Chonnam Med J. 2021 Jan;57(1):27-35. English.
Published online Jan 25, 2021.
© Chonnam Medical Journal, 2021
Review

Liposomes: Biomedical Applications

Eun-Mi Kim and Hwan-Jeong Jeong
    • Department of Nuclear Medicine, Molecular Imaging & Therapeutic Medicine Research Center, Research Institute of Clinical Medicine of Jeonbuk National University and Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea.
Received December 04, 2020; Revised December 22, 2020; Accepted December 23, 2020.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Liposomes, with their flexible physicochemical and biophysical properties, continue to be studied as an important potential a critical drug delivery system. Liposomes have overcome the challenges of conventional free drug therapy by encapsulating therapeutic agents, thereby improving in vivo biodistribution and reducing systemic toxicity. New imaging modalities and interpretation techniques, as well as new techniques for targetable system formulation technique, and tumor environmental information, have affected the search for a means of overcoming the difficulties of conventional liposome formulation. In this review, we briefly discuss how liposomal formulation has been applied across the biomedical field, particularly as a therapy, and the role it may play in the future, when paired with new developments in diagnosis and theranostics. The biological challenges that still remain and the translational obstacles are discussed.

Keywords
Liposomes; Drug Delivery Systems; Precision Medicine; Neoplasms

INTRODUCTION

The number of potential drug delivery systems that have been studied is vast, and includes carriers such as metallic, organic, inorganic, nanohybrid, and polymeric nanoparticles like micelles, dendrimers, and liposomes. Among these, liposomes, which have lipid bilayer spherical nanovesicles that resemble the structure of cell membrane, are a very promising carrier.1, 2, 3 Since the 1964 discovery of liposomes by Prof. Bangham, numerous studies have sought to understand their possible clinical applications. Liposomes have a number of advantages, which explains why they are present in a significant percentage of clinical-stage nanotherapeutics and pharmaceutical preparations. They are biodegradable, biocompatible, non-toxic, and composed of non-immunogenic compounds. They also improved solubility of lipophilic and amphiphilic drugs, passively target immune system cells, permit sustained release for systemic and local administration, and offer improved tissue penetration.3, 4 To date, more than eighteen liposomal drugs have been approved by FDA for the treatment of cancer, infectious disease, pain management, and age-related macular degeneration.2

Liposomes are comprised of a lipid bilayer structure and lipid monolayer structures called micelles (Fig. 1). A liposomal bilayer is typically composed of cholesterol and phospholipids.5 Due to their unique biphasic nature, these can be used to deliver both hydrophilic as well as lipophilic drugs. Liposomes can vary in size from very small to large (20 nm-2.5 µm). Vesicle size is a main parameter for determining the in vivo circulation half-life, as both the size and number of bilayers affects the amount of drug encapsulation possible in the liposomes.6, 7 Liposomes can be classified into two categories on the basis of their size and number of bilayers: multilamellar vesicles (>500 nm) referred as MLV and unilamellar vesicles. Unilamellar vesicles can be further classified as large (LUV, >100 nm) and small unilamellar vesicles (SUV, <100 nm).8 These structures are depicted in Fig. 2.

FIG. 1
Liposome (A) and micelle (B) structure.

FIG. 2
Structure of MLV (A), LUV (B), and SUV (C). MLV: multilamellar vesicles, LUV: large unilamellar vesicles, SUV: small unilamellar vesicles.

There are several methods of liposome preparation and drug loading, including passive loading techniques and an active loading technique. The most common passive loading methods are thin-film hydration, microemulsification, sonication, membrane extrusion, microfluidizer, freeze-thawing, ether injection, ethanol injection, reverse phase evaporation, dehydration-rehydration, and calcium-induced fusion.9, 10, 11 Of these, thin-film hydration is the most widely used. In this passive loading method, a water-soluble drug is encapsulated in the aqueous phase of a liposome, while a lipid-soluble drug is loaded into the lipid layer. With active loading, drugs are loaded by creating a diffusion gradient for the ions or drugs across the external to internal phases. This method, called “remote loading”, is used in liposomal formulations of doxorubicin, and offers significant improvements in the drugs' pharmacokinetics and safety profiles in humans.12, 13, 14, 15, 16

Although liposomes have been intensely investigated for 50 years, they continue to be the object of vigorous research. Liposomes have a reputation as an optimal delivery system for drugs or biologically active substances, and are regarded one of the most successful drug delivery systems identified to date. Despite their success, liposomes still face problems related to their stability, storage, drug leakage, rapid clearance, and specific targeting ability. In this review, we provide a brief overview of liposomes as they are applied in medicine.

LIPOSOME APPLICATIONS IN MEDICINE

1. Liposomes and cancer therapy

Cancer is a leading cause of death in many countries. Regrettably, the efficacy of standard treatments for a variety of cancers is not optimal.17 Many anticancer agents are highly toxic, and a number of cytotoxic chemotherapeutics have short half-lives in vivo due to their highly hydrophobic nature, which leads to side effects, noncompliance, and patient inconvenience as a result of difficulties in administration. This, in turn, limits their utility in cancer therapy.9, 17, 18 In cancer therapies, a liposomal delivery system is particularly useful. The incorporation of chemotherapeutic agents into liposomes can improve their specificity to cancer cells and tumor tissues through passive or ligand-mediated active targeting, resulting in a minimization of the drug's negative side-effects as well as enhanced anticancer efficacy from the increased accumulation of liposomes within the tumors.

A PEGylated liposomal formulation of doxorubicin (Doxil) was the first FDA-approved nanosized anti-cancer drug delivery system. After the approval of Doxil® in 1995, a number of other cytotoxic agent-containing liposomes were approved for clinical use, with a number of liposomal chemotherapeutic formulations having been approved by the European Medicines Agency (EMA) and the U.S. Food and Drug Administration (FDA) for the treatment of various cancers in the past few years alone.19, 20, 21, 22 A summary of liposome-based anticancer drugs is provided in Table 1. Several more anticancer liposome formulations are currently in various stages of clinical trials or awaiting approval (Table 2).

TABLE 1
Clinically used liposomal formulation products for cancer therapy

TABLE 2
Liposomes in ongoing clinical trials for cancer therapy

Despite these successes by liposomal anticancer drugs, which have been achieved by an enhanced permeation and retention (EPR) effect, there have been many reports of conventional liposomes that have failed for their lack of selectivity. In these cases, a minimum therapeutic concentration of liposomes is delivered within tumor tissues while the drug accumulates in healthy tissue and mucosa, resulting in treatment failure.23, 24

As an alternative, the active “ligand-mediated” targeting approach has been thoroughly investigated for its improved intracellular delivery of an encapsulated drug to a tumor tissue. A variety of molecules, including peptides, antibodies, proteins, and charged molecules, and some low molecular weight ligands, and aptamers, have been studied for use in conjunction with liposomes to enhance anticancer treatments. For further improvement, multi-functional liposomes with specific features like targeted delivery functionalities designed using a variety of surface functionalization and modification approaches, sustained and triggered release, will play a vital role in future tumor therapies.19, 25 Table 3 presents therapies using ligand-mediated targeted liposomes in the clinical trial stage.

TABLE 3
Examples of ligand-targeted liposome formulations undergoing clinical evaluation

2. Liposomes and infectious and vascular diseases

1) Liposomes and fungal infections

Fungal infections are categorized as either superficial or invasive. Invasive fungal infections, while less common, have a higher morbidity and mortality rate, especially in immunocompromised patients.26, 27 Amphotericin B, with a broad antifungal spectrum and minimal risk of resistance, has been considered the gold standard for the treatment of severe systemic fungal infections.28 Unfortunately, amphotericin B is associated with acute and chronic as well as dose-dependent toxicity. To solve this, amphotericin B encapsulated liposomes were introduced, which reduced its toxicity and allowed for its administration at higher doses.29

Three major classes of antifungal drugs exist: (a) polyenes, (b) azoles, and (c) echinocandins.30 Amphotericin B is a poorly water-soluble polyene produced by Streptomyces nodosus.31 Encapsulating amphotericin B in a liposome has been useful for overcoming the drug's disadvantages. Research in the 1980s-1990s ultimately led to the development and clinical introduction of three lipid formulations of amphotericin B: (a) liposomal amphotericin B (LAmB; AmBisome), (b) amphotericin B lipid complex (ABLC; Abelcet), and (c) amphotericin B colloidal dispersion (ABCD; Amphotec).32 AmBisome was developed in the 1980s and showed 30-fold reduction in toxicity while retaining the pharmacological effectivity of the active ingredient in preclinical models.33 Table 4 presents liposomal formulation products currently in clinical use to treat fungal infections.

TABLE 4
Clinically used liposomal formulation products for fungal infection

The current generation of commercially available AmB lipid formulations are expensive and require parenteral administration, resulting in longer hospital stays and increased healthcare costs. There is an urgent need to develop an orally available AmB formulation that decreases the systemic toxicity of the drug, avoids infusion-related adverse events, improves patient compliance, and reduces the costs associated with current commercial AmB formulations.29, 34

2) Liposomes and bacterial infections

Among the most serious problems in medicine today is the increase in drug-resistant bacterial pathogens, against which conventional therapies have only limited effectiveness.35 Biofilms are clusters of microorganisms and are a common cause of chronic, nosocomial, and hospital acquired infections like implantable medical devise. Biofilms are associated with high-level resistance to antimicrobials, frequent treatment failure, and increased morbidity and mortality. Both Staphylococcus aureus and Pseudomonas aeruginosa are known as major pathogens. Once a biofilm is formed, bacteria become up to 1000x more resistant to antibiotic treatment than their planktonic type.36 Localized delivery of antimicrobials is a promising tactic to treat challenging infections like biofilms and intracellular infections such as Salmonellosis.37, 38, 39 Many research results have shown that liposome encapsulation improves the efficacy of antibacterial drugs against a broad range of pathogens both in vitro and in vivo.

Liposome-encapsulated amikacin, marketed under the name Arikace, is clinically approved for the treatment of Mycobacterium avium complex lung disease.40 Arikace is a novel formulation of inhaled liposomal amikacin for the treatment of patients suffering from chronic Pseudomonas aeruginosa biofilms.41 Table 5 presents liposomal formulation products for bacterial infections in use or in clinical trials.

TABLE 5
Examples of liposome formulations undergoing clinical evaluation

3) Liposomes and vaccine formulation

Vaccination is the most cost effective prophylactic strategy against many types of diseases like as pathogenic infections (viral, bacterial, fungal or parasitic origin), cancerous lesions, and even rheumatoid arthritis.42 Gregoriadis and Allison were the first to report on the ability of liposomes to induce immune responses to incorporate as vaccine adjuvants or associated antigens.43 Since then, liposomes (natural, anionic, and cationic lipid), archaeosomes (polar glycerolipid plus lipid), and virosomes (viral extract and lipid) have become important vaccine systems, and interest in liposome-based vaccines has markedly increased.

Epaxal, Inflexal, and Mosquirix are now clinically approved liposome-based vaccine products.42, 44 Table 6 presents details associated with these three approved drugs. All three are classified as virosomes, which are liposomes comprised of a phosphatidylcholine membrane vesicle that incorporates a virus-derived protein. All three vaccines are safe, well-tolerated, and effective at generating an immune response. Advantages of particulate vaccines are greater protection of antigens from enzymatic degradation by encapsulation and also can deliver molecular adjuvants with antigen to antigen presenting cells (APC), thus promoting cellular and humoral immune responses.45

TABLE 6
Clinically used liposomal vaccine products for infection

Traditionally, vaccines have relied on the use of whole killed or live attenuated pathogens. Today, research is focused on the development of subunit vaccines that are better defined, easier to produce, and safer. Liposomes are promising delivery systems for subunit vaccines composed of fragment of a pathogen which can trigger immune response.43 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) based cationic liposomes have been extensively studied as potential vaccine delivery systems and adjuvants. Recently, combining liposomes with immuno-stimulating ligands has been studied as a potential next-step in the development of novel adjuvant systems.

3. Liposomes and angiogenesis in vascular disease

Vascular disease including atherosclerosis, thrombosis, stroke, coronary artery disease (CAD), and peripheral artery disease (PAD) is a leading cause of death and disability worldwide.46 Inflammation plays a main role in the initiation and progression of both PAD and CAD. CAD and PAD are common vascular diseases which are characterized by reduced blood supply caused by a buildup of plaque in the peripheral or coronary arteries, leading to an ischemic state, i.e., a deficient nutrient and oxygen supply to the head, organs and limbs.47 One potential approach to overcoming this disease is the promotion of angiogenesis in ischemic tissues via the delivery of angiogenic factors. Angiogenic factors promote the generation of new blood vessels and increase blood flow in damaged tissue in the ischemic state.48 Successful angiogenesis is highly dependent on delivery of vascular endothelial growth factor (VEGF) to an ischemic site.

Clinical trials of VEGF growth factor in patients failed to obtain a significant therapeutic effect and this was due to the short in vivo half-life and lack of specificity to the target site.49 Therefore, targeted delivery and then sustained release of growth factors in ischemic sites is a grand challenge. Inspired by the targeted nanocarriers in cancer therapy, various types of nanopaticles have been studied as potential therapies for ischemic lesions, including graphene oxide,49 silica nanoparticles,50 polyglutamic acid (PGA) polypeptides,51 ONO-1301 containing lipid nanoparticles,52 polylactic-co-glycolic acid (PLGA) nanoparticles,53 and ironoxide nanoparticles.54

Nam et al.55 injected VEGF protein-loaded Alexa Fluor 750-labeled liposome into the ischemic hind limb mouse model. The liposomes were delivered intravenously to target ischemic sites and achieved VEGF-influenced angiogenesis at ischemic sites. In our lab, when PEGylated liposomes loaded with angiogenic peptide from the VEGF sequence origin was used in a cerebral ischemia rat model, with vascular density increasing in response to angiogenic therapy.56 Furthermore, 99mTc-radiolabeled PEGylated liposomes loaded with angiogenic peptides as a theranostic agent were administered to hind limb ischemia in an animal model. The liposomal agent improved ischemic limb perfusion and promoted angiogenic responses.57 This agent was also applied to aischemia/reperfusion heart injury in an animal model. This liposome accumulated in the ischemic myocardium and induced its own protective effect against ischemia/reperfusion injury.58

4. Liposomes, theranostic applications, and diagnostic techniques

Theranostics is the combination of the terms “Therapeutics” and “Diagnostics,” and refers to technologies that include both diagnostic and therapeutic functions in one complex. Theranostics have attracted a great deal of attention and theranostic nano-carrier systems are considered to be a critical component of the next generation of medicine. Well-made theranostic nanoparticles allow for the monitoring of real-time drug delivery, accurate diagnosis and assessment of biological signals, easier determination of responses to a therapy, an increase in the use of minimally invasive procedures that rely on precision guidance, and better decisions concerning the end point of therapy, all of which promote the development of individualized medicine.59, 60

Theranostics is the combination of the terms “Therapeutics” and “Diagnostics,” and refers to technologies that include both diagnostic and therapeutic functions in one complex. Theranostics have attracted a great deal of attention and theranostic nano-carrier systems are considered to be a critical component of the next generation of medicine. Well-made theranostic nanoparticles allow for the monitoring of real-time drug delivery, accurate diagnosis and assessment of biological signals, easier determination of responses to a therapy, an increase in the use of minimally invasive procedures that rely on precision guidance, and better decisions concerning the end point of therapy, all of which promote the development of individualized medicine.59, 60

The development of new imaging modalities has led to the design of various diagnostic and theranostic nanodelivery systems. Today, imaging techniques including optical fluorescence imaging, magnetic resonance imaging (MRI), computed tomography (CT), positron emission tomography (PET) and single photon emission computed tomography (SPECT) are in wide use. Nuclear imaging is high sensitive, capable of imaging an entire body, without risk of issues associated with tissue penetration, and highly accurate. High sensitivity is particularly important in therapeutic nanomedicines. Unlike MRI and CT scans where gram quantity of a contrast medium is required, nuclear imaging is achieved with injections of micrograms or less.61 Nuclear imaging modalities are limited in that their spatial resolution in the range of below 10 mm, which is lower than optical or MR imaging. The majority of radionuclide imaging is performed by SPECT or PET.62, 63

Flexibility of liposomes in surface functionalization provide opportunities for labeling with various imaging probes which can co-deliver therapeutic drugs that allow them to act as both therapeutic and imaging agents (multifunctional system). In the initial stage, liposome systems mainly relied on the EPR effect, known as passive targeting. FDA-approved liposomes were non-targeted liposomes which reduced side effects and improved patient tolerability over conventional anti-cancer drugs, but largely failed to improve therapeutic outcomes and patient survival rates. These outcomes are considered due to the heterogeneity in EPR conditions individually. To improve the effectiveness of therapies that rely on the EPR effects, upgraded formulations and protocols will be needed. Imagebased or assisted nanomedicine is considered to be crucial for addressing the EPR's heterogeneities, ligand expression, or tumor environments. Clinically available technology, such as CTs and MRIs, might be used to image the EPR effect in patients and provide clinicians with valuable information (e.g., vessel density, vessel permeability, fluid pressure) for medication regimens and treatment planning. Imaging the EPR variation across patients may also provide a means of identifying patients likely to respond best to liposomal therapy.

Unfortunately, EPR imaging is currently only in the developmental stage. Until now, only a few studies have reported or clinically analyzed the EPR effect in patients. Børresen et al.64 recently investigated the correlation between the degree of the EPR effect (64Cu-liposome) and tumor neoangiogenesis (68Ga-RGD), fluid pressure, glycolytic activity (18F-FDG) and diffusivity (diffusion-weighted MRI) to identify potential biomarkers suitable for prediction of the EPR effect in cancer models. The researchers determined that 64Cu-liposome and 68Ga-RGD uptake were moderately correlated, and the authors ultimately concluded only that 68Ga-RGD does not qualify as a surrogate marker, and that 18F-FDG (metabolic activity) and 64Cu-liposome uptake were not correlated. Lee et al.65 introduced 64Cu-MM-DX-929 (untargeted, no-drug PEGylated liposome) as a universal companion diagnostic agent to prospectively select patients for liposomal therapeutics. 64Cu-MM-DX-929 was injected into an animal cancer model, then tumor deposition and intra-tumoral distributions were evaluated. The mouse received a median tumor deposition score and a liposomal drug or free drug (MM-302, PLD, Liposomal irinotecan, and doxorubicin) and was treated to identify an improved treatment response. These researchers ultimately recommended a quantitative PET for longitudinal imaging as providing the best estimation of liposomal drug delivery in tumors.

Moving beyond EPR effect-based strategies, active targeting is one means of delivering drugs or theranostics to a lesion site while avoiding normal tissue. Active targeting strategies include targeting a tumor cell surface receptor or targeting a tumor micro-environment, and stimuli-response strategies that rely on changes in pH, temperature, redox, enzyme, light, and ultrasound to trigger drug release. Releasing the encapsulated drug in the target area is particularly difficult as a result of a limit in the liposome system. The aforementioned stimuli-response strategies were designed to overcome this issue.

Antibodies and their fragments are among the most studied targeting agents in preclinical and clinical trials. Antibody conjugated liposomes, called immunoliposomes, will doubtlessly play a pivotal role in precision cancer diagnosis and treatment in the future. Current clinical trials of immunoliposomes are shown in Table 3. Among these is MM-302, a dox-loaded immunoliposome (ILs-Dox) that targets the human epithermal growth factor receptor-2 (HER-2) currently in Phase I clinical trials to determine the maximum tolerable dose. In this study, Lee et al.66 administered 64Cu-labeled MM-302 and employed hybrid PET/CT to visualize tumor accumulation and therapeutic response in HER2-positive metastatic breast cancer patients. High accumulated activity tumor lesions showed favorable treatment efficacy and were well correlated with preclinical data.

A deeper knowledge of the molecular events associated with cancer, liposome formulation techniques, and image interpretation techniques will facilitate the development of future theranostic systems.

5. Future research directions

Today, liposomal vesicles are among the most effective delivery options for various classes of drugs designed to treat everything from cancer to pain. Table 7 lists FDA-approved liposomal drugs that were not discussed in this paper. While this list in long, serious problems remain for conventional liposomes as they are used in biomedical applications, including the storage stability problems caused by lipid oxidation and hydrolysis, drug leakage from the vesicle, rapid clearance during blood circulation, and the absence of cancer cell-specific targeting.51 To survive on the market, pharmaceutical drugs must maintain their properties during storage, a problem that has attracted considerable attention by researchers. Liposomes in an aqueous state can be degraded by oxidation and hydrolysis, which can cause structural changes such as liposome sedimentation, aggregation, or fusion. To remediate these problems, researchers have tried freeze-drying, adding surfactants or antioxidants, modifying with chitosan, and changing their composition to improve rigidity.67 Despite these efforts, the challenge of ensuring stability remains unsolved. Stability can be influenced by pH, size, surface charge, lipid composition, and temperature, and these parameters also impact the drug encapsulation efficiency and the half-life of blood circulation in vivo.

TABLE 7
Clinically used various liposomal drugs

To improve therapeutic efficacy, researches have concentrated on developing multi-functional liposomes, capable of long circulation, increase accumulation at the target site, and increased cellular internalization. Various strategies have been adopted to achieve this, including strategies that rely on passive or active targeting. In passive targeting through EPR, several factors, including lipid composition, charge, size, individual heterogeneity of vascular condition, can influence. In order to improve EPR mediated therapy, gaining EPR relevant biomarkers through physical modalities is useful.

Active targeting strategies include targeting a tumor cell's surface receptors, targeting a tumor's micro-environment, and stimuli-response strategies including pH-, temperature-, redox-, enzyme-, light-, and ultrasound-triggered drug release.25 At this time, ThermoDox is the only thermosensitive liposomal formulation (encapsulated doxorubicin), and it is currently in phase 3 of its clinical trials,68 in which it has been provided to patients with hepatocellular carcinoma by radiofrequency ablation. Coupling targeting ligands to surface features such as proteins, low molecular weight ligands including peptides, carbohydrates, or monoclonal antibodies or their fragments has been studied, and VEGF, vascular cell adhesion molecule (VCAM), matrix metalloproteinases (MMP), and integrin have been evaluated as tumor environmental targets.25 Targeted therapies, however, rely on ligands presented by only a few types of tumors and have not yet overcome the problem of the heterogeneity of tumor cells and their surface markers. One possible direction for future research may be ligands coupling of different natures for more sensitive malignant lesion detection.

In addition to these developments, we will likely see increased focus by researchers on the ability to visually track both the process of a drug as it is injected the process of treatment. A focus on quantification, which is achievable through a variety of imaging instruments, will help practitioners offer precision or personalized treatments.

CONCLUSION

This paper has briefly summarized how and where liposomes are currently applied in the biomedical sciences. Liposomal drug formulations offer a means of overcoming the limitations of conventional therapies and have a wide variety of therapeutic applications, ranging from cancer to pain management. While many obstacles remain to the realization of their full potential, growing interest in the development of liposomal-based drug formulations may spur the development of the next generation of liposomes as drug carriers, and result in significant improvements to the quality of life of patients.

Notes

CONFLICT OF INTEREST STATEMENT:None declared.

ACKNOWLEDGEMENTS

This paper was supported by a Fund of Biomedical Research Institute, Jeonbuk National University Hospital (2019).

References

    1. Mohanty A, Uthaman S, Park IK. Utilization of polymer-lipid hybrid nanoparticles for targeted anti-cancer therapy. Molecules 2020;25:4377
    1. Yuba E. Development of functional liposomes by modification of stimuli-responsive materials and their biomedical applications. J Mater Chem B 2020;8:1093–1107.
    1. Bozzuto G, Molinari A. Liposomes as nanomedical devices. Int J Nanomedicine 2015;10:975–999.
    1. Beltrán-Gracia E, López-Camacho A, Higuera-Ciapara I, VelázquezFernández JB, Vallejo-Cardona AA. Nanomedicine review: clinical developments in liposomal applications. Cancer Nanotechnol 2019;10:11
    1. He H, Lu Y, Qi J, Zhu Q, Chen Z, Wu W. Adapting liposomes for oral drug delivery. Acta Pharm Sin B 2019;9:36–48.
    1. Peng Y, Bariwal J, Kumar V, Tan C, Mahato RI. Organic nanocarriers for delivery and targeting of therapeutic agents for cancer treatment. Adv Ther (Weinh) 2020;3:1900136
    1. Akbarzadeh A, Rezaei-Sadabady R, Davaran S, Joo SW, Zarghami N, Hanifehpour Y, et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett 2013;8:102
    1. Maja L, Željko K, Mateja P. Sustainable technologies for liposome preparation. J Supercrit Fluids 2020;165:104984
    1. Olusanya TOB, Haj Ahmad RR, Ibegbu DM, Smith JR, Elkordy AA. Liposomal drug delivery systems and anticancer drugs. Molecules 2018;23:907
    1. Panahi Y, Farshbaf M, Mohammadhosseini M, Mirahadi M, Khalilov R, Saghfi S, et al. Recent advances on liposomal nanoparticles: synthesis, characterization and biomedical applications. Artif Cells Nanomed Biotechnol 2017;45:788–799.
    1. Tikshdeep C, Sonia A, Bharat P, Abhishek C. Liposome drug delivery: a review. Int J Pharm Chem Sci 2012;1:1103–1113.
    1. Gonzalez Gomez A, Hosseinidoust Z. Liposomes for antibiotic encapsulation and delivery. ACS Infect Dis 2020;6:896–908.
    1. Sur S, Fries AC, Kinzler KW, Zhou S, Vogelstein B. Remote loading of preencapsulated drugs into stealth liposomes. Proc Natl Acad Sci U S A 2014;111:2283–2288.
    1. Bulbake U, Doppalapudi S, Kommineni N, Khan W. Liposomal formulations in clinical use: an updated review. Pharmaceutics 2017;9:12
    1. Gubernator J. Active methods of drug loading into liposomes: recent strategies for stable drug entrapment and increased in vivo activity. Expert Opin Drug Deliv 2011;8:565–580.
    1. Vakili-Ghartavol R, Rezayat SM, Faridi-Majidi R, Sadri K, Jaafari MR. Optimization of docetaxel loading conditions in liposomes: proposing potential products for metastatic breast carcinoma chemotherapy. Sci Rep 2020;10:5569
    1. Yingchoncharoen P, Kalinowski DS, Richardson DR. Lipid-based drug delivery systems in cancer therapy: what is available and what is yet to come. Pharmacol Rev 2016;68:701–787.
    1. Massing U, Fuxius S. Liposomal formulations of anticancer drugs: selectivity and effectiveness. Drug Resist Updat 2000;3:171–177.
    1. Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and challenges of liposome assisted drug delivery. Front Pharmacol 2015;6:286
    1. Bobo D, Robinson KJ, Islam J, Thurecht KJ, Corrie SR. Nanoparticle-based medicines: a review of FDA-approved materials and clinical trials to date. Pharm Res 2016;33:2373–2387.
    1. Wicki A, Witzigmann D, Balasubramanian V, Huwyler J. Nanomedicine in cancer therapy: challenges, opportunities, and clinical applications. J Control Release 2015;200:138–157.
    1. Anselmo AC, Mitragotri S. Nanoparticles in the clinic: an update. Bioeng Transl Med 2019;4:e10143
    1. Abu Lila AS, Ishida T. Liposomal delivery systems: design optimization and current applications. Biol Pharm Bull 2017;40:1–10.
    1. Mufamadi MS, Pillay V, Choonara YE, Du Toit LC, Modi G, Naidoo D, et al. A review on composite liposomal technologies for specialized drug delivery. J Drug Deliv 2011;2011:939851
    1. Deshpande PP, Biswas S, Torchilin VP. Current trends in the use of liposomes for tumor targeting. Nanomedicine (Lond) 2013;8:1509–1528.
    1. Kumar L, Verma S, Bhardwaj A, Vaidya S, Vaidya B. Eradication of superficial fungal infections by conventional and novel approaches: a comprehensive review. Artif Cells Nanomed Biotechnol 2014;42:32–46.
    1. Waghule T, Sankar S, Rapalli VK, Gorantla S, Dubey SK, Chellappan DK, et al. Emerging role of nanocarriers based topical delivery of anti-fungal agents in combating growing fungal infections. Dermatol Ther 2020;33:e13905
    1. Faustino C, Pinheiro L. Lipid systems for the delivery of amphotericin B in antifungal therapy. Pharmaceutics 2020;12:29
    1. Tollemar J, Klingspor L, Ringdén O. Liposomal amphotericin B (AmBisome) for fungal infections in immunocompromised adults and children. Clin Microbiol Infect 2001;7 Suppl 2:68–79.
    1. Roemer T, Krysan DJ. Antifungal drug development: challenges, unmet clinical needs, and new approaches. Cold Spring Harb Perspect Med 2014;4:a019703
    1. Svahn KS, Chryssanthou E, Olsen B, Bohlin L, Göransson U. Penicillium nalgiovense Laxa isolated from Antarctica is a new source of the antifungal metabolite amphotericin B. Fungal Biol Biotechnol 2015;2:1
    1. Adler-Moore J, Lewis RE, Brüggemann RJM, Rijnders BJA, Groll AH, Walsh TJ. Preclinical safety, tolerability, pharmacokinetics, pharmacodynamics, and antifungal activity of liposomal amphotericin B. Clin Infect Dis 2019;68 Suppl 4:S244–S259.
    1. Soo Hoo L. Fungal fatal attraction: a mechanistic review on targeting liposomal amphotericin B (AmBisome®) to the fungal membrane. J Liposome Res 2017;27:180–185.
    1. Cuddihy G, Wasan EK, Di Y, Wasan KM. The development of oral amphotericin B to treat systemic fungal and parasitic infections: has the myth been finally realized? Pharmaceutics 2019;11:99
    1. Bassetti M, Vena A, Russo A, Peghin M. Inhaled liposomal antimicrobial delivery in lung infections. Drugs 2020;80:1309–1318.
    1. Dong D, Thomas N, Thierry B, Vreugde S, Prestidge CA, Wormald PJ. Distribution and inhibition of liposomes on Staphylococcus aureus and Pseudomonas aeruginosa biofilm. PLoS One 2015;10:e0131806
    1. Rukavina Z, Vanić Ž. Current trends in development of liposomes for targeting bacterial biofilms. Pharmaceutics 2016;8:18
    1. Forier K, Raemdonck K, De Smedt SC, Demeester J, Coenye T, Braeckmans K. Lipid and polymer nanoparticles for drug delivery to bacterial biofilms. J Control Release 2014;190:607–623.
    1. Gao W, Chen Y, Zhang Y, Zhang Q, Zhang L. Nanoparticle-based local antimicrobial drug delivery. Adv Drug Deliv Rev 2018;127:46–57.
    1. Shirley M. Amikacin liposome inhalation suspension: a review in Mycobacterium avium complex lung disease. Drugs 2019;79:555–562.
    1. Waters V, Ratjen F. Inhaled liposomal amikacin. Expert Rev Respir Med 2014;8:401–409.
    1. Wang N, Chen M, Wang T. Liposomes used as a vaccine adjuvant-delivery system: from basics to clinical immunization. J Control Release 2019;303:130–150.
    1. Schwendener RA. Liposomes as vaccine delivery systems: a review of the recent advances. Ther Adv Vaccines 2014;2:159–182.
    1. Xing H, Hwang K, Lu Y. Recent developments of liposomes as nanocarriers for theranostic applications. Theranostics 2016;6:1336–1352.
    1. Fan Y, Sahdev P, Ochyl LJ, Akerberg J, Moon JJ. Cationic liposome-hyaluronic acid hybrid nanoparticles for intranasal vaccination with subunit antigens. J Control Release 2015;208:121–129.
    1. Sharma M, Dube T, Chibh S, Kour A, Mishra J, Panda JJ. Nanotheranostics, a future remedy of neurological disorders. Expert Opin Drug Deliv 2019;16:113–128.
    1. Gupta MK, Lee Y, Boire TC, Lee JB, Kim WS, Sung HJ. Recent strategies to design vascular theranostic nanoparticles. Nanotheranostics 2017;1:166–177.
    1. Kargozar S, Baino F, Hamzehlou S, Hamblin MR, Mozafari M. Nanotechnology for angiogenesis: opportunities and challenges. Chem Soc Rev 2020;49:5008–5057.
    1. Sun Z, Huang P, Tong G, Lin J, Jin A, Rong P, et al. VEGF-loaded graphene oxide as theranostics for multi-modality imaging-monitored targeting therapeutic angiogenesis of ischemic muscle. Nanoscale 2013;5:6857–6866.
    1. Kim J, Cao L, Shvartsman D, Silva EA, Mooney DJ. Targeted delivery of nanoparticles to ischemic muscle for imaging and therapeutic angiogenesis. Nano Lett 2011;11:694–700.
    1. O'Dwyer J, Murphy R, Dolan EB, Kovarova L, Pravda M, Velebny V, et al. Development of a nanomedicine-loaded hydrogel for sustained delivery of an angiogenic growth factor to the ischaemic myocardium. Drug Deliv Transl Res 2020;10:440–454.
    1. Yajima S, Miyagawa S, Fukushima S, Sakai Y, Iseoka H, Harada A, et al. Prostacyclin analogue-loaded nanoparticles attenuate myocardial ischemia/reperfusion injury in rats. JACC Basic Transl Sci 2019;4:318–331.
    1. Oduk Y, Zhu W, Kannappan R, Zhao M, Borovjagin AV, Oparil S, et al. VEGF nanoparticles repair the heart after myocardial infarction. Am J Physiol Heart Circ Physiol 2018;314:H278–H284.
    1. Bietenbeck M, Engel S, Lamping S, Hansen U, Faber C, Ravoo BJ, et al. Functionalization of clinically approved MRI contrast agents for the delivery of VEGF. Bioconjug Chem 2019;30:1042–1047.
    1. Nam M, Lee J, Lee KY, Kim J. Sequential targeted delivery of liposomes to ischemic tissues by controlling blood vessel permeability. ACS Biomater Sci Eng 2018;4:532–538.
    1. Hwang H, Jeong HS, Oh PS, Na KS, Kwon J, Kim J, et al. Improving cerebral blood flow through liposomal delivery of angiogenic peptides: potential of 18F-FDG PET imaging in ischemic stroke treatment. J Nucl Med 2015;56:1106–1111.
    1. Hwang H, Kim HS, Jeong HS, Rajasaheb BT, Kim M, Oh PS, et al. Liposomal angiogenic peptides for ischemic limb perfusion: comparative study between different administration methods. Drug Deliv 2016;23:3619–3628.
    1. Hwang H, Jeong HS, Oh PS, Kim M, Lee TK, Kwon J, et al. PEGylated nanoliposomes encapsulating angiogenic peptides improve perfusion defects: radionuclide imaging-based study. Nucl Med Biol 2016;43:552–558.
    1. Degrauwe N, Hocquelet A, Digklia A, Schaefer N, Denys A, Duran R. Theranostics in interventional oncology: versatile carriers for diagnosis and targeted image-guided minimally invasive procedures. Front Pharmacol 2019;10:450
    1. Xie J, Lee S, Chen X. Nanoparticle-based theranostic agents. Adv Drug Deliv Rev 2010;62:1064–1079.
    1. Man F, Gawne PJ, T M. Nuclear imaging of liposomal drug delivery systems: a critical review of radiolabelling methods and applications in nanomedicine. Adv Drug Deliv Rev 2019;143:134–160.
    1. Farzin L, Sheibani S, Moassesi ME, Shamsipur M. An overview of nanoscale radionuclides and radiolabeled nanomaterials commonly used for nuclear molecular imaging and therapeutic functions. J Biomed Mater Res A 2019;107:251–285.
    1. Peltek OO, Muslimov AR, Zyuzin MV, Timin AS. Current outlook on radionuclide delivery systems: from design consideration to translation into clinics. J Nanobiotechnology 2019;17:90
    1. Børresen B, Hansen AE, Fliedner FP, Henriksen JR, Elema DR, Brandt-Larsen M, et al. Noninvasive molecular imaging of the enhanced permeability and retention effect by 64Cu-liposomes: in vivo correlations with 68Ga-RGD, fluid pressure, diffusivity and 18F-FDG. Int J Nanomedicine 2020;15:8571–8581.
    1. Lee H, Gaddy D, Ventura M, Bernards N, de Souza R, Kirpotin D, et al. Companion diagnostic 64Cu-liposome positron emission tomography enables characterization of drug delivery to tumors and predicts response to cancer nanomedicines. Theranostics 2018;8:2300–2312.
    1. Lee H, Shields AF, Siegel BA, Miller KD, Krop I, Ma CX, et al. 64Cu-MM-302 positron emission tomography quantifies variability of enhanced permeability and retention of nanoparticles in relation to treatment response in patients with metastatic breast cancer. Clin Cancer Res 2017;23:4190–4202.
    1. Franzé S, Selmin F, Samaritani E, Minghetti P, Cilurzo F. Lyophilization of liposomal formulations: still necessary, still challenging. Pharmaceutics 2018;10:139
    1. Besse HC, Barten-van Rijbroek AD, van der Wurff-Jacobs KMG, Bos C, Moonen CTW, Deckers R. Tumor drug distribution after local drug delivery by hyperthermia, in vivo. Cancers (Basel) 2019;11:1512

Metrics
Share
Figures

1 / 2

Tables

1 / 7

PERMALINK