Recent advances in inhalable liposomes for treatment of pulmonary diseases: Concept to clinical stance

Recent advances in inhalable liposomes for treatment of pulmonary diseases: Concept to clinical stance

Journal Pre-proof Recent advances in inhalable liposomes for treatment of pulmonary diseases: Concept to clinical stance Piyush P. Mehta, Debjit Ghosh...

3MB Sizes 0 Downloads 3 Views

Journal Pre-proof Recent advances in inhalable liposomes for treatment of pulmonary diseases: Concept to clinical stance Piyush P. Mehta, Debjit Ghoshal, Atmaram P. Pawar, Shivajirao S. Kadam, Vividha S. Dhapte-Pawar PII:

S1773-2247(19)31859-3

DOI:

https://doi.org/10.1016/j.jddst.2020.101509

Reference:

JDDST 101509

To appear in:

Journal of Drug Delivery Science and Technology

Received Date: 1 December 2019 Revised Date:

28 December 2019

Accepted Date: 7 January 2020

Please cite this article as: P.P. Mehta, D. Ghoshal, A.P. Pawar, S.S. Kadam, V.S. Dhapte-Pawar, Recent advances in inhalable liposomes for treatment of pulmonary diseases: Concept to clinical stance, Journal of Drug Delivery Science and Technology (2020), doi: https://doi.org/10.1016/ j.jddst.2020.101509. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2020 Published by Elsevier B.V.

Recent advances in inhalable liposomes for treatment of pulmonary diseases: Concept to Clinical Stance

Piyush P. Mehta 1, Debjit Ghoshal 2, Atmaram P. Pawar 2, Shivajirao S. Kadam 3 and Vividha S Dhapte-Pawar 2*

1

Department of Quality Assurance, Poona College of Pharmacy, Bharati Vidyapeeth Deemed

University, Pune - 38, Maharashtra, India. 2

Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth Deemed

University, Pune - 38, Maharashtra, India. 3

Bharati Vidyapeeth Bhavan, Bharati Vidyapeeth Deemed University, LBS Road, Pune- 30,

Maharashtra, India.

*Corresponding author Dr. Vividha S Dhapte-Pawar, Associate Professor, Department of Pharmaceutics, Bharati Vidyapeeth University, Poona College of Pharmacy, Erandwane, Kothrud, Pune 411038. Email: [email protected]; [email protected]

1

Recent advances in inhalable liposomes for treatment of pulmonary diseases: Concept

2

to Clinical Stance

3 4

Graphical Abstract

5

6 7 8 9 10 11 12 13 14 15 16 17 1|Page

18

Recent advances in inhalable liposomes for treatment of pulmonary diseases: Concept

19

to Clinical Stance

20 21

Abstract

22

Liposomes and liposomes-based drug delivery systems are promising carriers in the rapidly

23

evolving field of nanomedicine. Liposomes are receiving growing attention in the scientific

24

domain owing to their distinctive structural characteristics, physiological features and

25

biological properties. These versatile lipid vesicles represent unique platforms for drug

26

delivery, targeting, diagnostics, imaging as well as theranostics. The main objective of the

27

present article is to present insights into the pulmonary delivery of these versatile cargos for

28

nanomedicine. Inhalable liposomes for pulmonary delivery present unique advantages as they

29

are formulated from phospholipids similar to endogenous pulmonary surfactant. The article

30

begins by unfolding the important background information about liposomes and their key

31

advantages as nanomaterials. In the subsequent section, various liposomes and proliposome

32

formulations are summarized with a special emphasis on their physicochemical properties,

33

aerosolization performance and in-vivo aerodynamic behavior. Additionally, this article

34

includes a segment devoted to recent status on clinical trials of liposomal formulations for

35

treating pulmonary diseases. Moreover, the present article contains a section dedicated to the

36

market potential and scientific challenges related to inhalable liposomes. In summary, this

37

article is a comprehensive report of inhalable liposomes to appear in recent years.

38 39

Keywords

40

Liposomes, proliposome, nanomedicine, nanocarriers, dry powder inhalers, aerodynamic

41

behavior, drug delivery

42

2|Page

43

Abbreviations

44

Chronic obstructive pulmonary disease (COPD); world health organization (WHO);

45

pressurized metered-dose inhalers (pMDIs); soft mist inhalers (SMI); dry powder inhalers

46

(DPI); novel drug delivery systems (NDDS); small unilamellar vesicles (SUVs); large

47

unilamellar vesicles (LUVs); giant unilamellar vesicles (GUVs); multilamellar vesicles

48

(MLV); multi-vesicular vesicles (MVV); double emulsion templating (DET); microfluidic

49

hydrodynamic focusing (MHF); reticuloendothelial system (RES); polyethylene glycol

50

(PEG); enhanced permeability and retention (EPR); solid lipid nanoparticles (SLNs);

51

nanostructured lipid carriers (NLCs); nanoparticles embedded microparticles (NEMs);

52

encapsulation

53

bromide (MTT); half-maximal inhibitory concentration (IC50); folate receptor alpha

54

(SPCA1); hydrogenated soy phosphatidylcholine (HSPC); 1,2-Distearoyl-sn-glycero-3-

55

phosphoglycerol (DSPG); fine particle fraction (FPF); emitted dose (ED); mass median

56

aerodynamic diameter (MMAD); human lung adenocarcinoma (A549); maximum tolerated

57

dose (MTD); area under curve (AUC); mean residence time (MRT); 1,2-dioleoyl-sn-glycero-

58

3-[phospho-L-serine] (DOPS); twin stage impinger (TSI); maximum drug concentration

59

(Cmax); 4-aminophenyl-alpha-D-manno-pyranoside (PAM); next generation impactor (NGI);

60

3-methylcholanthrene (MCA); diethyl nitrosamine (DEN); tumor necrosis factor-α (TNF-α);

61

vascular endothelial growth factor (VEGF); malondialdehyde (MDA); caspase-3 and B-cell

62

lymphoma 2 protein (BCL-2); 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES);

63

nuclear factor-κB (NF-κB); geometric standard deviation (GSD); idiopathic pulmonary

64

fibrosis (IPF); dipalmitoylphosphatidylcholine (DPPC); phosphatidylcholines (PC); the phase

65

transition temperature (Tm); normal human bronchial epithelial cells (NHBE); small airway

66

epithelial cells (SAEC); alveolar macrophages (AMs); enzyme linked immunosorbent assay

67

(ELISA); human lung cancer cell line cells (Calu-3); human alveolar basal epithelial cell

3|Page

efficiency

(EE);

3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium

68

(NR8383); chemistry, manufacturing and control (CMC); forced vital capacity (FVC); forced

69

expiratory volume (FEV); Tiffeneau-Pinelli index (FEV1/FVC); maximal voluntary

70

ventilation (MVV).

71 72

1. Introduction

73

Even with the continuous efforts and progress made in pulmonary research, pulmonary

74

ailments still remain a major health issue across the globe. Pulmonary diseases are leading

75

causes of morbidity and mortality worldwide [1]. Some of the most prevalent pulmonary

76

ailments are asthma, chronic obstructive pulmonary disease (COPD), pulmonary tuberculosis,

77

lung cancer, pulmonary hypertension, chronic bronchitis, emphysema and occupational lung

78

diseases. COPD is not one single disease but an umbrella phrase commonly utilized to

79

express diverse chronic lung disorders that cause limitations in pulmonary airflow [2].

80

Almost 65 million individuals suffer from COPD and more than 3 million fatalities are

81

reported annually making it the third major cause of mortality worldwide. According to the

82

World Health Organization (WHO) statistics, approximately 235 million people suffer from

83

asthma and it is the most ordinary pulmonary disease among the paediatric population [3].

84

Moreover, the family, not just the individual, suffer the impact and crisis of pulmonary

85

diseases. In brief, pulmonary diseases are prevalent in every part of the world; this public

86

health problem is just not limited to 2nd and 3rd income countries but affects all countries

87

irrespective of their developmental levels. In view of the impact of pulmonary diseases on a

88

universal scale, it is apparent that these ailments have raised a significant public health

89

challenge [4].

90 91

Various pulmonary delivery systems such as pressurized metered-dose inhalers (pMDIs), soft

92

mist inhalers (SMI), dry powder inhalers (DPI) and nebulizers have been designed, developed

4|Page

93

and studied for the treatment of pulmonary diseases. Among them, DPIs are more preferred

94

dosage forms because of their better physicochemical stability and capability to deliver the

95

drug into deep lungs using the patient’s respiration [5,6]. Each type of delivery system has

96

distinct strengths and weaknesses considering the disease pathophysiology, severity and type

97

of prescribed formulation. Furthermore, it has been well reported that the therapeutic success

98

of inhalation therapy is highly dependent on particle size distribution, inhalation flow rate,

99

inhaler device resistance and dispersion capability [7]. Therefore, to maximize the clinical

100

outcome of inhalation therapy, a formulation with apt physicochemical properties is vital [8].

101 102

With the continuous growth of various scientific fields such as particle engineering, materials

103

science, biotechnology, molecular biology, green chemistry and biomaterials, rapid

104

advancement has been achieved in the domain of novel drug delivery systems (NDDS) [9].In

105

addition, recent progress in nanotechnology has opened avenues for augmenting the clinical

106

value of inhalation therapy for different pulmonary diseases [10]. The application of

107

nanotechnology to the design of NDDS such as polymeric systems (polymer-drug conjugates,

108

polymeric nanoparticles, polymeric micelles) [11], nanoparticles [12,13], nanoaggregates,

109

nanocomposites [14], dendrimers, quantum dots, fullerenes, novel lipid vesicles (liposomes,

110

pro-liposomes, solid lipid nanoparticles, nanostructured lipid carriers, cochleates and lipidic

111

spherulites), ethosomes, cubosomes, lipomers, polymeric microparticles and microspheres

112

[15] have illustrated numerous therapeutic advantages over traditional pulmonary

113

formulations [9,16]. The design and development of NDDS based DPIs have the potential to

114

surpass issues associated with the carrier as a critical formulation component. These novel

115

formulations help in improving physicochemical stability, tissue distribution, in-vivo drug

116

deposition and bioavailability [11,17]. To sum up, such novel formulations represent

117

promising alternatives to traditional inhaled formulations.

5|Page

118 119

Among various novel bioactive platforms, lipid vesicles i.e. liposomes have attracted the

120

focus of formulation scientists as they can be prepared from substances endogenous to the

121

pulmonary airways as components of the lung with surfactant and other unique

122

physicochemical and biopharmaceutical properties [18]. The application of liposomes in drug

123

delivery has been studied and investigated by numerous formulation scientists [19]. Several

124

active scientists and clinicians have studied the significance of liposomes systems for drug

125

delivery, drug targeting, tissue engineering, diagnostics, detection of genetic material and

126

imaging [20-22]. In brief, the superior therapeutic efficacy of liposomes has been evidenced

127

either in laboratory investigations or in clinical analysis, particularly in the therapy of

128

pulmonary diseases.

129 130

By knowing this continues development, in this article, we have analyzed inhalable liposomal

131

formulations for the treatment of numerous pulmonary ailments. In the opening section of

132

this article, we have discussed liposomes, generations of liposomes and the importance of

133

liposomes-based nanomaterials. In the next segment, various dry powdered liposomal

134

formulations are summarized with a special emphasis on their physicochemical properties,

135

aerosolization performance and in-vivo behavior. In the third part, the importance of

136

respirable pro-liposome formulations and their influence on pulmonary drug delivery are

137

thoroughly summarized. Additionally, this article includes a section devoted to recent efforts

138

taken in clinical trials of liposomal formulations for the treatment of pulmonary diseases.

139

Moreover, this article concisely explores the market potential of inhaled liposomal

140

formulations. Furthermore, the present article contains a segment dedicated to the practical

141

concerns, technical limitations and scientific challenges related to inhalable liposomes. This

6|Page

142

review can be of potential significance from the perspective of both academicians and

143

industrial scientists.

144 145

2. Methodology

146

We performed a broad literature search for liposomes and pro-liposomes for pulmonary drug

147

delivery. This literature search included scientific journals, books and book chapters from

148

various electronic sources such as ScienceDirect, PubMed, Google Scholar, Springer and

149

Web of Science. Present article investigates various key aspects associated to inhalable

150

liposomes and pro-liposomes with special importance on the prepration methods, in-vitro

151

aerodynamic performance and pulmokinetic parameters. References enlisted in present article

152

contains 130 articles, containing peered review articles, original research articles, clinical trial

153

reports and book chapters. All articles were examined, screened cautiously and then selected

154

for the review. ChemDraw software (Version 12.2; Perkin Elmer) was used to draw the

155

figures.

156 157

3. Liposomes

158

Liposomes perhaps are the most extensively explored and characterized lipid-based drug

159

delivery systems. Liposomes are the sub-micron spherical phospholipid vesicles consisting of

160

single or multiple concentric lipid bilayers enclosing aqueous interior [23]. Liposomes were

161

first discovered and explained in 1965 by Bangham and his co-workers. Since then, they are

162

extensively studied as a drug reservoir from the last five to six decades [24]. Accordingly,

163

liposomes have become one of the most explored drug reservoirs in diagnosis, treatment and

164

imaging of several diseases [25]. Generally, on basis of vesicular arrangement liposome can

165

be classified as unilamellar liposomes (single bilayer lipid membrane) i.e. small unilamellar

166

vesicles (SUVs; 20-100 nm); large unilamellar vesicles (LUVs; 100 nm -1 mm) and giant

7|Page

167

unilamellar vesicles (GUVs; > 1 mm) or multilamellar liposomes (numerous bilayer lipid

168

membranes)

169

MVV is also known as vesosomes (Fig. 1) [26]. Phospholipids and cholesterol are the main

170

two components of liposomal bilayers. Cholesterol plays a vital role in the physical stability

171

of liposome vesicles with its ability to increase the phospholipid phase transition temperature

172

(TM) [27,28]. Many techniques have been explored for the design and development of

173

liposomes (Table1). The most popular techniques that have been explored to develop

174

liposomes are the thin-film (Bangham method) and the ethanol injection method [29]. Each

175

type of technique has its own unique strengths and weaknesses considering the type of lipid

176

and the nature of drugs that can be utilized. On account of this, many formulation scientists

177

are now actively engaged in developing strategies to improve the physicochemical quality of

178

the liposomes. Microfluidic technology, membrane contactor, electrospray mechanism are a

179

helpful alternative for the fabrication of liposomes [29]. Moreover, new techniques derived

180

from microfluidic approaches for fabrication of liposome mainly include double emulsion

181

templating (DET) [30], electro formation and hydration [31], pulsed jetting [32], ice droplet

182

hydration [33], extrusion [34], transient membrane ejection [35], droplet emulsion transfer

183

[36] and microfluidic hydrodynamic focusing (MHF) [37]. Among these methods, templating

184

is a good option to fabricate liposomes of consistent size and high encapsulation efficiency

185

[29].

i.e. multilamellar vesicles (MLV) or multi-vesicular vesicles (MVV) [26].

186 187

Additionally, based on structural modifications, liposomes can be classified into the different

188

‘generations’. The first generation liposomes mainly consist of phospholipids and cholesterol

189

vesicles without any structural modifications. The mean particle size of first-generation

190

liposomes is in the range of 50 to 450 nm [48]. As a nanoscale drug reservoir, the liposomes

191

are biodegradable, biocompatible with low toxicity. As well, the liposomes are also

8|Page

192

convenient considering the scale-up and the quality control specifications [49]. Liposomes

193

are capable to hold both, lipophilic as well as hydrophilic molecules. Thus, a wide range of

194

therapeutic compounds can be easily incorporated into the liposomes [50]. Various synthetic

195

drugs, semi-synthetic compounds, herbs, herb extracts and phytoconstituents can be easily

196

incorporated into the liposomes. Ambisome® (Astellas Pharma), Amphotec® (Intermune), and

197

Abelcet® (Enzon) [amphotericin B liposomes] are few successful, commercial liposomal

198

products approved for the treatment of fungal infections across the globe [51]. Yet, these

199

first-generation liposomes displayed noticeable limitations such as leakage of drugs from

200

liposome vesicles, low loading capacity for hydrophilic compounds and rapid clearance of

201

vesicles by the reticuloendothelial system (RES) in the systemic circulation. Therefore, the

202

first generation liposomes were almost abandoned until the active drug loading and sterically

203

stable second-generation liposomes were discovered [52].

204 205

The second-generation liposomes are long-circulating, surface modified liposomes. Usually,

206

liposomes are coated with inert polymers i.e. polysaccharides, oligosaccharides,

207

glycoproteins and synthetic polymers [53]. Among these polymers, polyethylene glycol

208

(PEG) is most commonly explored to stabilize the liposomes. PEG-modified (PEG-coated)

209

liposomes are often denoted as sterically stabilized or ‘stealth liposomesʼ [54]. Several active

210

drug loading techniques have been explored to achieve higher drug encapsulation efficiency.

211

The active drug loading techniques are distant encapsulation methods that load the drug into

212

the liposomes by the differential chemical gradient across the membrane of the liposomes.

213

The chemical gradients mainly comprise the pH of the system, ammonium sulfate gradient or

214

calcium acetate gradient. As an effect, the liposomes exhibited favourable physicochemical

215

and biological properties such as reduced drug leakage, prolonged systemic circulation,

216

modified release pattern and altered bio-distribution. The prolonged systemic circulation is

9|Page

217

mainly ascribed to the steric hindrance effect presented by a hydrophilic polymer which

218

could avoid the surface-modified liposomes from being rapidly purged by the RES [53].

219

Consequently, a number of surface-modified liposomes were studied successfully and few of

220

them are under various phases of clinical trials. Few pegylated doxorubicin liposomes such as

221

Doxil® (PEG 2000 surface-modified liposome), Caelyx® (liposomes with surface-bound

222

methoxy-polyethylene glycol) and Myocet® (liposome-encapsulated doxorubicin citrate

223

complex) have been approved and marketed for the treatment of cancers [55]. Even though

224

great commercial achievements have been made, the second generation liposomes

225

demonstrated few downsides such as poor cancer cells selectivity and low cellular uptake

226

[56].

227 228

The targeting liposomes are known as the third generation liposomes wherein the targeting

229

ligand molecules or functional moieties are incorporated into liposomes to target a specific

230

site. The third generation liposomes may possess passive as well as active targeting

231

properties. Both of the first and second generation liposomes fit into the passive targeting

232

reservoirs with high lymphatic affinity attributed to the lipophilic casing of liposomes. These

233

first and second-generation liposomes displayed the enhanced accumulation at tumor site

234

owing to the enhanced permeability and retention (EPR) consequence [57]. Active targeting

235

liposomes can target cells and/or cellular organelles by a precise interaction between a

236

targeting ligand attached to the liposomes and a cell receptor [58]. Additionally, the diverse

237

class of chemical moieties such as carbohydrates, vitamins, monoclonal antibodies, peptides,

238

proteins and aptamers can be used as targeting ligands [59]. The enhanced therapeutic

239

efficacy of third-generation liposomes had been verified in the laboratory model particularly

240

in the treatment of cancer [56]. Nowadays, a novel dual-functional liposomes have appeared

241

as a promising drug delivery system. Dual-functional liposomes are typically formulated

10 | P a g e

242

using a mixture of phospholipid and functional material by either chemical adaptation or

243

physicochemical inclusion of functional motifs against the membrane of liposomes. Current

244

investigations showed that the dual-functional liposomes are capable of improving the

245

therapeutic efficiency by modifying drug delivery as well as the mechanism of action [56].

246 247

For the pulmonary application, liposomes offer numerous benefits (Fig. 2) [60,61]. Superior

248

tolerability of liposomes in the pulmonary airways can be guaranteed, if lipids selected are

249

biodegradable resulting in non-toxic, endogenous degradation products [60,62]. Furthermore,

250

owing to their small (nm) size, they can be easily encapsulated into particles with suitable

251

aerosolization properties, which facilitates satisfactory deep lung deposition of a molecule.

252

Additionally, liposomes adhere to the mucosal surface of the airways for a longer time

253

compared to larger particles owing to the small size. Particle size, adhesion, accumulation

254

and retention in the pulmonary airways along with controlled release characteristics of

255

liposomes can lead to improved therapeutic outcomes leading to better patient compliance

256

[60,63]. This can assist an imperative function in the therapy for chronic diseases as many of

257

the existing inhalation formulations have to be applied at least twice a day because of the

258

comparatively less duration of the drug in the pulmonary airways [62,65]. With this

259

background, various liposomal DPIs are described in the following section with special

260

emphasis on their aerodynamic behavior and clinical outcomes.

261 262

4. Pulmonary drug delivery

263

It is fascinating to believe in pulmonary therapy as a modern strategy for drug delivery but

264

this practice has been well acknowledged in most of the ancient literature and it has a strong

265

history of more than 4000 years [66]. From the therapeutic perspective, pulmonary therapy

266

represents a few attractive benefits allied with the anatomical and physiological

11 | P a g e

267

characteristics of the lung. Pulmonary therapy has a better potential of treating various

268

intrapulmonary and extrapulmonary diseases, such as asthma, COPD, cystic fibrosis,

269

pulmonary hypertension, pneumonia, cancer [67], diabetics and tuberculosis [68]. The

270

reduced invasiveness of pulmonary therapy may further improve clinical outcomes and

271

patient compliance with the treatment. Furthermore, owing to the scientific and technological

272

advances in formulations (e.g. Technosphere® particles, AerosphereTM delivery, iSPERSE™

273

platform and/or Capsugel® Zephyr™), particle replication in nonwetting templates (PRINT),

274

inkjet-printed aerogel particles and inhaler devices (e.g. Advair Diskus®, ProAir®

275

Digihaler™, Spiriva® Handihaler® and TwinCapsTM Inhaler), pulmonary therapy has become

276

more ‘patient-friendly’ and economically favourable [68,69].

277 278

Many different types of delivery systems such as nanoparticles (polymeric nanoparticles,

279

polymeric micelles), microparticles (microspheres), solid lipid nanoparticles (SLNs),

280

nanostructured lipid carriers (NLCs), polymer-drug conjugates, macromolecules (dendrimers)

281

and lipid vesicles (liposomes and proliposomes) have been designed and developed for

282

pulmonary delivery of several therapeutics. They fulfill many biopharmaceutical

283

requirements i.e. sufficient drug loading, shielding of the actives from degradation,

284

biocompatibility, biodegradability and stability during aerosolization. However, one major

285

limitation of these systems is that they can be readily exhaled from the lungs after pulmonary

286

delivery. Table 2 denotes various advantages and limitations of novel carrier-based

287

pulmonary drug delivery [70]. However, several formulation techniques, such as nano-

288

agglomeration processes, nanoparticle-rooted microparticles or nanoparticles embedded

289

microparticles (NEMs) can be utilized to form nanoparticles with appropriate aerodynamic

290

diameters for pulmonary delivery. Knowing these facts, lipid vesicles (i.e. liposomes and

291

proliposomes) for pulmonary delivery have been a popularized theme for the last two-three

12 | P a g e

292

decades [8,11]. Accordingly, the present segment discusses and analyzes the available

293

literature on inhalable liposomes and proliposomes.

294 295

4.1 Liposomes based dry powder inhalers

296

Zhu et al (2019) developed folic-acid conjugated docetaxel (1:25) liposomes (LP-DTX-FA)

297

using phosphatidylcholine and cholesterol (6:1) by thin lipid film hydration method.

298

Furthermore, obtained LP-DTX-FA were spray-dried using mannitol (2 % w/v; carrier) and

299

leucine (anti-adherent) to obtain an inhalable dry powder. Spherical shaped, smooth surface

300

spray-dried LP-DTX-FA showed mean particle size, zeta potential and encapsulation

301

efficiency (EE) of 346.80 nm, -29.30 mV and 99.50 %, respectively. During the in-vitro

302

release study, LP-DTX-FA showed sustained release (>70 %) up to 50 h in phosphate buffer

303

saline (PBS; pH 7.4). In in-vitro cytotoxicity study using 3-(4,5-dimethylthiazole-2-yl)-2,5-

304

diphenyltetrazolium bromide (MTT) assay, LP-DTX-FA showed 3.3-fold superior half-

305

maximal inhibitory concentration (IC50) as compared to LP-DTX in high expression of folate

306

receptor alpha (SPCA1) cells. Additionally, during the in-vitro uptake study, LP-DTX-FA

307

showed significantly superior DTX uptake as compared to DTX alone due to conjugated

308

folic-acid. Moreover, during the in-vivo study after intratracheal administration in Sprague

309

Dawley rats, LP-DTX-FA showed 23.39-fold higher DTX deposition within the lung as

310

compared to intravenous LP-DTX-FA at the same dose of 1 mg/kg [71].

311 312

Gemcitabine intercalated liposomes (LP-GB) were formulated using a combination of lipids

313

i.e.

314

phosphoglycerol

315

emulsification solvent evaporation method. Gemcitabine liposomal powder (LP-GB-DPI)

316

was formulated using trehalose (1:2) as a cryoprotectant in the lyophilization technique.

hydrogenated

13 | P a g e

soy

(DSPG),

phosphatidylcholine cholesterol

and

(HSPC),

1,2-Distearoyl-sn-glycero-3-

mPEG2000-DSPE (5:2:2:0.8) by W/O

317

Spherical shape smooth surface LP-GB showed particle size and negative zeta potential of

318

339.49 nm and -53.1 mV, respectively while LP-GB-DPI demonstrated particle size of 8.79

319

µm with satisfactory flow properties. LP-GB-DPI showed satisfactory performance with fine

320

particle fraction (FPF), emitted dose (ED) and mass median aerodynamic diameter (MMAD)

321

of 56.12 %, 88.99 and 3.91 microns, respectively, measured at 60 L/min. LP-GB showed a

322

pH-dependent release profile over a period of 48 h. LP-GB displayed maximum (55.93 %)

323

drug release at pH 5.5 and pH 6.8 (41.45 %) whereas 7.4 (5.61 %) minimum drug release was

324

observed. Release kinetics showed that maximum gemcitabine release from liposome was

325

obtained at endosomal pH (5.5) or cancerous tissue pH (6.5). The reduced drug release at pH

326

7.4 confirmed the stability of liposomes within lung fluid. During the in-vitro toxicity study

327

using MTT assay on human lung adenocarcinoma (A549) cell 3.44-fold improvement in IC50

328

value was observed for LP-GB-DPI as compared to free drug. Moreover, maximum tolerated

329

dose (MTD) and edema index analysis showed retention of alveolar-capillary integrity and

330

negligible infiltration around bronchioles with LP-GB-DPI as compared to drug alone at the

331

same dose. Additionally, during the in-vivo study after intratracheal administration of

332

liposomes, a marked 8.31 and 5.69-fold improvement in area under curve (AUC) and mean

333

residence time (MRT) was observed as compared to drug alone at the same dose (2 mg/kg).

334

In brief, the formulated liposomes showed promising results for the management of lung

335

cancer [72].

336 337

In another study, a respirable lyophilized recombinant secretory leukocyte protease inhibitor

338

(RSLPI) loaded liposomes (RSLPI-DOPS-LP) was formulated using 1,2-dioleoyl-sn-glycero-

339

3-[phospho-L-serine] (DOPS)/ cholesterol (7:3) by thin-film hydration method. Additionally,

340

lyophilized RSLPI-DOPS-LP was micronized in the presence of mannitol (1:10). RSLPI-

341

DOPS-LP exhibited average particle size and EE of 153.60 nm and 74.10 %, respectively

14 | P a g e

342

while lyophilized RSLPI-DOPS-LP showed mean particle size and yield of 19.2 µm and

343

86.70 %, respectively. Micronization showed marked a 12-fold reduction in particle size of

344

lyophilized RSLPI-DOPS-LP. Micronized RSLPI-DOPS-LP powder retained the stability

345

and anti-neutrophil elastase activity of RSLPI without protein degradation as evident from

346

western blot analysis. During the in-vitro aerosolization study using low resistance

347

Spinhaler® device, micronized RSLPI-DOPS-LP powder showed FPF of 38.70 % by twin

348

stage impinger (TSI). In addition, micronized RSLPI-DOPS-LP powder showed FPF and

349

MMAD of 33.30 % and 2.44 µm, respectively, using cascade assembly at a flow rate of 90

350

L/min. This micronized RSLPI-DOPS-LP powder was more stable in terms of liposome size

351

for 5 months at room temperature [73]. In another study, Tang et al (2013) developed a pro-

352

drug oseltamivir phosphate (OP) loaded liposomes (1:10) using ovelecithin and cholesterol

353

by film dispersion method followed by spray drying (LP-OP-DPI). Smooth surface, spherical

354

OP liposomes displayed mean particle size, negative zeta potential and EE of 105.90 nm,-

355

13.65 mV and 60.43 %, respectively. Corrugated nonaggregating LP-OP-DPI showed

356

average particle size and yield of 3.5 µm and 55.37 % respectively, with acceptable flow

357

properties. During the in-vitro deposition study using TSI, LP-OP-DPI showed FPF of 35.4

358

%. In the in-vitro release study LP-OP-DPI displayed sustained release pattern for 20 hr in

359

PBS (pH 7.4) while OP solution showed more than 90 % drug release within the 2 h.

360

Moreover, during the in-vivo study after endotracheal administration, LP-OP-DPI showed a

361

marked 1.14 and 1.22-fold improvement in AUC and maximum drug concentration (Cmax), of

362

oseltamivir carboxylate (OSCA) respectively as compared to orally administered OP solution

363

at the same dose (12 mg/kg). The improved clinical outcome may be attributed to the

364

transformation of OP to OSCA in pulmonary airways by carboxylesterase enzyme [74].

365

15 | P a g e

366

Moxifloxacin loaded nanoliposomes (MFX-NL) were developed by a reversed-phase

367

evaporation method using L-α-phosphatidylcholine (type X-E) and cholesterol (7:3) lipids.

368

MFX-NL was decorated with targeting ligand 4-aminophenyl-alpha-D-manno-pyranoside

369

(PAM) to enhance the uptake of (NL) by alveolar macrophages. Additionally, MFX-NL was

370

embedded into microparticles using dextran as a carrier by spray drying. Corrugated surface

371

and dimple shaped MFX-NL microparticles showed particle size, zeta potential and EE of

372

277 nm, -12.31 mV and 66.25 %, respectively. During the in-vitro aerodynamic study,

373

microparticles showed higher respirable fraction (> 75 %) using the Aerolizer® device. In the

374

in-vitro release study, microparticles showed biphasic release pattern i.e. initial burst release

375

for first 2 h (~ 50 %) followed by controlled release (~ 100 %) up to 48 h in PBS (pH 7.4).

376

Moreover, during the in-vivo deposition study, after intrapulmonary administration (Penn-

377

century®) green fluorescence-labeled microparticles showed sufficiently higher MFX

378

deposition within the alveolar macrophages as compared to the upper respiratory tract.

379

Negatively charged MFX-NL provided higher anti-tubercular activity as compared to the

380

neutral NL whereas PAM decoration was capable enough to augment MFX alveolar delivery.

381

From a pharmacological viewpoint, higher alveolar deposition is important in the treatment

382

of pulmonary tuberculosis. Developed PAM decorated MFX-NL microparticles can be

383

suitable for the treatment of pulmonary tuberculosis and other pulmonary disorders [75].

384 385

Inhalable salbutamol sulphate (SS) loaded liposomal powder (SS-LP-DPI) was formulated

386

using soybean phosphatidylcholine by vesicular phospholipid gel technique followed by

387

lyophilization using lactose (1:5) as a cryoprotectant. In addition, the lyophilised powder was

388

lubricated with 0.5 % magnesium stearate and subjected to ball milling. SS-LP-DPI showed

389

80.71 and 44.35 % EE for SS before lyophilization and after rehydration, respectively.

390

Irregular shaped microparticles showed a marked 4-fold reduction in mean particle size after

16 | P a g e

391

ball milling. In an in-vitro drug release study using a dialysis bag technique SS-LP-DPI

392

displayed sustained release profile up to SS for 24 h in deionized water. During the in-vitro

393

aerodynamic study in TSI assembly using Spinhaler® device, ball-milled SS-LP-DPI

394

exhibited 2.53-fold superior FPF as compared to lyophilized powder. The present

395

investigation highlights the localized pulmonary delivery of liposomes containing anhydrous

396

SS [76]. Honmane et al (2018), prepared and optimized SS loaded liposomal DPI using

397

soybean lecithin and cholesterol (1:1) by thin-film hydration technique followed by spray

398

drying using lactose as a carrier (SS-LM-DPI). Optimized liposomes displayed mean particle

399

size, zeta potential and entrapment efficiency of 167.2 nm, 9.74 mV and 80.68 %,

400

respectively. While SS-LM-DPI showed a mean particle size of 6.35 µm suitable for

401

pulmonary drug delivery. During the in-vitro drug deposition study SS-LM-DPI

402

demonstrated marked 8.92-fold enhancement in FPF as compared to spray dried SS at a flow

403

rate of 60 L/min using the Rotahaler® inhaler device. Moreover, during the in-vitro

404

dissolution study, SS-LM-DPI displayed a controlled release profile for SS (~ 90 %) up to 14

405

hr in PBS (pH 7.4) [77]. Ye et al (2016), designed clarithromycin liposomes using a mixture

406

of soybean phosphatidylcholine: cholesterol: clarithromycin (4:1:2 w/w) by thin lipid film

407

hydration method. Inhalable powder formulation (CLA-LP-DPI) was formulated by

408

ultrasonic spray freeze drying (SFD) techniques using a combination of cryoprotectants i.e.

409

mannitol (15 % w/v) and sucrose (5 % w/v). Spherical shaped, porous CLA-LP-DPI showed

410

high drug recovery (85 %) with suitable particle size. During the in-vitro aerodynamic study

411

at a flow rate of 100 L/min, CLA-LP-DPI showed FPF and ED of 43.82 % and 53.78 %,

412

respectively. Additionally, during a three-month stability study at 25 °C and 60 % relative

413

humidity CLA-LP-DPI didn't show any major change in EE and mean particle size [78].

414

17 | P a g e

415

Curcumin (CUR) intercalated liposomal DPI (CUR-LP-DPI) were prepared using soybean

416

lecithin: cholesterol in the ratio of 5:1 by a film hydration method followed by freeze-drying

417

technique for the treatment of lung cancer. Irregular shaped microparticles showed a mean

418

particle size of 15.02 µm. During aerodynamic assessment CUR-LP-DPI showed FPF and

419

MMAD of 46.71 % and 5.81 µm using next-generation impactor (NGI). In in-vitro

420

cytotoxicity study using normal human bronchial epithelial cell (BEAS-2B) microparticles

421

showed marked 93-fold improvement in selection indices as compared to CUR alone at the

422

same dose (100 µmol/L) while microparticles showed satisfactory in-vitro anti-cancer cell

423

effect against A549 cells. Additionally, uptake of CUR from liposomes by A549 cells was

424

noticeably superior to that of CUR alone. For the in-vivo study, the primary lung cancer

425

model was developed using 3-methylcholanthrene (MCA) and diethylnitrosamine (DEN)

426

inducing agents. After pulmonary administration, CUR-LP-DPI displayed better anti-cancer

427

potential as compared to CUR alone and gemcitabine via regulating enzymatic markers such

428

as tumor necrosis factor-α (TNF-α), vascular endothelial growth factor (VEGF),

429

malondialdehyde (MDA), caspase-3 and B-cell lymphoma 2 protein (BCL-2) [79]. Dry

430

powders of liposomal encapsulated ciprofloxacin nanocrystals (CUR-LP-NC) were fabricated

431

using a freeze-thaw method followed by spray drying. CUR-LP-NC was formulated using

432

HSPC: cholesterol (7:3) and sucrose (2 % w/w) as a cryoprotectant. Corrugated dimple

433

shaped CUR-LP-NC microparticles showed particle size of 1.92 µm. In the in-vitro

434

dissolution study, CUR-LP-NC displayed a controlled release up to 12 h in HEPES (4-(2-

435

hydroxyethyl)-1-piperazineethanesulfonic acid) buffered saline (pH 7.4). During the in-vitro

436

aerodynamic study, CUR-LP-NC displayed higher FPF (69.70 %) as compared to CUR-LP-

437

NC formulated from 0.5 and 1 % w/w sucrose using Osmohaler device® at 105 L/min. The

438

developed formulation can be apt for a once-a-daily treatment schedule [80].

439

18 | P a g e

440

Li et al (2017) developed liposomal andrographolide powder (AG-LP-DPI) using soybean

441

lecithin and cholesterol (6:1) by the solvent injection method with subsequent by freeze-

442

drying using mannitol as a cryoprotectant. AG liposomes showed mean particle size and

443

negative zeta potential of 77.91 nm and -56.13 mV, respectively. While AG-LP-DPI showed

444

irregular and rough surface particles (7.44 µm) suitable for lung deposition. During the in-

445

vitro drug deposition study AG-LP-DPI showed a marked 2.75-fold improvement in FPF as

446

compared to conventional AG-DPI at 60 L/min flow rate using Twister® inhaler device.

447

Moreover, during in-vivo bacterial (S. aureus) study intratracheal administration of AG-LP-

448

DPI (1 mg) showed satisfactory anti-bacterial action at a 10-fold lower dose as compared to

449

AG (10 mg) alone. AG-LP-DPI considerably decreased pro-inflammatory cytokines such as

450

TNF-α, IL-1 and inhibited the phosphorylation of IκB-α in the nuclear factor-κB (NF-κB)

451

pathway (Fig. 3). In summary, phytoconstituent loaded inhaled liposomes are the potential

452

therapeutic agents for pulmonary therapy [81]. In another study, Viswanathan et al (2018),

453

fabricated inhalable liquorice acetone extract loaded liposomes (L-LP-DPI) using soybean

454

phosphatidylcholine by thin-film hydration method with further freeze-drying using trehalose

455

as cryoprotectant and carrier. Unilamellar spherical shaped liposomes showed a mean particle

456

size and EE of 210 nm and 75 %, respectively. During the multistage cascade analysis at 60

457

L/min flow rate using Lupihaler® device, L-LP-DPI showed the FPF, MMAD and geometric

458

standard deviation (GSD) of 54.68 %, 4.29 µm and 1.23 respectively. The in-vivo lung

459

deposition study using the nose-only apparatus in Swiss-albino mice showed more than 46 %

460

of drug deposited by L-LP-DPI in the lungs whereas only 16% of drug retained in the lungs

461

24 h of administration. Moreover, an in-vivo pharmacodynamic study in Mycobacterium

462

tuberculosis H37Rv infected Balb/c mice L-LP-DPI showed a satisfactory reduction in

463

bacterial count in lungs and spleen. In brief, liposomal containing liquorice extract found to

464

be a useful anti-tubercular medicine alone or as an adjunct to the existing standard drugs [82].

19 | P a g e

465 466

Chennakesavulu et al (2017) formulated colchicine (CL) and budesonide (BU) loaded

467

liposomal DPI using thin layer film hydration technique followed by lyophilization for

468

effective treatment of idiopathic pulmonary fibrosis (IPF). CL and BU liposomes were

469

prepared using a combination of various lipids i.e. DPPG: SPC: COL in a ration of 3:6:1 and

470

4:5:1, respectively. CL and BU liposomal DPI using mannitol as a carrier and glycine (10 %)

471

as an anti-adherent was developed. Spherical shaped, CL liposomes showed particle size, zeta

472

potential and drug entrapment of < 100 nm, -24.7 mV and 50.94 %, respectively while BU

473

liposomes showed particle size, zeta potential and drug entrapment of < 100 nm, -36.9 mV

474

and 74.22 %, respectively. During the in-vitro aerosolization study using Rotahaler® device,

475

CL and BU liposomal DPI showed FPF of 44.45and 48.62 %, respectively at a flow rate of

476

28.3 L/min. In-vitro diffusion study for both formulations showed sustained release up to 24

477

h and followed Higuchi’s diffusion-controlled kinetic model in PBS, pH 7.4 containing SLS

478

(1 % w/v). Additionally, during in-vivo studies, in bleomycin (2.5 U/kg) induced IPF rats, CL

479

and BU combined DPI showed 1.17 and 3.53-fold reduction in hydroxyproline content and

480

myeloperoxidase activity showing a positive effect against IPF. Moreover, six-month stability

481

studies at two different conditions i.e. long term (25 °C ± 2 °C, 60 % ± 5 % RH) and

482

refrigerated conditions (2-8 °C) confirmed the stability of lyophilized BU and CL liposomes.

483

Thus, liposomal based BU and CL DPI formulations can be potentially used for the treatment

484

of IPF [83]. Apart from these investigations, various, appealing case studies of inhalable

485

liposomal DPI are summarized in Table 3.

486 487

As summarized in Table 3, liposomes were thoroughly explored and studied as carriers for

488

the inhalation delivery of synthetic drugs [76,77], herbal extracts [82], phytoconstituents,

489

[79,81] vitamins, protein and peptides for the treatment of numerous pulmonary diseases and

20 | P a g e

490

pathological conditions. They were carefully explored for various pulmonary disorders

491

ranging from asthma, COPD to lung cancer. Several well-known and novel processing

492

techniques have been explored to deliver these as dry powder. In various scientific studies,

493

after judiciously selecting phospholipid composition and aerosolization technique, liposomes

494

retain their payload, mean size and do not aggregate after aerosolization. They reveal a

495

superior pulmonary deposition, satisfactory lung retention for a prolonged period after

496

inhalation and delivery of the payload inside the cells [10].

497 498

The drug distribution pattern within the pulmonary airways is highly depended on the size of

499

the aerosolized liposomes rather than vesicle size or type. Among the inhaled liposome

500

formulations, phosphatidylcholine was the most appropriate phospholipid to certify vesicle

501

stability and minimize lipid-drug interactions [91,92]. Normally, the addition of cholesterol in

502

liposomal vesicles enhances in-vivo stability with the decrease in drug release rate. The slow-

503

release pattern of liposomes extends the drug residence time in pulmonary airways and assists

504

the uptake by macrophages [92]. While the inclusion of phosphatidylglycerol assist the

505

spreading of liposomes at the alveolar interface and potentially improve the drug release [93].

506

Multilamellar liposomes are more suitable than unilamellar liposomes for sustained

507

pulmonary drug release. The fusion of lipid vesicles with the alveolar interface and/or lipid

508

exchange between lipid vesicles and pulmonary surfactants has a significant impact on the

509

drug release mechanism [93]. Moreover, liposomes appear to be more suitable for pulmonary

510

application if they are formulated from substances endogenous to the lung i.e. pulmonary

511

surfactant.

512

[dipalmitoylphosphatidylcholine (DPPC), phosphatidylglycerol, phosphatidylcholines (PC)]

513

and proteins where lipids account for >90 % of the surfactant by mass [94-96]. The fate of

514

liposomes deposited in the alveoli is mainly governed by clearance and re-utilization of lung

21 | P a g e

Pulmonary

surfactant

is

a

unique

mixture

of

lipids

515

surfactant. The rate and degree of pulmonary uptake of liposomes are a function of their

516

phospholipid

517

phospholipid showed superior pulmonary uptake [97,98] whereas, macrophage activity

518

played an important role in the clearance mechanism of liposomes [99]. The mean vesicle

519

size of liposome should be less than 400 nm as macrophage uptake of the formulation will

520

increase after 400 nm [100]. Furthermore, neutral or anionic and cationic liposomes showed

521

the different pharmacological outcome. No major adverse effects were found after the

522

delivery of neutral or anionic liposomes. But, cationic liposomes were found to be fatal to

523

human cells and can potentially initiate genetic abnormalities. Furthermore, side effects of

524

cationic liposomes considerably enhanced with an increase of positive charge of the carriers.

525

However, cationic liposomes usually form the almost neutral complexes with negatively

526

charged nucleic acids. Thus such structural modification of cationic carriers usually controls

527

adverse effects on the cells [101]. However, during storage liposomal formulation

528

experienced considerable loss of encapsulated drug and alteration in the physical integrity of

529

the liposomes [102]. To surpass the instability and delivery issues, liposomes can be dried

530

using spray drying, SFD or freeze-drying into proliposomes [63]. Proliposomes are free-

531

flowing particles that instantly form liposomes when in contact with aqueous media [92,103].

532

These delivery systems are thoroughly summarized in the following section.

composition.

In

general,

liposomes

containing

phosphatidylglycerol

533 534

4.2 Proliposomes

535

Proliposomes were initially explored by Payne and his co-workers in 1986 as dry

536

phospholipid formulations with better stability than liposomes [104]. The dry form of

537

proliposomes ease transportation and makes them a functional and efficient delivery system.

538

Proliposomes includes hydrophilic carriers that are layered with cholesterol and

539

phospholipid. They are superior for the entrapment/encapsulation of both hydrophilic as well

22 | P a g e

540

as lipophilic molecules. Later in 1991, the idea of proliposomes was extended to include

541

liquid phospholipid formulations that can produce liposomes upon addition of aqueous

542

medium. Proliposomes can be grouped into two types i.e. particulate-based proliposomes and

543

solvent-based proliposomes. The selection of an apt carrier is a key aspect for the formulation

544

of particulate-based proliposomes. Basically, the carrier is selected on account of its porosity

545

and capacity to hold phospholipids on its surface [105,106]. Whereas, solvent-based

546

proliposomes are fabricated using an organic solvent that dissolves lipids and all at once is

547

miscible with water. In this technique, high concentration of phospholipid in the organic

548

solvent is formulated and the resulting liposomes are collected by dispersion into the aqueous

549

phase. The company of phospholipid, ethanol and water initially produces a stacked bilayer

550

that transforms into liposomes by hydration. Ultimately, proliposomes can be transformed

551

into liposomes by the addition of aqueous phase above the phase transition temperature (Tm)

552

of the lipid followed by continuous shaking. In relation to traditional liposomal formulations,

553

proliposomes reveal more benefits in stability, solubility, drug entrapment and drug release.

554

In addition, production of proliposomes can be scaled up by routinely used techniques such

555

as spray drying, fluidized-bed coating and fluid-energy micronization (jet-milling). Various

556

inhalable proliposomes formulations have been fabricated and explored to augment the

557

aerosolization performance and physicochemical stability of various molecules [68,106].

558

Various studies on inhalable proliposomes formulations are described in the following

559

section.

560 561

Patil-Gadhe et al (2013), fabricated inhalable rifapentine loaded proliposomes for the

562

treatment of pulmonary TB using soy phosphatidylcholine (HSPC) and cholesterol by spray

563

drying and optimized using 32 experimental design. Developed proliposomes showed mean

564

particle size, entrapment efficacy and zeta potential of 578 nm, 72.08 % and 29.40 mV,

23 | P a g e

565

respectively. Smooth spherical, spray-dried rifapentine proliposomes showed satisfactory

566

flow properties. During the in-vitro aerosolization study using Rotahaler®, rifapentine loaded

567

proliposomes showed FPF and MMAD of 92.50 % and 2.62 microns, respectively at a flow

568

rate of 60 L/min. In in-vitro release study rifapentine proliposomes displayed controlled

569

release (~ 90 %) up to 24 h in PBS (pH 7.4). Moreover, during the in-vivo pulmonary

570

pharmacokinetic study after intratracheal administration rifapentine proliposomes displayed

571

13.99, 6.43, 6.40 and 2.35-fold improvement in AUC(0-∞), AUC(0-24), MRT and Cmax,

572

respectively as compared to drug alone at the same dose (250 µg) [107]. Rojanarat et al

573

(2011),

574

phosphatidylcholine and cholesterol (1:1) by spray drying technique with mannitol as an inert

575

carrier. Irregular shaped proliposomes demonstrated FPF and MMAD of 35 % and 2.99

576

microns, respectively, at a flow rate of 60 L/min. During the in-vitro toxicity study, in MTT

577

assay INH-proliposomes did not show any toxicity to pulmonary-associated cells i.e. growth

578

of normal human bronchial epithelial cells (NHBE) and small airway epithelial cells (SAEC).

579

In addition, INH-proliposomes did not activate the alveolar macrophages (AMs) to produce

580

inflammatory mediators i.e. interleukin-1β, tumor necrosis factor-α, and nitric oxide, at a

581

toxic level confirmed from enzyme-linked immunosorbent assay (ELISA) method. Most

582

significantly, INH-proliposomes showed superior anti-mycobacterial activity against M.

583

Bovis-infected AM as compared to INH alone [108].

formulated

inhalable

isoniazid

(INH)

proliposomes

using

soybean

584 585

Also, Rojanarat et al (2012), fabricated inhalable pyrazinamide (PAZ) proliposomes using

586

soybean phosphatidylcholine and cholesterol (1:1) by spray drying method with porous

587

mannitol as a carrier for releasing PAZ to AM infected with mycobacteria. Irregular shaped

588

pro-liposomes demonstrated FPF and MMAD of 29 % and 4.4 microns, respectively, at a

589

flow rate of 60 L/min. During in-vitro toxicity study, in MTT assay using A549, human lung

24 | P a g e

590

cancer cell line cells (Calu-3; as an upper airway cell) and human alveolar basal epithelial

591

cell (NR8383; as a lower airway cell), formulated pro-liposome did not show any toxicity up

592

to 250 µg/mL. Furthermore, formulated proliposome did not activate the AMs to produce

593

inflammatory mediators i.e. interleukin-1β, tumor necrosis factor-α, and nitric oxide, at a

594

toxic level confirmed from ELISA kits. Additionally, in-vivo studies in male Wistar rats after

595

intratracheal administration (4 mg/kg) proliposome did not show any liver or renal toxicity

596

[109]. The same research group also prepared fluoro-quinolone antibiotic (levofloxacin)

597

loaded proliposomes for the treatment of pulmonary TB. Irregular shaped proliposomes

598

showed FPF and ED of 38.10 % and 91.30 % respectively, at a flow rate of 60 L/min. During

599

the in-vitro toxicity study, in MTT assay, levofloxacin pro-liposome showed absences of

600

toxicity on Calu-3, NR8383 up to 5 µg/mL. Moreover, levofloxacin proliposomes could not

601

produce inflammatory mediators and hence, showed marked improvement in the minimum

602

inhibitory concentration value as compared to drug alone [110].

603 604

Proliposomes have illustrated better therapeutic outcomes in the treatment of pulmonary

605

diseases. As summarized in the above section, proliposomes are easily fabricated using a

606

combination of SPC or HSPC and cholesterol with mannitol, porous mannitol or lactose as an

607

inert

608

physicochemical, biological and aerosolization properties of various actives. In comparison to

609

all discussed proliposomes, the HSPC and lactose based proliposome have achieved a greater

610

improvement in FPF (>90%) for rifapentine. Therefore, the combination of a drug molecule

611

with carefully selected lipid moiety, carrier and method may be a perfect move to formulate

612

proliposomes with reasonable aerodynamic properties.

carrier

by

613 614

5. Clinical outcome

25 | P a g e

spray-drying

method.

Spray-dried

proliposomes

could

enhance

615

In the last few decades, formulation scientists have been dynamically exploring the field of

616

liposomes to strengthen the existing standard of therapeutics. This active growth and

617

development in liposomes call for sustained clinical translation and commercialization [111].

618

Even though the preclinical trials have revealed positive effects, there is uncertainty linked to

619

safety in human beings. Thus, it is necessary to conduct a clinical trial to realize the ways in

620

which liposomes interact with the pulmonary airways. Clinical trials are the key building

621

blocks of evidence-based medicine and therefore, nurturing the backbone of clinical practice

622

[112].

623 624

In the past few years, the regulatory bodies have approved some liposomes-based delivery

625

systems and more than 100 different liposomes are in various clinical phases. Thus, in the

626

present segment, we have discussed the liposome-based pulmonary formulation that has

627

reached various phases of clinical trials. Clinical trials related to ‘‘liposomes’’ have been

628

indexed

629

[https://clinicaltrials.gov/]) and European (EU Clinical Trials Register [https://www.clinical

630

trialsregister.eu/]). Clinical trials with inhaled liposomes have been executed for pulmonary

631

tuberculosis, cystic fibrosis, bronchiectasis, fungal infections, lung transplantation, cancer

632

(for osteosarcoma metastatic), analgesia (for post-operative pain), pulmonary moisturizer

633

along with the understanding of safety, efficacy and toxicity. Few remarkable clinical trials

634

are summarized in the following section while clinical efforts invested in inhaled liposomes

635

are listed in Table 4.

and

are

easily

accessible

in

the

US

FDA

(ClinicalTrials.gov

636 637

5.1 Liposomal amikacin (Arikayce®)

638

Insmed Incorporation (New Jersey, US) is a well-known global biopharmaceutical

639

organization with a focus on the design and development of various critical pharmaceutical

26 | P a g e

640

dosage forms. The lipid bilayer of Arikayce® consists of DPPC and cholesterol. The Insmed

641

studied inhaled liposomal amikacin(Arikayce®) to treat mycobacterium infections (phase 3;

642

NCT02344004) [113], cystic fibrosis (phase 3;NCT01316276) [114] and Pseudomonas

643

aeruginosa infection (phase 3; NCT01315678) [115]. The liposomal amikacin was expected

644

to modulate disease conditions. Similarly, Insmed also studied inhaled liposomal cisplatin for

645

the treatment of cancer i.e. osteosarcoma metastatic of lungs (phase 1; NCT00102531) [116].

646

All these studies are near completion and the Insmed is expecting positive outcomes.

647 648

5.2 Liposomal amphotericin B (Ambisome®)

649

Various medical institutes together with pharmaceutical industries investigated inhaled

650

liposomal amphotericin B (Ambisome®) for various pulmonary diseases. Ambisome® (~ 100

651

nm) is composed of amphotericin B, DSPG, hydrogenated soy phosphatidylcholine and

652

cholesterol in a 0.4:0.8:2:1 molar ratio. Interaction between amphotericin B and cholesterol

653

attributed to its sterol binding which is highly responsible for stabilization. Ambisome® was

654

studied for the treatment of allergic bronchopulmonary aspergillosis (phase 2;

655

NCT02273661) [117] and invasive pulmonary aspergillosis (phase 4; NCT00986713) [118].

656 657

5.3 Liposomal phospholipids (LipoAerosol©)

658

The weakening of airways surfaces fluid film (pulmonary surfactant) is one of the main

659

factors responsible for pulmonary disorders (e.g. asthma, COPD and pulmonary edema).

660

Thus, replenishment of pulmonary surfactants through a pharmacological treatment might be

661

an appropriate strategy in the treatment of pulmonary disorders. The lipid vesicles of

662

LipoAerosol® have composed of phospholipids i.e. phosphatidylcholine which is an integral

663

part of the natural pulmonary surfactant. LipoAerosol® provides moistening, warming and

664

cleaning of the upper and lower pulmonary airways as well as assists the natural moistening

27 | P a g e

665

of the film in airway diseases and irritations. Recently, Technische Universität München

666

(Germany)

667

complications (NCT02157129) [119] and hoping for positive results.

has

investigated

LipoAerosol® therapeutic

potential

for

tracheostomy

668 669

5.4 Aerosolized liposomal fentanyl (AeroLEF™)

670

AeroLEF™ (liposome loaded fentanyl) is a new aerosol that offers rapid, extended and

671

personalized analgesia for patients undergoing acute pain episodes. It is designed and

672

developed for the non-invasive route of administration. It can be used for the treatment of

673

moderate to severe pain. AeroLEF™ was assessed in a few clinical trials for controlling pain

674

and post-operative pain (NCT00791804) [120]. Moreover, YM BioSciences Inc. (Canadian

675

drug development company) has studied AeroLEF™ therapeutic potential with four different

676

inhaler devices for device characterization and qualification (NCT00794209) [121].

677 678

The clinical trial database of the US FDA reported no clinical trial on the inhaled liposomes

679

as dry powders. However, an extensive amount of time and effort has been devoted by active

680

scientists and clinicians over the years towards design, development and clinical assessment

681

of inhaled liposomes. All these active efforts signify a bridgehead for the clinical progress of

682

inhaled liposomes. Still, a number of human clinical trials of inhaled liposomes have yet to be

683

performed for a wide range of ailments and better pulmonary therapy.

684 685

6. Market overview

686

The excitement regarding liposomes and liposome-based products has accelerated gradually

687

over the past few years. Liposomes and liposome-based products have demonstrated a

688

beneficial impact on the pharmaceutical market. Inhaled liposomes have offered several key

689

advantages including local as well as systemic delivery, high drug loading capacity,

28 | P a g e

690

controlled release kinetics and good patient compliance. Furthermore, nanocarriers such as

691

liposomes or pro-liposomes also help to extend the patent life and consequently improves the

692

value of drug molecules. For example, the Doxil® (liposomal formulation of doxorubicin) has

693

shown a good impact on cancer management with great benefits for pharmaceutical

694

industries. While, DaunoXome® (liposomal daunorubicin, Galen, Craigavon, UK), Onivyde®

695

(liposomal irinotecan injection, Merrimack Pharmaceuticals, US), DepoCyt® (liposomal

696

cytarabine, Pacira Pharmaceuticals, US),Marqibo® (liposomal vincristine sulfate, Talon

697

Therapeutics, US), AmBisome® (liposomal Amphotericin B, NeXstar Pharmaceuticals, US),

698

Vyxeos® (daunorubicin and cytarabine encapsulated liposomes, Jazz Pharmaceutics, Ireland)

699

and Visudyne® (benzoporphyrin derivative liposomes, QLT Phototherapeutics, Canada) are

700

the known and FDA approved liposomal products [122]. Still, no inhaled liposomal as dry

701

powders were available in the market. However, bench-to-bedside translation of nanocarriers

702

and adopting the same into the mainstream level is often a critical task. There are several

703

chemistry, manufacturing and control (CMC) challenge along with regulatory issues that

704

need to be defeated before it can move on to extensive clinical applications and community

705

acceptance. Thus, by knowing this situation, in the upcoming segment of article authors

706

provide a view on the existing challenges and future directions for liposomes; especially for

707

pulmonary applications.

708 709

7. Discussion

710

Currently, nanocarriers such as liposomes as well as proliposomes are receiving growing

711

interest among formulation scientists for better pulmonary therapy. So far, several

712

formulation scientists and experts have invested their time and efforts in designing,

713

developing and analyzing respirable liposomes and proliposomes for better clinical outcomes.

714

However, investigation in this stage is still in the primitive phase. Many published articles

29 | P a g e

715

have evaluated and discussed particle size, surface charge, in-vitro aerosolization behavior

716

and pulmonary pharmacokinetics. Yet, various other challenges must be addressed when

717

formulating liposomes and proliposomes for inhalation. Several key benefits and challenges

718

of inhaled liposomes are listed in Fig. 4 [123].

719 720

Needless to say, but achieving adequate deep lung deposition and targeting the drug to

721

specific pulmonary airways, is still very tough [68]. Ahead of the complexity of respiratory

722

diseases and lung morphology, it should be kept in mind that disease severity, patient’s age,

723

breathing pattern, device configuration (single dose, multi-dose device) and design features

724

(capsule-based, pre-filed or disposable device) conclude the real fate of aerosol and the final

725

therapeutic outcome of inhaled therapy [68,124,125]. Normally, the successful aerosol

726

delivery depends on four mutually dependent factors: the formulation, the inhaler device

727

configuration, the metering system and lastly, the patient’s training/understanding [124,126].

728

In the above-reviewed articles, most scientists have addressed liposomes and proliposomes

729

manufacturing techniques, drug loading methods, drug targeting strategies and pulmonary

730

pharmacokinetic problems; however, they have merely discussed issues pertain to inhaler

731

devices. Beyond this, there is also a fascinating story to be told about inhaler devices.

732

Furthermore, the device dose metering system has great importance as the dose needed for

733

the effective therapeutic outcome of antimicrobial is too large (e.g. 500 µg) to be inhaled in a

734

single actuation [68]. In such cases, there is a strong need for disposable inhaler device for

735

safe, effective delivery and to avoid bacterial resistance. Basically, to attain satisfactory drug

736

deposition, the development of formulation and inhaler devices should be considered as a

737

whole medicinal product. As per our perceptive strong collaboration between formulation

738

experts and device, engineers is desired to accomplish complex devices/formulation

739

interfaces.

30 | P a g e

740 741

Similarly, to explore the complete therapeutic potential of these multifunctional nanocarriers,

742

exhaustive consideration is required for pulmokinetics, lung clearance rate and other

743

toxicological issues. These issues can be tackled by improving current animal models that can

744

simulate the pathology of the human pulmonary airways [126]. Further, the new imaging

745

tools and techniques can provide superior extrapolative pre-clinical models to understand

746

complex and difficult to treat pulmonary ailments [128-130]. In addition, regarding the

747

situation of respirable liposomes in human clinical trials, there are presently only a few

748

formulations. Hence, up to date data and knowledge of clinical measurements such as forced

749

vital capacity (FVC) and forced expiratory volume (FEV), Tiffeneau-Pinelli index

750

(FEV1/FVC) and maximal voluntary ventilation (MVV) will be crucial in understanding their

751

capability for treating respiratory ailments [68,11]. US FDA has recently published a

752

guidance document i.e. ʻLiposome Drug Productsʼ for a better understanding of the liposomal

753

drug. Likewise, to utilize the full potential of these multifunctional nanocarriers absolute

754

consideration must be paid to upcoming regulatory and CMC guidance. Simply, combined

755

research effort will be highly appreciated to surpass gaps in our understanding with the

756

intention that we can assemble and use all accessible tools and techniques across the pre-

757

clinical-translational-clinical-axis to deliver the best pulmonary dosage to the patient in the

758

most efficient manner.

759 760

8. Conclusion

761

With the growth of nanotechnology and other allied fields, a number of versatile liposomes

762

are designed, developed and investigated as drug delivery carriers. In the vicinity of

763

pulmonary drug delivery, many bioactive molecules have been effectively encapsulated into

764

different liposomal carriers. Additionally, numerous novel strategies have been adopted to

31 | P a g e

765

modify the liposomal drug release pattern, in-vivo drug targeting and biodistribution within

766

the pulmonary airways. The research carried until now proved a great potential for the use of

767

liposomes in the treatment of various intrapulmonary and extrapulmonary diseases. However,

768

it is apparent from the present article that there is a clear need to continue the efforts to

769

design, develop and systematically evaluate the inhaled liposomal formulations. The present

770

situation of inhaled liposomes highly demands formulation scientists to judge long term and

771

structured strategy to pave a way for successful clinical translation, regulatory clearance with

772

FDA approval.

773 774

9. Acknowledgments

775

The author is thankful to Bharati Deemed Vidyapeeth University, Poona College of

776

Pharmacy, Pune-38, India for support and institutional facilities.

777 778

10. Funding Information

779

No financial support and writing assistance was utilized in the production of this manuscript.

780 781

11. Conflicts of Interest

782

Author declare no conflict of interest.

783 784

12. References

785

1. Salvi S, Kumar GA, Dhaliwal RS, Paulson K, Agrawal A, Koul PA et al. The burden of

786

chronic respiratory diseases and their heterogeneity across the states of India: the Global

787

Burden of Disease Study 1990-2016. Lancet Glob Health. 2018;6(12):e1363-e1374.

788

32 | P a g e

789

2. World Health Organization, WHO, Chronic Respiratory Diseases (CRDs): Fact Sheets,

790

(2018) http://www.who.int/respiratory/en/.

791 792

3. Celli BR, Agusti A. COPD: time to improve its taxonomy?. ERJ Open Res. 2018;4(1). pii:

793

00132-2017.

794 795

4. Forum of International Respiratory Societies. The Global Impact of Respiratory Disease-

796

Second Edition. Sheffield, European Respiratory Society, 2017.

797 798

5. Mehta PP. Multi-dose dry powder inhaler: advance technology for drug delivery to

799

airways. Indian Drug, 2019;56 (11):59-62.

800 801

6. Mehta P, Bothiraja C, Kadam S, Pawar A. Effect of USP induction ports, glass sampling

802

apparatus, and inhaler device resistance on aerodynamic patterns of fluticasone propionate-

803

loaded engineered mannitol microparticles. AAPS PharmSciTech. 2019;20(5):197.

804 805

7. Behara SR, Larson I, Kippax P, Stewart P, Morton DA. Insight into pressure drop

806

dependent efficiencies of dry powder inhalers. Eur J Pharm Sci. 2012;46(3):142-148.

807 808

8. Mehta PP. Imagine the superiority of dry powder inhalers from carrier engineering. J Drug

809

Deliv. 2018;2018:5635010.

810 811

9. Mehta PP. Dry powder inhalers: a focus on advancements in novel drug delivery systems. J

812

Drug Deliv. 2016;2016:8290963.

33 | P a g e

813

10. Kuzmov A, Minko T. Nanotechnology approaches for inhalation treatment of lung

814

diseases. J Control Release. 2015;219:500-518.

815 816

11. Mehta P, Kadam S, Pawar A, Bothiraja C. Dendrimers for pulmonary delivery: current

817

perspectives and future challenges. New J. Chem. 2019;43(22):8396-8409.

818 819

12. Aguilera G, Berry C, West R, Gonzalez-Monterrubio E, Angulo-Molina A, Arias-

820

Carrión O et al. Carboxymethyl cellulose coated magnetic nanoparticles transport across a

821

human lung microvascular endothelial cell model of the blood-brain barrier. Nanoscale Adv.

822

2019;1:671-685.

823 824

13. Wongpinyochit T, Totten J, Johnstona B, Seib F. Microfluidic-assisted silk nanoparticle

825

tuning. Nanoscale Adv. 2019;1:873-883.

826 827

14. Abdelaziz HM, Gaber M, Abd-Elwakil MM, Mabrouk MT, Elgohary MM, Kamel NM, et

828

al. Inhalable particulate drug delivery systems for lung cancer therapy: nanoparticles,

829

microparticles, nanocomposites and nanoaggregates. J Control Release. 2018;269:374-392.

830 831

15. Muralidharan P, Malapit M, Mallory E, Hayes Jr D, Mansour HM. Inhalable

832

nanoparticulate powders for respiratory delivery. Nanomedicine. 2015;11(5):1189-1199.

833 834

16. Andrade F, Rafael D, Videira M, Ferreira D, Sosnik A, Sarmento B. Nanotechnology and

835

pulmonary delivery to overcome resistance in infectious diseases. Adv Drug Deliv

836

Rev. 2013;65(13-14):1816-1827.

837

34 | P a g e

838

17. Mehta P, Bothiraja C, Mahadik K, Kadam S, Pawar A. Phytoconstituent based dry

839

powder inhalers as biomedicine for the management of pulmonary diseases. Biomed

840

Pharmacother. 2018;108:828-837.

841 842

18. van Rijt SH, Bein T, Meiners S. Medical nanoparticles for next generation drug delivery

843

to the lungs. Eur Respir J. 2014;44(3):765-774.

844 845

19. Cipolla D, Gonda I, Chan HK. Liposomal formulations for inhalation. Ther

846

Deliv. 2013;4(8):1047-1072.

847 848

20. Jumeaux C, Kim E, Howes P, Kim H, Chandrawati R, Stevens M. Detection of

849

microRNA biomarkers via inhibition of DNA-mediated liposome fusion. Nanoscale Adv.

850

2019;1:532-536.

851 852

21. Willis L, Hayes D, Mansour HM. Therapeutic liposomal dry powder inhalation aerosols

853

for targeted lung delivery. Lung. 2012;190(3):251-262.

854 855

22. Khatib I, Khanal D, Ruan J, Cipolla D, Dayton F, Blanchard JD et al. Ciprofloxacin

856

nanocrystals liposomal powders for controlled drug release via inhalation. Int J

857

Pharm. 2019;566:641-651.

858 859

23. Laverman P, Boerman OC, Oyen WJ, Dams ET, Storm G, Corstens FH. Liposomes for

860

scintigraphic detection of infection and inflammation. Adv. Drug Deliv. Rev. 1999;37:225-

861

235.

862

35 | P a g e

863

24. Bangham AD, Standish MM, Watkins JC. Diffusion of univalent ions across the lamellae

864

of swollen phospholipids. J. Mol. Biol. 1965;13:238-252.

865 866

25. van der Meel R, Fens MH, Vader P, van Solinge WW, Eniola-Adefeso O, Schiffelers

867

RM. Extracellular vesicles as drug delivery systems: lessons from the liposome field. J.

868

Control. Release 2014;195:72-85.

869 870

26. Rideau E, Dimova R , Schwille P , Wurm FR, Landfester K. Liposomes and

871

polymersomes: a comparative review towards cell mimicking. Chem. Soc. Rev.

872

2018;47:8572-8610.

873 874

27. Chorilli M, Calixto G, Rimério TC, Scarpa MV. Caffeine encapsulated in small

875

unilamellar liposomes: characerization and in vitro release profile. J. Dispers. Sci. Technol.

876

2013;34:1465-1470.

877 878

28. Koynova R, Caffrey M. Phases and phase transitions of the phosphatidylcholines.

879

Biochim. Biophys. Acta. 1998;1376:91-145.

880 881

29. Van Tran V, Moon JY, Lee YC. Liposomes for delivery of antioxidants in

882

cosmeceuticals: Challenges and development strategies. J Control Release. 2019;300:114-

883

140.

884 885

30. Chu LY, Utada AS, Shah RK, Kim JW, Weitz DA. Controllable monodisperse multiple

886

emulsions. Angew. Chem. Int. Ed. 2007;46:8970-8974.

887

36 | P a g e

888

31. Aimon S, Manzi J, Schmidt D, Poveda Larrosa JA, Bassereau P, Toombes GE.

889

Functional reconstitution of a voltage-gated potassium channel in giant unilamellar vesicles.

890

PLoS One. 2011;6(10):e25529.

891 892

32. Stachowiak JC, Richmond DL, Li TH, Liu AP, Parekh SH, Fletcher DA. Unilamellar

893

vesicle formation and encapsulation by microfluidic jetting. Proc Natl Acad Sci U S

894

A. 2008;105(12):4697-4702.

895 896

33. Sugiura S, Kuroiwa T, Kagota T, Nakajima M, Sato S, Mukataka S et al. Novel method

897

for obtaining homogeneous giant vesicles from a monodisperse water-in-oil emulsion

898

prepared with a microfluidic device. Langmuir. 2008;24:4581-4588.

899 900

34. Mayer LD, Hope MJ, Cullis PR. Vesicles of variable sizes produced by a rapid extrusion

901

procedure. Biochim Biophys Acta. 1986;858(1):161-168.

902 903

35. Pautot S, Frisken BJ, Weitz DA. Engineering asymmetric vesicles. Proc Natl Acad Sci U

904

S A. 2003;100(19):10718-10721.

905 906

36. Ota S, Yoshizawa S, Takeuchi S. Microfluidic formation of monodisperse, cell‐sized, and

907

unilamellar vesicles. Angew Chem Int Ed Engl. 2009;48(35):6533-6537.

908 909

37. Mazzitelli S, Capretto L, Quinci F, Piva R, Nastruzzi C. Preparation of cell-encapsulation

910

devices in confined microenvironment. Adv Drug Deliv Rev. 2013;65(11-12):1533-1555.

911

37 | P a g e

912

38. Bangham AD, De Gier, Greville G. Osmotic properties and water permeability of

913

phospholipid liquid crystals. Chem. Phys. Lipids. 1967;1:225-246.

914 915

39. Grimaldi N, Andrade F, Segovia N, Ferrer-Tasies L, Sala S, Veciana J, et al. Lipid-based

916

nanovesicles for nanomedicine. ChemSoc Rev. 2016;45(23):6520-6545.

917 918

40. Grijalvo S, Mayr J, Eritja R, Díaz DD. Biodegradable liposome-encapsulated hydrogels

919

for biomedical applications: a marriage of convenience. Biomater Sci. 2016;4(4):555-574.

920 921

41. Szoka J, Papahadjopoulos D. Procedure for preparation of liposomes with large internal

922

aqueous space and high capture by reverse-phase evaporation. Proc. Natl. Acad. Sci. U. S. A.

923

1978;75:4194-4198.

924 925

42. Deamer DW. Preparation and properties of ether injection liposomes. Ann. N. Y. Acad.

926

Sci. 1978;308:250-258.

927 928

43. Jahn A, Vreeland W, Gaitan M, Locascio L. Controlled vesicle self-assembly in

929

microfluidic channels with hydrodynamic focusing. J. Am. Chem. Soc. 2004;126

930

:2674-2675.

931 932

44. van Swaay D, deMello AD. Microfluidic methods for forming liposomes. Lab

933

Chip. 2013;13(5):752-767.

934 935

45. Jaafar-Maalej C, Charcosset C, Fessi H. A new method for liposome preparation using a

936

membrane contactor. J. Liposome Res. 2011;21:213-220.

38 | P a g e

937 938

46. Duong AD, Collier MA, Bachelder EM, Wyslouzil BE, Ainslie KM. One Step

939

Encapsulation of Small Molecule Drugs in Liposomes via Electrospray-Remote Loading.

940

Mol Pharm. 2016;13(1):92-99.

941 942

47. Collier MA, Bachelder EM, Ainslie KM. Electrosprayed Myocet-like Liposomes: An

943

Alternative to Traditional Liposome Production. Pharm Res. 2017;34(2):419-426.

944 945

48. Etheridge ML, Campbell SA, Erdman AG, Haynes CL, Wolf SM, McCullough J. The big

946

picture on nanomedicine: the state of investigational and approved nanomedicine products.

947

Nanomedicine. 2013;9(1):1-14.

948 949

49. Simone EA, Dziubla TD, Muzykantov VR. Polymeric carriers: role of geometry in drug

950

delivery. Expert Opin Drug Deliv. 2008;5(12):1283-1300.

951 952

50. Coune A. Liposomes as drug delivery system in treatment of infectious diseases, potential

953

applications and clinical experience. Infection. 1988;16:141-147.

954 955

51. Plotnick AN. Lipid-based formulations of amphotericin B. J. Am. Vet. Med. Assoc.

956

2000;216:838-841.

957 958

52. de Pauw BE. Fungal infections: diagnostic problems and choice of therapy. Leuk. Suppl.

959

2012;1:S22-23.

960

39 | P a g e

961

53. Woodle MC. Sterically stabilized liposome therapeutics. Adv. Drug Deliver. Rev.

962

1995;16:249-265.

963 964

54. Papahadjopoulos D. Stealth liposomes: from steric stabilization to targeting. CRC Press,

965

Boca Raton FL USA; 1995.

966 967

55. Drummond DC, Meyer O, Hong K, Kirpotin DB, Papahadjopoulos D. Optimizing

968

liposomes for delivery of chemotherapeutic agents to solid tumors. Pharmacol Rev.

969

1999;51(4):691-743.

970 971

56. Mu LM, Ju RJ, Liu R, Bu YZ, Zhang JY, Li XQ, et al. Dual-functional drug liposomes in

972

treatment of resistant cancers. Adv Drug Deliv Rev. 2017;115:46-56.

973 974

57. Nogueira E, Gomes AC, Preto A, Cavaco-Paulo A. Design of liposomal formulations for

975

cell targeting. Colloids Surf B Biointerfaces. 20151;136:514-526.

976 977

58. Marqués-Gallego P, de Kroon AI. Ligation strategies for targeting liposomal

978

nanocarriers. Biomed Res Int. 2014;2014:129458.

979 980

59. Morshed M, Chowdhury E. Gene delivery and clinical applications, Reference module in

981

biomedical sciences, Encyclopedia of Biomedical Engineering. 2019:345-351.

982 983

60. Weber S, Zimmer A, Pardeike J. Solid Lipid Nanoparticles (SLN) and Nanostructured

984

Lipid Carriers (NLC) for pulmonary application: a review of the state of the art. Eur J Pharm

985

Biopharm. 2014;86(1):7-22.

40 | P a g e

986 987

61. Gaspar M, Bakowsky U, Ehrhardt C. Inhaled liposomes-current strategies and future

988

challenges. J. Biomed. Nanotechnol. 2008;4:1-13.

989 990

62. G. Pilcer, K. Amighi, Formulation strategy and use of excipients in pulmonary drug

991

delivery. Int. J. Pharm. 2010;392:1-19.

992 993

63. Misra A, Jinturkar K, Patel D, Lalani J, Chougule M. Recent advances in liposomal dry

994

powder formulations: preparation and evaluation. Expert Opin Drug Deliv. 2009;6(1):71-89.

995 996

65. Willis L, Hayes D, Mansour H. Therapeutic liposomal dry powder inhalation aerosols for

997

targeted lung delivery. Lung. 2012;190:251-262.

998 999

66. Sanders M. Inhalation therapy: an historical review. Prim Care Respir J. 2007;16:71-81.

1000 1001

67. Skupin Mrugalska P. Liposome-based drug delivery for lung cancer, in Kesharwani P

1002

(Eds.), Nanotechnology-based targeted drug delivery systems for lung cancer, Academic

1003

Press, 2019;123-160.

1004 1005

68. Mehta P, Bothiraja C, Kadam S, Pawar A. Potential of dry powder inhalers for

1006

tuberculosis therapy: facts, fidelity and future. Artif Cells Nanomed Biotechnol.

1007

2018;46(sup3):S791-S806.

1008 1009

69. Zhou Q, Tang P, Leung S, et al. Emerging inhalation aerosol devices and strategies:

1010

where are we headed? Adv Drug Deliv Rev. 2014;75:3-17.

41 | P a g e

1011 1012

70. Tagami T, Ozeki T. Recent Trends in Clinical Trials Related to Carrier-Based Drugs. J

1013

Pharm Sci. 2017;106(9):2219-2226.

1014 1015

71. Zhu X, Kong Y, Liu Q, Lu Y, Xing H, Lu X, et al. Inhalable dry powder prepared from

1016

folic acid-conjugated docetaxel liposomes alters pharmacodynamic and pharmacokinetic

1017

properties relevant to lung cancer chemotherapy. Pulm Pharmacol Ther. 2019;55:50-61.

1018 1019

72. Gandhi M, Pandya T, Gandhi R, Patel S, Mashru R, Misra A, Tandel H. Inhalable

1020

liposomal dry powder of gemcitabine-HCl: Formulation, in vitro characterization and in vivo

1021

studies. Int J Pharm. 2015;496(2):886-895.

1022 1023

73. Gibbons A, McElvaney NG, Cryan SA. A dry powder formulation of liposome-

1024

encapsulated recombinant secretory leukocyte protease inhibitor (rSLPI) for inhalation:

1025

preparation and characterisation. AAPS PharmSciTech. 2010;11(3):1411-1421.

1026 1027

74. Tang Y, Zhang H, Lu X, Jiang L, Xi X, Liu J, Zhu J. Development and evaluation of a

1028

dry powder formulation of liposome-encapsulated oseltamivir phosphate for inhalation. Drug

1029

Deliv. 2015;22(5):608-618.

1030 1031

75. Hamed A, Osman R, Al-Jamal KT, Holayel SM, Geneidi AS. Enhanced antitubercular

1032

activity, alveolar deposition and macrophages uptake of mannosylated stable nanoliposomes.

1033

J Drug Deliv Sci Technol. 2019;51:513-523.

1034

42 | P a g e

1035

76. Huang WH, Yang ZJ, Wu H, Wong YF, Zhao ZZ, Liu L. Development of liposomal

1036

salbutamol sulfate dry powder inhaler formulation. Biol Pharm Bull. 2010;33(3):512-517.

1037 1038

77. Honmane S, Hajare A, More H, Osmani RAM, Salunkhe S. Lung delivery of

1039

nanoliposomal salbutamol sulfate dry powder inhalation for facilitated asthma therapy. J

1040

Liposome Res. 2019;29(4):332-342.

1041 1042

78. Ye T, Yu J, Luo Q, Wang S, Chan H. Inhalable clarithromycin liposomal dry powders

1043

using ultrasonic spray freeze drying. Powder Technol. 2017;305:63-70.

1044 1045

79. Zhang T, Chen Y, Ge Y, Hu Y, Li M, Jin Y. Inhalation treatment of primary lung cancer

1046

using liposomal curcumin dry powder inhalers. Acta Pharm Sin B. 2018;8(3):440-448.

1047 1048

80. Khatib I, Khanal D, Ruan J, Cipolla D, Dayton F, Blanchard J, Chan H. Ciprofloxacin

1049

nanocrystals liposomal powders for controlled drug release via inhalation. Int J Pharm.

1050

2019;566:641-651.

1051 1052

81. Li M, Zhang T, Zhu L, Wang R, Jin Y. Liposomal andrographolide dry powder inhalers

1053

for treatment of bacterial pneumonia via anti-inflammatory pathway. Int J Pharm.

1054

2017;528(1-2):163-171.

1055 1056

82. Viswanathan V, Pharande R, Bannalikar A, Gupta P, Gupta U, Mukne A. Inhalable

1057

liposomes of Glycyrrhiza glabra extract for use in tuberculosis: formulation, in

1058

characterization, in vivo lung deposition, and in vivo pharmacodynamic studies. Drug Dev

1059

Ind Pharm. 2019;45(1):11-20.

43 | P a g e

vitro

1060 1061

83. Chennakesavulu S, Mishra A, Sudheer A, Sowmya C, Reddy C, Bhargav E. Pulmonary

1062

delivery of liposomal dry powderinhaler formulation for effective treatment ofidiopathic

1063

pulmonary fibrosis. Asian J. Pharm. Sci. 2018;13:91-100.

1064 1065

84. Ourique AF, Chaves Pdos S, Souto GD, Pohlmann AR, Guterres SS, Beck RC.

1066

Redispersible

1067

development, in vitro characterization and antioxidant activity. Eur J Pharm Sci.

1068

2014;65:174-182.

liposomal-N-acetylcysteine

powder

for

pulmonary

administration:

1069 1070

85. Khatib I, Khanal D, Ruan J, Cipolla D, Dayton F, Blanchard JD, Chan HK. Ciprofloxacin

1071

nanocrystals liposomal powders for controlled drug release via inhalation. Int J Pharm.

1072

2019;566:641-651.

1073 1074

86. Daman Z, Gilani K, Najafabadi A, Eftekhariand H, Barghi M. Formulation of inhalable

1075

lipid-based salbutamol sulfatemicroparticles by spray drying technique. Daru. 2014; 22(1):

1076

50.

1077 1078

87. Joshi M, Misra A. Dry powder inhalation of liposomal Ketotifen fumarate: formulation

1079

and characterization. Int J Pharm. 2001;223(1-2):15-27.

1080 1081

88. Chougule M, Padhi B, Misra A. Development of spray dried liposomal dry powder

1082

inhaler of Dapsone. AAPS PharmSciTech. 2008;9(1):47-53.

1083

44 | P a g e

1084

89. Chougule M, Padhi B, Misra A. Nano-liposomal dry powder inhaler of tacrolimus:

1085

preparation, characterization, and pulmonary pharmacokinetics. Int J Nanomedicine.

1086

2007;2(4):675-688.

1087 1088

90. Changsan N, Chan HK, Separovic F, Srichana T. Physicochemical characterization and

1089

stability of rifampicin liposome dry powder formulations for inhalation. J Pharm Sci.

1090

2009;98(2):628-639.

1091 1092

91. Giovagnoli S, Blasi P, Vescovi C, Fardella G, Chiappini I, Perioli L, Ricci M, Rossi C.

1093

Unilamellar

1094

physicochemical characterization. AAPS PharmSciTech. 2004;4(4):E69.

vesicles

as

potential

capreomycin

sulfate

carriers:

preparation

and

1095 1096

92. Parumasivam T, Chang RY, Abdelghany S, Ye TT, Britton WJ, Chan HK. Dry powder

1097

inhalable formulations for anti-tubercular therapy. Adv Drug Deliv Rev. 2016;102:83-101.

1098 1099

93. Taylor KM, Newton JM. Liposomes for controlled delivery of drugs to the lung. Thorax.

1100

1992 Apr;47(4):257-259.

1101 1102

94. King RJ. Isolation and chemical composition of pulmonary surfactant, in Robertson B,

1103

Van Golde LMG, Batenburg JJ (Eds.), Pulmonary surfactant, Amstedam, Elsevier, 1984:1-

1104

15.

1105 1106

95. Wright JR, Clements JA. Metabolism and turnover of lung surfactant. Am Rev Respir Dis

1107

1987;135:426-444.

1108

45 | P a g e

1109

96. Lopez-Rodriguez E, Pérez-Gil J. Structure-function relationships in pulmonary surfactant

1110

membranes: from biophysics to therapy. Biochim Biophys Acta. 2014;1838(6):1568-1585.

1111 1112

97. Oyarzun MJ, Clements JA, Baritussio A. Ventilation enhances the pulmonary alveolar

1113

clearance of radioactive dipalmitoylphosphatidylcholine in liposomes. Am Rev Respir Dis

1114

1980;121:709-721.

1115 1116

98. Morimoto Y, Adachi Y. Pulmonary uptake of liposomal phosphatidylcholine upon

1117

intratrachealadministation to rats. Chem Pharmaceut Bull 1982;30:2248-2251.

1118 1119

99. Martin WJ, Standing JE. Amiodarone pulmonary toxicity: biochemical evidence for a

1120

cellular phospholipidosis in the bronchoalveolar lavage of human subjects. J Pharmacol

1121

ExpTher 1988;244:774-779.

1122 1123

100. Geiser M, Casaulta M, Kupferschmid B, Schulz H, Semmler-Behnke M, Kreyling W.

1124

The role of macrophages in the clearance of inhaled ultrafine titanium dioxide particles. Am J

1125

Respir Cell Mol Biol. 2008;38(3):371-376.

1126 1127

101. Shah V, Taratula O, Garbuzenko OB, Patil ML, Savla R, Zhang M, Minko T.

1128

Genotoxicity of different nanocarriers: possible modifications for the delivery of nucleic

1129

acids. Curr Drug Discov Technol. 2013;10(1):8-15.

1130 1131

102. Cipolla D, Gonda I, Chan HK. Liposomal formulations for inhalation. Ther Deliv.

1132

2013;4(8):1047-1072.

1133

46 | P a g e

1134

103. Agnihotri SA, Soppimath KS, Betageri GV. Controlled release application of

1135

multilamellar vesicles: a novel drug delivery approach. Drug Deliv. 2010;17(2):92-101.

1136 1137

104. Payne NI, Timmins P, Ambrose CV, Ward MD, Ridgway F. Proliposomes: a novel

1138

solution to an old problem. J Pharm Sci 1986;75:325-329.

1139 1140

105. Elhissi A, Phoenix D, Ahmed W. Some approaches to large-scale manufacturing of

1141

liposomes, in Ahmed W and Jackson MJ (Eds.), Emerging Nanotechnologies for

1142

Manufacturing, 2nd Eds, William Andrew Publishing, 2015: 402-417.

1143 1144

106. Khan I, Elhissi A, Shah M, Alhnan MA, Ahmed W. Liposome-based carrier systems and

1145

devices used for pulmonary drug delivery, in Davim JP (Eds.), Biomaterials and Medical

1146

Tribology, Woodhead Publishing, 2013:395-443.

1147 1148

107. Patil-Gadhe A, Pokharkar V. Single step spray drying method to develop proliposomes

1149

for inhalation: a systematic study based on quality by design approach. Pulm Pharmacol

1150

Ther. 2014;27(2):197-207.

1151 1152

108. Rojanarat W, Changsan N, Tawithong E, Pinsuwan S, Chan HK, Srichana T. Isoniazid

1153

proliposome powders for inhalation-preparation, characterization and cell culture studies. Int

1154

J Mol Sci. 2011;12(7):4414-4434.

1155 1156

109.

Rojanarat

1157

Inhaled pyrazinamide proliposome for targeting alveolar macrophages. Drug Deliv. 2012;

1158

19(7):334-345.

47 | P a g e

W, Nakpheng

T, Thawithong

E, Yanyium

N, Srichana

T.

1159 1160

110. Rojanarat W, Nakpheng T, Thawithong E, Yanyium N, Srichana T. Levofloxacin-

1161

proliposomes: opportunities for use in lung tuberculosis. Pharmaceutics. 2012;4(3):385-412.

1162 1163

111. Hassan S, Prakash G, Ozturk AB, Saghazadeh S, Sohail MF, Seo J, et al. Evolution and

1164

clinical translation of drug delivery nanomaterials. Nano Today. 2017;15:91-106.

1165 1166

112. Masic I, Miokovic M, Muhamedagic B. Evidence based medicine–new approaches and

1167

challenges. Acta Informatica Medica. 2008;16(4):219.

1168 1169

113. Study to Evaluate Efficacy of LAI When Added to Multi-drug Regimen Compared to

1170

Multi-drug

1171

https://clinicaltrials.gov/ct2/show/NCT02344004?term=NCT02344004&rank=1.

Regimen

Alone

(CONVERT).

1172 1173

114. Extension Study of Liposomal Amikacin for Inhalation in Cystic Fibrosis (CF) Patients

1174

With

1175

https://clinicaltrials.gov/ct2/show/NCT01316276?term=NCT01316276&rank=1

Chronic

Pseudomonas

Aeruginosa

(Pa)

Infection.

1176 1177

115. Study to Evaluate Arikayce™ in CF Patients With Chronic Pseudomonas Aeruginosa

1178

Infections.

1179

https://clinicaltrials.gov/ct2/show/NCT01315678?term=NCT01315678&rank=1

1180 1181

116. Inhalation SLIT Cisplatin (Liposomal) for the Treatment of Osteosarcoma Metastatic to

1182

the Lung. https://clinicaltrials.gov/ct2/show/NCT00102531?term=NCT00102531&rank=1

1183

48 | P a g e

1184

117. Evaluation of a Therapeutic Strategy Including Nebulised Liposomal Amphotericin B

1185

(Ambisome®) in Maintenance Treatment of Allergic BronchopulmonaryAspergillosis

1186

(Cystic Fibrosis Excluded). (NEBULAMB)

1187

https://clinicaltrials.gov/ct2/show/NCT02273661?term=NCT02273661&rank=1

1188 1189

118. Value of Amphotericin B Inhalation for Prophylaxis of Invasive Pulmonary

1190

Aspergillosis

1191

Transplantation.https://clinicaltrials.gov/ct2/show/NCT00986713?term=NCT00986713&rank

1192

=1

After

Renal

1193 1194

119.LipoAerosol©

1195

Tracheostomy.https://clinicaltrials.gov/ct2/show/NCT02157129?term=NCT02157129&rank=

1196

1

Inhalation

After

1197 1198

120. Phase II Study of Inhaled AeroLEF for Analgesia After ACL Knee Surgery (Pain).

1199

https://clinicaltrials.gov/ct2/show/NCT00791804?term=NCT00791804&rank=1.

1200 1201

121. Study Evaluating Inhaled AeroLEF Delivered in 4 Aerosol Delivery Devices in Healthy

1202

Volunteers

1203

https://clinicaltrials.gov/ct2/show/NCT00794209?term=NCT00794209&rank=1.

(LEF-07).

1204 1205

122.

Farjadian

1206

Nanopharmaceuticals and

1207

opportunities. Nanomedicine (Lond). 2019;14(1):93-126.

1208

49 | P a g e

F, Ghasemi

A, Gohari

O, Roointan

nanomedicines currently on

A, Karimi

M, Hamblin

MR.

the market: challenges and

1209

123. Limeres MJ, Moretton MA, Bernabeu E, Chiappetta DA, Cuestas ML. Thinking small,

1210

doing big: Current success and future trends in drug delivery systems for improving cancer

1211

therapy with special focus on liver cancer. Mater Sci Eng C Mater Biol Appl. 2019;95:328-

1212

341.

1213 1214

124. Giovagnoli S, Schoubben A, Ricci M. The long and winding road to inhaled TB therapy:

1215

not only the bug’s fault. Drug Dev. Ind. Pharm. 2017;43(3):347-363.

1216 1217

125. Mehta PP, Kadam SS, Pawar AP. Influence of modified induction port, modified DUSA

1218

assembly and device air-inlet geometry on the aerosolization pattern of a dry powder inhaler.

1219

J Drug Deliv Sci Technol. 2020;55:101416.

1220 1221

126. Rogueda P, Traini D. The future of inhalers: how can we improve drug delivery in

1222

asthma and COPD? Expert Rev Respir Med. 2016; 10:1041-1044.

1223 1224

127. Guillon A, Sécher T, Dailey LA, Vecellio L, de Monte M, Si-Tahar M et al. Insights on

1225

animal models to investigate inhalation therapy: relevance for biotherapeutics. Int J

1226

Pharm. 2018;536(1):116-126.

1227 1228

128. Vulović A, Šušteršič T, Cvijić S, Ibrić S, Filipović N. Coupled in silico platform:

1229

Computational fluid dynamics (CFD) and physiologically-based pharmacokinetic (PBPK)

1230

modelling. Eur J Pharm Sci. 2018;113:171-184.

1231

50 | P a g e

1232

129. Backman P, Arora S, Couet W, Forbes B, de Kruijf W, Paudel A. Advances in

1233

experimental and mechanistic computational models to understand pulmonary exposure to

1234

inhaled drugs. Eur J Pharm Sci. 2018;113:41-52.

1235 1236

130. Mehta PP. Dry powder inhalers: upcoming platform technologies for formulation

1237

development. Ther Deliv. 2019 Sep;10(9):551-554.

1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256

51 | P a g e

1257

Figure captions

1258 1259

Graphical Abstract - Applications and interaction of liposomes with the pulmonary

1260

membrane. Liposomes are synthesized from substances endogenous to pulmonary airways

1261

such as pulmonary surfactants and hence, explored in the pulmonary drug delivery field

1262

owing to their versatile attributes. By interacting with the pulmonary membrane,

1263

multifunctional liposomes can adhere to the mucosal surface for a longer time, open

1264

intercellular tight junctions in pulmonary epithelia along with modified drug release kinetics

1265

and reduced dosing frequency to improve therapeutic outcomes.

1266 1267

Figure 1 - Liposome types based on lamellarity and size. SUV: small unilamellar vesicles,

1268

LUV: large unilamellar vesicles, GUV: giant unilamellar vesicles, MLV: multilamellar

1269

vesicles and MVV: multivesicular vesicles (vesosomes). Moreover, a fraction of classic lipid

1270

bilayer with hydrophilic head groups and hydrophobic alkyl chains shown.

1271 1272

Figure 2 - Applications and interaction of liposomes with the pulmonary membrane.

1273

Liposomes are synthesized from substances endogenous to pulmonary airways such as

1274

pulmonary surfactants and hence, explored in the pulmonary drug delivery field owing to

1275

their versatile attributes. By interacting with the pulmonary membrane, multifunctional

1276

liposomes can adhere to the mucosal surface for a longer time, open intercellular tight

1277

junctions in pulmonary epithelia along with modified drug release kinetics and reduced

1278

dosing frequency to improve therapeutic outcomes. (MRT: mean resisdance time; Cmax: the

1279

maximum concentration of the drug achieved in the plasma and PEG: polyethylene glycol).

1280

52 | P a g e

1281

Figure 3 -Graphical representation depicting the mechanism of action for liposomal

1282

andrographolide DPI used in regulating immune responses (Lie et al., 2017) [81]. The figure

1283

showed that liposomal andrographolide DPI regulates immune responses by downregulating

1284

various inflammatory pathways such as inhibit phosphorylation of kappa B (IκB-α) inhibitor

1285

and reduced the expression of intercellular adhesion molecule (ICAM) family 1, Interferon-γ

1286

(IFN-γ) in immunocytes.

1287 1288

Figure 4 -Benefits and challenges of multifunctional inhaled liposomes. The figure showed

1289

crucial technical, biological and formulations benefits as well as challenges related to the

1290

design and development of multifunctional inhaled liposomes. This figure emphasis various

1291

key factors such as chemistry, manufacturing and control (CMC), center for devices and

1292

radiological health (CDRH), forced vital capacity (FVC) and forced expiratory volume

1293

(FEV). (IPR: intellectual property rights, ADME: absorption, distribution, metabolism, and

1294

excretion and MRT: mean resisdance time).

53 | P a g e

Table 1 - Advantages and disadvantages of liposome preparation methods Preparation

Particle size Advantages

method

(nm)

Disadvantages

References

Conventional method Thin-lipid

film 100-1000

hydration

A

simple,

conventional Low yield, difficult to scale up, low [38-40]

process

encapsulation

efficacy,

heterogeneous

particle size Reverse-phase

100-1000

High entrapment efficacy

Organic

evaporation Ethanol

solvent

traces,

difficult

to [39-41]

manufacture at large scale injection 70-200

Ability to control vesicle size

method

Reproducibility, difficult to remove ethanol [39,40,42] traces, heterogeneous particle size, require high temperature

Novel methods Microfluidic technology

100-300

High

entrapment

suitable homogenous

1|Page

for

efficacy, Prerequisites

of

equipment,

various [29,40,

scale-up, processing conditions; thus need more 43,44]

particle

size, optimization

appropriate

for

template-

based manufacturing Membrane

~ 100

contactor

Homogenous high

particle

entrapment

size, Encapsulation requires optimization

[40,45]

efficacy,

suitable for scale up Electrospray technology

100-500

Continuous

method,

encapsulation efficiency

high Prerequisites processing optimization

2|Page

of

equipment,

parameters;

thus

various [29,46,47] critical

Table 2 - Advantages and limitations of novel carrier-based pulmonary drug delivery system Advantages

Limitations

Protection from biological environment

Complex and multi-step process

(enzymes, hydration, pH variation) Masking of immunogenicity resulting from API

High cost

Both hydrophilic and hydrophobic API can be easily loaded

Complex control of reproducibility i.e. particle size distribution and/or surface charge

Controlled and modified drug release

Need to conduct toxicological studies

Triggered released in response to physical or chemical different Need to gather and understand ADME pattern stimuli (pH/temperature) API: active pharmaceutical ingredient; ADME: absorption, distribution, metabolism and elimination

3|Page

Table 3- Developed liposomal formulations for pulmonary delivery Drug

Method Key ingredient

Particle

size Device

FPF (%)

References

(nm) SS

FD

SPC, α-LM and MgSt (0.5 %)

137.00 (LUVs)

Spinhaler®

41.51

[76]

Docetaxel

SD

PC, CHOL, mannitol and leucine

346.80 (LUVs)

Reusable

10.10

[71]

device N-acetylcysteine

SD

SPC, CHOL and α-LM

117.00 (LUVs)

HandiHaler®

35.34

[84]

Ciprofloxacin

SD

HSPC, CHOL and sucrose

131.30 (LUVs)

Osmohaler®

69.70

[85]

Liquorice extract FD

SPC, CHOL and trehalose

212.60 (LUVs)

Lupihaler®

54.68

[82]

Andrographolide

FD

Soybean lecithin, CHOL and mannitol

77.91 (SUVs)

Twister®

23.03

[81]

SS

SD

SPC, CHOL and α-LM

167.20 (LUVs)

Rotahaler®

64.01

[77]

SS

SD

DPPC, CHOL and α-LM

-

Cyclohaler®

42.70

[86]

Ketotifen

FD

EPC, CHOL and sucrose

-

Rotahaler®

21.59

[87]

Dapsone

SD

DPPC, CHOL and α-LM

137.00 (LUVs)

-

75.60

[88]

Clarithromycin

FD

SPC, CHOL, mannitol (15 %) and 370.00 (LUVs)

-

53.78

[78]

fumarate

4|Page

sucrose (5 %) Gemcitabine

FD

HSPC, DSPG, CHOL mPEG2000- 331.42 (LUVs)

-

56.12

[72]

DSPE and trehalose Curcumin

FD

SPC, CHOL and mannitol

94.65 (SUVs)

-

46.71

[79]

Oseltamivir

SD

Ovelecithin, CHOL and leucine

105.90 (LUVs)

-

35.40

[74]

Tacrolimus

SD

HSPC, CHOL and trehalose

140.00 (LUVs)

-

71.10

[89]

Rifampicin

FD

SPC, CHOL and mannitol

255.00 (LUVs)

-

66.80

[90]

Moxifloxacin

SD

PC, CHOL and dextran

272.00 (LUVs)

-

72.08

[75]

phosphate

SS: salbutamol sulphate; SD: spray drying; FD: freeze drying; SPC: Soya phosphatidylcholine; CHOL: cholesterol; HSPC: hydrogenated soybean

phosphatidylcholine;

DSPG:

1,2-Distearoyl-sn-glycero-3-phosphoglycerol;

dipalmitoylphosphatidylcholine and EPC: egg phosphatidylcholine

5|Page

PC:

phosphatidylcholine;

DPPC:

Table 4 - A summary of ongoing clinical trials for liposomal formulations Product

Condition or disease

Intervention/ treatment

Phase

Identifier

Sponsor

Arikayce™

Cystic fibrosis

280 mg of matching placebo

I and II

NCT00777296

Insmed Incorporated., US

Arikayce™

Cystic fibrosis

70 mg; 140 mg and 560 mg

I and II

NCT00558844

Insmed Incorporated., US

Cisplatin

Osteosarcoma

24 mg/m2 once daily

I and II

NCT00102531

Insmed Incorporated., US

liposomes

metastatic

for 14 days

Cyclosporine

Bronchiolitis obliterans

2.5 mg/10 mg x 2/day

I and II

NCT01334892

Pari

liposomes

for 96 weeks

Cyclosporine

Lung

liposomes

and

transplantation -

Pharma

GmbH,

Germany I and II

NCT01650545

bronchiolitis

University of Maryland, US

obliterans Arikayce™

Cystic fibrosis

560 mg once daily dose for 6 II

NCT03905642

Insmed Incorporated., US

NCT03038178

Kevin Winthrop, Insmed

cycles over 18 months Arikayce™

MBI and NMBI

590 mg for 12 months

II

Incorporated and etc. Ambisome®

6|Page

Allergic

25 mg x 1/ week for 6 months

II

NCT02273661

Poitiers

University

bronchopulmonary

Hospital, France

aspergillosis Ambisome®

Lung transplantation

-

II

NCT01254708

University

Health

Network, Toronto Liposomal Nitro-20

9- Lung cancer (S)-

5 consecutive days per week II

NCT00250120

X 8 weeks, every 10 weeks

University

of

New

Mexico, US

camptothecin Amikacin

MBI and NMBI

590 mg/day for 12 months

II

NCT03038178

liposomes

Kevin Winthrop and Insmed Incorporated., US and etc.

Arikayce™

MBI and NMBI

590 mg administered

III

NCT02344004

Insmed Incorporated., US

III

NCT03270514

Kathirvel Subramaniam

III

NCT02628600

Insmed Incorporated., US

once daily Bupivacaine

Coronary artery disease

liposomes Liposomal amikacin

7|Page

Bupivacaine liposomes 226 mg

Non-tuberculous for infections

590 mg/day

inhalation Ambisome®

Lung

transplantation 1 mg/kg/day for 4 days

III

NCT00177710

and fungal infections

University of Pittsburgh and Astellas Pharma US, Inc.

Ciprofloxacin

Non-cystic

fibrosis -

liposomes

bronchiectasis

III

NCT01515007

Grifols

(ORBIT-3) Cyclosporine

Bronchiolitis obliterans

Twice daily inhalation for a III

NCT01439958

maximum of three years

Ambisome®

Chronic

with

aspergillosis

for

24

weeks)

Pari

Pharma

GmbH,

Germany

pulmonary Ambisome (25 mg x 2/week III

itraconazole

NCT03656081

and

Poitiers

University

Hospital, France

itraconazole (200 mg x 2/day)

Ciprofloxacin

Non-cystic

liposomes

bronchiectasis

fibrosis -

III

NCT02104245

Aradigm Corporation Grifols

(ORBIT-4)

8|Page

Therapeutics

LLC

liposomes

Liposomal

Aradigm Corporation and

Therapeutics

LLC Lung cancer

-

-

NCT00277082

University

of

New

camptothecin LipoAerosol®

Mexico, US Tracheostomy complications

5x/d for 30 min

-

NCT02157129

Technical University of Munich, Germany

ArikayceTM : amikacin liposome inhalation suspension; Ambisome ®: amphotericin B liposomal; MBI: mycobacterial infection and NMBI: nonmycobacterial infection.

9|Page

AUTHOR DECLARATION TEMPLATE

We wish to draw the attention of the Editor to the following facts which may be considered as potential conflicts of interest and to significant financial contributions to this work. We confirm that the manuscript has been read and approved by all named authors and that there are no other persons who satisfied the criteria for authorship but are not listed. We further confirm that the order of authors listed in the manuscript has been approved by all of us. We confirm that we have given due consideration to the protection of intellectual property associated with this work and that there are no impediments to publication, including the timing of publication, with respect to intellectual property. In so doing we confirm that we have followed the regulations of our institutions concerning intellectual property. We understand that the Corresponding Author is the sole contact for the Editorial process (including Editorial Manager and direct communications with the office). He/she is responsible for communicating with the other authors about progress, submissions of revisions and final approval of proofs. We confirm that we have provided a current, correct email address which is accessible by the Corresponding Author and which has been configured to accept email from [email protected]; [email protected]

Corresponding author; Dr. Vividha Dhapte-Pawar, Associate Professor, Department of Pharmaceutics, Bharati Vidyapeeth University, Poona College of Pharmacy, Pune -411038 Email: [email protected]; [email protected]