You are on page 1of 107

Accepted Manuscript

Lipid based nanocarriers: A Translational Perspective

Dinesh K. Mishra, Ruchita Shandilya, P.K. Mishra

PII: S1549-9634(18)30478-7
DOI: doi:10.1016/j.nano.2018.05.021
Reference: NANO 1826
To appear in: Nanomedicine: Nanotechnology, Biology, and Medicine
Received date: 1 May 2018
Accepted date: 28 May 2018

Please cite this article as: Dinesh K. Mishra, Ruchita Shandilya, P.K. Mishra , Lipid based
nanocarriers: A Translational Perspective. Nano (2018), doi:10.1016/j.nano.2018.05.021

This is a PDF file of an unedited manuscript that has been accepted for publication. As
a service to our customers we are providing this early version of the manuscript. The
manuscript will undergo copyediting, typesetting, and review of the resulting proof before
it is published in its final form. 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.
ACCEPTED MANUSCRIPT

Lipid based nanocarriers: A Translational Perspective

Dinesh K. Mishra1*, Ruchita Shandilya, P. K. Mishra2


1
NMIMS, School of Pharmacy & Technology Management, Shirpur (Maharashtra) 425405,

India
2
Department of Molecular Biology ICMR-National Institute for Research in Environmental

T
Health, Bhopal, India.

IP
CR
* Corresponding author
US
AN
Dr. Dinesh Kumar Mishra

Associate Professor,
M

NMIMS, School of Pharmacy & Technology Management, Shirpur (Maharashtra) 425405


ED

dineshdops@gmail.com; Tel/Fax: +91-2563286545/+91-2563286552


PT

# The author(s) declare(s) that there is no conflict of interest regarding the publication of this
CE

article.

Word count:
AC

Abstract- 146

Manuscript (body text & figure legends)-9025

References- 200

Number of figures-12

Number of tables-05

1
ACCEPTED MANUSCRIPT

Abstract

Over the recent couple of decades, pharmaceutical field has embarked most phenomenal

noteworthy achievements in the field of medications as well as drug delivery. The rise of

Nanotechnology in this field has reformed the existing drug delivery for targeting, diagnostic,

remedial applications and patient monitoring. The convincing usage of nanotechnology in the

T
conveyance of medications that prompts an extension of novel lipid-based nanocarriers and non-

IP
liposomal systems has been discussed. Present review deals with the late advances and updates

CR
in lipidic nanocarriers, their formulation strategies, challenging aspects, stability profile, clinical

applications alongside commercially available products and products under clinical trials. This

US
exploration may give a complete idea viewing the lipid based nanocarriers as a promising choice
AN
for the formulation of pharmaceutical products, the challenges looked by the translational

process of lipid-based nanocarriers and the combating methodologies to guarantee the headway
M

of these nanocarriers from bench to bedside.


ED

Key words: Drug delivery, Lipid carrier, Nanotechnology, Nanocarrier, Controlled release
PT
CE
AC

2
ACCEPTED MANUSCRIPT

Abbreviations:

NEMS: Nano electromechanical systems


MEMS: Micro-electromechanical systems
NE: Nanoemulsions
NPs: Nanoparticles
QD: Quantum dots

T
PLA: Polylactic acid

IP
PGA: Polyglycolic acid

CR
PLGA: Co-polymers of lactic and glycolic acid
PCL: Poly(ε-caprolactone)

US
PACA: Poly-alkyl cyanoacrylate
PEI: Polyethyleneimine
AN
PTX: Paclitaxel
M

GIT: Gastrointestinal tract


PEG: Polyethylene glycol
ED

RES: Reticuloendothelial system


FA: Folic acid
PT

EPR: Enhanced penetration and retention


CE

PAMAM: Polyamidoamine
PG: Poly L-glutamic acid
AC

PPI: Polypropyleneimine
LA: Lactobionic acid
CNTs: Carbon nanotubes
DOX: Doxorubicin
SW: Single walled
MW: Multiwalled
MNPs: Metallic nanoparticles

3
ACCEPTED MANUSCRIPT

LBDDS: Lipid based drug delivery systems


DSC: Differential scanning calorimeter
ME: Microemulsions
MMEs: Mucoadhesive ME
AB: Amlodipine besilate
SEDDS: Self emulsifying drug delivery systems

T
ADV: Adefovirdipivoxil

IP
PE: Pickering emulsions

CR
PC: Phosphatidylcholine
MLVs: Multilamellar vesicles

US
SUVs: Small unilamellar vesicles
LUVs: Large unilamellar vesicles
AN
HACE: Hyaluronic acid-ceramide
M

SF: Sorafenib
Gd: Gadolinium
ED

MDR: Multidrug resistance


Tf: Transferrin
PT

DOX: Doxorubicin
OVA: Ovalbumin
CE

SC: Stratum corneum


AC

MTX: Methotrexate
LS: Lipospheres
SLMs: Solid lipid microparticles
SLN: Solid lipid nanoparticles
CNS: Central nervous system
NLC: Nanostructured lipid carriers
LDCS: Lipid drug conjugates

4
ACCEPTED MANUSCRIPT

BHT: Butylatedhydroxytoluene
BHA: Butylatedhydroxyanisole
MPS: Mononuclear phagocytic system

T
IP
CR
US
AN
M
ED
PT
CE
AC

5
ACCEPTED MANUSCRIPT

1. Nanotechnology

The journey of drug from its development to its translation into the market is very tedious and

experiences incredibly little accomplishment owing to numerous factors like toxicity of the

therapeutic agents, inadequate solubility leading to lesser bioavailability and consequently

reduced efficiency. In order to combat this, nanotechnology based drug delivery systems is one

T
of the approaches that has been investigated for this purpose. Nanotechnology is the science and

IP
technology of precise manipulation in the materials, devices or systems at nanometer scale

CR
(usually less than 100 nm) (Figure 1)1, 2. The last several decades have witnessed the emergence

of nanomedicine as one of the major field of academic research providing direct benefit to

US
human health through clinical and commercial development. The broadened applications of
AN
nanomedicine and exploitation of nanotherapeutics in the clinic is only possible due to increased

understanding of the disease and designing of advanced nanoscale materials. Nanomedicines


M

include nanopharmaceuticals (i.e. intended for drug delivery), nanodiagnostics (i.e. used for
ED

imaging and diagnostics), nanotheranostics (i.e. combined therapeutic and diagnostic), and

nanobiomaterials (i.e. medical implants). Among these, nanopharmaceuticals are predominant


PT

and represent 75% of the market share of approved nanomedicines. The ever growing field of
CE

development of nanoscale delivery systems for biotherapeutics represents a major sector of

academic research and is beginning to contribute to the future progress in modern health care in
AC

terms of disease diagnosis, treatment, and prevention. Although significant progress has been

made in these fields but the clinical translation of next-generation nanopharmaceuticals remains

challenging and needs advanced exploration of the manufacturing and regulatory perspectives.

Nanotechnology (nanocarriers) based drug delivery systems may lead to better half-life, precise

6
ACCEPTED MANUSCRIPT

release completed short or extended durations, and identical site specific targeted delivery of

therapeutic compounds3-7.

Attributes of nanocarriers5

 Drugs protection from metabolic degradation.

 Availability of products with superior characteristics.

T
 Dosing frequency may be reduced.

IP
 Efficient and improved treatment with less expense.

CR
 Increased potential of solubilization.

 Drugs rejected earlier or difficult to administer can be utilized by these carriers.

 Feasibility of drug targeting.


US
AN
 Enhanced oral bioavailability of the agents which are not effective after oral

administration.
M

 Flexibility of surface manipulation.


ED

 Capability of designing specific to drug and disease.

2. Classification of nanocarriers
PT

Nanotechnology basically offers two kinds of nanotools viz. nanomaterials and nanodevices, that
CE

offer a foremost contribution in the area of pharmaceuticals and related areas (Figure 2).

Nanomaterials can be enriched by addition of functionalities to interface with biological


AC

molecules or structures. Nanomaterials are subcategorized into nanocrystalline and

nanostructured materials. Nanodevices are minute devices at nanoscale and a few of them

include nano and micro-electromechanical systems (NEMS/ MEMS), microfluidics, and

microarrays. Nanocarrier can be broadly classified as organic-based, inorganic-based or a hybrid

combination of both. Organic nanocarriers comprise of polymeric frameworks, lipid-based

7
ACCEPTED MANUSCRIPT

frameworks e.g., liposomes and nanoemulsions (NE), dendrimers, and carbon-based nanocarriers

e.g., fullerenes and carbon nanotubes. Inorganic nanocarriers comprise of metallic

nanostructures, silica nanoparticles (NPs), and quantum dots (QD)8.

2.1.1 Polymeric NPs:

Polymeric NPs are synthesized by different types of natural and synthetic polymers, which are

T
often biocompatible and biodegradable. The merits of these polymeric particles as compared to

IP
other nanocarriers include stability in various microenvironments, slow release of the drug as a

CR
function of the polymer degradation, as well as their versatility in terms of polymer type and type

of drug to be encapsulated. The hydrophobicity and hydrophilicity inside the polymeric

US
framework can be controlled to suit wide assortment of medication molecules 9,10. Commonly
AN
employed natural polymers include gelatin, dextran, albumin, chitosan, alginate while synthetic

biodegradable polymers are polylactic acid (PLA), polyglycolic acid (PGA), copolymers of
M

lactic and glycolic acid (PLGA), poly(ε-caprolactone) (PCL), poly-alkyl cyanoacrylate (PACA),
ED

polyethyleneimine (PEI), poly(L-lysine), poly(aspartic acid), etc.1,11.

DOXIL, Abraxane and Genexol-PM exists among these first generation NPs, established for
PT

therapy of cancer. Abraxane (nab-paclitaxel) is based on the NP albumin-bound (nab) platform


CE

led to the second class of therapeutic NP which was approved by the FDA in 2005. Abraxane

revealed considerably greater tumour response rates (33% vs. 19%) and elongated times to
AC

tumour progression (23.0 vs. 16.9 weeks) among metastatic breast cancer patients who did not

respond to combination therapy as compared to paclitaxel (PTX) formulated with Cremophor EL

(Taxol), 12.

Surface manipulation like PEGylation, co-polymerization with functional polymers,

polyelectrolyte coatings and ligand anchoring of polymeric NPs is also done to attain desirable

8
ACCEPTED MANUSCRIPT

characteristics. The hydrophobicity and surface charge of the formulated NPs can be modified to

transform the assimilation energy from the gastrointestinal tract (GIT). It includes surface

functionalization. Peracchia et al. investigated the biodistribution characteristics of polyethylene

glycol (PEG) coated polycyanoacrylate NPs that exhibited prolonged circulation, reduced

hepatic accumulation and in-vitro toxicity. Xie et al. reported a PEG conjugated Fe3O4 NPs that

T
demonstrated targeted delivery with concomitant reduction in the macrophage uptake by

IP
reticuloendothelial system (RES) 13.

CR
The properties of polymeric NPs could be advantageous for the treatment of several life-

threatening diseases including cancer, neurodegenerative disorders, cardiovascular diseases, and

US
even other diseases, such as viral infections and osteoporosis. Dhas et al. investigated a prostate
AN
cancer targeted delivery systems of bicalutamide-loaded folic acid (FA) conjugated chitosan

functionalized PLGA nanocarriers14.


M

2.1.2 Dendrimers:
ED

Initially proposed by Tomalia in the year 1985, dendrimers are unimolecular, monodisperse,

micellar with well-defined size and structure, which are created from repeating units, like
PT

polymers, but they basically vary from classical polymers. There are three fundamental reasons
CE

that legitimize the utilization of dendrimers for drug delivery:

 Firstly, the presence of multiple copies of a drug induce a multivalency effect,


AC

reminiscent to the polyvalent interactions extensively arising in biological systems.

 Secondly, enhancement of the solubility and thus achieving increased the bioavailability.

 Thirdly, the varying size that may exceeds the renal threshold, and may induce the so-

called enhanced penetration and retention (EPR) effect15.

9
ACCEPTED MANUSCRIPT

However, the deficiency of this carrier is its synthesis because of their complex design. Polymers

that are commonly used in dendrimers for drug delivery are: polyamidoamine (PAMAM),

melamine, poly L-glutamic acid (PG), polyethyleneimine (PEI), polypropyleneimine (PPI), and

(PEG), Chitin etc.16,17. There have been several studies reporting the enhanced therapeutic

efficacy and better targetability of drugs when administered as dendrimers. Fu et al. detailed the

T
advancement of a lactobionic acid (LA)-modified multifunctional generation 5 (G5) PAMAM

IP
dendrimer-based carrier system for targeted therapy of liver cancer cells 18.Cao et al. reported

CR
doxorubicin encapsulated, folate-functionalized, degradable amphiphilic dendrimer-like star

polymer (FA-DLSP) to be used as a nanoscale carrier for cancer cell targeted drug

US
delivery19.Wang et al. reported a targeted anticancer effect via development of multifunctional
AN
PAMAM dendrimer-based nanocarrier20.

2.1.3 Micelles:
M

Micelles have become one of the main players in nanoparticle research to be utilized in advanced
ED

drug delivery systems. These are supramolecular assembly or systems constituting hydrophobic

core enclosed by hydrophilic corona. For the past several years, polymer-based micelles have
PT

been the main focus of researchers as carrier systems considering their numerous benefits such as
CE

better stability, high drug loading capacity, improved solubilization, prolonged circulation,

smaller size and targeting potential. Their small size (1-50 nm) makes them superior for
AC

intravenous delivery. Additionally they are more stable in comparison to liposomes and

biocompatible. Once ready to be administered, micelles are injected and enter target cells by

endocytosis through ligand-receptor interactions. Targeted delivery of docetaxol, doxorubicin

(DOX) to tumor cells by means of micelles was well demonstrated by Chen et al21.

10
ACCEPTED MANUSCRIPT

2.1.4 Polymer drug conjugates:

Polymer-medicate conjugates are the new medication conveyance frameworks made out of

covalently connected medication particles with the polymer backbone. Apart from drug

molecules, additional purposeful additives like diagnostic agent, targeting ligand, PEG chains to

enhance hydrophilicity can also be housed in the polymer backbone which brings about

T
arrangement of nanocargos, specific to disease condition. Further, targeting potential offered by

IP
polymer-drug conjugate is especially noteworthy if there should be an occurrence of growth

CR
treatment because of greatly lethal impacts of anticancer agents to the normal tissues of the body.

The classical approach for formulation of polymer–drug conjugates can be comprehensively

US
arranged as PEGylation that involves covalent binding of PEG chains with drug molecules. It is
AN
found that PEGylation may improve solubility, permeability and minimized presystemic

metabolism4, 17, 23. Parvizikhosroshahi and Can developed polymeric drug conjugates to attain
M

targeted delivery of bioactive molecules22.


ED

2.1.5 Carbon nanotubes (CNTs):

CNTs have emerged as a new delivery system and their specific design as well as
PT

physicochemical properties represents as potential drug carrier system. They belong to fullerenes
CE

(made up of carbon atom and present as hollow sphere, tube) that are cylindrical in shape and

consist of a hexagonal arrangement. According to structure and arrangement of atoms they are of
AC

two types: single walled (SW) CNTs and multiwalled (MW) MWCNTs. Depending upon

functionalization they may be further categorized as target oriented, ligand attached, solvent

dispersed and surfactant grafted. They have extensive applications in healthcare sector including

from pharmaceutical companies in developing target oriented drug delivery medicines, biotech

companies in developing numerous novel vaccine and blood products as well as biomedical

11
ACCEPTED MANUSCRIPT

industries in designing nano-sensors (glucose sensors, blood pressure/temperature sensor), nano

diagnostic tools, nano-probes and nano-robots23-25.

A wide array of studies have been put forward proposing CNTs as promising candidates for drug

delivery. Ji et al. attempted to achieve targeted controllable loading/release of anti-cancer agent

doxorubicin (DOX) via construction of chitosan modified folic acid conjugated SWNTs26. Ren et

T
al. fabricated a dual-targeting system consisting angiopep-2 functionalized oxidized MWCNTs

IP
for delivery of doxorubicin for treatment of brain glioma27. Lu et al. developed a magnetic dual-

CR
targeted nanocarrier combining the advantages of MWCNTs and iron oxide magnetic NPs for

delivery of doxorubicin to the target tumor cells28.

2.1.6 Metallic nanoparticles (MNPs):


US
AN
MNPs are very promising as novel carrier and contrast agents for the treatment of cancer. The

unique features of MNPs like high surface-volume ration, wide optical properties, easy synthesis
M

and simple surface chemistry and functionalization cling to assurance in the clinical ground for
ED

cancer treatment. MNPs (gold, silver or blend of two) show highly tunable optical characteristics

which can be easily tuned to required wavelength and enable them for imaging and photothermal
PT

applications. Current developments have directed to site specific targeting and drug delivery by
CE

these particles29.

Several studies proving the ability of metallic nanoparticles to achieve improved drug delivery
AC

and therapeutic efficacy have been reported. Paciottiet al.30 and Maoet al.31 reported targeted and

improved delivery of drugs by means of gold nanoparticle. Hadjipanayis et al. reported the use of

iron oxide NPs conjugated to an anti-synthetic peptide antibody (EGFRvIIIAb) for MRI contrast

enhancement of human glioblastoma cells 32.

12
ACCEPTED MANUSCRIPT

2.1.7 Quantum dots (QDs):

They are inorganic fluorescent semiconductor nanoparticles having size range of 2-10 nm that

comprise a center of hundreds to thousands atoms. QDs with cadmium selenide center and a zinc

sulphide shell, encompassed by a covering of organizing ligand and an amphiphilic polymer are

most extensively used for biological application. They possess certain benefits over conventional

T
fluorescent dyes: narrower emission spectra, bigger strokes shift, high quantum yield, greater

IP
photostability. QDs can release toxic cadmium via photolysis on excitation that is not desirable.

CR
To overcome this problem system can be coated with biocompatible polymers which may

improve colloidal stability. The surface of QDs can be engineered or manipulated to improve

US
solubility, sensitivity, specificity and visualization in target tissue8, 33.
AN
Many targeted, image-guided drug delivery studies have been reported by Feng et al.34, Justin et

al.35 Muhammad et al.36 and Al-Nahain et al.37 proving the potential of QDs for multimodal
M

therapy.
ED

2.1.8 Silica nanoparticles:

Silica materials are the carriers which mostly preferred for biological purposes among inorganic
PT

NPs. The mechanism by which drug loading can be done into mesoporous silica material is a
CE

chemical or physical adsorption. These processes can incorporate various types of drugs,

including anticancer drugs antibiotics, and cardiac drugs into MSNs. These particles have
AC

applications in catalysis, drug delivery and imaging38. Targeted and improved doxorubicin drug

delivery was reported by Meng et al.39, Pan et al.40 and Meng et al. 41 by means of functionalized

mesoporous silica NPs.

13
ACCEPTED MANUSCRIPT

2.2 Nanodevices:

Microarray is very vital technology in the area of diagnostics, particularly in genetic analysis. It

can be defined as a matrix of numerous entity elements immobilized on a solid support by means

of bulk phase mass transfer approach specifically by direct spotting of biomolecules on the

support. They display persuasive apparatuses for biomedical scientists and medication, as

T
exhibits can be designed to look at the event of atomic marks in a to a great degree parallel mold

IP
and can be arranged to look for either for nucleic acids (DNA microarrays) or proteins (immune

CR
response based microarrays) and additionally different kinds of cells. On the other hand

microfluidics shows the capability to evaluate miniature volumes (micro, nano or even pico-

US
liters) of sample and diminish costly reagent expenditure as well as automatic sample preparation
AN
with reduced sample processing time. The combination of microarray technologies and rising

area of microfluidics offers numerous benefits like minimization of reagent cost, less time
M

requirement for hybridization assay, high-throughput processing of sample, integration and


ED

automation capacities of the front-end sample processing steps42, 43.

3. Lipid based drug delivery systems (LBDDS)


PT

Lipids are generally organic compounds which contain hydrocarbons and extracted from plants
CE

and animals by low polarity solvents. Common examples of lipids are oils, waxes, cholesterol,

sterols, monoglycerides, diglycerides, triglycerides, phospholipids and fat soluble vitamins.


AC

Recently lipids are becoming very popular as potential excipients for drug delivery as well as

cosmetics44. Among the array of NPs being as of now examined by pharmaceutical researchers,

lipid nanoparticles have led the pack on account of clear preferences of higher level of

biocompatibility and flexibility. These frameworks are monetarily appropriate to define

pharmaceuticals for topical, oral, pneumonic or parenteral organization and delivery. Lipid nano

14
ACCEPTED MANUSCRIPT

formulations can be custom fitted to meet extensive variety of product requirements directed by

disease condition, route of administration and contemplations of cost, product stability, toxicity

and viability. These systems emerge as an alluring candidate for the formulation of

pharmaceuticals, as well as immunizations, diagnostics and nutraceuticals as a result of their

demonstrated safety and efficacy45.

T
Based on chemical composition solid lipids can be categorized as:

IP
a. Homolipids

CR
Examples: Cerides (Waxes- beeswax, carnauba wax etc.), Glycerides (fats and oils) and Sterides

(esters of cholesterol with fatty acids)

b. Heterolipids
US
AN
Examples: Phospholipids (Phosphoglycerides, Phosphosphingolipids-ceramide), Glycolipids and

Sulfolipids
M

c. Complex lipids
ED

Examples: Lipoproteins, Chylomicrons

3.1 Useful properties of lipids in the formulation of LBDDS44


PT

1. Crystallinity and polymorphism


CE

Appropriate crystallinity is a criterion for a good quality lipid particulate delivery systems.

Triglycerides, which are principally utilized as lipid networks solidify in various polymorphic
AC

structures.

2. Melting characteristics

A pure triacylglycerol is having single melting point which occurs at a definite temperature.

However certain lipids contain different types of triacylglycerols with multiple melting points

which results their melting over a broad range of temperatures. It produces a wide endothermic

15
ACCEPTED MANUSCRIPT

transition in differential scanning calorimeter (DSC). Highly pure lipids with sharp melting

transitions do not include drugs on recrystallization.

Solubility is a great challenge for the formulation scientists. Hence, LBDDS have gained much

importance in the recent years due to their capability to improve the solubility and ultimately

bioavailability. As of now huge endeavors have been made to investigate the possibilities of

T
LBDDS since it gives the reasonable methods for site specific as well as time controlled delivery

IP
of drugs with different molecular weight, either little or huge, and furthermore the bioactive

CR
operators 46 47.

3.1 Reason behind exploration of lipid based systems48, 49:

 Adaptability of lipidic excipients


US
AN
 Versatility in designing of formulation

 Low hazard profile


M

 Greater oral bioavailability with minimum plasma profile variation


ED

 Superior permeation in case of topical delivery

 Possibility of commercialization
PT

 Better representation of lipidic excipients


CE

 High market appealing qualities for products with innovation

 Improved ability to address the key issues of technology transfer and manufacture scale
AC

up.

3.2 Attributes of LBDDS

 Controlled and targeted drug release.

 Pharmaceutical stability.

 High and improved drug content as compared to other carrier systems.

16
ACCEPTED MANUSCRIPT

 Capability for incorporation of both lipophilic as well as hydrophilic drugs.

 Biodegradable and biocompatible.

 Better dose uniformity.

 Requirement of low therapeutic dose owing to better drug absorption.

 Low risk profile.

T
 Easy scale up and sterilization.

IP
 Easy validation.

CR
 Economic than polymeric or surfactant based systems.

 Lowering of therapeutic dose due to improved drug absorption.

US
 Administration versatility i.e., can be administered via various routes such as oral,
AN
parenteral, ocular, intranasal, dermal/transdermal, and vaginal.

3.3 Mechanism of drug transport & Formulation strategies46, 48, 50


M

There are numerous factors which needs consideration for the formulation of LBDDS includes
ED

absorption, compatibility, cost, digestion, dispersion, melting point, purity, regulatory aspect,

solubility, stability, toxicity. Many approaches have been put forward for the formulation of lipid
PT

based drug delivery systems as shown in Figure 3a.


CE

Various transfer mechanisms are involved in the transportation of drug across the membrane by

means of lipid based drug delivery systems as shown in Figure 3b. These may include:
AC

i. Paracellular absorption,

ii. M cell mediated transport,

iii. Fluid phase macropinocytosis,

iv. Endocytosis (clathrin/caveolin mediated),

v. Ligand mediated transport, and

17
ACCEPTED MANUSCRIPT

vi. Enhanced uptake by size regulation.

3.4 Classification of LBDS:

LBDDS are classified as follows Figure 3c& Table 1

3.4.1 Emulsion based systems:

3.4.1.1 Microemulsions (ME)

T
The concept of ME was introduced by Hoar and Schulman in the year 1940. They are

IP
characterized as straightforward, less gooey, thermodynamically steady, optically isotropic

CR
arrangement of oil and water balanced out by an interfacial film of amphiphilic mixes, for

example, surfactant and co-surfactant. The key difference between emulsion and ME is the size

US
and shape of dispersed particles. ME size ranges from 10-200 nm while emulsions size ranges
AN
from 1-20 µm. ME have created great interest as prospective drug delivery systems as well as

cosmetic and cosmeceutical carriers for skin and hair preparations. Some benefits associated
M

with these systems are thermodynamic stability, great solubilization potential, clarity, easy
ED

preparation and scale up51. Several studies have shown the impact of ME based delivery through

various routes providing better targetability. Topical drug delivery of clobetasol propionate by
PT

ME based systems was reported by Patel et al.52 ME-base hydrogel formulation for topical
CE

delivery of ibuprofen. Shah et al. meant to figure ME and mucoadhesive ME (MMEs) of

rivastigmine for nose to brain delivery and to compare percentage drug diffused for the two
AC

frameworks utilizing as a part of vitro and ex-vivo contemplate 53.

3.4.1.2 Nanoemulsions (NEs)

NEs are emulsions having droplet size in the range of 100 nm. Classic NEs consists of oil, water

and an emulsifier. An emulsifier is very essential for the formation of minute sized droplets

because it reduces the interfacial tension i.e., the surface energy per unit area, among the oil and

18
ACCEPTED MANUSCRIPT

aqueous phases of the emulsion. They also engaged in stabilization of NEs by means of repulsive

electrostatic interactions and steric hindrance. The emulsifier employed is usually a surfactant,

but proteins and lipids are also efficient in the formation of NEs. NEs have been recognized over

few decades owing to their extraordinary characteristics like high surface area, transparent

occurrence, dynamic stability and tunable rheology. NEs are biodegradable, biocompatible, easy

T
to prepare and transporters for lipophilic drugs which are susceptible to hydrolysis. NEs have

IP
been applied in different fields such as drug delivery, food, cosmetics, pharmaceuticals, and

CR
material synthesis54.

Mahajan et al. developed an intranasal NE for enhanced bioavailability and CNS targeting of

US
saquinavirmesylate (SQVM)55. Modi et al. investigated the anti-inflammatory potential of a NE
AN
formulation for topical delivery of aceclofenac56.Chhabra et al. reported to achieve enhanced

solubility and oral bioavailability of amlodipine besilate (AB) by a development of a targeted NE


M

based formulation57.
ED

3.4.1.3 Self emulsifying drug delivery systems (SEDDS)

They are isotropic mixtures of oil, surfactant, co-surfactant and the therapeutic substance which
PT

emulsify under gentle agitation. SEDDS quickly distribute in the GIT with moderate agitation
CE

provided by gastric motility which cause formation of emulsion. SEDDS usually construct

emulsions with cloudy appearance, and droplet size between 200 nm to 5 µm. They are superior
AC

in comparison to lipid solution due to the existence of surfactants in the preparations leading to a

more uniform and reproducible bioavailability. Specific advantages associated with these carriers

include better steady drug absorption, specific targeting potential, drug protection from the gut

environment, controlled delivery, minimized variability accounting food effects, improved oral

19
ACCEPTED MANUSCRIPT

bioavailability with decreased dose and elevated drug loading capacity. SEDDS is one of the

potential techniques for oral delivery of water insoluble or poorly soluble compounds58.

Chen et al. developed a supersaturatable SEDDS for the enhanced oral bioavailability of

indirubin59.Milovi´c et al. investigated a solid SEDDS, as potential delivery system for poorly

water soluble drug carbamazepine (CBZ)60. To overcome the limitations of poor solubility,

T
stability and bioavailability SEDDS of curcumin was developed by Wu et al.61 Balakumar et al.

IP
developed SEDDS of Rosuvastatin calcium62. Seo et al. investigated the improved bioavailability

CR
and chemotherapeutic potential of Docetaxel loaded SEDDS when compared with Taxotere®, a

commercial product of docetaxel63. Gupta et al. reported the development of Adefovirdipivoxil

US
(ADV) loaded SEDDS exhibiting increased bioavailability due to enhancement of its intestinal
AN
permeability and minimized effect of pH with improved therapeutic efficacy64.

3.4.1.4 Pickering emulsions (PE)


M

PE are very promising preparations owing to its simplicity and similarity to the well-recognized
ED

surfactant-based emulsions. They may be any type of emulsions either oil-in water (o/w), water-

in-oil (w/o), or even multiple, which is stabilized by solid particles instead of surfactants. The
PT

main advantage of the stabilization by solid particles is high resistance to coalescence. PE are
CE

emulsions based on lipid having inner nanostructures stabilized by solid particles like silica,

clays, calcium carbonate, titanium dioxide etc. These emulsions can be substituted for a typical
AC

emulsion in most of the applications because PE hold the basic properties of typical emulsions.

They are well suited to particular fields like cosmetic and pharmaceutical where surfactant may

cause adverse effects51.

PE with its attractive advantageous attribute of stabilization present an effective strategy to

design potent carriers in biomedical area. The stability and release of curcumin have been

20
ACCEPTED MANUSCRIPT

65
evaluated in nanoparticle stabilized PE by Tikekar et al. and Shah et al.66 during storage and

simulated gastric and intestinal digestion.

3.4.2 Vesicles based systems

3.4.2.1 Liposomes

Liposomes can be defined as microscopic, colloidal, concentric bilayered vesicles comprising of

T
an aqueous compartment enclosed by a bilayer of lipids which can be of either natural or

IP
synthetic lipids (Figure 4a).The essential components of liposomal drug delivery system include

CR
phospholipids like phosphatidylcholine (PC) and cholesterol where cholesterol acts as a fluidity

buffer. In spite of the fact that cholesterol don't take part in bilayer formation, it can be added to

US
PC up to 1:1 or even 2:1 molar ratio of cholesterol to PC. They can be formulated and
AN
characterized for various sizes, number of lamellae, structure, composition, lipids

physicochemical property and different payloads. As there are numerous types of phospholipids,
M

it is feasible to alter the size, charge, surface properties, upon addition of new ingredients to the
ED

lipid mixture.

They can be categorized in to different types depending on their size and number of bilayers
PT

includes multilamellar vesicles (MLVs), small unilamellar vesicles (SUVs) and large unilamellar
CE

vesicles (LUVs). Liposomes contain lipid bilayers which are biocompatible and may improve the

solubility and stability of encapsulated agents, also modify tissue distribution, targeted to
AC

particular sites, augment therapeutic response and minimize toxicity. They are a versatile tool for

delivery of large array of bioactives ranging from drugs to high molecular weight proteins and

peptides. They can be explored for oral, ocular, pulmonary and transdermal delivery of drugs.

The amphipathic nature of liposomes renders them to be explored as a non-invasive delivery

agent for vaccines. Thus they can be structured to present control over properties like elimination

21
ACCEPTED MANUSCRIPT

half-lives, permeability, biodistribution and targeting specificity67, 68. The last few decades have

witnessed wide array of development in liposomes providing better therapeutic efficacy and

better targetability along with imaging.

Park et al. reported a nanohybrid approach consisting doxorubicin loaded liposomes coated with

amphiphilic hyaluronic acid-ceramide (HACE) for targeted delivery of anticancer drug and in

T
vivo cancer imaging69. Ninomiya et al. demonstrated an ultrasound-triggered TSP liposomes for

IP
drug delivery to HepG2 cancer cells70. Yan et al. reported to achieve enhanced delivery of

CR
doxorubicin to cells residing in a sub physiologic pH environment through a pH-responsive

liposomal nanocarrier71. Xiao et al. developed a sorafenib (SF) and gadolinium (Gd) co-loaded

US
liposomes (SF/Gd-liposomes) with an intent to achieve improved solubility of sorafenib, to
AN
monitor its distribution and the early feedback effects on its in vivo treatment efficacy72. Meng et

al. first reported a co-encapsulation of Resveratrol with PTX in a PEGylated liposome to


M

overcome multidrug resistance (MDR) in tumor cells73. Deshpande et al. reported a R8 and
ED

transferrin (Tf) modified DOX-loaded liposomes to achieve improved targeting of A2780

ovarian carcinoma cells via the over-expressed Tf receptors (TfRs) with R8-mediated
PT

intracellular DOX delivery74.


CE

Many mechanisms are involved in the transportation as well as enhanced availability of drug

when delivered as liposomal and non-liposomal lipid based systems as shown in Figure 4b.
AC

These may involve

a) Drug free mechanism in which the drug permeates through the membrane independently

after exiting from the vesicles.

b) Penetration mechanism(as in ethosomes), in which a penetration enhancer (like ethanol)

interacts with lipid molecules of the membrane leading to increased fluidity and

22
ACCEPTED MANUSCRIPT

decreased density of the membrane followed by penetration and permeation through

opened new pathways.

c) Ability to deform and pass through narrow constriction and reform, a characteristic

feature of transfersomes without measurable loss.

3.4.2.2 Niosomes

T
Niosomes, showing structural resemblance with liposomes, are minuscule multi-lamellar non-

IP
ionic surfactant vesicles formed as a result of addition of non-ionic surfactants (primarily alkyl or

CR
dialkylpolyglycerol ether class) to cholesterol with subsequent hydration in aqueous media

(Figure 4c). Niosomes exhibit increased size and charge on the vesicles due to addition of non-

US
ionic surfactants which further leads to increase their entrapment efficiency. These systems are
AN
very close to liposomes in functions and also enhance the bioavailability of the substances with

reduced clearance. They alleviate the limitations related to classic liposomes like chemical
M

instability, variable purity of phospholipids and high cost. Additionally, they have more
ED

penetrating potential than the classic liposomes. He et al. reported to improve the bioavailability

and chemical stability of Paeonol by formation of PEGylated niosomes resulting in its prolonged
PT

cellular uptake and enhanced synergistic anticancer effects with 5-Fu75. Jyoti et al. prepared
CE

inhalable curcumin loaded cationic small unilamellar niosomes (Cur-C-SUNS) to surmount the

poor physicochemical and biopharmaceutical limitations for effective drug delivery in lung
AC

cancer cells76. Mehta et al. reported to prepare biocompatible niosomal formulation of anti-

retroviral drug, Nevirapine by a biological surfactant, tyloxapol for the achievement of for better

patient compliant pharmacotherapy for acquired immune deficiency syndrome77. Auda et al.

developed different niosomal gel preparations of celecoxib that showed significant anti-

inflammatory effect concluding to be as potential carriers78. Qumbar et al. formulated lacidipine

23
ACCEPTED MANUSCRIPT

(LAC) loaded niosomal formulation using transdermal delivery for the management of

hypertension79.

3.4.2.3 Pharmacosomes

They are the colloidal dispersions of drugs with covalent bonding to lipids. Since this system is

developed by a drug to a carrier hence termed as pharmacosomes. They may exist as ultrafine

T
vesicles, micelles or hexagonal aggregates depend upon the chemical structure of the drug-lipid

IP
complex. They could be an efficient tool to acquire desired purposes like drug targeting and

CR
controlled delivery80. Semalty et al. reported to formulate a diclofenac-phospholipid complex

(pharmacosomes) exhibiting improved solubility improved solubility and dissolution profile with

US
reduced gastrointestinal toxicity of the drug81. Kamlesh D et al. formulated ketoprofen loaded
AN
pharmacosomes to achieve improved solubility, bioavailability, protect active ingredients from

degradation in the gastrointestinal tract and avoid GI disturbance of ketoprofen82. With an


M

objective of achieving enhanced solubility, absorption and bioavailability of BCS class III drug,
ED

Pal et al. formulated and evaluated pharmacosomes of Rosuvastatin Calcium83.

3.4.2.4 Phytosomes/herbosomes
PT

Phytosomes also known as herbosomes are cell like design which is generated from the
CE

stoichiometric interaction of phospholipids with standard extract or polyphenolic constituents.

They are well absorbed than traditional herbal extracts. Due to its amphiphilic characteristics
AC

herbosomal formulations may increase the bioavailability of active phytochemical constituents

since they can readily permeate and cross the lipid membrane. The active constituent of herbal

extract is also secured from enzymatic degradation as well as gut bacteria. These systems have

gained wide attention in various areas like pharmaceuticals, cosmeceuticals and nutraceuticals 67.

Yu et al. a self-assembly technique was employed for the preparation of berberine-phospholipid

24
ACCEPTED MANUSCRIPT

complex loaded phytosomes to cater the limitations of low oral bioavailability and poor

gastrointestinal absorption84. Abdelkader et al. reported to develop hyaluronic acid-phytosomes

loaded with L-carnosine as an alternative approach for the prodrug N-acetyl-L-carnosine as a

novel ocular delivery system85. Mirzaei et al. reported to improve the oral bioavailability of

curcumin by means of phytosomal preparations86.

T
3.4.2.5 Elastic liposomes

IP
A new type of manipulated liposome with an optimum quantity of edge activator or surfactant

CR
which gives elasticity has been developed (Transfersome ®; Idea AG). This type of novel

vesicular system was initially described by Cevc and Blume (Figure 4d). Elastic liposomes

US
(transfersomes) are particularly optimized, ultradeformable lipid supramolecular aggregates
AN
possessing the ability for drug deliverance into or through the skin and ultimately reaches to the

systemic circulation87. Elastic liposomes were basically designed to get the benefits of
M

phospholipids vesicles as transdermal drug carrier. Their transdermal delivery is dependent on


ED

the choice of administration or application, with high efficiency. Their elastic nature makes him

appropriate for skin delivery and also minimizes the risk of complete vesicle rupture. It follows
PT

the natural water gradient across the epidermis for skin permeation under non-occlusive
CE

condition. Elastic liposomes can accommodate hydrophobic as well as hydrophilic moieties due

to its infrastructure. They can go about as a bearer for low and in addition high sub-atomic
AC

weight substances e.g. analgesic, anesthetic, corticosteroids, sex hormone, anticancer, insulin,

gap junction protein, and albumin. Since, they are made up of natural phospholipids hence

biocompatible and biodegradable in nature. They also possess greater entrapment efficiency and

shield the enveloped moieties from metabolic degradation. They also act as depot therefore

releases their contents slowly. They may be applied for local as well as systemic delivery of

25
ACCEPTED MANUSCRIPT

drugs and easy in formulation. Although it possess numerous benefits but also suffers from

certain limitations like susceptible to oxidative degradation, purity of lipids, and cost of

development88-90.

Transfersomes based transdermal drug delivery system of clindamycin was reported by

Abdellatif et al.91providing good stability characteristics and enhanced transdermal delivery.

T
Salama et al. investigated an intranasal formulation of olanzapine encapsulated transfersomes for

IP
brain targeted drug delivery92. Sarwa et al. investigated the biopotential of an antiarthritic agent,

CR
capsaicin loaded transfersomes in arthritic rats93. Various formulation parameters and processing

conditions affecting the properties of transfersomes were investigated by Ahmed et al.94

3.4.2.5 Ethosomes
US
AN
Ethosomes or ethanolic liposomes, are new lipid based, non-invasive carrier system that helps to

deliver biologically active agents to deeper layer of skin layer and systemic circulation (Figure
M

4e). These vesicular systems are mainly consists of phospholipids, higher concentration of
ED

ethanol (20-50%) and water95, 96. In comparison to well established liposomal system they show

greater transdermal flux. Although the actual mechanism lying behind the transdermal
PT

permeation of ethosomes is not clear but it is believed that phospholipids and higher percentage
CE

of ethanol are responsible for efficient skin permeation. Generally ethanol in ethosomal systems

disrupt the skin lipid bilayer organization resulting in enhanced skin penetration. Due to higher
AC

percentage of ethanol the lipid membrane is less tightly packed as compared to classic vesicles

but stability is same. They are soft malleable vesicles and enhance drug distribution capacity in

skin. These ethanolic vesicles are also able to incorporate hydrophilic, lipophilic or amphiphilic

molecules88, 97.

26
ACCEPTED MANUSCRIPT

Ethosomes have been known to show high transdermal permeability providing better skin

tolerability of drugs like lamivudine, melatonin, and repaglinide as reported by Jain et al.98,

Dubey et al.95, and Bodade et al.99, respectively.

3.4.2.7 Archaeosomes

Archaeosomes are also nothing but liposomes made up of fully saturated bipolar tetra ether

T
lipids. They are found to be highly stable as compared to conventional lipids in various

IP
conditions like high temperature, acidic or alkaline pH, and oxidative stress etc. They are nano

CR
range vesicles formulated from lipids extracted from Archaeobacteria or synthetic archaeal

lipid100. They provide a promising approach for drug delivery. Alavi et al. provided an

US
archaeosomal approach for delivery of paclitaxel in breast cancer cells with increased therapeutic
AN
efficacy and reduced side effects101. Another approach for in vitro skin permeation was put

forward by Moghimipour et al. of the archaeosomes, prepared using lipid extracted from
M

Sulfolobusacidocaldarius102.
ED

3.4.2.8 Vesosomes

They can be defined as a multi-compartment array frameworks possessing discrete inner


PT

compartments alienated from the external membrane are the characteristic features of vesosomes.
CE

Each compartment can incorporate diverse materials and have different bilayer composition.

They can entrap colloidal as well as biomacromolecules effectively. Multiple compartments of


AC

this framework offer better protection to the encapsulated contents in physiological system and

shows extended release of drug 67.

3.4.2.9 Colloidosomes

These are microcapsules whose cell contains tightly packed colloidal particles. Their physical

characteristics like permeability, mechanical strength, and biocompatibility can be specifically

27
ACCEPTED MANUSCRIPT

controlled by means of proper selection of colloids and formulation conditions for their

assembly. The better control over their physical characteristics makes these system attractive

arrangements for encapsulation and controlled drug release103. Shah et al explained a versatile

technique using stimuli-responsive microgel particles as building blocks, aqueous droplets, and

microfluidic devices for fabrication of stimuli-responsive colloidosomes104. Nan et al. reported to

T
prepare monodisperse colloidosomes based on chitosan-coated alginate particles for oral delivery

IP
of insulin105. Bollhorst et al. reported a synthetic route for synthesis of bifunctional

CR
colloidosomes from superparamagnetic iron oxide nanoparticles and fluorescent silica

nanoparticles106. Similarly, a straightforward approach was put forward by Zhou et al. for

US
fabrication of thermal stimuli based colloidosomes using thermo-sensitive triblock copolymer107.
AN
3.4.2.10 Aquasomes

Aquasomesor ceramic nanoparticles, self-assembled nanostructures (Figure 4f) as triple layers,


M

comprising of a solid phase nanocrystalline core, coated with an oligomeric film (made up of
ED

carbohydrate) on which biochemically active molecules are adhered with or without

modification. The nanocrystalline core comprises polymers such as acrylate, gelatin, or albumin,
PT

or ceramic such as diamond particles, brushite (calcium phosphate), and tin oxide that provide
CE

structural stability. Coating materials commonly used are trehalose, cellobiose, sucrose,

pyridoxal 5 phosphate, citrate, chitosan, and so forth108. Aquasomes are widely utilized for the
AC

delivery of insulin, haemoglobin109, and enzymes like serratiopeptidase. Pandey et al. reported

the preparation and evaluation of carbohydrate modified ultrafine aquasomes in specific

immunotherapy using ovalbumin (OVA) as an allergen model110.

28
ACCEPTED MANUSCRIPT

3.4.2.11 Cubosomes:

Cubosomes can be characterized as self-assembled, liquid crystalline, thermodynamically stable

nano frameworks, made up of certain amphiphilic lipids in definite proportions, known as

biocompatible carriers in drug delivery. In presence of polar solvents, these particles assemble as

curved bicontinuous lipid bilayers and is divided into two internal aqueous channels that can be

T
exploited by various bioactive moieties. Cubosomes have appeared as one of the promising

IP
vehicles for various routes of administration offering attractive attributes such as the ability to

CR
encapsulate hydrophilic, hydrophobic and amphiphilic substances, thermodynamic stability,

bioadhesion, and controlled release through functionalization. The term “cubosomes” was coined

US
by Larsson, which reflects the cubic molecular crystallography and structural and functional
AN
resemblance with liposomes108, 111.

Cubosomes have been widely used for enhancement of drug delivery via various routes such as
M

oral, topical, ophthalmic etc. For instance, Esposito et al. reported a percutaneous administration
ED

of indomethacin for anti-inflammatory action via cubosome dispersions112. Ophthalmic drug

delivery with low irritancy and high bioavailability of flurbiprofen and dexamethasone as
PT

cubosomes was achieved as reported by Han et al.113 and Gan et al114. Cubosomes as an
CE

alternative approach for the oral drug delivery of amphotericin B was put forward by Yang et

al.115
AC

3.4.2.12 Sphingosomes.

Sphingosomes can be defined as bilayered vesicles, mainly composed of natural or synthetic

sphingolipids and cholesterol in varying ratio, entirely enclosing an aqueous compartment.

Various types of sphingolipids have been used for the fabrication of sphingosomes such as

sphinganines, hexadecasphinganine, lysosphingomyelins, and lysoglycosphingolipids, n-

29
ACCEPTED MANUSCRIPT

acylsphingosines, gangliosides, glucuronosphingolipids, phosphoglycosphingolipids, and so

forth108. Recently, Tiwari et al. suggested an investigative study of sphingosomes for the delivery

of anti-cancer drug such as Dox116.

3.4.13. Ufasomes:

Ufasomes appears as promising approach for efficient topical/ transdermal delivery of drugs,

T
proteins, peptides, hormones, etc. as they enhance penetration of drug into viable skin through

IP
stratum corneum (SC). Ufasomes containing lipid carriers that attached to the skin surface and

CR
allows lipid exchange between the outermost layers of the SC.

Initially named as “ufasomes,”reflecting unsaturated fatty acid liposomes was first reported by

US
Gebicki and Hicks in 1973. Ufasomes are suspensions of closed lipid bilayers that are composed
AN
of fatty acids and ionic surfactants108. Cutaneous delivery of dexamethasone loaded ufasomes for

anti-inflammatory activity was reported by Mittal et al117. To overcome the limitations of oral
M

administration of cinnarizine, intranasal ufasomes based formulation was reported by Salama et


ED

al118.

3.4.14. Emulsomes
PT

Emulsomes are nanocarriers consisting of a solid fat core enclosed by ones or more phospholipid
CE

bilayers with an integrated properties of emulsion and liposomes. Vyas et al. reported a novel

formulation based on emulsomes for the liver-targeted delivery of anti-viral drug, zidovudine119.
AC

A chylomicron mimicking carrier based on emulsomal approach was developed by Paliwal et al.
120
for oral lymphatic delivery of methotrexate (MTX) . Brain targeted drug delivery via

intranasal route was achieved by oxcarbamazepine loaded lipidic emulsomes by El-Zaafarany et

al.121

30
ACCEPTED MANUSCRIPT

3.4.15. Novasomes

Novasomes, a type of liposomal based nanocarrier with diameter ranging from 0.1 up to 1

micron, consist of a large amphipathic core enclosed by 2-7 bilayers and are produced by

mixture of polyoxyethylene fatty acids (as monoester), free fatty acids and cholesterol. The

surface charge of novasomes could be neutral, positive or negative122. Radwa et al. reported an

T
intranasal delivery of a novasomal based formulation encapsulating zolmitriptan for brain

IP
targeting for management of acute migraine attacks123.

CR
3.4.3. Particulate system

3.4.3.1 Lipospheres

US
Lipid microspheres, also called as lipospheres (LS), are newer type encapsulation based on
AN
lipidic system comprising of water dispersible solid microparticles, with diameter ranging

between 0.1 -100 μm, developed for delivery of therapeutically active compounds particularly
M

lipophilic substances.. These consist of solid hydrophobic core (triglyceride fat) which is
ED

stabilized by phospholipids embedded in its surface. The innermost core comprises of

therapeutic compound dispersed in the lipid matrix. These are utilized for the controlled delivery
PT

of various types of drugs including local anesthetics, anti-inflammatory compounds, anticancer,


CE

vaccines, and antibiotics agents67, 124. LS display better physical stability due to avoidance of

coalescence in the formulation. LS are widely used for parenteral nutrition and possess stability
AC

over a time period at room temperature and possessed no particular undesirable effects, even at

high dose levels. These can be prepared by melt dispersion technique, solvent evaporation

technique, solvent evaporation method, multiple ME, sonication method and roto evaporation

method125. LS exhibiting attractive attributes such as better stability and sustained release of the

drugs such as carbamazepine126, aceclofenac127 were reported. Attama et al. reported the

31
ACCEPTED MANUSCRIPT

preparation of PEGylated Ceftriaxone sodium loaded LS exhibiting high permeability and better

oral bioavailability128. Acoustically active LS have been reported by Fang et al.129 providing

better therapeutic delivery.

3.4.3.2 Solid lipid microparticles (SLMs)

SLMs are defined as spherical solid lipid particles having size range from 1 to 1000 µm. These

T
contain polymeric, biodegradable synthetic polymers or waxy protective materials, and modified

IP
natural excipients such as gums, starches, fats, waxes and proteins. These are physicochemically

CR
compatible, physiologically stable and therefore these can be formulated at large scale at a

comparatively low manufacturing cost compared to other vesicular systems or other

US
microparticles. These micron sized particles comprised of solid polymeric core based on
AN
naturally occurring lipids and stabilized by dispersed surfactant molecules. These are having

numerous advantages like controlled drug release, targeting potential, drug protection form
M

degradation, and incorporation of both the hydrophilic and/or hydrophobic drugs. These can be
ED

prepared by: high pressure homogenization; cold homogenization; solvent evaporation and spray

drying techniques. These can be administered via oral route, parenteral route, pulmonary
PT

administration, topical route, ocular administration and rectal administration124, 125. El-Kamel et
CE

al. reported the fabrication and characterization of testosterone SLMs for the transdermal

delivery130. Controlled drug delivery via pulmonary route was demonstrated by Jaspart et al.
AC

taking salbutamol acetonide as a model drug131. The anti-inflammatory effect of curcumin loaded

SLMs was studied by Yadav et al. for the treatment of inflammatory bowel disease132.

3.4.3.3 Solid lipid nanoparticles (SLN)

SLN are novel colloidal delivery systems comprise of biocompatible lipid nucleus and an

amphiphilic surfactant in the outer shell with size range of 50 to 1,000 nm (Figure 4g). These

32
ACCEPTED MANUSCRIPT

symbolize an unconventional carrier system to conventional drug delivery systems, such as

emulsions, liposomes, and polymeric micro- and nanoparticles. In the 1980s, Speiser and co-

workers first reported SLN for drug delivery applications. These are aqueous colloidal

dispersions, comprising matrix made of solid biodegradable lipids and provide dispersion site for

both hydrophilic as well as hydrophobic drug moiety. These solid lipids are used instead of

T
liquid oils proffer good drug dispersion and controlled drug release, as the drug mobility in a

IP
solid lipid should be very much lower contrast to liquid oil68, 133, 134.

CR
SLN are very much advantageous as in nano-size range these proffer relatively narrow size

distribution requisite for biological prospect for site specific targeted drug delivery; provide

US
protection against chemical degradation of drug in formulation, these are free from organic
AN
solvents and suitable for industrial scale production. These can be formulated by techniques like

high pressure homogenization, cold homogenization, solvent evaporation, supercritical fluid


M

technology, ME based techniques and spray drying techniques. Possible routes of administration
ED

for SLN include peroral administration, parenteral administration and transdermal

administration. The drug distribution in SLN governs the type of release pattern which can be
PT

achieved by selecting proper ingredient and the type of technique for SLN production. Possibly
CE

three strategies which offer the drug distribution pattern and consequently the release behavior

patterns includes
AC

(a) Homogeneous matrix: these are formed when the melting points of drug and lipid becomes in

steady state;

(b) Lipid-enriched core: these are resulting when lipid melts at higher temperature than that of

drug; and

33
ACCEPTED MANUSCRIPT

(c) Drug encapsulated core: these are formed when lipids liquefy at an early point than that of

drug.

Drug loading may bring about strong changes of the SLN attributes (particle size distribution,

zeta potential, lipid modifications etc.). Notwithstanding, a few elective joining locales (micelles,

mixed micelles, liposomes, drug nanosuspensions) exist in addition to the complex

T
physicochemical status of the lipid (supercooled melt and several modifications). SLN are

IP
successfully utilized for cancer therapy, vaccine delivery, drug targeting, gene delivery and it is a

CR
choice of carrier for central nervous system (CNS) targeting. Various drugs are also utilized for

delivery through SLN and are commercially available for easy and simple delivery43, 97
.

US
Sarmento et al. reported the delivery of insulin via oral and pulmonary by means of solid lipid
AN
nanoparticles with enhanced absorption135. Various anticancer drugs have been delivered by

means of solid lipid nanoparticles such as 5-Flurouracil136, doxorubicin137 etc. There have been
M

several reports demonstrating solid lipid nanoparticles used for brain targeting. Dhawan et al.
ED

and Martins et al. reported the brain targeted delivery of quercetin and camptothecin138, 139. Solid

lipid nanoparticles for topical delivery of drugs have been reported by Bhalekar et al.140.
PT

3.4.3.4 Nanostructured lipid carriers (NLC)


CE

This system was developed from SLN and it is advanced form of lipid nanoparticles. They are

colloidal carriers and a combination of solid lipids and liquid lipids which yield lipid matrix.
AC

This nanostructure enhance drug loading and maintain the drug stability during storage.

Additionally drug release can be modulated by alteration in lipid matrix composition. This

system can be a better alternative than SLN owing to superior controlled drug release and greater

stability profile. There are three possible types of NLC which are imperfect type, amorphous

type and multiple type67, 141. Various routes have been employed for the delivery of drugs via

34
ACCEPTED MANUSCRIPT

nanostructured lipid carriers with improved bioavailability. For instance, ocular drug delivery by

nanostructured lipid carriers was reported by Shen et al.142 and Luo et al.143. Topical drug

delivery of drugs like meloxicam using nanostructured lipid frameworks has been reported by

Khurana et al.144. Delivery of drugs via oral route for enhancement of bioavailability have been

reported by Fathi et al.145, Jawahar et al.146

T
3.4.3.5 Lipid drug conjugates (LDCS)

IP
LDCS, a novel lipid based nanoparticles which have been broadly opted for the delivery through

CR
site specific targeting. These have been utilized for many features in the field of pharmaceutical

drug therapy. These proffer potential site for hydrophilic and lipophilic drugs, and suitable for

US
CNS targeting. The particles were surface modified to target them to the CNS which builds the
AN
lipid drug conjugates a promising delivery system147. Paliwal et al. reported the development of

lipid drug conjugate of methotrexate to overcome the poor bioavailability and associated
M

gastrointestinal toxicity148. Cancer targeting by means of lipid drug conjugates were reported by
ED

Sharma et al. and Abu-Fayyad et al.149, 150

3.5 Characterization of LBDDS:


PT

LBDDS can be characterized following methods which are given below (Table 2):
CE

3.6 Challenges and stability aspect

LBDDS confront various difficulties in the utilization of other targeted nanocarriers. One of the
AC

real difficulties related with lipids is their helplessness to oxidation particularly for unsaturated

triglycerides and unsaturated fats. It may occur during processing or storage and cause loss in

quality of products. When lipids are encountered with environmental conditions like light, air or

temperature, auto oxidation may occur and lead to changes in texture, color, flavor, low quality

and the formation of toxic compounds with health issues for patients. Other degradation

35
ACCEPTED MANUSCRIPT

pathways are triggered by lipoxygenases enzymes. Metal traces may have a considerable impact

in encouraging oxidation. Nitrogen flushing can be a suitable way to prevent oxidation in closed

systems. Metal based reactions can be evaded by the use of chelating agent (EDTA or citric

acid). Physical and chemical stability of LBDDS that pose challenge of oxidation can be reduced

by the use of saturated triglycerides and suitable antioxidants. Antioxidants can avoid oxidation

T
reaction by diversified mechanisms. Some examples of antioxidants include alpha-tocopherol,

IP
butylatedhydroxytoluene (BHT), butylatedhydroxyanisole (BHA), and propyl gallate etc. To

CR
analyze the effects of antioxidants on oxidative stability various analytical approaches can be

utilized like peroxide value, p-anisidine value, thermogravimetry and scanning calorimetry.

US
Other challenges imparted by LBDDS are rapid clearance from bloodstream, burst drug release,
AN
non-specific uptake by the mononuclear phagocytic system (MPS), elevated production cost and

deficient of controlled-release properties. These challenges and stability issues are limiting in
M

translation of LBDDS to the clinic151.


ED

3.7 Clinical applications

LBDDS having applications in various fields like pharmaceuticals, cosmetics, nutraceuticals etc.
PT

but few of the products has reached to market or still in clinical testing. Some important clinical
CE

applications are given below (Table 3 and Table 4):

4. Contribution by various researchers in the proposed area


AC

Various researchers have contributed in the field of drug delivery using lipid system.

Contributions by the researchers using different therapeutic agents are compiled in the given

table (Table 5).

36
ACCEPTED MANUSCRIPT

5. Conclusion

The last few decades have witnessed an intensive increase in interest in nanotechnology,

specifically in the application of nanostructure based system for drug delivery via various routes.

The advances in the diagnosis and therapy along with the possibility of monitoring of drug

distribution, are all the result of fusion of nanotechnology, materials science, and biotechnology.

T
Pharmaceutical nanotechnology has offered tweaked finding and centered ailment treatment.

IP
Still some moral, logical, social and administrative issues are posturing challenges in the

CR
possibility of pharmaceutical nanotechnology. As such there are no specific guidelines or

directive to control products based on pharmaceutical nanotechnology. Overall these challenges

US
needs to be addressed for regulation of these nanotechnology based products and delivery
AN
devices.

LBDDS have emerged as the most promising candidate of drug delivery and have been the
M

longest-studied carriers. LBDDS offer the enormous array of possibilities for safe, economical
ED

and an encouraging approach for the delivery of hydrophobic drugs having poor bioavailability

that may be largely due to environmentally responsive linkers specifically temperature, pH, ionic
PT

strength, enzyme or oxidative/reductive environment. The distinctive functioning of target site


CE

like slightly acidic/alkaline pH, hypoxia (in case of solid tumors), and overexpression of some

enzymes (e.g., sialidase, matrix metalloproteinase) suggests a great array of opportunities for the
AC

designing of environmentally manipulated delivery systems for drug targeting. They also have

the capability to enhance absorption, sustain the drug release and target at particular site resulting

in improved therapeutic benefits with minimum side effects. The advancement of these

frameworks needs exhaustive comprehension of the physicochemical idea of the medication and

the lipid excipients and gastrointestinal processing. Owing to their biocompatibility, industrial

37
ACCEPTED MANUSCRIPT

scalability and production feasibility, lipid-based nanocarriers provide a great potential of

overcoming various technological and stability limitations of predecessor delivery systems like

liposomes or polymeric NPs, and especially in case of delivery of highly lipophilic and low

soluble actives. The second generation of lipid nanocarriers is emerging day-by-day offering an

awesome potential to speak to a change in outlook in nanomedicine. There are various non-

T
liposomal lipidic nanocarriers for example niosomes, transfersomes and ethosomes with

IP
promising in vitro and in vivo brings about an assortment of utilizations. Currently, there are

CR
very few non-liposomal lipid based nanocarriers under clinical trials or commercialized, but their

precursors, liposomes, have just demonstrated the maximum capacity as nanocarriers both for

US
manufactured medications and biopharmaceuticals deliverance, venturing from the lab seat to the
AN
clinical practice. Liposomes have also undergone various modifications to prepare targeted,

stimuli-responsive, magnetism activated, and many acoustic signals mediated systems for
M

delivery of therapeutic agents. These modified liposomes may include stealth liposomes,
ED

immunoliposomes, genosomes, marinosomes, ultrasomes etc. Among the non-liposomal lipidic

nanocarriers, sphingosomes and virosomes have appeared to be the first prosperous ones by
PT

achieving clinical utilize. Apart from second generation non-liposomal carriers, nanocarriers like
CE

nanocapsules and core-shell NPs have been developed that are a hybrid of lipid and polymeric

carriers, which provide a ‘smart’ environmentally responsive delivery and offering a great
AC

opportunity of integration of their unique physicochemical properties leading to a lipid-based

polymeric materials.

Commercially available lipid based formulations have been prepared by conventional methods,

yielding final products however their transformation from lab scale to the clinic is often faced by

limitations such as lack of homogeneity, contamination with remaining organic solvents,

38
ACCEPTED MANUSCRIPT

complexity of the scale up process from lab scale to large scale manufacturing along with

variable reproducibility of high-quality products . In this frame, the utilization of novel

technologies such as Pastillation, DepoFoam® technology, AKVANO® technology, lipid

multiparticulate systems, Lipidots®, Lipopearl™ etc. are few venturing stones towards

development of promising alternatives approaches with affordable cost, simplicity of the process,

T
high throughputs of lipid based nanoformulations with very much requested physicochemical

IP
properties and relatively unimportant leftover solvents, and additionally their bewildering

CR
flexibility and basic adaptability make them perfect skill for pharmaceutical organizations. In

conclusion LBDDS are very promising and can be potential candidates for drug delivery.

US
Despite of tremendous efforts, only a few formulations gets promoted for clinical use. Therefore,
AN
the current approaches and strategies employed for design and development of these LBDDS for

the delivery of hydrophobic drugs must be keenly taken into consideration.


M

6. Future prospect/ Translational perspective


ED

Pharmaceutical industries are more concerned in the designing of an appropriate drug delivery

system that could be adequately versatile and viable for scale up of the formulation. A drug
PT

delivery system can only be successful when technology transfer will be up-scaled from lab-
CE

scale to large-scale. Drug delivery is restricted to the academia, which should enter clinical

translation and required to be up scaled to industry. Nanotechnology is fast progressing field and
AC

has immense potential especially in deliverance of drug. It has bunches of difficulties to

overcome from innovative and regulatory perspective. However the prospective benefits to

enhancing human wellbeing, over an extensive variety of utilizations, are huge. Still

nanotechnology is in initial stages, yet it is significantly watched that NPs are promising

apparatuses for the better targeted delivery. The safety of many of nanotechnological systems is

39
ACCEPTED MANUSCRIPT

yet to be determined. More consideration to be given to the qualities of different lipid based

formulations available, with the goal that reasonable guidelines and methodology can be created.

The need for future research ought to be to direct human bioavailability examines and to

complete more fundamental examinations on the mechanisms of action of this captivating and

distinctive kind of formulations. There are numerous treatments available today that take a

T
considerable measure of time and exorbitant too. Using nanotechnology based LBDDS, faster,

IP
patient friendly and economic treatments can be established. Usually, drugs work through the

CR
entire body before they reach to the affected part but by the use of these systems drug can be

effectively targeted to specific location. It will lead to efficient drug delivery with minimum side

US
effects. In the blink of an eye present day advancement, showcase acknowledgment of an
AN
assortment of pharmaceutical nanotools and all inclusive intrigue appeared by researchers,

governments and businesses ensure that there is momentous potential and probability of nano
M

lipid based medication conveyance framework in not so distant future. For the translation of lipid
ED

based nanocarriers to the clinic, the various critical aspects such as formulation issues,

toxicological aspects related to the use of nanomaterials, accurate evaluation of absorption,


PT

biodistribution, metabolism and degradation, accumulation and potential negative effects must be
CE

taken into consideration and steps to develop triggering modalities amenable to human

applications and alternative strategies for in vivo stabilization of drug nanocarriers must be
AC

taken. Further functionalized nanocarriers for targeted and triggered release as well as

technological improvements in nanocarrier composition can be expected in the upcoming years.

There is no dithering to accept that in coming years market will be overflowed with these

frameworks and gadgets. However, the clinical evaluation of LBDDS is still in its “infancy.”

Future refinements related to issues in constitution, methodology, characterization, and stability

40
ACCEPTED MANUSCRIPT

at product development stage will unwrap new avenues for lipid based system as a potential

molecular vector for clinical translation.

Figure legends:

Figure 1 Nanotechnology dimensions

T
Figure 2 Classification of nanocarriers

IP
Figure 3a Formulation strategies of lipid based drug delivery systems

CR
Figure 3b Mechanism of transport of drugs by lipid based drug delivery systems

Figure 3c Classification of lipid based systems

Figure 4a Structure of liposomes


US
AN
Figure 4b Mechanism of transport of drug via liposomal and non-liposomal drug delivery

systems
M

Figure 4c Structure of niosomes


ED

Figure 4d Structure of elastic liposomes

Figure 4e Structure of ethosomes


PT

Figure 4f Structure of aquasomes


CE

Figure 4g Structure of SLN


AC

41
ACCEPTED MANUSCRIPT

References:

1. Niemirowicz K, Car H. Nanocarriers as modern transport media in drug delivery systems.

Chemik 2012; 66(8):868-81.

2. Duggal D. Role of nanotechnology in new drug delivery systems. Int J Drug Dev Res

T
2011;3(4):4-8.

IP
3. Ragelle H, Danhier F, Preat V, Langer R, Anderson DG. Nanoparticle-based drug

CR
delivery systems: a commercial and regulatory outlook as the field matures. Expert Opin

US
Drug Deliv 2017;14(7):851-64.

4. Safari J, Zarnegar Z. Advanced drug delivery systems: Nanotechnology of health design


AN
A review. J Saudi Chem Soc 2014;18(2):85-99.
M

5. Jain N, Jain R, Thakur N, Gupta BP, Jain DK, Banveer J, et al. Nanotechnology: a safe
ED

and effective drug delivery system. Asian J Pharm Clin Res 2010;3:159-65.
PT

6. Khan DR. The use of nanocarriers for drug delivery in cancer therapy. J Cancer Sci Ther

2010;2:58-62.doi:10.4172/1948-5956.1000024
CE

7. Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an


AC

emerging platform for cancer therapy. Nat Nanotechnol 2007;2(12):751-

60.doi:1038/mnano.2007.387

42
ACCEPTED MANUSCRIPT

8. Jabr-Milane L, van Vlerken L, Devalapally H, Shenoy D, Komareddy S, Bhavsar M, et

al. Multi-functional nanocarriers for targeted delivery of drugs and genes. J Control Rel

2008;130(2):121-8.

9. Thanki K, Gangwal RP, Sangamwar AT, Jain S. Oral delivery of anticancer drugs:

challenges and opportunities. J Control Rel 2013;170(1):15-40.

T
IP
10. Beck-Broichsitter M, Merkel OM, Kissel T. Controlled pulmonary drug and gene

CR
delivery using polymeric nano-carriers. J Control Rel 2012;161(2):214-24.

US
11. Desai PP, Date AA, Patravale VB. Overcoming poor oral bioavailability using

nanoparticle formulations-opportunities and limitations. Drug Discovery Today Technol


AN
2012;9(2):e87-e95.https://doi.org/10.1016/j.ddtec.2011.12.001
M

12. Kamaly N, Xiao Z, Valencia PM, Radovic-Moreno AF, Farokhzad OC. Targeted

polymeric therapeutic nanoparticles: design, development and clinical translation. Chem


ED

Soc Rev 2012;41(7):2971-3010.doi:10.1039/C2CS15344K


PT

13. Xie J, Xu C, Kohler N, Hou Y, Sun S. Controlled PEGylation of Monodisperse Fe3O4


CE

Nanoparticles for Reduced Non‐Specific Uptake by Macrophage Cells. Adv Material

2007;19(20):3163-6.https://doi.org/10.1002/adma.200701975
AC

14. Dhas NL, Ige PP, Kudarha RR. Design, optimization and in-vitro study of folic acid

conjugated-chitosan functionalized PLGA nanoparticle for delivery of bicalutamide in

prostate cancer. Powder Tech 2015;283:234-

45.https://doi.org/10.1016/j.powtec.2015.04.053

43
ACCEPTED MANUSCRIPT

15. Caminade A-M, Turrin C-O. Dendrimers for drug delivery. J Materials Chem B

2014;2:4055-66.doi:10.1039/C4TB00171K

16. Raj R, Mongia P, Kumar Sahu S, Ram A. Nanocarriers based anticancer drugs: current

scenario and future perceptions. Curr Drug Target2017;17(2):206-28.

T
17. Ruiz ME, Gantner ME, Talevi A. Applications of nanosystems to anticancer drug therapy

IP
(Part II. Dendrimers, micelles, lipid-based nanosystems). Recent Pat Anticancer Drug

CR
Discov 2014;9(1):99-128.

US
18. Fu F, Wu Y, Zhu J, Wen S, Shen M, Shi X. Multifunctional lactobionic acid-modified

dendrimers for targeted drug delivery to liver cancer cells: investigating the role played
AN
by PEG spacer. ACS Appl Mater Interfaces 2014;6(18):16416-25.
M

19. Cao W, Zhou J, Mann A, Wang Y, Zhu L. Folate-functionalized unimolecular micelles

based on a degradable amphiphilic dendrimer-like star polymer for cancer cell-targeted


ED

drug delivery. Biomacromolecules 2011;12(7):2697-707.


PT

20. Wang Y, Guo R, Cao X, Shen M, Shi X. Encapsulation of 2-methoxyestradiol within


CE

multifunctional poly(amidoamine) dendrimers for targeted cancer therapy.

Biomaterials2011;32(12):3322-9.
AC

21. Chen Q, Zheng J, Yuan X, Wang J, Zhang L. (2017). Folic acid grafted and tertiary

amino based pH-responsive pentablock polymeric micelles for targeting anticancer drug

delivery.Materials Sci EngC2018;82:1-9.

22. Parvizikhosroshahi S, Can HK.Synthesis and characterization of novel polymer-drug

conjugate based on the anhydride containing copolymer as a potential method for drug

44
ACCEPTED MANUSCRIPT

carrier. J Macromol Sci Part A. 2018;55(2):192-

204.https://doi.org/10.1080/10601325.2017.1410064

23. Jain AK, Kumar Mehra N, Lodhi N, Dubey V, Mishra DK, Jain PK, et al. Carbon

nanotubes and their toxicity. Nanotoxicology 2009;1(3):167-97.

T
24. Liu Z, Tabakman S, Welsher K, Dai H. Carbon Nanotubes in Biology and Medicine: In

IP
vitro and in vivo Detection, Imaging and Drug Delivery. Nano Res 2009;2(2):85-120.

CR
25. Mehra NK, Jain AK, Lodhi N, Raj R, Dubey V, Mishra D, et al. Challenges in the use of

US
carbon nanotubes for biomedical applications. Crit Rev Ther Drug Carrier Syst

2008;25(2):169-206.
AN
26. Ji Z, Lin G, Lu Q, Meng L, Shen X, Dong L, et al. Targeted therapy of SMMC-7721 liver
M

cancer in vitro and in vivo with carbon nanotubes based drug delivery system. J Colloid

Interface Sci 2012;365(1):143-9.


ED

27. Ren J, Shen S, Wang D, Xi Z, Guo L, Pang Z, et al. The targeted delivery of anticancer
PT

drugs to brain glioma by PEGylated oxidized multi-walled carbon nanotubes modified


CE

with angiopep-2. Biomaterials 2012;33(11):3324-33.


AC

28. Lu YJ, Wei KC, Ma CC, Yang SY, Chen JP. Dual targeted delivery of doxorubicin to

cancer cells using folate-conjugated magnetic multi-walled carbon nanotubes. Colloids

Surf B Biointerfaces 2012;89:1-9.

29. Bhattacharya R, Mukherjee P. Biological properties of "naked" metal nanoparticles. Adv

Drug Deliv Rev 2008;60(11):1289-306.

45
ACCEPTED MANUSCRIPT

30. Paciotti GF, Zhao J, Cao S, Brodie PJ, Tamarkin L, Huhta M, et al. Synthesis and

evaluation of paclitaxel-loaded gold nanoparticles for tumor-targeted drug

delivery.Biocon Chem 2016;27(11): 2646-2657.

31. Mao W, Kim HS, Son YJ, Kim SR, Yoo Hs. Doxorubicin encapsulated clicked gold

nanoparticle clusters exhibiting tumor-specific disassembly for enhanced tumor

T
IP
localization and computerized tomographic imaging.J Controlled Rel2018;269: 52-62.

CR
32. Hadjipanayis CG, Machaidze R, Kaluzova M, Wang L, Schuette AJ, Chen H, et al.

EGFRvIII antibody-conjugated iron oxide nanoparticles for magnetic resonance imaging-

US
guided convection-enhanced delivery and targeted therapy of glioblastoma. Cancer Res
AN
2010;70(15):6303-12.doi: 10.1158/0008-5472.CAN-10-1022

33. Yong KT, Wang Y, Roy I, Rui H, Swihart MT, Law WC, et al. Preparation of quantum
M

dot/drug nanoparticle formulations for traceable targeted delivery and therapy.


ED

Theranostics 2012;2(7):681-94.
PT

34. Feng T, Ai X, An G, Yang P, Zhao Y. Charge-Convertible Carbon Dots for Imaging-

Guided Drug Delivery with Enhanced in Vivo Cancer Therapeutic Efficiency. ACS Nano
CE

2016;10(4):4410-20.
AC

35. Justin R TK, Román S, Chen D, Xu Y, Geng X, et al. Photoluminescent and

superparamagnetic reduced graphene oxide–iron oxide quantum dots for dual-modality

imaging, drug delivery and photothermal therapy. Carbon 2016;97:54-70.

46
ACCEPTED MANUSCRIPT

36. Muhammad F, Guo M, Guo Y, Qi W, Qu F, Sun F, et al. Acid degradable ZnO quantum

dots as a platform for targeted delivery of an anticancer drug. J Materials

Chem2011;21(35):13406-12.

37. Abdullah Al N, Lee JE, In I, Lee H, Lee KD, Jeong JH, et al. Target delivery and cell

imaging using hyaluronic acid-functionalized graphene quantum dots. Mol Pharm

T
IP
2013;10(10):3736-44.

CR
38. Amato G. Silica-encapsulated efficient and stable si quantum dots with high

biocompatibility. Nanoscale Res Lett 2010;5(7):1156-60.

39.
US
Meng H, Liong M, Xia T, Li Z, Ji Z, Zink JI, et al. Engineered design of mesoporous
AN
silica nanoparticles to deliver doxorubicin and P-glycoprotein siRNA to overcome drug

resistance in a cancer cell line. ACS Nano 2010;4(8):4539-50.


M

40. Pan L, He Q, Liu J, Chen Y, Ma M, Zhang L, et al. Nuclear-targeted drug delivery of


ED

TAT peptide-conjugated monodisperse mesoporous silica nanoparticles. J Am Chem Soc


PT

2012;134(13):5722-25.
CE

41. Meng H, Mai WX, Zhang H, Xue M, Xia T, Lin S, et al. Codelivery of an optimal

drug/siRNA combination using mesoporous silica nanoparticles to overcome drug


AC

resistance in breast cancer in vitro and in vivo. ACS Nano 2013;7(2):994-1005.

42. Sun H, Chen GY, Yao SQ. Recent advances in microarray technologies for proteomics.

Chem Biol 2013;20(5):685-99.

43. Miller MB, Tang YW. Basic concepts of microarrays and potential applications in

clinical microbiology. Clin Microbiol Rev 2009;22(4):611-33.

47
ACCEPTED MANUSCRIPT

44. Gonnade YR, Niranjane, K., Ambatkar, A. Lipid: an emerging platform for lipid based

drug delivery system. World J Pharm Pharmaceutical Sci 2014;3:572-89.

45. Attama AA, Momoh MA, Builders PF. Lipid Nanoparticulate Drug Delivery Systems: A

Revolution in Dosage Form Design and Development. In: Sezer AD, editor. Recent

Advances in Novel Drug Carrier Systems. Rijeka: InTech; 2012, p. 1-34. 10.5772/50486

T
IP
46. Shrestha H, Bala R, Arora S. Lipid-Based Drug Delivery Systems. J Pharm

CR
2014;2014:10.

US
47. Chakraborty S, Shukla D, Mishra B, Singh S. Lipid-an emerging platform for oral

delivery of drugs with poor bioavailability. Eur J Pharm Biopharm 2009;73(1):1-15.


AN
48. Cerpnjak K, Zvonar A, Gasperlin M, Vrecer F. Lipid-based systems as a promising
M

approach for enhancing the bioavailability of poorly water-soluble drugs. Acta Pharm

2013;63(4):427-45.
ED

49. Pouton CW, Porter CJ. Formulation of lipid-based delivery systems for oral
PT

administration: materials, methods and strategies. Adv Drug Deliv Rev 2008;60(6):625-
CE

37.
AC

50. Mullertz A, Ogbonna A, Ren S, Rades T. New perspectives on lipid and surfactant based

drug delivery systems for oral delivery of poorly soluble drugs. J Pharm Pharmacol

2010;62(11):1622-36.

51. S Saroj, DA Baby, M Sabitha. Current trends in lipid based delivery systems and its

applications in drug delivery. Asian J Pharm Clin Res 2012;5:4-9.

48
ACCEPTED MANUSCRIPT

52. Patel HK, Barot BS, Parejiya PB, Shelat PK, Shukla A. Topical delivery of clobetasol

propionate loaded microemulsion based gel for effective treatment of vitiligo: ex vivo

permeation and skin irritation studies. Colloids Sur B Biointerface 2013;102:86-94.

53. Thakkar H, Nangesh J, Parmar M, Patel D. Formulation and characterization of lipid-

based drug delivery system of raloxifene-microemulsion and self-microemulsifying drug

T
IP
delivery system. J Pharm Bioallied Sci 2011;3(3):442-8.

CR
54. Lai F, Pireddu R, Corrias F, Fadda AM, Valenti D, Pini E, et al. Nanosuspension

improves tretinoin photostability and delivery to the skin. Int J Pharm 2013;458(1):104-

US
19.
AN
55. Mahajan HS, Mahajan MS, Nerkar PP, Agrawal A. Nanoemulsion-based intranasal drug

delivery system of saquinavir mesylate for brain targeting. Drug Deliv 2014;21(2):148-
M

54.
ED

56. Modi JD, Patel JK. Nanoemulsion-based gel formulation of aceclofenac for topical
PT

delivery. Int J Pharm Pharm Sci Res2011;1(1):6-12.


CE

57. Chhabra G, Chuttani K, Mishra AK, Pathak K. Design and development of nanoemulsion

drug delivery system of amlodipine besilate for improvement of oral bioavailability.


AC

Drug Dev Ind Pharm 2011;37(8):907-16.

58. Wadhwa J, Nair A, Kumria R. Emulsion forming drug delivery system for lipophilic

drugs. Acta Pol Pharm 2012;69(2):179-91.

49
ACCEPTED MANUSCRIPT

59. Chen ZQ, Liu Y, Zhao JH, Wang L, Feng NP. Improved oral bioavailability of poorly

water-soluble indirubin by a supersaturatable self-microemulsifying drug delivery

system. Int J Nanomed 2012;7:1115-25.

60. Milovic M, Djuris J, Djekic L, Vasiljevic D, Ibric S. Characterization and evaluation of

solid self-microemulsifying drug delivery systems with porous carriers as systems for

T
IP
improved carbamazepine release. Int J Pharm 2012;436(1-2):58-65.

CR
61. Wu X, Xu J, Huang X, Wen C. Self-microemulsifying drug delivery system improves

curcumin dissolution and bioavailability. Drug Dev Ind Pharm 2011;37(1):15-23.

62.
US
Balakumar K, Raghavan CV, selvan NT, prasad RH, Abdu S. Self nanoemulsifying drug
AN
delivery system (SNEDDS) of rosuvastatin calcium: design, formulation, bioavailability

and pharmacokinetic evaluation. Colloid Sur B Biointerface 2013;112:337-43.


M

63. Seo YG, Kim DH, Ramasamy T, Kim JH, Marasini N, Oh Y-K, et al. Development of
ED

docetaxel-loaded solid self-nanoemulsifying drug delivery system (SNEDDS) for


PT

enhanced chemotherapeutic effect. Int J Pharm 2013;452(1):412-20.


CE

64. Gupta S, Chavhan S, Sawant KK. Self-nanoemulsifying drug delivery system for

adefovir dipivoxil: Design, characterization, in vitro and ex vivo evaluation. Colloid Sur
AC

A: Physicochem Eng Asp 2011;392(1):145-55.

65. Tikekar RV PY, Nitin N. Fate of curcumin encapsulated in silica nanoparticle stabilized

Pickering emulsion during storage and simulated digestion. Food Res Int

2013;51(1):370-7.https://doi.org/10.1016/j.foodres.2012.12.027

50
ACCEPTED MANUSCRIPT

66. Shah BR, Li Y, Jin W, An Y, He L, Li Z, et al. Preparation and optimization of Pickering

emulsion stabilized by chitosan-tripolyphosphate nanoparticles for curcumin

encapsulation. Food Hydrocolloids 2016;52:369-77.

67. Chime SA, Onyishi, I.V. Lipid based drug delivery systems (LDDS): recent advances and

applications of lipids in drug delivery. African J Pharm Pharmacol 2013;7:3034-59.

T
IP
68. Shamant BS, Moin A, Gowda DV, R Rashmi, Hiremath R. Lipid based drug delivery

CR
systems in arthritis and allied conditions. World J Pharm Sci 2016;4(4):61-68.

US
69. Park JH, Cho HJ, Yoon HY, Yoon IS, Ko SH, Shim JS, et al. Hyaluronic acid derivative-

coated nanohybrid liposomes for cancer imaging and drug delivery. J Control Rel
AN
2014;174:98-108.
M

70. Ninomiya K, Kawabata S, Tashita H, Shimizu N. Ultrasound-mediated drug delivery

using liposomes modified with a thermosensitive polymer. Ultrason Sonochem


ED

2014;21(1):310-16.https://doi.org/10.1016/j.ultsonch.2013.07.014
PT

71. Yan L, Crayton SH, Thawani JP, Amirshaghaghi A, Tsourkas A, Cheng Z. A pH-
CE

Responsive Drug-Delivery Platform Based on Glycol Chitosan-Coated Liposomes. Small

2015;11(37):4870-4.https://doi.org/10.1002/smll.201501412
AC

72. Xiao Y, Liu Y, Yang S, Zhang B, Wang T, Jiang D, et al. Sorafenib and gadolinium co-

loaded liposomes for drug delivery and MRI-guided HCC treatment. Colloids Surf B

Biointerface 2016;141:83-92.

51
ACCEPTED MANUSCRIPT

73. Meng J, Guo F, Xu H, Liang W, Wang C, Yang XD. Combination Therapy using Co-

encapsulated Resveratrol and Paclitaxel in Liposomes for Drug Resistance Reversal in

Breast Cancer Cells in vivo. Sci Rep 2016;6:22390.doi:10.1038/srep22390

74. Deshpande P, Jhaveri A, Pattni B, Biswas S, Torchilin V. Transferrin and octaarginine

modified dual-functional liposomes with improved cancer cell targeting and enhanced

T
IP
intracellular delivery for the treatment of ovarian cancer. Drug Deliv 2018;25(1):517-32.

CR
75. He R-X, Ye X, Li R, Chen W, Ge T, Huang T-Q, et al. PEGylated niosomes-mediated

drug delivery systems for Paeonol: preparation, pharmacokinetics studies and synergistic

US
anti-tumor effects with 5-FU. J Liposome Res 2017;27(2):161-70.
AN
76. Kiran Jyoti RSP, Jitender Madan, Upendra Kumar Jain. Inhalable cationic niosomes of

curcumin enhanced drug delivery and apoptosis in lung cancer cells. Ind J Pharm Edu
M

Res 2016;50(2):S21-S31.
ED

77. Mehta SK, Jindal N. Tyloxapol niosomes as prospective drug delivery module for
PT

antiretroviral drug nevirapine. AAPS PharmSciTech 2015;16(1):67-

75.https://doi.org/10.1208/s12249-014-0183-y
CE

78. Auda SH, Fathalla D, Fetih G, El-Badry M, Shakeel F. Niosomes as transdermal drug
AC

delivery system for celecoxib: in vitro and in vivo studies. Polymer Bulletin

2016;73(5):1229-45.

79. Qumbar M, Ameeduzzafar, Imam SS, Ali J, Ahmad J, Ali A. Formulation and

optimization of lacidipine loaded niosomal gel for transdermal delivery: In-vitro

characterization and in-vivo activity. Biomed Pharmacother 2017;93:255-66.

52
ACCEPTED MANUSCRIPT

80. Chiranjeevi G, Muthukumaran, M., Krishnamoorthy, B. A review on potency of vesicular

systems in targeting drug delivery. Res J Pharm Biol Chem Sci 2013;4:156-70.

81. Semalty A, Semalty M, Singh D, Rawat MS. Development and physicochemical

evaluation of pharmacosomes of diclofenac. Acta Pharm 2009;59(3):335-44.

T
82. Mali Kamalesh DBD, Baviskar Kiran, Wagh Kalpesh. Formulation and evaluation of

IP
pharmacosomes of ketoprofen. Indo American J Pharm Res 2014;4(3):1363-8.

CR
83. Pal T, Mishra J, Podder A. Design, fabrication and evaluation of rosuvastatin

US
pharmacosome-a novel sustained release drug delivery system. Eur J Pharm Med Res

2016;3(4):332-50.
AN
84. Yu F, Li Y, Chen Q, He Y, Wang H, Yang L, et al. Monodisperse microparticles loaded
M

with the self-assembled berberine-phospholipid complex-based phytosomes for

improving oral bioavailability and enhancing hypoglycemic efficiency. Eur J Pharm


ED

Biopharm 2016;103:136-48.
PT

85. Abdelkader H, Longman MR, Alany RG, Pierscionek B. Phytosome-hyaluronic acid


CE

systems for ocular delivery of L-carnosine. Int J Nanomed 2016;11:2815-27.


AC

86. Mirzaei H, Shakeri A, Rashidi B, Jalili A, Banikazemi Z, Sahebkar A. Phytosomal

curcumin: A review of pharmacokinetic, experimental and clinical studies. Biomed

Pharmacother2017;85:102-12.

87. Cevc G, Gebauer D, Stieber J, Schatzlein A, Blume G. Ultraflexible vesicles,

Transfersomes, have an extremely low pore penetration resistance and transport

53
ACCEPTED MANUSCRIPT

therapeutic amounts of insulin across the intact mammalian skin. Biochim Biophys Acta

1998;1368(2):201-15.

88. Dubey V, Mishra D, Nahar M, Jain NK. Vesicles as tools for the modulation of skin

permeability. Expert Opin Drug Deliv 2007;4(6):579-

93.https://doi.org/10.1517/17425247.4.6.579

T
IP
89. Mishra D, Garg M, Dubey V, Jain S, Jain NK. Elastic liposomes mediated transdermal

CR
delivery of an anti-hypertensive agent: propranolol hydrochloride. J Pharm

Sci2007;96(1):145-55.https://doi.org/10.1002/jps.20737

90.
US
Mishra DK, Dhote V, Mishra PK. Transdermal immunization: biological framework and
AN
translational perspectives. Exp Opin Drug Deliv 2013;10(2):183-

200.https://doi.org/10.1517/17425247.2013.746660
M

91. Abdellatif AAH, Tawfeek HM. Transfersomal Nanoparticles for Enhanced Transdermal
ED

Delivery of Clindamycin. AAPS PharmSciTech2016;17(5):1067-74.


PT

92. Salama HA, Mahmoud AA, Kamel AO, Abdel Hady M, Awad GAS. Brain delivery of
CE

olanzapine by intranasal administration of transfersomal vesicles. J Liposome Res

2012;22(4):336-45.
AC

93. Sarwa KK, Mazumder B, Rudrapal M, Verma VK. Potential of capsaicin-loaded

transfersomes in arthritic rats. Drug Deliv2015;22(5):638-46.

94. Ahmed TA. Preparation of transfersomes encapsulating sildenafil aimed for transdermal

drug delivery: Plackett–Burman design and characterization. J Liposome

Res2015;25(1):1-10.

54
ACCEPTED MANUSCRIPT

95. Dubey V, Mishra D, Jain NK. Melatonin loaded ethanolic liposomes: physicochemical

characterization and enhanced transdermal delivery. Eur J Pharm

Biopharm2007;67(2):398-405.

96. Touitou E, Dayan N, Levi‐Schaffer F, Piliponsky A. Novel lipid vesicular system for

enhanced delivery. J Lip Res 1998;8(113).

T
IP
97. Mishra D, Mishra P, Dabadghao S, Dubey V, Nahar M, Jain N. Comparative evaluation

CR
of hepatitis B surface antigen-loaded elastic liposomes and ethosomes for human

dendritic cell uptake and immune response. Vaccine 2009;6(1):110-18.

US
https://doi.org/10.1016/j.nano.2009.04.003
AN
98. Jain S, Tiwary AK, Sapra B, Jain NK. Formulation and evaluation of ethosomes for

transdermal delivery of lamivudine. AAPS PharmSciTech2007;8(4):E111.


M

99. Bodade SS, Shaikh KS, Kamble MS, Chaudhari PD. A study on ethosomes as mode for
ED

transdermal delivery of an antidiabetic drug. Drug Delivery 2013;20(1):40-6.


PT

100. Asadujjaman MD, Mishuk AU. Novel approaches in lipid based drug delivery systems. J
CE

Drug Del Ther 2013;3:124-30.


AC

101. Alavi SE, Mansouri H, Esfahani MKM, Movahedi F, Akbarzadeh A, Chiani M.

Archaeosome: as new drug carrier for delivery of paclitaxel to breast cancer. Ind J

Clinical Biochem2014;29(2):150-3.

102. Moghimipour E, Kargar M, Ramezani Z, Handali S. The potent in vitro skin permeation

of archaeosome made from lipids extracted of sulfolobus acidocaldarius. Archaea

2013;2013:7.http://dx.doi.org/10.1155/2013/782012

55
ACCEPTED MANUSCRIPT

103. Daeyeon L, David AW.Double emulsion-templated nanoparticle colloidosomes with

selective permeability. Adv Mater 2008;20:3498-

503.https://doi.org/10.1002/adma.200800918

104. Shah RK, Kim J-W, Weitz DA. Monodisperse stimuli-responsive colloidosomes by self-

assembly of microgels in droplets. Langmuir 2010;26(3):1561-5.

T
IP
105. Nan F, Wu J, Qi F, Fan Q, Ma G, Ngai T. Preparation of uniform-sized colloidosomes

CR
based on chitosan-coated alginate particles and its application for oral insulin delivery. J

Materials Chem B 2014;2(42):7403-9.

106.
US
Bollhorst T, Shahabi S, Worz K, Petters C, Dringen R, Maas M, et al. Bifunctional
AN
Submicron Colloidosomes Coassembled from Fluorescent and Superparamagnetic

Nanoparticles. Angewandte Chemie2015;127(1):120-


M

25.https://doi.org/10.1002/ange.201408515
ED

107. Zhou S, Fan J, Datta SS, Guo M, Guo X, Weitz DA. Thermally Switched Release from
PT

Nanoparticle Colloidosomes. Adv Functional Materials 2013;23(47):5925-9.


CE

108. Jain S, Jain V, Mahajan SC. Lipid Based Vesicular Drug Delivery Systems. Adv

Pharm2014;2014:12.
AC

109. Patil S, Pancholi SS, Agrawal S, Agrawal GP. Surface-modified mesoporous ceramics as

delivery vehicle for haemoglobin. Drug Delivery 2004;11(3):193-9.

110. Pandey RS, Sahu S, Sudheesh MS, Madan J, Kumar M, Dixit VK. Carbohydrate

modified ultrafine ceramic nanoparticles for allergen immunotherapy. Int

Immunopharmacol 2011;11(8):925-31.

56
ACCEPTED MANUSCRIPT

111. Karami Z, Hamidi M. Cubosomes: remarkable drug delivery potential. Drug Discov

Today 2016;21(5):789-801.

112. Esposito E, Cortesi R, Drechsler M, Paccamiccio L, Mariani P, Contado C, et al.

Cubosome dispersions as delivery systems for percutaneous administration of

indomethacin. Pharm Res 2005;22(12):2163-73.

T
IP
113. Han S, Shen J-q, Gan Y, Geng H-m, Zhang X-x, Zhu C-l, et al. Novel vehicle based on

CR
cubosomes for ophthalmic delivery of flurbiprofen with low irritancy and high

bioavailability. Acta Pharmacologica Sinica 2010;31:990.

114.
US
Gan L, Han S, Shen J, Zhu J, Zhu C, Zhang X, et al. Self-assembled liquid crystalline
AN
nanoparticles as a novel ophthalmic delivery system for dexamethasone: Improving

preocular retention and ocular bioavailability. Int J Pharm 2010;396(1):179-87.


M

115. Yang Z, Chen M, Yang M, Chen J, Fang W, Xu P. Evaluating the potential of cubosomal
ED

nanoparticles for oral delivery of amphotericin B in treating fungal infection. Int J


PT

Nanomed 2014;9:327-36.
CE

116. Tiwari B, Dangi Y, Jain A, Jain A. Sphingosomes: Potential Anti-Cancer Vectors for the

Delivery of Doxorubicin. Int J Pharmacol Pharm Sci 2016;10(12):1.10.1999/1307-


AC

6892/56520

117. Mittal R, Sharma A, Arora S. Ufasomes Mediated Cutaneous Delivery of

Dexamethasone: Formulation and Evaluation of Anti-Inflammatory Activity by

Carrageenin-Induced Rat Paw Edema Model. J Pharm 2013;2013:12.

57
ACCEPTED MANUSCRIPT

118. Salama AH, Aburahma MH. Ufasomes nano-vesicles-based lyophilized platforms for

intranasal delivery of cinnarizine: preparation, optimization, ex-vivo histopathological

safety assessment and mucosal confocal imaging. Pharm Dev Tech 2016;21(6):706-15.

119. Vyas SP, Subhedar R, Jain S. Development and characterization of emulsomes for

sustained and targeted delivery of an antiviral agent to liver. J Pharm Pharmacol

T
IP
2006;58(3):321-6.

CR
120. Paliwal R, Paliwal SR, Mishra N, Mehta A, Vyas SP. Engineered chylomicron

mimicking carrier emulsome for lymph targeted oral delivery of methotrexate. Int J

US
Pharm 2009;380(1):181-8.
AN
121. El-Zaafarany GM, Soliman ME, Mansour S, Awad GAS. Identifying lipidic emulsomes

for improved oxcarbazepine brain targeting: In vitro and rat in vivo studies. Int J Pharm
M

2016;503(1):127-40.
ED

122. Alavi M, Karimi N, Safaei M. Application of Various Types of Liposomes in Drug


PT

Delivery Systems. Adv Pharm Bull 2017;7(1):3-9.


CE

123. Abd-Elal RM, Shamma RN, Rashed HM, Bendas ER. Trans-nasal zolmitriptan

novasomes: in-vitro preparation, optimization and in-vivo evaluation of brain targeting


AC

efficiency. Drug Deliv 2016;23(9):3374-86.

124. Chime SA, Attama AA, Builders PF, Onunkwo GC. Sustained-release diclofenac

potassium-loaded solid lipid microparticle based on solidified reverse micellar solution:

in vitro and in vivo evaluation. J Microencapsul 2013;30(4):335-45.

58
ACCEPTED MANUSCRIPT

125. Umeyor EC, Kenechukwu FC, Ogbonna JD, Chime SA, Attama A. Preparation of novel

solid lipid microparticles loaded with gentamicin and its evaluation in vitro and in vivo. J

Microencapsul 2012;29(3):296-307.

126. Barakat NS, Yassin AEB. In Vitro Characterization of Carbamazepine-Loaded Precifac

Lipospheres. Drug Deliv 2006;13(2):95-104.

T
IP
127. Nasr M, Mansour S, Mortada ND, El Shamy AA. Lipospheres as Carriers for Topical

CR
Delivery of Aceclofenac: Preparation, Characterization and In Vivo Evaluation. AAPS

PharmSciTech 2008;9(1):154-62.

128.
US
Attama AA, Okafor CE, Builders PF, Okorie O. Formulation and in vitro evaluation of a
AN
PEGylated microscopic lipospheres delivery system for ceftriaxone sodium. Drug Deliv

2009;16(8):448-57.
M

129. Fang J-Y, Hung C-F, Liao M-H, Chien C-C. A study of the formulation design of
ED

acoustically active lipospheres as carriers for drug delivery. Eur J Pharm


PT

Biopharm2007;67(1):67-75.
CE

130. El-Kamel AH, Al-Fagih IM, Alsarra IA. Testosterone solid lipid microparticles for

transdermal drug delivery. Formulation and physicochemical characterization. J


AC

Microencapsul 2007;24(5):457-75.

131. Jaspart S, Bertholet P, Piel G, Dogné J-M, Delattre L, Evrard B. Solid lipid

microparticles as a sustained release system for pulmonary drug delivery. Eur J Pharm

Biopharm 2007;65(1):47-56.

59
ACCEPTED MANUSCRIPT

132. Yadav VR, Suresh S, Devi K, Yadav S. Novel formulation of solid lipid microparticles of

curcumin for anti-angiogenic and anti-inflammatory activity for optimization of therapy

of inflammatory bowel disease. J Pharm Pharmacol2009;61(3):311-21.

133. Bhargava A, Mishra D, Khan S, Varshney SK, Banerjee S, Mishra PK. Assessment of

tumor antigen-loaded solid lipid nanoparticles as an efficient delivery system for

T
IP
dendritic cell engineering. Nanomed (Lond). 2013;8(7):1067-

CR
84.https://doi.org/10.2217/nnm.12.164

134. Mishra H, Mishra D, Mishra PK, Nahar M, Dubey V, Jain NK. Evaluation of solid lipid

US
nanoparticles as carriers for delivery of hepatitis B surface antigen for vaccination using
AN
subcutaneous route. J Pharm Pharm Sci 2010;13(4):495-

509.http://dx.doi.org/10.18433/J3XK53
M

135. Sarmento B, Martins S, Ferreira D, Souto EB. Oral insulin delivery by means of solid
ED

lipid nanoparticles. Int J Nanomed. 2007;2(4):743-9.


PT

136. Yassin AE, Anwer MK, Mowafy HA, El-Bagory IM, Bayomi MA, Alsarra IA.

Optimization of 5-flurouracil solid-lipid nanoparticles: a preliminary study to treat colon


CE

cancer. Int J Med Sci 2010;7(6):398-408.


AC

137. Jain A, Agarwal A, Majumder S, Lariya N, Khaya A, Agrawal H, et al. Mannosylated

solid lipid nanoparticles as vectors for site-specific delivery of an anti-cancer drug. J

Control Rel 2010;148(3):359-67.

60
ACCEPTED MANUSCRIPT

138. Dhawan S, Kapil R, Singh B. Formulation development and systematic optimization of

solid lipid nanoparticles of quercetin for improved brain delivery. J Pharm Pharmacol.

2011;63(3):342-51.

139. Martins S, Tho I, Reimold I, Fricker G, Souto E, Ferreira D, et al. Brain delivery of

camptothecin by means of solid lipid nanoparticles: Formulation design, in vitro and in

T
IP
vivo studies. Int J Pharm 2012;439(1):49-62.

CR
140. Bhalekar MR, Pokharkar V, Madgulkar A, Patil N, Patil N. Preparation and evaluation of

miconazole nitrate-loaded solid lipid nanoparticles for topical delivery. AAPS

US
PharmSciTech 2009;10(1):289-96.
AN
141. Ravani L, Esposito E, Bories C, Moal VL, Loiseau PM, Djabourov M, et al.

Clotrimazole-loaded nanostructured lipid carrier hydrogels: thermal analysis and in vitro


M

studies. Int J Pharm 2013;454(2):695-702.


ED

142. Shen J, Deng Y, Jin X, Ping Q, Su Z, Li L. Thiolated nanostructured lipid carriers as a


PT

potential ocular drug delivery system for cyclosporine A: Improving in vivo ocular

distribution. Int J Pharm 2010;402(1):248-53.


CE

143. Luo Q, Zhao J, Zhang X, Pan W. Nanostructured lipid carrier (NLC) coated with
AC

Chitosan Oligosaccharides and its potential use in ocular drug delivery system. Int J

Pharm 2011;403(1):185-91.

144. Khurana S, Jain NK, Bedi PMS. Development and characterization of a novel controlled

release drug delivery system based on nanostructured lipid carriers gel for meloxicam.

Life Sci 2013;93(21):763-72.doi:10.1016/j.lfs.2013.09.027

61
ACCEPTED MANUSCRIPT

145. Fathi HA, Allam A, Elsabahy M, Fetih G, El-Badry M. Nanostructured lipid carriers for

improved oral delivery and prolonged antihyperlipidemic effect of simvastatin. Colloids

Surfaces B Biointerface2018;162:236-45.https://doi.org/10.1016/j.colsurfb.2017.11.064

146. Jawahar N, Hingarh PK, Arun R, Selvaraj J, Anbarasan A, S S, et al. Enhanced oral

bioavailability of an antipsychotic drug through nanostructured lipid carriers. Int JBiol

T
IP
Macro 2018;110:269-75. https://doi.org/10.1016/j.ijbiomac.2018.01.121

CR
147. Mohammed M, Adel, M., Magda, S. Pharmaceutical particulate carriers: lipid-based

carriers. Nat J Physio Pharm Pharmacol 2012;2:10-22.

148.
US
Paliwal R, Rai S, Vyas S. Lipid Drug Conjugate (LDC) Nanoparticles as
AN
Autolymphotrophs for Oral Delivery of Methotrexate. J Biomed Nanotech 2011;7(1):

130-31.10.1166/jbn.2011.1235
M

149. Sharma P, Dube B, Sawant K. Synthesis of cytarabine lipid drug conjugate for treatment
ED

of meningeal leukemia: development, characterization and in vitro cell line studies. J


PT

Biomed Nanotech 2012; 8(6):928-37.https://doi.org/10.1166/jbn.2012.1464


CE

150. Abu-Fayyad A, Kamal MM, Carroll JL, Dragoi A-M, Cody R, Cardelli J, et al.

Development and in-vitro characterization of nanoemulsions loaded with paclitaxel/γ-


AC

tocotrienol lipid conjugates. Int J Pharm 2018;536(1):146-57.

151. Cattaneo AG, Gornati R, Sabbioni E, Chiriva-Internati M, Cobos E, Jenkins MR, et al.

Nanotechnology and human health: risks and benefits. J Appl Toxicol 2010;30(8):730-44.

152. Mahalingam D, Nemunaitis JJ, Malik L, Sarantopoulos J, Weitman S, Sankhala K, et al.

Phase I study of intravenously administered ATI-1123, a liposomal docetaxel formulation

62
ACCEPTED MANUSCRIPT

in patients with advanced solid tumors. Cancer Chemother Pharmacol 2014;74(6):1241-

50.

153. Lu C, Perez-Soler R, Piperdi B, Walsh GL, Swisher SG, Smythe WR, et al. Phase II

study of a liposome-entrapped cisplatin analog (L-NDDP) administered intrapleurally

and pathologic response rates in patients with malignant pleural mesothelioma. J Clin

T
IP
Oncol 2005;23(15):3495-501.

CR
154. Dragovich T, Mendelson D, Kurtin S, Richardson K, Von Hoff D, Hoos A. A Phase 2

trial of the liposomal DACH platinum L-NDDP in patients with therapy-refractory

US
advanced colorectal cancer. Cancer Chemother Pharmacol 2006;58(6):759-64.
AN
155. Hamaguchi T, Matsumura Y, Nakanishi Y, Muro K, Yamada Y, Shimada Y, et al.

Antitumor effect of MCC-465, pegylated liposomal doxorubicin tagged with newly


M

developed monoclonal antibody GAH, in colorectal cancer xenografts. Cancer Sci


ED

2004;95(7):608-13.
PT

156. Matsumura Y, Gotoh M, Muro K, Yamada Y, Shirao K, Shimada Y, et al. Phase I and

pharmacokinetic study of MCC-465, a doxorubicin (DXR) encapsulated in PEG


CE

immunoliposome, in patients with metastatic stomach cancer. Ann Oncol


AC

2004;15(3):517-25.

157. Xu L, Tang WH, Huang CC, Alexander W, Xiang LM, Pirollo KF, et al. Systemic p53

gene therapy of cancer with immunolipoplexes targeted by anti-transferrin receptor scFv.

Mol Med 2001;7(10):723-34.PMC1949994

63
ACCEPTED MANUSCRIPT

158. Camp ER, Wang C, Little EC, Watson PM, Pirollo KF, Rait A, et al. Transferrin receptor

targeting nanomedicine delivering wild-type p53 gene sensitizes pancreatic cancer to

gemcitabine therapy. Cancer Gene Ther 2013;20(4):222-8.

159. Kim SS, Rait A, Kim E, Pirollo KF, Chang EH. A tumor-targeting p53 nanodelivery

system limits chemoresistance to temozolomide prolonging survival in a mouse model of

T
IP
glioblastoma multiforme. Nanomed Nanotech Biol Med 2015;11(2):301-

CR
11. https://doi.org/10.1016/j.nano.2014.09.005

160. Tam YY, Chen S, Cullis PR. Advances in lipid nanoparticles for siRNA delivery.

US
Pharmaceutics 2013;5(3):498-507.10.3390/pharmaceutics5030498
AN
161. Leung AK, Tam YY, Cullis PR. Lipid nanoparticles for short interfering RNA delivery.

Adv Gene 2014;88:71-110.https://doi.org/10.1016/B978-0-12-800148-6.00004-3


M

162. Wetzler M, Thomas DA, Wang ES, Shepard R, Ford LA, Heffner TL, et al. Phase I/II
ED

trial of nanomolecular liposomal annamycin in adult patients with relapsed/refractory


PT

acute lymphoblastic leukemia. Clin Lymphoma Myeloma Leuk 2013;13(4):430-4.


CE

163. Yang SH, Lin CC, Lin ZZ, Tseng YL, Hong RL. A phase I and pharmacokinetic study of

liposomal vinorelbine in patients with advanced solid tumor. Invest New Drugs
AC

2012;30(1):282-9.

164. Lin YY, Kao HW, Li JJ, Hwang JJ, Tseng YL, Lin WJ, et al. Tumor burden talks in

cancer treatment with PEGylated liposomal drugs. PLoS One 2013;8(5):e63078.

165. Mamot C, Ritschard R, Wicki A, Stehle G, Dieterle T, Bubendorf L, et al. Tolerability,

safety, pharmacokinetics, and efficacy of doxorubicin-loaded anti-EGFR

64
ACCEPTED MANUSCRIPT

immunoliposomes in advanced solid tumours: a phase 1 dose-escalation study. Lancet

Oncol 2012;13(12):1234-41.

166. Suzuki R, Takizawa T, Kuwata Y, Mutoh M, Ishiguro N, Utoguchi N, et al. Effective

anti-tumor activity of oxaliplatin encapsulated in transferrin-PEG-liposome. Int J Pharm

2008;346(1-2):143-50.

T
IP
167. van der Meel R, Vehmeijer LJ, Kok RJ, Storm G, van Gaal EV. Ligand-targeted

CR
particulate nanomedicines undergoing clinical evaluation: current status. Adv Drug Deliv

Rev 2013;65(10):1284-98.

168.
US
Fasol U, Frost A, Buchert M, Arends J, Fiedler U, Scharr D, et al. Vascular and
AN
pharmacokinetic effects of EndoTAG-1 in patients with advanced cancer and liver

metastasis. Ann Oncol 2012;23(4):1030-6.


M

169. Lohr JM, Haas SL, Bechstein WO, Bodoky G, Cwiertka K, Fischbach W, et al. Cationic
ED

liposomal paclitaxel plus gemcitabine or gemcitabine alone in patients with advanced


PT

pancreatic cancer: a randomized controlled phase II trial. Ann Oncol 2012;23(5):1214-22.


CE

170. Awada A, Bondarenko IN, Bonneterre J, Nowara E, Ferrero JM, Bakshi AV, et al. A

randomized controlled phase II trial of a novel composition of paclitaxel embedded into


AC

neutral and cationic lipids targeting tumor endothelial cells in advanced triple-negative

breast cancer (TNBC). Ann Oncol 2014;25(4):824-31.

171. Duffaud F, Borner M, Chollet P, Vermorken JB, Bloch J, Degardin M, et al. Phase II

study of OSI-211 (liposomal lurtotecan) in patients with metastatic or loco-regional

65
ACCEPTED MANUSCRIPT

recurrent squamous cell carcinoma of the head and neck. An EORTC New Drug

Development Group study. Eur J Cancer 2004;40(18):2748-52.

172. Batist G, Gelmon KA, Chi KN, Miller WH, Jr., Chia SK, Mayer LD, et al. Safety,

pharmacokinetics, and efficacy of CPX-1 liposome injection in patients with advanced

solid tumors. Clin Cancer Res 2009;15(2):692-700.

T
IP
173. Slingerland M, Guchelaar HJ, Rosing H, Scheulen ME, van Warmerdam LJ, Beijnen JH,

CR
et al. Bioequivalence of Liposome-Entrapped Paclitaxel Easy-To-Use (LEP-ETU)

formulation and paclitaxel in polyethoxylated castor oil: a randomized, two-period

US
crossover study in patients with advanced cancer. Clin Ther 2013;35(12):1946-54.
AN
174. Serisier DJ, Bilton D, De Soyza A, Thompson PJ, Kolbe J, Greville HW, et al. Inhaled,

dual release liposomal ciprofloxacin in non-cystic fibrosis bronchiectasis (ORBIT-2): a


M

randomised, double-blind, placebo-controlled trial. Thorax 2013;68(9):812-7.


ED

175. Drummond DC, Noble CO, Guo Z, Hong K, Park JW, Kirpotin DB. Development of a
PT

highly active nanoliposomal irinotecan using a novel intraliposomal stabilization strategy.

Cancer Res 2006;66(6):3271-7.


CE

176. Kang MH, Wang J, Makena MR, Lee JS, Paz N, Hall CP, et al. Activity of MM-398,
AC

nanoliposomal irinotecan (nal-IRI), in Ewing's family tumor xenografts is associated with

high exposure of tumor to drug and high SLFN11 expression. Clin Cancer Res

2015;21(5):1139-50.

177. Roy AC, Park SR, Cunningham D, Kang YK, Chao Y, Chen LT, et al. A randomized

phase II study of PEP02 (MM-398), irinotecan or docetaxel as a second-line therapy in

66
ACCEPTED MANUSCRIPT

patients with locally advanced or metastatic gastric or gastro-oesophageal junction

adenocarcinoma. Ann Oncol 2013;24(6):1567-73.https://doi.org/10.1093/annonc/mdt002

178. Feldman EJ, Lancet JE, Kolitz JE, Ritchie EK, Roboz GJ, List AF, et al. First-in-man

study of CPX-351: a liposomal carrier containing cytarabine and daunorubicin in a fixed

5:1 molar ratio for the treatment of relapsed and refractory acute myeloid leukemia. J

T
IP
Clin Oncol 2011;29(8):979-85. 10.1200/JCO.2010.30.5961

CR
179. Cortes JE, Goldberg SL, Feldman EJ, Rizzeri DA, Hogge DE, Larson M, et al. Phase II,

multicenter, randomized trial of CPX-351 (cytarabine:daunorubicin) liposome injection

US
versus intensive salvage therapy in adults with first relapse AML. Cancer
AN
2015;121(2):234-42.https://doi.org/10.1002/cncr.28974

180. May JP, Li SD. Hyperthermia-induced drug targeting. Expert Opin Drug
M

Deliv2013;10(4):511-27.https://doi.org/10.1517/17425247.2013.758631
ED

181. Boulikas T. Clinical overview on Lipoplatin: a successful liposomal formulation of


PT

cisplatin. Expert Opin Invest Drugs 2009;18(8):1197-

218.https://doi.org/10.1517/13543780903114168
CE

182. Stathopoulos GP, Antoniou D, Dimitroulis J, Michalopoulou P, Bastas A, Marosis K, et


AC

al. Liposomal cisplatin combined with paclitaxel versus cisplatin and paclitaxel in non-

small-cell lung cancer: a randomized phase III multicenter trial. Ann Oncol

2010;21(11):2227-32.https://doi.org/10.1093/annonc/mdq234

67
ACCEPTED MANUSCRIPT

183. Clancy JP, Dupont L, Konstan MW, Billings J, Fustik S, Goss CH, et al. Phase II studies

of nebulised Arikace in CF patients with Pseudomonas aeruginosa infection. Thorax

2013;68(9):818-25.http://dx.doi.org/10.1136/thoraxjnl-2012-202230

184. Lu B, Xiong S-B, Yang H, Yin X-D, Zhao R-B. Mitoxantrone-loaded BSA nanospheres

and chitosan nanospheres for local injection against breast cancer and its lymph node

T
IP
metastases. II: Tissue distribution and pharmacodynamics. Int J Pharm2006;307(2):175-

CR
81.https://doi.org/10.1016/j.ijpharm.2005.09.037

185. Low PS, Henne WA, Doorneweerd DD. Discovery and development of folic-acid-based

US
receptor targeting for imaging and therapy of cancer and inflammatory diseases. Acc
AN
Chem Res 2008;41(1):120-29.10.1021/ar7000815

186. Liu Z, Chen K, Davis C, Sherlock S, Cao Q, Chen X, et al. Drug delivery with carbon
M

nanotubes for in vivo cancer treatment. Cancer Res 2008;68(16):6652-60.10.1158/0008-


ED

5472.CAN-08-1468
PT

187. Chien AJ, Illi JA, Ko AH, Korn WM, Fong L, Chen LM, et al. A phase 1 study of a 2 day

lapatinib chemosensitization pulse preceding nanoparticle albumin-bound paclitaxel for


CE

advanced solid malignancies. Clinical Cancer Research2009;15(17):5569-


AC

75.10.1158/1078-0432.CCR-09-0522

188. Zhu L, Huo Z, Wang L, Tong X, Xiao Y, Ni K. Targeted delivery of methotrexate to

skeletal muscular tissue by thermosensitive magnetoliposomes. Int J Pharm 2009;370(1-

2):136-43.https://doi.org/10.1016/j.ijpharm.2008.12.003

68
ACCEPTED MANUSCRIPT

189. Conlin AK, Seidman AD, Bach A, Lake D, Dickler M, D'Andrea G, et al. Phase II trial of

weekly nanoparticle albumin-bound paclitaxel with carboplatin and trastuzumab as first-

line therapy for women with HER2-overexpressing metastatic breast cancer. Clinical

Breast Cancer 2010;10(4):281-7.https://doi.org/10.3816/CBC.2010.n.036

190. Kang KW, Chun MK, Kim O, Subedi RK, Ahn SG, Yoon JH, et al. Doxorubicin-loaded

T
IP
solid lipid nanoparticles to overcome multidrug resistance in cancer therapy. Nanomed

CR
Nanotech Biol Med 2010;6(2):210-13.https://doi.org/10.1016/j.nano.2009.12.006

191. Liu F, Park JY, Zhang Y, Conwell C, Liu Y, Bathula SR, et al. Targeted cancer therapy

US
with novel high drug-loading nanocrystals. J Pharmsci 2010;99(8):3542-
AN
51.https://doi.org/10.1002/jps.22112

192. Khdair A, Chen D, Patil Y, Ma L, Dou QP, Shekhar MP, et al. Nanoparticle-mediated
M

combination chemotherapy and photodynamic therapy overcomes tumor drug resistance.


ED

J Control Rel 2010;141(2):137-44.https://doi.org/10.1016/j.jconrel.2009.09.004


PT

193. Yu YH, Kim E, Park DE, Shim G, Lee S, Kim YB, et al. Cationic solid lipid

nanoparticles for co-delivery of paclitaxel and siRNA. Eur J Pharm Biopharm


CE

2012;80(2):268-73.https://doi.org/10.1016/j.ejpb.2011.11.002
AC

194. Patel PA, Patil SC, Kalaria DR, Kalia YN, Patravale VB. Comparative in vitro and in

vivo evaluation of lipid based nanocarriers of Huperzine A. Int J Pharm 2013;446(1-

2):16-23.https://doi.org/10.1016/j.ijpharm.2013.02.014

69
ACCEPTED MANUSCRIPT

195. Lacatusu I, Niculae G, Badea N, Stan R, Popa O, Oprea O, et al. Design of soft lipid

nanocarriers based on bioactive vegetable oils with multiple health benefits. Chem Eng J

2014;246:311-21.https://doi.org/10.1016/j.cej.2014.02.041

196. Aditya NP, Macedo AS,Doktorovova S, Souto EB, Kim S, Chang PS, et al. Development

and evaluation of lipid nanocarriers for quercetin delivery: A comparative study of solid

T
IP
lipid nanoparticles (SLN), nanostructured lipid carriers (NLC), and lipid nanoemulsions

CR
(LNE). FoodSciTech 2014;59:115-21.https://doi.org/10.1016/j.lwt.2014.04.058

197. Gautier J, Allard-Vannier E, Munnier E, Souce M, Chourpa I. Recent advances in

US
theranostic nanocarriers of doxorubicin based on iron oxide and gold nanoparticles. J
AN
Control Rel 2013;169(1-2):48-61.https://doi.org/10.1016/j.jconrel.2013.03.018

198. Lacatusu I, Badea N, Badea G, Oprea O, Mihaila MA, Kaya DA, et al. Lipid nanocarriers
M

based on natural oils with high activity against oxygen free radicals and tumor cell
ED

proliferation. Mater Sci Eng C 2015;56:88-94.https://doi.org/10.1016/j.msec.2015.06.019


PT

199. Li L, Jiang G, Yu W, Liu D, Chen H, Liu Y, et al. A composite hydrogel system containing

glucose-responsive nanocarriers for oral delivery of insulin. Mater Sci Eng C 2016;69:37-
CE

45.https://doi.org/10.1016/j.msec.2016.06.059
AC

200. Du F, Honzke S, Neumann F, Keilitz J, Chen W, Ma N, et al. Development of

biodegradable hyperbranched core-multishell nanocarriers for efficient topical drug

delivery. J Control Rel 2016;242:42-49.https://doi.org/10.1016/j.jconrel.2016.06.048

70
ACCEPTED MANUSCRIPT

Table 1: Lipid based nanocarriers with their salient features and limitations

Lipid based Composition Salient features Benefits Limitations/chall


carrier system enges

Emulsion based

ME Water, oil  Transparenc  Thermodyn  Thermal


and y amic stress can

T
amphiphile stability break the
 Less ME

IP
viscosity  Great
solubilizati  High

CR
 Thermodyna on potential surfactant
mic stability levels are
 Clarity required

US
 Easy  Higher
preparation costing
AN
and scale
up  Elevated
risks of
M

 Increased adverse
solubilizati events
ED

on such as
 Improved irritation
and/or
PT

bioavailabil
ity and poor taste
efficacy  Cosolvent
CE

 Improved s may be
physical costly and
AC

stability have
unwanted
 Reversible toxicities
under
thermal  Affected
stress by the
conditions ionic
strength
of the GIT

71
ACCEPTED MANUSCRIPT

NE Oils Submicron  Lack of  Manufact


(acylglycerol emulsion flocculation uring via
s) and other , high
bioactive sedimentati energy
components, on & methods
Surfactants creaming utilize
(phospholipid costly and
s, proteins,  Reduced complicat
dosing

T
polysaccharid ed
es, or frequency equipment

IP
minerals),
 Sustained ’s like
aqueous

CR
and homogeni
component zer,
controlled
(water), microfuidi
release

US
texture zer, which
modifier,  Manufactur adds to
weighting ed by manufactu
AN
agent, and minimum ring cost.
ripening amount of
retardant surfactant,  Still better
M

hence less understan


toxic to ding of
ED

biological droplets
membranes generation
mechanis
PT

 Nanosize m using
droplets various
makes them methods
CE

resistant is required
towards to
AC

destabilizin efficiently
g place this
phenomeno system in
n market

 Used in  Stability
replacemen is of short
t for term due
liposomes to less
and amount of
vesicles as surfactant

72
ACCEPTED MANUSCRIPT

they are used


more
stable.  lack of
understan
ding of
basic
chemistry
involved
in

T
productio

IP
n of NE
has

CR
created
misconcep
tions in

US
mind of
researcher
AN
s leading
to
avoidance
M

in
working
ED

on NE or
terminatio
n of
PT

ongoing
works
CE

SEDDS Mixture of  GRAS  Improved  Lack of


oil & status oral robust in
surfactants bioavailabil vitro
AC

 Simple ity model


 Excellent  Selective  Oxidation
capsule targeting potential
compatibilit of lipid
y  Drug componen
protection t
 Unlikely to from the
lose solvent hostile  High ratio
capacity on environmen of lipid
and

73
ACCEPTED MANUSCRIPT

dispersion t in gut surfactant


may lead
 Drug  Reduced to
absorption dosing complex
without reactions
digestion and
ultimately
serious in-

T
vivo
issues

IP
 High

CR
concentrat
ion of
surfactant

US
may lead
to serious
AN
toxicity
by
altering
M

the
protein
ED

structure
and
malfuncti
PT

oning the
enzyme
and
CE

phospholi
pid
membrane
AC

PE Water, oil  Emulsifiers  Formulatio  Nanostruc


and used are of n at ture get
amphiphile nanometric nanoscale severely
size structures affected
are possible on higher
 The surfactant
stabilization  Recently concentrat
through manufactur ion
surface ed as

74
ACCEPTED MANUSCRIPT

adsorption. magnetic  When the


balls particle
size
 Easy distributio
preparation n is very
and scale broad and
up particles
 Lack of are used

T
flocculation at a non-
limiting

IP
,
sedimentati amount,

CR
on & only a
creaming small
fraction

US
of
particles
could
AN
actually
act as
Pickering
M

emulsion
stabilizers
ED

, whereas
broader
fractions
PT

would
remain
CE

non
adsorbed

Vesicles based systems


AC

Liposomes Phospholipid  Biological  Offer targeted  Less


s, ly inert drug delivery stability

cholesterol,  Biodegrad  Increase  Low


able and efficacy and solubility
water
weakly therapeutic
immunog index of drug  Short
enic half-life
 Non-toxic,
 Phospholi

75
ACCEPTED MANUSCRIPT

 Passive flexible, pids


targeting biocompatible undergoes
to REC by , completely oxidation,
pegylation biodegradable hydrolysis
possibility
 Help reduce  Leakage
to avoid
RES by exposure of and fusion
pegylation sensitive
tissues to  High

T
 Increased toxic drug productio

IP
plasma n cost
circulatio  Size can be

CR
varied to Allergic
n and reactions
increased incorporate
smaller or may occur
half life

US
larger drug to
 Active molecules liposomal
targeting constituen
AN
reduced  Can ts
toxicity incorporate
both water  They are
and
M

and lipid not


increased suitable
efficacy soluble drugs
ED

for
 Can be transderm
administered al delivery
PT

through
various routes
CE

Niosomes Non-ionic  Osmotical  Niosomes are  Physical


surfactant ly active. osmotically instability
and active,
AC

cholesterol  Site  Aggregati


specific  Chemically on
delivery stable and
have long  Fusion
 Biocompa storage time
tible,  Leaking
compared to of
biodegrad liposomes
able and entrapped
non-  They have drug
immunog high  Hydrolysi

76
ACCEPTED MANUSCRIPT

enic. compatibility s of
with encapsulat
 Less toxic biological ed drugs
and systems which
improves limiting
the  Low toxicity the shelf-
therapeuti because of life of the
c index of their non- dispersion
drug ionic nature

T
 They are

IP
Biodegradabl not
e and non- suitable

CR
immunogenic for
 They can transderm
al delivery

US
entrap
lipophilic
drugs into
AN
vesicular
bilayer
membranes
M

and
hydrophilic
ED

drugs in
aqueous
compartments
PT

 Sustained
release drug
CE

delivery
possible
AC

 Access to raw
materials is
convenient

Pharmacosomes Drug, lipid,  Physical  Less time  A


and required for compound
High purity
chemical formulation can be
solvents
stable than other synthesise
system vesicular d
systems depending

77
ACCEPTED MANUSCRIPT

 Consist of  The process on the


both of drug amphiphil
hydrophili release is ic nature
c and hydrolysis
hydropho rather than  They
bic drugs bilayer require
diffusion, superficial
 Can surface as well as
permeate mass

T
desorption, or
through degradation drug-lipid

IP
the cell as in case of interaction
membrane .

CR
liposomes.
, walls, or
tissues  The  Covalent
entrapment type of

US
 The rate efficiency of bond is
of pharmacosom required
degradatio to restrict
AN
es remain
n relies on unaffected by drug
size, the volume of leakage
nature of
M

inclusion  Pharmaco
functional
group  The somes are
ED

present in membrane susceptibl


the drug fluidity of e to get
fused,
PT

molecule pharmacosom
es is aggregate,
 Can be dependent on or
CE

administer conjugate hydrolyse


ed via phase by
topical, transition chemicals
AC

oral, temperature. on storage


extra- or
intravascu  There is no
lar route drug leakage
or
sedimentation
due to
covalent
binding of the
drug to the

78
ACCEPTED MANUSCRIPT

carrier

Phytosomes/her Herbal  Better  Phosphatidylc  Bioavaila


bosomes extracts, PC bioavailab holine acts a bility is
ility carrier as well limited in

T
andabsorp as has case of
tion hepatoprotecti orally or

IP
ve effect topical
 Easy applicatio

CR
manufactu  The ns
ring absorption
processes and  Require

US
bioavailabilit superficial
 Ingredient y of water as well as
s are of soluble mass
AN
natural phytoconstitu drug-lipid
origin ents is interaction
M

increased.
 Covalent
 Better type of
ED

therapeutic bond is
effects required
to restrict
PT

 Drug drug
targeting leakage
CE

 These are
susceptibl
AC

e to get
fused,
aggregate,
or
hydrolyse
by
chemicals
on storage

79
ACCEPTED MANUSCRIPT

Elastic Phospholipid  These can  They have  Unable to


liposomes s, surfactants accommo higher deliver
and edge date entrapment high-
activators hydropho efficiency molecular
bic as -weight
 They allow
well as complex
hydrophili enhanced molecules

T
c moieties permeation of to the

IP
drug through inner
 Due to skin layers of

CR
their skin
 They can
deformable
nature they increase the  Resistance

US
can transdermal to deliver
squeeze flux hydrophili
through  Prolonging c drug
AN
narrow the release moiety
constriction and  SC the
without
M

improving the outermost


measurable site-
loss layer and
specificity of
ED

interface
 Give bioactive with the
better molecules outside
PT

penetratio  They can world – is


n of intact accommodate considere
vesicles. d a major
CE

drug
molecules hydropho
 Can act as a
with a wide bic
carrier for
AC

range of crystalline
low as barrier to
well as solubility
TD. Thus,
high it is even
molecular more of a
weight hurdle to
substance deliver
s hydrophili
 Are made c and
up of high-

80
ACCEPTED MANUSCRIPT

natural molecular
phospholi -weight
pids hence complex
are molecules
biocompat to the
ible and inner
biodegrad strata of
able the skin

T
 Higher

IP
entrapmen
t

CR
efficiency

Ethosomes Phospholipid  Permeatio  Can deliver to  Poor yield

US
s,water, high n of drug targeted site
 In case if
quantity of through by non-
ethanol skin invasive shell
AN
means locking is
 Can ineffective
deliver  Large mol. then the
M

large wt.
molecules compounds ethosomes may
coalescence and
ED

(peptides, can be deliver


protein fall apart on
 High transfer into
molecules
PT

) is permeation water
possible rate observed
 Loss of
CE

 Ethosoma product
l system is during
passive, transfer
AC

non- form
invasive organic to
water
media

Transfersomes Phospholipid  These are  Large mol. wt  These are


s and edge self compounds chemicall
activators aggregates, can be deliver y unstable
(EAs) like with an at efficient because of
sodium ultra rate their

81
ACCEPTED MANUSCRIPT

cholate flexible  Due to its predisposi


(NaCo), membrane elastic nature tion to
sodium which leakage of oxidative
deoxycholate delivers the drug is degradatio
drug minimum n
reproducibl
 Efficient  Expensive
y into or
through the transdermal manufactu
delivery ring

T
skin

IP
 More elastic
than the

CR
standard
liposomes

US
 Suitable for
peptides
delivery
AN
Arachaeosomes Archaeal  Can be  The archaeal  Less pH
lipids sterilized lipids are stability
M

by more stable
 Expensive
autoclavin than other
manufactu
ED

g phospholipids
ring
 Have  Stability
PT

more toward
thermo- oxidative
labile degradation
CE

stability in would be
environm improved by
ent saturated
AC

alkyl chains
 Archaeal
lipids act  It does not
as self- require
adjuvant cholesterol in
drug the
delivery formulation
systems

Vesosomes Aqueous core  Multi-  Steric  Premature

82
ACCEPTED MANUSCRIPT

containing compartm stabilization contents


unilamellar ent release in
 pH loading of
vesicles structure physiologi
that drugs cal
function  Intrinsic environme
as internal biocompatibil nts. This
compartm ity premature
ents release is
 Better

T
which likely
contain protection to due to

IP
the drug the interior enzyme
and which contents in degradatio

CR
can vary serum, n or
in leading to protein
extended

US
compositi insertion
on from release of into the
each other encapsulated liposome
AN
drug membrane
 Easy to , which
produce significant
M

and offers ly
the increases
flexibility
ED

the bilayer
to deliver permeabili
multiple ty.
PT

drugs
within a
single
CE

carrier

Colloidosomes Microcapsule  Excellent  Offers great  Poor yield


AC

s control of potential in of
sizing controlling particles
Colloidal
the
particles  Flexible permeability  If shell
and of the locking is
adjustable entrapped insufficien
yield species and t then the
strength to allows the colloidoso
withstand selective and mes may
varying collapse

83
ACCEPTED MANUSCRIPT

mechanic time release and fall


al loads apart on
 Control of transfer
 Easily compactibilti into water.
constructa y allows
ble encapsulation  Large
of fragile and proportion
sensitive of
ingredients, colloidoso

T
such as mes

IP
biomolecules seems to
and cells. be lost on

CR
the
transfer
from

US
organic to
water
AN
media.

Lipospheres Tri Caprin,  Better  Extended  Low drug


Tri Lauren, physical release of loading
M

Stearic acid, stability loaded drug. capacity


of
ED

Hydrogenate  Low cost  High lipophilic


d vegetable of its entrapment of proteins.
oil, Tri componen hydrophobic
PT

Stearin, Ethyl ts. drug.  Variable


kinetics.
Stearate.  Ease of  Reduced
CE

preparatio mobility of  Drug


n. entrapped degradatio
AC

drug. n due to
 High high
degree of  Offers better pressure.
dispersibil biocompatibil
ity in ity and being  Insufficie
aqueous physiological nt
medium substance. stability.
loading of
lipophilic  Offers high  Different
drug. degree of lipid
variability modificati

84
ACCEPTED MANUSCRIPT

due to ons and


different colloidal
degree of species
esterification coexist
and chain that may
lengths or cause
even mixtures different
of lipid solubility

T
components. and
melting

IP
 Administrable point of
by various active and

CR
routes such as auxiliary
oral, I.V, I.M species.
and topical

US
route.

SLM Solid  Use of  Possibility of  Possibility


AN
hydrophobic biodegrad controlled of particle
fat core able lipids drug release growth
(triglyceride) and drug
M

 Allows targeting  SLMs


hydrophili dispersion
ED

c and/or  Protection of s have


hydropho incorporated high water
bic drugs labile drug content
PT

to be against
incorporat chemical  They
adjust the
CE

e degradation
release
 There is profile of
AC

chemical and the


physical incorporat
storage ed drug.
stability (for
both drug and  Safety
carrier profile
system) may be
uncertain

 Drug
loading

85
ACCEPTED MANUSCRIPT

may be
limited
due to the
solubility
and
miscibility
of the
drug in

T
the melted
lipid,

IP
chemical
and

CR
physical
structure

US
of lipid
materials,
and their
AN
polymorp
hic state.
M

SLN Physiological  Avoidanc  Organic  Low drug


lipid, e of solvents not loading
ED

dispersed in organic required for capacity


water or in an solvents formulation
aqueous  Drug
 Potential  Greater expulsion
PT

surfactant
wide stability and during
solution applicatio bioavailabilit storage
CE

n y
spectrum(  High
dermal, water
AC

per oral, content


intraveno  Low
us) and capacity
 High to load
pressure hydrophili
homogeni c drug
zation as
an
establishe

86
ACCEPTED MANUSCRIPT

d
productio
n method

T
IP
CR
US
AN
M
ED
PT
CE
AC

87
ACCEPTED MANUSCRIPT

Table 2 Characterization of nanocarriers


S.
Characterization parameters Instrumentation/ Analytical methods
No
Transmission electron microscopy (TEM)
Scanning electron microscopy (SEM)
1 Shape and surface morphology Phase contrast optical microscopy (PCM)
Atomic force microscopy (AFM)

T
Freeze fracture microscopy (FFM)

IP
2 Molecular weight Gel permeation chromatography (GPC)

CR
3 Surface charge and electrophoretic mobility Laser light scattering technique
Electron microscopy (SEM/TEM)

US
4 Vesicle size and size distribution Optical microscopy
Photon correlation spectroscopy (PCS)
Hydrophobic interaction chromatography
AN
Two phase partition
5 Surface hydrophobicity Radiolabel probe
M

Contact angle measurement


X-ray photoelectron spectroscopy
ED

Zeta potential measurement


6 Electrical surface potential and surface pH
pH sensitive probes
PT

7 Density Gas pycnometer


8 Rheology Viscometer
CE

Dialysis membrane
9 In-vitro release
Dissolution test apparatus
AC

88
ACCEPTED MANUSCRIPT

Table 3 Lipid based formulation under clinical trials (CT)

Product name Phase of Drug delivered Indication Reference


CT
151
Alocrest Phase I Vinorelbine NSCLC and breast cancers,
(Optisomes) non-Hodgkin’s lymphoma,
Hodgkin’s disease

T
152
ATI-1123 Phase I Docetaxel NSCLC, gastric, pancreatic

IP
(Protein stabilized cancer, and soft
Liposomes) tissue sarcoma

CR
153,154
Aroplatin Phase I/II Analog of Advanced pancreatic and
(MLVs) cisplatin colorectal cancer,

US
malignant pleural
mesothelioma, advanced
AN
solid malignancies
155,156
MCC-465 Phase I Doxorubicin Stomach cancer
M

(Antibody
conjugated
ED

PEGylated
liposomes)
PT

157-159
SGT-53 Phase I p53 DNA plasmid Solid tumors
(Anti-TfR
CE

conjugated
cationic liposomes)
AC

TKM-080301 Phase I/II PLK1 siRNA Gastrointestinal 160,161

(TKM-PLK1) neuroendocrine tumors,


(Cationic adrenocortical carcinoma,
PEGylated hepatocellular carcinoma
Liposomes)
162-164
Liposome- Phase I/II annamycin breast cancer, acute
annamycin lymphocytic leukemia

89
ACCEPTED MANUSCRIPT

NanoVNB Phase I Vinorelbine Advanced solid tumors


(PEGylated
liposomes)
165
Anti-EGFR Phase I Doxorubicin solid tumors
immunoliposomes
166-167
MBP-426 Phase II Oxaliplatin Gastric, gastroesophageal,
(Tf-conjugated esophageal

T
IP
liposomes) adenocarcinomas
168-170
EndoTAG-1 Phase II Paclitaxel Solid tumors

CR
(Cationic
liposomes)

US
171
OSI-211 Phase II Lurtotecan NSCLC, breast, colorectal,
(SUVs) ovarian, head and neck
AN
cancers
172
CPX-1 Phase II Irinotecan and Advanced solid tumors,
M

(ULVs) floxuridine (1:1) including


colorectal cancer
ED

173
LEP-ETU Phase II Paclitaxel Metastatic breast cancer
(Anionic liposomes)
PT

174
Pulmaquin/Lipoquin Phase II/III ciprofloxacin non-cystic fibrosis
bronchiectasis
CE

175-177
MM-398 (PEP02) Phase III Irinotecan Metastatic pancreatic
(PEGylated cancer
AC

liposomes)
178, 179
CPX-351 Phase III Cytarabine and Acute myeloid leukemia
(Bilamellar daunorubicin (5:1)
liposomes)
180
Thermodox Phase III Doxorubicin Hepatocellular carcinoma
(Lyso-lipid and breast cancer
temperature
sensitive liposomes)

90
ACCEPTED MANUSCRIPT

181,182
Lipoplatin Phase III Cisplatin NSCLC, gastric,
(PEGylated pancreatic, breast, head and
liposomes) neck cancers
183
Arikace Phase III amikacin lung infection

T
IP
CR
US
AN
M
ED
PT
CE
AC

91
ACCEPTED MANUSCRIPT

Table 4 List of marketed nanocarrier products

Nanocarrier
Active ingredient Indication Trade name Company
system
Enzon
P T
Pharmaceuticals Inc., Bridgewater, NJ,
Amphotericn B Fungal infections

Lymphomatous
Abelcet
USA
R I
Cytarabine
Meningitis
Depocyt

S CEnzon Pharmaceuticals Inc., NJ, USA

Amphotericn B

Daunorubicin
Fungal infections

Kaposi’s sarcoma
AmBisome

Daunoxome
N U Gilead Sciences Inc., Foster City, CA, USA

Gilead Sciences Inc., CA, USA


Doxorubicin Advanced breast Myocet
A Zeneus/Cephalon, Inc., Frazer, PA, USA
Inactivated Hepatitis cancer M
A virus
Hepatitis A

E D Epaxal Berna Biotech, Bern, Switzerland

T
Inactivated influenza
Influenza Inflexal V Berna Biotech, Bern, Switzerland
surface antigen
Morphine
E P
Analgesia DepoDur EKR Therapeutics, Bedminster, NJ, USA

Verteporfin
C C Age-related macular
degeneration
Visudyne
QLT Inc., Vancouver, British Colombia,
Canada; Norvatis, Basel, Switzerland

Liposomes
A
Doxorubicin
Ovarian cancer &
Kaposi’s sarcoma
Doxil Ortho Biotech, Bridgewater, NJ, USA

Ovarian cancer,
Doxorubicin Kaposi’s arcoma & Caelyx Schering-Plough, Kenilworth, NJ, USA
breast cancer

92
ACCEPTED MANUSCRIPT

Menopausal Hot
Estradiol Estrasorb Novavax, Rockville, MD, USA
flushes
Beractant (bovine Respiratory distress
Survanta Abbott Laboratories, IL, USA
lung homogenate) syndrome
Bovactant(bovine Respiratory distress
P T
Boehringer Ingelheim GmbH, Ingelheim
lung lavage)
Poractant
syndrome
alfa Respiratory distress
Alveofact
I
Germany
R
(porcine lung syndrome Curosurf
S C Chiesi Farmaceutici SpA, Parma, Italy

Polymeric
homogenate)
Paclitaxel
Cancer
Genexol-PM
N U Samyang Pharmaceutical, Daejeon City, Korea
micelles chemotherapy
A
Rapamune Sirolimus
M
Immuno suppressant
Elan Corporation, Dublin,
Pharmaceutical, Madison, NJ, USA
Ireland; yeth

Emend Aprepitant
E D Antiemetic
Elan Corporation, Dublin, Ireland; Merck and

P T Co., Inc. Whitehouse Station, NJ, USA


Elan Corporation, Dublin, Ireland; Abbott Labs,
Nanocrystalline
Drugs
Tricor

C E Fenofibrate Hyperlipidemia
Illinois, USA

Megace

A C Megestrol acetate Anorexia, Cachexia


Elan Corporation, Dublin,
Pharmaceuticals, Woodcliff Lake, NJ, USA
Ireland;

Protein
Abraxis BioScience, Los Angeles, CA, USA;
(albumin) Abraxane Paclitaxel Metastatic breast cancer
Astra Zeneca, London, UK
nanoparticles

93
ACCEPTED MANUSCRIPT

Lipid colloidal
Amphotec Amphotericin B Fungal infections InterMune, Brisbane, CA, USA
dispersion
Lidocaine &
ORAQIX® prilocaine Periodontal Anesthesia Dentsply Pharmaceutical

T
Microemulsion
periodontal gel
Neoral Cyclosporine Anticancer
I P
Wolters Kluwer

Lesion-directed
Aminolevulinic acid field-directed
and

C
treatment
R
Nanoemulsion AMELUZ
hydrochloride of actinic
U S
keratoses
Biofrontera

Phytosome Greenselect®
(AKs)

A
Complex of green Antioxidant, N liver
thermogenic Thorne Research
M
tea polyphenols & protective,
phosphatidylcholine (fat burning) effects

Transfersomes Diractin
Ketoprofen
E D in
Analgesic Idea AG

P T
Transfersome gel
Vitamin C and E Used in skin care and
Aquasome Aquasome EC-30

C E
derivatives anti-aging agent
Nikkol

A C

94
ACCEPTED MANUSCRIPT

Table 5 Contribution in the proposed area

Therapeutic
Nanocarriers Disease Contribution References
agent

Breast

P T
cancer  Comparative evaluation based on tissue distribution,

I
184
Nanospheres Mitoxantrone and lymph node acute toxicity, therapeutic efficacy against breast
metastases.
R
cancer and its lymph node metastases.

C
 Exploration of novel methods for selective drug
Folate-hapten
conjugate
Hapten Mice liver delivery
U S 185

 Investigation
N
 Identification of optimal targeting ligand

A of functionalized SWCNTs for

M
tumour targeting

D
Murine 4T1
SWCNTs PTX  CNTs as promising carrier with high entrapment 186
breast cancer

T E and less side effects

E P  Possibility of future cancer therapy


 Potential results in phase I clinical study of

C C escalating doses of the lapatinib in patients with


Albumin
NPs
bound
PTX
A Advanced solid
malignancies
advanced solid tumors
 Dynamic contrast enhanced magnetic resonance
187

imaging studies in a subset of patients confirmed a


decrease in tumor vascular permeability
 Formulation of thermosensitive magneto-liposomes 188
Thermo-sensitive MTX Skeletal

95
ACCEPTED MANUSCRIPT

magneto- muscular tissue (TMs) containing MTX


liposome  Superior magnetic targeting effect and rapid drug
release in response to hyperthermia
 Potential for cancer therapy
HER-2 over
P T
Albumin bound Carboplatin/ expressing
paclitaxel nanoparticles
R I
 Evaluation of efficacy and safety of albumin-bound
189
NPs trastuzumab metastatic breast
cancer C
 High activity in HER2-overexpression
S
Metastatic
N U
 Development of doxorubicin loaded SLN
 Assessment of in vitro haemolytic effect
SLN DOX
breast cancer A
 Enhanced apoptotic cell death through higher
190

M
accumulation of doxorubicin in cancer cells

E D  Formulation of novel nanocrytals for hydrophobic


Pluronic
polymer
F127
PTX and Murine
P T breast
drugs
 Excellent antitumor activity 191

nanocrystals
camptothecin

C E
cancer
 Low toxicity

Aerosol-OT A C Balb/c mice


 Easy scale-up

 Reported combinatorial anticancer activity of DOX


bearing Drug
(surfactant) and methylene blue 192
DOX Resistant
(AOT), and  Enhanced tumor accumulation of both DOX and
syngeneic JC
Sodium alginate methylene blue
tumors

96
ACCEPTED MANUSCRIPT

 Considerable inhibition of tumor cell proliferation


For co-delivery  Formulation of cationic SLN for co-delivery of PTX
Epithelial 193
Cationic SLN of PTX and and siRNA
carcinoma
siRNA  Enhanced tumor inhibition

P T
Exploration of nanocarrier mediated transdermal

Lipid based Topical


R I
delivery of Huperzine A (HupA) for the treatment

C
194
Huperzine A of Alzheimer's disease
nanocarriers administration

S
Formulation and characterization of ME, SLNs,
and NLCs
U

 A N
Designing of nanocarrier
Soft
nanocarriers
lipid Bioactive
vegetable oils
Antioxidant
activity M
Superior in vitro antioxidant activity mediated by
nanocarrier
195

ED Improved photoprotective effect

SLN, NLC, and T


Nutraceutical
P  Fabrication of lipid carrier as SLN, NLC and NE 196
lipid NE
Quercetin

C E
compound
delivery
 Greater entrapment & maximum bioaccessibility

Nanocarrier- A C  Emergence of hybrid (organic/inorganic)


nanoparticles to build “theranostic” systems
Cancer

197
Plasmonic gold DOX Highlighted superparamagnetic iron oxide
treatment
NPs nanoparticles (SPIONs) and plasmonic gold
nanoparticles (Au-NPs) for cancer treatment

97
ACCEPTED MANUSCRIPT

High activity
 Formulation of functional lipid nanocarriers with
against oxygen
Lipid unique features 198
natural oils free radicals and
nanocarriers  Greater antioxidant activity with nanocarrier
tumor cell
proliferation
 Better therapeutic effect

P T
Glucose 
 R I
Preparation of glucose-responsive nanocarriers
responsive
hydrogel- Insulin
Oral delivery of
insulin
Encapsulation of

S C insulin-loaded
responsive nanocarriers into a HA hydrogel
glucose-
199

nanocarriers
system

U
Better option for diabetes treatment via oral
ingestion
N
Biodegradable A
hyperbranched For efficient

M
Formulation of nanocarrier for topical delivery

core-multishell Dexamethasone topical drug


E D 

Better skin permeation by nanocarrier
Improved stability, low cytotoxicity, and easy
200

nanocarriers
(CMS)
delivery

P T scale up

C E
A C

98
ACCEPTED MANUSCRIPT

Highlights:

 Nanotechnology is the science and technology of precise manipulation in the materials,


devices or systems at nanometer scale.
 Pharmaceutical nanotechnology has offered fine-tuned diagnosis and focused disease
treatment.

T
 Recently lipids are becoming very popular as potential excipients for drug delivery as

IP
well as cosmetics.

CR
 Using nanotechnology based LBDDS, faster, patient friendly and economic treatments
can be established.

US
In recent years significant efforts have been made to explore the potentials of LBDDS.
 LBDDS having applications in various fields like pharmaceuticals, cosmetics,
nutraceuticals.
AN
 One of the major challenges faced by LBDDS is their susceptibility to oxidation.
 Pharmaceutical industries are more concerned in the designing of an appropriate drug
M

delivery system that could be adequately versatile and viable for scale up of the
ED

formulation.
 The safety of many of nanotechnological systems is yet to be determined.

PT

Lipid based system could be a potential molecular vector for clinical translation.
CE
AC

99
Graphics Abstract
Figure 1
Figure 2
Figure 3ab
Figure 3c
Figure 4ad
Figure 4eg

You might also like