calcitonin: in vitro‐ in vivo evaluation. Journal of Clinical Pharmacy and Therapeutics 2000;25:435–43. [10] Sun H, Liu K, Liu W, Wang W, Guo C, Tang B, Gu J et al. Development and characterization of a novel nanoemulsion drug-delivery system for potential application in oral delivery of protein drugs. International Journal of Nanomedicine 2012;7:5529. [11] Cheng M-B, Wang J-C, Li Y-H, Liu X-Y, Zhang X, Chen D-W, Zhou S-F et al. Characterization of water-in-oil microemulsion for oral delivery of earthworm fibrinolytic enzyme. Journal of Controlled Release 2008;129:41–8. [12] Constantinides PP, Scalart J-P, Lancaster C, Marcello J, Marks G, Ellens H, Smith PL. Formulation and intestinal absorption enhancement evaluation of water-in-oil microemulsions incorporating medium-chain glycerides. Pharmaceutical Research 1994;11:1385–90. [13] Chen C, Fan T, Jin Y, Zhou Z, Yang Y, Zhu X, Zhang Z-r et al. Orally delivered salmon calcitonin-loaded solid lipid nanoparticles prepared by micelle-double emulsion method via the combined use of different solid lipids. Nanomedicine 2013;8:1085–100. [14] Yuan H, Jiang S-P, Du Y-Z, Miao J, Zhang X-G, Hu F-Q. Strategic approaches for improving entrapment of hydrophilic peptide drugs by lipid nanoparticles. Colloids and Surfaces B: Biointerfaces 2009;70:248–53. [15] Kashanian S, Rostami E. PEG-stearate coated solid lipid nanoparticles as levothyroxine carriers for oral administration. Journal of Nanoparticle Research 2014;16:1–10. [16] Zhang Z-H, Zhang Y-L, Zhou J-P, Lv H-X. Solid lipid nanoparticles modified with stearic acid–octaarginine for oral administration of insulin. International Journal of Nanomedicine 2012;7:3333. [17] Fan T, Chen C, Guo H, Xu J, Zhang J, Zhu X, Yang Y et al. Design and evaluation of solid lipid nanoparticles modified with peptide ligand for oral delivery of protein drugs. European Journal of Pharmaceutics and Biopharmaceutics 2014;88:518–28. [18] Yang R, Gao R, Li F, He H, Tang X. The influence of lipid characteristics on the formation, in vitro release, and in vivo absorption of protein-loaded SLN prepared by the double emulsion process. Drug Development and Industrial Pharmacy 2011;37:139–48. [19] Yang R, Gao R-C, Cai C-F, Xu H, Li F, He H-B, Tang X. Preparation of gel-core-solid lipid nanoparticle: a novel way to improve the encapsulation of protein and peptide. Chemical and Pharmaceutical Bulletin 2010;58:1195–202. [20] Christophersen PC, Zhang L, Yang M, Nielsen HM, Müllertz A, Mu H. Solid lipid particles for oral delivery of peptide and protein drugs I–Elucidating the release mechanism of lysozyme during lipolysis. European Journal of Pharmaceutics and Biopharmaceutics 2013;85:473–80. [21] Agrawal AK, Harde H, Thanki K, Jain S. Improved stability and antidiabetic potential of insulin containing folic acid functionalized polymer stabilized multilayered liposomes following oral administration. Biomacromolecules 2013;15:350–60. [22] Li Z, Chen J, Sun W, Xu Y. Investigation of archaeosomes as carriers for oral delivery of peptides. Biochemical and Biophysical Research Communications 2010;394:412–7. [23] Makhlof A, Fujimoto S, Tozuka Y, Takeuchi H. In vitro and in vivo evaluation of WGA– carbopol modified liposomes as carriers for oral peptide delivery. European Journal of Pharmaceutics and Biopharmaceutics 2011;77:216–24.
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[24] Thongborisute J, Tsuruta A, Kawabata Y, Takeuchi H. The effect of particle structure of chitosan-coated liposomes and type of chitosan on oral delivery of calcitonin. Journal of Drug Targeting 2006;14:147–54. [25] Takeuchi H, Matsui Y, Sugihara H, Yamamoto H, Kawashima Y. Effectiveness of submicronsized, chitosan-coated liposomes in oral administration of peptide drugs. International Journal of Pharmaceutics 2005;303:160–70. [26] Parmentier J, Thewes B, Gropp F, Fricker G. Oral peptide delivery by tetraether lipid liposomes. International Journal of Pharmaceutics 2011;415:150–7. [27] Zhang X, Qi J, Lu Y, He W, Li X, Wu W. Biotinylated liposomes as potential carriers for the oral delivery of insulin. Nanomedicine: Nanotechnology, Biology and Medicine 2014;10:167–76. [28] Niu M, Lu Y, Hovgaard L, Guan P, Tan Y, Lian R, Qi J et al. Hypoglycemic activity and oral bioavailability of insulin-loaded liposomes containing bile salts in rats: the effect of cholate type, particle size and administered dose. European Journal of Pharmaceutics and Biopharmaceutics 2012;81:265–72. [29] Niu M, Tan Y, Guan P, Hovgaard L, Lu Y, Qi J, Lian R et al. Enhanced oral absorption of insulin-loaded liposomes containing bile salts: a mechanistic study. International Journal of Pharmaceutics 2014;460:119–30. [30] Hu S, Niu M, Hu F, Lu Y, Qi J, Yin Z, Wu W. Integrity and stability of oral liposomes containing bile salts studied in simulated and ex vivo gastrointestinal media. International Journal of Pharmaceutics 2013;441:693–700. [31] Moghassemi S, Parnian E, Hakamivala A, Darzianiazizi M, Vardanjani MM, Kashanian S, Larijani B et al. Uptake and transport of insulin across intestinal membrane model using trimethyl chitosan coated insulin niosomes. Materials Science and Engineering: C 2015;46:333–40. [32] Zhang N, Ping QN, Huang GH, Xu WF. Investigation of lectin-modified insulin liposomes as carriers for oral administration. International Journal of Pharmaceutics 2005;294:247–59. [33] Li H, An JH, Park J-S, Han K. Multivesicular liposomes for oral delivery of recombinant human epidermal growth factor. Archives of Pharmacal Research 2005;28:988–94. [34] Karamanidou T, Bourganis V, Kammona O, Kiparissides C. Lipid-based nanocarriers for the oral administration of biopharmaceutics. Nanomedicine 2016;11:3009–32. [35] Moroz E, Matoori S, Leroux J-C. Oral delivery of macromolecular drugs: Where we are after almost 100 years of attempts. Advanced Drug Delivery Reviews 2016;101:108-121. [36] Renukuntla J, Vadlapudi AD, Patel A, Boddu SHS, Mitra AK. Approaches for enhancing oral bioavailability of peptides and proteins. International Journal of Pharmaceutics 2013;447:75–93. [37] Goldberg M, Gomez-Orellana I. Challenges for the oral delivery of macromolecules. Nature Reviews Drug Discovery 2003;2:289–95. [38] Rehman K, Akash MS, Akhtar B, Tariq M, Mahmood A, Ibrahim M. Delivery of Therapeutic Proteins: Challenges and Strategies. Current Drug Targets 2015;17(10): 1172-1188. [39] Rekha, Sharma CP. Oral delivery of therapeutic protein/peptide for diabetes–future perspectives. International Journal of Pharmaceutics 2013;440:48–62. [40] Mahajan A, Rawat AS, Bhatt N, Chauhan MK. Structural Modification of Proteins and Peptides. Indian Journal of Pharmaceutical Education and Research 2014;48:34–47. [41] Laffleur F, Bernkop-Schnürch A. Strategies for improving mucosal drug delivery. Nanomedicine 2013;8:2061–75. [42] Cone RA. Barrier properties of mucus. Advanced Drug Delivery Reviews 2009;61:75–85. [43] Fricker G, Drewe J. Current concepts in intestinal peptide absorption. Journal of Peptide Science 1996;2:195–211. [44] Di L. Strategic approaches to optimizing peptide ADME properties. The AAPS Journal 2015;17:134–43. [45] Witt KA, Gillespie TJ, Huber JD, Egleton RD, Davis TP. Peptide drug modifications to enhance bioavailability and blood-brain barrier permeability. Peptides 2001;22:2329–43. [46] Schmitz T, Huck CW, Bernkop-Schnürch A. Characterisation of the thiol–disulphide chemistry of desmopressin by LC, μ-LC, LC-ESI-MS and Maldi-Tof. Amino Acids 2006;30:35– 42. [47] Leonaviciute G, Bernkop-Schnürch A. Self-emulsifying drug delivery systems in oral (poly) peptide drug delivery. Expert Opinion on Drug Delivery 2015;12:1703–16.
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[48] Bernkop-Schnürch A. The use of inhibitory agents to overcome the enzymatic barrier to perorally administered therapeutic peptides and proteins. Journal of Controlled Release 1998;52:1– 16. [49] Bernkop-Schnurch A, Schmitz T. Presystemic metabolism of orally administered peptide drugs and strategies to overcome it. Current Drug Metabolism 2007;8:509–17. [50] Werle M, Bernkop-Schnürch A. Strategies to improve plasma half life time of peptide and protein drugs. Amino Acids 2006;30:351–67. [51] Sousa IP de, Bernkop-Schnürch A. Pre-systemic metabolism of orally administered drugs and strategies to overcome it. Journal of Controlled Release 2014;192:301–9. [52] Netsomboon K, Bernkop-Schnürch A. Mucoadhesive vs. mucopenetrating particulate drug delivery. European Journal of Pharmaceutics and Biopharmaceutics 2016;98:76–89. [53] Zhang J, Lv Y, Wang B, Zhao S, Tan M, Lv G, Ma X. Influence of Microemulsion–Mucin Interaction on the Fate of Microemulsions Diffusing through Pig Gastric Mucin Solutions. Molecular Pharmaceutics 2015;12:695–705. [54] Bernkop-Schnürch A. The use of multifunctional polymers for non-invasive peptide and protein application. Expert Opinion on Therapeutic Patents 2000;10:1357–66. [55] Zhang L, Bulaj G. Converting peptides into drug leads by lipidation. Current Medicinal Chemistry 2012;19:1602–18. [56] Rutkowska E, Pajak K, Jóźwiak K. Lipophilicity--methods of determination and its role in medicinal chemistry. Acta Poloniae Pharmaceutica 2012;70:3–18. [57] El Tayar N, Mark AE, Vallat P, Brunne RM, Testa B, van Gunsteren WF. Solvent-dependent conformation and hydrogen-bonding capacity of cyclosporin A: evidence from partition coefficients and molecular dynamics simulations. Journal of Medicinal Chemistry 1993;36:3757– 64. [58] Leahy DE, Taylor PJ, Wait AR. Model solvent systems for QSAR Part I. Propylene glycol dipelargonate (PGDP). A new standard solvent for use in partition coefficient determination. Quantitative Structure‐ Activity Relationships 1989;8:17–31. [59] Sabatka JJ, Minick DJ, Shumaker TK, Hodgson GL, Brent DA. Measurement of lipophilicity by high-performance liquid chromatography: Comparison with calculated lipophilicity values. Journal of Chromatography A 1987;384:349–56. [60] Cheng W, Satyanarayanajois S, Lim L-Y. Aqueous-soluble, non-reversible lipid conjugate of salmon calcitonin: synthesis, characterization and in vivo activity. Pharmaceutical Research 2007;24:99–110. [61] Cheng W, Lim L-Y. Lipeo-sCT: a novel reversible lipidized salmon calcitonin derivative, its biophysical properties and hypocalcemic activity. European Journal of Pharmaceutical Sciences 2009;37:151–9. [62] Cheng W, Lim L-Y. Comparison of reversible and nonreversible aqueous-soluble lipidized conjugates of salmon calcitonin. Molecular Pharmaceutics 2008;5:610–21. [63] Williams DK, Meadows CW, Bori ID, Hawkridge AM, Comins DL, Muddiman DC. Synthesis, characterization, and application of iodoacetamide derivatives utilized for the ALiPHAT strategy. Journal of the American Chemical Society 2008;130:2122–3. [64] Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Advanced Drug Delivery Reviews 2012;64:4–17. [65] Shen W-C, Wang J. Method and compositions for lipidization of hydrophilic molecules: U.S. Patent No. 6,093,692. Washington, DC: U.S. Patent and Trademark Office, 25 Jul. 2000 [66] Ekrami HM, Kennedy AR, Shen W-C. Water-soluble fatty acid derivatives as acylating agents for reversible lipidization of polypeptides. FEBS Letters 1995;371:283–6. [67] Patel A, Gaudana R, Mitra AK. A novel approach for antibody nanocarriers development through hydrophobic ion-pairing complexation. Journal of Microencapsulation 2014;31:542–50. [68] Gaudana R, Gokulgandhi M, Khurana V, Kwatra D, Mitra AK. Design and evaluation of a novel nanoparticulate-based formulation encapsulating a HIP complex of lysozyme. Pharmaceutical Development and Technology 2013;18:752–9. [69] Gaudana R, Khurana V, Parenky A, Mitra AK. Encapsulation of protein-polysaccharide HIP complex in polymeric nanoparticles. Journal of Drug Delivery 2011;2011.
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[70] Yan Q, Bennick A. Identification of histatins as tannin-binding proteins in human saliva. Biochemical Journal 1995;311:341–7. [71] Lu Y, Bennick A. Interaction of tannin with human salivary proline-rich proteins. Archives of Oral Biology 1998;43:717–28. [72] Joly L, Antoine R, Albrieux F, Ballivian R, Broyer M, Chirot F, Lemoine J et al. Optical and Structural Properties of Copper− Oxytocin Dications in the Gas Phase. The Journal of Physical Chemistry B 2009;113:11293–300. [73] Joly L, Antoine R, Allouche A-R, Broyer M, Lemoine J, Dugourd P. Optical properties of isolated hormone oxytocin dianions: ionization, reduction, and copper complexation effects. The Journal of Physical Chemistry A 2009;113:6607–11. [74] Jiang Y, Shi K, Xia D, Piao H, Quan P, Song T, Cui F. Protamine modified metal ion–protein chelate microparticles for sustained release of interferon. International Journal of Pharmaceutics 2011;407:31–7. [75] Liu D, Seuthe AB, Ehrler OT, Zhang X, Wyttenbach T, Hsu JF, Bowers MT. Oxytocinreceptor binding: why divalent metals are essential. Journal of the American Chemical Society 2005;127:2024–5. [76] Prabhu S, Jacknowitz AI, Stout PJ. A study of factors controlling dissolution kinetics of zinc complexed protein suspensions in various ionic species. International Journal of Pharmaceutics 2001;217:71–8. [77] Nelson RW, Henley K, Cole C. Field safety and efficacy of protamine zinc recombinant human insulin for treatment of diabetes mellitus in cats. Journal of Veterinary Internal Medicine 2009;23:787–93. [78] Della Maggiore A, Nelson RW, Dennis J, Johnson E, Kass PH. Efficacy of protamine zinc recombinant human insulin for controlling hyperglycemia in dogs with diabetes mellitus. Journal of Veterinary Internal Medicine 2012;26:109–15. [79] Gietz U, Arvinte T, Mader E, Oroszlan P, Merkle HP. Sustained release of injectable zincrecombinant hirudin suspensions: development and validation of in vitro release model. European Journal of Pharmaceutics and Biopharmaceutics 1998;45:259–64. [80] Gietz U, Arvinte T, Häner M, Aebi U, Merkle HP. Formulation of sustained release aqueous Zn–hirudin suspensions. European Journal of Pharmaceutical Sciences 2000;11:33–41. [81] Dolińska B, Ryszka F. Preparation and properties of selected Zn (II)–peptide complexes in suspension. Il Farmaco 2003;58:1131–6. [82] Ryszka F, Dolinska B. Pulsatile and sustained peptides release from Zn (II) suspensions. Bollettino Chimico Farmaceutico 2003;142:319–23. [83] Ryszka F, Dolińska B, Szulc B, Zientarska G. Preparation and properties of buserelin suspensions. Bollettino Chimico Farmaceutico 1994;133:156–9. [84] Otzen D. Protein–surfactant interactions: a tale of many states. Biochimica et Biophysica Acta (BBA)-Proteins and Proteomics 2011;1814:562–91. [85] Li P, Nielsen HM, Fano M, Müllertz A. Preparation and characterization of insulin–surfactant complexes for loading into lipid‐ based drug delivery systems. Journal of Pharmaceutical Sciences 2013;102(8), 2689-2698. [86] Cui F, Shi K, Zhang L, Tao A, Kawashima Y. Biodegradable nanoparticles loaded with insulin–phospholipid complex for oral delivery: preparation, in vitro characterization and in vivo evaluation. Journal of Controlled Release 2006;114:242–50. [87] Dai W-G, Dong LC. Characterization of physiochemical and biological properties of an insulin/lauryl sulfate complex formed by hydrophobic ion pairing. International Journal of Pharmaceutics 2007;336:58–66. [88] Kalhapure RS, Mocktar C, Sikwal DR, Sonawane SJ, Kathiravan MK, Skelton A, Govender T. Ion pairing with linoleic acid simultaneously enhances encapsulation efficiency and antibacterial activity of vancomycin in solid lipid nanoparticles. Colloids and Surfaces B: Biointerfaces 2014;117:303–11. [89] Karamanidou T, Karidi K, Bourganis V, Kontonikola K, Kammona O, Kiparissides C. Effective incorporation of insulin in mucus permeating self-nanoemulsifying drug delivery systems. European Journal of Pharmaceutics and Biopharmaceutics 2015;97:223–9.
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[90] Zupančič O, Leonaviciute G, Lam HT, Partenhauser A, Podričnik S, Bernkop-Schnürch A. Development and in vitro evaluation of an oral SEDDS for desmopressin. Drug Delivery 2016: 23(6), 2074-2083. [91] Zupančič O, Partenhauser A, Lam HT, Rohrer J, Bernkop-Schnürch A. Development and in vitro characterisation of an oral self-emulsifying delivery system for daptomycin. European Journal of Pharmaceutical Sciences 2016;81:129–36. [92] Wang J, Chow D, Heiati H, Shen W-C. Reversible lipidization for the oral delivery of salmon calcitonin. Journal of Controlled Release 2003;88:369–80. [93] Wang J, Shen D, Shen W-C. Preparation, purification, and characterization of a reversibly lipidized desmopressin with potentiated anti-diuretic activity. Pharmaceutical Research 1999;16:1674–9. [94] Moriyama Y, Kawasaka Y, Takeda K. Protective effect of small amounts of sodium dodecyl sulfate on the helical structure of bovine serum albumin in thermal denaturation. Journal of Colloid and Interface Science 2003;257:41–6. [95] Yonath A, Podjarny A, Honig B, Sielecki A, Traub W. Crystallographic studies of protein denaturation and renaturation. 2. Sodium dodecyl sulfate induced structural changes in triclinic lysozyme. Biochemistry 1977;16:1418–24. [96] Jones MN. Surfactant interactions with biomembranes and proteins. Chemical Society Reviews 1992;21:127–36. [97] Busti P, Scarpeci S, Gatti CA, Delorenzi NJ. Interaction of alkylsulfonate ligands with βlactoglobulin AB from bovine milk. Journal of Agricultural and Food Chemistry 1999;47:3628– 31. [98] Andersen KK, Oliveira CL, Larsen KL, Poulsen FM, Callisen TH, Westh P, Pedersen JS et al. The role of decorated SDS micelles in sub-CMC protein denaturation and association. Journal of Molecular Biology 2009;391:207–26. [99] Sjögren H, Ericsson CA, Evenäs J, Ulvenlund S. Interactions between charged polypeptides and nonionic surfactants. Biophysical Journal 2005;89:4219–33. [100] Jan BF. Polymer–micelle interactions: physical organic aspects. Chemical Society Reviews 1993;22:85–92. [101] Brackman JC, van Os NM, Engberts JB. Polymer-nonionic micelle complexation. Formation of poly (propylene oxide)-complexed n-octyl thioglucoside micelles. Langmuir 1988;4:1266–9. [102] Pardakhty A. Non-Ionic Surfactant Vesicles (Niosomes) as New Drug Delivery Systems. Pharmaceutical Sciences: Breakthroughs in Research and Practice: Breakthroughs in Research and Practice 2016:154. [103] Han BS, Kim SY, Lee KR, Seo HH, Moh SH. Stability of Various Liposome Formulation Containing the Phytochemical-Peptide Derivatives. Journal of the Society of Cosmetic Scientists of Korea 2016;42:135–43. [104] Manosroi A, Tangjai T, Sutthiwanjampa C, Manosroi W, Werner RG, Götz F, Sainakham M et al. Hypoglycemic activity and stability enhancement of human insulin–tat mixture loaded in elastic anionic niosomes. Drug Delivery 2016;23:3157–67. [105] Quintanar-Guerrero D, Allémann E, Fessi H, Doelker E. Applications of the ion-pair concept to hydrophilic substances with special emphasis on peptides. Pharmaceutical Research 1997;14:119–27. [106] Meyer JD, Manning MC. Hydrophobic ion pairing: altering the solubility properties of biomolecules. Pharmaceutical Research 1998;15:188–93. [107] Powers ME, Matsuura J, Brassell J, Manning MC, Shefter E. Enhanced solubility of proteins and peptides in nonpolar solvents through hydrophobic ion pairing. Biopolymers 1993;33:927–32. [108] Mahjub R, Dorkoosh FA, Rafiee-Tehrani M, Bernkop Schnürch A. Oral self-nanoemulsifying peptide drug delivery systems: impact of lipase on drug release. Journal of Microencapsulation 2015;32:401–7. [109] Hintzen F, Perera G, Hauptstein S, Müller C, Laffleur F, Bernkop-Schnürch A. In vivo evaluation of an oral self-microemulsifying drug delivery system (SMEDDS) for leuprorelin. International Journal of Pharmaceutics 2014;472:20–6.
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[110] Leonaviciute G, Zupančič O, Prüfert F, Rohrer J, Bernkop-Schnürch A. Impact of lipases on the protective effect of SEDDS for incorporated peptide drugs towards intestinal peptidases. International Journal of Pharmaceutics 2016; 508(1): 102-108. [111] Choi SH, Park TG. Hydrophobic ion pair formation between leuprolide and sodium oleate for sustained release from biodegradable polymeric microspheres. International Journal of Pharmaceutics 2000;203:193–202. [112] Sang Yoo H, Gwan Park T. Biodegradable nanoparticles containing protein‐ fatty acid complexes for oral delivery of salmon calcitonin. Journal of Pharmaceutical Sciences 2004;93:488–95. [113] Matsuura J, Powers ME, Manning MC, Shefter E. Structure and stability of insulin dissolved in 1-octanol. J. Am. Chem. Soc. 1993;115:1261–4. [114] Iqbal J, Vigl C, Moser G, Gasteiger M, Perera G, Bernkop-Schnürch A. Development and in vivo evaluation of a new oral nanoparticulate dosage form for leuprolide based on polyacrylic acid. Drug Delivery 2011;18:432–40. [115] Vaishya RD, Mandal A, Gokulgandhi M, Patel S, Mitra AK. Reversible hydrophobic ionparing complex strategy to minimize acylation of octreotide during long-term delivery from PLGA microparticles. International Journal of Pharmaceutics 2015;489:237–45. [116] Ijaz M, Bonengel S, Zupančič O, Yaqoob M, Hartl M, Hussain S, Huck CW et al. Development of oral self nano-emulsifying delivery system (s) of lanreotide with improved stability against presystemic thiol-disulfide exchange reactions. Expert Opinion on Drug Delivery 2016; 13(7), 923-929. [117] Alcock R, Blair JA, O’Mahony DJ, Raoof A, Quirk AV. Modifying the release of leuprolide from spray dried OED microparticles. Journal of Controlled Release 2002;82:429–40. [118] Rastogi R, Anand S, Koul V. Evaluation of pharmacological efficacy of 'insulin-surfoplex' encapsulated polymer vesicles. International Journal of Pharmaceutics 2009;373:107–15. [119] Aungst BJ. Absorption enhancers: applications and advances. The AAPS journal 2012;14:10– 8. [120] Karsdal MA, Byrjalsen I, Riis BJ, Christiansen C. Optimizing bioavailability of oral administration of small peptides through pharmacokinetic and pharmacodynamic parameters: the effect of water and timing of meal intake on oral delivery of Salmon Calcitonin. BMC Pharmacology and Toxicology 2008;8:5. [121] Karsdal MA, Byrjalsen I, Henriksen K, Riis BJ, Lau EM, Arnold M, Christiansen C. The effect of oral salmon calcitonin delivered with 5-CNAC on bone and cartilage degradation in osteoarthritic patients: a 14-day randomized study. Osteoarthritis and Cartilage 2010;18:150–9. [122] Maher S, Brayden DJ. Overcoming poor permeability: translating permeation enhancers for oral peptide delivery. Drug Discovery Today: Technologies 2012;9:e113-e119. [123] Morel S, Ugazio E, Cavalli R, Gasco MR. Thymopentin in solid lipid nanoparticles. International Journal of Pharmaceutics 1996;132:259–61. [124] Fricker G, Kromp T, Wendel A, Blume A, Zirkel J, Rebmann H, Setzer C et al. Phospholipids and lipid-based formulations in oral drug delivery. Pharmaceutical Research 2010;27:1469–86. [125] Maiti K, Mukherjee K, Gantait A, Saha BP, Mukherjee PK. Curcumin–phospholipid complex: preparation, therapeutic evaluation and pharmacokinetic study in rats. International Journal of Pharmaceutics 2007;330:155–63. [126] Yue P-F, Yuan H-L, Li X-Y, Yang M, Zhu W-F. Process optimization, characterization and evaluation in vivo of oxymatrine–phospholipid complex. International Journal of Pharmaceutics 2010;387:139–46. [127] Zhang J, Peng Q, Shi S, Zhang Q, Sun X, Gong T, Zhang Z. Preparation, characterization, and in vivo evaluation of a self-nanoemulsifying drug delivery system (SNEDDS) loaded with morinphospholipid complex. International Journal of Nanomedicine 2011;6:3405–14. [128] Smith PL, Wall DA, Gochoco CH, Wilson G. (D) Routes of delivery: case studies:(5) Oral absorption of peptides and proteins. Advanced Drug Delivery Reviews 1992;8:253–90. [129] Hackett MJ, Zaro JL, Shen W-C, Guley PC, Cho MJ. Fatty acids as therapeutic auxiliaries for oral and parenteral formulations. Advanced Drug Delivery Reviews 2013;65:1331–9.
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[130] Maher S, Leonard TW, Jacobsen J, Brayden DJ. Safety and efficacy of sodium caprate in promoting oral drug absorption: from in vitro to the clinic. Advanced Drug Delivery Reviews 2009;61:1427–49. [131] Zupančič O, Grieβinger JA, Rohrer J, Sousa IP de, Danninger L, Partenhauser A, Sündermann NE et al. Development, in vitro and in vivo evaluation of a self-emulsifying drug delivery system (SEDDS) for oral enoxaparin administration. European Journal of Pharmaceutics and Biopharmaceutics 2016;109:113–21. [132] Zupančič O, Grießinger JA, Lam HT, Bernkop-Schnürch A. Storage stability of bivalirudin: hydrophilic vs. lipophilic solutions. Journal of Pharmaceutical Sciences 2017, in press. [133] Hauptstein S, Prüfert F, Bernkop-Schnürch A. Self-nanoemulsifying drug delivery systems as novel approach for pDNA drug delivery. International Journal of Pharmaceutics 2015;487:25–31. [134] Kim SK, Lee S, Jin S, Moon HT, Jeon OC, Lee DY, Byun Y. Diabetes correction in pancreatectomized canines by orally absorbable insulin-deoxycholate complex. Molecular Pharmaceutics 2010;7:708–17. [135] Lee DY, Lee J, Lee S, Kim SK, Byun Y. Liphophilic complexation of heparin based on bile acid for oral delivery. Journal of Controlled Release 2007;123:39–45. [136] Hegg P-O. Precipitation of egg white proteins below their isoelectric points by sodium dodecyl sulphate and temperature. Biochimica et Biophysica Acta (BBA)-Protein Structure 1979;579:73–87. [137] Yoo HS, Choi H, Park TG. Protein–fatty acid complex for enhanced loading and stability within biodegradable nanoparticles. Journal of Pharmaceutical Sciences 2001;90:194–201. [138] Griesser J, Hetényi G, Moser M, Demarne F, Jannin V, Bernkop-Schnürch A. Hydrophobic ion pairing: Key to highly payloaded self-emulsifying peptide drug delivery systems. International Journal of Pharmaceutics 2017;520(1), 267-274. [139] Zhang Q, He N, Zhang L, Zhu F, Chen Q, Qin Y, Zhang Z et al. The in vitro and in vivo study on self-nanoemulsifying drug delivery system (SNEDDS) based on insulin-phospholipid complex. Journal of Biomedical Nanotechnology 2012;8:90–7. [140] Rao SVR, Yajurvedi K, Shao J. Self-nanoemulsifying drug delivery system (SNEDDS) for oral delivery of protein drugs: III. In vivo oral absorption study. International Journal of Pharmaceutics 2008;362:16–9. [141] Rao SVR, Shao J. Self-nanoemulsifying drug delivery systems (SNEDDS) for oral delivery of protein drugs: I. Formulation development. International Journal of Pharmaceutics 2008;362:2–9. [142] Rao SVR, Agarwal P, Shao J. Self-nanoemulsifying drug delivery systems (SNEDDS) for oral delivery of protein drugs: II. In vitro transport study. International Journal of Pharmaceutics 2008;362:10–5. [143] Cort JH, Schück O, Stříbrná J, Škopková J, Jošt K, Mulder JL. Role of the disulfide bridge and the C-terminal tripeptide in the antidiuretic action of vasopressin in man and the rat. Kidney International 1975;8:292–302. [144] Gazal S, Gelerman G, Ziv O, Karpov O, Litman P, Bracha M, Afargan M et al. Human somatostatin receptor specificity of backbone-cyclic analogues containing novel sulfur building units. Journal of Medicinal Chemistry 2002;45:1665–71. [145] Afargan M, Janson ET, Gelerman G, Rosenfeld R, Ziv O, Karpov O, Wolf A et al. Novel long-acting somatostatin analog with endocrine selectivity: potent suppression of growth hormone but not of insulin. Endocrinology 2001;142:477–86. [146] ORLOWSKI RC, EPAND RM, STAFFORD AR. Biologically potent analogues of salmon calcitonin which do not contain an N‐ terminal disulfide‐ bridged ring structure. European Journal of Biochemistry 1987;162:399–402. [147] Wang Y, Dou H, Cao C, Zhang N, Ma J, Mao J, Wu H. Solution structure and biological activity of recombinant salmon calcitonin S-sulfonated analog. Biochemical and Biophysical research Communications 2003;306:582–9. [148] Dasgupta P, Mukherjee R. Lipophilization of somatostatin analog RC‐ 160 with long chain fatty acid improves its antiproliferative and antiangiogenic activity in vitro. British Journal of Pharmacology 2000;129:101–9.
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[149] Timofeeva OA, Gaponenko V, Lockett SJ, Tarasov SG, Jiang S, Michejda CJ, Perantoni AO et al. Rationally designed inhibitors identify STAT3 N-domain as a promising anticancer drug target. ACS Chemical Biology 2007;2:799–809. [150] Dasgupta P, Singh AT, Mukherjee R. Lipophilization of somatostatin analog RC-160 improves its bioactivity and stability. Pharmaceutical Research 1999;16:1047–53. [151] Sato Y, Yang P, An Y, Matsukawa K, Ito K, Imanishi S, Matsuda H et al. A palmitoyl conjugate of insect pentapeptide Yamamarin arrests cell proliferation and respiration. Peptides 2010;31:827–33. [152] Asada H, Douen T, Mizokoshi Y, Fujita T, Murakami M, Yamamoto A, Muranishi S. Stability of acyl derivatives of insulin in the small intestine: relative importance of insulin association characteristics in aqueous solution. Pharmaceutical Research 1994;11:1115–20. [153] Asada H, Douen T, Waki M, Adachi S, Fujita T, Yamamoto A, Muranishi S. Absorption characteristics of chemically modified‐ insulin derivatives with various fatty acids in the small and large intestine. Journal of Pharmaceutical Sciences 1995;84:682–7. [154] Hashimoto M, Takada K, Kiso Y, Muranishi S. Synthesis of palmitoyl derivatives of insulin and their biological activities. Pharmaceutical Research 1989;6:171–6. [155] Kurtzhals P, Havelund S, Jonassen I, Kiehr B, Larsen UD, Ribel U, Markussen J. Albumin binding of insulins acylated with fatty acids: characterization of the ligand-protein interaction and correlation between binding affinity and timing of the insulin effect in vivo. Biochemical Journal 1995;312:725–31. [156] Maletinska L, Neugebauer W, Pérodin J, Lefebvre M, Escher E. Angiotensin analogues palmitoylated in positions 1 and 4. Journal of Medicinal Chemistry 1997;40:3271–9. [157] Yang Z, Attygalle AB. LC/MS characterization of undesired products formed during iodoacetamide derivatization of sulfhydryl groups of peptides. Journal of Mass Spectrometry 2007;42:233–43. [158] Honeycutt L, Wang J, Ekrami H, Shen W-C. Comparison of pharmacokinetic parameters of a polypeptide, the Bowman-Birk protease inhibitor (BBI), and its palmitic acid conjugate. Pharmaceutical Research 1996;13:1373–7. [159] Yuan L, Wang J, Shen W-C. Lipidization of human interferon-alpha: a new approach toward improving the delivery of protein drugs. Journal of Controlled Release 2008;129:11–7. [160] Yuan L, Wang J, Shen W-C. Reversible lipidization prolongs the pharmacological effect, plasma duration, and liver retention of octreotide. Pharmaceutical Research 2005;22:220–7. [161] Wang J, Wu D, Shen W-C. Structure-activity relationship of reversibly lipidized peptides: studies of fatty acid-desmopressin conjugates. Pharmaceutical Research 2002;19:609–14. [162] Wang J, Hogenkamp DJ, Tran M, Li W-Y, Yoshimura RF, Johnstone TBC, Shen W-C et al. Reversible lipidization for the oral delivery of leu-enkephalin. Journal of Drug Targeting 2006;14:127–36. [163] Rezai T, Yu B, Millhauser GL, Jacobson MP, Lokey RS. Testing the conformational hypothesis of passive membrane permeability using synthetic cyclic peptide diastereomers. Journal of the American Chemical Society 2006;128:2510–1. [164] Marepalli HR, Antohi O, Becker JM, Naider F. Solution Structures of i to i+ 3 cyclized model peptides: building blocks mimicking specific conformations. Journal of the American Chemical Society 1996;118:6531–9. [165] Wang B, Wang W, Zhang H, Shan D, Nimkar K, Gudmundsson O, Gangwar S et al. Synthesis and evaluation of the physicochemical properties of esterase‐ sensitive cyclic prodrugs of opioid peptides using coumarinic acid and phenylpropionic acid linkers. The Journal of Peptide Research 1999;53:370–82. [166] McMartin C, Hutchinson LEF, Hyde R, Peters GE. Analysis of structural requirements for the absorption of drugs and macromolecules from the nasal cavity. Journal of Pharmaceutical Sciences 1987;76:535–40. [167] Vlieghe P, Lisowski V, Martinez J, Khrestchatisky M. Synthetic therapeutic peptides: science and market. Drug Discovery Today 2010;15:40–56. [168] Kotz J. Bringing macrocycles full circle. SciBX: Science-Business eXchange 2012;5.
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[169] Gudmundsson OS, Pauletti GM, Wang W, Shan D, Zhang H, Wang B, Borchardt RT. Coumarinic acid-based cyclic prodrugs of opioid peptides that exhibit metabolic stability to peptidases and excellent cellular permeability. Pharmaceutical Research 1999;16:7–15. [170] Cain C. Excited about cycling: BioCentury; 2012. [171] Montalbetti CA, Falque V. Amide bond formation and peptide coupling. Tetrahedron 2005;61:10827–52. [172] Parenty A, Moreau X, Campagne J-M. Macrolactonizations in the total synthesis of natural products. Chemical Reviews 2006;106:911–39. [173] White CJ, Yudin AK. Contemporary strategies for peptide macrocyclization. Nature chemistry 2011;3:509–24. [174] Pauletti GM, Gangwar S, Okumu FW, Siahaan TJ, Stella VJ, Borchardt RT. Esterase-sensitive cyclic prodrugs of peptides: evaluation of an acyloxyalkoxy promoiety in a model hexapeptide. Pharmaceutical Research 1996;13:1615–23. [175] Bak A, Gudmundsson OS, Friis GJ, Siahaan TJ, Borchardt RT. Acyloxyalkoxy-based cyclic prodrugs of opioid peptides: evaluation of the chemical and enzymatic stability as well as their transport properties across Caco-2 cell monolayers. Pharmaceutical Research 1999;16:24–9. [176] Gudmundsson OS, Nimkar K, Gangwar S, Siahaan T, Borchardt RT. Phenylpropionic acidbased cyclic prodrugs of opioid peptides that exhibit metabolic stability to peptidases and excellent cellular permeation. Pharmaceutical Research 1999;16:16–23. [177] Maletinska L, Neugebauer W, Parê M, Pêrodin J, Pham D, Escher E. Lipid masking and reactivation of Angiotensin analogues. Helvetica Chimica Acta 1996;79:2023–34. [178] Kahns AH, Buur A, Bundgaard H. Prodrugs of peptides. 18. Synthesis and evaluation of various esters of desmopressin (dDAVP). Pharmaceutical Research 1993;10:68–74. [179] Lalatsa A, Lee V, Malkinson JP, Zloh M, Schätzlein AG, Uchegbu IF. A prodrug nanoparticle approach for the oral delivery of a hydrophilic peptide, leucine5-enkephalin, to the brain. Molecular Pharmaceutics 2012;9:1665–80. [180] Schroeder U, Sommerfeld P, Sabel BA. Efficacy of oral dalargin-loaded nanoparticle delivery across the blood–brain barrier. Peptides 1998;19:777–80. [181] Ogawa T, Araki M, Miyamae T, Okayama T, Hagiwara M, Sakurada S, Morikawa T. Synthesis and antinociceptive activity of orally active opioid peptides: improvement of oral bioavailability by esterification. Chemical and Pharmaceutical Bulletin 2003;51:759–71. [182] Flagg EW, Coates RJ, Eley JW, Jones DP, Gunter EW, Byers TE, Block GS et al. Dietary glutathione intake in humans and the relationship between intake and plasma total glutathione level 1994; 33-46. [183] Hoensch H, Morgenstern I, Petereit G, Siepmann M, Peters WH, Roelofs HM, Kirch W. Influence of clinical factors, diet, and drugs on the human upper gastrointestinal glutathione system. Gut 2002;50:235–40. [184] Loguercio C, Romano M, Di Sapio M, Nardi G, Taranto D, Grella A, Blanco CDV. Regional variations in total and nonprotein sulfhydryl compounds in the human gastric mucosa and effects of ethanol. Scandinavian Journal of Gastroenterology 1991;26:1042–8. [185] Zupančič O, Rohrer J, Lam HT, Bernkop-Schnürch A. Development and in vitro characterization of self emulsyfying drug delivery system (SEDDS) for oral opioid peptide delivery. Drug Development and Industrial Pharmacy, under review. [186] Trotta M, Cavalli R, Carlotti ME, Battaglia L, Debernardi F. Solid lipid micro-particles carrying insulin formed by solvent-in-water emulsion–diffusion technique. International Journal of Pharmaceutics 2005;288:281–8. [187] Zhang N, Ping Q, Huang G, Xu W, Cheng Y, Han X. Lectin-modified solid lipid nanoparticles as carriers for oral administration of insulin. International Journal of Pharmaceutics 2006;327:153– 9. [188] Sarmento B, Martins S, Ferreira D, Souto EB. Oral insulin delivery by means of solid lipid nanoparticles. International Journal of Nanomedicine 2007;2:743. [189] Garcıa-Fuentes M, Torres D, Alonso MJ. Design of lipid nanoparticles for the oral delivery of hydrophilic macromolecules. Colloids and Surfaces B: Biointerfaces 2003;27:159–68. [190] Salmaso S, Bersani S, Elvassore N, Bertucco A, Caliceti P. Biopharmaceutical characterisation of insulin and recombinant human growth hormone loaded lipid submicron
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
particles produced by supercritical gas micro-atomisation. International Journal of Pharmaceutics 2009;379:51–8. [191] Christophersen PC, Zhang L, Müllertz A, Nielsen HM, Yang M, Mu H. Solid lipid particles for oral delivery of peptide and protein drugs II–the digestion of trilaurin protects desmopressin from proteolytic degradation. Pharmaceutical Research 2014;31:2420–8. [192] Mohanraj VJ, Barnes TJ, Prestidge CA. Silica nanoparticle coated liposomes: a new type of hybrid nanocapsule for proteins. International Journal of Pharmaceutics 2010;392:285–93. [193] Katayama K, Kato Y, Onishi H, Nagai T, Machida Y. Double liposomes: hypoglycemic effects of liposomal insulin on normal rats. Drug Development and Industrial Pharmacy 2003;29:725–31. [194] Parmentier J, Hartmann FJ, Fricker G. In vitro evaluation of liposomes containing bioenhancers for the oral delivery of macromolecules. European Journal of Pharmaceutics and Biopharmaceutics 2010;76:394–403. [195] Cheng W, Lim L. Synthesis, characterization and in vivo activity of salmon calcitonin coconjugated with lipid and polyethylene glycol. Journal of Pharmaceutical Sciences 2009;98:1438–51. [196] Muranishi S, Murakami M, Hashidzume M, Yamada K, Tajima S, Kiso Y. Trials of lipid modification of peptide hormones for intestinal delivery. Journal of Controlled Release 1992;19:179–88. [197] Yodoya E, Uemura K, Tenma T, Fujita T, Murakami M, Yamamoto A, Muranishi S. Enhanced permeability of tetragastrin across the rat intestinal membrane and its reduced degradation by acylation with various fatty acids. Journal of Pharmacology and Experimental Therapeutics 1994;271:1509–13. [198] Bu P, Narayanan S, Dalrymple D, Cheng X, Serajuddin ATM. Cytotoxicity assessment of lipid-based self-emulsifying drug delivery system with Caco-2 cell model: Cremophor EL as the surfactant. European Journal of Pharmaceutical Sciences 2016;91:162–71. [199] Li P, Tan A, Prestidge CA, Nielsen HM, Müllertz A. Self-nanoemulsifying drug delivery systems for oral insulin delivery: in vitro and in vivo evaluations of enteric coating and drug loading. International Journal of Pharmaceutics 2014;477:390–8. [200] Ma H, Liu Z, Zheng C. In vitro and in vivo evaluation of a novel oral insulin formulation1. Acta Pharmacologica Sinica 2006;27:1382–8. [201] Guo L, Ma E, Zhao H, Long Y, Zheng C, Duan M. Preliminary evaluation of a novel oral delivery system for rhPTH1-34: in vitro and in vivo. International Journal of Pharmaceutics 2011;420:172–9. [202] Mahmood A, Prüfert F, Efiana NA, Ashraf MI, Hermann M, Hussain S, Bernkop-Schnürch A. Cell-penetrating self-nanoemulsifying drug delivery systems (SNEDDS) for oral gene delivery. Expert Opinion on Drug Delivery 2016;13:1503–12. [203] Fujita T, Kawahara I, Quan Y-s, Hattori K, Takenaka K, Muranishi S, Yamamoto A. Permeability characteristics of tetragastrins across intestinal membranes using the Caco-2 monolayer system: Comparison between acylation and application of protease inhibitors. Pharmaceutical Research 1998;15:1387–92. [204] Russell-Jones GJ. Oral vaccine delivery. Journal of Controlled Release 2000;65:49–54. [205] Chadwick S, Kriegel C, Amiji M. Nanotechnology solutions for mucosal immunization. Advanced Drug Delivery Reviews 2010;62:394–407. [206] Nordly P, Madsen HB, Nielsen HM, Foged C. Status and future prospects of lipid-based particulate delivery systems as vaccine adjuvants and their combination with immunostimulators. Expert Opinion on Drug Delivery 2009;6:657–72. [207] Souza Rebouças J de, Esparza I, Ferrer M, Sanz ML, Irache JM, Gamazo C. Nanoparticulate adjuvants and delivery systems for allergen immunotherapy. BioMed Research International 2012;2012. [208] Mowat AMI, Donachie AM, Jägewall S, Schön K, Löwenadler B, Dalsgaard K, Kaastrup P et al. CTA1-DD-immune stimulating complexes: A novel, rationally designed combined mucosal vaccine adjuvant effective with nanogram doses of antigen. The Journal of Immunology 2001;167:3398–405.
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
[209] Lycke N. From toxin to adjuvant: The rational design of a vaccine adjuvant vector, CTA1‐ DD/ISCOM. Cellular Microbiology 2004;6:23–32. [210] Chen W, Huang L. Induction of cytotoxic T-lymphocytes and antitumor activity by a liposomal lipopeptide vaccine. Molecular Pharmaceutics 2008;5:464.
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
Graphical abstract