Vous êtes sur la page 1sur 4

Targeted

Cationic Liposomes,
Technologies, and
Developments
Anas El-Aneed

C
ationic liposomes are widely used to transfer genetic ma-
terials into specific cells. A good liposomal formulation
for gene therapy should encapsulate and protect the nu-
cleic acid materials, escape endosomal degradation, and
reach the tumor site. The last goal can be achieved by incorpo-
rating a tumor-specific ligand that can deliver DNA to the tar-
geted tissue.
The same strategies that are applied when using anionic
liposomes to develop tissue-specific formulations also can be
PHOTODISC, INC.

applied when using targeted cationic liposomes. Because cationic


and noncationic liposomes have similar composition and struc-
ture, ligand attachment strategies also are similar, and any dis-
cussion in this area cannot separate the two liposomal groups.
The two main strategies in developing targeted liposomes are
Cationic liposomes are widely used in the attachment of a monoclonal antibody (mAb) (i.e., im-
gene therapy as a safe alternative to highly munoliposomes) or the attachment of a tissue-specific ligand
immunogenic viral vectors. Attachment of a to the surface of the liposomes. This article briefly reviews liver-
targeted and tumor-specific liposomes as examples of targeted
tissue-specific ligand to the surface of the liposomes. This article also describes liposome–ligand attach-
liposomes can increase specificity and ment techniques.
reduce undesired transfection. Targeted
liposomes can be categorized as either Immunoliposomes
immunoliposomes or ligand-targeted Antibodies are soluble proteins that are produced by B cells of
liposomes. The author provides a brief the immune system to bind to the antigens mediating their de-
struction. This process is accomplished either directly or with
review of tumor-specific and liver-targeted the help of other immune-system components—namely, Fab
cationic liposomes and the strategies for the (fragment antigen binding), which are fragments responsible
development of liposome–ligand for antigen recognition, and Fc (fragment crystallizable), which
complexes. are fragments that play a role in biological activity (1).
Immunoliposomes are studied because of their relative ease
of preparation and high specificity. In the early 1980s, researchers
Anas El-Aneed is a doctoral student in first linked antibodies or Fab fragments to liposomes by at-
biochemistry at the School of Pharmacy at taching them directly to lipids (2,3). Because of their short half
Memorial University of Newfoundland,
300 Prince Philip Dr., St. John’s, NL,
lives, immunoliposomes are used mainly in long circulating pe-
A1B 3V6 Canada, tel. 709.737.4331, gylated liposomes (4). Antibodies can be attached to the sur-
fax 709.777.7044, anas@pharm.mun.ca. face of the pegylated vesicles either at the terminal end of a poly-
ethylene glycol (PEG) chain (5,6) or directly onto the lipids (7).
58 Pharmaceutical Technology DECEMBER 2003 www.phar mtech.com
The former attachment methodology is used ex- In the case of hepatocyte cells, the main
tensively and favored because PEG serves as a spacer
Abbreviations challenge is to divert the liposomes from
between the ligand and the liposomal surface, DNA:deoxyribonucleic acid the lung “trap.” A conventional lip-
thereby providing easy access to the antibodies. PEG mAb:monoclonal antibody some–DNA complex (lipoplex) (21–23)
chains cause steric barriers when the mAb or Fab is PEG:polyethylene glycol and the novel liposomal preparation LPD
attached to the lipids. It has been shown that PEG EGFR:epidermal growth factor receptor (liposomes–protamine–DNA) (24–26)
2000 will mask the lipid-linked antibody to a lesser mRNA:messenger ribonucleic acid tend to be trapped by capillary embolism
degree than will the longer PEG 5000 chain (8). In LPD:liposomes/ Protamine/ DNA in the lungs where transfection may occur.
addition, a comparison study of PEG-linked and LDL:low density lipoprotein Liver accumulation of lipoplexes can be
lipid-linked antibodies has shown that coupling is ASGP-R:asialoglycoprotein receptors enhanced by manipulating the size of the
more efficient with the the use of PEG chains (9). AF:asialofetuin particles (27–29) or lowering the complex
Although the coupling reaction to PEG usually oc- hAAT:human alpha antitrypsin surface charge (30). Recent studies have
curs after the preparation of the liposomes, anchor HCC:hepatocellular carcinoma shown that transfection occurs mainly in
lipid molecules are attached to the antibody before the liver with the development of the
assembly into the liposomal structure during preparation (in serum resistant poly(cationic lipid) (31). However, one must en-
the case of direct linkage of the antibody to the lipids). How- sure that liposomes have actually reached the parenchymal liver
ever, a novel and simple preparation method for immunolipo- cells rather than the phagocytic kupffer cells. Some studies have
somes has been developed that involves transferring the lipid- shown that kupffer cells were actually the main destination for
conjugated mAb or Fab micelles to preformed, drug-loaded liposomes in the liver (32,33). This problem may be circum-
liposomes under specified conditions of temperature and pH vented by attaching a receptor-specific ligand to the surface of
(10–12). This method is referred to as the postinsertion tech- the liposomes. In such a case, the ligand binds to its receptors
nique. on the parenchymal cells before internalization occurs.
In two cancer gene therapy studies, researchers significantly Asialoglycoprotein receptors (ASGP-R) are abundant on the
enhanced gene expression in tumors using immunoliposome mammalian parenchymal liver cells. Their major role is to clear
technology instead of conventional liposomes (13,14). In an- glycoproteins and lipoproteins from circulation. The receptor
other study, the life span of mice bearing aggressive brain tu- contains a carbohydrate-recognition domain that can bind to
mors was increased by 100% after treatment with epidermal galactose derivatives (34).
growth factor receptor (EGFR) antisense mRNA delivered by Asialofetuin (AF) is a natural ligand for ASGP-R. It is a gly-
intravenous injection of immunoliposomes (15). coprotein with several terminal galactose sugar chains (35) and
In those studies, antibodies were covalently linked to the li- has been incorporated into the liposomal surface by covalently
posomal surface. However, noncovalent linkages also have been attaching a hydrophobic moiety (palmitic acid) as an anchor
used by simple mixing of the antibody with the liposomal vesi- among the lipids of the liposomal vesicles (AF-liposomes)
cles resulting in two- to four-fold increases in the transfection (36,37). In one study, the AF-liposomal uptake by the liver in
efficiency of the reporter gene in a glioma cell line (16,17). More- mice was increased 11 times in comparison with unmodified
efficient noncovalent linkages were obtained through avidin– liposomes (38). Similarly, AF-liposomal-mediated transfection
biotin binding. Biotinated lipids were bound to strepavidin, of the human alpha antitrypsin (hAAT) gene was significantly
which contains four biotin binding sites, and the system then enhanced in comparison with regular liposomes. After one year
was attached to biotinated mAb by simple incubation (9,18). of the treatment, hAAT mRNA in the liver was detected in all
Immunogenicity is the main concern associated with im- animals transfected with AF-liposomes versus only 25% with
munoliposome applications. This drawback was minimized those treated with regular liposomes, with more than a 4000-
with the use of Fab subunits instead of the whole antibodies or fold increase in the case of the AF-liposomes condition (39). In
the fully humanized mAb first produced in the 1980s (19,20). this study, AF was covalently linked to an anchor lipid on the
The linkage techniques of Fab fragments are identical to those surface of preformed liposomes. More recently, LPD was coated
applied on the complete mAb, covalently (3,13) or noncova- with the AF through charge–charge interactions, which signif-
lently (17,18). These liposome–ligand attachment methods also icantly increased the HepG2 cells’ uptake of the encapsulated
are applicable on all other peptide and protein ligands. DNA (40). AF, however, can induce immunogenic reaction.
Therefore, simpler glycosylated liposomes were developed and
Ligand-targeted liposomes evaluated for liver targeting (41). In another study, glycosylated
Ligand-targeted liposomes have lower immunogenicity in com- cholesterol, for example, was synthesized and incorporated into
parison with immunoliposomes. Ligands vary according to the the cationic liposomal vesicles, thereby resulting in a 10-fold
targeted tissues. One popular target is the liver, which is asso- increase in gene expression in the liver (42).
ciated with many genetically based diseases such as hemophilia, Liver cancer is another important target for gene delivery.
lipoprotein receptor deficiency, 1-antitrypsin deficiency, and HCC (hepatocellular carcinoma) is a leading cause of cancer-
liver cancer. Because many receptors, namely low-density related deaths worldwide (43). Gene therapy can provide a new
lipoprotein (LDL) and asialoglycoprotein receptors, are ex- approach to treat this fatal disease. In addition to the various
pressed on the surface of the liver, the discussion in this section receptors on the hepatocyte cells, there are some receptors that
focuses on liver-targeted liposomes. are over-expressed in hepatoma cells. One example is transfer-
60 Pharmaceutical Technology DECEMBER 2003 www.phar mtech.com
rin (TF) receptors, which are also elevated in other malignant Those Made by a New Postinsertion Approach,” Biochim. Biophys.
cells (44). TF-liposomes will not only target the cancerous cells, Acta. 1513 (2), 207–216 (2001).
12. T. Ishida, D.L. Iden, and T.M. Allen, “A Combinatorial Approach to
but will also reduce undesired transfection levels in the sur- Producing Sterically Stabilized (Stealth) Immunoliposomal Drugs,”
rounding normal tissues. This process can be exaggerated by FEBS Lett. 460 (1), 129–133 (1999).
hepatic arterial injection of TF-liposomes mediating DNA de- 13. C.H. Lee et al., “Enhanced Gene Delivery to Her-2-Overexpressing
livery (45). TF-liposome complexes are typically prepared Breast Cancer Cells by Modified Immunolipoplexes Conjugated with
through charge–charge interactions by simple mixing and in- the Anti-Her-2 Antibody,” J. Biomed. Sci. 10 (3), 337–344 (2003).
14. G.Y. Kao, L.J. Change, and T.M. Allen, “Use of Targeted Cationic Li-
cubation (45–47). Tumor growth was inhibited as much as 70% posomes in Enhanced DNA Delivery to Cancer Cells,” Cancer Gene
in liver tumor xenografts after treatment with TF-liposomes Ther. 3 (4), 250–256 (1996).
containing the antiangiogenesis gene, endostatin (46). Linkages 15. Y. Zhang, C. Zhu, and W.M. Pardridge, “Antisense Gene Therapy of
to the PEG terminal end of pegylated liposomes also were used Brain Cancer with an Artificial Virus Gene Delivery System,” Mol.
for TF-liposome preparations (48,49). Ther. 6 (1), 67–72 (2002).
16. J. Yoshida and M. Mizuno, “Simple Preparation and Characterization
of Cationic Liposomes Associated with Monoclonal Antibody Against
Closing remarks Glioma-Associated Antigen (Immunoliposomes),” J. Liposome Res. 5
Because cationic liposomes have a lower transfection efficiency (4), 981–995 (1995).
than viruses, modifications that may increase transfection lev- 17. M. Mizuno and J. Yoshida, “Repeated Exposure to Cationic Im-
els are always favored for the development of liposomal prepa- munoliposomes Activates Effective Gene Transfer to Human Glioma
Cells,” Neurol. Med. Chir. 36 (3), 141–144 (1996).
rations for gene transfer. One possible alteration is the intro- 18. H. Loughrey, M.B. Bally, and P.R. Cullis, “A Noncovalent Method of
duction of a tissue-specific ligand on the surface of the Attaching Antibodies to Liposomes,” Biochim. Biophys. Acta. 901 (1),
liposomes. This modification can enhance liposomal specificity 157–160 (1987).
and reduce the undesired delivery associated with toxic effects. 19. M. Verhoeyen, C. Milstein, and G. Winter, “Reshaping Human Anti-
Ligands vary according to the targeted tissues. Peptides, pro- bodies: Grafting an Antilysozyme Activity,” Science 239 (4847),
1534–1536 (1988).
teins, and sugar moieties have been explored as potential tar- 20. P.T. Jones et al., “Replacing the Complementarity-Determining Re-
geting ligands. Targeting specific tissues by altering the physi- gions in a Human Antibody with Those from a Mouse,” Nature 321
cal properties of liposomes also can be combined with ligand (6069), 522–525 (1986).
attachment for optimum targeting outcomes. Research in this 21. M. Iyer et al., “Noninvasive Imaging of Cationic Lipid-Mediated De-
area is expected to expand and increasingly enter the stage of livery of Optical and PET Reporter Genes in Living Mice,” Mol. Ther.
6 (4), 555–562 (2002).
clinical evaluation in humans. 22. J.W. McLean et al., “Organ-Specific Endothelial Cell Uptake of Cationic
Liposome-DNA Complexes in Mice,” Am. J. Physiol. 273 (1 Pt 2),
References H387–404 (1997).
1. E. Benjamini, R. Coico, and G. Sunshine, Immunology: A Short Course 23. G. Osaka et al., “Pharmacokinetics, Tissue Distribution, and Expres-
(Wiley–Liss, New York, NY, 4th ed., 2000). sion Efficiency of Plasmid [33P] DNA Following Intravenous Ad-
2. L.D. Leserman et al., “Targeting to Cells of Fluorescent Liposomes Co- ministration of DNA–Cationic Lipid Complexes in Mice: Use of a
valently Coupled with Monoclonal Antibody or Protein A,” Nature Novel Radionuclide Approach,” J. Pharm. Sci. 85 (6), 612–618 (1996).
288 (5791), 602–604 (1980). 24. Z. Ma et al.,“Lipid-Mediated Delivery of Oligonucleotide to Pulmonary
3. T.D. Heath, R.T. Fraley, and D. Papahdjopoulos, “Antibody Targeting Endothelium,” Am. J. Respir. Cell Mol. Biol. 27 (2), 151–159 (2002).
of Liposomes: Cell Specificity Obtained by Conjugation of F(ab')2 to 25. B. Li et al., “Lyophilization of Cationic Lipid-Protamine-DNA (LPD)
Vesicle Surface,” Science 210 (4469), 539–541 (1980). Complexes,” J. Pharm. Sci. 89 (3), 355–364 (2000).
4. G. Bendas, “Immunoliposomes: A Promising Approach to Targeting 26. S. Li and L. Huang, “In Vivo Gene Transfer via Intravenous Adminis-
Cancer Therapy,” Bio. Drugs 15 (4), 215–224 (2001). tration of Cationic Lipid-Protamine-DNA (LPD) Complexes,” Gene
5. N. Shi, R.J. Boado, and W.M. Pardridge, “Receptor-Mediated Gene Ther. 4 (9), 891–900 (1997).
Targeting to Tissues In Vivo Following Intravenous Administration 27. S.H. Hwang et al., “Liver-Targeted Gene Transfer into a Human He-
of Pegylated Immunoliposomes,” Pharm. Res. 18 (8), 1091–1095 (2001). patoblastoma Cell Line and In Vivo by Sterylglucoside-Containing
6. T.M. Allen et al., “A New Strategy for Attachment of Antibodies to Cationic Liposomes,” Gene Ther. 8 (16), 1276–1280 (2001).
Sterically Stabilized Liposomes Resulting in Efficient Targeting to Can- 28. S.F. Alino et al., “Human Alpha 1-Antitrypsin Gene Transfer to In Vivo
cer Cells,” Biochim. Biophys. Acta. 1237 (2), 99–108 (1995). Mouse Hepatocytes,” Hum. Gene Ther. 7 (16), 531–536 (1996).
7. A.L. Klibanov et al., “Activity of Amphipathic Poly(ethylene glycol) 29. J. Wu and M.A. Zern, “Modification of Liposomes for Liver Target-
5000 to Prolong the Circulation Time of Liposomes Depends on the ing,” J. Hepatol. 24 (6), 757–763 (1996).
Liposome Size and Is Unfavorable for Immunoliposome Binding to 30. R.I. Mahato et al., “Biodistribution and Gene Expression of Lipid–
Target,” Biochim. Biophys. Acta. 1062 (2), 142–148 (1991). Plasmid Complexes after Systemic Administration,” Hum. Gene Ther.
8. A. Mori et al., “Influence of the Steric Barrier Activity of Amphipathic 9 (14), 2083–2099 (1998).
Poly(ethyleneglycol) and Ganglioside GM1 on the Circulation Time 31. L. Liu et al., “Poly(cationic lipid)-Mediated In Vivo Gene Delivery to
of Liposomes and on the Target Binding of Immunoliposomes In Mouse Liver,” Gene Ther. 10 (2), 180–187 (2003).
Vivo,” FEBS Lett. 284 (2), 263–266 (1991). 32. D.C. Litzinger et al., “Fate of Cationic Liposomes and Their Complex
9. C.B. Hansen et al., “Attachment of Antibodies to Sterically Stabilized with Oligonucleotide In Vivo,” Biochim. Biophys. Acta. 1281 (2),
Liposomes: Evaluation, Comparison, and Optimization of Coupling 139–149 (1996).
Procedures,” Biochim. Biophys. Acta. 1239 (2), 133–144 (1995). 33. F. Roerdink et al., “The Involvement of Parenchymal, Kupffer, and En-
10. T.M. Allen, P. Sapra, and E. Moase, “Use of the Postinsertion Method dothelial Liver Cells in the Hepatic Uptake of Intravenously Injected
for the Formation of Ligand-Coupled Liposomes,” Cell Mol. Bio. Lett. Liposomes. Effects of Lanthanum and Gadolinium Salts,” Biochim.
7 (3), 889–894 (2002). Biophys. Acta. 677 (1), 79–89 (1981).
11. D.L. Iden and T.M. Allen, “In Vitro and In Vivo Comparison of Im- 34. J. Wu, M.H. Nantz, and M.A. Zern, “Targeting Hepatocytes for Drug
munoliposomes Made by Conventional Coupling Techniques with
Continued on page 79
62 Pharmaceutical Technology DECEMBER 2003 www.phar mtech.com
and Gene Delivery: Emerging Novel Approaches and Applications,” 44. H.N. Keer et al., “Elevated Transferrin Receptor Content in Human
Front. Biosci. 7, 717–725 (1 March 2002). Prostate Cancer Cell Lines Assessed In Vitro and In Vivo,” J. Urol. 143
35. R.G. Spiro, “Glycoproteins,” Adv. Protein Chem. 27, 349–467 (1973). (2), 381–385 (1990).
36. T. Hara et al., “Specific Uptake of Asialofetuin-Labeled Liposomes by 45. J.G. Seol et al., “Selective Gene Expression in Hepatic Tumor with
Isolated Hepatocytes,” Int. J. Pharm. 42 (1), 69–75 (1988). Transarterial Delivery of DNA–Liposome–Transferrin Complex,” In
37. S. Tsuchiya et al., “Preparation and Disposition of Asialofetuin- Vivo 14 (4), 513–517 (2000).
Labeled Liposome,” Biopharm. Drug Dispos. 7 (6), 549–558 (1986). 46. X. Li et al.,“Potent Inhibition of Angiogenesis and Liver Tumor Growth
38. J. Wu et al., “Increased Liver Uptake of Liposomes and Improved Tar- by Administration of an Aerosol Containing a Transferrin-Liposome-
geting Efficacy by Labeling with Asialofetuin in Rodents,” Hepatology Endostatin Complex,” World J. Gastroenterol. 9 (2), 262–266 (2003).
27 (3), 772–778 (1998). 47. M. Seki et al.,“p53 and PTEN/MMAC1/TEP1 Gene Therapy of Human
39. F. Dasi et al., “Asialofetuin Liposome-Mediated Human Alpha1-An- Prostate PC-3 Carcinoma Xenograft Using Transferrin-Facilitated
titrypsin Gene Transfer In Vivo Results in Stationary Long-Term Gene Lipofection Gene Delivery Strategy,” Hum. Gene Ther. 13 (6), 761–773
Expression,” J. Mol. Med. 79 (4), 205–212 (2001). (2002).
40. M.A. Arangoa, N. Duzgunes, and C. Tros de Ilarduya, “Increased Re- 48. A.S. Derycke and P.A. De Witte, “Transferrin-Mediated Targeting of
ceptor-Mediated Gene Delivery to the Liver by Protamine-Enhanced- Hypericin Embedded in Sterically Stabilized PEG-Liposomes,” Int. J.
Asialofetuin Lipoplexes,” Gene Ther. 10 (1), 5–14 (2003). Oncol. 20 (1), 181–187 (2002).
41. S. Kawakami et al., “Glycosylated Cationic Liposomes for Cell-Selec- 49. H. Iinuma et al., “Intracellular Targeting Therapy of Cisplatin-
tive Gene Delivery,” Crit. Rev. Ther. Drug Carrier Syst. 19 (2), 171–190 Encapsulated Transferrin-Polyethylene Glycol Liposome on Peritoneal
(2002). Dissemination of Gastric Cancer,” Int. J. Cancer 99 (1), 130–137 (2002).
42. S. Kawakami et al., “In Vivo Gene Delivery to the Liver Using Novel PT
Galactosylated Cationic Liposomes,” Pharm. Res. 17 (3), 306–313
(2000).
43. F.X. Bosch, “Global Epidemiology of Hepatocellular Carcinoma,” in
Liver Cancer, K. Okuda and E. Tabor, Ed. (Churchill Livingstone, New
York, NY, 1st ed., 1997), pp. 13–28.

Pharmaceutical Technology DECEMBER 2003 63

Vous aimerez peut-être aussi