SlideShare a Scribd company logo
1 of 58
Niosomes and pharmacosomes 
Under the esteemed guidance of 
Mrs.Yasmin begum(M.Pharm, Ph.D) 
Presentation by 
P.Soujanya 
256212886026 
M.Pharm(pharmaceutics) 
Mallareddy College of Pharmacy
CONTENTS OF NIOSOMES 
 INTRODUCTION 
 STRUCTURE OF NIOSOMES 
 ADVANTAGES 
 MECHANISM OF NIOSOMES 
 FORMULATION 
 CHARACTERIZATION 
 EVALUATION 
 THERAPEUTIC APPLICATIONS 
 MARKETED PRODUCTS
INTRODUCTION 
• Niosomes are non-ionic surfactant vesicles obtained 
on hydration of synthetic nonionic surfactants, with 
or without incorporation of cholesterol or other 
lipids. 
• They are vesicular systems similar to liposomes that 
can be used as carriers of amphiphilic and lipophilic 
drugs 
• These are less toxic and improves the therapeutic 
index of drug by restricting its action to target cells
• Niosomes are a novel drug delivery system, in which the 
medication is encapsulated in a vesicle. 
• The niosomes are very small, and microscopic in size. Their size 
lies in the nanometric scale. 
• Niosomes are unilamellar or multilamellar vesicles.The vesicle is 
composed of a bilayer of non-ionic surface active agents and 
hence the name niosomes. 
• A diverse range of materials have been used to form niosomes 
such as sucrose ester surfactants and polyoxyethylene alkyl ether 
surfactants, alkyl ester, alkyl amides, fatty acids and amino acid 
compound.
Structure of Niosomes 
• Niosomes are microscopic lamellar structures which are formed on 
the admixture of non-ionic surfactant of the alkyl or dialkyl 
polyglycerol ether class and cholesterol with subsequent hydration in 
aqueous media. 
• The bilayer in the case of niosomes is made up of non-ionic surface 
active agents rather than phospholipids as seen in the case of 
liposome. 
• The niosome is made of a surfactant bilayer with its hydrophilic ends 
exposed on the outside and inside of the vesicle, while the 
hydrophobic chains face each other within the bilayer.
• Examples of non ionic surfactants used in the 
preparation of niosomes : 
1.Sorbitan alkyl ester (spans) 
2.Polyoxyethylene glycol Sorbitan alkyl ester 
(polysorbates) 
3.Polyoxyethylene glycol alkyl ether (Brij) 
4.Fatty alcohols example cetyl alcohol , stearyl alcohol 
5.Glycerol alkyl ester example glyceryl laurate
• Bilayer formation : 
Hydrophobic chains of surfactants are responsible for bilayer formation. 
It is explained by critical packaging parameter for surfactants Pc. It is 
given by 
Pc = v/ao.lc 
• the minimum interfacial area occupied by the head group- ao; 
• the volume of the hydrophobic tail (s)- v; 
• the maximum extended chain length of the tail in the micelle core-lc. 
If Pc is < 0.33 the expected aggregate structure is spherical micelles. 
If Pc is 0.5 to 1 the expected aggregate structure is vesicle bilayer 
structure. 
* So formed bilayer vesicle mimic biological cell membrane and have 
similar interactions which help in adsorption of vesicles on the cell 
membranes.
Role of cholesterol 
• Cholesterol, a natural steroid, is the 
most commonly used membrane 
additive 
• Stabilize the system against the 
formation of aggregates by repulsive 
steric or electrostatic effects 
• Prevents the transition from the gel 
state to liquid phase in niosomes 
systems by increasing hydrophobic 
interactions. 
• Niosomes become less leaky as 
cholesterol seals the pores or spaces on 
the bilayer. 
Cholesterol
Small 
Unilamellar 
Vesicle 
(SUV) 
Large 
Unilamellar 
Vesicle 
(LUV) 
Multilamellar 
vesicles(MLV) 
Typical Size Ranges: ULV: 20-50 nm – MLV:100-1000 nm
Advantages of niosomes 
• They possess an infrastructure consisting of hydrophobic and 
hydrophilic moieties together and as a result can accommodate 
drug molecules with a wide range of solubilities 
• They improve the therapeutic performance of the drug 
molecules by delaying the clearance from the circulation, 
protecting the drug from biological environment, and 
restricting effects to target cells 
• Handling and storage of surfactants requires no special 
conditions.
• Enhance skin penetration of drugs 
. 
• The vesicle suspension being water based offers greater 
patient compliance over oil based systems 
• Act as a depot to release drug slowly 
• The surfactants are biodegradable, biocompatible, and non 
immunogenic.
Advantages of niosomes over 
liposomes 
o In case of liposomes ,Ester bonds of phospholipids are easily 
hydrolyzed, at low pH 
o Peroxidation of unsaturated phospholipids. 
oAs liposomes have purified phospholipids they are to be 
stored and handled at inert(N2) atmospheres where as 
Niosomes are are made of non ionic surfactants and are 
easy to handle and store. 
oPhospholipid raw materials are naturally occurring 
substances and as such require extensive purification thus 
making them costly
Mechanism of Niosome 
1.Acts as Permeation enhancer In Topical 
preparation : Increases penetration of drug 
by destabilizing the stratum corneum. 
2.Acts as targeting agent : drug targeting 
includes both passive targeting and active 
targeting. 
•Passive targeting seen incase of solid tumors. 
• Active targeting includes : Ligand mediated 
targeting and physical mediated targeting (pH 
or temp changes)
Stealth niosomes 
• Any foreign matter is recognized by human immune system i.e, 
macrophages engulf them and provide protection. 
• Opsonins are proteins which help macrophages in engulfing the foreign 
matter ( including niosomes) . 
• Opsonins attach on niosomes and helps macrophages to recognize the 
niosomes for phagocytosis, Which result in degradation of niosomal 
preparation. 
• Stealth niosomes are one which prevent the attachment of opsonins on 
it. 
• Stealth niosomes are prepared by coating the niosomes with 
hydrophillic and non ionic polymers for ex PEG and its derivatives.
• Absence of electrostatic and hydrophobic forces and also steric 
hindrance of PEG opsonins cannot bind to niosomes. 
• PEG have flexible and extended conformation which prevents 
the opsonins in binding to the niosomes by creating strong 
repulsive forces.
Formulation of niosomes 
1.Ether injection method. 
It is essentially based on slow injection of surfactant : cholesterol in 
ether (20ml) through a 14 gauge needle at approximately 0.25 
ml/min. into a pre-heated aqueous phase maintained at 60°C. 
Vaporization of ether leads to formation of single layered vesicles. 
Later it results in the formation of a bilayer sheet, which eventually 
folds on itself to form sealed vesicles. 
 Depending upon the conditions used, the diameter of the vesicle 
range from 50 to 1000 nm.
2.. Hand shaking method (Thin film hydration technique). 
The mixture of vesicles forming ingredients like surfactant and 
cholesterol are dissolved in 10 ml of volatile organic solvent (diethyl 
ether, chloroform or methanol) in a round bottom flask. 
 The organic solvent is removed at room temperature (20°C) using 
rotary evaporator leaving a thin layer of solid mixture deposited on the 
wall of the flask. 
 The dried surfactant film can be rehydrated with aqueous phase by 
gentle agitation. 
 This process forms typical multilamellar niosomes. The liquid volume 
entrapped in vesicles appears to be small, i.e. 5-10%.
3. Sonication. 
 A typical method of production of the vesicles is by sonication of 
solution. 
In this method an aliquot of drug solution in buffer is added to the 
surfactant/cholesterol mixture in a 10-ml glass vial. 
 The mixture is probe sonicated at 60°C for 3 minutes using a 
sonicator with a titanium probe to yield niosomes. 
The method involves the formation of MLVs which are 
subjected to ultrasonic vibrations. 
Probe sonicator - when the volume of sample is small. 
Bath sonicator - when the volume of sample is large. 
Vesicles obtained are unilamellar in shape . 
*Care must be taken while working with temperature sensitive 
solute.
4.The “Bubble” Method. 
 It is novel technique for the one step preparation of liposome and 
niosomes without the use of organic solvents. 
The bubbling unit consists of round-bottomed flask with three necks 
positioned in water bath to control the temperature. 
 Water-cooled reflux and thermometer is positioned in the first and 
second neck and nitrogen supply through the third neck. 
Cholesterol and surfactant are dispersed together in this buffer (pH 7.4) 
at 70°C, the dispersion mixed for 15 seconds with high shear 
homogenizer and immediately afterwards “bubbled” at 70°C using 
nitrogen gas
5.Reverse Phase Evaporation Technique (REV) 
LUVs can also be prepared by forming a water in oil 
emulsion of surfactants and buffer in excess organic 
phase followed by removal of the organic phase under 
reduced pressure ( so called reverse phase 
evaporation) 
 The two phases are usually emulsified by sonication 
 The lipid or surfactant forms a gel first and 
subsequently hydrates to form vesicles. Free drug 
(unentrapped) is generally removed by dialysis.
Dialysis 
Separation of 
unentrapped 
material from 
niosomes 
Gel filtration Centrifugation 
Filtration
Separation of Unentrapped Drug 
1. Dialysis 
The aqueous niosomal dispersion is dialyzed in a dialysis tubing against 
phosphate buffer or normal saline or glucose solution. 
2. Gel Filtration 
The unentrapped drug is removed by gel filtration of niosomal dispersion 
through a Sephadex-G-50 column and elution with phosphate buffered 
saline or normal saline. 
3. Centrifugation 
The niosomal suspension is centrifuged and the supernatant is separated. 
The pellet is washed and then resuspended to obtain a niosomal 
suspension free from unentrapped drug.
Characterization 
1.Entrapment efficiency 
After preparing niosomal dispersion, unentrapped drug is separated by 
dialysis and the drug remained entrapped in niosomes is determined by 
complete vesicle disruption using 50% n-propanol or water soluble 
marker 0.1% Triton X-100 and analysing the resultant solution by 
appropriate assay method for the drug. 
2.Size, Shape and Morphology : 
Electron Microscopy and light scattering :- Morphological studies of 
vesicles 
Gel chromatography : size distribution
3.Niosomal drugloading and encapsulation efficiency 
The niosomal aqueous suspension was ultracentrifuged,supernant was 
removed and sediment was washed twice with distilled water in order to 
remove the adsorbed drug. 
Amount of drug in niosomes 
Entrapment efficiency (%)= --------------------------------------------X 100 
Amount of Drug used 
4. Vesicle Suface Charge 
Determined by measurement of electrophoretic mobility and expressed 
in terms of zeta potential
5. Niosomal drug release 
The simplest method to determine invitro release kinetics of the 
loaded drug is by incubating a known quantity of drug loaded 
niosomes in a buffer of suitable pH at 37°c with continuous stirring 
,withdrawing samples periodically and analysed the amount of 
drug by suitable analytical technique.dialysis bags or dialysis 
membranes are commonly used to minimize interference.
Evaluation of Niosomes 
In-vitro release : 
 A method of in-vitro release rate study includes the use of 
dialysis tubing. 
 A dialysis sac is washed and soaked in distilled water. The vesicle 
suspension is pipetted into a bag made up of the tubing and 
sealed. 
 The bag containing the vesicles is placed in 200 ml of buffer 
solution in a 250 ml beaker with constant shaking at 25°C or 37°C. 
 At various time intervals, the buffer is analyzed for the drug 
content by an appropriate assay method of vesicles during the 
cycle.
Franz diffusion cell: 
• In a Franz diffusion cell, the cellophane membrane is 
used as the dialysis membrane. 
• The niosomes are dialyzed through a cellophane 
membrane against suitable dissolution medium at room 
temperature. 
• The samples are withdrawn at suitable time intervals 
and analyzed for drug content.
Limitations 
• Aggregation on storage may be the drawback for the 
niosome preparation . 
• To avoid this instability Proniosomes are discovered. 
Proniosomes are dry formulation of surfactant coated 
carrier, which can be measured out as needed and 
rehydrated by brief agitation in hot water. 
Advantages: 
• Convenience of storage, transport and dosing. 
• More stable formulations. 
• Being dry powders makes processing and packaging easier.
Method of formation 
To create proniosomes, a water soluble carrier such as sorbitol is 
first coated with the surfactant. The coating is done by preparing a 
solution of the surfactant with cholesterol in a volatile organic 
solvent, which is sprayed onto the powder of sorbitol kept in a 
rotary evaporator. 
 The evaporation of the organic solvent yields a thin coat on the 
sorbitol particles. The resulting coating is a dry formulation in 
which a water soluble particle is coated with a thin film of dry 
surfactant. This preparation is termed Proniosome. 
The niosomes can be prepared from the proniosomes by adding 
the aqueous phase with the drug to the proniosomes with brief 
agitation at a temperature greater than the mean transition phase 
temperature of the surfactant.
THERAPEUTIC APPLICATIONS 
TREATMENT OF NEOPLASIA 
The anthracyclic antibiotic Doxorubicin, with broad 
spectrum anti tumour activity, shows a dose dependent 
irreversible cardio toxic effect. 
The halflife of drug increased by its niosomal entrapment of the drug and 
also prolonged its circulation and its metabolisom altered 
If the mice bearing S-180 tumour is treated with niosomal 
delivery of this drug it was observed that their life span 
increased and the rate of proliferation of sarcoma 
decreased. 
Methotrexate entrapped in niosomes if administered 
intravenously to S-180 tumour bearing mice results in total 
regression of tumour and also higher plasma level and 
slower elimination.
• Treatment of mitochondrial disorders : 
Drug should released intracellular and should be protected from 
lysosomal degradation. In this case niosomal preparation of 
medication (ubiquinone,ubiquinol) by using DOPE-PEG as coating 
material which prevents lysosomal degradation of the preparation.
• Treatment of vitiligo : 
Topical application of corticosteroids and calcium modulators using 
niosomal preparation. 
Niosomes known to enhance the permeation and facilitate the drug 
transport across the skin. 
• Treatment of glaucoma : 
Ophthalmic preparations of a acetazolamide niosomal preparation 
results in Increased corneal permeability and increases drug delivery.
Leishmaniasis therapy 
• Derivatives of antimony are most commonly prescribed 
drugs for the treatment of leishmaniasis. 
• These drugs in higher concentrations – can cause liver, 
cardiac and kidney damage. 
• Use of niosomes as a drug carrier showed that it is 
possible to overcome the side effects at higher 
concentration also and thus showed greater efficacy in 
Treatment.
• Magnetic Niosomes acts as contrasting agent in MRI imaging. 
• Niosomes as immunological adjuvant
List of Drugs formulated as Niosomes 
Intravenous route Doxorubicin, Methotrexate, Sodium Stibogluconate, Vincristine, 
Flurbiprofen, , Indomethacin, Colchicine,Rifampicin, Tretinoin, 
Transferrin and Glucose ligands, Zidovudine, Insulin,Cisplatin, Amarogentin, 
Daunorubicin, Amphotericin B, 5-Fluorouracil, Camptothecin 
Transdermal route Flurbiprofen, Piroxicam, Estradiol 
Levonorgestrol,Nimesulide,Dithranol, Ketoconazole, Enoxacin, Ketorolac 
Ocular route Timolol Maleate, Cyclopentolate 
Nasal route Sumatriptan, Influenza Viral Vaccine 
Inhalation All - trans retinoic acids
Marketed formulations 
• Lancome has come out with a variety of anti-ageing products 
which are based on niosome formulations . 
Anti ageing cream consists of D-Contraxol it is said to reduce the 
depth of wrinkles by working on the neuro -transmitters 
responsible for muscle contraction
PHARMACOSOMES
INTRODUCTION: 
• Pharmacosomes are the colloidal dispersions of drugs covalently 
bound to lipids, and may exist as ultrafine vesicular, micellar, or 
hexagonal aggregates, depending on the chemical structure of 
drug-lipid complex. 
• Pharmacosomes are amphiphilic phospholipid complexes of drugs 
bearing active hydrogen that bind to phospholipids. 
• Pharmacosomes impart better biopharmaceutical properties to 
the drug, resulting in improved bioavailability.
• Pharmacosomes have been prepared for various non-steroidal anti-inflammatory 
drugs, proteins, cardiovascular and antineoplastic drugs. 
• Developing the pharmacosomes of the drugs has been found to 
improve the absorption and minimize the gastrointestinal toxicity.
IMPORTANCE: 
• Pharmacosomes have importance in escaping the tedious steps of 
removing the free unentrapped drug from the formulation. 
• Pharmacosomes provide an efficient method for delivery of drug 
directly to the site of infection, leading to reduction of drug toxicity 
with no adverse effects and also reduces the cost of therapy by 
improved bioavailability of medication, especially in case of poorly 
soluble drugs. 
• Pharmacosomes are suitable for incorporating both hydrophilic and 
lipophilic drugs. 
• Entrapment efficiency is not only high but predetermined, because 
drug itself in conjugation with lipids forms vesicles
• There is no need of following the tedious, time-consuming step for 
removing the free, unentrapped drug from the formulation. 
• Since the drug is covalently linked, loss due to leakage of drug, does 
not take place. 
• No problem of drug incorporation 
• Encaptured volume and drug-bilayer interactions do not influence 
entrapment efficiency, in case of pharmacosomes
• In pharmacosomes, membrane fluidity depends upon the phase 
transition temperature of the drug lipid complex, but it does not affect 
release rate since the drug is covalently bound. 
• The drug is released from pharmacosome by hydrolysis (including 
enzymatic). 
• The physicochemical stability of the pharmacosome depends upon the 
physicochemical properties of the drug-lipid complex. 
• Following absorption, their conversion into active drug molecule 
depends to a great extent on the size and functional groups of drug 
molecule, the chain length of the lipids, and the spacer.
PREPARATION: 
• Two methods have been used to prepare vesicles: 
1. The hand-shaking method 
2. The ether-injection method 
• In the hand-shaking method, the dried film of the drug–lipid complex is 
deposited in a round-bottom flask and upon hydration with aqueous 
medium, readily gives a vesicular suspension. 
• In the ether-injection method, an organic solution of the drug–lipid 
complex is injected slowly into the hot aqueous medium, wherein the 
vesicles are readily formed.
FORMULATION OF PHARMACOSOMES: 
• Drug salt was converted into the acid form to provide an active 
hydrogen site for complexation. 
• Drug acid was prepared by acidification of an aqueous solution of 
drug salt, extraction into chloroform, and subsequent 
recrystallization. 
• Drug -PC complex was prepared by associating drug acid with an 
equimolar concentration of PC.
• The equimolar concentration of PC and drug acid were placed in a 
100-mL round bottom flask and dissolved in dichloromethane. 
• The solvent was evaporated under vacuum at 40°C in a rotary 
vacuum evaporator. 
• The pharmacosomes were collected as the dried residue and 
placed in a vacuum desiccator overnight and then subjected to 
characterization.
EVALUATION OF PHARMACOSOMES: 
• Solubility 
• Drug content 
• Differential scanning calorimetry 
• X-ray powder diffraction (XRPD)
Solubility: 
• To determine the change in solubility due to complexation, solubility 
of drug acid and drug-PC complex was determined in pH 6.8 
phosphate buffer and n-octanol by the shake-flask method. 
• Drug acid (50 mg) (and 50 mg equivalent in case of complex) was 
placed in a 100-mL conical flask. Phosphate buffer pH 6.8 (50 mL) was 
added and then stirred for 15 minutes. 
• The suspension was then transferred to a 250 mL separating funnel 
with 50 mL n-octanol and was shaken well for 30 minutes. 
• Then the separating funnel was kept still for about 30 minutes. 
Concentration of the drug was determined from the aqueous layer 
spectrophotometrically at 276 nm.
Drug content: 
• To determine the drug content in pharmacosomes of drug (e.g.: 
diclofenac-PC complex), a complex equivalent to 50 mg diclofenac 
was weighed and added into a volumetric flask with 100 mL of pH 6.8 
phosphate buffer. 
• Then the volumetric flask was stirred continuously for 24 h on a 
magnetic stirrer. At the end of 24 h, suitable dilutions were made and 
measured for the drug content at 276 nm UV spectrophotometrically.
Differential scanning calorimetry 
(DSC): 
• Thermograms of drug acid, phosphatidylcholine (80 %) and the drug 
-PC complex were recorded using a 2910 Modulated Differential 
Scanning Calorimeter V4.4E (TA Instruments, USA). 
• The thermal behavior was studied by heating 2.0 ± 0.2 mg of each 
individual sample in a covered sample pan under nitrogen gas flow. 
The investigations were carried out over the temperature range 25– 
250 °C at a heating rate of 10 °C min–1.
X-ray powder diffraction (XRPD): 
• The crystalline state of drug in the different samples was 
evaluated using X-ray powder diffraction. Diffraction patterns 
were obtained on a Bruker Axs- D8 Discover Powder X-ray 
diffractometer, Germany. 
• The X-ray generator was operated at 40 kV tube voltages and 40 
mA tube current, using lines of copper as the radiation source. 
The scanning angle ranged from 1 to 60° of 2q in the step scan 
mode (step width 0.4° min–1). 
• Drug acid, phosphatidylcholine 80 % (Lipoid S-80) and the 
prepared complex were analyzed.
APPLICATIONS: 
• The approach has successfully improved the therapeutic 
performance of various drugs i.e. pindolol maleate, bupranolol 
hydrochloride, taxol, acyclovir, etc. 
• The phase transition temperature of pharmacosomes in the 
vesicular and Micellar state could have significant influence on 
their interaction with membranes. 
• Pharmacosomes can interact with biomembranes enabling a 
better transfer of active ingredient. This interaction leads to 
change in phase transition temperature of biomembranes thereby 
improving the membrane fluidity leading to enhance 
permeations.
Conclusion 
• The concept of incorporating the drug into niosomes for a better 
targeting of the drug at appropriate tissue destination . 
• They presents a structure similar to liposome and hence they can 
represent alternative vesicular systems with respect to liposomes 
• Niosomes are thoughts to be better candidates drug delivery as 
compared to liposomes due to various factors like cost, stability etc. 
Various type of drug deliveries can be possible using niosomes like 
targeting, ophthalmic, topical, parentral, etc.
• Vesicular systems have been realized as extremely useful carrier 
systems in various scientific domains. Over the years, vesicular 
systems have been investigated as a major drug delivery system, 
due to their flexibility to be tailored for varied desirable purposes. 
• In spite of certain drawbacks, the vesicular delivery systems still play 
an important role in the selective targeting, and the controlled 
delivery of various drugs. 
• Researchers all over the world continue to put in their efforts in 
improving the vesicular system by making them steady in nature, in 
order to prevent leaching of contents, oxidation, and their uptake by 
natural defense mechanisms.
References: 
• S.P. Vyas R.K.Khar, Targeted And Controlled Drug Delivery novel carrier 
systems ; niosomes (p no.249-276) 
• N.K.JAIN , Controlled and Novel Drug Delivery; niosomes 
p no.(292-301) 
• H.A. Lieberman, M.M. Rieger, and G.S. Banker, 
Pharmaceutical Dosage Forms: Disperse Systems p. 163. 
• International Journal of Biopharmaceutics.2011;2(1):47-53; NIOSOMES 
FORMULATION AND EVALUATION
• Review article of PHARMACOSOMES: A Potential vesicular drug delivery 
system BY de pintu kumar,de arnab 
• Review article of PHARMACOSOMES:opening new doors for drug delivery 
BY anu goyal
Niosomes and pharmacosomes

More Related Content

What's hot

Nucleic acid based therapeutic drug delivery system
Nucleic acid based therapeutic  drug delivery systemNucleic acid based therapeutic  drug delivery system
Nucleic acid based therapeutic drug delivery systemtadisriteja9
 
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMS
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMSLIPOSOMES USED AS AN DRUG DELIVERY SYSTEMS
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMSROHIT
 
Microencapsulation
Microencapsulation Microencapsulation
Microencapsulation suhailk11
 
Solid lipid nanoparticle
Solid lipid nanoparticleSolid lipid nanoparticle
Solid lipid nanoparticleSagar Savale
 
Targeted drug delivery system
Targeted drug delivery systemTargeted drug delivery system
Targeted drug delivery systemHemant Khandoliya
 
Targeted Drug Delivery Systems
Targeted Drug Delivery SystemsTargeted Drug Delivery Systems
Targeted Drug Delivery SystemsSURYAKANTVERMA2
 
Coacervation Phase Separation Techniques
Coacervation Phase Separation TechniquesCoacervation Phase Separation Techniques
Coacervation Phase Separation TechniquesGargi Nanda
 
Bilosomes and emulsomes.pptx
Bilosomes and emulsomes.pptxBilosomes and emulsomes.pptx
Bilosomes and emulsomes.pptxParimal Hadge
 
Nanoparticles, types, preparation and evaluation ppt.pptx
Nanoparticles, types, preparation and evaluation ppt.pptxNanoparticles, types, preparation and evaluation ppt.pptx
Nanoparticles, types, preparation and evaluation ppt.pptxmanjureddy62
 
Mucoadhesive drug delivery system
Mucoadhesive drug delivery systemMucoadhesive drug delivery system
Mucoadhesive drug delivery systemAnita Duduskar
 
TRANSDERMAL DRUG DELIVERY SYSTEM
TRANSDERMAL DRUG DELIVERY SYSTEMTRANSDERMAL DRUG DELIVERY SYSTEM
TRANSDERMAL DRUG DELIVERY SYSTEMDr Gajanan Sanap
 

What's hot (20)

Liposome preparation and evaluation
Liposome preparation and evaluationLiposome preparation and evaluation
Liposome preparation and evaluation
 
Liposomes
LiposomesLiposomes
Liposomes
 
Nucleic acid based therapeutic drug delivery system
Nucleic acid based therapeutic  drug delivery systemNucleic acid based therapeutic  drug delivery system
Nucleic acid based therapeutic drug delivery system
 
Buccal drug delivery system
Buccal drug delivery systemBuccal drug delivery system
Buccal drug delivery system
 
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMS
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMSLIPOSOMES USED AS AN DRUG DELIVERY SYSTEMS
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMS
 
Aquasomes
AquasomesAquasomes
Aquasomes
 
Nanoparticles
NanoparticlesNanoparticles
Nanoparticles
 
Microencapsulation
Microencapsulation Microencapsulation
Microencapsulation
 
Aquasome ppt
Aquasome pptAquasome ppt
Aquasome ppt
 
Solid lipid nanoparticle
Solid lipid nanoparticleSolid lipid nanoparticle
Solid lipid nanoparticle
 
Targeted drug delivery system
Targeted drug delivery systemTargeted drug delivery system
Targeted drug delivery system
 
Niosomes
NiosomesNiosomes
Niosomes
 
Targeted Drug Delivery Systems
Targeted Drug Delivery SystemsTargeted Drug Delivery Systems
Targeted Drug Delivery Systems
 
Microcapsules and microspheres
Microcapsules and microspheresMicrocapsules and microspheres
Microcapsules and microspheres
 
Coacervation Phase Separation Techniques
Coacervation Phase Separation TechniquesCoacervation Phase Separation Techniques
Coacervation Phase Separation Techniques
 
Bilosomes and emulsomes.pptx
Bilosomes and emulsomes.pptxBilosomes and emulsomes.pptx
Bilosomes and emulsomes.pptx
 
Transdermal drug delivery system
Transdermal drug delivery systemTransdermal drug delivery system
Transdermal drug delivery system
 
Nanoparticles, types, preparation and evaluation ppt.pptx
Nanoparticles, types, preparation and evaluation ppt.pptxNanoparticles, types, preparation and evaluation ppt.pptx
Nanoparticles, types, preparation and evaluation ppt.pptx
 
Mucoadhesive drug delivery system
Mucoadhesive drug delivery systemMucoadhesive drug delivery system
Mucoadhesive drug delivery system
 
TRANSDERMAL DRUG DELIVERY SYSTEM
TRANSDERMAL DRUG DELIVERY SYSTEMTRANSDERMAL DRUG DELIVERY SYSTEM
TRANSDERMAL DRUG DELIVERY SYSTEM
 

Similar to Niosomes and pharmacosomes

Similar to Niosomes and pharmacosomes (20)

Niosomes
NiosomesNiosomes
Niosomes
 
Ppt.niosomebychetan
Ppt.niosomebychetanPpt.niosomebychetan
Ppt.niosomebychetan
 
NIOSOMES
NIOSOMESNIOSOMES
NIOSOMES
 
Niosomes by vbr
Niosomes  by vbrNiosomes  by vbr
Niosomes by vbr
 
Niosome
Niosome Niosome
Niosome
 
Niosomes by kalyan
Niosomes by kalyanNiosomes by kalyan
Niosomes by kalyan
 
Niosomes
NiosomesNiosomes
Niosomes
 
Niosomes Naveen Balaji
Niosomes Naveen BalajiNiosomes Naveen Balaji
Niosomes Naveen Balaji
 
niosome anshul vishwakarma .pptx
niosome anshul vishwakarma .pptxniosome anshul vishwakarma .pptx
niosome anshul vishwakarma .pptx
 
Niosomes
NiosomesNiosomes
Niosomes
 
Liposomes & Niosomes.pptx
Liposomes & Niosomes.pptxLiposomes & Niosomes.pptx
Liposomes & Niosomes.pptx
 
Niosomes, Aquasomes, Phytosomes,electrosomes Molecular pharmaceutics (MPH 201...
Niosomes, Aquasomes, Phytosomes,electrosomes Molecular pharmaceutics (MPH 201...Niosomes, Aquasomes, Phytosomes,electrosomes Molecular pharmaceutics (MPH 201...
Niosomes, Aquasomes, Phytosomes,electrosomes Molecular pharmaceutics (MPH 201...
 
Targeted dds
Targeted ddsTargeted dds
Targeted dds
 
Niosome drug delivery
Niosome drug deliveryNiosome drug delivery
Niosome drug delivery
 
niosome by shubham
niosome by shubhamniosome by shubham
niosome by shubham
 
Niosomes
NiosomesNiosomes
Niosomes
 
NIOSOMAL DRUG DELIVERY SYSTEM
NIOSOMAL DRUG DELIVERY SYSTEMNIOSOMAL DRUG DELIVERY SYSTEM
NIOSOMAL DRUG DELIVERY SYSTEM
 
Liposomes-Classification, methods of preparation and application
Liposomes-Classification, methods of preparation and application Liposomes-Classification, methods of preparation and application
Liposomes-Classification, methods of preparation and application
 
BASIC AND ADVANCE CONCEPT OF NIOSOME
BASIC AND ADVANCE CONCEPT OF NIOSOMEBASIC AND ADVANCE CONCEPT OF NIOSOME
BASIC AND ADVANCE CONCEPT OF NIOSOME
 
LIPOSOME
LIPOSOMELIPOSOME
LIPOSOME
 

More from Malla Reddy College of Pharmacy (20)

Rna secondary structure prediction
Rna secondary structure predictionRna secondary structure prediction
Rna secondary structure prediction
 
Proteomics
ProteomicsProteomics
Proteomics
 
Proteins basics
Proteins basicsProteins basics
Proteins basics
 
Protein structure classification
Protein structure classificationProtein structure classification
Protein structure classification
 
Protein identication characterization
Protein identication characterizationProtein identication characterization
Protein identication characterization
 
Protein modeling
Protein modelingProtein modeling
Protein modeling
 
Primerdesign
PrimerdesignPrimerdesign
Primerdesign
 
Phylogenetic studies
Phylogenetic studiesPhylogenetic studies
Phylogenetic studies
 
Multiple sequence alignment
Multiple sequence alignmentMultiple sequence alignment
Multiple sequence alignment
 
Homology modeling tools
Homology modeling toolsHomology modeling tools
Homology modeling tools
 
Homology modeling
Homology modelingHomology modeling
Homology modeling
 
Genome assembly
Genome assemblyGenome assembly
Genome assembly
 
Genome analysis2
Genome analysis2Genome analysis2
Genome analysis2
 
Genome analysis
Genome analysisGenome analysis
Genome analysis
 
Fasta
FastaFasta
Fasta
 
Drug design intro
Drug design introDrug design intro
Drug design intro
 
Drug design
Drug designDrug design
Drug design
 
Data retrieval
Data retrievalData retrieval
Data retrieval
 
Blast
BlastBlast
Blast
 
Biological databases
Biological databasesBiological databases
Biological databases
 

Niosomes and pharmacosomes

  • 1. Niosomes and pharmacosomes Under the esteemed guidance of Mrs.Yasmin begum(M.Pharm, Ph.D) Presentation by P.Soujanya 256212886026 M.Pharm(pharmaceutics) Mallareddy College of Pharmacy
  • 2. CONTENTS OF NIOSOMES  INTRODUCTION  STRUCTURE OF NIOSOMES  ADVANTAGES  MECHANISM OF NIOSOMES  FORMULATION  CHARACTERIZATION  EVALUATION  THERAPEUTIC APPLICATIONS  MARKETED PRODUCTS
  • 3. INTRODUCTION • Niosomes are non-ionic surfactant vesicles obtained on hydration of synthetic nonionic surfactants, with or without incorporation of cholesterol or other lipids. • They are vesicular systems similar to liposomes that can be used as carriers of amphiphilic and lipophilic drugs • These are less toxic and improves the therapeutic index of drug by restricting its action to target cells
  • 4. • Niosomes are a novel drug delivery system, in which the medication is encapsulated in a vesicle. • The niosomes are very small, and microscopic in size. Their size lies in the nanometric scale. • Niosomes are unilamellar or multilamellar vesicles.The vesicle is composed of a bilayer of non-ionic surface active agents and hence the name niosomes. • A diverse range of materials have been used to form niosomes such as sucrose ester surfactants and polyoxyethylene alkyl ether surfactants, alkyl ester, alkyl amides, fatty acids and amino acid compound.
  • 5. Structure of Niosomes • Niosomes are microscopic lamellar structures which are formed on the admixture of non-ionic surfactant of the alkyl or dialkyl polyglycerol ether class and cholesterol with subsequent hydration in aqueous media. • The bilayer in the case of niosomes is made up of non-ionic surface active agents rather than phospholipids as seen in the case of liposome. • The niosome is made of a surfactant bilayer with its hydrophilic ends exposed on the outside and inside of the vesicle, while the hydrophobic chains face each other within the bilayer.
  • 6.
  • 7. • Examples of non ionic surfactants used in the preparation of niosomes : 1.Sorbitan alkyl ester (spans) 2.Polyoxyethylene glycol Sorbitan alkyl ester (polysorbates) 3.Polyoxyethylene glycol alkyl ether (Brij) 4.Fatty alcohols example cetyl alcohol , stearyl alcohol 5.Glycerol alkyl ester example glyceryl laurate
  • 8. • Bilayer formation : Hydrophobic chains of surfactants are responsible for bilayer formation. It is explained by critical packaging parameter for surfactants Pc. It is given by Pc = v/ao.lc • the minimum interfacial area occupied by the head group- ao; • the volume of the hydrophobic tail (s)- v; • the maximum extended chain length of the tail in the micelle core-lc. If Pc is < 0.33 the expected aggregate structure is spherical micelles. If Pc is 0.5 to 1 the expected aggregate structure is vesicle bilayer structure. * So formed bilayer vesicle mimic biological cell membrane and have similar interactions which help in adsorption of vesicles on the cell membranes.
  • 9. Role of cholesterol • Cholesterol, a natural steroid, is the most commonly used membrane additive • Stabilize the system against the formation of aggregates by repulsive steric or electrostatic effects • Prevents the transition from the gel state to liquid phase in niosomes systems by increasing hydrophobic interactions. • Niosomes become less leaky as cholesterol seals the pores or spaces on the bilayer. Cholesterol
  • 10. Small Unilamellar Vesicle (SUV) Large Unilamellar Vesicle (LUV) Multilamellar vesicles(MLV) Typical Size Ranges: ULV: 20-50 nm – MLV:100-1000 nm
  • 11. Advantages of niosomes • They possess an infrastructure consisting of hydrophobic and hydrophilic moieties together and as a result can accommodate drug molecules with a wide range of solubilities • They improve the therapeutic performance of the drug molecules by delaying the clearance from the circulation, protecting the drug from biological environment, and restricting effects to target cells • Handling and storage of surfactants requires no special conditions.
  • 12. • Enhance skin penetration of drugs . • The vesicle suspension being water based offers greater patient compliance over oil based systems • Act as a depot to release drug slowly • The surfactants are biodegradable, biocompatible, and non immunogenic.
  • 13. Advantages of niosomes over liposomes o In case of liposomes ,Ester bonds of phospholipids are easily hydrolyzed, at low pH o Peroxidation of unsaturated phospholipids. oAs liposomes have purified phospholipids they are to be stored and handled at inert(N2) atmospheres where as Niosomes are are made of non ionic surfactants and are easy to handle and store. oPhospholipid raw materials are naturally occurring substances and as such require extensive purification thus making them costly
  • 14. Mechanism of Niosome 1.Acts as Permeation enhancer In Topical preparation : Increases penetration of drug by destabilizing the stratum corneum. 2.Acts as targeting agent : drug targeting includes both passive targeting and active targeting. •Passive targeting seen incase of solid tumors. • Active targeting includes : Ligand mediated targeting and physical mediated targeting (pH or temp changes)
  • 15. Stealth niosomes • Any foreign matter is recognized by human immune system i.e, macrophages engulf them and provide protection. • Opsonins are proteins which help macrophages in engulfing the foreign matter ( including niosomes) . • Opsonins attach on niosomes and helps macrophages to recognize the niosomes for phagocytosis, Which result in degradation of niosomal preparation. • Stealth niosomes are one which prevent the attachment of opsonins on it. • Stealth niosomes are prepared by coating the niosomes with hydrophillic and non ionic polymers for ex PEG and its derivatives.
  • 16. • Absence of electrostatic and hydrophobic forces and also steric hindrance of PEG opsonins cannot bind to niosomes. • PEG have flexible and extended conformation which prevents the opsonins in binding to the niosomes by creating strong repulsive forces.
  • 17. Formulation of niosomes 1.Ether injection method. It is essentially based on slow injection of surfactant : cholesterol in ether (20ml) through a 14 gauge needle at approximately 0.25 ml/min. into a pre-heated aqueous phase maintained at 60°C. Vaporization of ether leads to formation of single layered vesicles. Later it results in the formation of a bilayer sheet, which eventually folds on itself to form sealed vesicles.  Depending upon the conditions used, the diameter of the vesicle range from 50 to 1000 nm.
  • 18. 2.. Hand shaking method (Thin film hydration technique). The mixture of vesicles forming ingredients like surfactant and cholesterol are dissolved in 10 ml of volatile organic solvent (diethyl ether, chloroform or methanol) in a round bottom flask.  The organic solvent is removed at room temperature (20°C) using rotary evaporator leaving a thin layer of solid mixture deposited on the wall of the flask.  The dried surfactant film can be rehydrated with aqueous phase by gentle agitation.  This process forms typical multilamellar niosomes. The liquid volume entrapped in vesicles appears to be small, i.e. 5-10%.
  • 19. 3. Sonication.  A typical method of production of the vesicles is by sonication of solution. In this method an aliquot of drug solution in buffer is added to the surfactant/cholesterol mixture in a 10-ml glass vial.  The mixture is probe sonicated at 60°C for 3 minutes using a sonicator with a titanium probe to yield niosomes. The method involves the formation of MLVs which are subjected to ultrasonic vibrations. Probe sonicator - when the volume of sample is small. Bath sonicator - when the volume of sample is large. Vesicles obtained are unilamellar in shape . *Care must be taken while working with temperature sensitive solute.
  • 20. 4.The “Bubble” Method.  It is novel technique for the one step preparation of liposome and niosomes without the use of organic solvents. The bubbling unit consists of round-bottomed flask with three necks positioned in water bath to control the temperature.  Water-cooled reflux and thermometer is positioned in the first and second neck and nitrogen supply through the third neck. Cholesterol and surfactant are dispersed together in this buffer (pH 7.4) at 70°C, the dispersion mixed for 15 seconds with high shear homogenizer and immediately afterwards “bubbled” at 70°C using nitrogen gas
  • 21. 5.Reverse Phase Evaporation Technique (REV) LUVs can also be prepared by forming a water in oil emulsion of surfactants and buffer in excess organic phase followed by removal of the organic phase under reduced pressure ( so called reverse phase evaporation)  The two phases are usually emulsified by sonication  The lipid or surfactant forms a gel first and subsequently hydrates to form vesicles. Free drug (unentrapped) is generally removed by dialysis.
  • 22. Dialysis Separation of unentrapped material from niosomes Gel filtration Centrifugation Filtration
  • 23. Separation of Unentrapped Drug 1. Dialysis The aqueous niosomal dispersion is dialyzed in a dialysis tubing against phosphate buffer or normal saline or glucose solution. 2. Gel Filtration The unentrapped drug is removed by gel filtration of niosomal dispersion through a Sephadex-G-50 column and elution with phosphate buffered saline or normal saline. 3. Centrifugation The niosomal suspension is centrifuged and the supernatant is separated. The pellet is washed and then resuspended to obtain a niosomal suspension free from unentrapped drug.
  • 24. Characterization 1.Entrapment efficiency After preparing niosomal dispersion, unentrapped drug is separated by dialysis and the drug remained entrapped in niosomes is determined by complete vesicle disruption using 50% n-propanol or water soluble marker 0.1% Triton X-100 and analysing the resultant solution by appropriate assay method for the drug. 2.Size, Shape and Morphology : Electron Microscopy and light scattering :- Morphological studies of vesicles Gel chromatography : size distribution
  • 25. 3.Niosomal drugloading and encapsulation efficiency The niosomal aqueous suspension was ultracentrifuged,supernant was removed and sediment was washed twice with distilled water in order to remove the adsorbed drug. Amount of drug in niosomes Entrapment efficiency (%)= --------------------------------------------X 100 Amount of Drug used 4. Vesicle Suface Charge Determined by measurement of electrophoretic mobility and expressed in terms of zeta potential
  • 26. 5. Niosomal drug release The simplest method to determine invitro release kinetics of the loaded drug is by incubating a known quantity of drug loaded niosomes in a buffer of suitable pH at 37°c with continuous stirring ,withdrawing samples periodically and analysed the amount of drug by suitable analytical technique.dialysis bags or dialysis membranes are commonly used to minimize interference.
  • 27. Evaluation of Niosomes In-vitro release :  A method of in-vitro release rate study includes the use of dialysis tubing.  A dialysis sac is washed and soaked in distilled water. The vesicle suspension is pipetted into a bag made up of the tubing and sealed.  The bag containing the vesicles is placed in 200 ml of buffer solution in a 250 ml beaker with constant shaking at 25°C or 37°C.  At various time intervals, the buffer is analyzed for the drug content by an appropriate assay method of vesicles during the cycle.
  • 28. Franz diffusion cell: • In a Franz diffusion cell, the cellophane membrane is used as the dialysis membrane. • The niosomes are dialyzed through a cellophane membrane against suitable dissolution medium at room temperature. • The samples are withdrawn at suitable time intervals and analyzed for drug content.
  • 29. Limitations • Aggregation on storage may be the drawback for the niosome preparation . • To avoid this instability Proniosomes are discovered. Proniosomes are dry formulation of surfactant coated carrier, which can be measured out as needed and rehydrated by brief agitation in hot water. Advantages: • Convenience of storage, transport and dosing. • More stable formulations. • Being dry powders makes processing and packaging easier.
  • 30. Method of formation To create proniosomes, a water soluble carrier such as sorbitol is first coated with the surfactant. The coating is done by preparing a solution of the surfactant with cholesterol in a volatile organic solvent, which is sprayed onto the powder of sorbitol kept in a rotary evaporator.  The evaporation of the organic solvent yields a thin coat on the sorbitol particles. The resulting coating is a dry formulation in which a water soluble particle is coated with a thin film of dry surfactant. This preparation is termed Proniosome. The niosomes can be prepared from the proniosomes by adding the aqueous phase with the drug to the proniosomes with brief agitation at a temperature greater than the mean transition phase temperature of the surfactant.
  • 31.
  • 32. THERAPEUTIC APPLICATIONS TREATMENT OF NEOPLASIA The anthracyclic antibiotic Doxorubicin, with broad spectrum anti tumour activity, shows a dose dependent irreversible cardio toxic effect. The halflife of drug increased by its niosomal entrapment of the drug and also prolonged its circulation and its metabolisom altered If the mice bearing S-180 tumour is treated with niosomal delivery of this drug it was observed that their life span increased and the rate of proliferation of sarcoma decreased. Methotrexate entrapped in niosomes if administered intravenously to S-180 tumour bearing mice results in total regression of tumour and also higher plasma level and slower elimination.
  • 33. • Treatment of mitochondrial disorders : Drug should released intracellular and should be protected from lysosomal degradation. In this case niosomal preparation of medication (ubiquinone,ubiquinol) by using DOPE-PEG as coating material which prevents lysosomal degradation of the preparation.
  • 34. • Treatment of vitiligo : Topical application of corticosteroids and calcium modulators using niosomal preparation. Niosomes known to enhance the permeation and facilitate the drug transport across the skin. • Treatment of glaucoma : Ophthalmic preparations of a acetazolamide niosomal preparation results in Increased corneal permeability and increases drug delivery.
  • 35. Leishmaniasis therapy • Derivatives of antimony are most commonly prescribed drugs for the treatment of leishmaniasis. • These drugs in higher concentrations – can cause liver, cardiac and kidney damage. • Use of niosomes as a drug carrier showed that it is possible to overcome the side effects at higher concentration also and thus showed greater efficacy in Treatment.
  • 36. • Magnetic Niosomes acts as contrasting agent in MRI imaging. • Niosomes as immunological adjuvant
  • 37. List of Drugs formulated as Niosomes Intravenous route Doxorubicin, Methotrexate, Sodium Stibogluconate, Vincristine, Flurbiprofen, , Indomethacin, Colchicine,Rifampicin, Tretinoin, Transferrin and Glucose ligands, Zidovudine, Insulin,Cisplatin, Amarogentin, Daunorubicin, Amphotericin B, 5-Fluorouracil, Camptothecin Transdermal route Flurbiprofen, Piroxicam, Estradiol Levonorgestrol,Nimesulide,Dithranol, Ketoconazole, Enoxacin, Ketorolac Ocular route Timolol Maleate, Cyclopentolate Nasal route Sumatriptan, Influenza Viral Vaccine Inhalation All - trans retinoic acids
  • 38. Marketed formulations • Lancome has come out with a variety of anti-ageing products which are based on niosome formulations . Anti ageing cream consists of D-Contraxol it is said to reduce the depth of wrinkles by working on the neuro -transmitters responsible for muscle contraction
  • 40. INTRODUCTION: • Pharmacosomes are the colloidal dispersions of drugs covalently bound to lipids, and may exist as ultrafine vesicular, micellar, or hexagonal aggregates, depending on the chemical structure of drug-lipid complex. • Pharmacosomes are amphiphilic phospholipid complexes of drugs bearing active hydrogen that bind to phospholipids. • Pharmacosomes impart better biopharmaceutical properties to the drug, resulting in improved bioavailability.
  • 41. • Pharmacosomes have been prepared for various non-steroidal anti-inflammatory drugs, proteins, cardiovascular and antineoplastic drugs. • Developing the pharmacosomes of the drugs has been found to improve the absorption and minimize the gastrointestinal toxicity.
  • 42. IMPORTANCE: • Pharmacosomes have importance in escaping the tedious steps of removing the free unentrapped drug from the formulation. • Pharmacosomes provide an efficient method for delivery of drug directly to the site of infection, leading to reduction of drug toxicity with no adverse effects and also reduces the cost of therapy by improved bioavailability of medication, especially in case of poorly soluble drugs. • Pharmacosomes are suitable for incorporating both hydrophilic and lipophilic drugs. • Entrapment efficiency is not only high but predetermined, because drug itself in conjugation with lipids forms vesicles
  • 43. • There is no need of following the tedious, time-consuming step for removing the free, unentrapped drug from the formulation. • Since the drug is covalently linked, loss due to leakage of drug, does not take place. • No problem of drug incorporation • Encaptured volume and drug-bilayer interactions do not influence entrapment efficiency, in case of pharmacosomes
  • 44. • In pharmacosomes, membrane fluidity depends upon the phase transition temperature of the drug lipid complex, but it does not affect release rate since the drug is covalently bound. • The drug is released from pharmacosome by hydrolysis (including enzymatic). • The physicochemical stability of the pharmacosome depends upon the physicochemical properties of the drug-lipid complex. • Following absorption, their conversion into active drug molecule depends to a great extent on the size and functional groups of drug molecule, the chain length of the lipids, and the spacer.
  • 45. PREPARATION: • Two methods have been used to prepare vesicles: 1. The hand-shaking method 2. The ether-injection method • In the hand-shaking method, the dried film of the drug–lipid complex is deposited in a round-bottom flask and upon hydration with aqueous medium, readily gives a vesicular suspension. • In the ether-injection method, an organic solution of the drug–lipid complex is injected slowly into the hot aqueous medium, wherein the vesicles are readily formed.
  • 46. FORMULATION OF PHARMACOSOMES: • Drug salt was converted into the acid form to provide an active hydrogen site for complexation. • Drug acid was prepared by acidification of an aqueous solution of drug salt, extraction into chloroform, and subsequent recrystallization. • Drug -PC complex was prepared by associating drug acid with an equimolar concentration of PC.
  • 47. • The equimolar concentration of PC and drug acid were placed in a 100-mL round bottom flask and dissolved in dichloromethane. • The solvent was evaporated under vacuum at 40°C in a rotary vacuum evaporator. • The pharmacosomes were collected as the dried residue and placed in a vacuum desiccator overnight and then subjected to characterization.
  • 48. EVALUATION OF PHARMACOSOMES: • Solubility • Drug content • Differential scanning calorimetry • X-ray powder diffraction (XRPD)
  • 49. Solubility: • To determine the change in solubility due to complexation, solubility of drug acid and drug-PC complex was determined in pH 6.8 phosphate buffer and n-octanol by the shake-flask method. • Drug acid (50 mg) (and 50 mg equivalent in case of complex) was placed in a 100-mL conical flask. Phosphate buffer pH 6.8 (50 mL) was added and then stirred for 15 minutes. • The suspension was then transferred to a 250 mL separating funnel with 50 mL n-octanol and was shaken well for 30 minutes. • Then the separating funnel was kept still for about 30 minutes. Concentration of the drug was determined from the aqueous layer spectrophotometrically at 276 nm.
  • 50. Drug content: • To determine the drug content in pharmacosomes of drug (e.g.: diclofenac-PC complex), a complex equivalent to 50 mg diclofenac was weighed and added into a volumetric flask with 100 mL of pH 6.8 phosphate buffer. • Then the volumetric flask was stirred continuously for 24 h on a magnetic stirrer. At the end of 24 h, suitable dilutions were made and measured for the drug content at 276 nm UV spectrophotometrically.
  • 51. Differential scanning calorimetry (DSC): • Thermograms of drug acid, phosphatidylcholine (80 %) and the drug -PC complex were recorded using a 2910 Modulated Differential Scanning Calorimeter V4.4E (TA Instruments, USA). • The thermal behavior was studied by heating 2.0 ± 0.2 mg of each individual sample in a covered sample pan under nitrogen gas flow. The investigations were carried out over the temperature range 25– 250 °C at a heating rate of 10 °C min–1.
  • 52. X-ray powder diffraction (XRPD): • The crystalline state of drug in the different samples was evaluated using X-ray powder diffraction. Diffraction patterns were obtained on a Bruker Axs- D8 Discover Powder X-ray diffractometer, Germany. • The X-ray generator was operated at 40 kV tube voltages and 40 mA tube current, using lines of copper as the radiation source. The scanning angle ranged from 1 to 60° of 2q in the step scan mode (step width 0.4° min–1). • Drug acid, phosphatidylcholine 80 % (Lipoid S-80) and the prepared complex were analyzed.
  • 53. APPLICATIONS: • The approach has successfully improved the therapeutic performance of various drugs i.e. pindolol maleate, bupranolol hydrochloride, taxol, acyclovir, etc. • The phase transition temperature of pharmacosomes in the vesicular and Micellar state could have significant influence on their interaction with membranes. • Pharmacosomes can interact with biomembranes enabling a better transfer of active ingredient. This interaction leads to change in phase transition temperature of biomembranes thereby improving the membrane fluidity leading to enhance permeations.
  • 54. Conclusion • The concept of incorporating the drug into niosomes for a better targeting of the drug at appropriate tissue destination . • They presents a structure similar to liposome and hence they can represent alternative vesicular systems with respect to liposomes • Niosomes are thoughts to be better candidates drug delivery as compared to liposomes due to various factors like cost, stability etc. Various type of drug deliveries can be possible using niosomes like targeting, ophthalmic, topical, parentral, etc.
  • 55. • Vesicular systems have been realized as extremely useful carrier systems in various scientific domains. Over the years, vesicular systems have been investigated as a major drug delivery system, due to their flexibility to be tailored for varied desirable purposes. • In spite of certain drawbacks, the vesicular delivery systems still play an important role in the selective targeting, and the controlled delivery of various drugs. • Researchers all over the world continue to put in their efforts in improving the vesicular system by making them steady in nature, in order to prevent leaching of contents, oxidation, and their uptake by natural defense mechanisms.
  • 56. References: • S.P. Vyas R.K.Khar, Targeted And Controlled Drug Delivery novel carrier systems ; niosomes (p no.249-276) • N.K.JAIN , Controlled and Novel Drug Delivery; niosomes p no.(292-301) • H.A. Lieberman, M.M. Rieger, and G.S. Banker, Pharmaceutical Dosage Forms: Disperse Systems p. 163. • International Journal of Biopharmaceutics.2011;2(1):47-53; NIOSOMES FORMULATION AND EVALUATION
  • 57. • Review article of PHARMACOSOMES: A Potential vesicular drug delivery system BY de pintu kumar,de arnab • Review article of PHARMACOSOMES:opening new doors for drug delivery BY anu goyal