Sei sulla pagina 1di 15

Progress in Lipid Research 41 (2002) 392406 www.elsevier.

com/locate/plipres

Review

A lipid based depot (DepoFoam1 technology) for sustained release drug delivery
Sankaram Mantripragada*
SkyePharma Inc., 10450 Science Center Drive, San Diego, CA 92121, USA Accepted 18 February 2002

Abstract Encapsulation of drugs into multivesicular liposomes (DepoFoam1) oers a novel approach to sustained-release drug delivery. While encapsulation of drugs into unilamellar and multilamellar liposomes, and complexation of drugs with lipids, resulted in products with better performance over a period lasting several hours to a few days after intravascular administration, DepoFoam-encapsulation has been shown to result in sustained-release lasting over several days to weeks after non-vascular administration. The routes of administration most viable for delivery of drugs via DepoFoam formulations include intrathecal, epidural, subcutaneous, intramuscular, intra-atricular, and intraocular. DepoFoam particles are distinguished structurally from unilamellar vesicles, multilamellar vesicles, and neosomes in that each particle comprises a set of closely packed non-concentric vesicles. The particles are tens of microns in diameter and have large trapped volume, thereby aording delivery of large quantities of drugs in the encapsulated form in a small volume of injection. A number of methods based on a manipulation of the lipid and aqueous composition can be used to control the rate of sustained-release from a few days to several weeks. # 2002 Elsevier Science Ltd. All rights reserved.

Contents 1. Introduction ........................................................................................................................................................... 393 1.1. Structure ........................................................................................................................................................ 394 2. Method of preparation........................................................................................................................................... 397 3. Methods for controlled release............................................................................................................................... 399

* Tel.: +1-858-625-2424; fax: +1-858-678-3982. E-mail address: sankaram@skyepharma.com


0163-7827/02/$ - see front matter # 2002 Elsevier Science Ltd. All rights reserved. PII: S0163-7827(02)00004-8

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

393

4. Mechanism of release ............................................................................................................................................. 402 5. Sustained release pharmacokinetics ....................................................................................................................... 405 References ................................................................................................................................................................... 405

1. Introduction In the area of injectable drug delivery systems, research into liposomes played a major role in the past few decades. Signicant eorts in basic and applied research institutions led to the clinical development and ultimate approval by regulatory agencies for human use of a lipid complex (Abelcet1, Amphotec1) and three liposome formulations, AmbiSome1, DaunoXome11 and a Stealth1 liposome (DOXIL1) [1,2]. These products have been developed for intravascular administration. The advantages conferred by the lipid complex, liposome and Stealth1 liposome technologies include enhancement of circulation times, and reduced toxicity by lipid encapsulation. This article reviews the scientic basis of DepoFoam1 technology, a lipid-based drug delivery system. A sustained release depot product (DepoCyt1) utilizing this technology has been clinically developed and recently approved for clinical use by regulatory agencies [3]. DepoFoam formulations of drugs are intended for non-vascular, but parenteral administration of drugs by injection. In the published scientic literature, the technology has been described as multivesicular liposomes (MVL). Both terms will be used synonymously in this article. DepoFoam or MVL are distinguished from liposomes such as unilamellar vesicles (ULV) and multilamellar vesicles (MLV) by the characteristic structure and composition of MVL (Fig. 1).

Fig. 1. Comparison of the internal structure of a unilamellar liposome, a multilamellar liposome and a DepoFoam1 particle (multivesicular liposome, MVL) [28].
1 Abelcet is the registered trademark of Elan (previously Liposome Technology Inc.). Ambisome and DaunoXome are registered trademarks of Gilead (previously NeXtar), Amphotec and DOXIL are registered trademarks of ALZA Inc. (previously Sequus Pharmaceuticals).

394

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

While ULV are liposomes with a single bilayer surrounding an aqueous compartment and MLV are liposomes with concentric lipid bilayers, DepoFoam particles (or MVL) are composed of non-concentric multiple lipid layers. It has been suggested that the non-concentric nature of the arrangement of lipid layers confers an increased level of stability and longer duration of drug release [4]. This is because only breaches in the outermost membranes of an MVL result in release of encapsulated drug to the external medium, and release of drug from the internal vesicles results in a redistribution of drug inside the particle without drug release from the particle. The interconnected network of the multivesiculated structure also ensures that the vesicles can rearrange themselves internally without release of drug by internal fusion and division. These events are characteristic of and are possible with MVL system due to the non-concentric, close-packed nature of the particle, but not with the concentric multivesicular liposomes, the single-vesicle based liposomes, or with the non-concentric but not closely packed plurilamellar liposomes. A detailed structural characterization of MVL is discussed later. 1.1. Structure One of the earlier published reports on the internal structure of MVL was based on light microscopy and thin-section transmission electron microscopy [5]. Morphologically, MVL were described as spheroids with granular internal structures under the light microscope. The details of the membrane were seen best with transmission electron microscopy, which revealed that a bilayer formed the outermost membrane and the internal space was divided into numerous compartments by bilayer septa. It was hypothesized that the essential component, neutral oil, became part of corners or edges where membranes meet each other, thus stabilizing membrane boundaries analogous to planar black lipid membranes that also require a neutral lipid. Some of the putative corners were seen in the electron micrographs as osmiophilic buttons which were considered to be collections of triglycerides. In this respect the MVL dier from all other types of liposomes. A detailed quantitative study using freeze-fracture electron microscopy showed that the structure of MVL is governed by general topological constraints in analogy to gas-liquid foams, simple liquids, and metallic glasses [6]. Freeze-fracture images of MVL provided direct visual conrmation that the compartment contacts are tetrahedrally coordinated, as dictated by rules set down by Plateau in the last century (Fig. 2). From the experimentally measured distribution of polyhedral facets of the MVL, it has been shown that the distribution of compartment shapes is in near perfect agreement with theoretical predictions and computer simulations of random close packing of polytetrahedra. This polyhedral random close packing of the internal vesicles is a direct analog of the way bubbles are packed in a gasliquid foam. The close packing of vesicles formed by lipids and the encapsulated aqueous phase are also apparent in uorescent micrographs [7]. In this study, multivesicular liposomes were labeled with a lipid probe, rhodamine-DHPE, and an aqueous probe, Bodipy disulfonate. As seen in Fig. 3, while rhodamine-DHPE revealed the lipid matrix surrounding aqueous chambers, the Bodipy disulfonate showed discrete non-concentric aqueous compartments inside the multivesicular liposome. At the molecular level, the distribution of phospholipids and triglycerides in MVL was determined using laser scanning confocal microscopy and 13C NMR [8]. Confocal microscopy with

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

395

two phospholipid uorescent probes, Rhodamine-DHPE and NBD-PG, and a triglyceride uorescent probe, Bodipy-triglyceride, was used to visualize the distribution of the lipid components in MVL. The micrographs revealed a uniform distribution of the two phospholipid probes in the plane of the membrane, whereas the triglyceride probe accumulated at discrete locations. In the NMR spectra of MVL, components attributable to triolein in a liquid-like phase were observed,

Fig. 2. Freeze fracture electron micrograph of a multivesicular liposome showing random close packing of the internal chambers. Examples of faceted structures surrounding the internal chambers that are closely packed and tetrahedrally coordinated are outlined and identied [6].

Fig. 3. Confocal micrographs of DepoFoam particles recorded with a red uorescent dye (Rhodamine DHPE) labeling the lipids, a green uorescent dye (Bodipy disulfonate) labeling the aqueous phase. In the merged image, the distribution of both the lipids and the encapsulated aqueous phases is seen [7]. The bar indicates 10 mm.

396

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

and the intensity of these components accounted for the triolein present in the sample. The NMR and microscopy results are consistent with a structural model for MVL in which triolein acts as a hydrophobic space ller at bilayer intersection points and stabilizes these junctions, and is also present as oil droplets dispersed in the encapsulated aqueous compartments (Fig. 4). In a 31P NMR study, temperature dependence of the spectra was monitored [9]. At 15  C, The 31 P NMR spectrum was a partially averaged anisotropic powder pattern typically seen for bilayers with a low degree of curvature. When the temperature rose to 25  C, an isotropic spectral component appeared. The intensity of the isotropic component relative to that of the anisotropic powder pattern increased as the temperature was raised to 85  C. The temperature dependence was reversible in that upon cooling to 15  C, only the powder pattern was observed. Interestingly, light scattering and microscopy experiments ruled out the possibility that the isotropic component was due to formation of small vesicles. A cubic phase is probably not involved since the

Fig. 4. Schematic illustrating locations of the triglycerides in MVL based on NMR and confocal microscopy [8].

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

397

viscosity of the sample does not increase with temperature. Further, there is no evidence for a thermotropic phase transition from a lamellar phase to an inverted hexagonal phase for MVL. It is likely that the isotropic component does not represent a separate population of small structures; rather it is due to the polyhedral symmetry of the internal vesicles seen in freeze-fracture electron microscopy. There is at least a theoretical precedence for the appearance of isotropic spectral components arising from the symmetry of internal structures through which lipid molecules laterally diuse, without the presence of small structures with macroscopic isotropic motion.

2. Method of preparation MVL are produced by the use of a double emulsication process as shown in Fig. 5 [5,6]. The rst step is making a water-in-oil emulsion by dissolving amphipathic lipids containing at least one neutral lipid in one or more volatile organic solvents for the lipid component, adding to the lipid component an immiscible rst aqueous component and a biologically active substance to be encapsulated. The mixture is then emulsied, and then mixed with a second immiscible aqueous component followed by mechanical mixing to form solvent spherules suspended in the second

Fig. 5. Process ow diagram for the preparation of multivesicular liposomes.

398

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

aqueous component. The solvent spherules contain multiple aqueous droplets with the substance to be encapsulated dissolved in them. The organic solvent is removed from the spherules, generally by evaporation, by reduced pressure or by passing a stream of gas over or through the suspension. When the solvent is completely removed, the spherules become MVL. Various types of lipids can be used to make MVL, and the only requirements regarding lipids are that at least one amphipathic lipid (e.g. phospholipids) and one neutral lipid (e.g. triglyceride) be included in the lipid component. When the neutral lipid is omitted, conventional multilamellar vesicles or unilamellar vesicles are formed instead of multivesicular liposomes [10]. It is also critical that the double emulsication process be employed in order to form the multivesiculated structure. When other well-known methods for the preparation of liposomes are employed, even when the required mixture of phospholipids and triglycerides is used, MLV and ULV are formed instead of multivesicular liposomes. Not only is a triglyceride required for the formation of MVL, but the concentration of triglyceride also has a profound eect on capture volume and encapsulation eciency (Fig. 6). In a mixture with phosphatidylcholine, cardiolipin and cholesterol (4:5:1:4.5 mole ratio) maximal capture volume and encapsulation eciency are obtained in the triolein mole fraction range of 0.010.08 [5]. At lower triolein mole fractions the capture eciency is reduced markedly.

Fig. 6. Eect of mole fraction of triolein with respect to other lipids in MVL on capture volume (ml/mg) and encapsulation (%). In addition to triolein, the lipid combination consisted of phosphatidylcholine, cardiolipin and cholesterol in the relative proportions of 4.5:1: 4.5 mmol, respectively. Data are from [5].

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406 Table 1 Individual and sequential unit operations during manufacturing scale-up Unit operation First emulsion Independent variables Mixing speed, mixing time, energy input per unit volume, temperature Mixing speed, mixing time, impeller diameter, impeller design, temperature, volume of second aqueous phase Gas ow rate prole, degree of foaming, temperature Process ow rates, recirculation mass ow, volume of buer exchange Dependent variables Viscosity, conductivity, particle size

399

Second emulsion

Particle size, particle size distribution

Solvent extraction

Rate of solvent removal, extent of solvent removal, step yield Processing time, buer ow rate, step yield

Microltration

Overall process

Comparison to pilot scaleParticle size, loading, overall yield, product stability, drug release (in vitro and in vivo) prole

Independent variables optimized for each unit operation and the dependent variables used to determine completion of optimization are also listed [11].

In order to produce sterile commercial scale batches of a DepoFoam product conforming to regulatory requirements, aseptic processing techniques are employed [1115]. The production method is treated as a set of four sequential unit operations namely, rst emulsion, second emulsion, solvent extraction, and microltration. Table 1 lists the independent variables to be optimized for each unit operation, along with a dependent variable utilized to judge the quality of material produced at the end of the unit operation.

3. Methods for controlled release Both the lipid components and the composition of the encapsulated aqueous phase can be used to modulate the rate of sustained release from MVL. The release rate of the biologically active compound can be modied by utilizing in the preparation of MVL, a neutral lipid component (typically a triglycerides) that provides the desired rate of release in the type of uid in which the MVL are to be used (Fig. 7). It has been found that long chain triglycerides result in slower release rates from the MVL formulations than short chain triglycerides do [16], presumably due to a stabilizing eect on the structure of the particle by increased chainlength. In mixtures, the rate of release of the biologically active compound decreases in proportion with the increase in the ratio of the slow release neutral lipid to the fast release neutral lipid [16]. The slow release neutral lipid is selected from triglycerides having mono-unsaturated fatty acid ester moieties containing about 1418 carbons in the acyl chain and generally having a molecular

400

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

Fig. 7. Eect of the composition and chainlength of triglycerides on in vivo pharmacokinetics [16]. Pharmacokinetics of morphine MVL formulations manufactured with dierent mole ratios of triolein to tricaprylin and injected into the epidural space of Beagle dogs. Morphine released from the MVL was determined by measuring the level of morphine in the adjacent, dura membrane-separated, cerebrospinal uid. Results from an injection of 5 mg unencapsulated morphine (sulfate) in saline (^); 40 mg of morphine in MVL with a triolein:tricaprylin mole ratio of 10:0 (!), 20 mg of morphine in MVL with a triolein:tricaprylin mole ratio of 1:4 (*), 1:9 (&), and 0:10 ().

weight about 725885, and those with saturated fatty acid ester moieties containing about 10 12 carbons in the acyl chain and generally having a molecular weight about 554639. Cholesterol esters and esters of propylene glycol can also be used. While triolein, tripalmitolein, trimyristolein, trilaurin and tricaprin fall into this category, triolein or tripalmitolein are most preferred. The neutral lipid inducing fast release is selected from triglycerides having saturated fatty acid ester moieties containing from about 6 to 8 carbons in the acyl chain and having a molecular weight from about 387 to 471. Tricaprylin, and mixtures of tricaprylin and tricaproin, or mixed chain C6 to C8 triglycerides are most preferred. However, the use of neutral lipids with less than six carbons (e.g. tricaproin) in the acyl chain results in a rapid release of the encapsulated compound. By adjusting the slow release to fast release lipid ratio, the rate of release could be adjusted for a number of molecules such as G-CSF, GM-CSF, recombinant human insulinlike growth factor-1 (IGF-1), recombinant human insulin, morphine, cytosine arabinoside, a 15mer interleukin-6 antisense oligonucleotide, Escherichia coli plasmid PBR 322, tetracaine, and sucrose. The osmolarity of the aqueous phase containing the drug that is mixed with the immiscible solvent phase containing lipids is an important determinant of in vivo rates of release [17]. The rate of release of the active substance into the surrounding in vivo environment can be decreased by increasing osmolarity of this solution (Table 2). The decay half-life of the amount of cytarabine in the intraperitoneal cavity of mice was 0.64 h for an MVL formulation prepared with an aqueous solution containing 82 mM cytarabine. The half-life was 13.6 h for an MVL preparation made with 246 mM cytarabine solution. If the mechanism of release in vivo is exclusively classical diusion, one would expect that the rate of release is faster for the formulation with the greater

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

401

Table 2 Concentration of cytarabine in the pellet fractions of the peritoneal uid, and the total amount of cytarabine in the peritoneal cavity, at dierent times subsequent to intraperitoneal injection to mice of MVL formulations prepared using a solution containing either 82 mM cytarabine or 246 mM cytarabinea Hours after intraperitoneal injection Concentration in pellet fraction, mg/ml 82 mM formulation 0 0.5 4 15 64 108 t1/2 (r2) 9380 9925371 11151,108 1015922 ND ND 1.7 (0.974) 246 mM formulation 9685 6896 1,083 13,360 761 11,493 1493 132 634518 29.6 (0.904) Total amount (mg) 82 mM formulation 2372 110867 150 14 76 58 ND ND 0.64 (0.989) 246 mM formulation 2791 1202 48 1493 132 790 419 388 191 58 34 13.6 (0.892)

ND, not determined; t1/2, decay half-life in hours. a A group size of three was used [17]. The pellet fraction contains closely packed MVL particles, and analysis of pellet allows one to determine the concentration of drug inside the particles. Table 3 Release in vitro of MVL-encapsulated drug (cytosine arabinoside) into human plasma at 37  C from various DepoFoam formulationa Acid Perchloric Nitric Phosphoric Formic Trichloroacetic Acetic Triuoroacetic Sulfuric Half life in days 37.2 8.0 54.5 5.7 6.5 0.2 5.6 0.2 5.5 0.6 4.8 0.5 3.4 0.4 1.6 0.5

a Each formulation was prepared with a constant concentration of cytarabine (20 mg/mL), and a constant concentration (136 mM) of one of the acids listed in the table [18].

concentration gradient of drug (246 mM formulation) between the inside of the particle and the peritoneal uid than for the 82 mM formulation with the a lower gradient. The fact that the results are in the opposite direction indicates that the in vivo release is dominated by mechanisms other than simple diusion. Use of dierent acids in the rst aqueous solution has a profound eect on release rates [18]. The half-life for in vitro release into human plasma from MVL formulations ranged from 1.6 to 37.2 h depending on the acid added to a solution of cytarabine at a constant concentration (Table 3). In another study, it was found that the addition of a polyhydroxy organic acid (eg., glucuronic acid), or triprotic mineral acids (e.g. phosphoric acid) provides a synergistic eect with respect to loading of bupivacaine [19]. With an MVL preparation of bupivacaine prepared using this method, a half-life of 12 h was reported for bupivacaine at the intracutaneous injection site in

402

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

Table 4 Pharmacokinetic (PK) parameters for a subcutaneous injection of either unencapsulated methotrexate (MTX) or a DepoFoam (MVL)-encapsulated cyclodextrin (CD)-complex of methotrexate (MVLCDMTX)a MTX Injection site Amount t1/2 (h) Plasma Cmax S.D. (mM) Concentration t1/2 (h) AUC (mM h)
a

MVLCDMTX 50.4

0.16

17.4 5.2 0.53 17.3

0.138 0.061 109 24.5

PK parameters obtained by measurements both at the subcutaneous injection site and in plasma are given in the table [20].

guinea pigs. In comparison, an unencapsulated free bupivacaine hydrochloride solution gave a half-life of 1.3 h. The half-life values decrease roughly as both the charge and the size of the counterion increase, suggesting that permeation of the salt complex is an important determinant of drug release. Water-soluble compounds such as methotrexate can interact with cyclodextrins to form an inclusion complex. Encapsulation of the inclusion complex into MVL results in a release rate for the drug that is slower than that from an MVL with just the drug encapsulated without formation of the cyclodextrin inclusion complex [20]. The apolar cavity of the cyclodextrin sequesters the compound suciently to slow the rate of release from MVL. The periphery of the inclusion complex is hydrophilic with the result that the inclusion complex forms a solution in aqueous media. Upon intrathecal injection in rats of 100 mg unencapsulated methotrexate, the amount of drug in the cerebrospinal uid decayed with a half-life of 0.03 days. For an MVL preparation with a cyclodextrin inclusion complex of methotrexate encapsulated, the corresponding half-life increased to 9 days (Table 4). The areas under the curve (AUC) are comparable for the MVL formulation and the unencapsulated drug suggesting that the relative bioavailability of the drug is not altered by encapsulation.

4. Mechanism of release The evolution of the structure of MVL with time as determined by freeze-fracture electron microscopy and in vitro release of encapsulated drug was followed in both human serum and articial cerebrospinal uid [21]. Over a period of 9 days, the internal compartments in the MVL grew in size and the average number of compartments per MVL decreased as cytarabine was released, suggesting that internal coalescence plays a role in drug release (Fig. 8). The overall MVL size decreased during aging, and some particles adopted irregular shapes. However, a signicant fraction of MVL retained their original multivesicular structure even after near-complete release of cytarabine. These results suggested that permeation of drug through the vesicle membranes is an important determinant of in vitro drug release. While cytarabine and most other molecules studied with MVL are water-soluble compounds with low octanolwater partition

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

403

Fig. 8. Coalescence of internal chambers within a multivesicular liposome during incubation in human serum at 37  C (J.A. Zasadzinski, personal communication). Multiple freeze fracture images were recorded on each day on samples withdrawn from incubation in human plasma at 37  C. The number of internal chambers in a given size range were counted on for a total count of about a thousand chambers per sample. The normalized size distributions are plotted against the diameter of the internal chamber.

coecients, it is likely that membrane partitioning will play a critical role for molecules with high partition coecients. Both during in vitro incubation in human plasma and in vivo in a subcutaneous hollow ber implant study in rats, lipid reorganization leading to coalescence of internal chambers followed by formation of blebs at the surface of excess lipid were observed [22]. The internal chamber size was determined by uorescence confocal microscopy. This study also suggested that the hydrocarbon chain length of the triglyceride and the concentration of the shorter versus longer chain triglycerides determine the rate at which such internal membrane reorganization and coalescence take place. Cytarabine release over a 2-week period at 37  C was found to be dependent on the medium of incubation [23,24]. The media investigated in this study included human plasma, articial cerebrospinal uid, simulated gastric uid, phosphate buers at pH 6 and 7.4, phthalate buers at pH 3, 4 and 5, and hydrochloric acid at pH 2. The release rate in human plasma was much faster than in the other low-protein media or buers of similar pH. In low pH media (simulated gastric uid and pH 2 solution), large aggregates were formed as determined by particle size measurements. Incubation at pH 3 had an intermediate eect on aggregation. Over the period of 2 weeks, particle size remained fairly constant for pH 47.4 buers and for ACSF, while in plasma particle size increased slightly. Particle concentration showed time-dependent behavior similar to that of drug release for all media of pH 6 and above. For the low pH hypotonic media, drug release did not correlate with particle concentration.

404

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

Fig. 9. Biodistribution of tritiated cholesterol in MVL from the subcutaneous injection site into surrounding tissues [25]. Remaining radioactivity at the injection site (^) and the carcass (&) are shown.

The in vitro drug release, particle size and particle concentration data are consistent with the overall mechanism of release composed of at least three components namely, diusion, erosion and attrition [24]. For the diusion component, particle number (measured by hemocytometry) remains constant but the amount of drug retained within the MVL particles decreases over time. Particle size remains essentially constant, except when the particles act as osmometers sensing the loss of solute. For the erosion component, both particle concentration and internal drug concentration remain constant while particle size decreases. When the attrition part of the mechanism is operative, particle concentration decreases while internal drug concentration and size remain constant. In an in vivo study, male mice were injected subcutaneously with amikacin encapsulated in DepoFoam particles prepared with tritium labeled cholesterol [25]. At time points over 21 days, the level of tritium was determined for the site of injection, plasma, several organs involved with cholesterol processing or storage, and the residual carcass. At day 7, a decrease in radioactivity levels was observed at the site of injection (Fig. 9). Concomitantly, an increase in tritium levels was detected at day 7 in the plasma, organs, and residual carcass. After 21 days, 20% of the initial dose remained at the site of injection, 20% of the initial dose had accumulated in the residual carcass, and detectable levels of radioactivity were present in the plasma and all organs. Cholesterol is known to move spontaneously between membranes faster than phospholipids do [26]. As a result, the amount of cholesterol remaining at the injection site may not be an indictor of the number of DepoFoam particles remaining at the injection site. However, the fact that signicant residual cholesterol is detected at the injection site after 21 days suggests that the DepoFoam particles do remain at the site at least for 21 days.

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

405

Table 5 Comparison of elimination half-life values obtained for a single bolus injection of DepoFoam formulations with those obtained for a single bolus injection of unencapsulated drugsa
Drug Species Elimination half-life, t1/2 (h) DepoFoam Cytarabine Cytarabine Cytarabine Cytarabine Cytarabine Methotrexate Methotrexate Methotrexate Morphine IGF-1 Interferon a-2b Bleomycin 20 30 Deoxycytidine 5-FUMP
a

Route of administration

Possible clinical use

Unencapsulated 0.74 0.26 2.7 0.16 0.2 7.2 0.16 0.54 2.6 4 1.5 0.13 1.1 4.5 Intrathecal Intraperitoneal Intrathecal Subcutaneous Subconjuctival Intrathecal Subcutaneous Intraperitoneal Epidural Subcutaneous Intraperitoneal Subcutaneous Intraventricular Intravitreal Cancer Cancer Cancer Cancer Cancer Cancer Cancer Cancer Pain Management Growth Factor Viral Infections, Cancer Antibiotic Viral Infections Viral Infections

Monkey Mouse Rat Mouse Rabbit Rat Mouse Mouse Rat Rat Mouse Mouse Rat Rabbit

156 21 148 96 52.5 96 50.4 45.6 82 26 20 31.8 23 124

For intracavitary delivery (intrathecal, intraventricular, intravitreal and epidural), the pharmacokinetic prole shows an initial distribution and a later elimination phase. Only elimination half-life values are reported in the table. t1/2 Values were calculated using the concentration of drug versus time prole [7]. All formulations contained the phospholipids dioleoyl phosphatidylcholine, dipalmitoyl phosphatidylglycerol and cholesterol. They also contained triloein, except the IGF-1 formulation which was prepared with tripalmitolein. The typical amounts of the lipids are given in [5,6].

5. Sustained release pharmacokinetics Pharmacokinetic data from DepoFoam formulations of a number of dierent drugs have been reviewed extensively elsewhere [7,27]. These studies cover a wide variety of routes of injection such as intrathecal, intraperitoneal, subcutaneous, intramuscular, epidural, subconjuctival and intravitreal routes. The classes of molecules for which sustained release pharmacokinetics with DepoFoam formulations have been demonstrated include antineoplastic agents (cytarabine, methotrexate, leuprolide), antibiotics (amikacin, gentamicin), analgesics and anesthetics (bupivacaine, morphine, enkephalin), cytokines (interferon, IGF-1, IL-2, G-CSF, GM-CSF), DNA and oligonucleotides. In general, single bolus injections of unencapsulated drugs result in plasma or serum elimination half-lives ranging from 0.13 h to 11 h (Table 5). Depending on the drug, dose administered and the route of injection, single bolus injections of the corresponding DepoFoam formulations increase the elimination half-lives by a factor of 6600. The longest reported half-life is 156 h for DepoFoam-encapsulated cytarabine injected intrathecally in a monkey model. References
[1] Gabizon A, Barenholz Y. Liposomal anthracyclinesfrom basics to clinical approval of PEGylated liposomal doxorubicin. In: Jano AS, editor. Liposomes: rational design. New York: Marcel Dekker; 1999. pp. 34362. [2] Sankaram MB. Commercial sustained-release injectable formulations by encapsulation. In: Senior J and Radomsky M, editors. Sustained-release injectable products. Denver, CO: Interpharm Press; 2000. pp. 2539.

406

S. Mantripragada / Progress in Lipid Research 41 (2002) 392406

[3] Chiron Corporation. Physicians Desk Reference 2000;54:9358. [4] New RRC. Inuence of liposome characteristics on their properties and fate. In: Philippot JR and Schuber F, editors. Liposomes as tools in basic research and industry. Boca Raton: CRC Press; 1995. pp. 320. [5] Kim S, Turker MS, Chi EY, Sela S, Martin GM. Biochim Biophys Acta 1983;728:33948. [6] Spector MS, Zasadzinski JA, Sankaram MB. Langmuir 1996;12:47048. [7] Mantripragada S. Drug Delivery Systems and Sciences 2001;1:1316. [8] Ellena JF, Le M, Caso D, Solis RM, Langston M, Sankaram MB. Drug Delivery 1999;6:97106. [9] Ellena JF, Solis RM, Caso D, Sankaram MB. Biophysical Journal 1998;74:A372. [10] Schneider M. Process for the preparation of liposomes in aqueous solution. US Patent No. 4,224,179; 1980. [11] Pepper C, Patel M, Hartounian H. Pharmaceutical Engineering 1999;19:818. [12] Pepper C, Patel M, Hartounian H. BioPharm 1999;12:2634. [13] Pepper C, Patel M, Hartounian H. BioPharm 1999;12:3655. [14] Pepper C, Patel M, Hartounian H. BioPharm 1999;12:524. [15] Pepper C, Patel M, Hartounian H. BioPharm 2000;13:2834. [16] Willis RC. Method for utilizing neutral lipids to modify in vivo release from multivesicular liposomes. US Patent No. 5,891,467; 1999. [17] Sankaram MB, Kim S. Preparation of multivesicular liposomes for controlled release of encapsulated biologically active substances. US Patent No. 5,993,850; 1999. [18] Sankaram MB, Kim S. Multivesicular liposomes with controlled release of encapsulated biologically active substances. US Patent No. 5,766,627; 1998. [19] Kim S, Kim T, Murdandi S. Sustained-release liposomal anesthetic compositions. US Patent No. 6,045,824; 2000. [20] Kim S. Cyclodextrin liposomes encapsulating pharmacologic compounds and methods for their use. US Patent No. 5,759,573; 1998. [21] Keller S, Strahler D, Spector M, Walker S, Kennedy M, Zasadzinski JA. Biophysical Journal 1997;72:A400. [22] Willis RC, Gai W, McAllister DL, Samaniego AC. Proceed Intl Symp Control Rel Bioact Mater 1998;25:3901. [23] Longenecker JP, Willis RC, Thrift R, Sankaram MB. Proceed Intl Symp Control Rel Bioact Mater 1997;24:2067. [24] Thrift R, Solis RM, Lewcock K, Sankaram MB. Proceed Intl Symp Control Rel Bioact Mater 1998;25:42930. [25] Brownson EA, Langston M, Tsai AG, Gillespie T, Davis TP, Intaglietta M. Proceed Intl Symp Control Rel Bioact Mater 1998;25:423. [26] Bar LK, Barenholz Y, Thompson TE. Biochemistry 1986;25:67015. [27] Mantripragada S, Howell SB. Sustained release drug delivery with DepoFoam. In: Brown D, editor. Drug delivery systems for cancer treatment. New York: Humana Press; 2001 (in press). [28] Sankaram MB, Kim S. Multivesicular liposomes with controlled release of encapsulated biologically active substances. US Patent No. 5,766,627; 1998.

Potrebbero piacerti anche