Elsevier

Progress in Lipid Research

Volume 39, Issue 5, September 2000, Pages 409-460
Progress in Lipid Research

Current status of pH-sensitive liposomes in drug delivery

https://doi.org/10.1016/S0163-7827(00)00011-4Get rights and content

Introduction

Liposomes are being increasingly utilized to deliver drugs, enzymes, antisense oligonucleotides, and genes to various therapeutic targets. Liposome-mediated delivery of therapeutic molecules has continued to make considerable progress in the transition from the laboratory to the clinic [1], [2], [3], [4]. Circulation lifetimes have been greatly increased by incorporating polymer-coated lipids in the formulation to form sterically stabilized liposomes (SSL) and reduce uptake by macrophages of the reticuloendothelial system (RES) [5], [6], [7]. Advances in drug loading technologies have allowed for loading efficiencies approximating 100% of some amphipathic drugs using pH or ammonium sulfate gradients [8], [9], [10], [11], [12]. In addition, a more detailed understanding of the interactions of different drugs with liposomal membranes has resulted in more stable liposomal drug preparations [13], [14], [15], [16], [17]. Finally, specific targeting to internalizing cell surface receptors has increased both in vitro cytotoxicity [18], [19], [20], [21] and in vivo efficacy of liposomal drugs [4], [22]. These advances have contributed greatly to the development of current liposomal drugs either approved or presently in clinical trials; Doxil® (Alza Corp.; Palo Alto, CA), Evacet™ (Liposome Co.; New Brunswick, NJ), Daunoxome® (Nexstar Pharm.; Boulder, CO), and conventional liposomal vincristine [23], [24], and brightened the prospects for future liposome-based therapies.

However, while significant advances have been made in overcoming many of the barriers associated with liposomal drug delivery, an elusive problem has been the ability to selectively increase the bioavailability of the drug at the target tissue, while maintaining stability in the circulation. For pegylated liposomal doxorubicin (SSL-DOX), the drug is thought to leak very slowly from the carrier and thus be similar to a slow infusion of the drug specifically near the tumor [25], [26]. The exact mechanisms responsible for liposome breakdown remain speculative but may include lysosomal processing by tumor-residing macrophages [27], a destabilizing environment provided by the low pH, lipases, enzymes, and oxidizing agents found in the tumor interstitium [28], or catalysis by free ammonium sulfate [29]. Regardless of the mechanism, the release is slow and may not apply to other classes of drugs, more specifically highly charged water-soluble drugs. In addition, following release from the liposomal carrier, the drug or gene often needs to be delivered to cytosolic or nuclear targets. While amphipathic drugs are able to freely cross biological membranes, DNA, peptides, and water-soluble drugs are likely to be excluded completely from the cell or trapped in internal organelles.

To overcome these problems a variety of approaches have been employed; including complexation of DNA with cationic lipids [30], [31], [32], the design of thermosensitive liposomes capable of releasing their contents in response to small changes in temperature [33], [34], [35], and the development of pH-sensitive liposomes [36], [37], [38]. With regards to vectors, complexation and condensation of nucleic acids with cationic lipids still holds the greatest promise for gene delivery in vivo. The near complete entrapment or complexation of DNA to cationic lipids is considerably greater than the low encapsulation efficiencies observed with neutral or anionic liposomes. However, problems with in vivo stability of the complexes, rapid clearance from the circulation (t1/2 on the order of minutes), and primary distribution to the lung are just beginning to be addressed and much more work needs to be completed to increase the efficiency of delivery in vivo and make this a viable approach with systemic administration [32], [39], [40], [41]. Thermosensitive liposomes rely on a relatively sharp phase transition in the lipid phase at temperatures only a few degrees above 37° [33], [34], [35]. Packing defects observed at the gel-to-liquid crystalline phase transition result in increased permeability to entrapped solutes or drugs [33], [42]. Thermosensitive liposomes require the addition of an external stimulus (heat) in a spatially well defined region, surrounding and including the tumor. This creates a problem for distant metastasis and thus, may be limited in effectiveness to the primary tumor or substantial and detectable metastasis. Studies are in progress using whole body hyperthermia in an attempt to overcome this problem (Kirpotin, personal communication). The final approach and the focus of this review is the use of pH-sensitive liposomes to release their contents into the cytoplasm following endocytosis. This approach relies on selective destabilization of liposomes following acidification of the surrounding medium. The initial rationale for the design of pH-sensitive liposomes was to exploit the acidic environment of tumors to trigger destabilization of liposomal membranes [43]. However, the sites of greatest acidity in tumors are often the most distant from the tumor microvasculature, where liposomes often fail to reach [44], [45], [46]. In addition, the pH of the tumor interstitium rarely declines below pH 6.5 and therefore, makes it technically difficult to engineer liposomes that become disrupted in such a narrow window of pH. Endosomes and lysosomes, on the other hand, can reach values below 5.0 [47], [48], [49] and liposomes can be internalized by cells on the tumor periphery.

One fate of liposomes upon reaching the target tissues is they can be recognized and taken up by endocytosis [50]. However, upon uptake by the cell, they are eventually delivered to lysosomes (Fig. 1) [47], where the liposome and its contents may be degraded by various hydrolases and peptidases [51], [52]. The result can be diminished or lost biological activity of the delivered drug or macromolecule. pH-Sensitive liposomes have been designed to circumvent this problem by releasing their contents prior to reaching the lysosomes and at least partly, into the cytosol, where they can diffuse to their cytosolic or nuclear targets. Endosomes and lysosomes are acidified by proton-translocating ATPases [48], [53] to an average pH of approximately 5.0 [48], but which can be as low as 4.6 in macrophages [47], [49]. pH-Sensitive liposomes release their contents into the cytosol by one, or a combination, of several potential mechanisms (Fig. 1). These liposomes can be induced to undergo a pH-induced fusion of liposomal membranes with endosomal membranes, directly releasing liposomal contents into the cytosol. Alternatively, liposomes can become destabilized and cause the destabilization of endosomal membranes, resulting in leakage of the drug or liposomal “cargo” into the cytosol. In the case of certain encapsulated molecules, pH-sensitive liposomes may also become destabilized and release their contents solely inside the endosome, where amphipathic molecules may diffuse across endosomal membranes and other molecules (antifolates and nucleoside analogues) may be translocated by membrane transporters located in the endosomal membrane [54], [55], [56], [57].

Four basic classes of pH-sensitive liposomes have been previously described. The first class combines polymorphic lipids, such as unsaturated phosphatidylethanolamines, with mildly acidic amphiphiles that act as stabilizers at neutral pH. Although this class of pH-sensitive liposomes has been the most intensively studied, it may prove to be the least useful for in vivo drug delivery due to the effects of serum components on the formulation stability or pH-sensitivity, and their rapid clearance from the circulation. The second class includes liposomes composed of “caged” lipid derivatives [58], [59], [60], [61], [62]. Acid-induced hydrolysis of specifically engineered chemical bonds results in an increased presence of membrane destabilizing lipid components in the liposome membrane, and thus increased permeability to encapsulated solutes. A third class of pH-sensitive liposomes utilizes pH-sensitive peptides or reconstituted fusion proteins to destabilize membranes at low pH [63], [64], [65], [66], [67]. The final and most current class of pH-sensitive liposomes uses pH-titratable polymers to destabilize membranes following change of the polymer conformation at low pH [38], [68], [69], [70]. All four classes offer unique advantages and disadvantages that may vary in importance depending on the desired application. A careful comparison of these differences, limitations, and potential applications will be the focus of this review.

Section snippets

Mildly acidic amphiphiles

Mildly acidic amphiphiles can be combined with unsaturated phosphatidylethanolamines (PE), such as dioleoylphosphatidylethanolamine (DOPE) to form stable liposomes at neutral pH (for reviews see [37], [71], [72]). N-succinyldioleoylphosphatidylethanolamine (suc-DOPE), oleic acid (OA), palmitoylhomocysteine (PHC), cholesteryl hemisuccinate (CHEMS), and 1,2-dioleoyl- or 1,2-dipalmitoyl-sn-3-succinylglycerol (DOSG or DPSG) are a few of the stabilizers that have been used to prepare pH-sensitive

“Caged” liposomes

A novel strategy for designing pH-sensitive liposomes for drug delivery has been described using “caged” pH-labile N-maleylphosphatidylethanolamine derivatives (Fig. 8) [59], [60] or alkylether bonds [61], [152], [153]. “Caged” liposomes refer to liposomes that reversibly express a particular property, which may include the ability to form fusion competent non-bilayer phases or more simply drug permeable membranes. The “caging” process has involved both the reversible covalent modification of a

pH-sensitive peptides and virosomes

Recently, intense research has been directed towards mimicking the characteristics of certain viruses or toxins, in order to improve the efficiency of vaccination, drug and gene delivery. One of the most desirable properties of viral-based vectors is their ability to promote the destabilization of the host cell membrane to allow access of drugs, genes, and antigens to their intracellular targets. Indeed, enveloped viruses employ two basic routes of entry into host cells [183], [184], [185]. One

pH-sensitive polymer-liposome systems

In recent years synthetic polymers have received growing scientific attention in the formulation of liposomal preparations. For instance, coating liposomes with specifically designed hydrophilic polymers such as PEG has been shown to improve liposome stability and increase their circulation time in vivo [296], [297], [298], [299], [300], [301]. One interesting feature of polymers is that they can be tailored to participate actively in the release of drugs upon an external stimulation.

Pairing the delivery system with the molecule to be delivered

The design of the liposome to undergo one of the different mechanisms of release is extremely dependent on the type of molecule to be delivered. Large molecules, such as DNA that form complexes with cationic lipids in lipid-based particles or proteins when encapsulated in liposomes, may not benefit from delivery strategies which include a transient destabilization of the carrier's membrane. Fusion or disruption of the endosomal membrane is essential in these cases to ensure the entrapped

Conclusions and future directions

Considerable advances have been made in the design of pH-sensitive liposomal delivery systems since their first mention by Yatvin et al. [43]. Increased cytoplasmic delivery, rapid and pH-optimal contents release, and improvements in the types of liposomes that may be modified for pH-dependent contents release have all been achieved with a certain degree of success. However, many barriers remain before any of these systems may be utilized for in vivo drug or gene delivery. Studies have

Acknowledgements

D.C.D. was supported in part by a postdoctoral fellowship from the California Breast Cancer Research Program of the University of California, Grant Number 4FB-0154. M.Z. was supported in part by a postdoctoral fellowship from the FNSRS. J.C.L. was supported in part by the American Foundation for Pharmaceutical Education (AFPE) and the Burroughs Wellcome Fund.

First page preview

First page preview
Click to open first page preview

References (382)

  • J.W. Park et al.

    Adv Pharmacol.

    (1997)
  • A.L. Klibanov et al.

    FEBS Lett.

    (1990)
  • T.M. Allen et al.

    Biochim Biophys Acta

    (1991)
  • J.W. Nichols et al.

    Biochim Biophys Acta

    (1976)
  • T.D. Madden et al.

    Chem Phys Lipids

    (1990)
  • G. Haran et al.

    Biochim Biophys Acta

    (1993)
  • D.D. Lasic et al.

    Biochim Biophys Acta

    (1995)
  • P.R. Cullis et al.

    Biochim Biophys Acta

    (1997)
  • M.S. Webb et al.

    Biochim Biophys Acta

    (1998)
  • R.J. Lee et al.

    Biochim Biophys Acta

    (1995)
  • J.W. Park et al.

    Cancer Lett.

    (1997)
  • A.R. Horowitz et al.

    Biochim Biophys Acta

    (1992)
  • G. Storm et al.

    Waalkes, M.v.B., Crommelin D.J.A. Biochim Biophys Acta

    (1988)
  • K.L. Hong et al.

    FEBS Lett.

    (1997)
  • M.H. Gaber et al.

    Int J Radiat Oncol Biol Phys

    (1996)
  • R.M. Straubinger et al.

    FEBS Lett.

    (1985)
  • D.C. Litzinger et al.

    Biochim Biophys Acta

    (1992)
  • O. Meyer et al.

    FEBS Lett.

    (1998)
  • Y. Liu et al.

    J Biol Chem.

    (1995)
  • D. Papahadjopoulos et al.

    Biochim Biophys Acta

    (1973)
  • B. Tycko et al.

    Cell

    (1982)
  • D.L. Daleke et al.

    Biochim Biophys Acta

    (1990)
  • R.M. Straubinger et al.

    Cell

    (1983)
  • A. Huang et al.

    J Biol Chem.

    (1983)
  • J. Dijkstra et al.

    Biochim Biophys Acta

    (1984)
  • P.M. Brown et al.

    Biochim Biophys Acta

    (1990)
  • D. Kirpotin et al.

    FEBS Lett.

    (1996)
  • D.C. Drummond et al.

    Chem Phys Lipids

    (1995)
  • O.V. Gerasimov et al.

    Biochim Biophys Acta

    (1997)
  • R.A. Parente et al.

    J Biol Chem.

    (1988)
  • C. Puyal et al.

    Biochim Biophys Acta

    (1994)
  • A.L. Bailey et al.

    Biochim Biophys Acta

    (1997)
  • K. Kono et al.

    Biochim Biophys Acta

    (1994)
  • M. Zignani et al.

    Biochim Biophys Acta

    (2000)
  • D. Collins et al.

    Biochim Biophys Acta

    (1989)
  • D.P. Siegel

    Biophys J.

    (1993)
  • L. Chernomordik

    Chem Phys Lipids

    (1996)
  • R. Sundler et al.

    Biochim Biophys Acta

    (1981)
  • J.M. Boggs

    Biochim Biophys Acta

    (1987)
  • D. Papahadjopoulos et al.

    Biochim Biophys Acta

    (1974)
  • D. Collins et al.

    Biochim Biophys Acta

    (1990)
  • R. Nayar et al.

    Biochim Biophys Acta

    (1988)
  • D.C. Drummond et al.

    Pharmacol Rev.

    (1999)
  • Bally MB, Lim H, Cullis PR, Kirpotin D. J Liposome Res...
  • D.D. Lasic et al.

    Medical applications of liposomes

    (1998)
  • D. Papahadjopoulos et al.

    Proc Natl Acad Sci USA

    (1991)
  • E. Mayhew et al.

    Cancer Treat Rep

    (1979)
  • Y. Barenholz et al.

    Med Res Rev.

    (1993)
  • Y. Barenholz et al.

    J Liposome Res.

    (1995)
  • Barenholz Y. In: Lasic DD, Papahadjopoulos D., editors. Medical applications of liposomes. New York: Elsevier Science...
  • Cited by (424)

    • Lipid membrane-based therapeutics and diagnostics

      2021, Archives of Biochemistry and Biophysics
    View all citing articles on Scopus
    View full text