Antibody-directed liposomes as drug-delivery vehicles

47
Advanced Drug Delivery Reviews, 3 (1989) 343-389 343 Elsevier ADR 00026 Antibody-directed liposomes as drug-delivery vehicles Stephen Wright and Leaf Huang Department of Biochemistry, University of Tennessee, Knoxville, TN, U.S.A. (Received February 5, 1988) (Accepted February 15, 1988) Key words: Liposome; Drug targeting; Antibody-liposome conjugation; Liposome-cell interaction; Li- posome targeting Contents Summary ................................................................................................................. 344 I. Introduction ................................................................................................... 345 II. Preparation of antibody-targeted liposomes .......................................................... 347 1. Covalent conjugation to pre-formed liposomes ................................................ 349 2. Incorporation of lipophilic antibodies ............................................................ 352 III. Immunoliposome binding and drug delivery in vitro ............................................... 353 1. Binding to immobilized antigens ................................................................... 353 Abbreviations: RES, reticuloendothelial system; SUV, small unilamellar vesicle; PE, phosphatidyl- ethanolamine; EDCI, 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide; SPDP, N-hydroxysuccinimidyl- 3-(2-pyridyldithio)propionate; DTP, dithiopyridine; MPB, N-[4-(p-maleimidophenyl)butyryl]; LUV, large unilamellar vesicle; REV, reverse-phase evaporation vesicle; SATA, succinimidyl-S-acetylthioac- etate; SAMSA, S-acetylmercaptosuccinic anhydride; AETA, aminoethylthioacetyl; NHSP, N-hydrox- ysuccinimide ester of palmitic acid; FITC, fluorescein isothiocyanate; MLV, multilamellar vesicle; MTX, methotrexate; DNP-cap, N-dinitrophenylamino caproyl; CF, carboxyfluorescein; DOPC, dioleoyl- phosphatidylcholine; TNP, trinitrophenyl moiety; PHC, palmitoyl homocysteine; CHEMS, cholesteryl hemisuccinate; DOPE, dioleoylphosphatidylethanolamine; RET, resonance energy transfer; OA, oleic acid; ANTS, 1-aminonaphthalane-3,6,8-trisulphonic acid; DPX, N,N'-p-xylylenebis (pyridinium bro- mide); NBD, 7-nitro-2,1,3-benzoxadiazol-4-yl;ADP, adenosine diphosphate; HSV, herpes simplex vi- rus; TK, thymidine kinase; PEP, phosphoenolpyruvate; CK, carboxykinase; CAT, chloramphenicol acetyltransferase; AFP, anti-human c~-fetoprotein; MHC, major histocompatibility complex; DPPC, dipalmitoylphosphatidylcholine; DSPC, distearoylphosphatidylcholine; TPE, phosphatidylethanolam- ine transphosphatidylated from egg phosphatidylcholine Correspondence: L. Huang, Department of Biochemistry, Biology Business Office, University of Ten- nessee, M407 Waiters Life Sciences Building, 125 Austin Peay Boulevard, Knoxville, TN 37996-0840, U.S.A. 0169-409X/89/$03.50 © 1989 Elsevier Science Publishers B.V. (Biomedical Division)

Transcript of Antibody-directed liposomes as drug-delivery vehicles

Page 1: Antibody-directed liposomes as drug-delivery vehicles

Advanced Drug Delivery Reviews, 3 (1989) 343-389 343 Elsevier

ADR 00026

Antibody-directed liposomes as drug-delivery vehicles

Stephen Wright and Leaf Huang Department of Biochemistry, University of Tennessee, Knoxville, TN, U.S.A.

(Received February 5, 1988) (Accepted February 15, 1988)

Key words: Liposome; Drug targeting; Antibody-liposome conjugation; Liposome-cell interaction; Li- posome targeting

Contents

Summary ................................................................................................................. 344

I. Introduction ................................................................................................... 345

II. Preparation of antibody-targeted liposomes .......................................................... 347 1. Covalent conjugation to pre-formed liposomes ................................................ 349 2. Incorporation of lipophilic antibodies ............................................................ 352

III. Immunoliposome binding and drug delivery in vitro ............................................... 353 1. Binding to immobilized antigens ................................................................... 353

Abbreviations: RES, reticuloendothelial system; SUV, small unilamellar vesicle; PE, phosphatidyl- ethanolamine; EDCI, 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide; SPDP, N-hydroxysuccinimidyl- 3-(2-pyridyldithio)propionate; DTP, dithiopyridine; MPB, N-[4-(p-maleimidophenyl)butyryl]; LUV, large unilamellar vesicle; REV, reverse-phase evaporation vesicle; SATA, succinimidyl-S-acetylthioac- etate; SAMSA, S-acetylmercaptosuccinic anhydride; AETA, aminoethylthioacetyl; NHSP, N-hydrox- ysuccinimide ester of palmitic acid; FITC, fluorescein isothiocyanate; MLV, multilamellar vesicle; MTX, methotrexate; DNP-cap, N-dinitrophenylamino caproyl; CF, carboxyfluorescein; DOPC, dioleoyl- phosphatidylcholine; TNP, trinitrophenyl moiety; PHC, palmitoyl homocysteine; CHEMS, cholesteryl hemisuccinate; DOPE, dioleoylphosphatidylethanolamine; RET, resonance energy transfer; OA, oleic acid; ANTS, 1-aminonaphthalane-3,6,8-trisulphonic acid; DPX, N,N'-p-xylylenebis (pyridinium bro- mide); NBD, 7-nitro-2,1,3-benzoxadiazol-4-yl; ADP, adenosine diphosphate; HSV, herpes simplex vi- rus; TK, thymidine kinase; PEP, phosphoenolpyruvate; CK, carboxykinase; CAT, chloramphenicol acetyltransferase; AFP, anti-human c~-fetoprotein; MHC, major histocompatibility complex; DPPC, dipalmitoylphosphatidylcholine; DSPC, distearoylphosphatidylcholine; TPE, phosphatidylethanolam- ine transphosphatidylated from egg phosphatidylcholine

Correspondence: L. Huang, Department of Biochemistry, Biology Business Office, University of Ten- nessee, M407 Waiters Life Sciences Building, 125 Austin Peay Boulevard, Knoxville, TN 37996-0840, U.S.A.

0169-409X/89/$03.50 © 1989 Elsevier Science Publishers B.V. (Biomedical Division)

Page 2: Antibody-directed liposomes as drug-delivery vehicles

344 S. WRIGHT AND L. HUANG

2. Drug delivery to cells in culture ................................................................... 358

IV. Controlled release immunoliposomes .................................................................. 363 1. Acid-sensitive liposomes ............................................................................. 363 2. Temperature-sensitive liposomes .................................................................. 369 3. Target-sensitive liposomes ........................................................................... 370

V. Animal studies with immunoliposomes ................................................................ 372

VI. Conclusions .................................................................................................... 377

VII. Future perspectives .......................................................................................... 378

Acknowledgements .................................................................................................... 379

References ............................................................................................................... 379

Summary

The field of l iposome targeting has developed rapidly in the past 10 years as a multidisciplinary approach to the controlled delivery of bioactive molecules. Li- posomes as delivery vehicles offer several benefits over the administration of free compounds as therapeutic devices. Advantages such as the ability to encapsulate a wide variety of polar, nonpolar, and amphipathic agents make the l iposome an attractive delivery device for many applications. Encapsulation protects the drug from metabolic degradation and protects host tissue from non-specific effects until their arrival at the site of consumption. The large relative carrying capacity of the l iposomes permits the transfer of thousands of drug molecules to a given cell. Re- gardless of the type of l iposome or its route of administration, the pr imary con- sumer of either empty or drug-laden liposomes are the highly phagocytic members of the reticuloendothelial system. Thus, except for these 'natural ' target cells, li- posomes remain relatively non-specific transporters. Conjugation of targeting li- gands such as antibodies to the drug or drug carrier confers specificity of that com- plex for a certain cell or organ expressing the targeted antigenic determinant. However , direct conjugation of drug with antibody has met with limited success, due in part to the required chemical coupling between the drug and antibody. Al- ternatively, by first entrapping a native drug into a liposome by a non-perturbing method, followed by conjugation of antibodies to the surface of the liposome, some of the advantages of either independent approach may be realized without many of the disadvantages. The pros and cons of this general approach have been de- bated during the past decade in many comprehensive reviews and monographs that have been published on the subject of l iposome targeting. The pr imary emphasis of the current effort is to focus on one particular strategy for liposome targeting, i.e., the use of antibodies as vector molecules for targeted liposome drug delivery systems. In the sections that follow we try to describe the progress realized and the pitfalls and the limitations encountered with this approach to drug targeting.

Page 3: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 345

I. Introduct ion

Perhaps the foremost problem of contemporary chemotherapy is the lack of specificity of a drug for its target organ and the profound side effects resulting from administration of the large dosages needed to reach the target site at acceptable levels. The large variety of potent chemotherapeutic agents that have been devel- oped in the last two decades have shown great potential but are too frequently just as toxic to the host cells as they are to the target cells. The desirability of a system which can mediate the site-specific delivery of drugs is not new. In fact, it was the recognized father of chemotherapy, Paul Ehrlich, who over 80 years ago envi- sioned the ability of drugs to be selectively taken up at infected sites. He later foresaw the use of "a carrier by which to bring therapeutically active groups to the organ in question" [1]. The number and diversity of 'magic bullets' and drug car- riers that have since been devised are too numerous to catalog here and several reviews and monographs have appeared on the topic [2-5]. All of the rapidly emerging technologies in the field of drug targeting are primarily designed for one purpose, to concentrate the drug or other bioactive molecule at the target site with minimal effects on surrounding normal tissue. Optimally, the role of the targeting vehicle is to act as a guidance system which allows the drug or other cytodestruc- tive element to home in on its target from an otherwise random distribution. Since at this time we have no intelligent homing devices with an a priori knowledge of which direction the target lies, we must settle on a system of 'close encounters'. Under this scenario, the targeted drug or drug carrier wanders about the body with its 'key' projecting outward until it encounters the 'lock' for which it was designed. Once this specific interaction has occurred, presumably at the cell surface, then the drug may elicit its response either topically or after internal processing by the target cell. Presumably, no response will commence upon bumping into a non-tar- geted cell, which, in our analogy has either no 'lock' or at least the wrong 'lock' for our roaming 'key'.

Among the many schemes devised for controlled drug delivery, two quite dif- ferent approaches have proved particularly promising. One approach involves di- rect targeting of the drug by conjugation with a ligand having a distinct affinity for a particular cell or tissue. Such ligands might include hormones, carbohydrates, lectins, peptides, and receptor agonists or antagonists and immunoglobulins. An- tibody targeting is a type of ligand-mediated targeting which is dependent on a li- gand-receptor interaction to mediate the binding of drug to target cells. Immu- noglobulins are ideal targeting molecules because of their specificity and high affinity for distinct targets and the relative ease with which antibodies can be pre- pared against a variety of different antigen molecules. Further, with the advent of the technology to produce monoclonal antibodies, clonal populations of antibody can now be generated against very specific epitopes of the target antigen. This technology now also includes the ability to produce human monoclonal antibodies [6]. Conjugation of drugs to antibodies for therapeutic applications has been only partially successful (reviewed in Ref. 7). Many times the drug's activity is altered as the result of the new bonding and there may not be mechanisms operable for

Page 4: Antibody-directed liposomes as drug-delivery vehicles

346 S. WRIGHT AND L. HUANG

cleavage of this bond which is often required for drug activity. Alternatively, con- centration of a drug within microreservoirs can be considered another approach to targeting in that the cells which take up and process the microreservoirs are af- fected to a greater extent than the surrounding tissue. In addition, encapsulation isolates the drug from the internal milieu until the carrier either starts to leak or releases its contents following cellular uptake. Microreservoirs include microcap- sules, red blood cells and their ghosts, and lipid vesicles. Lipid vesicles or lipo- somes have been widely used as delivery devices for ions, proteins, genetic ma- terial, and drugs ranging from anticancer, antibacterial, antifungal and antiviral to immunoregulators (see Refs. 8-17 for reviews).

The potential of liposomes as microscopic carriers was first realized following the discovery by Bangham and colleagues [18] that these synthetic vesicular struc- tures could entrap water-soluble compounds. However, these devices are not lim- ited to encapsulation of hydrophilic agents; water-insoluble compounds such as amphipathic drugs can also be successfully incorporated into liposomes. These hy- drophobic agents tend to partition into the nonpolar fatty-acyl region of the mem- brane bilayer where they reside in equilibrium with the phospholipids. Liposomes have been used to entrap a wide variety of compounds ranging from simple metal ions to enzymes and DNA plasmids. Benefits of using liposomes in drug delivery schemes include the ability to encapsulate a large number of drug molecules into a biodegradable reservoir which protects the drug from metabolism until it reaches its destination. In addition, the pharmacokinetics and biodistribution of the lipo- somes-associated drug are different from those of the free drug. This may be par- ticularly beneficial for coadministration of multiple drugs in combination regi- mens.

Although there have recently been many procedures devised for the efficient encapsulation of a variety of materials into liposomes, the concurrent develop- ment of the necessary technology needed to direct the carrier to specific targets has lagged far behind. However, if substances entrapped in liposomes are to be delivered successfully to selected sites, then efficient targeting techniques must be developed. Several different approaches have been used to target liposomes to specific organs or cells. The most direct approach was to deposit the drug-bearing liposomes directly to the site where therapy is desired. Since this method is some- times impossible and rarely practical, other liposome-targeting strategies had to be devised. Administration of liposomes intravenously or subcutaneously invariably results in their accumulation in liver and spleen or the lymphatics, respectively. This strategy of natural targeting can be utilized for treatment of pathologies lo- calized to the reticuloendothelial system (RES) but can represent a significant bar- rier for delivery to other organ systems.

A combination of two different approaches that have been mentioned above, vector or ligand conjugation and liposome encapsulation, would have many ad- vantages over either method alone; the ligand-conferring specificity and the lipo- some-contributing volume capacity. For example, addition of a vector molecule such as an antibody to the liposome surface would facilitate the specific binding of the liposome to its target cell where it can potentially release thousands of drug

Page 5: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 347

molecules as the result of a single binding event. The efficacy of using antibodies as biospecific targeting agents is well established and it was perhaps inevitable that they be utilized to give some direction to the inert lipid carriers. These antibody- targeted liposomes, or immunoliposomes as we shall hereafter refer to them, have been the subject of previous reviews [9,19-22]. In the sections which follow we will describe first the procedures devised to bring antibody and liposome together and then the in vitro and in vivo studies which have evaluated its utility as a drug delivery vehicle.

II. Preparation of antibody-targeted liposomes

There have been many methods developed over the past decade for the con- jugation proteins to l iposomes and the reader is advised to consult any of several recent thorough review [9,23,24]. Many of these coupling approaches are ideal- ized in Fig. 1. The most basic approach to antibody immobilization on liposomes is simple adsorption. Adsorpt ion can be viewed as either an intentional coupling method or the undesirable side reaction of a more specific coupling procedure. As the first procedure devised for targeting liposomes with antibodies, Weissmann and

Acylated antibody ~ Thiolated antibody

y Fab' (~ ~--"~ Protein A Fc Phospholipid ~ Biotin

Avidin

Fig. 1. Possible methods of conjugation of antibodies to the liposome surface (left to right). Binding of thiolated antibody to derivatized PE; binding of antibody to phospholipid through Schiff base re- duction or glutaraldehyde conjugation; incorporation of fatty acid-derivatized antibody; binding of Fab' to derivatized PE; indirect antibody conjugation through binding to protein A which has been thiolated and bound to derivatized PE, indirect binding of biotinylated antibody to biotin-PE mediated by

avidin.

Page 6: Antibody-directed liposomes as drug-delivery vehicles

348

TABLE I

EXAMPLES OF ANTIBODY-CONJUGATED LIPOSOMES

S. WRIGHT AND L. HUANG

Antibody Protein modification Liposome composi- Conjugation method Refs. tion

IgG,F(ab')2 none PC/Chol / Ganglio- Reduced Schiff base 40,41 side 45:45:10 between oxidized gan- (REV) glioside and protein.

Polyclonal none PC/Chol/PE Glutaraldehyde cross- 36 antibody 6:2:2 (SUV) linking between

amines. Mohoclonal SPDP thiolation PC/Chol/DTP-PE Disulfide 46 antibody 11.3:6.6:0.2 (SUV) Rabbit Fab' reduction PC/Chol/DTP-PE Disulfide 45

9:10:1 (REV) reduction PC/Chol/MPB-PE Thioether 49

10:9.5:0.5 (REV) SPDP thiolation PC/Chol/MPB-PE Thioether 47

64:35:1 (SUV) SAMSA thiolation PC/Chol/DCP/IA- Disulfide 34

(AETA)2-PE 2:1.5:0.2:0,2 (SUV)

",/-globulin SATA thiolation PC/Chol/DCP/ Thioether 54 MPB-PE 19:16:4:1 (REV) PC (SUV)

Rabbit Fab'

(Protein A)

Rabbit IgG

Beuce-Jones IA-DL-PE alkyla- monomer tion

F(ab')2 citraconylation and conjugation to PE with CDI

Monoclonal IgG NHSP acylation

PC/Chol 2:1 (SUV)

PE/PHC 8:2 or PC/Chol 2:1 (SUV or modified REV)

Spontaneous incorpo- 62 ration of protein into preformed liposomes. Spontaneous incorpo- 37,38 ration of protein into preformed liposomes. Detergent or etherme- 66-68,71 diated incorporation with dialysis.

Abbreviations: PC, phosphatidylcholine; PE, phosphatidylethanolamine; DCP, dicetyl phosphate; Chol, cholesterol; DTP, dithiopyridine; MPB, (maleimidophenyl) butyryl; IA, iodoacetyl; AETA, amino- thioacetyl; DL, dansyllysyl; CDI, carbodiimide; SAMSA, S-acetylmercaptosuccinic anhydride; SATA, Succinimidyl-S-acetyl thioacetate; NHSP, N-hydroxysuccinimide ester or palmitic acid.

co-workers [25,26] observed that aggregated immunoglobul ins would coat and partially insert into l iposomes with the Fc region exposed to the surrounding me- dium. Significant adsorption of antibodies to small unilamellar vesicles (SUV's) can be achieved ei ther during [27-29] or after [26,30-33] their sonication. Adsorp t ion was greater for small [30] negatively charged [28] l iposomes suggesting that inser- tional or ionic mechanisms were involved. Sonication of the ant ibody in the pres- ence or absence of lipid had no significant effect on their binding capacity [28]. Adsorp t ion has recently been re-emphasized by Senior et al. [30] following their detect ion of large non-specific binding of native ant ibody to l iposomes which had been pre-derivat ized for covalent conjugat ion with thiolated antibodies. F rom 34 to 89% of added IgG could be bound on the vesicle surface and there was no leak-

Page 7: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 349

age of vesicle contents reported. Thiolated IgG exhibits very little non-specific binding to SUV's ]34]. This method is attractive in its simplicity and the adsorp- tively coated liposomes exhibit considerable stability even in the presence of serum [27,28,30]. However, the mode of attachment is non-specific and the coating ef- ficiency is highly variable. Even though Huang and Kennel [28] found no large in- terspecies differences in the adherent capacity of normal IgG, there may be dif- ferences in IgG subtype for a given liposome [30].

There are two major approaches which have been developed for the specific and controlled coating of liposomes with antibody. The first approach is to derivatize lipid, typically the amino group of phosphatidylethanolamine (PE), which, follow- ing incorporation into the liposome, will covalently attach antibody which is sub- sequently applied. The second major approach is to convert an otherwise hydro- philic protein antibody molecule into an amphipathic molecule which can then non- covalently intercalate into the liposomal bilayer. We will discuss briefly only some of the more popular techniques along with their most recent modifications. Ex- amples of these procedures are listed in Table I.

H.1. Covalent conjugation to pre-formed liposomes

One of the first successful attempts at covalent attachment of antibody to lipo- somes was achieved by glutaraldehyde conjugation. The traditional use of glutar- aldehyde has been for cell fixation in which proteins are effectively cross-linked via their amino groups. Under the proper conditions, glutaraldehyde can also be used for controlled conjugation of amino groups between antibody and PE. Following their earlier success with liposome immobilized a-chymotrypsin [35], Torchilin and co-workers [36] used glutaraldehyde to conjugate polyclonal Ig to PE-containing SUV's. These immunoliposomes retained antigen binding capacity both in vitro and in vivo and although up to 60% coupling efficiency was achieved, the net antibody incorporation was low. Potential undesirable side reactions would be homocou- piing between proteins or between liposomes which would limit its general use- fulness.

Another fundamental conjugation method involves coupling of protein carbox- ylic groups with the primary amine of PE on the liposome. Coupling between an- tibody and liposome has been achieved with a water soluble carbodiimide (EDCI) [37]. A major drawback is potential protein-protein cross-linking but protection of the protein amines by prior citraconylation minimizes this side reaction ]37,38]. Further, this method is inefficient and produces unstable liposomes which retain little of entrapped contents [37,38]. More recently, Kung and Redemann [39] have used carboxyacyl derivatives of PE for conjugation to protein amines using EDCI. PE was derivatized with C4 to C18 dicarboxylic acids which provide spacer arms of various chain lengths. Maximum antibody incorporation was found for 1,12-do- decanedicarboxylic acid (C16). Incorporation was dependent upon the spacer length and the shortest derivative, succinic (C6), produced no detectable protein binding.

Protein attachment to liposomes can also be achieved via simple Schiff base for- mation under the proper conditions [40--42]. Oxidation by periodate of the car-

Page 8: Antibody-directed liposomes as drug-delivery vehicles

350 S. W R I G H T A N D L. H U A N G

bohydrate moieties on glycolipid-containing liposomes to the corresponding al- dehydes facilitates Schiff base formation with protein amines. The conjugate is then stabilized by subsequent reduction with sodium borohydride or sodium cyanobo- rohydride. Coupling efficiencies of up to 200 Ixg IgG per p.mol lipid has been achieved [40,41]. These liposomes showed greater binding capacity to target cells than immunoliposomes produced by earlier conjugation techniques. Potential drawbacks of this technique are that 9-20 mol% of glycolipid must be included in the liposome and that periodate addition to the pre-formed liposomes may oxidize the fatty-acyl chains of the lipid and/or the contents within the liposome. In ad- dition, up to a 60% reduction of immunoreactivity of antibodies following period- ate oxidation has been observed [42]. Aldehyde groups can alternatively be gen- erated from the carbohydrate groups on the constant region of the heavy chain of immunoglobulins by mild oxidation with periodate or glucose oxidase [43]. Ad- dition of oxidized antibody to liposomes containing a hydrazide group resulted in up to 535 ~g of IgMs per i~mol phospholipid [43].

One of the most popular conjugation methods utilizes the heterobifunctional agent N-hydroxysuccinimidyl-3-(2-pyridyldithio)propionate (SPDP) [44]. In this scheme, SPDP is reacted with liposomal-PE to produce the dithiopyridine deriv- ative, DTP-PE, which can then react by disulfide interchange with any protein having a free thiol [44-47]. Immunoglobulin Fab' fragments are thus potential coupling partners with yields up to 600 Ixg Fab' per ~mol lipid [45]. Typically 1-5 mol% of DTP-PE is incorporated into the liposome with complete retention of en- trapped molecules [44-47]. If intact antibody is used (following SPDP modifica- tion) then greater than 60% of antibody-conjugated liposomes could be precipi- tated by Staphylococcus aureus indicating exposed antibody Fc regions on the liposome surface. Conjugation of antibody Fab' via the free SH is thus disadvan- tageous for in vivo applications because if the Fc portion remains intact and ex- posed, the immunoliposomes can be trapped by Fc receptor-bearing cells present in the host. Advantages of this method are a lack of homocoupling between pro- teins or liposomes, provision of a spacer arm, and the stability of liposomal-PE- DTP and antibody-DTP. The primary disadvantage is that the disulfide product is susceptible to reductive cleavage by endogenous thiols and would therefore be generally unsuitable as an in vivo targeting vehicle.

Goundalkar et al. [48] demonstrated that derivatized fatty acid could also be used as an anchor for antibody conjugation to pre-formed liposomes. Stearylamine was reacted with SPDP and then incorporated into liposomes. Thiolated antibody could then be conjugated to the liposomes with a 24-32% binding efficiency. Approx. 20% of an encapsulated drug leaked out during the latter binding reaction. In 1982, Martin and Papahadjopoulos [49] introduced a new sulfhydryl-reactive lipid deriv- ative N-[4-(p-maleimidophenyl)butyryl]phosphatidylethanolamine (MPB-PE). When reacted with a thiol-bearing antibody or Fab' fragment, a stable thioether bond is formed between the two by sulfhydryl addition across the maleimide dou- ble bond. Coupling efficiency is 20-30%, producing up to 600 I~g Fab' per ~xmol lipid corresponding to about 3000 Fab' per 0.2 Ixm vesicle. The advantage of this protocol over that with SPDP is that the conjugation is essentially irreversible and

Page 9: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 351

the covalently linked product is stable in serum [49]. For most of the earlier tar- geting strategies, SUV's were most often employed. However, large unilamellar vesicles (LUV's) such as those produced by the reverse-phase evaporation tech- nique [50] have larger internal volume and better trapping efficiencies. Reverse- phase evaporation vesicles (REV's) conjugated to antibody with this derivative are stable and do not aggregate or leak [49]. In addition, they exhibit good binding and retention of contents even in 50% serum [49]. However, depending upon the lipid composition, high concentrations of the derivative can perturb membranes. Coupling of Fab' to liposomes with 5 mol% MPB-PE caused increases in size, po- lydispersity and resulted in up to 95% loss of entrapped carboxyfluorescein [51]. Others have reported that up to 10 mol% MPB-PE could be used without any ad- verse effects on bilayer structure [49]. Again, these discrepancies are probably largely due to differences in lipid composition and methods used for preparation of liposomes.

MPB-PE or DTP-PE have been used with success for conjugation of Fab' which has a free thiol available for conjugation following cleavage of F(ab')2 with di- thiothreitol (DTT). If the protein has no free sulfhydryl groups available, then it can be thiolated with a variety of agents (Table II). In fact, SPDP was originally designed to introduce sulfhydryl groups into proteins for reversible pro- tein-protein conjugation [52] and was therefore the early choice to thiolate anti- bodies [44,46,47]. It is necessary that soluble F(ab')2 or Ig-DTP be first reduced, the products of which are unstable and must be used immediately. In 1983, Dun- can et al. [53] introduced a new thiolating agent, succinimidyl-S-acetylthioacetate (SATA). This agent has the advantage that deprotection of the sulfhydryl is ac- complished with hydroxylamine at neutral pH and does not necessitate removal of strong maleimide-reactive reducing agents prior to addition to derivatized lipo- somes. SATA has been used to conjugate up to 300 Ixg Ig per p~mol lipid into MPB- PE sensitized liposomes [54]. S-acetylmercaptosuccinic anhydride (SAMSA) is very similar to SATA but alters the electrostatic properties of the protein. Traut 's re- agent (2-iminothiolane) [55] has been used to thiolate many proteins including monoclonal antibody [56] and has the advantage that no deprotection is required.

There have been several novel methods developed for the indirect coupling of

TABLE II

METHODS OF INTRODUCTION OF THIOLS INTO PROTEINS

Protein Thiolation reagent Activating agent Refs.

Polyclonal (rabbit) N-succinimidyl-3-(2-pyridyldi- dithiothreitol (DTT) 52 antibodies thio)propionate (SPDP) Monoclonal IgG SPDP DTT 44,46 Protein A SPDP DTT 44,47 ~ - g lobu l i n Succinimidyl-S-acetyl thioacetate hydroxylamine 53,54

(SATA) IgG S-acetylmercaptosuccinic anhy- hydroxylamine 34

dride (SAMSA) Monoclonal IgG 2-Iminothiolane (Traut's reagent) none 55,56

Page 10: Antibody-directed liposomes as drug-delivery vehicles

352 S. WRIGHT AND L. HUANG

antibody with liposomes. These systems are designed primarily for targeting of li- posomes to cells with derivatized antibody already bound to their surface. Two principal schemes have been utilized based upon the affinity between protein A of S. aureus and antibody Fc, and the affinity of biotin for avidin. Leserman and co- workers [44,57,58] conjugated protein A to liposomes after both had been deri- vatized with SPDP. Protein A-bearing liposomes are added to cells which had been pre-treated with the targeting antibody resulting in specific binding of the lipo- somes of the cells with the newly added Fc. A similar mechanism is employed with the biotin-avidin system [59-61]. Typically, liposomes and antibody are both de- rivatized with biotin. Target cells bound with biotinylated antibody are then treated with avidin followed by the biotinylated liposomes. Avidin has the capacity to bind up to four biotin molecules with very high affinity, thus serving to cross-link the two biotinylated components. Streptavidin has recently been used to avoid the non- specific binding properties of native avidin [59].

H.2. Incorporation of lipophilic antibodies

The second major approach to incorporate antibody into liposomes is to add a hydrophobic moiety to the antibody which facilitates its association with the am- phipathic bilayer. Sinha and Karush [62] were the first to attempt this approach utilizing PE and the hydrophobic anchor. An alkylating phospholipid derivative, N-(N-iodoacetyl-N-dansyllysyl)-PE, was used to derivatize Bence-Jones mono- mers which then spontaneously attached to either preformed liposomes or red cell ghosts. However, this iodoacetyl derivative of PE is not an effective crosslinking agent for coupling thiolated protein into pre-formed liposomes [34]. Addition of one or two aminoethylthioacetyl (AETA) groups as spacers between iodoacetyl and PE groups makes it a reasonably efficient surface target for conjugation with thiolated antibody [34]. Periodate oxidation of the carbohydrate unit of phospha- tidylinositol to a free aldehyde permits Schiff base formation between protein and phospholipid [63]. Derivatized protein can then be incorporated into liposomes by detergent dialysis. Yet another approach utilizes a polysaccharide derivative of cholesterol as the anchoring molecule [64]. The polysaccharide used in these stud- ies, pullulan, enhanced the stability of the immunoliposomes without affecting its target specificity.

The most commonly used nonpolar anchor is a fatty acid such as palmitic acid. Two different protocols have been developed for conjugating fatty acid onto an- tibody. The N-hydroxysuccinimide ester of palmitic acid (NHSP) is one popular choice for modifying antibody to make it more lipophilic. Huang, A. et al. [65] were the first to use this agent for incorporation of monoclonal antibody into li- posomes. The antibody is first reacted with NHSP with the extent of derivatization controlled by the input ratio of NHSP to Ig and/or the reaction time [66]. Optimal coupling stoichiometry for incorporation into detergent dialysis liposomes was 4-5 palmitoyl chains per IgG [66-68]. Under these conditions, about 90% of the acyl chains were located on the heavy chain of the antibody. Antigen binding capacity of the antibody was reduced 3-4-fold but the liposomes exhibited good target cell

Page 11: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 353

specificity [65-68]. Slow infusion of palmitoyl IgG in detergent into a suspension of DPPC liposomes (DOC:PC = 60:1, mol/mol) at their main phase-transition temperature resulted in up to 52% incorporation [70]. Acylated antibody can also be incorporated into modified REV liposomes [71]. Antibodies modified by this technique have recently been shown to stabilize otherwise unstable lipid assem- blies into forming liposomes [72]. The major benefit of this targeting scheme is that all procedures may be carried out in the absence of harsh coupling reagents and that potential homocoupling is not a concern.

Utilizing palmitic chloroanhydride to acylate their protein, Torchilin et al. [73] were able to mediate protein incorporation either during or subsequent to lipo- some formation. The advantage of this latter approach is that it can be completed in the absence of detergent although incorporation is slow and of low efficiency [73].

III. Immunoliposome binding and drug delivery in vitro

111.1. Binding to immobilized antigens

The various techniques that were developed to conjugate antibodies to the li- posome surface were devised for the ultimate goal of enhancing drug delivery. Ef- fective drug delivery with liposomes will involve three phases: transport through blood and interstitium to the target site, binding to target antigens on the cell sur- face, and internalization and intracellular release of encapsulated drug. We will first discuss the in vitro situation in which the model is tested under near ideal con- ditions thus involving only the latter two aspects, binding and internalization.

The initial step in any effort at liposome targeting is to evaluate the specificity of binding between carrier and target. The addition of antibody to the liposome surface greatly enhances its avidity for antigen bearing cells, but sometimes at the expense of also increasing the nonspecific binding to nontarget cells. Therefore it is usually necessary to compare the interaction between immunoliposomes and its target with either the immunoliposome and a nonantigenic cell or between the tar- get cell and a nonspecific immunoliposome.

The first reported successful attempt at antibody-mediated targeting was achieved by Gregoriadis and Neerunjun [29]. Sonicated liposomes containing 111I-labeled bleomycin were coated with adsorbed 125I-labeled IgG raised against whole cells. When anti-HeLa cell antibodies were used, the uptake by HeLa cells of both la- bels was 25-fold greater than that for fibroblasts. Specific binding of antibody-coated liposomes to targeted fibroblasts and AKR-A cells was also demonstrated [29].

Targeting to human erythrocytes was examined with periodate conjugated im- munoliposomes. Up to 8000 liposomes (0.2 ~m SUV) coated with 143 anti-human erythrocyte rabbit F(ab')2 molecules could bind to each human erythrocyte [40]. This represents binding of about 80% of the added immunospecific liposomes compared to less than 1% binding of added nonspecific immunoliposomes. Im- munospecific targeting to sheep erythrocytes has also been reported for liposomes coupled with palmitic acid esterified antibodies [69]. Liposome binding to the sheep

Page 12: Antibody-directed liposomes as drug-delivery vehicles

354

TABLE III

DELIVERY WITH pH-SENSITIVE IMMUNOLIPOSOMES

S. WRIGHT AND L. HUANG

Liposome Antibody Entrapped agent Target c e l l Comments Refs.

PE/PHC 8:2 palmitoyl anti- calcein L-929 very efficient de- 83 (modified REV) H2K k livery, blocked

at low temp or with lysosomo- tropic agents

PE/OA 8:2 palmitoyl anti- ara-C MTX L-929 only drugs in 87 (modified REV) H2K k targeted pH-sen-

sitive immunoli- posome were beneficial, prely- sosomal delivery

PE/OA 8:2 palmitoyl anti- DT-A L-929 only targeted 88 (DRV) H2K k pH-sensitive im-

munoliposomes were toxic, blocked by free antibody or im- munoliposome

PE/Chol/OA palmitoyl anti- plasmid DNA L-929 ( tk) targeting en- 94 4:4:2 (detergent H2K k (adsorbed) hanced transfor- dialysis) mation PE/Chol/OA palmitoyl anti- plasmid DNA RDM-4 efficient in vivo 92 4:4:2 (detergent H2K k (adsorbed) delivery, con- dialysis) trolled expres-

sion

red blood cells was fast (within 1 min) and produced some changes in morphology in the cells.

The effectiveness of monoclonal antibody for liposome targeting to cultured cells was demonstra ted by Huang et al. [65]. Using an N-hydroxysuccinimide ester of palmitic acid, monoclonal antibody to the mouse histocompatibility antigen H-2K k was acylated and incorporated into phosphatidylcholine (PC) liposomes (0.09 Ixm) by detergent dialysis. These liposomes showed specific binding to H-2K k express- ing L-929 cells but not to A-31 cells which expressed the H-2K d antigen. For either native liposomes or liposomes derivatized with nonspecific antibody there was no significant binding to either cell type reported. The liposome preparat ion was het- erogeneous with respect to antibody density and only those liposomes which had greater than 67 antibody molecules per liposome became cell associated. Subse- quent studies indicated that l iposome-associated antibody containing 4-5 palmi- toyl chains per IgG had an apparent dissociation constant 6-7-fold higher than na- tive antibody [66]. Optimal incorporation of these antibodies into liposomes was achieved using an initial lipid/protein ratio of 10/1 (w/w). These liposomes which had only about 48 IgG molecules per liposome demonstrated saturable binding to L-929 cells. At saturation there was about 5 times as much antibody associated to

Page 13: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 355

liposome-treated cells as there was to cells incubated with free underivatized an- tibody. These acylated antibodies could also be efficiently (over 80%) incorpo- rated into REV vesicles (0.15-0.45 p~m) which had a high drug capture efficiency [71]. Immunoliposome (200 IgG per liposome) binding to RDM-4 non-adherent lymphoma cells (H-2K k) was saturable with respect to antibody or lipid binding and could be blocked by free anti-H-2K k IgG. When incubated at 4°C, about 90% of the immunoliposomes taken up by RDM-4 cells could be removed by proteases but only 30% could be released from cells incubated at 37°C [74]. This suggests that at 4°C most of the liposomes are surface bound and not internalized as they are at the higher temperature. Cytochalasin B or a combination of 2-deoxyglucose and NaN 3 could inhibit the uptake at 37°C. Specific intracellular delivery to target but not control cells was demonstrated with encapsulated carboxyfluorescein (an impermeable aqueous fluorophore) which was diluted and dequenched when re- leased into the cytosol subsequent to liposome internalization and processing at 37°C (but not at 4°C) [74].

Indirect targeting with protein A conjugated liposomes also proved to be an ef- fective means to achieve binding to target cells [44]. Human leukocytes were shown to bind with protein A liposomes (SPDP conjugated) only in the presence of anti- human 13-2-microglobulin antibodies. Direct conjugation of this IgG to liposomes (via SPDP) also produced selective binding to human but not mouse cells [44]. In- corporation of up to 4.4 mol dithiopyridine per mol IgG had no appreciable effect on binding via Fab or Fc regions [46]. Only one molecule of coupled antibody per liposome was apparently sufficient to mediate liposome binding to either target cells or to S. aureus .

In contrast to immunoliposomes in which both Fab and Fc portions are exposed, liposomes conjugated with Fab' will bind only to antigen and not to Fc receptor- bearing cells. Martin and co-workers [45] conjugated antihuman erythrocyte Fab' fragments to SPDP-derivatized reverse phase vesicles (6000 Fab' per 0.2 p~m ve- sicle). Binding of up to 5000 vesicles per erythrocyte was specific for human cells and was moderately stable in the presence of 50% serum. Serum stability could be improved with an irreversible conjugation method utilizing MPB-PE as the lipid anchor [49]. Vesicles retained their aqueous contents during conjugation and bind- ing [45,49]. Immunoliposomes could also be targeted to erythrocytes with anti-gly- cophorin A antibodies [75]. Binding was increased 80-fold and was inhibited only by soluble anti-glycophorin and not its Fab' fragment. Targeting of liposomes with anti-rat erythrocyte F(ab')2 was found to enhance their binding to erythrocytes in rat blood [76]. The permeability properties of the liposomes were not affected by binding to target cells.

For any antibody-targeting strategy to succeed there must be an epitope on the surface of the intended cell which is somehow unique so that molecules can be di- rected to or processed by that cell specifically. The field of immunoliposome tar- geting as it has developed has been directed primarily at, although not exclusively for, antitumor therapy. However, there has been little effort to target these car- riers specifically to tumor cells. Most of the in vitro delivery studies with model systems, although utilizing tumor cell lines, have been based upon targeting di-

Page 14: Antibody-directed liposomes as drug-delivery vehicles

356 S. WRIGHT AND L. HUANG

rected toward epitopes such as Fc receptors or histocompatibility antigens. This is not for a lack of targetable sites on tumor cells. There are presently dozens of mol- ecules which have been identified as tumor-specific or more frequently as tumor- associated antigens, and most of these are surface molecules to which antibodies have been generated. These targets may include modified or overexpressed pro- teins or modified glycolipids. Transformation-associated glycolipids may be the product of either incomplete synthesis resulting in accumulated precursors or due to induction of synthesis of foreign glycolipids. Urdal and Hakamori [61] indirectly targeted antibody or antibody-associated liposomes to the tumor-associated gly- colipid ganglio-N-triosylceramide found on L5178c127 or Kirsten virus-trans- formed 3T3 cells. Coupling between antibody and liposome was dependent upon the high-affinity binding of avidin to biotin. Cells were first incubated with bioti- nylated anti-glycolipid antibody which was then followed by addition of drug-con- taining liposomes to which had been conjugated either avidin or biotin (plus free avidin as the cross-linker in the latter case). Avidin was covalently coupled to gly- colipid-containing liposomes by periodate oxidation, whereas biotin was incorpo- rated by using biotinyl-PE. In the presence of free avidin as cross-linker, the an- tibody-treated L5178c127 cells were not at all sensitive to actinomycin D encapsulated in biotin-liposomes, although they were sensitive to the targeted drug, biotinyl neocarzinostatin. Neocarzinostatin was believed to be cytotoxic without being internalized [61] based upon previous reports that its effects can be me- diated at the surface of the cell [77]. Transformed 3T3 cells were only moderately sensitive to the actinomycin-D liposomes although it was demonstrated that they did bind to the target cells. This suggests that either the bound liposomes were not internalized or if they were, then the contents were not effectively delivered into the cytosol.

Torchilin and colleagues [78,79] have examined the potential of antibody-con- jugated liposomes to bind to extracellular matrices. Antibodies against fibronectin or laminin were modified by the palmitoylchloride procedure [73] and incorpo- rated into PC liposomes with trace amounts of [3H]cholesterol or [14C]cholesterol oleate (lipid markers) by detergent dialysis. Using protein monolayers as the tar- get, anti-laminin or antifibronectin immunoliposomes bound with nearly the same specificity and affinity to the glycoproteins as the free antibody. This targeting scheme offers the possibility of targeting to areas of vascular damage where pro- teins of the extracellular matrix are exposed. Deendothelialization and exposure of subendothelial components may be the consequence of trauma, surgery or var- ious cardiovascular pathologies. These events invariably lead to platelet aggrega- tion and thrombus formation at the site of damage which could represent a threat to patients at risk for coronary thrombosis. In fact, perfusion studies with partially denuded arterial segments demonstrated in situ that immunoliposome targeting was feasible for these applications [79]. Liposomes which were conjugated with human anti-collagen type-I antibodies (biotin-avidin coupling) could be visualized in den- uded areas of the vessels [61]. Although drug delivery to luminal lesions was not attempted in these studies, the potential for liposome delivery of anti-platelet or related drugs is apparent.

Page 15: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 357

Since most immunoliposomes have several antibody molecules attached to their surface, it might be expected that, depending upon the antigen density, a single liposome could be bound to the target cell at several points. Even derivatized an- tibodies have high lateral diffusional mobilities within the liposome bilayer [80]. It is therefore plausible that subsequent to the initial binding event, additional an- tibodies may diffuse into the area and form multipoint attachments or multivalent binding. This might be expected to appear as an increase in the binding affinity of the intact liposome. In fact, that is what is observed experimentally [43,45,81]. Fluorescence quenching of fluorescein by anti-fluorescein antibodies was utilized by Heath et al. [43] to examine possible multivalent interactions between lipo- some-associated antibody (periodate conjugated and only 40% active) and target cells. Binding to fluorescein isothiocyanate (FITC)-labeled erythrocytes (108 FITC/RBC) was rapid (within 5 min at 37°C) and was insensitive to antibody den- sity above 230 active binding sites per liposome. They found that there was up to a 1000-fold greater affinity for immunoliposomes (K a = 109 M -1) than free anti- body (Ka = 106 M -~) for the target cells over a wide range of cell densities al- though at higher cell densities apparent hemagglutination reduced the available binding sites [43].

Quantitative analysis of immunoliposome binding to antigens immobilized on polystyrene microtitre plates has been reported by Klibanov et al. [81]. Liposomes conjugated with palmitoyl-anti-IgG (with 40% of protein being trypsin sensitive) bound to an antigen monolayer with an apparent dissociation constant of (1.5-5).10 -9 M liposomes (for a 100 nm liposome particle of 105 lipid molecules). For native antibody the K d was 2.5 • 10 .8 M antibody, 10-fold lower than for the immunoliposomes (which had about 30 exposed antibodies per liposome). The mere 10-fold enhancement of binding affinity conferred by liposome incorporation is much lower than the 1000-fold difference reported by Heath et al. [42] but is due to the different ways the authors express their values. Heath et al. relate protein association constants (intrinsic vs. functional) whereas Klibanov et al. [81] were relating the dissociation constants of binding particles (antibodies vs. liposomes). In either situation it can be concluded that a drug encapsulated in an antibody- targeted liposome would have a greater probability of interacting with the target cell than it would have if simply attached directly to the antibody.

In studies where the antigen density on target cells was manipulated there was not a simple relationship between liposome binding and antigen density. Lipo- somes incorporating variable amounts of palmitic acid derivatized antibody (anti- H-2K k IgG) [65] were incubated with RDM-4 lymphoma cells in which the num- ber of surface H-2K k molecules was controlled by treatment of cells with protein- ase k [82]. For liposomes (90 nm diameter) with 0 to 55 total antibodies incor- porated, binding of the lipid component (labeled with 3H-cholestanyl ether) of antibody-associated liposomes to cells of any antigen density was proportional to surface density of antibody on the liposome. However, binding of immunolipo- somes with high antibody density did not vary considerably between cells with 8.7 • 103 and 6.4 • 1 0 4 sites per cell, although as the antibody density was lowered the difference became larger. Therefore, immunoliposomes with low antibody density

Page 16: Antibody-directed liposomes as drug-delivery vehicles

358 S. WRIGHT AND L. HUANG

may be better at discriminating between high and low antigen density cell popu- lations.

Another potential mechanism for enhancing cell specificity through multipoint binding is to target to multiple antigens on a given cell. This would be of advan- tage for cells which have a unique combination of antigens rather than a specific single antigen for targeting. Liposomes conjugated with antiglycophorin A anti- bodies have been shown to interact with the human leukemia cells line K562 either through their Fc receptors or by binding to glycophorin [75]. Binding of immu- noliposomes was only partially blocked by excess anti-glycophorin Fab' or excess human IgG and was completely prevented only when both types of competing an- tibodies were present. In addition, both ligands were needed to block delivery of cytotoxic drugs to these cells [75]. This scheme may be especially useful for deliv- ery to tumor cells which shed surface antigen which could block binding of lipo- somes targeted only to that antigen. The idea of multitargeted binding has also been studied from a different approach by Trubetskoy et al. [83]. An indirect tar- geting protocol was used where binding of biotinylated liposomes to protein monolayers bound with different biotinylated antibodies was mediated by avidin. Addition of antifibrinogen, antifibronectin, or anti-LDL antibodies to an immo- bilized mixture of target proteins resulted in binding of fewer liposomes than if all three antibodies were present simultaneously. The effects were subadditive. An alternative to the use of multiple antibodies against individual targets might be found in the use of hybrid antibodies. The recent generation of these biospecific antibodies having independent Fab"s permits simultaneous binding to two differ- ent antigens [84].

111.2. Drug delivery to cells in culture

Some of the earliest studies utilizing immunoliposomes were aimed at delivery to enzyme-deficient cells as a model for treatment of lysosomal storage diseases. For example, multilamellar vesicles (MLV's) containing horseradish peroxidase and coated with non-specific heat-aggregated IgM were given to dogfish phagocytes which are normally low in peroxidase activity [25]. These cells bound the lipo- somes via their FC receptors, endocytosed the complex, and were subsequently able to express peroxidase activity. This was the first demonstration of effective delivery of liposome contents to a target cell. This method was also found to be effective for the in vitro introduction of hexosaminidase A into leukocytes of Tay- Sachs patients [26].

Leserman and co-workers utilized a similar targeting scheme to examine the ability of liposomes to deliver cytotoxic drugs to three fundamentally different types of cells [85]. Antibody Fc receptors are rapidly internalized by the highly phago- cytic tumor cell line P388D1 but not by the nonphagocytic P388 line. Liposomes containing the antifolate drug methotrexate (MTX) and the antigenic lipid N-di- nitrophenylaminocaproyl-PE (DNP-cap-PE) were mixed with target cells that had been pre-incubated with anti-DNP antibodies which became cell associated via their Fc region. Unlike cells that were either non-phagocytic (P388) or receptor nega-

Page 17: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 359

tive (EL4), the P388D1 cells were much more susceptible to the MTX encapsu- lated in the immunoliposomes than to the free drug as determined by [3H]deoxyuridine incorporation [85]. Although it was the initial premise that drug delivery to the cell was imminent if one could just achieve tight binding of the li- posome to the target cell, a comparison between the P388 and the P388D1 data suggested otherwise. It was apparent that an active means of internalization of surface-bound liposomes was required.

The requirement for factors other than binding were also demonstrated by Weinstein and co-workers [86]. In this study the binding of liposomes to human lymphocytes was mediated by bivalent antibody which was directed against surface components common to the two opposing structures (trinitrophenyl moiety). The use of the self-quenching dye carboxyfluorescein (CF) entrapped inside the lipo- somes permitted quantitative determination of liposome binding and, more im- portantly, permitted one to distinguish between delivery of entrapped marker to the cell cytosol from simple binding. Although there were from 5000 to 14000 di- oleoylphosphatidylcholine (DOPC) SUV's bound to each cell, there was no re- lease of contents into the lymphocytes. Since the dye itself is relatively imper- meant, it was concluded that the liposomes were not interacting with the target cell beyond the initial binding step. For efficient cytoplasmic delivery of contents, the liposomes must either fuse at the cell surface or be internalized where it can eventually release its contents directly into the cell interior during cellular proc- essing. The target antigen used in this early study was a trinitrophenyl moiety (TNP) which had been randomly incorporated into the lymphocyte membrane. Failure to achieve delivery was probably due to exclusion of the TNP-antibody-liposome complex from endocytic vacuoles. Thus, even though the liposome may be tightly bound to the cell, if there is no additional processing to accommodate drug trans- fer, then the system will be of little use as a therapeutic modality. However, this limited interaction can be utilized for cytological studies where fiuorophore-con- taining liposomes can be used as a sensitive means to immunolabel cells [87,88].

The introduction of efficient methods of antibody coupling to liposomes gen- erated great expectations for the therapeutic potential of immunoliposomes. Pre- vious studies up to this point had utilized the Fc portion of the antibody most fre- quently to target the liposomes to cells bearing Fc receptors. In 1981 Leserman et al. [89] targeted liposomes to murine histocompatibility antigens by covalent at- tachment of antibodies to SUV's using SPDP. Binding and delivery was followed by encapsulating CF and MTX in the liposomes, respectively. It was found that upon targeting to mitogen-induced spleen blast cells there was not a direct cor- relation between the number of liposomes bound and the effect of encapsulated MTX. Liposomes targeted to other histocompatibility antigens were more effec- tive at drug delivery than those targeted to H-2K k, even though more H-2Kk-tar - geted liposomes were bound to the cells. The lysosomotropic amine, NH4C1, in- hibited the effects of encapsulated but not free drug, suggesting that endocytosis was the mechanism of uptake. Therefore, if this type of liposome is bound to a surface component which is not actively internalized then it will not be an effective delivery vehicle.

Page 18: Antibody-directed liposomes as drug-delivery vehicles

360 S. WRIGHT AND L. HUANG

Using the same targeted liposomes, this group utilized this system not so much for its therapeutic potential, but more as a means to measure endocytosis apart from surface binding. In studies designed to examine the fate of MHC-encoded determinants, they observed differential endocytosis by T and B lymphocytes of an identical specific surface antigen [90]. Liposomes containing MTX and targeted to cells expressing H-2K or surface molecules of molecular weight 94 000 and 180 000 were much more cytotoxic for T cells than B cells. However, even though T-cells extensively internalized the antibody-targeted liposomes, there was no enhance- ment of MTX cytotoxicity by encapsulation in targeted liposomes as compared to the free drug. Each cell type was equally sensitive to free drug. These results sug- gest that for effective chemotherapy with antibody-coated liposomes, targeting to an antigen which has a much higher internalization rate than other surface mole- cules may be sufficient to generate selective toxicity. Thus, targeting with anti- bodies to tumor-specific antigens might not be necessary if the endocytic rate was simply higher for a common antigen on the tumor cell.

Rather than generate a large panel of specifically targeted immunoliposomes, an indirect targeting strategy was devised in which liposomes coated with protein A from S. aureus could be used against cells already targeted with free antibody [57,89,91,92]. The primary advantage of this approach is that a single population of liposomes can be used against a variety of target antigens. For DPPC liposomes containing 34% cholesterol and 1% DTP-PE, up to 15 protein A molecules could be conjugated to SUV's following SPDP modification [91]. As before, it was found that B-cells were insensitive to liposome-entrapped MTX even though they had a greater surface density of the target antigen, H-2K k, than T-cells which were quite sensitive to these cytotoxic liposomes. However, in addition to displaying cell specificity the effects were also antigen specific. If the liposomes were instead tar- geted to I-EK molecules on the B-cells, then MTX induced inhibition of 3H-dUrd was observed [91]. The two cell types were equally sensitive to free MTX but only the T cells were more sensitive to encapsulated over free drug, regardless of target antigen. Using gold-labeled protein A bound to surface-attached antibody, Machy et al. [93] have recently shown that T-lymphocytes spontaneously internalize 20-40% of their MHC-encoded class I molecules in a process involving coated pits and coated vesicles. No internalization was observed in B cells. These observa- tions point out that internalization of a given surface antigen may be drastically different between cell types and if endocytosis is to be relied upon as the principal means of uptake, then more consideration should be given as to the turnover rate of target antigen than to the surface density of the antigen.

The rate of endocytosis of liposomes bound to MHC-encoded proteins can be controlled by drugs that interfere with energy metabolism, microfilament organi- zation or phospholipase activity [58]. Intracellular release of CF from liposomes bound to cells via protein-A-antibody was inhibited by lysosomotropic NH4C1, the metabolic poison phenylarsenoxide, and the Ca2+-dependent enzyme phospholi- pase A:. There was also a partial requirement for extracellular Ca 2+ for endocy- tosis of liposomes. More importantly from a drug delivery standpoint, they were able to demonstrate that the calmodulin antagonist trifluoperazine enhanced the

Page 19: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 361

apparent rate of endocytosis. Increasing internalization augments the effects of cy- totoxic liposomes.

There are strict limitations on the size of particles that can be taken up by en- docytosis and liposome size has been shown to be a critical factor in determining the extent of liposomal drug delivery. LUV and SUV conjugated to anti-Thy 1.1 antibody were tested for their ability to deliver MTX-y-asp to either AKR or R1.1 T-lymphoma cells [94]. Although either type of liposome bound to the target cells, the LUV's (0.45 txm) failed to enhance the drug effects for either cell type whereas SUV's (0.053 jxm) were 22-40 times more effective than the free drug. The role of liposome size in drug delivery was also examined for cultured tumor cells using the indirect targeting strategy with protein-A conjugated liposomes. For liposomes of 80, 200 and 400 nm entrapping MTX or CF there was an inverse relationship between liposome size and the ability to deliver a cytotoxic drug to cells targeted with anti-H-2K k IgG [92]. This did not correlate with cell binding as there was more CF associated with the larger liposomes than there was for the smaller liposomes bound to cells. The enhanced cytotoxicity of the small liposomes was thought to result from size restrictions of the endocytic vesicles such that only the smaller li- posomes could be engulfed. The average size of coated pits are estimated at about 150 nm in diameter so that liposomes smaller than this should be readily accepted. In fact, there was no difference in drug delivery for liposomes of 60 or 80 nm in diameter [92]. In addition, RDM-4 cells or lymphoblasts did not process immu- noliposomes of 200 nm or larger although L-929 cells were sensitive to the larger (200 and 400 nm) liposomes, although the smaller carriers were still more effective [92]. This may suggest that the endocytic size restrictions are cell-type dependent. Moreover, fibroblasts such as L929 cells, are known to internalize their Class-I molecules via uncoated cell surface vesicles and tubular invaginations rather than coated pits [95].

The antigen selectivity of antibody-coated liposomes can be implemented for immunoselection protocols. Machy et al. [57] have recently used protein-A con- jugated cytotoxic liposomes to select for cells lacking a specific surface marker which had been tagged with antibody. In the presence of anti-H2K k IgG RDM-4 cell mu- tants which no longer expressed the H-2K k determinant were not sensitive to pro- tein-A-coated liposomes containing MTX whereas the wild-type cells were elimi- nated within 48 h. Some of the mutant cells could be induced to augment their expression of H-2K k in the presence of interferon-y [57]. Therefore, the use of in- terferon-y/ could improve the efficacy of liposomes targeted to interferon-sensitive cell-surface molecules, although interferon-insensitive mutants which lack the tar- get molecule would not be affected and could continue development.

A novel targeting strategy employs conjugation of antigen to the liposome sur- face for targeting to sensitized lymphocytes [96]. A palmitylated nanopeptide of myelin basic protein was incorporated into liposomes containing MTX and incu- bated in vitro with T-lymphocytes obtained from guinea pigs sensitized to the an- tigen. Targeted cells were significantly more susceptible to the drug than nontar- geted cells.

One of the goals of targeted drug delivery is to develop a system which mini-

Page 20: Antibody-directed liposomes as drug-delivery vehicles

362 S. WRIGHT AND L. HUANG

mizes the toxicity of a drug to nontargeted host tissue. When using liposomes, host toxicity could arise from leakage of drug from the liposome during transit to its target. One way of overcoming this problem other than enhancing the serum sta- bility of the liposome is to encapsulate an agent which would be nontoxic to cells even if it did leak out. Heath and co-workers introduced the use of so-called li- posome-dependent cytotoxic agents in 1983 [97]. These drugs are thus classified because they are very poorly permeable and must be delivered into the cytosol by indirect routes, presumably via endocytosis. Methotrexate-~-asparate (MTX-~-asp) is an example of a liposome-dependent drug. MTX is a folate analog which is rap- idly taken up by cells by an energy-dependent facilitated transport mechanism [98]. MTX-~/-asp is a derivative of MTX which is equipotent in its inhibition of dihy- drofolate reductase but has an influx Km of at least 100-fold greater than that of the parent compound [99]. As a result of its low permeability, MTX-~/-asp in its free form is 200-fold less toxic to L1210 cells than MTX. Liposomes containing MTX-~-asp were targeted to L-929 cells with anti-H2K k antibodies producing a 6-12-fold enhancement of binding. The cytotoxicity of MTX-~-asp was increased 10-fold (IC5o -- 0.066 ~M) over the free drug (IC50 = 0.66 ~M) when encapsulated in the appropriately targeted liposome. Further, the cytotoxic effects were inhib- ited by antibody-coated empty vesicles or NH4C1, suggesting internalization via an endocytic route. Even more pronounced effects of targeting with this drug are ob- served if the liposomes are conjugated with protein A rather than with antibody directly [100]. In addition, for antibody-pretreated AKR/J SL2 cells the IC5o for MTX-~-asp encapsulated in protein A liposomes was 9-fold lower than for lipo- somes conjugated with goat anti-mouse immunoglobulin. For antibody-directed protein A liposomes, cytotoxicity was proportional to the antibody concentration and incubation time. lgG 1 was about 15-fold less effective than the other three murine isotypes at mediating protein A liposome delivery. Although it could take up to 4-8 h for liposome binding to plateau, up to one-third of the cell-associated liposomes could be internalized after 2 h at 37°C [100]. Internalization could be followed by removing any surface-bound liposomes by acidification to pH 3 and quantitating the remaining liposome markers. Cytotoxicity was proportional to the amount internalized and only subsaturating amounts of liposomes were needed.

Another liposome-dependent cytotoxic agent recently introduced by Heath et al. [101] is 5-fluoroorotate. This drug is a derivative of 5-fluoruracil and has no known transport pathway. Drug encapsulated in large, negatively charged lipo- somes was demonstrated to be 14-35-fold more cytotoxic to L929 or CV1-P cells than free drug [101].

A novel type of cytocidal immunoliposome, the phototoxic liposome, has re- cently been developed which has the advantage of being selectively activated sub- sequent to binding [102]. Phototoxic LUV's were prepared by reverse-phase eva- porization of a PC/cholesterol mix containing 2.5 mol% of 3-palmitoyl-2-[1- pyrenedecanoyl]-L-alpha-PC and a small amount of MPB-PE for antibody conju- gation. Human T lymphocytes treated with the phototoxic liposomes targeted to a surface protein lost their ability to proliferate in response to phytohemagglu- tinin. The primary advantage of this system is that the liposomes need not be en-

Page 21: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 363

docytosed although they believe that endocytosis and subsequent damage to ly- sosomes may contribute to their mode of action. Cytotoxicity can be selective because the phototoxic drug is only active at the site of irradiation.

IV. Controlled release liposomes

Controlled release liposomes refer to those liposomes which are designed to re- lease entrapped aqueous agents in a controlled manner through environmental manipulations. Controlled release hposomes are a type of special function lipo- some which can be used for localized drug delivery even in the absence of ligand- mediated targeting. The concept of controlled drug release from liposomes implies that delivery can be directly manipulated by the design of the mechanism mediat- ing release. Examples of this include liposomes which release upon encountering an area which has been locally heated or those which release after entering a mildly acidic environment. Although either type of selective release occurs independent of direct targeting, these liposomes which can actually be triggered to release their aqueous contents under certain environmental conditions represent a type of phys- ical targeting of liposome. One such type of physical targeting is found with pH- sensitive liposomes. These liposomes are constructed such that when certain mem- brane components become protonated under low pH conditions the membrane structure is altered resulting in increased permeability and/or enhanced fusogenic activity. A similar system which responds to heat rather than pH are the temper- ature-sensitive liposomes and these will be discussed in a later section.

IV.1. Acid-sensitive liposomes

All of the initial studies and much of the recent work with antibody-targeted li- posomes have been based upon liposomes composed of PC or PC/cholesterol. These compositions are frequently utilized primarily because of their high degree of sta- bility, particularly in the presence of serum components. However, this liposome composition may not be the best choice for efficient release of entrapped drugs upon arriving at their target. It is now well established that endocytosis is the pri- mary mechanism of liposome internalization but it is not known at which point subsequent to the initial uptake that the liposomes are able to deliver their con- tents to the cytosol. As the endocytic vesicle containing the membrane-bound li- posome pinches off from the plasma membrane it now assumes the role of a tar- geted carrier itself which is destined for the lysosomes. However, soon after departing from the cell surface, the endosome or transport vesicle starts to undergo many changes which will continue until it eventually fuses with the lysosome where its remaining contents can be completely hydrolyzed. One of the early events of this transition is an acidification of the endosome interior, reaching a pH of ap- prox. 5.0--6.5 [103,104]. The lysosomal pH is even lower and many drugs such as cytosine arabinoside may be acid labile [105] and if delivered all the way to the lysosomes are frequently inactivated under its degradative conditions [74]. There- fore, release of the drug out of the liposome and endosome from a pre-lysosomal

Page 22: Antibody-directed liposomes as drug-delivery vehicles

364 S. WRIGHT AND L. HUANG

site is necessary for the efficient delivery of this type of drug. Liposomes which are pH sensitive have been developed to meet this need for selective release.

Liposomes made pH-sensitive were first described by Yatvin et al. in 1980 [106]. The pH-dependent trigger which was used was the N-acylamino acid palmitoyl homocysteine (PHC). When PHC was incorporated into PC liposomes with en- capsulated calcein, an isothermal pH-induced leakage of the dye could be dem- onstrated. The underlying mechanism was originally thought to be thiolactone ring formation by homocysteine which is known to occur at low pH. However, sub- sequent studies suggested that electrostatic headgroup interactions or lateral phase separations of the components were more likely involved [107,108]. Headgroup composition, acyl chain length, and the amount of acylamino acid incorporated were all important for the pH sensitivity [108]. Internalization of these PC-based liposomes into acidic compartments should facilitate leakage and release of con- tents into the vacuole. However, for most applications the drug must then escape to the cytosol to reach its target. Some drugs are more likely to penetrate through the endosomal membrane than others. For example, weakly anionic drugs such as MTX could leak more rapidly when located in acidic environment because the drug would have a higher permeability when neutralized by protons. Drugs which are less permeable could remain sequestered inside the endosome/lysosome unless there is an alternative mechanism for release.

Fusion of the liposome with the endosomal membrane would mediate direct re- lease of vesicle contents into the cytosol. Although many studies using PC-based liposomes have demonstrated the efficacy of drug delivery with liposomes and im- munoliposomes, superior delivery might be achieved by using liposome composi- tions which are more prone than PC to undergo fusion with endosomal mem- branes at reduced pH. Generation of fusion-competent liposomes which were pH- sensitive was achieved by incorporating a titratable component into liposomes composed of PE rather than PC. Unsaturated PE does not form stable bilayer ves- icles at neutral pH [109-111] but rather assumes the hexagonal (HII) phase. How- ever, PE can be induced to form bilayers by the addition of molecules such as phospholipids [112,113}, fatty acids [114-116], acylated amino acids [107,117,118], or cholesterol derivatives [118-126]. There are many other stabilizing molecules available (proteins and other phospholipids) but the above stabilizing molecules are of particular importance for pH-sensitive liposomes because all are weakly an- ionic at neutral pH but upon entry into an acidic environment they become neu- tralized and lose their bilayer stabilizing ability. When these PE liposomes become physically unstable they start to leak and/or become fusion competent due to the phase transition towards the H~I phase. In studies utilizing cholesteryl hemisuccin- ate (CHEMS) for membrane stabilization, Szoka and co-workers [119,121] dem- onstrated that PE/CHEMS liposomes aggregate and then leak in a lipid concen- tration-dependent manner in response to acidification to pH 5 and below.

The ability of protein-free liposomes composed of DOPE and at least 20 mol% PHC to undergo rapid pH-dependent fusion was first demonstrated by Connor et al. [107]. Using electron microscopy, gel filtration and resonance energy transfer (RET), these authors observed apparent fusion between SUV's within a pH range

Page 23: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 365

similar to that found in the endosome (pH <6.5). In addition, these liposomes (PE/PHC = 8:2) were very permeable to calcein under the same conditions. How- ever, addition of 40 mol% cholesterol significantly reduced the acid-induced leak- age with minimal effects on fusion [107]. Others have shown that addition of cho- lesterol to PE-containing liposomes can actually increase its fusion capacity [122]. The results with PHC-stabilized liposomes are in contrast to pH-sensitive lipo- somes with oleic acid (OA) as the stabilizer which were found to be quite leaky under similar conditions. Liposomes composed of DOPE/chol/OA (4:4:2) leaked more than 85% of entrapped calcgln within 5 min at pH 5.5 [114]. Liposomes composed of PE prepared by transphosphatidylation of egg PC (TPE) and OA (7:3 mole ratio) have been shown to aggregate, become destabilized, and fuse below pH 6.5 [115]. These acid-sensitive liposomes (DOPE/OA, 7:3) have been shown to mediate cytoplasmic delivery of calcein. As evidenced by diffuse intracellular fluorescence [123] up to 30 mol% cholesterol could be incorporated into the li- posome without inhibiting cytoplasmic delivery. PC/OA liposomes were stable at pH 4-8 and were unable to achieve cytoplasmic delivery of the dye. Acid respon- sive liposomes which are fusogenic can also be prepared with bioactive lipids such as arachidonic acid and 1-oleoyl-2-acetylglycerol [124]. Liposomes composed of mixed chain egg PE/OA (7:3) became destabilized when the pH was lowered to less than 6.5 or upon addition of calcium [115]. Characteristics of destabilization included rapid proton-induced aggregation of liposomes coincident with release of internal aqueous contents and membrane fusion.

Fusion between two liposomes involves bilayer-bilayer contact followed by con- solidation of the two opposed membranes into a continuous bilayer which involves mixing of lipid components and intermixing of the aqueous contents from the two liposomes. The initial contact between liposomes leads to aggregation into larger structures which can be followed by light scattering or turbidity measurements. The two aspects of actual fusion can be monitored with the use of aqueous or lipid markers which are fluorescently labeled. Intermixing of separately entrapped aqueous markers 1-aminonaphthalane-3,6,8-trisulfonic acid (ANTS) and N,N'-p- xylylenebis (pyridinium bromide) (DPX) results in quenching of the fluorescent signal from ANTS by DPX upon vesicle fusion [125]. A leakage assay can also be developed with these markers by following the dequenching of ANTS by DPX when they leak out from liposomes from which they have been coencapsulated [115,119]. For the RET lipid mixing assay a donor/acceptor pair of labeled lipids 7-nitro-2,1,3- benzoxadiazol-4-yl-PE (NBD-PE, acceptor) and No(lissamine rhodamine B sul- fonyl)-PE (Rh-PE, donor) are each incorporated into one population of liposomes such that there is significant NBD-PE quenching by Rh-PE. When fused with an unlabeled population the probes are diluted and dequenched resulting in fluores- cence signal enhancement [126]. Development of these novel biophysical tech- niques have been very important in elucidating the probable mechanism of action of this type of liposome.

An alternate type of pH-sensitive liposome has recently been described in which the destabilization characteristics apparently do not involve membrane fusion. Li- posomes composed of N-succinyldioleoyl-PE and DOPE (3:7) start to leak at pH

Page 24: Antibody-directed liposomes as drug-delivery vehicles

366 S. WRIGHT AND L. HUANG

5 and below but do not fuse [112]. Release was believed to result from bilayer des- tabilization induced by pH-dependent packing defects of the lipids and electro- static interactions between the stabilizing molecules. The mechanism of delivery by pH-sensitive liposomes is dependent upon the phase behavior of the liposome which in turn is based on the liposome composition. Addition of targeting mole- cules to the bilayer may alter its phase dynamics and thus its ability to encapsulate, retain and specifically release its contents.

However, a recent report describing the preparation of biotin-bearing pH-sen- sitive liposomes suggests that these liposomes can also be effectively targeted while retaining both their contents and phase behavior [127]. Incorporation of less than 1 mol% biotinaminocaproyl-PE into PE/cholesterol/OA (7:3:3) liposomes did not significantly change the pH-dependent leakage profile. These liposomes could be agglutinated with acetylated avidin and bound with very high affinity to avidin- coated microtitre plates.

Liposomes sensitive to pH can also be conjugated with acylated antibody with- out altering their permeability response to acidification [116], although calcium- induced fusion activity was reduced by antibody incorporation [128]. However, targeting of calcein-containing pH-sensitive liposomes (PE/PHC, 8:2) with acy- lated anti-H-2K k IgG resulted in delivery of the dye into the cytoplasm of L-929 ceils [117]. Dye was dispersed throughout the cell when given with pH-sensitive immunoliposomes but showed only punctate patterns for pH-insensitive immu- noliposomes (PC based). A cytoplasmic calcein concentration of 50 ixM was de- termined using a microscope-associated photometer. This concentration repre- sented dye transfer from approx. 1000 liposomes per cell, indicating a very efficient delivery from approx. 50% of the internalized liposomes.

The therapeutic potential of antibody-conjugated acid-sensitive liposomes has been demonstrated in vitro by the efficient target-specific delivery of antitumor drugs and cytotoxins. Connor and Huang [118] selectively delivered MTX or ara- C to L-929 cells with anti-H-2K k targeted liposomes (56 IgG per 130 nm liposome) composed of DOPE/OA (8:2). Moreover, targeted pH-sensitive liposome delivery was more efficient at cell killing than either that of the free drugs or targeted PC liposomes. Pretreatment with chloroquine or NH4C1, each a weak base which neu- tralizes acidic organelles, inhibited cytoplasmic delivery of calcein [117] or ara-C [118] by the immunoliposomes, indicating liposome uptake into base-sensitive or- ganelles, presumably endocytic vesicles.

A tryptic fragment of diphtheria toxin, fragment A (DTA), is a potent inhibitor of protein synthesis. DTA is normally impermeable to cells because it lacks the fragment-mediating cell binding and entry and therefore is not cytotoxic as a free protein. However, upon gaining entry into the cell interior elongation factor 2 is selectively inactivated by a reaction involving the adenosine diphosphate (ADP)- ribosyl transferase activity of the catalytic A fragment leading to cell death [129,130]. It has been determined that the effects are stoichiometric, i.e., only one toxin molecule is sufficient to kill the cell [131]. The characteristics of low perme- ability and high lethality make DTA a natural candidate for delivery with lipo- somes. In early studies using proteoliposomes containing Sendai glycoproteins,

Page 25: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 367

Okada and co-workers [131,132] demonstrated enhanced cytotoxicity of DTA, presumably as the result of direct liposome-cell fusion. Using the fusogenic pH- sensitive immunoliposomes Collins and Huang [116] have successfully delivered the DTA to murine cells which are normally toxin-resistant. DTA was encapsulated into DOPE/OA (8:2) vesicles by a modified dehydration/rehydration procedure [133]. Acid-sensitive liposomes conjugated with anti-H-2K k but not non-targeted or targeted pH-insensitive liposomes were able to deliver DTA specifically to L- 929 cells as determined by inhibition of 3H-leucine incorporation. The effects could be blocked by pretreating the cells with NH4C1 or chloroquine.

The potential of gene therapy that has been realized in the past few years [134] has led to intensive efforts in the development of techniques for the efficient de- livery of DNA into cells. The currently available methods such as microinjection, electroporation, chromosome-mediated insertion, microcell-mediated gene trans- fer, DNA-mediated gene transfer, infection with recombinant RNA or DNA vi- ruses, spheroblast fusion, and cell fusion are considered inefficient and relatively non-specific. Moreover, none of these methods are acceptable for in vivo trans- fection. Liposomes have also been utilized to introduce nucleic acids into several different cell types, although with minimal benefits compared to conventional methods. Effective liposome-mediated transfer has been reported into bacteria [135,136], fungal [137], and animal cells [138-147]. Transfer is frequently initiated by the addition of a membrane fusogen such as DMSO or PEG. The established ability to introduce drugs or proteins into cells with pH-sensitive immunolipo- somes suggests that this system may also be applicable for the specific delivery of other macromolecules such as plasmids via similar mechanisms. Using such a sys- tem, Wang and Huang [145-147] have demonstrated efficient delivery and expres- sion of exogenous DNA to targeted cells both in vitro and in vivo. A plasmid (pPCTK-6A) containing the herpes simplex virus thymidine kinase gene (HSV-TK) was encapsulated in acid-sensitive liposomes (DOPE/cholesterol/OA, 4:4:2 mole ratio) targeted with acylated IgG against the routine H-2K k histocompatibility complex [145]. Liposomes prepared by detergent dialysis with a mean diameter of 290 nm entrapped approx. 20% of the added DNA, corresponding to about 3-5 8.2 kb plasmids per liposome. So that the expression of this gene could be regu- lated following incorporation into the host genome, the endogenous promoter was removed and replaced with a promoter containing a cAMP regulatory region [148]. Therefore, by adjusting the cAMP levels the introduced promoter from a rat phosphoenolpyruvate carboxykinase (PEP-CK) gene would turn on or off the transcription of HSV-TK. Since all mammalian cells normally have the salvage en- zyme thymidine kinase, tk- mutant mouse L cells were used as the target cells so that gene expression could be followed by monitoring regulated TK activity. An- tibody-targeted liposomes which were pH-sensitive were about 8-fold more effec- tive at restoring thymidine kinase activity to the Ltk- cells than pH-insensitive im- munoliposomes [145]. The presence of antibody increased the effectiveness of the pH-sensitive liposomes 6-fold compared to those not targeted. Expression of en- zyme activity was under the control of cAMP indicating that TK activity was the result of transformation and not reversion of the mutant. When long term trans-

Page 26: Antibody-directed liposomes as drug-delivery vehicles

368 S. W RI GHT AND L. H U A N G

formation by pH-sensitive immunoliposomes was compared with that by a tradi- tional method such as calcium phosphate transfection, the liposomes were found to produce a transformation efficiency of 9% vs. 1% for the salt precipitation method. Liposome administration was also much less toxic to the cells than in- cubation with high concentrations of calcium phosphate. In addition, DNA blot hybridization studies of long-term transformants indicated that the method of de- livery may dictate at which site the plasmid DNA integrates into the host genome [145]. Efficient transfection by pH-sensitive immunoliposomes was also observed if DNA was simply adsorbed to the surface of preformed liposomes rather than encapsulated within during liposome formation [147]. Induced kinase activity was elevated 30-fold for entrapped and 20-fold for adsorbed compared to free DNA. Efficient liposome-mediated transfection with adsorbed DNA has recently been described by others [149]. Unless cells susceptible to passive targeting are the tar- gets, then antibody conjugation should further increase the delivery efficiency.

Huang and co-workers have also found that targeted pH-sensitive liposomes could be used to deliver DNA to lymphoma cells transplanted to the peritoneal cavity of nude mice [146]. A 4.6 kb plasmid (pBB0.6-CAT) containing the Esch- erichia coli chloramphenicol acetyltransferase (CAT) gene under the control of the PEP-CK promoter used previously was encapsulated in pH-sensitive immunoli- posomes. RDM-4 lymphoma cells (H-2K k positive) which had been injected i.p. into immunodeficient mice were able to take up about 20% of an i.p. injected dose of immunoliposomes. When activated with 8-bromo-cAMP and 3-isobutyl-l-meth- ylxanthine, CAT-specific activity in these cells transformed with the liposomes was 12-fold higher than that in the liver and 5-fold higher than that detected in the spleen. No activity was found in peritoneal macrophages although these cells ac- cumulated a significant amount of liposomal lipid. PC-based immunoliposomes (pH- insensitive) produced only one-fourth the activity in target cells that pH-sensitive vesicles could. These results have important implications for gene therapy and cancer therapy for they demonstrate that genetic material can be effectively deliv- ered in vivo to targeted cells with little incorporation into the genome of nontar- geted host cells. The ability to deliver DNA to cells in vivo is the greatest advan- tage of using liposomes over other methods.

Mechanistically, the premise that pH-sensitive liposomes fuse with the endo- some membrane and release their contents directly into the cytosol is most strongly supported by the studies demonstrating intracellular delivery of macromolecules which are too large to permeate across intact membrane [116,145]. Alternatively, simultaneous endosome and liposome membrane rupture is conceivable which leads to dispersal of entrapped contents into the cytosol. This mechanism should also proceed with empty pH-sensitive liposomes, yet no cytotoxicity is observed with these liposomes as one might expect following rupture of the acidic endosomes. However, the ability to deliver DNA adsorbed to the surface of pH-sensitive im- munoliposomes [147] suggests that this mechanism is possible.

The rate and extent of endocytosis and cellular processing including sorting and recycling of internalized targeted liposomes has not been examined. Gruenberg and Howell [150] have recently reported that an internalized transmembrane protein

Page 27: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 369

(G protein of VSV) resides for less than 5 min in a fusion-competent endosome. The fusion-competent compartment was determined to be the early endosome where sorting of the protein for recycling or degradation occurs, which, for the G protein, was 50% in either direction. Visualization of endosomes reveal that they move bidirectionally along linear tracks of microtubules by discontinuous salta- tians until they cluster along with lysosomes in the perinuclear region of the mi- crotubule-organizing center [151].

The mechanism and dynamics of PE-phase behavior and the role of Hn for- mation in bilayer destabilization has been examined in detail by Szoka and co- workers [119,120,152,153] and recently reviewed by Siegel [154]. It was observed that pH-dependent liposome destabilization and leakage was mediated by lipo- some aggregation [119,120]. Moreover, they reported that destabilization is greater between two different kinds of liposomes than between pure pairs [120] which may have relevance for interaction between liposomal and cellular membranes.

IV.2. Temperature-sensitive immunoliposomes

Other schemes have been developed which also rely on the dynamics of lipid phase behavior and are of particular use for cells with inherently low endocytic activity and thus inappropriate candidates for pH-sensitive liposomes. Utilization of temperature-sensitive liposomes was one of the first attempts at physical tar- geting of liposomes. Taking advantage of the well-known observation that the permeability of liposomes is highest at the temperature at which they undergo a phase change from gel to liquid-crystalline [155], Yatvin and co-workers [156] ex- amined the ability of liposomes to release entrapped dye contents as a function of temperature. DPPC was chosen for these studies because it has a phase-transition temperature (41°C) which can be reached with only mild hyperthermia to the an- imal. Multilamellar vesicles of DPPC leaked 85% of drug when heated near their phase-transition temperature in the presence of serum. The transition temperature is also sensitive to the number of lamellae in the liposome, the presence of serum, and the addition of any other lipids or hydrophobic components [157]. For ex- ample, the behavior of this system in the presence of amphipathic drugs will be drug-dependent because of the potential effects of membrane-interacting drugs on the phase behavior of the liposomes [158,159]. The drug delivery potential of this system was initially ascertained by the enhanced ability to kill E. coli with neo- mycin encapsulated in DPPC/DSPC (3:1) vesicles when heated to 41°C [156]. Ma- gin and Weinstein and co-workers [157,160,161] have demonstrated the ability of heat-sensitive liposomes to deliver MTX to tumor cells following local hyper- thermia. This therapy is somewhat selective in that only the liposomes in the lo- cally heated area release their drugs. The effectiveness of heat-sensitive liposomes can be enhanced by targeting with antibody which will bring the liposome into di- rect contact with the cell surface prior to release of its contents. Heat-sensitive im- munoliposomes were prepared by Sullivan and Huang [70] by injection of palmitic acid-derivatized IgG into a suspension of DPPC near its phase-transition temper- ature. Although incorporation of antibody changed the temperature at which dye

Page 28: Antibody-directed liposomes as drug-delivery vehicles

370 S. WRIGHT AND L. HUANG

Fig. 2. Proposed mechanism of release from heat-sensitive immunoliposomes. Phase boundaries formed by the co-existence of gel and fluid-phase lipids at the phase-transition temperature facilitates leakage

of soluble drugs from liposomes bound to the cell surface.

release became maximal in buffer, in the presence of 5% serum the release tem- perature was the same (40°C) for bare or conjugated liposomes. Moreover, im- munoliposomes which were bound to target cells retained their ability to release entrapped contents upon heating [70,162]. Using this protocol they demonstrated that 3H-uridine encapsulated in heat-sensitive immunoliposomes could be taken up more efficiently by the target cells than free nucleoside [162]. Since uptake could be mostly eliminated with nucleoside transport inhibitors the mechanism of deliv- ery was presumed to involve surface release from bound liposomes producing a locally high concentration of substrate which was then taken up by the normal transport routes (see Fig. 2).

IV.3. Target-sensitive immunoliposomes

Target-sensitive immunoliposomes as described by Ho et al. [72] represent a composite of pH-sensitive and heat-sensitive liposomes. These liposomes are con- structed much like their pH-sensitive counterparts in that they are based on PE, yet they have no stabilizers other than the targeting molecule, such as palmitoyl IgG. However, like the heat-sensitive liposomes, the site of delivery is at the cell surface. Thus, therapeutically, these liposomes were also designed for drug deliv- ery to cells with either generally low endocytic activity or poorly internalized sur-

Page 29: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 371

face targets. Although they have similar potential targets the suggested mecha- nism of release for this system is quite novel and different from those which are temperature sensitive. One of the initial observations important for the develop- ment of this system was by Taraschi et al. [163] who reported that PE liposomes stabilized by incorporated glycoproteins could be destabilized and induced back into their native non-bilayer structures (Hit) following their binding to lectins. Other amphipathic molecules such as the lipid hapten DNP-cap-PE has also been found to stabilize D O P E when incorporated at a level of greater than 12 mol% [164]. If these antigenic liposomes were loaded with quenched calcein and added to im- mobilized anti-DNP antibodies they leaked in an antibody concentration-depend- ent manner [164]. The mechanism of release was presumed to be multivalent bind- ing between liposome and antibody leading to contact capping of the stabilizer at the binding site. This redistribution of the stabilizing hapten is believed to result in reorganization of the membrane lipids toward the H n phase which produced highly permeable areas where the stabilizer had vacated (see Fig. 3). Similar find- ings were obtained with glycophorin-stabilized PE liposomes which could be dis- rupted when treated with trypsin [165]. It is of interest that glycophorin can sta- bilize D O P E whereas if added to DOPC it increases bilayer permeability [166].

Antibody-dependent target-sensitive liposomes were introduced when it was found that under certain well-defined conditions fatty acid-derivatized IgG could itself stabilize D O P E [72]. Liposomes composed of PE and acylated IgG against the gD envelope protein of HSV loaded with calcein exhibited target-induced lysis

o o

Fig. 3. Proposed mechanism of release from target-sensitive immunoliposomes. Subsequent to bind- ing to surface antigens the stabilizing acylated antibodies undergo contact capping which leads to lipid instability in the vacated regions. The PE-based liposome then is susceptible to contact-dependent des- tabilization which pushes the bilayer toward the hexagonal equilibrium phase which is characterized by rapid leakage from intermediate states. Drugs released from this pericellular site at locally high con-

centrations are then taken up by cellular transport systems.

Page 30: Antibody-directed liposomes as drug-delivery vehicles

372 S. WRIGHT AND L. HUANG

when incubated with HSV [167] or HSV-infected L cells [72]. In addition, these liposomes were able to deliver specifically ara-C or acyclovir to virus-infected cells resulting in enhanced antiviral activity compared to the free drug [168]. As before, the mechanism was thought to involve multivalent binding followed by pericellular lysis. Uptake of surface-released drug was apparently through the nucleoside car- rier and could be effectively blocked by specific nucleoside-transport inhibitors. Kinetic and ultrastructural studies implied that collision among bound liposomes was essential for liposome destabilization [169]. This system also has potential for the development of liposome-based homogenous immunoassays [170-172].

V. Animal studies with immunoliposomes

One of the first obstacles encountered with liposome administration in vivo is stability of the particle in blood. Successful drug delivery with immunoliposomes requires that the drug remains with its carrier until it reaches the target site, at which point it should dissociate from its carrier so that it may then unite with the effector molecule which is the ultimate target. There are a variety of interactions between liposome and blood components which can disrupt the integrity of the li- posome resulting in leakage and premature loss of the drug to be delivered. Thor- ough reviews on this topic have been presented by Scherphof et al. [173], Juliano et al. [174], and Bonte and Juliano [175].

The physical properties and ultimate fate of blood-borne liposomes can be in- fluenced by interactions with serum components such as albumin, immunoglobu- lins, clotting factors, fibronectin, and C-reactive protein [176,177]. However, the most prominent means of liposome destabilization in blood is interaction with serum lipoproteins. Scherphof and co-workers [178] have shown that the phospholipid constituents of liposomes can actually be extracted by high-density lipoproteins re- suiting in leakage of entrapped contents. The extent of leakage depends upon the HDL/phospholipid ratio [173,177]. Although the mechanism of interaction is still unresolved, it appears that phospholipid transfer to HDL or an HDL-like particle is multi-phasic with at least one saturable component. In addition, a small amount of phospholipid transfer out of liposomes can also be mediated by low and/or very low density lipoproteins [177].

It is evident that the ability of serum proteins to interact with the liposome is heavily dependent upon the bilayer physical state. What effects the addition of an- tibody has on the physical properties of the liposome depends upon the method of conjugation. Due to packing considerations, coupling to liposomal lipids might not be expected to be as perturbing as the incorporation of multiacylated antibod- ies. Liposomes with protein coupled to glycolipids by the periodate procedure were just as stable (nonleaky) in buffer as liposomes treated with periodate alone [41]. In contrast, incorporation of peptides modified with palmitic acid into liposomes results in the alteration of several bilayer properties [179]. Sullivan and Huang [70] observed that incorporation of palmitoyl-IgG into DPPC altered the rate and ex- tent of heat-induced carboxyfluorescein release. When heated in buffer, the tem- perature for maximal rate of dye release was lowered and the total amount of dye

Page 31: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 373

released was decreased from 80 to 45%. In the presence of 5% serum the release temperature was now the same for both free and antibody-coated liposome al- though the extent of dye release was still lower for the latter group. Liposomes with Fab' fragments attached via DTP-PE [45] or MPB-PE [49] are relatively non- leaky in the presence of serum. The mechanism of serum-induced bilayer disrup- tion for antibody-conjugated liposomes has not been examined in any detail. It is expected that the lipid components of targeted liposomes are susceptible to the same attack by serum proteins that the antibody-free liposomes are. However, the presence of the foreign antibodies on the surface of the liposome increases its an- tigenicity and thus becomes more susceptible to complement-mediated lysis [180]. Furthermore, antibody attached to liposomes could fix complement itself and bring about a complement-mediated lysis [180].

The interactions between liposomes and serum components are governed to a large extent by the size and composition of the liposome. Inclusion of 30-50 mol% cholesterol in the liposome enhances its stability in serum and is now routinely in- cluded in most liposome preparations to be used in the presence of serum [181]. Cholesterol increases liposomal stability by increasing the lipid packing density which eliminates any phase boundaries (and phase transitions) and thus decreases potential interactions with serum lipoproteins. Glycolipids can reduce plasma-in- duced leakage in proportion to their sialic acid content [182]. The effects of adding both cholesterol and gangliosides are synergistic with in vitro half-lives in serum of up to 24 h obtainable for SUV's at 37°C (PC/chol/Gx= 10:5:1) [180]. Even greater serum stability has been reported for sphingomyelin/cholesterol mixtures [176]. Incorporation of PS [182], sulfatides [182], or sphingomyelin [183], altering phospholipid structure [184], or utilizing polymerizable lipids [176,185] can also enhance serum stability. Allen and Chonn [186] have recently demonstrated that LUV composed of ganglioside GM 1 and sphingomyelin had significantly reduced uptake by RES components in vivo. In addition, LUV's retain their contents longer than SUV's in the presence of plasma [182] although others have reported that SUV's exhibit much longer half lives in blood than MLV's [187,188]. The small radius of curvature of SUV's influences bilayer packing and thus susceptibility to disintegration by serum proteins.

Fatty acid-stabilized pH-sensitive immunoliposomes (PE/OA) are unstable in serum where they will spontaneously aggregate and release entrapped calcein [189,190]. Fatty acids may not be an appropriate choice as a PE-stabilizer for in vivo studies, since it can be removed from the liposome by serum components [113]. For example, oleic acid is known readily to partition out of liposomes in the pres- ence of albumin [191]. Albumin is known to induce leakage of inulin from PC li- posomes [192] and it can also rapidly destabilize PE/OA liposomes (Collins, D. and Huang, L., unpublished observations). However, the pH-sensitive liposomes are apparently stable in vivo if cholesterol is also incorporated [146]. Rather than adding a third component such as cholesterol to enhance serum stability, substi- tution with a different pH trigger such as a double-chain amphiphile [112,193] can also be used for the preparation of serum stable acid-sensitive liposomes [193].

Although serum stability is an important parameter for liposome carriers, this

Page 32: Antibody-directed liposomes as drug-delivery vehicles

374 s. WRIGHT AND L. HUANG

does not necessarily mean that more stable liposomes are more effective carriers in general. For some applications where sustained blood levels of drug is the goal, then slow controlled release from the liposome is desired. In fact, liposomes can act as small continuous infusion devices because encapsulated drugs generally have a longer plasma half-life than the free drugs [189]. A slow release mechanism would be particularly beneficial for the delivery of cell cycle-dependent drugs and where a reduced frequency of administration is desired. The ability to anchor the lipo- some by antibody targeting can effectively prolong the drug effects at the target site. Slow release of acyclovir from liposomes targeted with antibody to herpes vi- rus-infected corneal cells in culture has been shown to be an effective means of inhibiting virus replication [194,195] although its effectiveness as a topical treat- ment in vivo has yet to be demonstrated.

Liposome encapsulation has previously been found to be a means by which to reduce some of the major side effects of potent chemotherapeutic agents. The dose- limiting cardiotoxicity of adriamycin can be attenuated by entrapment in lipo- somes of cardiolipin [196,197] or other anionic phospholipids [198-200] resulting in an overall enhancement of its therapeutic index. Encapsulation of the very toxic anti-fungal agent amphotericin B increases its therapeutic index [201] apparently by mediating selective drug transfer to fungal cells [202]. In spite of the many promising in vitro studies demonstrating the efficacy of liposomes as drug delivery devices, the widely anticipated benefits of liposome use in vivo have not, in gen- eral, been realized. There are of course exceptions such as for the successful treat- ment of Leishmaniasis with liposome encapsulated antimonials [203,204] and the delivery of amphotericin B for systemic fungal infections [201,205-207]. Although the selectivity of treatment with these liposome-encapsulated drugs is high, at- tempts at further enhancing their therapeutic index by surface targeting has not been reported.

There have been very few reports on the in vivo use of immunoliposomes for delivery of chemotherapeutic drugs. This apparently stems from the problem of compartmentalization. In general, the distribution of liposomes is restricted to the compartment in which they are administered. For example, to extravasate from the circulation, liposomes injected intravenously must be transported through or around capillary endothelial cells where they will then usually encounter the basal lamina as a second barrier [4,207]. Only particles much smaller than limit vesicles are capable of escaping out of the circulation through sinus discontinuities be- tween the endothelial cells [208,209]. However, in the liver and spleen SUV's are able to penetrate the relatively large sinusoids (no basement membrane) and ac- cumulate in both parenchymal and non-parenchymal cells. These openings into the space of Wisse [210] are about 100 nm so that there is a size restriction here as elsewhere because MLV's are taken up less readily then SUV's [194]. Extravasa- tion elsewhere should be possible, since leukocytes are known to cross unruptured capillaries through the ameboid movement involved in diapedesis. The apparent inability of liposomes to escape the circulation is a major impediment for their use against extravascular targets such as solid tumors. Effort must now be focused on targetable sites confined to the cardiovascular system for which targeted tiposomes

Page 33: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 375

may be of benefit as therapeutic or diagnostic agents. Alternate routes of administration have not been examined so closely as the in-

travenous approach. Liposomes injected s.c. are rapidly drained to the regional lymph node where, depending upon size, they may enter into the circulation [211]. Liposomes injected into the subarachnoid space have been shown to prolong the drug retention time significantly with no display of undesirable toxic effects (Huang, L., unpublished results). Intraperitoneal tumors have been a favorite target for in vivo studies of liposome targeting [146,212]. Administration of pH-sensitive im- munoliposomes i.p. to mice carrying RDM-4 lymphoma cells as an ascites tumor results in accumulation of approx. 30% of the injected dose in ascites cells [146]. Targeting increased the uptake by non-adherent ascites cells (primarily RDM-4 cells) 2-fold without altering accumulation by adherent cells. The next largest site of uptake was the stomach with approx. 15%. Injection of either non-targeted pH- sensitive liposomes into tumor-bearing animals or targeted pH-sensitive liposomes into tumor-free animals resulted in significant uptake by the spleen which was even greater than that found in the ascites cells. Antibody targeting increased accu- mulation in the heart 3-5-fold. Liver, lung and kidney each accumulated less than 5% of the injected dose of any liposome type.

One of the first attempts at using antibody-targeted liposomes for anti-tumor therapy in an animal model was reported by Hashimoto et al. [212]. Liposomes were prepared by immobilizing an IgMs fragment against a mouse mammary tu- mor-associated antigen on SUV's containing actinomycin D and were found to be selectively cytotoxic in vitro. Intraperitoneal injection of these chemoimmunoli- posomes into mice with an i.p. transplanted tumor (MM46) resulted in more ef- fective therapy than either free drug or BSA-coated drug-containing liposomes [212]. However, the relevancy of the i.p.-i.p, system as a general in vivo model is questionable and reflects primarily the potential of the vehicle for local therapy. The authors also examined potential systemic effects by i.v. administration of li- posomes targeted against a subcutaneous MM46 tumor [212]. A single i.v. dose of liposomes for treatment of tumors implanted 4 days earlier was found to be mod- erately effective in reducing tumor weights in mice compared to the effects of con- trol liposomes. A more significant tumor reduction was obtained if the mice were pre-dosed with unmodified MLV's prior to the injection of the antibody-conju- gated liposomes. These effects were attributed to specific binding to the tumor cells based upon their in vitro results. However, since extravasation of the targeted li- posomes was unlikely the effects were more probably due to sustained serum lev- els of drug from the liposome. Although blood clearance values for liposomes were not determined, the ability of the animal to clear colloidal carbon from the cir- culation was inhibited by MLV predosing [208]. Circulating macrophages and res- ident macrophages in each organ are apparently the primary cell types responsible for liposome uptake at these sites [213]. The augmented effects on targeted cells that can be achieved with MLV predosing is apparently due to saturation of phag- ocytic cells so that their capacity to engulf the immunoliposomes is impaired. Blockage of RES components by predosing with dextran sulfate [213,214], SUV's [215], REV's [216] and MLV's [217] have been demonstrated effective in altering

Page 34: Antibody-directed liposomes as drug-delivery vehicles

376 S. WRIGHT AND L. HUANG

the circulation time and organ distribution of liposomes. Rather than predosing with lipid for RES blockade Aragnol and Leserman [218]

have evaluated the ability of an anti-Fc receptor antibody to influence the distri- bution of injected liposomes in the presence of accelerated immune elimination. They demonstrated that it is possible in vivo to enhance Fc receptor-mediated up- take of ligand-bearing liposomes by the use of liposome-specific monoclonal an- tibodies or to reduce Fc receptor-mediated uptake with a monoclonal antibody to the Fc receptor. They also evaluated the influence of passive or elicited anti-DNP antibodies on the circulation and tissue distribution of i.v. injected DNP-bearing liposomes. These liposomes had increased uptake to the liver which could be ab- rogated by injection of an anti-Fc receptor antibody.

Gregoriadis et al. [219] have recently examined the ability of anti-Thyl IgG- bearing liposomes to interact with circulating target AKR-A cells which had been injected by the same route. They concluded that antibody-targeted liposomes are cleared more rapidly than bare liposomes from the circulation of mice which had or had not been injected with the target cells. Radioactive and fluorescent markers from antibody-bearing liposomes were cleared from target-bearing animals faster than from the control animals. Approx. 15% of the injected dose interacted with the circulating AKR-A cells. Konno et al. [220] recently examined the therapeutic potential of antibody targeted liposomes against a solid tumor in vivo. Anti-hu- man alpha-fetoprotein (AFP) monoclonal antibody were conjugated to SUV's (via SPDP/MBS) composed of PC/chol/PA and loaded with adriamycin for targeting to AFP-positive human hepatoma cells (Li-7) maintained in nude mouse as xeno- transplanted subcutaneous tumors [222]. Reduction in tumor weight and histo- pathological evidence indicated that the drug-loaded liposomes with immobilized antibody were therapeutically more effective than unconjugated liposomes. Treat- ment was initiated by administration of three i.v. injections of liposomes 10-14 days after inoculation when tumor weight exceeded 100 rag. Tumor (Li-7) reduction with the immunoliposomes was enhanced by targeting with tumor-specific antibodies whereas antibody specificity made no difference for immunoliposomes given against an AFP-negative strain (MX1).

The ability of immunoliposome to reach target cells depends on the extent of RES uptake and accessibility of the target within that compartment. Encapsula- tion of chemotherapeutic agents in targeted liposomes can alter its biodistribution and, depending upon its dose-limiting toxicity, the maximum tolerated dose. For example, ADR dosages of up to 7.5 mg per kg can be tolerated in encapsulated form when administered i.v., since the normal toxic distribution of the drug to the heart is reduced [220]. However, the lower uptake by the heart that was reported was due to the encapsulation and could not be attributable to the presence of an- tibody on the liposome. In fact, the addition of the antibody to the liposome did not significantly alter the drug levels in serum, spleen, lung, kidney, and most no- tably, the tumor. Liver accumulation was found to be initially higher for the tar- geted liposome which lasted up until about 8 h postinjection. In addition, previous studies have indicated that protein-coated liposomes [221] and antibody-coated li- posomes [222] are cleared faster from the circulation to the liver than unconju-

Page 35: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 377

gated liposomes [221,223]. Injection of antibody conjugated liposomes into anti- gen-negative mice circulated for the same period as control liposomes whereas they rapidly disappeared from the plasma in mice expressing the targeted determinant [223]. Debs et al. [222] found that antibody-mediated enhancement of liver uptake resulted in decreased uptake in a number of organs outside the RES. The lipo- somes used were coated with an antibody against a murine lymphoid differentia- tion antigen (Thy 1.1) present in blood, thymus, lymph nodes and spleen of Thy 1.1 mice (AKR-J). Uptake of antibody-coated SUV's by lymph nodes of AKR-J mice was enhanced 7.4-fold by using antigen-specific antibody and uptake at this site was 3 times that in antigen-negative mice (AKR-Cu). Predosing with a large amount of unmodified liposomes prior to i.v. injection of the targeted liposomes reduced liver uptake of the coated liposomes but did not alter uptake by the lymph nodes.

Murine biodistribution of antibody-conjugated pH-sensitive liposomes has been examined by Connor and Huang [189] and was found to be essentially the same as for antibody-free liposomes. Lipos0mes labeled with 3H-cholestanyl ether (lipid marker) and t25I-tyraminyl-inulin (aqueous marker) were injected into the tail vein of antigen-positive (C3H) or antigen negative (Balb.c) mice. Acid-insensitive li- posomes (PC/OA) which were antibody-free or coated with palmitoyl-anti-H-2K k IgG displayed similar clearance rates and organ distribution in either type of mouse. However, in contrast to pH-insensitive liposomes which were taken up primarily by liver and spleen, a significant amount of 3H-CE from pH-sensitive liposomes was found in the lung as well as in the liver. After about 6 h the marker was dis- tributed the same as for the pH-insensitive profile. The presence of antibody on the PE/OA liposomes resulted in a slightly faster liver accumulation which was not antigen-dependent.

VI. Conclusions

(1) There have now been developed a sufficient number and diversity of pro- cedures for targeting of liposomes with antibodies or antibody fragments such that this aspect of the targeted delivery problem is no longer a major concern. More- over, most of these methods are relatively nonperturbing with respect to encap- sulated drugs.

(2) Delivery of anticancer drugs by immunoliposomes to cells in vitro has been shown by several groups to be advantageous in many aspects over simple drug en- capsulation or administration of free drug. Delivery of molecules such as toxins and plasmids is also augmented by immunoliposome encapsulation.

(3) Although binding to the target antigen is a prerequisite, it alone is not suf- ficient for immunoliposome delivery into the cell. The efficiency of drug delivery by antibody-targeted liposomes depends upon the method of antibody targeting, surface density and internalization of the target antigen, and the physical prop- erties of the liposome, including size, charge, and phase behavior.

(4) Special function or controlled release immunoliposomes such as those which are pH-sensitive or temperature-sensitive are now available which can mediate de-

Page 36: Antibody-directed liposomes as drug-delivery vehicles

378 S. W RI GHT AND L. H U A N G

livery either at intracellular or pericellular sites. The mechanism of action of these phase-sensitive, polymorphic liposomes is just beginning to be understood.

(5) Although antibody targeting can apparently enhance delivery in vivo, the same problems of compartmentalization and RES uptake that exist for non-tar- geting liposome carriers also remain for the targeted carriers. However, the sta- bility of immunoliposomes in serum can be controlled by using appropriate lipo- some constituents.

VII. Future perspectives

Perhaps the most significant barrier yet to be surmounted in the field of lipo- somal drug delivery is the rapid scavenging by components of the RES. This prob- lem applies to non-targeted as well as targeted liposomes. For antibody-conju- gated liposomes, RES capture suggests that those liposomes which are available initially to interact with their target and fail to do so on their first pass will prob- ably be engulfed before encountering the target for a second time. Although some progress has been achieved in reducing RES uptake through measures such as li- posome modification or RES saturation by predosing, liposome capture by RES components remains a formidable problem. The recent success in RES avoidance by utilizing a combination of membrane stabilizers and/or size modifications sug- gests the possibility of overcoming this barrier.

A second major obstacle faced by liposome-based drug therapy is anatomical compartmentalization. Even though liposomes can be administered through a va- riety of routes the fact that these subcellular sized structures cannot permeate ep- ithelial and/or endothelial boundaries severely limits their utility particularly for disseminated solid tumors. However, some macromolecular ligands are able to be taken up, delivered to the other side of the cell and released by a process of trans- cytosis. The intriguing idea that liposomes may be capable of crossing some of these boundaries by transcytosis must be vigorously pursued.

The development of novel controlled release liposomes has given new impetus into the area of drug delivery using targeted liposomes. Although using immu- noliposomes which are sensitive to temperature or pH appears to be promising for drug delivery, they are not idealized carriers. For pH-sensitive immunoliposomes in particular much more must be done to elucidate the precise mechanism of in- tracellular delivery so that the system may be optimized as a delivery vehicle. The ability of antibody-targeted pH-sensitive liposomes to efficiently deliver macro- molecules such as plasmids (discussed below) in addition to smaller drugs indicates that these liposomes can be widely utilized as a drug delivery vehicle. These stud- ies open up the possibility of using DNA as a new class of drug. For example, genes coding for toxic protein products or antisense RNA would be cytotoxic when de- livered to and expressed by the target cell. Genes coding for secreted products such as peptide hormones or proteins used as vaccines could be used.

Perhaps the brightest future for immunoliposome delivery lies in its unique abil- ity to deliver DNA to specific cells in vivo. Use of DNA as a chemotherapeutic drug offers at least the following three advantages over the use of the conventional

Page 37: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 379

drugs such as the small molecular-weight drugs, peptides and proteins: (a) the ac- t ion of gene is highly amplified th rough transcript ion and translation. Theoret i - cally a few copies of a gene are sufficient to p roduce significant biological modi- fications to a given cell; (b) the expression of a gene can be tightly regulated by choosing an appropr ia te p romote r and enhancers ; (c) if the D N A leaks out of the l iposome carrier it would be rapidly des t royed by the serum nucleases, and thus produce minimal toxicity to the normal cells. Target-specific immunoliposomes have now offered the oppor tun i ty for this novel class of chemotherapeut ic drug.

Much progress has been made iff the past decade toward an unders tanding of what role l iposomes may play as a drug delivery device. It is now clear that lipo- somes will not be the universal magic bullet as was earlier thought . More practical applications of l iposomes are now being pursued. Many l iposome-based drug for- mulat ions are soon scheduled for clinical trials backed by a newly spawned com- petit ive industry.

Acknowledgements

W o r k done in this labora tory is suppor ted by a contract f rom L i p o G e n , Inc. , N I H grants No. CA24553 and AI25834. L .H. was a recipient of a Research Career D e v e l o p m e n t A w a r d f rom N I H (CA00718). S.W. is a recipient of an N I H fellow- ship 1 F32 CA08442-01. We thank Carolyn Drake for excellent technical assist- ance.

References

1 Ehrlich, P. (1906) Collected Studies on Immunology, Vol. 2, John Wiley & Sons, New York. 2 Goldberg, E.P. (Ed.) (1983) Targeted drugs, John Wiley & Sons, New York. 3 Gregoriadis, G. (1977) Targeting of drugs, Nature 265, 407411. 4 Poznansky, M.J. and Juliano, R.L. (1984) Biological approaches to the controlled delivery of drugs:

a critical review, Pharmacol. Rev. 36, 277-336. 5 Poste, G. and Kirsh, R. (1983) Site-specific targeted drug delivery in cancer therapy, Biotechnology

869-878. 6 James, K. and Bell, G.T. (1987) Human monoclonal antibody production. Current status and fu-

ture prospects, J. Immunol. Methods 100, 5-40. 7 Edwards, D.C. and Mclntosh, D.P.(1986) Targeting potential of antibody conjugates. In: (G.M.

Ihler Ed.), Methods of drug delivery, Pergamon Press, New York, pp. 83-120. 8 Weinstein, J.N. (1983) Target-direction of liposomes: four strategies for attacking tumor cells. In:

B. Chadner (Ed.), Rational basis for chemotherapy, Alan R. Liss, New York, pp. 441~173. 9 Torchilin, V.P. (1987) Liposomes as targetable drug carriers, CRC Crit. Rev. Ther. Drug Carrier

Systems 2, 65-115. 10 Schneider, M. (1985) Liposomes as drug carriers: 10 years of research. In: P. Buri and A. Gumma

(Eds.), Drug targeting, Elsevier Science Publishers, Amsterdam, pp. 119-134. 11 Ostro, M.J. (Ed.) (1983) Liposomes, Marcel Dekker, New York. 12 Schmidt, K.H. (Ed.) (1986) Liposomes as drug carriers, Georg Thieme Verlag, Stuttgart. 13 Baldwin, H.T. and Six, H.R. (Eds.) (1980) Liposomes and immunobiology, Elsevier/North-Hol-

land, Amsterdam. 14 Gregoriadis, G. and Allison, A.C. (Eds.) (1980) Liposomes in biological systems, John Wiley &

Sons, Chichester. 15 Yagi, K. (Ed.) (1986) Medical application of liposomes, Japan Societies Press, Tokyo.

Page 38: Antibody-directed liposomes as drug-delivery vehicles

380 S. WRIGHT AND L. HUANG

16 Weinstein, J.N. and Leserman, L.D. (1984) Liposomes as drug carriers in cancer chemotherapy, Pharmacol. Ther. 24,207-233.

17 Ostro, M.J. (Ed.) (1987) Liposomes: from biophysics to therapeutics, Marcel Dekker, Inc., New York.

18 Bangham, A.D., Standish, M.M. and Watkins, J.C. (1965) Diffusion of univalent ions across the lamellae of swollen phospholipids, J. Mol. Biol. 13,238-252.

19 Connor, J., Sullivan, S. and Huang, L. (1985) Monoclonal antibody and liposomes. In: J.C, Venter and C.M. Fraser (Eds.), Pharmacology and Therapeutics, Vol. 28, Pergamon Press, London, pp. 341-365.

20 Sullivan, S., Connor, J. and Huang, L. (1986) Immunoliposomes: preparation, properties, and ap- plications, Medic. Res. Rev. 6, 171-195.

21 Collins, D., Norley, S., Rouse, B. and Huang, L. (1988) Liposomes as carriers for antitumor and antiviral drugs: pH°sensitive immunoliposomes and sustained release immunoliposomes, In: G. Gregoriadis (Ed.), Liposomes as drug carriers, John Wiley & Sons, New York, in press.

22 Collins, D. and Huang, L. (1988) Drug delivery by immunoliposomes. In: S. Ohki, T.D. Flanagan, N. Doyle, S.W. Hui and E. Mayhew (Eds.), Molecular mechanisms of membrane fusion, Plenum Publishing Co., New York, in press.

23 Torchilin, V.P. and Klibanov, A.L. (1981) Immobilization of proteins on liposome surface, En- zyme Microb. Technol. 3,297-304.

24 Heath, T.D. and Martin, F.J. (1986) The development and application of protein-liposome con- jugation techniques, Chem. Phys. Lipids 40, 347-358.

25 Weissmann, G., Bloomgarden, D., Kaplan, R., Cohen, C., Hoffstein, S., Collins, T., Gotlieb, A. and Nagle, D. (1975) A general method for the introduction of enzymes by means of immuno- globulin-coated liposomes into lysosomes of deficient cells, Proc. Natl. Acad. Sci. USA 72, 88-92.

26 Cohen, C.M., Weissmann, G., Hoffstein, S., Awasthi, Y.C. and Srivastava, S.K. (1976) Introduc- tion of purified hexosaminidase A into Tay-Sachs leukocytes by means of immunoglobulin-coated liposomes, Biochemistry 15,452-460.

27 Gregoriadis, G., Meehan, A. and Mah, M.M. (1981) Interaction of antibody-bearing small unila- mellar liposomes with target free antigen in vitro and in vivo, Biochem. J. 200, 203-210.

28 Huang, L. and Kennel, S.J. (1979) Binding of immunoglobulin G to phospholipid vesicles by son- ication, Biochemistry 18, 1701-1707.

29 Gregoriadis, G. and Neerunjun, E.D. (1975) Homing of liposomes to target cells, Biochem. Bio- phys. Res. Commun. 65, 53%544.

30 Senior, J., Waters, J.A. and Gregoriadis, G. (1986) Antibody-coated liposomes, the role of non- specific antibody adsorption, FEBS Lett. 196, 54-58.

31 Hashimoto, Y., Endoh, H. and Sugawara, M. (1984) Chemical methods for the modification of li- posomes with proteins or antibodies. In: G. Gregoriadis (Ed.), Liposome technology, Vol. 3, CRC Press, Boca Raton, FL, pp. 41-50.

32 Hashimoto, Y., Sugawara, M., Kamiya, T. and Suzuki, S. (1986) Coating of liposomes with sub- units of monoclonal IgM antibody and targeting of the liposomes, Methods Enzymol. 121,817-828.

33 Carroll, T.R., Davison, A. and Jones, A.G. (1986) Functional cholesteryl binding agents: synthe- sis, characterization and evaluation of antibody binding to modified phospholipid vesicles, J. Med. Chem. 29, 1821-1826.

34 Hashimoto, K., Loader, J.E. and Kinsky, S.C. (1986) Iodoacetylated and biotinylated liposomes: effect of spacer length on sulfhydryl ligand binding and avidin precipitability, Biochim. Biophys. Acta 856, 556--565.

35 Torchilin, V.P., Goldmacher, V.S. and Smirnov, V.N. (1978) Comparative studies on covalent and noncovalent immobilization of protein molecules on the surface of liposomes, Biochem. Biophys. Res. Commun. 85,983-990.

36 Torchilin, V.P., Khaw, B.A., Smirnov, V.N. and Haber, E. (1979) Preservation of antimyosin an- tibody activity after covalent coupling to liposomes, Biochim. Biophys. Res. Commun. 89, 1114--1119.

37 Jansons, V.K. and Mallett, P.L. (1981) Targeted liposomes: a method for preparation and analysis, Anal. Biochem. 111, 54-59.

38 Jansons, V.K. (1984) Preparation and analysis of antibody-targeted liposomes. In: G. Gregoriadis

Page 39: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 381

(Ed.), Liposome Technology, Vol. 2, CRC Press, Boca Raton, FL, Ch. 5, pp. 63-74. 39 Kung, V.T. and Redemann, C.T. (1986) Synthesis of carboxyacyl derivatives of phosphatidyle-

thanolamine and use as an efficient method for conjugation of protein to liposomes, Biochim. Bio- phys. Acta 862, 435-439.

40 Heath, T.D., Fraley, R.T. and Papahadjopoulos, D. (1980) Antibody targeting of liposomes: cell specificity obtained by conjugation of F(ab')2 to vesicle surface, Science 210, 53%541.

41 Heath, T.D., Macher, B.A. and Papahadjopoulos, D. (1981) Covalent attachment of immuno- globulins to liposomes via glycosphingolipids, Biochim. Biophys. Acta 640, 66-81.

42 Heath, T.D., Fraley, R.T., Bentz, J., Voss, E.W., Jr., Herron, J.N. and Papahadjopoulos, D. (1984) Antibody-directed liposomes: ~etermination of affinity constants for soluble and liposome-bound antifluorescein, Biochim. Biophys. Acta 770, 148-158.

43 Chua, M.-M., Fan, S.-T. and Karush, F. (1984) Attachment of immunoglobulin to liposomal mem- brane via protein carbohydrate, Biochim. Biophys. Acta 800, 291-300.

44 Leserman, L.D., Barbet, J., Kourilsky, F. and Weinstein, J.N. (1980) Targeting to ceils of flu- orescent liposomes covalently coupled with monoclonal antibody or protein A, Nature 288, 602-604.

45 Martin, F.J., Hubbell, W.L. and Papahadjopoulos, D. (1981) Immunospecific targeting of lipo- somes to cells: a novel and efficient method for covalent attachment of Fab' fragments via disulfide bonds, Biochemistry 20, 4229-4238.

46 Barbet, J., Machy, P. and Leserman, L.D. (1981) Monoclonal antibody covalently coupled to li- posomes: specific targeting to cells, J. Supramol. Struct. Cell, Biochem. 16,243-258.

47 Leserman, L.D., Machy, P. and Barbet, J. (1984) Covalent coupling of monoclonal antibodies and protein A to liposomes: specific interaction with cells in vitro and in vivo. In: G. Gregoriadis (Ed.), Liposome Technology, Vol. 3, Boca Raton, FL, pp. 29-40.

48 Goundalkar, A., Ghose, T. and Mezei, M. (1984) Covalent binding of antibodies to liposomes us- ing a novel lipid derivative, J. Pharm. Pharmacol. 36, 465-466.

49 Martin, F.J. and Papahadjopoulos, D. (1982) Irreversible coupling of immunoglobulin fragments to preformed vesicles: an improved method for liposome targeting, J. Biol. Chem. 10, 286-288.

50 Szoka, F. and Papahadjopoulos, D. (1978) Procedure for preparation of liposomes with large in- ternal aqueous space and high encapsulation by reverse-phase evaporation, Proc. Natl. Acad. Sci. USA 75, 4194-4198.

51 Bredehorst, R., Ligler, F.S., Kusterbeck, A.W., Chang, E.L., Gaber, B.P. and Vogel, C.-W. (1986) Effect of covalent attachment of immunoglobulin fragments on liposomal integrity, Biochemistry 25, 5693-5698.

52 Carlsson, J., Drevin, H. and Axen, R. (1978) Protein thiolation and reversible protein-protein con- jugation, Biochem. J. 173,723-737.

53 Duncan, R.J., Weston, P.D. and Wrigglesworth, R. (1983) A new reagent which may be used to introduce sulfhydryl groups, into proteins, and its use in the preparation of conjugates for immu- noassay, Anal. Biochem. 132, 68--73.

54 Derksen, J.T.P. and Scherphof, G.L. (1985) An improved method for the covalent coupling of pro- teins to liposomes, Biochim. Biophys. Acta 814, 151-155.

55 Traut, R.R., Bollen, A., Sun, T.-T., Hershey, J.W.B., Sundberg, J. and Pierce, LR. (1973) Methyl 4-mercaptobutyrimidate as a cleavable cross-linking reagent and its application to the Escherichia coli 30S ribosome, Biochemistry 12, 3266-3273.

56 Bl~ittler, W.A., Kuenzi, B.S., Lambert, J.M. and Senter, P.D. (1985) New heterobifunctional pro- tein cross-linking reagent that forms an acid-labile link, Biochemistry 24, 1517-1524.

57 Machy, P., Arnold, B., Alino, S. and Leserman, L.D. (1986) Interferon sensitive and insensitive MHC variants of a murine thymoma differentially resistant to methotrexate-containing antibody- directed liposomes and immunotoxin, J. lmmunol. 136, 3110-3115.

58 Truneh, A., Mishal, Z. and Leserman, L.D. (1984) A calmodulin antagonist increases the apparent rate of endocytosis of liposomes bound to MHC molecules via monoclonal antibodies, Exper. Cell Res. 155, 50-63.

59 Loughrey, H., Bally, M.B. and Cullis, P.R. (1987) A noncovalent method of attaching antibodies to liposomes, Biochim. Biophys. Acta 901, 157-160.

60 Godfrey, W., Doe, B., Wallace, E.E., Bredt, B. and Wofsy, L. (1981) Affinity targeting of mem- brane vesicles to cell surfaces, Exper. Cell Res~ 135, 137-145.

Page 40: Antibody-directed liposomes as drug-delivery vehicles

382 S. WRIGHT AND L. HUANG

61 Urdal, D.L. and Hakomori, S.-I. (1980) Tumor-associated Ganglio-N-triosylceramide target for an- tibody-dependent, avidin-mediated drug killing of tumor cells, J. Biol. Chem. 255, 10509-10516.

62 Sinha, D. and Karush, F. (1979) Attachment to membranes of exogenous immunoglobulin conju- gated to a hydrophobic anchor, Biochem. Biophys. Res. Commun. 90, 554-560.

63 Torchilin, V.P., Klibanov, A.I. and Smirnov, V.N. (1982) Phosphatidylinositol may serve as the hydrophobic anchor for immobilization of proteins on liposome surface, FEBS Lett. 138, 117-120.

64 Sunamoto, J., Sato, T., Hirota, M., Fukushima, K., Hiratani, K. and Hara, K. (1987) A newly developed immunoliposome - an egg phosphatidylcholine coated with puUulan bearing both a cho- lesterol moiety and an IgMs fragment, Biochim. Biophys. Acta 898, 323-330.

65 Huang, A., Huang, L. and Kennel, S.J. (1980) Monoclonal antibody covalently coupled with fatty acid - a reagent for in vitro liposome targeting, J. Biol. Chem. 255, 8015-8018.

66 Huang, A., Tsao, Y.S., Kennel, S.J. and Huang, L. (1982) Characterization of antibody covalently coupled to liposomes, Biochim. Biophys. Acta 716, 140-150.

67 Huang, A., Kennel, S.J. and Huang, L. (1984) Preparation of immunoliposomes. In: E. Reid, G.M.W. Cook and D.J. Morr'e (Eds.), Investigation of membrane-located receptors, Plenum Press, New York, pp. 351-355.

68 Huang, L., Huang, A. and Kennel, S.J. (1984) Coupling of antibodies with liposomes. In: G. Gre- goriadis (Ed.), Liposome technology, Vol. 3, CRC Press, Boca Raton, FL, pp. 51-62.

69 Harsch, M., Walther, P. and Weder, H.G. (1981) Targeting of monoclonal antibody-coated lipo- somes to sheep red blood cells, Biochem. Biophys. Res. Commun. 103, 1069-1076.

70 Sullivan, S.S. and Huang, L. (1985) Preparation and characterization of heat-sensitive immunoli- posomes, Biochim. Biophys. Acta 812, 116-126.

71 Shen, D.-F., Huang, A. and Huang, L. (1982) An improved method for covalent attachment of antibody to liposomes, Biochim. Biophys. Acta 689, 31-37.

72 Ho, R.J.Y., Rouse, B.T. and Huang, L. (1986) Target-sensitive immunoliposomes: preparation and characterization, Biochemistry 25, 5500-5506.

73 Torchilin, V.P., Omel'yanenko, V.G., Klibanov, A.L., Mikhailov, A.I., Gol'danskii, V.I. and Smirnov, V.N. (1980) Incorporation of hydrophilic protein modified with hydrophobic agent into liposome membrane, Biochim. Biophys. Acta 602,511-521.

74 Huang, A., Kennel, S.J. and Huang, L. (1983) Interactions of immunoliposomes with target cells, J. Biol. Chem. 258, 14034-14040.

75 Bragman, K.S., Heath, T.D. and Papahadjopoulos, D. (1983) Simultaneous interaction of mono- clonal antibody-targeted liposomes with two receptors on K562 cells, Biochim. Biophys. Acta 730, 187-195.

76 Singhal, A., Bali, A. and Gupta, C.M. (1986) Antibody-mediated targeting of liposomes to eryth- rocytes in whole blood, Biochim. Biophys. Acta 880, 72-77.

77 Lazarus, H., Raso, V. and Samy, T.S.A. (1977) In vitro inhibition of human leukemic cells (CCRF- CEM) by agarose-immobilized neocarzinostatin, Cancer Res. 37, 3731-3736.

78 Torchilin, V.P., Klibanov, A.L., lvanov, N.N., Gluckhova, M.A., Koteliansky, V.E., Kleinman, H.K. and Martin, G.R. (1985) Binding of antibodies in liposomes to extracellular matrix antigens, J. Cell. Biochem. 28, 23-29.

79 Smirnov, V.N., Domagatsky, S.P., Dolgov, V.V., Hvatov, V.B., Klibanov, A.L., Koteliansky, V.E., Muzykantov, V.R., Repin, V.S., Samokhin, G.P., Shekhonin, B.V., Smirnov, M.D., Sviridov, D.D., Torchilin, V.P. and Chazov, E.I. (1986) Carrier-directed targeting of liposomes and erythrocytes to denuded areas of vessel wall, Proc. Natl. Acad. Sci. USA 83, 6603-6607.

80 Huang, L. (1985) Incorporation of acylated antibody into planar lipid multilayers: characterization and cell binding, Biochemistry 24, 29-34.

81 Klibanov, A.L., Muzykantov, V.R., Ivanov, N.N. and Torchilin, V.P. (1985) Evaluation of quan- titative parameters of the interaction of antibody-bearing liposomes with target antigens, Anal. Biochem. 150,251-257.

82 Houck, K.S. and Huang, L. (1987) The role of multivalency in antibody mediated liposome tar- geting, Biochem. Biophys. Res. Commun. 145, 1205-1210.

83 Trubetskoy, V.S., Berdichevsky, V.R., Efremov, E.E. and Torchilin, V.P. (1987) On the possi- bility of the unification of drug targeting systems. Studies with liposome transport to the mixtures of target antigens, Biochem. Pharmacol. 36, 839-842.

Page 41: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 383

84 Tiebout, R.F., Van Boxtel-Oosterhof, F., Stricker, E.A.M. and Zeijlemaker, W.P. (1987) A hu- man hybrid hybridoma, J. Immunol. 139, 3402-3405.

85 Leserman, L.D., Weinstein, J.N., Blumenthal, R. and Terry, W.D. (1980) Receptor-mediated endocytosis of antibody-opsonized liposomes by tumor cells, Proc. Natl. Acad. Sci. USA 77, 4089-4093.

86 Weinstein, J.N., Blumenthal, R., Sharrow, S.O. and I-[enkart, P.A. (1978) Antibody-mediated targeting of liposomes: binding to lymphocytes does not ensure incorporation of vesicle contents into the cells, Biochim. Biophys. Acta 509,272-288.

87 Huang, A., Kennel, S.J. and Huang, L. (1981) Immunoliposome labeling: a sensitive and specific method for cell surface labeling, J. Immunol. Methods 46, 141-151.

88 Truneh, A., Machy, P. and Horan, P.K. (1987) Antibody-bearing liposomes as multicolor im- munofluorescence markers for flow cytometry and imaging, J. Immunol. Methods 100, 59-71.

89 Leserman, L.D., Machy, P. and Barber, J. (1981) Cell-specific drug transfer from liposomes bear- ing monoclonal antibodies, Nature 293,226--228.

90 Machy, P., Barbet, J. and Leserman, L.D. (1982) Differential endocytosis of T and B lymphocyte surface molecules evaluated with antibody-bearing fluorescent liposomes containing methotrexate, Proc. Natl. Acad. Sci. USA 79, 4148-4152.

91 Machy, P., Pierres, M., Barbet, J. and Leserman, L.D. (1982) Drug transfer into lymphoblasts mediated by liposome bound to distinct sites on H-2 encoded I-A, 1-E and K molecules, J. Im- munol. 129, 2098-2102.

92 Machy, P. and Leserman, L.D. (1983) Small liposomes are better than large liposomes for specific drug delivery in vitro, Biochim. Biophys. Acta 730, 313-320.

93 Machy, P., Truneh, A., Gennaro, D. and Hoffstein, S. (1987) Major histocompatibility complex class I molecules internalized via coated pits in T- lymphocytes, Nature 328,724-726.

94 Matthay, K.K., Heath, T.D. and Papahadjopoulos, D. (1984) Specific enhancement of drug de- livery to AKR lymphoma by antibody-targeted small unilamellar vesicles, Cancer Res. 44, 1880-1886.

95 Huet, C., Ash, J.F. and Singer, S.J. (1980) The antibody-induced clustering and endocytosis of HLA antigens on cultured human fibroblasts, Cell 21,429-438.

96 Boggs, J.M., Goundalkar, A., Doganoglu, F., Samji, N., Kurantsin-Mills, J. and Koshy, K.M. (1985) Antigen-targeted liposome-encapsulated methotrexate specifically kills lymphocytes sensi- tized to the nonapeptide of myelin basic protein, J. Neuroimmunol. 17, 35-48.

97 Heath, T.D., Montgomery, J.A., Piper, J.R. and Papahadjopoulos, D. (1983) Antibody-treated liposomes: Increase in specific toxicity of methotrexate-~-aspartate, Proc. Natl. Acad. Sci. USA 80, 1377-1381.

98 Goldman, D. and Matherly, L.H. (1984) Cellular pharmacology of methotrexate, Pharmac. Ther. 28, 77-102.

99 Piper, J.R., Montgomery, J.A., Sirotnak, F.M. and Chello, P.L. (1982) Synthesis of c~- and ~- substituted amides, peptides, and esters of methotrexate and their evaluation as inhibitors of fol- ate metabolism, J. Med. Chem. 25, 182-187.

100 Matthay, K.K., Heath, T.D., Badger, C.C., Bernstein, I.D. and Papahadjopoulos, D. (1986) An- tibody-directed liposomes: comparisons of various ligands for association, endocytosis, and drug delivery, Cancer Res. 46, 4904-4910.

101 Heath, T.D., Lopez, N.G., Stern, W.H. and Papahadjopoulos, D. (1985) 5-Fluoroorotate: a new liposome-dependent cytotoxic agent, FEBS Lett. 187, 73-75.

102 Yemul, S., Berger, C., Estabrook, A., Suarez, S., Edelson, R. and Bayley, H. (1987) Selective killing of T-lymphocytes by phototoxic liposomes, Proc. Natl. Acad. Sci. USA 84, 246-250.

103 Ohkuma, S. and Poole, B. (1978) Fluorescent probe measurements of the intralysosomal pH in living cells and the perturbation by various agents, Proc. Natl. Acad. Sci. USA 80, 3334-3337.

104 Tycko; B. and Maxfield, F.R. (1982) Rapid acidification of endocytic vesicles containing a ma- croglobulin, Cell 28,643-648.

105 Rustum, Y.M., Mayhew, E., Szoka, F. and Campbell, J. (1981) Inability of liposomes encapsu- lated 1-13-D-arabinofuranosyl-cytosine nucleotides to overcome drug resistance in L1210 cells, Eur. J. Cancer Clin. Oncol. 17,809-814.

106 Yatvin, M.B., Kreutz, W., Horwitz, B.A. and Shinitzky, M. (1980) pH-sensitive liposomes: pos- sible clinical implications, Science 210, 1253.

Page 42: Antibody-directed liposomes as drug-delivery vehicles

384 S. WRIGHT AND L. HUANG

107 Connor, J., Yatvin, M.B. and Huang, L. (1984) pH-sensitive liposomes: acid-induced liposome fusion, Proc. Natl. Acad. Sci. USA 81, 1715-1718.

108 Yatvin, M., Cree, T. and Tegmo-Larsson, I.-M. (1984) Theoretical and practical considerations in preparing liposomes for the purpose of releasing drug in response to changes in temperature and pH. In; G. Gregoriadis (Ed.), Liposome technology, Vol. 3, CRC Press, Boca raton, FL, pp. 157-176.

109 Gruner, S. (1985) Lipid polymorphism: the molecular basis of nonbilayer phases, Annu. Rev. Bio- phys. Biophys. Chem. 14, 211-238.

110 Cullis, P.R., Hope, M.J. and Tilcock, C.P.S. (1985) Lipid polymorphism: the molecular basis of nonbilayer phases, Annu. Rev. Biophys. Biophys. Chem. 14, 211-238.

111 Cullis, P.R., Hope, M.J. and Tilcock, C.P.S. (1986) Lipid polymorphism and the roles of lipids in membranes, Chem. Phys. Lipids 40, 127-144.

112 Nayar, R. and Schroit, A.J. (1985) Generation of pH-sensitive liposomes: use of large unilamellar vesicles containing N-succinyldioleoylphosphatidylethanolamine, Biochemistry 24, 5967-5971.

113 Leventis, R., Diacovo, T. and Silvius, J.R. (1987) pH-dependent stability and fusion of liposomes combining protonatable double-chain amphiphiles with phosphatidylethanolamine, Biochemistry 26, 3267-3276.

114 Wang, C.-Y., Hughes, K.W. and Huang, L. (1986) Improved cytoplasmic delivery to plant pro- toplasts via pH-sensitive liposomes, Plant Physiol. 82, 179-184.

115 Diigfines, N., Straubinger, R.M., Baldwin, P.A., Friend, D.S. and Papahadjopoulos, D. (1985) Proton-induced fusion of oleic acid-phosphatidylethanolamine liposomes, Biochemistry 24, 3091-3098.

116 Collins, D. and Huang, L. (1987) Cytotoxicity of diphtheria toxin A fragment to toxin-resistant murine ceils delivered by pH-sensitive immunoliposomes, Cancer Res. 47,735-739.

117 Connor, J. and Huang, L. (1985) Efficient cytoplasmic delivery of a fluorescent dye by pH-sen- sitive immunoliposomes, J. Cell Biol. 101,582-589.

118 Connor, J. and Huang, L. (1986) pH-sensitive immunoliposomes as an efficient and target-specific carrier for antitumor drugs, Cancer Res. 46, 3431-3435.

119 Ellens, H., Bentz, J. and Szoka, F.C. (1984) pH-induced destabilization of phosphatidylethano- lamine-containing liposomes: role of bilayer contact, Biochemistry 23, 1532-1538.

120 Bentz, J., Ellens, H. and Szoka, F. (1987) Destabilization of phosphatidylethanolamine-contain- ing liposomes: hexagonal phase and asymmetric membranes, Biochemistry 26, 2105-2116.

121 Lai, M.-Z., Vail, W.J. and Szoka, F.C. (1985) Acid- and calcium-induced structural changes in phosphatidylethanolamine membranes stabilized by cholesterol hemisuccinate, Biochemistry 24, 1654-1661.

122 Scheule, R.K. (1986) Fusion of Sindbis virus with model membranes containing phosphatidyle- thanolamine: implications for protein-induced membrane fusion, Biochim. Biophys. Acta 899, 185-195.

123 Straubinger, R.M., DiizgiJnes, N. and Papahadjopoulos, D. (1985) pH-sensitive liposomes me- diate cytoplasmic delivery of encapsulated macromolecules. FEBS Lett. 179, 148.

124 Tsusaki, B.E., Kanda, S. and Huang, L. (1986) Stimulation of superoxide release in neutrophils by 1-oleoyl-2-acetylglycerol incorporated into pH-sensitive liposomes, Biochem. Biophys. Res. Commun. 136, 242-246.

125 Ellens, E., Bentz, J. and Szoka, F.C. (1985) H ÷- and Ca2+-induced fusion and destabilization of liposomes, Biochemistry 24, 3099-3106.

126 Struck, D.K., Hoekstra, D. and Pagano, R.E. (1981) Use of resonance energy transfer to monitor membrane fusion, Biochemistry 20, 4093-4099.

127 Klibanov, A.L., Bogdanov, A.A., Jr. and Torchilin, V.P. (1989) Biotin-bearing pH-sensitive li- posomes: high-affinity binding to avidin layer, in press.

128 Huang, L., Connor, J. and Wang, C.-Y. (1987) pH-sensitive immunoliposomes, Methods En- zymol. 149, 88.

129 Pappenheimer, A.M., Jr. (1977) Diphtheria toxin, Annu. Rev. Biochem. 46, 69-94. 130 Ward, W.H.J. (1987) Diphtheria toxin: a novel cytocidal enzyme, Trends Biochem. Sci. 12, 28--31. 131 Uchida, T., Yamaizumi, M. and Okada, Y. (1977) Reassembled HVJ (Sendai virus) envelopes

Page 43: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 385

containing non-toxic mutant proteins of diphtheria toxin show toxicity to mouse L cells, Nature 266, 839-841.

132 Uchida, T., Kim, J., Yamaizumi, M., Miyake, Y. and Okada, Y. (1979) Reconstitution of lipid vesicles associated with HVJ (Sendai virus) spikes: purification and some properties of vesicles containing nontoxic fragment A of diphtheria toxin, J. Cell Biol. 80, 10-15.

133 Kirby, J. and Gregoriadis, G. (1984) A simple procedure for preparing liposomes capable of high encapsulation efficiency under mild conditions. In: G. Gregoriadis (Ed.), Liposome technology, Vol. 1, CRC Press, Boca Raton, FL, pp. 19-28.

134 Cline, M.J. (1984) Gene therapy, Mol. Cell. Biochem. 59, 3-10. 135 Fraley, R.T., Fornari, C.S. and Kaplan, S. (1979) Entrapment of a bacterial plasmid in phospho-

lipid vesicles: potential for gene transfer, Proc. Natl. Acad. Sci. USA 76, 3348--3352. 136 Makin, J.F. and Holt, J. (1981) Liposome-mediated transformation of streptomycetes by chro-

mosomal DNA, Nature (Lond.) 293,671-673. 137 Radford, A., Pope, S., Sazci, A., Frazer, M.J. and Parish, J.A. (1981) Liposome mediated ge-

netic transformation of neurospora crassa, Molec. Gen. Genet. 184,567-568. 138 Dimitriadis, G.J. (1978) Introduction of ribonucleic acids into cell by means of liposomes, Nucl.

Acids Res. 5, 1381-1389. 139 Ostro, M.J., Gracomoni, D., Lavella, D., Paxton, W. and Dray, S. (1978) Evidence for transla-

tion of rabbit globin mRNA after liposome-mediated delivery in a human cell line, Nature (Lond.) 247,923-928.

140 Wang, T.K., Nicolau, C. and Hofschneider, P.H. (1980) Appearance of 13-1actamase activity in animal cells upon liposome mediated gene transfer, Gene 10, 87-93.

141 Fraley, R.T. and Papahadjopoulos, D. (1982) Liposomes: the development of a new carrier sys- tem for introducing nucleic acids into plant and animal cells, Curr. Top. Microbiol. Immunol. 96, 171-190.

142 Schaefer-Ridder, M., Wong, Y. and Hofschneider, P.H. (1982) Liposomes as gene carriers: effi- cient transformation of mouse L ceils by thymidine kinase gene, Science 215, 166--168.

143 Soriano, S., Dijkstra, J., Legrand, A., Spanjer, H., Londos-Gagliardi, D., Roerdink, F., Scher- phof, G. and Nicolau, C. (1983) Targeted and nontargeted liposomes for in vivo transfer to rat liver cells of a plasmid containing the preproinsulin I gene, Proc. Natl. Acad. Sci. USA 80, 7128-7131.

144 Cudd, A. and Nicolau, C. (1985) Intracellular fate of liposome-encapsulated DNA in mouse liver. Analysis using EM autoradiography and subcellular fractionation, Biochim. Biophys. Acta 845, 477-491.

145 Wang, C.-Y. and Huang, L. (1988) Highly efficient DNA delivery mediated by pH-sensitive im- munoliposomes, in press.

146 Wang, C.-Y. and Huang, L. (1987) pH-sensitive immunoliposomes mediate target cell-specific de- livery and controlled expression of a foreign gene in mouse, Proc. Natl. Acad. Sci. USA 84, 7851-7855.

147 Wang, C.-Y. and Huang, L. (1987) Plasmid DNA adsorbed to pH-sensitive liposomes efficiently transforms the target cells, Biochem. Biophys. Res. Commun. 147,980-985.

148 Wynshaw-Boris, A., Lugo, T.G., Short, J.M., Rournier, R.E.K. and Hanson, R.W. (1984) Iden- tification of a cAMP regulatory region in the gene for rat cytosolic phosphoenolpyruvate carboxy- kinase (GTP), J. Biol. Chem. 259, 12161-12169.

149 Feigner, P.L., Gadek, T.R., Holm, M., Roman, R., Chan, H.W., Wenz, M., Northrop, J.P., Ringold, G.M. and Danielsen, M. (1987) Lipofection: a highly efficient, lipid-mediated DNA- transfection procedure, Proc. Natl. Acad. Sci. USA 84, 7413-7417.

150 Gruenberg, J. and Howell, K.E. (1987) An internalized transmembrane protein resides in a fu- sion-competent endosome for less than 5 minutes, Proc. Natl. Acad. Sci. USA 84, 5758-5762.

151 Matteoni, R. and Kreis, T.E. (1987) Translocation and clustering of endosomes and lysosomes depends on microtubules, J. Cell Biol. 105, 1253-1265.

152 Ellens, H., Bentz, J. and Szoka, F.C. (1986) Destabilization of phosphatidylethanolamine lipo- somes at the hexagonal phase transition temperature, Biochemistry 25,285-294.

153 Lai, M.-Z., Vail, W.J. and Szoka, F.C. (1985) Acid- and calcium-induced structural changes in

Page 44: Antibody-directed liposomes as drug-delivery vehicles

386 S. WRIGHT AND L. HUANG

phosphatidylethanolamine membranes stabilized by cholesteryl hemisuccinate, Biochemistry 24, 1654.

154 Siegel, D.P. (1988) Membrane-membrane interactions via intermediates in lamellar-to-inverted hexagonal phase transitions. In: A.E. Sowers (Ed.), Membrane Fusion, Plenum Publishing Co., New York, in press.

155 Blok, M.C., Van der Neut-Kok, C.M., Van Deenean, L.M., and De Gier, J. (1975) The effect of chain length and lipid phase transition on the selective permeability properties of liposomes, Biochim. Biophys. Acta 406, 187-203.

156 Yatvin, M.B., Weinstein, J.N., Dennis, W.H. and Blumenthal, R. (1978) Design of liposomes for enhanced local release of drugs by hyperthermia, Science 202, 1290-1291.

157 Magin, R. and Weinstein, J. (1984) The design and characterization of temperature-sensitive li- posomes. In: G. Gregoriadis (Ed.), Liposome technology, Vol. 3, CRC Press, Boca Raton, FL, pp. 137-156.

158 Wright, S.E. and White, J.C. (1986) Teniposide-induced changes in the physical properties of phosphatidylcholine liposomes: a calorimetric study, Biochem. Pharmacol. 35, 2731-2735.

159 Jain, M.K. and Wu, N.M. (1977) Effect of small molecules on the dipalmitoyl lecithin liposomal bilayer: III. Phase transition in lipid bilayer, Membrane Biol. 34, 157-201.

160 Weinstein, J.N., Magin, R.L., Yatvin, M.B. and Saharko, D.S. (1978) Liposomes and local hy- perthermia: selective delivery of methotrexate to heated tumors, Science 204, 188-190.

161 Weinstein, J.N., Magin, R.L., Cysyk, R.L. and Zaharko, D.S. (1980) Treatment of solid L1210 murine tumors with local hyperthermia and temperature-sensitive liposomes containing metho- trexate, Cancer Res. 40, 1388-1395.

162 Sullivan, S.S. and Huang, L. (1986) Enhanced delivery to target cells by heat-sensitive immuno- liposomes, Proc. Natl. Acad. Sci. USA 83, 6117-6121.

163 Taraschi, T.F., Van der Steen, A.T.M., De Kruijff, B., Tellier, C. and Verkleij, A.J. (1982) Lec- tin-receptor interaction in liposomes: evidence that binding of wheat germ agglutinin to glycopro- tein-phosphatidylethanolamine vesicles induce non-bilayer structure, Biochemistry 21, 5656-5674.

164 Ho, R.J.Y. and Huang, L. (1985) Interactions of antigen-sensitized liposomes with immobilized antibody: a homogeneous solid-phase immunoliposome assay, J. Immunol. 134, 4035-4040.

165 Hu, L.-R., Ho, R.J.Y. and Huang, L. (1986) Trypsin induced destabilization of liposomes com- posed of dioleoylphosphatidylethanolamine and glycophorin, Biochem. Biophys. Res. Commun. 141,973-978.

166 Van der Steen, A.T.M., De Kruijff, B. and De Gier, J. (1982) Glycophorin incorporation in- creases the bilayer permeability of large unilamellar vesicles in a lipid-dependent manner, Biochim. Biophys. Acta 691, 13-23.

167 Ho, R.J.Y., Rouse, B.T. and Huang, L. (1987) Interactions of target-sensitive immunoliposomes with herpes simplex virus: the foundation of a sensitive immunoliposome assay for the virus, J. Biol. Chem. 262, 13979-13984.

168 Ho, R.J.Y., Rouse, B.T. and Huang, L. (1987) Target-sensitive immunoliposomes as an efficient drug carrier for antiviral activity, J. Biol. Chem. 262, 13973--13978.

169 Ho, R.J.Y., Ting-Beall, H.P., Rouse, B.T. and Huang, L. (1988) Kinetic and ultrastructural stud- ies of interactions of target-sensitive immunoliposomes with herpes simplex virus, Biochemistry 27,500-504.

170 Pinnaduwage, P. and Huang, L. (1988) A homogeneous, liposome-based signal amplification for assays involving enzymes, Clinical Chemistry 34, 268-272.

171 Pinnaduwage, P. and Huang, L. (1988) [3-Galactosidase-induced destabilization of liposome com- posed of phosphatidylethanolamine and ganglioside GM1, Biochim. Biophys. Acta 939, 375-382.

172 Ho, R.J.Y. and Huang, L. (1988) Immunoliposome assays: perspectives, progress and potential. In: G. Gregoriadis (Ed.), Liposomes as drug carriers: trends and progress, John Wiley & Sons, in press.

173 Scherphof, G., Damen, J. and Wilschut, J. (1984) Interactions of liposomes with plasma proteins. In: G. Gregoriadis (Ed.), Liposome technology, Vol. 3, CRC Press, Boca Raton, FL, pp. 205-224.

174 Juliano, R.L. (1983) Interactions of proteins and drugs with liposomes. In: M.J. Ostro (Ed.), Mar- cel Dekker, New York, pp. 53-86.

Page 45: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 387

175 Bonte, F. and Juliano, R.L. (1986) Interactions of liposomes with serum proteins, Chem. Phys. Lipids 40, 359-372.

176 Bonte, F., Hsu, M.J., Papp, A., Wu, K., Regen, S.L. and Juliano, R.L. (1987) Interactions of polymerizable phosphatidylcholine vesicles with blood components: relevance to biocompatibility, Biochim, Biophys. Acta 900, 1-9.

177 Senior, J. and Gregoriadis, G. (1984) Methodology in assessing liposomal stability in the presence of blood, clearance from the circulation of injected animals, and uptake by tissues. In: G. Gre- goriadis (Ed.), Liposome technology, Vol. 3, CRC Press, Boca Raton, FL, pp. 263-282.

178 Scherphof, G., Roerdink, F., Waite, M. and Parks, J. (1978) Disintegration of phosphatidylcho- line liposomes in plasma as a result of interaction with high density lipoproteins, Biochim. Bio- phys. Acta 542, 296-303.

179 Babbitt, B., Huang, L. and Freire, E. (1984) Thermotropic and dynamic characterizations of in- teractions of acylated c~-bungarotoxin with phospholipid bilayer membranes, Biochemistry 23, 3920-3926.

180 Alving, C.R. and Richards, R.L. (1983) Immunologic aspects of liposomes. In: M.J. Ostro (Ed.), Liposomes, Marcel Dekker, New York, p. 209.

181 Kirby, C., Clarke, J. and Gregoriadis, G. (1980) Effect of the cholesterol content of small uni- lamellar liposomes on their stability in vivo and in vitro, Biochem. J. 186,591-597.

182 Allen, T.M., Ryan, J.L. and Papahadjopoulos, D. (1985) Gangliosides reduce leakage of aqueous- space markers from liposomes in the presence of human plasma, Biochim. Biophys. Acta 818, 205-210.

183 Sweeny, S.A. and Jonas, A. (1985) Substrate specificity of human plasma phospholipid transfer protein, Biochim. Biophys. Acta 835,279-290.

184 Agarwal, K., Bali, A. and Gupta, C.M. (1986) Effect of phospholipid structure on stability and survival times of liposomes in circulation, Biochim. Biophys. Acta 883,468-475.

185 Freeman, F.J., Hayward, J.A. and Chapman, D. (1987) Permeability studies on liposomes formed from polymerisable diacetylenic phospholipids and their potential applications as drug delivery systems, Biochim. Biophys. Acta 924,341-351.

186 Allen, T.M. and Chonn, A. (1987) Large unilamellar liposomes with low uptake into the reticu- loendothelial system, FEBS Lett. 223, 42-46.

187 Juliano, R. and Stamp, D. (1978) Pharmacokinetics of liposome-encapsulated anti-tumor drugs, Biochem. Pharmacol. 27, 22-27.

188 Abra, R.M. and Hunt, C.A. (1981) Liposome distribution in vivo: III dose and vesicle size effects, Biochim. Biophys. Acta 666,493-503.

189 Connor, J. and Huang, L. (1987) Acid-induced fusion of liposomes. In: A.E. Sowers (Ed.), Cell Fusion, pp. 285-289.

190 Connor, J., Norley, N. and Huang, L. (1986) Biodistribution of pH-sensitive immunoliposomes, Biochim. Biophys. Acta 884,474-481.

191 Hamilton, J.A. and Cistola, D.P. (1986) Transfer of oleic acid between albumin and phospholipid vesicles, Proc. Natl. Acad. Sci. USA 83, 82-86.

192 Scherphof, G., Roerdink, F., Hoekstra, D., Zborowski, J. and Wisse, E. (1980) Stability of li- posomes in presence of blood constituents: consequences for uptake of liposomal lipid and en- trapped compounds by rat liver cells. In: G. Gregoriadis and A.C. Allison (Eds.), Liposomes in biological systems, John Wiley and Sons, Chichester, pp. 179-209.

193 Leventis, R., Diacovo, T. and Silvius, J.R. (1987) pH-dependent stability and fusion of liposomes combining protonatable double-chain amphiphiles with phosphatidylethanolamine, Biochemistry 26, 3267-3276.

194 Norley, S.G., Sendele, D., Huang, L. and Rouse, B.T. (1987) Inhibition of herpes simplex virus replication in the mouse cornea by drug containing immunoliposomes, Invest. Ophthalmol. Vis. Sci. 28, 591-595.

195 Norley, S.G., Huang, L. and Rouse, B.T. (1986) Targeting of drug loaded immunoliposomes to herpes simplex virus infected corneal cells: and effective means of inhibiting virus replications in vitro, J. Immunol. 136, 681-685.

196 Herman, E.H., Rahman, A., Ferrans, V.J., Vick, J.A. and Schein, P.S. (1983) Prevention of

Page 46: Antibody-directed liposomes as drug-delivery vehicles

388 S. WRIGHT AND L. HUANG

chronic doxorubicin cardiotoxicity in beagles in liposomal encapsulation, Cancer Res. 43, 5427-5432. 197 Rahman, A., White, G., More, N. and Schein, P.S. (1985) Pharmacological, toxicological, and

therapeutic evaluation in mice of doxorubicin entrapped in cardiolipin liposomes, Cancer Res. 45, 796-803.

198 Forssen, E.A. and Tokes, Z.A. (1981) Use of anionic liposomes for the reduction of chronic dox- orubicin-induced cardiotoxicity, Proc. Natl. Acad. Sci. USA 78, 1873-1877.

199 Gabizon, A., Dagan, A., Goren, D., Barenholz, Y. and Fuks, Z. (1982) Liposomes as in vivo carriers of adriamycin:reduced cardiac uptake and preserved antitumor activity in mice, Cancer Res. 42, 4734-4739.

200 Forssen, E.A. and Tokes, Z.A. Improved therapeutic benefits of doxorubicin by entrapment in anionic liposomes, Cancer Res. 43, 546-550.

201 Lopez-Berestein, G., Hopfer, R.I., Mehta, R., Mehta, K., Hersh, E.M. and Juliano, R.L. (1984) Liposome-encapsulated amphotericin B for treatment of disseminated candidiasis in neutropenic mice, J. Infect. Dis. 150, 278-285.

202 Juliano, R., Grant, C., Barber, K. and Kalp, M. (1987) Mechanism of the selective toxicity of amphotericin B incorporated into liposomes, Mol. Pharm. 31, 1-11.

203 Alving, C.R. (1986) Liposomes as drug carriers for treatment of leishmaniasis. In: K. Yagi (Ed.), Medical application of liposomes, Japan Scientific Society Press, Tokyo, pp. 105-112.

204 Alving, C.R. (1987) Liposomes as carriers for vaccines. In: M.J. Ostro (Ed.), Liposomes: from Biophysics to Therapeutics, Marcel Dekker, New York, pp. 195-218.

205 Lopez-Berestein, G., Mehta, R., Hopfer, R.L., Mills, K., Kasi, L., Mehta, K., Fainstein, V., Luna, M., Hersh, E.M. and Juliano, R. (1983) Treatment and prophylaxis of disseminated infection due to Candida albicans in mice and liposome-encapsulated amphotericin B, J. Infect. Dis. 147, 939-945.

206 Lopez-Berestein, G. and Juliano, R.E. (1987) Application of liposomes to the delivery of anti- fungal agents. In: M.J. Ostro (Ed.), Liposomes: from Biophysics to Therapeutics, Marcel Dekker, New York, pp. 253-276.

207 Hwang, K.J. (1987) Liposome pharmacokinetics. In: M.J. Ostro (Ed.), Liposomes: from Bio- physics to Therapeutics, Marcel Dekker, New York, pp. 109-156.

208 Poste, G., Bucana, C., Raz, A., Bogleski, P., Kirsh, R. and Fidler, I.J. (1982) Analysis of the fate of systemically administered liposomes and implications for their use in drug delivery, Cancer Res. 42, 1412-1420.

209 Poste, G. (1983) Liposome targeting in vivo: problems and opportunities, Biol. Cell 47, 19-38. 210 Wisse, E. (1970) An electron microscopic study of the fenestrated endothelium lining of rat liver

sinusoids, J. Ultrastruct. Res. 31, 125-142. 211 Tumer, A., Kirby, C., Senior, J. and Gregoriadis, G. (1983) Fate of cholesterol rich liposomes

after subcutaneous injection into rats, Biochim. Biophys. Acta 760, 119-125. 212 Hashimoto, Y., Sugawara, M., Masuko, T. and Hojo, H. (1983) Antitumor effect of actinomycin

D entrapped in liposomes bearing subunits of tumor~specific monoclonal immunoglobulin M an- tibody, Cancer Res. 43, 5328--5334.

213 Patel, K.R., Li, M.P. and Baldeschweiler, J.D. (1983) Suppression of liver uptake of liposomes by dextran-sulfate 500, Proc. Natl. Acad. Sci. USA 80, 6518-6522.

214 Souhami, R.L., Patel, H.M. and Ryman, B.E. (1981) The effect of reticuloendothelial blockade on the blood clearance and tissue distribution of liposomes, Biochim. Biophys. Acta 674, 354-371.

215 Proffitt, R.T., Williams, L.E., Presant, C.A., Tin, G.W., Uliana, J.A., Gamble, R.C. and Bal- deschwieler, J.D. (1983) Liposomal blockade of the reticuloendothelial system: improved tumor imaging with small unilamellar vesicles 220, 502-503.

216 Ellens, H., Mayhew, E. and Rustum, Y.M. (1982) Reversible depression of the reticuloendo- thelial system by liposomes, Biochim. Biophys. Acta 714,479-485.

217 Hwang, K.J. (1986) Kinetic evidences of enhancing interaction of small liposomes with hepatic parenchymal cells mediated by large liposomes blockade, Res. Commun. Chem. Path. Pharmacol. 54, 417-420.

218 Aragnol, D. and Leserman, L.D. (1986) Immune clearance of liposomes inhibited by an anti-Fc receptor antibody in vivo, Proc. Natl. Acad. Sci. USA 83, 2699-2703.

219 Gregoriadis, G., Senior, J. and Wolff, B. (1986) Targeting of liposomes: optimization of vesicle

Page 47: Antibody-directed liposomes as drug-delivery vehicles

ANTIBODY-DIRECTED LIPOSOMES AS DRUG-DELIVERY VEHICLES 389

behavior in vivo. In: K. Yagi (Ed.), Medical application of liposomes, Japan Scientific Society Press, Tokyo.

220 Konno, H., Suzuki, H., Tadakuma, T., Kumai, K., Yasuda, T., Kubota, T., Ohta, S., Nagaike, K., Hosokawa, S., Ishibiki, K., Abe, O. and Saito, K. (1987) Antitumor effect of adriamycin en- trapped in liposomes conjugated with anti-human c~-fetoprotein monoclonal antibody, Cancer Res. 47, 4471-4477.

221 Lai, M.Z., Szoka, F.C. and Hunt, C.A. (1982) Surface attached transferrin enhances the binding of liposomes to erythroid cell in vitro but not in vivo, Pharm. Sci. 12, 1312-1319.

222 Debs, R.J., Heath, T.D. and Papahadjopoulos, D. (1987) Targeting of anti-Thy 1.1 monoclonal antibody conjugated liposomes in Thy 1.1 mice after intravenous administration, Biochim. Bio- phys. Acta 901, 183-190.

223 Leserman, L.D., Machy, P., Devaux, C. and Barbet, J. (1983) Antibody-bearing liposomes: tar- geting in vivo, Biol. Cell 47, 111-115.