Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Send Orders for Reprints to reprints@benthamscience.

net
Current Applied Polymer Science, 2020, 4, 1-9 1
REVIEW ARTICLE

Lipoidal-Nano Architecture for Parental Drug Delivery: Formulation De-


velopment and Regulatory Concerns
Vikas Jhawat1,*, Anil Kumar Sharma1, Vandana Garg2, Monika Gulia1 and Rohit Dutt1

1
School of Medical and Allied Sciences, GD Goenka University, Gurugram, Haryana, India; 2Department of Pharma-
ceutical Sciences, Mahrishi Dyananad University, Rohtak, Haryana, India

Abstract: The nanoparticles as drug carriers have demonstrated enhanced targeting, and sustained/-
controlled drug release, as evident from numerous investigations that have shown promising out-
comes facilitating the wellbeing of humans in the desired manner. The lipid-based nanoparticles
are biodegradable and considered biocompatible by virtue of being composed of lipid moieties
ARTICLE HISTORY
mimicking physiological lipids of biological systems which is their prime advantage over the other
Received: April 10, 2020 polymeric systems. A variety of such lipid carriers have been reported to be delivered from the par-
Revised: June 09, 2020 enteral route. However, there are certain pitfalls which are associated with lipid nanoparticles such
Accepted: June 11, 2020
as toxicity, poor scale up potential, immunological reactions and absence of straight forward regula-
DOI: tory guidelines that address the issues of lipoidal nanocarriers such as their classification, approval
10.2174/2452271604999200706014809 and compliance of governmental policies. Therefore attention must be given to address the techno-
logical and regulatory challenges associated with lipoidal nano-formulations for parenteral adminis-
tration to smoothen the approval process throughout the world and bringing the same to the termi-
nal users on time.

Keywords: Lipids carriers, nanotechnology, parenteral route, regulatory challenges, lipid nanoparticulates, lipoidal nanocarri-
ers.

1. INTRODUCTION carriers are one among them which, when processed at the
The parenteral route has been considered as the most ef- nano scale can result in biocompatible drug carriers. In
fective and fastest route for drug delivery and action of a the present manuscript, we reviewed the applications of
drug inside the body because of many reasons such as no nanotechnology in the area of parenteral drug delivery using
need for drug absorption phase, hundred percent bioavailabil- lipoidal carriers for improved drug delivery.
ity, no first-pass effect and formulation advantages. But de-
2. NANOTECHNOLOGY
spite many advantages, this route also has some limitations
related to the physicochemical properties of drugs such as Application of nanotechnology in the area of drug deliv-
the problem of solubility in blood components, drug release ery (nanocarrier drug delivery) could be considered promis-
from the formulation, non-target adherence of drug ing in circumventing above mentioned problems. Nanotech-
molecules, drug toxicity, burst effect and many more. The nology is the science which deals with the molecules at the
majority of the newly discovered drug molecules being hy- nano scale (1-100 nm) where molecules exhibit some specif-
drophobic in nature have low aqueous solubility, low bioa- ic size and shape-dependent magnetic, electrical and optical
vailability, and impaired pharmacokinetic behavior inside properties that are not generally observed in the bulk mate-
the body and tissue toxicity [1, 2]. During the drug develop- rial of the same compound [5, 6]. Nanotechnology allows us
ment phase, we cannot make a choice for the lead molecules to manipulate physicochemical properties in such a manner
to be hydrophilic or lipophilic and as per a study; there are that the molecule (nanoparticles) behaves as per the require-
more chances of the drug molecules being lipophilic which ments under different environmental conditions [7, 8]. Nano-
further may become the reason of rejection in later stages of
carrier drug delivery systems are of various types based on
the drug development due to poor water solubility [3, 4].
the polymeric systems utilized in the manufacturing and can
Therefore to avoid such problems, we need some modifica-
tions in the drug formulation to get the desired effect with be administered via different routes such as oral, nasal, ocu-
minimum adverse events. Nanotechnology can be of great lar, intraperitoneal, rectal, transdermal and parenteral route
importance in developing the desired formulation utilizing [9-12].
various synthetic and natural polymers carriers. Lipoidal
2.1. Parenteral Route
The focus of the current review is on drug delivery
* Address correspondence to this author at the School of Medical and Al-
lied Sciences, GD Goenka University, Gurugram, Haryana, India; through the parenteral route. This route offers several advan-
Tel: +91-9729216101; E-mail jhawat231287@gmail.com tages over other routes such as [13]:

2452-2716/20 $65.00+.00 © 2020 Bentham Science Publishers


2 Current Applied Polymer Science, 2020, Vol. 4, No. 0 Jhawat et al.

Dose accuracy with immediate onset of action poly (alkyl cyanoacrylates), poly (isobutyl cyanoacry-
Pain-free delivery of larger doses lates), poly (butylcyano acrylates), poly methyl
Easy to administer in unconscious and non-coopera- (methcyanoacrylates).
tive patients.
The unpleasant and bitter taste of the drug is no 2.2. Lipoidal Nanocarriers for Drugs
problem.
Lipoidal nanoparticulate delivery system can deliver the
Nanocarrier drug delivery systems play an important drug in such a manner that it may overcome the problems as-
role in improved parenteral administration of the drugs for sociated with the new drug candidates such as drug transpor-
the specific need. Nanoparticles delivered through parenteral tation problem, first pass metabolism, short residence time,
route have additional advantages such as [14]: poor absorption and permeation of the drug from blood ves-
sels to the tissues [17, 18]. When these lipid carriers are ad-
ministered through the parenteral route, it provides an excel-
Enhanced drug circulation time in the body which lent opportunity to inject the drug directly into the blood
prolongs the residence time of the drug for pro- stream for faster onset of action, site-specific delivery of the
longed therapeutic effect drug and encapsulation of both hydrophilic and lipophilic
Targeting and release of the drug at the desired site drugs [19, 20]. Lipoidal nanoparticles have demonstrated bet-
Improved physicochemical properties of drugs ter biocompatibility with the biological system than the or-
Bypassing the first-pass effect of drugs via oral ad- ganic nanoparticles because of the lipid nature of the biologi-
ministration. cal membranes [21]. Lipoidal nano-carrier drug delivery sys-
tem for parenteral administration can be formulated into dif-
Nanocarriers can be prepared using different biodegrad- ferent types of assemblies such as solid-lipid nanoparticles
able and biocompatible polymers such as [15, 16]: [16, 21, 22], nanoemulsions [23, 24], nanosuspensions [25],
niosomes [26], liposomes [27], lipid nanocapsule [28], lipid
[a] Natural lipids e.g. triglycerides, fatty acids, partial core micelles [29, 30], and nanostructured lipid carriers [31].
glycerides, steroids and waxes. The purpose of all the drug delivery systems is to maximize
[b] Hydrophilic polymers from natural origin e.g. ge- the efficacy of the drug at lower doses and to minimize the
latin, albumin, lecithin, legumin, alginate, dextran, side effects. Therefore, a suitable drug delivery system is de-
chitosan, agarose and pullulan. signed based on the physicochemical properties of the drug
[c] Synthetic hydrophobic polymer e.g. polyesters, poly molecules. Diverse lipid nanocarrier systems suitable for par-
(lactic acid), poly (lactide-co-glycolide), polystyrene, enteral administration are given below (Fig. 1):

Fig. (1). Lipoidal nanoarchitecture that can be used for a novel drug delivery system through parenteral route.
Lipoidal-Nano Architecture for Parental Current Applied Polymer Science, 2020, Vol. 4, No. 0 3

Solid lipid nanoparticles (SLN): Solid lipid nanoparti- with the cell membranes and release its content where re-
cles are made up of a mixture of solid lipids and liquid quired [48, 49].
lipids, which is stabilized by the addition of emulsifying
Lipid nanocapsule: Lipid nanocapsules are the vesicu-
agents. SLNs have unique properties such as small size (10
lar structure made up of three components namely; oily
nm to 1000 nm), large surface area, biocompatible and
phase such as triglycerides, aqueous phase such as purified
biodegradable, high drug loading capacity, and can load
water containing sodium chloride salt and non-ionic surfac-
both hydrophilic and lipophilic drugs [32, 33]. SLNs are pre-
tants for the stabilization of the nanocapsule [50-52]. The
pared from fatty acids, triglycerides, steroids and waxes us-
vesicles of lipid nanocapsule form an inner lipid core that en-
ing soybean lecithin, phosphatidylcholine, and sodium gluco-
traps the drug molecules and an open hydrophilic surfactant
nate as emulsifying agents [34].
tail outside. Nanocapsules can be used for sustained or con-
Nanoemulsion: Nanoemulsion is a fine to ultrafine ther- trolled delivery of the drug [53]. Lipid nanocapsules reduce
modynamically stable dispersed system which is made up of the drug degradation and increase the drug’s stability and wa-
water in oil (w/o) or oil in water (o/w) emulsion stabilized ter solubility [54, 55], and when administered through the
by emulsifying agents and have a size range of 50nm to parenteral route, lipid nanoparticles provide the direct access
1000nm [35]. Nanoemulsions, when seen through the naked of the drug into the blood which enables quick action and tar-
eye, appear transparent as fine globules in the emulsion, can- geting of the drug at the desired site [56].
not scatter the light rays [36]. Nanoemulsion is non-toxic
and non-irritant to the biological system and can be utilized Lipid core micelles: Along with the polymeric micelles,
as the drug carrier for a number of drugs [37]. Nanometric lipid core micelles formed by the phospholipids can also be
size of the dispersed phase in the nanoemulsion confers it utilized as the drug delivery carrier for poorly water-soluble
least chances of cracking, coalescence, flocculation, cream- drugs. The long-chain fatty acyl group present in the phos-
ing and sedimentation of the emulsion. pholipid can be formulated into a hydrophobic micelle core
capped by the hydrophilic polymer such as PEG, which
Nanosuspension: It is the biphasic dispersion system in forms the external hydrophilic part of the lipid micelle
which nano-sized particles (200nm-600nm) of the pure drug [57-59]. Phospholipids are a biocompatible and biodegrad-
are uniformly dispersed in the aqueous vehicle stabilized by able carrier and considered as safe drug delivery systems as
the surfactants. Reduction in the particle size of the drug to compared to polymeric systems. Therefore lipid-polymer
nanoscale increases the surface area and saturation solubility conjugate micelles prepared from phospholipids-PEG have a
of the drug particles and therefore, poorly water lipid-solu- size range of 7nm-35nm and at Critical Micelle Concentra-
ble drugs can be easily formulated with high bioavailability tion (CMC), the hydrophobic part forms the core of the mi-
profile [38-40]. celles that can uptake the poorly soluble drugs surrounded
Niosomes: Niosomes are unilamellar or multilamellar by the polymeric hydrophilic part. This type of micelles can
vesicles formed by the non-ionic surfactants with the combi- also be attached with the targeting moieties for a particular
nation of cholesterol or other lipids [41]. Cholesterol forms type of organ, tissue or cells to achieve the maximum con-
the hydrogen bond with the hydrophilic head of the non-ion- centration of the drug at that particular site [60].
ic surfactant, which impacts the physical properties of the Nanostructured lipid carrier: Nano-Structured Lipid
vesicles such as entrapment efficiency, stability, and drug re- Carriers (NLC) are similar in composition to the Solid Lipid
lease characters [42, 43]. These have the size range of 20 nm Nanoparticles (SLN) but NLCs have been designed to over-
to 200 nm and can encapsulate both hydrophilic and lipophil- come the limitations of SLN such as poor drug loading due
ic drugs for delivery to the human body. Niosomes are a sta- to crystalline nature of SLN, drug expulsion from the SLN,
ble, low cost, material easily available for preparation [44]. and up to 30% particle concentration in the aqueous environ-
These prolong the circulation time of the drug in the blood ment [61-63]. The NLCs are composed of surfactants, solid
and can easily penetrate the cells through different peptide lipidse.g., fats and liquid lipid e.g., oils complex at the room
channels, having a higher distribution rate [45]. temperature that has the size in the nanometer range. The
Liposomes: In contrast to the niosomes, liposomes are SLN contains up to 30% w/w of solid lipids content dis-
vesicular structure made up of phospholipids or sphin- persed in liquid aqueous part, but in NLC, solid lipid content
golipids, which contain an inner hydrophilic core protected may increase up to 95% w/w [64]. The NLC formulation
by an external hydrophobic lipid bilayer. Because of the contains liquid lipid (oil) in the core of the particles, which
presence of dual moieties, liposomes can be utilized for the increases the solubility and loading capacity of drugs. More-
delivery of both hydrophilic and lipophilic drugs [46]. Based over, complex lipids of NLC have very slow polymorphic
on the arrangement of lipid bilayers, liposomes can be classi- transitions, therefore lower chances of crystallization [65,
fied into unilamellar (containing single bilayer) and multil- 66]. The lipid complexity of NLC improves the encapsula-
amellar (more one lipid bilayer separated by aqueous space) tion efficiency, drug loading capacity, bioavailability along
[47]. The structure and composition of the lipid bilayer of li- with the physical stability and chemical stability of the drugs
posomes are almost similar to that of the biological mem- [64, 67]. NLCs, when administered through the parenteral
branes, which makes liposomes more biocompatible and route, provide higher bioavailability and pharmacological ac-
safer as compared to niosomes and other vesicles. Because tivity as compared to plain drug and provide protection to
of the structural similarity, liposomal bilayer can easily fuse the encapsulated drugs [68].
4 Current Applied Polymer Science, 2020, Vol. 4, No. 0 Jhawat et al.

Table 1. Drug delivery through different kinds of lipoidal nano architect systems

Sr. No. Drug Polymer carrier Formulation Ref


1. Doxorubicin Phosphatidylcholine Liposomes 72
2. Zidovudine Phosphatidylcholine, Cholesterol Liposomes 73
3. Clobetasol propionate Monostearin SLN 74
4. Cortisone Trimyristin-phospholipid mixture SLN 75
5. Itraconazole Tristearin and triolein Lipid nanoparticles 76
6. All-trans retinoic acid (ATRA) or Cetyl palmitate and soybean oil NLC 77
Tretinoin
7. Silybin Glycerol monostearate-triglyceride NLC 78
8. Docetaxel Stearic acid, glyceryl NLC 79
monostearate, soya lecithin and oleic acid
9. Paclitaxel - Nanoemulsion 80
10. Thalidomide Olive oil, soybean oil and soybean lecithin Nanoemulsion 81
11. Docetaxel Soya lecithin, Glycerol, ceramide Nanosuspension 82
12. Docetaxel Soya lecithin, DSPE-PEG-2000-amine and DSPE-PEG-2000-methoxy Nanosuspension 83
13. Zidovudine Cholesterol, Tweens, Spans, dicetylphosphate Niosome 84
14. Tamoxifen Caprylic-capric acid triglyceride and Soybean lecithin Lipid nanocapsule 85
15. Tretinoin Caprylic-capric acid triglyceride and Polycaprolactone Lipid nanocapsule 86
16. Docetaxel PEG 660 12-hydroxy stearate, Hydrogenated soybean phosphatidylcholine Lipid nanocapsule 87
17. Taxol Sodium glycocholate, phosphatidylcholine Lipid core micelle 88
18. Lipid core micelle Phosphatidylcholine and bile salt Lipid core micelle 89

2.3. Lipid Nano Carrier Delivery area, charge), pharmacokinetic (ADME) and pharmacologi-
Lipid nanocarriers, as discussed above, have the im- cal properties of the drug and polymers so that these can inte-
mense potential to deliver the variety of molecules for differ- grate into a complex system in a manner to target the drug at
ent purposes inside the body e.g., delivery of hydrophilic a specific site crossing the different biological barriers and
and hydrophobic drugs, gene, DNA, vitamins, hormones, deliver the drug at a specific rate [92, 93]. But along with
proteins, peptides and nutrients [69]. Lipoidal drug delivery many advantages, nanotechnology-based modified products
systems are preferred over the polymeric system because being at nano-size range have direct interaction with the hu-
lipid content of the delivery system resembles the lipid con- man biological system and living cells which may prove
tent of biological cell membranes, which provides easy ac- lethal because the body’s response to the nanoparticulate sys-
cess to the drug delivered through the different barriers [70, tems cannot be foreseen and it may also induce an immuno-
71]. However, lipoidal systems have their own limitations, logical and inflammatory reaction, toxicity and oxidative
which pose a hindrance in the way of the lipoidal drug deliv- stress [94, 95], accumulation of nanoparticles in the liver,
ery system to become the universal carrier. These limitations kidney or other organs [96], risk of dose dumping from the
can be overcome using an alternate lipoidal carrier or by in- nanoparticles, leakage of a drug at the undesired site, escape
corporating synthetic polymers in the lipoidal system. Differ- of the nanoparticulate system form the circulation, and im-
ent types of molecules delivered using the lipoidal carrier proper release of a drug from the nanoparticles [94]. Lipid
through the parenteral route are given in the below Table 1: content of the lipoidal nanocarrier system (cationic lipids)
can be toxic for the organ systems in which these accumu-
2.4. Challenges in the way of Lipodal Nano Architect Sys- late by interfering in the normal functioning of the cell mem-
tems brane. Nanoparticles administered via the intravenous route
get absorbed into the lymphatic system at the injection site
2.4.1. Toxicity and Side Effects and the situation worsens when cationic nano lipids adminis-
tered via parenteral route initiate inflammation-induced im-
Nanotechnology has wider applications in the area of
drug delivery, which offers many advantages over conventio-
munological reaction via activation of the phagocytic
nal drug delivery systems. It also has become successful to macrophages [97-99].
some extent and few nanotechnology-based products have
2.5. Technology Transfer Related Challenges
been introduced in the market [90, 91]. Scientists from all
over the world are working continuously on this novel sci- Technology transfer from the laboratory scale to the in-
ence with the hope of developing more efficient and safer dustrial scale for mass production of the nanoparticulate
drug delivery systems [91]. As nanoparticulate drug delivery products is also a big problem as it is very difficult to pro-
systems have been altered at the molecular level to alter the cess the materials at the nano scale without any variation
physico-chemical (particle size, shape, composition, surface [90]. Nanoparticles are refined entities that have some specif-
Lipoidal-Nano Architecture for Parental Current Applied Polymer Science, 2020, Vol. 4, No. 0 5

ic predefined features for which these are designed, such as mented evidence. But the approval of the novel nanoparticu-
the size, shape, surface area, integrity, toxicity, biodegrada- late drug delivery systems is a major concern which the regu-
bility, drug loading capacity, controlled drug release and in- latory agencies are facing due to lack of data regarding the
tegrity of the multicomponent system. There are higher classification of the new product, the chemistry of the prod-
chances of these features to be lost at large scale production uct, manufacturing process and control parameters of the
which makes the product unacceptable for physicians and pa- product [107]. For approval, a product must be categorized
tients. Therefore, the selection of polymers, solvents, meth- as either a medicine or a medical device. A medicine is an
ods of preparation and processing parameters needs to be re- entity that shows its intrinsic metabolic, pharmacological
addressed during scaling up operations, which is a challeng- and immunological activity while a medical device acts via
ing task [100, 101]. Other than technical challenges, there providing physical support or initiating chemical reaction
are many other challenges in technology transfer from labo- and may be a drug delivery carrier [108]. But the nanopartic-
ratory to the industry such as lack of funds, lack of adequate ulate drug delivery systems may have both types of activi-
laboratory setup, lack of marketing of the product, lack of ties as the drug contained in it performs pharmacological ac-
consumer for the product, lack of skilled manpower because tivity and the delivery system itself may act as a device
of nanotechnology being multidisciplinary, lack of analyti- which helps the drug to traverse across the body, providing
cal instruments, lack of knowledge on Intellectual Property
targeting potential and also acting as the diagnostic agent
Rights (IPR) to protect the product, lack of infrastructure re-
(Fig. 2).
quired and sustainability of the product in the market
[102-106]. Therefore, for the regulatory agencies, it would be very
difficult to categorize the nanoparticulate system. Likewise,
2.6. Regulatory Challenges the chemistry of the drug in the formulation determines its
To get the approval of any new product, its quality, safe- quality, safety and efficacy. So a novel nanoparticulate
ty and efficacy parameters must comply with the guidelines based product is expected to have a similar quality, safety
of the regulatory agency and must be supported with docu- and efficacy parameters to get approval from the regulatory

Fig. (2). Challenges for the approval of lipid nanoachitect drug delivery systems from the regulatory body.
6 Current Applied Polymer Science, 2020, Vol. 4, No. 0 Jhawat et al.

authority but the technical conditions of the nanoparticulate CONCLUSION


system with respect to chemistry, manufacturing method Nanotechnology has the potential to deliver drugs
and its controlling parameters are different from the conven- through different routes to serve the purpose of effective and
tional formulations which are difficult to comply as per cur- safe delivery. Different types of nanoformulations have been
rent regulatory conditions and therefore pose a problem in developed throughout the world to target the drugs at a spe-
getting approval [107]. Regulatory authorities also require cific site and to avoid problems encountered in conventional
data from toxicological studies, preclinical studies and clini- systems. Lipid-based nanoformulations are one of them
cal studies as proof of desired quality, safety and efficacy, which have been developed in recent years due to their bio-
but the assessment of novel nanoparticulate system is not compatibility with living system and biodegradable nature.
conducted in a manner as in conventional systems. Nanopar- Though nanotechnology offers several advantages over con-
ticles are reported to interfere with the normal physiological ventional drug carriers, yet it is not entirely devoid of nega-
system of the body, such as the administration of nanoparti- tive effects and poses many technical and regulatory chal-
cles induces immune-inflammatory and immune-modulatory lenges that need to be addressed. The technical complexity
responses, increases the cytokine release [94, 95] and even to manufacture and administer lipoidal nanoarchitecture
some nanoparticles may affect gene or gene expression and through the parenteral route requires special attention. Apart
induce cytotoxicity in the normal cells. So, the current regu- from the technical difficulties, the unique attributes of these
latory requirement for the conventional system cannot pro- dosage forms have resulted in non-uniformity of regulations
vide the approval to the nanoparticulate delivery systems. In as the regulatory bodies across the globe have found it diffi-
the same way, the limited knowledge about the clinical out- cult to categorize these lipoidal nanoarchitectures, formulate
comes of the nanoparticulate system restricts the regulatory user guidelines and comply governmental policies. There-
authority to approve these products. It is very difficult to de- fore, there is a need to address these challenges in the devel-
opment of lipoidal nano-formulations so as to facilitate drug
sign a clinical study and bioequivalence study for new prod-
delivery in a safe and effective manner and to bring these for-
ucts due to the lack of prior knowledge about the number of mulations in the market on time for the betterment of
patients required, study protocol, the initial dose required, mankind.
unavailability of biosimilars for comparison. Another most
important hurdle in the way of nanoparticulate drug delivery
systems is its size. The behavior of any chemical entity LIST OF ABBREVIATIONS
varies from macromolecular state to nano molecular state
but as per the regulatory guidelines, if any substance has a SLN = Solid Lipid Nanoparticles
similar molecular and chemical features (i.e. number of CMC = Critical Micelle Concentration
atoms, atomic and molecular arrangement of bonds and pat-
tern of bonding), it will be considered as the existing NLC = Nano Structured Lipid Carriers
molecule irrespective of the difference in the physical and bi- ATRA = All-trans Retinoic Acid
ological activities which may place the nanomaterial at a
greater risk than its macromolecular form. Moreover, high IPR = Intellectual Property Rights
surface area to volume ratio of nanomaterialsrenders them
highly reactive as they can pass through living biological CONSENT FOR PUBLICATION
membranes easily in high concentration and may cause sys- Not applicable.
temic toxicity in the doses similar to that of the macromolec-
ular form [109]. One such example is of iron oxide, which
FUNDING
has been used in the form of parenteral iron colloidal as a
conventional dosage form, but the competitors launched a None.
nanotechnology-based iron oxide parenteral product, which
is not therapeutically equivalent to the conventional form at CONFLICT OF INTEREST
same dose strength. But conventional regulatory guidelines
cannot deal with this kind of situation which compromises The authors have no conflicts of interest, financial or
the quality, safety and efficacy of the same drug in the same otherwise.
dose but at different size scales of the system [110, 111]. Dif-
ferences in the nature of material required for conventional ACKNOWLEDGEMENTS
dosage forms and novel nanotechnology-based dosage Declared none.
forms also create problems as the fate of new biodegradable
and non-biodegradable polymers inside the body is un- REFERENCES
known. Therefore, differences in the technical requirement
of conventional and novel drug delivery systems make the [1] Ahire E, Thakkar S, Darshanwad M, Misra M. Parenteral nanosus-
pensions: a brief review from solubility enhancement to more nov-
approval of the novel products challenging with the regulato- el and specific applications. Acta Pharm Sin B 2018; 8(5): 733-55.
ry guideline of the conventional system. http://dx.doi.org/10.1016/j.apsb.2018.07.011 PMID: 30245962
[2] Verma S, Burgess D. Solid nanosuspensions: the emerging tech-
nology and pharmaceutical applications as nanomedicine.Pharma-
ceutical suspensions. New York: Springer 2009; pp. 285-318.
Lipoidal-Nano Architecture for Parental Current Applied Polymer Science, 2020, Vol. 4, No. 0 7

[3] Lu Y, Li Y, Wu W. Injected nanocrystals for targeted drug deliv- ticles formulated from nano-emulsion templates-a review. J Con-
ery. Acta Pharm Sin B 2016; 6(2): 106-13. trol Release 2008; 128(3): 185-99.
http://dx.doi.org/10.1016/j.apsb.2015.11.005 PMID: 27006893 http://dx.doi.org/10.1016/j.jconrel.2008.02.007 PMID: 18374443
[4] Shegokar R, Müller RH. Nanocrystals: industrially feasible multi- [24] Rossi J. Principles in the development of intravenous lipid emul-
functional formulation technology for poorly soluble actives. Int J sions.Role of Lipid Excipients in Modifying Oral and Parenteral
Pharm 2010; 399(1-2): 129-39. Drug Delivery. Hoboken, New Jersey: Wiley-Interscience 2007;
http://dx.doi.org/10.1016/j.ijpharm.2010.07.044 PMID: 20674732 pp. 88-123.
[5] Nguyen KC, Willmore WG, Tayabali AF. Cadmium telluride [25] Wang Z, Li Z, Zhang D, Miao L, Huang G. Development of eto-
quantum dots cause oxidative stress leading to extrinsic and intrin- poside-loaded bovine serum albumin nanosuspensions for paren-
sic apoptosis in hepatocellular carcinoma HepG2 cells. Toxicolo- teral delivery. Drug Deliv 2015; 22(1): 79-85.
gy 2013; 306: 114-23. http://dx.doi.org/10.3109/10717544.2013.871600 PMID:
http://dx.doi.org/10.1016/j.tox.2013.02.010 PMID: 23485651 24401038
[6] Schmid G, Decker M, Ernst H, et al. Small dimensions and mate- [26] Doijad RC, Manvi FV, Swati S, Rony MS. Niosomal drug deliv-
rial properties A definition of nanotechnology. Europäische ery of Cisplatin: Development and characterization. Indian Drugs
Akademie 2003; pp. 1-135. 2008; 45: 713-8.
[7] Stylios GK, Giannoudis PV, Wan T. Applications of nanotechnolo- [27] Mamot C, Drummond DC, Hong K, Kirpotin DB, Park JW. Lipo-
gies in medical practice. Injury 2005; 36 (Suppl. 4): S6-S13. some-based approaches to overcome anticancer drug resistance.
http://dx.doi.org/10.1016/j.injury.2005.10.011 PMID: 16291325 Drug Resist Updat 2003; 6(5): 271-9.
[8] Yokoyama M. Drug targeting with nano-sized carrier systems. J http://dx.doi.org/10.1016/S1368-7646(03)00082-7 PMID:
Artif Organs 2005; 8(2): 77-84. 14643297
http://dx.doi.org/10.1007/s10047-005-0285-0 PMID: 16094510 [28] Marchiori ML, Lubini G, Dalla Nora G, et al. Hydrogel contain-
[9] Suri SS, Fenniri H, Singh B. Nanotechnology-based drug delivery ing dexamethasone-loaded nanocapsules for cutaneous administra-
systems. J Occup Med Toxicol 2007; 2: 16. tion: preparation, characterization, and in vitro drug release study.
http://dx.doi.org/10.1186/1745-6673-2-16 PMID: 18053152 Drug Dev Ind Pharm 2010; 36(8): 962-71.
[10] De Jong WH, Borm PJA. Drug delivery and nanoparticles:applica- http://dx.doi.org/10.3109/03639041003598960 PMID: 20590450
tions and hazards. Int J Nanomedicine 2008; 3(2): 133-49. [29] Bae Y, Kataoka K. Intelligent polymeric micelles from functional
http://dx.doi.org/10.2147/IJN.S596 PMID: 18686775 poly(ethylene glycol)-poly(amino acid) block copolymers. Adv
[11] Patra JK, Das G, Fraceto LF, et al. Nano based drug delivery sys- Drug Deliv Rev 2009; 61(10): 768-84.
tems: recent developments and future prospects. J Nanobiotechnol- http://dx.doi.org/10.1016/j.addr.2009.04.016 PMID: 19422866
ogy 2018; 16(1): 71. [30] Tyrrell ZL, Shen YQ, Radosz M. Fabrication of micellar nanopar-
http://dx.doi.org/10.1186/s12951-018-0392-8 PMID: 30231877 ticles for drug delivery through the self-assembly of block copoly-
[12] Surenya RS, Nair SKV, Kumar LV. Pharmaceutical nano drug de- mers. Prog Polym Sci 2010; 35: 1128-43.
livery route administration for cancer therapy. Adv Sci Eng Med http://dx.doi.org/10.1016/j.progpolymsci.2010.06.003
2015; 7: 739-45. [31] Radtke M, Muller RH. Novel concept of topical cyclosporine de-
http://dx.doi.org/10.1166/asem.2015.1767 livery with supersaturated SLN creams. Int Symp Control Rel
[13] Verma P, Thakur AS, Deshmukh K, Jha AK, Verma S. Routes of Bioact Mater 2001; 28: 470-1.
drug administration. Int J Pharm Stud Res 2010; 1: 54-9. [32] Cavalli R, Caputo O, Gasco MR. Solid lipospheres of doxorubicin
[14] Barenholz Y. Doxil®--the first FDA-approved nano-drug: lessons and idarubicin. Int J Pharm 1993; 89: R9-R12.
learned. J Control Release 2012; 160(2): 117-34. http://dx.doi.org/10.1016/0378-5173(93)90313-5
http://dx.doi.org/10.1016/j.jconrel.2012.03.020 PMID: 22484195 [33] zur Muhlen A, Mehnert W. Drug release and release mechanism
[15] Jawahar N, Meyyanathan SN. Polymeric nanoparticles for drug de- of prednisolone loaded solid lipid nanoparticles. Pharmazie 1998;
livery and targeting: A comprehensive review. Int J Health Allied 53: 552-5.
Sci 2012; 1: 217-23. [34] Zhang L, Pornpattananangku D, Hu CMJ, Huang CM. Develop-
http://dx.doi.org/10.4103/2278-344X.107832 ment of nanoparticles for antimicrobial drug delivery. Curr Med
[16] Mehnert W, Mäder K. Solid lipid nanoparticles: production, char- Chem 2010; 17(6): 585-94.
acterization and applications. Adv Drug Deliv Rev 2001; 47(2-3): http://dx.doi.org/10.2174/092986710790416290 PMID: 20015030
165-96. [35] Shah P, Bhalodia D, Shelat P. Nanoemulsion: A Pharmaceutical
http://dx.doi.org/10.1016/S0169-409X(01)00105-3 PMID: Review. Syst Rev Pharm 2010; 1: 24-32.
11311991 http://dx.doi.org/10.4103/0975-8453.59509
[17] Braddock M. Nanomedicines: design, delivery and detection. Cam- [36] McClements DJ. Colloidal bases of emulsion colour. Curr Opin
bridge: Royal Society of Chemistry 2016. Colloid In Sci 2002; 7: 451-5.
http://dx.doi.org/10.1039/9781782622536 http://dx.doi.org/10.1016/S1359-0294(02)00075-4
[18] Tian X, Li H, Zhang D, et al. Nanosuspension for parenteral deliv- [37] Aboofazeli R. Nanometric-scaled emulsions (nanoemulsions).
ery of a p-terphenyl derivative: preparation, characteristics and Iran J Pharm Res 2010; 9(4): 325-6.
pharmacokinetic studies. Colloids Surf B Biointerfaces 2013; 108: PMID: 24381596
29-33. [38] Patravale VB, Date AA, Kulkarni RM. Nanosuspensions: a
http://dx.doi.org/10.1016/j.colsurfb.2013.02.038 PMID: 23528604 promising drug delivery strategy. J Pharm Pharmacol 2004; 56(7):
[19] Panayiotis PC, Mahesh VC, Robert S. Advances in lipid nanodis- 827-40.
persions for parenteral drug delivery and targetting. Science 2008; http://dx.doi.org/10.1211/0022357023691 PMID: 15233860
60: 757-67. [39] Pu X, Sun J. Moli and Zhonggui He. Formulation of nanosuspen-
[20] Joshi MD, Müller RH. Lipid nanoparticles for parenteral delivery sion as a new approach for the delivery of poorly soluble drugs.
of actives. Eur J Pharm Biopharm 2009; 71(2): 161-72. Curr Nanosci 2009; 5: 417-27.
http://dx.doi.org/10.1016/j.ejpb.2008.09.003 PMID: 18824097 http://dx.doi.org/10.2174/157341309789378177
[21] Müller RH, Mäder K, Gohla S. Solid lipid nanoparticles (SLN) for [40] Muthu MS, Singh S. Poly (D, L-lactide) nanosuspensions of ris-
controlled drug delivery - a review of the state of the art. Eur J peridone for parenteral delivery: formulation and in-vitro evalua-
Pharm Biopharm 2000; 50(1): 161-77. tion. Curr Drug Deliv 2009; 6(1): 62-8.
http://dx.doi.org/10.1016/S0939-6411(00)00087-4 PMID: http://dx.doi.org/10.2174/156720109787048302 PMID: 19418957
10840199 [41] Uchegbu IF, Vyas SP. Non-ionic surfactant based vesicles (nio-
[22] Wissing SA, Kayser O, Müller RH. Solid lipid nanoparticles for somes) in drug delivery. Int J Pharm 1998; 172: 33-70.
parenteral drug delivery. Adv Drug Deliv Rev 2004; 56(9): http://dx.doi.org/10.1016/S0378-5173(98)00169-0
1257-72. [42] Akhilesh D, Bini KB, Kamath JV. Review on span-60 based non-
http://dx.doi.org/10.1016/j.addr.2003.12.002 PMID: 15109768 ionic surfactant vesicles (niosomes) as novel drug delivery. Int J
[23] Anton N, Benoit JP, Saulnier P. Design and production of nanopar- Res Pharm Biomed Sci 2012; 3: 6-12.
8 Current Applied Polymer Science, 2020, Vol. 4, No. 0 Jhawat et al.

[43] Seleci DA, Seleci M, Walter JG, Stahl F, Scheper T. Niosomes as http://dx.doi.org/10.1038/355279a0 PMID: 1731228
nanoparticular drug carriers: fundamentals and recent applica- [61] Hwang SR, Lim SJ, Park JS, Kim CK. Phospholipid-based microe-
tions. J Nanomater 2016; •••: 1-13. mulsion formulation of all-trans-retinoic acid for parenteral admin-
http://dx.doi.org/10.1155/2016/7372306 istration. Int J Pharm 2004; 276(1-2): 175-83.
[44] Fang JY, Hong CT, Chiu WT, Wang YY. Effect of liposomes and http://dx.doi.org/10.1016/j.ijpharm.2004.02.025 PMID: 15113624
niosomes on skin permeation of enoxacin. Int J Pharm 2001; [62] Müller RH, Radtke M, Wissing SA. Nanostructured lipid matrices
219(1-2): 61-72. for improved microencapsulation of drugs. Int J Pharm 2002;
http://dx.doi.org/10.1016/S0378-5173(01)00627-5 PMID: 242(1-2): 121-8.
11337166 http://dx.doi.org/10.1016/S0378-5173(02)00180-1 PMID:
[45] Carafa M, Marianecci C, Di Marzio L, et al. Potential dopamine 12176234
prodrug-loaded liposomes: preparation, characterization, and in [63] Radtke M, Muller RH. NLC-nanostructured lipid carriers: the new
vitro stability studies. J Liposome Res 2010; 20(3): 250-7. generation of lipid drug carriers. New Drugs 2001; 2: 97-138.
http://dx.doi.org/10.3109/08982100903384129 PMID: 19958070 [64] Pardeike J, Hommoss A, Müller RH. Lipid nanoparticles (SLN,
[46] Sharma A, Sharma US. Liposomes in drug delivery: progress and NLC) in cosmetic and pharmaceutical dermal products. Int J
limitations. Int J Pharm 1997; 154: 123-40. Pharm 2009; 366(1-2): 170-84.
http://dx.doi.org/10.1016/S0378-5173(97)00135-X http://dx.doi.org/10.1016/j.ijpharm.2008.10.003 PMID: 18992314
[47] Vemuri S, Rhodes CT. Preparation and characterization of lipo- [65] Shidhaye SS, Vaidya R, Sutar S, Patwardhan A, Kadam VJ. Solid
somes as therapeutic delivery systems: a review. Pharm Acta Helv lipid nanoparticles and nanostructured lipid carriers--innovative
1995; 70(2): 95-111. generations of solid lipid carriers. Curr Drug Deliv 2008; 5(4):
http://dx.doi.org/10.1016/0031-6865(95)00010-7 PMID: 7651973 324-31.
[48] Zhang L, Granick S. How to stabilize phospholipid liposomes (us- http://dx.doi.org/10.2174/156720108785915087 PMID: 18855604
ing nanoparticles). Nano Lett 2006; 6(4): 694-8. [66] Müller RH, Radtke M, Wissing SA. Solid lipid nanoparticles (SL-
http://dx.doi.org/10.1021/nl052455y PMID: 16608266 N) and nanostructured lipid carriers (NLC) in cosmetic and derma-
[49] Cevc G. Transfersomes, liposomes and other lipid suspensions on tological preparations. Adv Drug Deliv Rev 2002b; 54 (Suppl. 1):
the skin: permeation enhancement, vesicle penetration, and trans- S131-55.
dermal drug delivery. Crit Rev Ther Drug Carrier Syst 1996; http://dx.doi.org/10.1016/S0169-409X(02)00118-7 PMID:
13(3-4): 257-388. 12460720
http://dx.doi.org/10.1615/CritRevTherDrugCarrierSyst.v13.i3-4.3 [67] Teeranachaideekul V, Müller RH, Junyaprasert VB. Encapsula-
0 PMID: 9016383 tion of ascorbyl palmitate in nanostructured lipid carriers (NLC)---
[50] Minkov I, Ivanova T, Panaiotov I, Proust J, Saulnier P. Reorgani- effects of formulation parameters on physicochemical stability. Int
zation of lipid nanocapsules at air-water interface: Part 2. Proper- J Pharm 2007; 340(1-2): 198-206.
ties of the formed surface film. Colloids Surf B Biointerfaces http://dx.doi.org/10.1016/j.ijpharm.2007.03.022 PMID: 17482778
2005; 44(4): 197-203. [68] Jaiswal P, Gidwani B, Vyas A. Nanostructured lipid carriers and
http://dx.doi.org/10.1016/j.colsurfb.2005.07.001 PMID: 16081256 their current application in targeted drug delivery. Artif Cells
[51] Vonarbourg A, Saulnier P, Passirani C, Benoit JP. Electrokinetic Nanomed Biotechnol 2016; 44(1): 27-40.
properties of noncharged lipid nanocapsules: influence of the dipo- http://dx.doi.org/10.3109/21691401.2014.909822 PMID:
lar distribution at the interface. Electrophoresis 2005; 26(11): 24813223
2066-75. [69] Solaro R, Chiellini F, Battisti A. Targeted Delivery of Protein
http://dx.doi.org/10.1002/elps.200410145 PMID: 15852355 Drugs by Nanocarriers. Materials (Basel) 2010; 3: 1928-80.
[52] Huynh NT, Passirani C, Saulnier P, Benoit JP. Lipid nanocap- http://dx.doi.org/10.3390/ma3031928
sules: a new platform for nanomedicine. Int J Pharm 2009; 379(2): [70] Din FU, Aman W, Ullah I, et al. Effective use of nanocarriers as
201-9. drug delivery systems for the treatment of selected tumors. Int J
http://dx.doi.org/10.1016/j.ijpharm.2009.04.026 PMID: 19409468 Nanomedicine 2017; 12: 7291-309.
[53] Hoarau D, Delmas P, David S, Roux E, Leroux JC. Novel long-cir- http://dx.doi.org/10.2147/IJN.S146315 PMID: 29042776
culating lipid nanocapsules. Pharm Res 2004; 21(10): 1783-9. [71] Li J, Wang X, Zhang T, et al. A review on phospholipids and their
http://dx.doi.org/10.1023/B:PHAM.0000045229.87844.21 PMID: main applications in drug delivery systems. Asian J Pharm Sci
15553223 2015; 10: 81-98.
[54] Fachinetto JM, Ourique AF, Lubini G, Tedesco SB, Silva ACF, http://dx.doi.org/10.1016/j.ajps.2014.09.004
Beck RCR. Tretinoin-loaded polymeric nanocapsules: Evaluation [72] Gabizon AA. Applications of liposomal drug delivery systems to
of the potential to improve the antiproliferative activities on Alli- cancer therapy In nanotechnology for cancer therapy. Boca Raton:
um cepa root-tip compared to the free drug. Lat Am J Pharm CRC Press 2007; pp. 595-611.
2008; 27: 668-73. [73] Kaur CD, Nahar M, Jain NK. Lymphatic targeting of zidovudine
[55] Fontana MC, Coradini K, Guterres SS, Pohlmann AR, Beck RCR. using surface-engineered liposomes. J Drug Target 2008; 16(10):
Nanoencapsulation as a way to control the release and to increase 798-805.
the photostability of clobetasol propionate: influence of the nanos- http://dx.doi.org/10.1080/10611860802475688 PMID: 19005941
tructured system. J Biomed Nanotechnol 2009; 5(3): 254-63. [74] Hu FQ, Yuan H, Zhang HH, Fang M. Preparation of solid lipid
http://dx.doi.org/10.1166/jbn.2009.1030 PMID: 20055007 nanoparticles with clobetasol propionate by a novel solvent diffu-
[56] Constantinides PP, Chaubal MV, Shorr R. Advances in lipid nan- sion method in aqueous system and physicochemical characteriza-
odispersions for parenteral drug delivery and targeting. Adv Drug tion. Int J Pharm 2002; 239(1-2): 121-8.
Deliv Rev 2008; 60(6): 757-67. http://dx.doi.org/10.1016/S0378-5173(02)00081-9 PMID:
http://dx.doi.org/10.1016/j.addr.2007.10.013 PMID: 18096269 12052697
[57] Klibanov AL, Maruyama K, Torchilin VP, Huang L. Amphipathic [75] Westesen K, Bunjes H, Koch MHJ. Physicochemical characteriza-
polyethyleneglycols effectively prolong the circulation time of li- tion of lipid nanoparticles and evaluation of their drug loading ca-
posomes. FEBS Lett 1990; 268(1): 235-7. pacity and sustained release potential. J Control Release 1997; 48:
http://dx.doi.org/10.1016/0014-5793(90)81016-H PMID: 2384160 223-36.
[58] Trubetskoy VS, Torchilin VP. Use of polyoxyethylene-lipid conju- http://dx.doi.org/10.1016/S0168-3659(97)00046-1
gates as long-circulating carriers for delivery of therapeutic and di- [76] Kim JK, Park JS, Kim CK. Development of a binary lipid nanopar-
agnostic agents. Adv Drug Deliv Rev 1995; 16: 311-20. ticles formulation of itraconazole for parenteral administration and
http://dx.doi.org/10.1016/0169-409X(95)00032-3 controlled release. Int J Pharm 2010; 383(1-2): 209-15.
[59] Lasic DD, Woodle MC, Martin FJ, Valentincic T. Phase behavior http://dx.doi.org/10.1016/j.ijpharm.2009.09.008 PMID: 19747966
of ‘stealth-lipid’ decithin mixtures. Period Biol 1991; 93: 287-90. [77] Chinsriwongkul A, Chareanputtakhun P, Ngawhirunpat T, et al.
[60] Lasic DD. Mixed micelles in drug delivery. Nature 1992; Nanostructured lipid carriers (NLC) for parenteral delivery of an
355(6357): 279-80. anticancer drug. AAPS PharmSciTech 2012; 13(1): 150-8.
Lipoidal-Nano Architecture for Parental Current Applied Polymer Science, 2020, Vol. 4, No. 0 9

http://dx.doi.org/10.1208/s12249-011-9733-8 PMID: 22167418 [94] Sharma A, Madhunapantula SV, Robertson GP. Toxicological con-
[78] Jia L, Zhang D, Li Z, et al. Nanostructured lipid carriers for paren- siderations when creating nanoparticle-based drugs and drug deliv-
teral delivery of silybin: Biodistribution and pharmacokinetic ery systems. Expert Opin Drug Metab Toxicol 2012; 8(1): 47-69.
studies. Colloids Surf B Biointerfaces 2010; 80(2): 213-8. http://dx.doi.org/10.1517/17425255.2012.637916 PMID:
http://dx.doi.org/10.1016/j.colsurfb.2010.06.008 PMID: 20621458 22097965
[79] Liu D, Liu Z, Wang L, Zhang C, Zhang N. Nanostructured lipid [95] Dhawan A, Sharma V. Toxicity assessment of nanomaterials:
carriers as novel carrier for parenteral delivery of docetaxel. Col- methods and challenges. Anal Bioanal Chem 2010; 398(2):
loids Surf B Biointerfaces 2011; 85(2): 262-9. 589-605.
http://dx.doi.org/10.1016/j.colsurfb.2011.02.038 PMID: 21435845 http://dx.doi.org/10.1007/s00216-010-3996-x PMID: 20652549
[80] Constantinides PP, Lambert KJ, Tustian AK, et al. Formulation de- [96] Yang Y, Qin Z, Zeng W, et al. Toxicity assessment of nanoparti-
velopment and antitumor activity of a filter-sterilizable emulsion cles in various systems and organs. Nanotechnol Rev 2017; 6:
of paclitaxel. Pharm Res 2000; 17(2): 175-82. 279-89.
http://dx.doi.org/10.1023/A:1007565230130 PMID: 10751032 http://dx.doi.org/10.1515/ntrev-2016-0047
[81] Araújo FA, Kelmann RG, Araújo BV, Finatto RB, Teixeira HF, [97] Puri A, Loomis K, Smith B, et al. Lipid-based nanoparticles as
Koester LS. Development and characterization of parenteral na- pharmaceutical drug carriers: from concepts to clinic. Crit Rev
noemulsions containing thalidomide. Eur J Pharm Sci 2011; Ther Drug Carrier Syst 2009; 26(6): 523-80.
42(3): 238-45. http://dx.doi.org/10.1615/CritRevTherDrugCarrierSyst.v26.i6.10
http://dx.doi.org/10.1016/j.ejps.2010.11.014 PMID: 21130164 PMID: 20402623
[82] Wang T, Feng L, Yang S, Liu Y, Zhang N. Ceramide lipid-based [98] Miller CR, Bondurant B, McLean SD, McGovern KA, O’Brien
nanosuspension for enhanced delivery of docetaxel with synergis- DF. Liposome-cell interactions in vitro: effect of liposome surface
tic antitumor efficiency. Drug Deliv 2017; 24(1): 800-10. charge on the binding and endocytosis of conventional and sterical-
http://dx.doi.org/10.1080/10717544.2016.1225853 PMID: ly stabilized liposomes. Biochemistry 1998; 37(37): 12875-83.
28502199 http://dx.doi.org/10.1021/bi980096y PMID: 9737866
[83] Wang L, Li M, Zhang N. Folate-targeted docetaxel-lipid-based-- [99] Thanos C, Sandor M, Jong Y, et al. Inter-species uptake of poly-
nanosuspensions for active-targeted cancer therapy. Int J meric particles. Proc MRS 1999; 550: 65-70.
Nanomedicine 2012; 7: 3281-94. http://dx.doi.org/10.1557/PROC-550-65
PMID: 22802688 [100] Galindo-Rodríguez SA, Puel F, Briançon S, Allémann E, Doelker
[84] Ruckmani K, Sankar V, Sivakumar M. Tissue distribution, phar- E, Fessi H. Comparative scale-up of three methods for producing
macokinetics and stability studies of zidovudine delivered by nio- ibuprofen-loaded nanoparticles. Eur J Pharm Sci 2005; 25(4-5):
somes and proniosomes. J Biomed Nanotechnol 2010; 6(1): 357-67.
43-51. http://dx.doi.org/10.1016/j.ejps.2005.03.013 PMID: 15916889
http://dx.doi.org/10.1166/jbn.2010.1101 PMID: 20499831 [101] Colombo AP, Briançon S, Lieto J, Fessi H. Project, design, and
[85] Allard E, Passirani C, Garcion E, et al. Lipid nanocapsules loaded use of a pilot plant for nanocapsule production. Drug Dev Ind
with an organometallic tamoxifen derivative as a novel drug-carri- Pharm 2001; 27(10): 1063-72.
er system for experimental malignant gliomas. J Control Release http://dx.doi.org/10.1081/DDC-100108369 PMID: 11794809
2008; 130(2): 146-53. [102] Purushotham H. Transfer of nanotechnologies from R&D institu-
http://dx.doi.org/10.1016/j.jconrel.2008.05.027 PMID: 18582507 tions to SMEs in India. Tech Monitor 2012; pp. 23-33.
[86] Ourique AF, Azoubel S, Ferreira CV, et al. Lipid-core nanocap- [103] Khan MU. Problems of technol¬ogy transfer from laboratory to in-
sules as a nanomedicine for parenteral administration of tretinoin: dustry and policy issues in India. Int J Serv Technol Manag 2000;
development and in vitro antitumor activity on human myeloid 1: 375-94.
leukaemia cells. J Biomed Nanotechnol 2010; 6(3): 214-23. http://dx.doi.org/10.1504/IJSTM.2000.001583
http://dx.doi.org/10.1166/jbn.2010.1120 PMID: 21179938 [104] Mazzola L. Commercializing nanotechnology. Nat Biotechnol
[87] Khalid MN, Simard P, Hoarau D, Dragomir A, Leroux JC. Long 2003; 21(10): 1137-43.
circulating poly(ethylene glycol)-decorated lipid nanocapsules de- http://dx.doi.org/10.1038/nbt1003-1137 PMID: 14520392
liver docetaxel to solid tumors. Pharm Res 2006; 23(4): 752-8. [105] McNeil RD, Lowe J, Mastroianni T, Cronin J, Ferk D. Report-Bar-
http://dx.doi.org/10.1007/s11095-006-9662-5 PMID: 16550475 riers to nanotechnology commercialization. U.S. Department of
[88] Alkan-Onyuksel H, Ramakrishnan S, Chai HB, Pezzuto JM. A Commerce Technology Administration 2007.
mixed micellar formulation suitable for the parenteral administra- [106] Aithal PS, Aithal S. Nanotechnology Innovations and Commercial-
tion of taxol. Pharm Res 1994; 11(2): 206-12. ization –Opportunities, Challenges & Reasons for Delay. MPRA
http://dx.doi.org/10.1023/A:1018943021705 PMID: 7909371 2016; pp. 1-9.
[89] Hjelm RP, Thiyagarajan P, Onyuksel HA. Organization of phos- [107] Jain S, Edwards M, Spencer L. Advances and challenges in the de-
phatidycholine and bile salt in rodlike mixed micelles. J Phys velopment of drug delivery systems – A European perspective.
Chem 1992; 96: 8653-61. Regulatory Rapporteur 2016; 13: 4-8.
http://dx.doi.org/10.1021/j100200a080 [108] Medicines and Healthcare products Regulatory Agency (MHRA).
[90] Sahoo BK, Pattajoshi SP. Challenges of Nano Drug Delivery and Guidance: Decide if your product is a medicine or a medical de-
its Safety Issues. Int J Pharm and Pharm Res 2016; 6: 523-31. vice , 2019https://www.gov.uk/guidance/decide-if-your-produc-
[91] Winter E, Pizzol CD, Locatelli C, Crezkynski-Pasa TB. Develop- t-is-a-medicine-or-a-medical-device [05 April, 2020]; Published
ment and Evaluation of Lipid Nanoparticles for Drug Delivery: on 21 June 2013 and updated on 3 April 2019.
Study of Toxicity In vitro and In Vivo. J Nanosci Nanotechnol [109] Mühlebach S. Regulatory challenges of nanomedicines and their
2015; 15: 1-10. follow-on versions: A generic or similar approach? Adv Drug De-
PMID: 27433582 liv Rev 2018; 131: 122-31.
[92] Powers KW, Brown SC, Krishna VB, Wasdo SC, Moudgil BM, http://dx.doi.org/10.1016/j.addr.2018.06.024 PMID: 29966685
Roberts SM. Research strategies for safety evaluation of nanomate- [110] Mühlebach S, Borchard G, Yildiz S. Regulatory challenges and ap-
rials. Part VI. Characterization of nanoscale particles for toxicolog- proaches to characterize nanomedicines and their follow-on simi-
ical evaluation. Toxicol Sci 2006; 90(2): 296-303. lars. Nanomedicine (Lond) 2015; 10(4): 659-74.
http://dx.doi.org/10.1093/toxsci/kfj099 PMID: 16407094 http://dx.doi.org/10.2217/nnm.14.189 PMID: 25723097
[93] Oberdoster G. Safety assessment for nanotechnology and [111] David B Fischer. Nanotechnology - Scientific and Regulatory
nanomedicine: concept of nanotoxicology. J Intern Med 2009; Challenges, 19 Vill. Envtl LJ 2008; 315https://digitalcommon-
267: 89-105. s.law.villanova.edu/elj/vol19/iss2/2 Available at:
http://dx.doi.org/10.1111/j.1365-2796.2009.02187.x

You might also like