You are on page 1of 7

Journal of Drug Delivery Science and Technology 45 (2018) 408–414

Contents lists available at ScienceDirect

Journal of Drug Delivery Science and Technology


journal homepage: www.elsevier.com/locate/jddst

Controlled delivery of rivastigmine using transdermal patch for effective T


management of alzheimer's disease
Mayank Kumar Malaiya, Ashish Jain, Hurkat Pooja, Anki Jain, Dharmendra Jain∗
Department of Pharmaceutical Sciences, Dr. H. S. Gour Central University, Sagar, 470003, India

A R T I C LE I N FO A B S T R A C T

Keywords: Alzheimer's disease is a chronic neurodegenerative disease. Preparation of transdermal patches is a useful
Alzheimer's disease strategy for management of the disease. The transdermal patches bearing Rivastigmine tartarate were prepared
Rivastigmine using solvent casting method to provide good drug content uniformity. Transdermal patches were characterized
Transdermal patches for various attributions viz. patch weight, patch thickness, folding endurance, surface morphology study, X-Ray
Solvent casting method
diffraction study, Differential Scanning Calorimetry, tensile strength, surface pH, percent swelling, percent
Folding endurance
moisture sorption, drug content uniformity, In vitro release study, and ex-vivo study. Results of in vitro drug
release studies clearly indicate that the drug release governed polymer interaction. No lag time was observed
then reduction in drug release rate was observed. Extended drug release of 75.15 ± 1.43% within 24 h was
observed with formulation. The results of the permeation study indicated that the rate of drug permeation was
slow and it was found that 61 ± 1.43% of Rivastigmine tartrate could permeate through the skin membrane in
24 h. In the light of the above mentioned considerations, it is clear that the developed transdermal patch bearing
Rivastigmine tartrate was retained in skin layer and delivers the therapeutic amount of drug in blood for longer
time. This indicates the potential use of transdermal patches in the treatment of Alzheimer disease.

1. Introduction positron Emission Tomography (FDG-PET) measurements of decline in


the metabolic rate of glucose, PET measurements of brain Aβ by
Alzheimer's disease (AD) accounts for 60–70% of cases of dementia Pittsburgh Compound B (PIB) [23].
[4]. It is a chronic neurodegenerative disease that usually starts slowly Scientists do not yet fully understand what causes AD, but it is clear
and gets worse over time [8]. Alois Alzheimer recorded the first case of that it develops because of a complex series of events that take place in
Alzheimer's disease (AD) in 1906 [17]. The most common early the brain over a long period of time. It is likely that the causes include
symptom is complexity in remembering recent events (short term genetic, environmental, and lifestyle factors. There is no cure for
memory loss). As the disease advances, symptoms can include: pro- Alzheimer's disease; available treatments offer relatively small symp-
blems with language, disorientation (including easily getting lost), tomatic benefit but remain palliative in nature. Current treatments can
mood swings, loss of motivation, not managing self-care, and beha- be divided into pharmaceutical, psychosocial and care giving. Common
vioural issues [4]. As a person's condition declines they often withdraw medications are only symptomatic and aim to restore the disrupted
from family and society. Gradually, bodily functions are lost, ultimately neurotransmission between the degenerated neurons [16]. Current
leading to death [20]. The pathophysiological hallmarks of AD include therapies for AD include the Acetylcholinesterase inhibitors (Donepezil,
extracellular β-amyloid protein (Aβ) deposition in the forms of senile Galantamine and Rivastigmine), and a low-affinity NMDA (N-methyl-d-
plaques and intracellular deposits of the microtubule-associated protein aspartate) receptor antagonist (Memantine) for moderate to severe AD.
tau as neurofibrillary tangles in the brain. The acetylcholinesterase inhibitors mediate their effects by re-
Presently, a definitive diagnosis of AD is possible only after death, mediating, in part, the cholinergic deficit in AD, whereas the precise
when the presence of plaques and neurofibrillary tangles in the brain mechanism of action of memantine remains to be elucidated. Overall,
parenchyma can be revealed after pathological examination. The best the effects of these drugs are limited as they modestly improve some of
established measurements for the detection and tracking of AD include the symptoms but do not treat the underlying causes of the disease [13].
cerebrospinal fluid (CSF) measures of Aβ and tau, structural magnetic At present, the most common form of delivery of drugs is the oral route.
resonance imaging (MRI) of hippocampal atrophy, fluorodeoxyglucose While this has the notable advantage of easy administration, it also has


Corresponding author. Department of Pharmaceutical Sciences, Dr. H. S. Gour Central University, Sagar, 470003, India.
E-mail address: dharmjain@gmail.com (D. Jain).

https://doi.org/10.1016/j.jddst.2018.03.030
Received 2 August 2017; Received in revised form 17 November 2017; Accepted 25 March 2018
Available online 10 April 2018
1773-2247/ © 2018 Elsevier B.V. All rights reserved.
M.K. Malaiya et al. Journal of Drug Delivery Science and Technology 45 (2018) 408–414

significant drawbacks-namely poor bioavailability due to hepatic me- medication through the skin and into bloodstream. Transdermal patch
tabolism (first pass) and the tendency to produce rapid blood level bearing Rivastigmine tartarate were prepared by solvent casting
spikes (both high and low), leading to a need for high and/or frequent method.
dosing, which can be both cost prohibitive and inconvenient.
Reduction in the activity of the cholinergic neurons is a well-known 2.2.1. Preparation of backing membrane
feature of Alzheimer's disease [1]. Acetylcholinesterase inhibitors are The backing membrane was prepared using an aqueous solution of a
employed to reduce the rate at which acetylcholine (ACh) is broken 6% w/v of polyvinyl alcohol (PVA). Weighed amount of polyvinyl al-
down, thereby increasing the concentration of ACh in the brain and cohol (600 mg) was added to a 10 ml of warm distilled water and was
combating the loss of ACh caused by the death of cholinergic neurons constantly stirred on a magnetic stirrer at 60 °C for 5 min without for-
[2]. There is evidence for the efficacy of these medications in mild to mation of bubbles to attain a homogeneous solution. Then 3 ml of
moderate Alzheimer's disease, [14, 15] and some evidence for their use homogeneous solution was poured into a glass bangle placed on the
in the advanced stage. surface of aluminium foil in a Petridish. The rate of evaporation of the
The blood-brain barrier (BBB) is a common obstacle in treating CNS solvent was controlled by inverting the funnel over the Petridish. Glass
diseases. The BBB represents such an insurmountable obstacle for most spherical Petridish mold (1.4 cm depth, 5.2 cm internal diameter) to
drugs and contrast agents, an effective treatment or diagnosis of many one of the open end and circumference was wrapped by aluminum foil.
brain diseases is difficult or not possible. Therefore, a number of dif- Smooth uniform, transparent backing membrane was obtained after
ferent strategies such as osmotic opening of the tight junctions, direct keeping the mold at 60 °C for 6 h [5,6].
surgical administration of molecules into the brain, and the use of
prodrugs or carrier systems interacting with specific transporters and/
or receptors expressed at the luminal (blood) side of the endothelial 2.2.2. Formation of drug matrix over the backing membrane
cells employed during the past years to overcome this barrier [18,26]. Blend of polymers polyvinyl pyrolidone (PVP) and ethyl cellulose
Woo et al. [25] and Ravi & Gupta [22] also presented transdermal (EC) were measured in the requisite ratios of 1:2 and were dissolved in
patches as treatment option for Alzheimer's disease. To overcome the 5 ml of chloroform and a homogeneous solution was prepared using
limitations posed by BBB, transdermal patches can be used to deliver a magnetic stirrer. Then the plasticizer Dibutyl phthalate (DBP) and
specific dose of medication through the skin into bloodstream. Trans- Rivastigmine tartrate (100 mg) were added to the mixture and stirred
dermal delivery provides controlled, constant administration of the for 20 min until a homogeneous suspension was obtained. Then 5 ml of
drug, and allows continuous input of drugs with short biological half- the homogeneous suspension was casted on the prepared backing
lives and eliminates pulsed entry into systemic circulation [27]. membrane and dried at room temperature for 24 h, which yielded a
Nausea, vomiting and diarrhoea are much less common side effects uniform, flat medicated matrix patch of Rivastigmine tartrate [7,10].
with the patch than with the capsule and tablets. They provide ex- Formulation shows maximum water vapor transmission in 24 h i.e.
tended therapy with a single application, improving compliance over 4.76 × 10−3 g/cm2 and it shows extended drug release i.e. 36 h.
other dosage forms requiring more frequent dose administration. The
drug rivastigmine possess suitable characteristics for transdermal de- 3. Characterization of transdermal patches
livery such as low daily doses (1.5–6 mg twice daily), short half-life
(1.5 h), low molecular weight (400.43 gms). The rivastigmine patch 3.1. Patch weight
provides a viable treatment option for patients with mild to moderate
AD; it has an improved tolerability profile compared with rivastigmine For evaluation of patch weight three patches of each formulation
capsule doses providing comparable drug exposure and efficacy. This were taken and weighed individually and average weight was calcu-
has been mainly attributed to the favourable properties of lack of first lated.
pass metabolism effects of liver, better patient compliance, steady re-
lease profile and lowered pill burden in transdermal system. 3.2. Patch thickness
Therefore, it is envisaged that transdermal patches bearing
Rivastigmine can overcome most of demerits of conventional delivery For evaluation of patch thickness, 3 patches of formulation were
and will be able to release the drug at predetermined rate into systemic taken and the patch thickness was measured using screw gauge of three
circulation for a prolong period of time, thereby minimizing its adverse different places and the mean thickness of patches was calculated.
effect and improving patient compliance.
The main components to a transdermal patch are polymer matrix,
drug, permeation enhancers, adhesive, backing laminates, release liner, 3.3. Surface morphology (SEM)
other excipients like plasticizers and solvents [28].
Scanning Electron Microscope (SEM) was used as visualizing aid for
2. Materials and methods surface morphology. The patches were mounted on an aluminium
sample support by means of a conductive and double sided adhesive
2.1. Materials tape. The morphologies of the patch surfaces analyzed using a Philips
XL30 TMP SEM (LEO 435 UP, Eindhoven, The Netherlands) at an ac-
The drug Rivastigmine tartrate was obtained as a gift sample from celeration voltage of 30 kV, the photomicrographs were taken at sui-
Cipla limited, Mumbai India. Polyvinyl pyrolidone and Ethyl cellulose table magnification. The images were taken without the use of a metal
(Central Drug House, Ltd), polyvinyl alcohol and Chloroform (Himedia coating. Photomicrographs (Fig. 1) shows the SEM image of the patch.
Laboratories), Dibutyl phthalate (Ranbaxy Laboratories Limited) was
obtained from Drug Delivery Research Laboratory, Department of 3.4. X- ray diffraction pattern (X-RD study)
Pharmaceutical Sciences, Sagar. Other chemicals, solvents and reagents
were of analytical grade. X-ray diffraction study was carried on pure drug and transdermal
patch containing drug using the XRD technique (model X'Pert-the
2.2. Methods Netherlands). XRD study was performed on the samples by expose them
to Cu K-α-1 radiation (45 kV, 40 mA) and scanned from 2 to 50° 2θ, at a
A transdermal patch used for targeting a drug to a particular region step size of 0.0170 2θ and a step time of 20.0271 s. Fig. 2 shows the
of body for extended period of time is used to deliver a specific dose of XRD pattern of pure drug and transdermal patch.

409
M.K. Malaiya et al. Journal of Drug Delivery Science and Technology 45 (2018) 408–414

Fig. 1. (A) SEM images of transdermal film, (B) SEM image of after transdermal film after dissolution of 4 h.

3.5. Differential scanning calorimetry the patch could be folded at the same place without breaking/cracking
gave the value of folding endurance [9].
The physicochemical compatibility between Rivastigmine and
polymers used in the patches was studied using differential scanning
calorimetry (Jupiter STA 449 F1 Nersch). In DSC analysis, the samples 3.7. Tensile strength
were weighed (2 mg), hermetically sealed in flat-bottom aluminum
pans, and heated over a temperature range of 50–250 °C (in case of pure A tensile strength study for patch is total weight necessary to break
drug) and 50–150 °C (in case of patch) in an atmosphere of nitrogen or rupture the patch and this was done by a device has rectangular
(50 mL/min) at a constant increasing rate of 10 °C/min. The frame with two plates made up of Plexiglas's. The one plate is in front
Thermograms (Fig. 3) obtained for Rivastigmine and transdermal patch and is movable part of device and can be pulled by loading weights on
and compared. the string, which is connected to movable part, the 2 × 1 cm2 plate. The
force needed to fracture the patch was determined by measuring the
total weight loaded in the string. The weight loaded corresponds to
3.6. Folding endurance break the patches were taken as tensile strength and the values are
given in Table 1. Following formula was used to calculate the me-
The folding endurance is important to check the ability of sample to chanical properties of the patches.
withstand folding was measured manually for the prepared patches. It
is expressed as number of times the patch is folded at the same place Force of break(g)
Tensile Strength =
either to break the patch or to develop visible cracks. This also gives an Initial cross sectional of the sample(cm2)
indication of brittleness [21]. This was determined by repeatedly
folding one patch at the same place till it break. The number of times

Fig. 2. X ray diffraction pattern of (A) Pure drug and (B) Drug loaded film.

410
M.K. Malaiya et al. Journal of Drug Delivery Science and Technology 45 (2018) 408–414

Fig. 3. (A): Differential scanning calorimetry of pure drug Rivastigmine. (B) Differential scanning calorimetry of transdermal film.

411
M.K. Malaiya et al. Journal of Drug Delivery Science and Technology 45 (2018) 408–414

Table 1
Evaluation of transdermal patches. 6 Percent moisture sorpƟon
Parameters Observation 5

Percent moisture sorpƟon


4
Film Weight (mg) 35.23 ± 0.19
Thickness (mm) 00.38 ± 0.01 3
Folding Endurance 276 ± 08 2
Tensile strength (gcm−2) 287 ± 1.8
Surface pH 5.4 ± 0.08 1
Drug content uniformity (%) 95.87 ± 2.3 0
1 2 3
N = 3, Mean ± SD. Time (In days)

3.8. Surface pH Fig. 5. Percent moisture sorption of different transdermal patches (n = 3).

Normally skin pH is around 5.8. Surface pH of the patches was


determined by the method described by Ref. [3]. The patches were 3.11. Drug content uniformity
allowed to swell by keeping them in contact with phosphate buffer
(pH5.8) for 2 h in glass tubes at room temperature. The surface pH was From the prepared drug contained patches specified surface area
then noted by bringing a combined glass electrode near the surface of (1 cm2) were cut and dissolved in 100 ml of pH 7.4 phosphate buffer,
the patch and allowing it to equilibrate for 1 min. and vigorously shaken for 2hrs, and then centrifuged at 5000 rpm for
30 min. Then the drug contained polymeric solution was filtered
through 42 number whatman filter paper, and drug content was de-
3.9. Swelling
termined at ƛmax 264 nm using double beam UV–Visible spectro-
photometer (Shimadzu 1601, Japan). Respected Placebo patch was
Three patches of formulation was allowed to swell on the surface of
taken as blank solution preparation and results in reported in Table 1.
agar plate kept in an incubator at 37 ± 0.2ᵒC. Increase in weight of the
patch (n = 3) was determined at every hour upto 6 h. The percent
swelling was calculated using the following equation: 3.12. In vitro drug release study

Wt − Wo In vitro drug release study is a prerequisite for evaluating the In vivo


Percent Swelling =
Wo performance of a drug delivery system because the in vitro drug release
where, Wt is the weight of the swollen patch after time t, and Wo is the profile provides the most sensitive and reliable information for in vivo
initial patch weight at zero time (The mean values of three readings are evaluation that helps in ascertaining the future behaviour of the de-
calculated and reported in Fig. 4). signed formulation with regards to its drug release pattern and the time
duration of its action in a biological system.
3.10. Percent moisture sorption The in vitro release was carried out with the dialysis membrane
using Franz diffusion cell (Fig. 7). The cell consists of two chambers, the
Three patches (1 × 1cm size) were dried in the oven at 30 ± 2ᵒC. donor and the receptor compartment. The donor compartment was
After drying the weight of each patch was measured. The patches were open at the top and exposed to the atmosphere. The temperature was
successively transferred to desiccators over saturated salt solution of maintained at 37 ± 0.5 °C and receptor compartment was provided
sodium nitrite (75 ± 5% RH) at 25ᵒC. After each 1, 3, 5 days, the with sampling port. The diffusion medium used was PBS pH 7.4 solu-
conditioned patches were weighed and noted the weight and placed tion. The drug containing patch with a support of backing membrane
back to desiccators. Percentage moisture sorption was calculated using was kept in the donor compartment and it was separated from the re-
following formula: ceptor compartment by dialysis membrane. The dialysis membrane was
previously soaked for 24 h in PBS (pH 7.4). The donor and receptor
we − wc
Percentage moisture sorption = ×100 compartments were hold together using clamp. The receptor compart-
wc ment with 15 ml of PBS pH 7.4 was maintained at 37 ± 0.5 °C and
where, We = Weight of exposed patch, and Wc = Weight of conditioned stirred on magnetic stirrer, to prevent the formation of concentrated
patch. drug solution layer below the dialysis membrane. The release study was
The mean values of three readings are calculated and reported in performed for 24 h. After every 1 h, 1 ml samples were withdrawn from
Fig. 5. receptor compartment and immediately replaced with fresh media. The
withdrawn samples were filtered; 1 ml of the filtrate was diluted to
10 ml using PBS (pH 7.4). The samples were analyzed spectro-
Time Percent Swelling
photometrically at 264 nm. In vitro percentage cumulative release of
80 drug from transdermal patch formulation is shown in Fig. 6.
70
Percent swelling

60 3.13. Ex- vivo drug permeation studies


50
40 3.13.1. Preparation of excised goat skin
30 Goat skin was procured from the local slaughter house (Sagar). The
20 pouch was washed and rinsed with the normal saline to remove any
10 looser material from a pouch. A thin layer of mucosa of about 30 mm
0 diameter was stripped with the help of forceps. The mucosa was flat-
1 2 3 4 5 6
tened, kept in normal saline at refrigerated temperature overnight.
Time (in Hours)

3.13.2. Modified diffusion cell


Fig. 4. Percent swelling of transdermal patch (n = 3). For exvivo drug permeation study, a modified diffusion cell was

412
M.K. Malaiya et al. Journal of Drug Delivery Science and Technology 45 (2018) 408–414

Fig. 8. Ex Vivo drug permeation profile of transdermal patch through goat


membrane (n = 3).

Fig. 6. In vitro release profile of rivastigmine tartarate from transdermal patch.


transmission in 24 h.
These transdermal patches were characterized for various attribu-
tions viz. patch weight, patch thickness, folding endurance, surface
morphology study, X-Ray diffraction study, Differential Scanning
Calorimetry (DSC), tensile strength, surface pH, percent swelling, per-
cent moisture sorption, drug content uniformity, In vitro release study,
and ex-vivo study.
The patch weight and thickness was found to be 35.23 ± 0.19 mg
and 00.38 ± 0.01 mm, respectively. The folding endurance measures
the ability of patch to withstand rupture. It was found to be 276 ± 08.
The folding endurance values of patches were found to be optimum and
therefore the patches exhibited good physical and mechanical proper-
ties. The result indicated that the patches would not break and maintain
their integrity with general skin folding when applied.
In order to examine the surface morphology, formulations were
examined under Scanning Electron Microscope (SEM). It was found that
the surface morphology of patch was smooth surface as shown by
Fig. 7. Franz diffusion cell. photomicrographs (Fig. 1A). On keeping the patches in the dissolution
medium the pore were formed that are clearly seen in the SEM pho-
used. It considered of an open-ended glass cylinder of approximate 4 cm tograph of the patch after 240 min (Fig. 1B). The pore formation on
diameter and a beaker filled with 100 ml PBS (pH 7.4). The excised patch surface may be due to the dissolution of the hydrophilic polymer
mucosa was tied delicately on to one end of the glass cylinder. The of patch.
cylinder acts as donor compartment while the beaker act as a receptor The X-ray diffraction gives us about the crystalline and amorphous
compartment. The glass cylinder was fixed vertically in the glass beaker nature of the drug within the transdermal patches. The sharp peaks
in such a way that the mucus membrane just baths the content of the present in diffractogram indicates crystalline of the compound which
beaker. This assembly set up was placed over a magnetic stirrer with a were absent in case of amorphous compounds. In case of Fig. 2 both
teflon bead inside the receptor compartment for constant stirring the (pure drug and transdermal patch) have the sharp peak so it indicates
content of media and temperature of 37 ± 0.5ᵒ C was maintained that the drug is in crystalline state in the patch.
throughout the experiment. DSC studies were carried out for Rivastigmine tartarate pure drug
The ex-vivo drug permeation study of transdermal patch through and its physical mixture form of transdermal patch. The DSC thermo-
excised goat buccal mucosa was studied using the modified diffusion gram (Fig. 3A and B) of drug showed an exothermic peak at 107.19 °C
cell. A 1 × 1 cm size patch was placed intimate contact with the goat corresponding to its melting temperature, which was absent in the
buccal mucosa. The sample were withdrawn at every hour, filtered, thermograms of physical mixture, signifying no interaction between
diluted suitably and then absorbance were taken. Ex-vivo percent cu- drug and the polymers.
mulate drugs permeated from formulation (Fig. 8). The tensile strength was determined to evaluate mechanical
strength to the patches and found to be 287 ± 1.8. Appropriate swel-
ling behaviour of transdermal patches is an essential property for uni-
4. Results and discussion form and prolonged release of the drug and for effective skin adhesion.
The study of the hydration of polymers used in sustained release ap-
Solvent casting method was selected for the preparation because of plications has been an area of interest because it is believed that it af-
easy availability of the processing requirements in the laboratory, fects drug release from controlled release matrix. The consequence of
economical scale up and good drug content uniformity. water uptake could be the formation of empty spaces within the patch
Ethyl cellulose (EC), polyvinyl pyrolidone (PVP) and polyvinyl al- that could make its structure less resistant to mechanical stresses. It
cohol (PVA) were selected for the preparation of rivastigmine trans- varied from 35.13 ± 1.09 to 68.13 ± 2.43 after 1 h–6 h. The swelling
dermal patch because of Compatible with drug, economic, nonirritating ability may vary with nature and composition of patches. Hydrophilic
to skin and good patch forming capacity. Dibutyl phthalate was selected polymer showed considerable swelling, as it increased the surface
as the plasticizer in the present study as it is economic with good wettability and consequently water penetration within the matrix.
elasticity to make the patches. It also provides adhesiveness to the The surface pH of the patch was found to be 5.5 ± 0.08.
patches. Considering the fact that acidic or alkaline pH may cause irritation to
Formulation showed4.76 × 10−3 g/cm2 maximum water vapor the skin mucous membrane and influence the degree of hydration of

413
M.K. Malaiya et al. Journal of Drug Delivery Science and Technology 45 (2018) 408–414

polymers, the surface pH of the transdermal patch was determined to Conflicts of interest
optimize the drug permeation and skin adhesion. Attempts were made
to keep the surface pH close to skin pH as possible. The surface pH of None.
formulations was within the range of skin pH. Hence the patches will
not cause any irritation to the skin after its application. The surface pH Acknowledgement
of formulations was determined in order to investigate the possibility of
any side effects. Authors acknowledge Cipla Ltd Mumbai for providing Rivastigmine
Drug content was found to be 95.87 ± 2.3% of Rivastigmine tar- and All India Council of Technical Education (AICTE) for providing
trate for formulations. On this basis it was found that the drug dispersed financial assistance (JRF) to one of the authors.
uniformly throughout the patch.
The drug release profile of rivastigmine tartrate from formulation is Appendix A. Supplementary data
shown in Fig. 6. Results of drug release study clearly indicate that the
drug release governed polymer interaction. No lag time was observed as Supplementary data related to this article can be found at http://dx.
the patch was directly exposed to the dissolution medium. Slow drug doi.org/10.1016/j.jddst.2018.03.030.
release rate was observed as the presence of ethyl cellulose in the for-
mulation. This may be due to the hydrophobic nature of the polymer, References
which reduces the penetration of the dissolution medium into the
patches and retarded the drug release. Extended drug release [1] Y. Hirashima, The roles of platelet-activating factor (PAF) and its related signaling
and metabolism in a, in: A.A. Farooqui, T. Farooqui (Eds.), Neurological Diseases in
75.15 ± 1.43% within 24 h was observed with formulation.
Molecular Aspects of Neurodegeneration and Neuroprotectione, 2011, pp. 90–101
The results of the permeation study indicated that the rate of drug ISBN: 978-1-60805-092-5.
permeation was slow and it was found that 61 ± 1.43% of [2] J. Birks, Cholinesterase inhibitors for Alzheimer's disease, Cochrane Database Syst.
Rev. 1 (2006) CD005593.
Rivastigmine tartrate could permeate through the skin membrane in [3] P. Bottenberg, R. Cleymact, C.D. Muynck, et al., Development and testing of
24 h. bioadhesive fluoride containing slow release tablets for oral use, J. Pharm.
Pharmacol. 43 (1991) 457–464.
[4] A. Burns, S. Iliffe, Alzheimer's disease, BMJ (Clinical research ed.) 338 (2009) b158.
[5] Y.W. Chien, Novel Drug Delivery Systems, Drugs and the Pharmaceutical Sciences,
5. Conclusions vol. 50, Marcel Dekker, New York, NY, 1992797.
[6] K.P.R. Choudhary, R.A.S. Naidu, Studies on permeability of ethyl cellulose films for
transdermal use, East. Pharm. 34 (405) (1991) 119–121.
Alzheimer's disease (AD) is one of the most common neurodegen- [7] Nicholas A. Boon, Nicki R. Colledge, Brian R. Walker (Eds.), Davidson’s Principles
erative disorders, comprising 50–70% of all reported cases of dementia and Practice of Medicine 20th Edition, 1999 International Editor John A.A. Hunter
pg 1217, 1188, 1189.
[12] and affecting the middle to old-aged individuals [11]. The [8] Dementia Fact sheet N°362" (http://www.who.int/mediacentre/factsheets/fs362/
blood–brain barrier (BBB) is a highly regulated interface that separates en/). Who.int. April 2012. Retrieved 28 November 2014.
the peripheral circulation and central nervous system (CNS). It is a [9] V.K. Devi, S. Saisivam, G.R. Maria, P.U. Deepti, Design and evaluation of matrix
diffusion controlled transdermal patches of Verapamil hydrochloride, Drug Dev.
dynamic and functional neurovascular unit. The tight junctions be- Ind. Pharm. 29 (5) (2003) 495–503.
tween the endothelial cells serve to restrict blood-borne substances [10] K. Devi, K.L.K. Paranjothy, Pharmacokinetic profile of a new matrix type trans-
dermal delivery system. Diclofenacdiethylammonium patch, Drug Dev. Ind. Pharm.
from entering the brain. Nanotechnology based strategies have gained 25 (1999) 695–700.
tremendous importance as some of them are capable of overcoming the [11] Y. Feng, X. Wang, Antioxidant therapies for Alzheimer's disease, Oxid. Med. Cell.
limitations inherent to BBB passage. These include various types of li- Longev. 472932 (2012) 2012.
[12] J. Gotz, A. Eckert, M. Matamales, L.M. Ittner, X. Liu, Modes of A β toxicity in
pidic, polymeric, inorganic and other types of nanoparticles (NPs) for Alzheimer's disease, Cell. Mol. Life Sci. 68 (20) (2011) 3359–3375.
controlled drug delivery and release pertinent to various CNS condi- [13] E. Karran, M. Mercken, B. De Strooper, The amyloid cascade hypothesis for
Alzheimer's disease: an appraisal for the development of therapeutics, Nat. Rev.
tions [19,24]. Today about 74% of drugs are taken orally and are found
Drug Discov. 10 (9) (2011) 698–712.
not to be as effective as desired. To improve such characters trans- [14] S.A. Lipton, Paradigm shift in neuroprotection by NMDA receptor blockade:
dermal drug delivery system was emerged. Drug delivery through the memantine and beyond, Nat. Rev. Drug Discov. 5 (2) (2006) 160–170.
[15] Memantine"(http://www.nlm.nih.gov/medlineplus/druginfo/meds/a604006.
skin to achieve a systemic effect of a drug is commonly known as html). US National Library ofMedicine (Medline). 4 January 2004. Accessed 3
transdermal drug delivery and differs from traditional topical drug February 2010.
delivery. Transdermal drug delivery systems (TDDS) are dosage forms [16] A. Nazem, G.A. Mansoori, Nanotechnology solutions for Alzheimer's disease: ad-
vances in research tools, diagnostic methods and therapeutic agents, J. Alzheimers
which involves drug transport to viable epidermal and or dermal tissues Dis. 13 (2) (2008) 199–223.
of the skin for local therapeutic effect while a very major fraction of [17] T.T. Nguyen, V.V. Giau, T.K. Vo, Current advances in transdermal delivery of drugs
for Alzheimer's disease, Indian J. Pharmacol. 49 (2) (2017) 145–154.
drug is transported into the systemic blood circulation. The adhesive [18] W.M. Pardridge, Brain drug development and brain drug targeting, Pharm. Res. 24
character of the transdermal drug delivery system is critical to the (9) (2007) 1729–1732.
safety, efficacy and quality of the product. Topical administration of [19] M.M. Patel, B.R. Goyal, S.V. Bhadada, J.S. Bhatt, A.F. Amin, Getting into the brain:
approaches to enhance brain drug delivery, CNS Drugs 23 (1) (2009) 35–58.
therapeutic agents offers many advantages over conventional oral and [20] H.W. Querfurth, F.M. LaFerla, Alzheimer's disease, N. Engl. J. Med. 362 (4) (2010)
invasive methods of drug delivery. Several important advantages of 329–344.
transdermal drug delivery are limitation of hepatic first pass metabo- [21] S. Raghuraman, R. Velrajan, B. Ravi, D. Jeyabalan, J. Benito, V. Sankar, Design and
evaluation of Propranolol hydrochloride buccal films, Indian J. Pharm. Sci. 64 (1)
lism, enhancement of therapeutic efficacy and maintenance of steady (2002) 32–36.
plasma level of the drug. It is expected that this approach will improve [22] G. Ravi, N.V. Gupta, The treatment of alzheimers disease by using donopezil loaded
transdermal patch, J. Chem. Pharmaceut. Res. 7 (3) (2015) 806–813.
management of drug therapy in alzheimer's patients, minimize the drug [23] E.M. Reiman, W.J. Jagust, Brain imaging in the study of Alzheimer's disease,
toxicity and improve patient's compliance by delivering the drug at Neuroimage 61 (2) (2012) 505–516.
controlled rate for prolonged period of time at a desired site. It is [24] H.L. Wong, X.Y. Wu, R. Bendayan, Nanotechnological advances for the delivery of
CNS therapeutics, Adv. Drug Deliv. Rev. 64 (7) (2012) 686–700.
concluded that transdermal patch bearing Rivastigmine tartarate was [25] F.Y. Woo, M. Basri, H.R. Masoumi, et al., Formulation optimization of galantami-
able to deliver the drug at a controlled rate for an extended period of nehydrobromide loaded gel drug reservoirs in transdermal patch for Alzheimer's
disease, Int. J. Nanomed. 10 (2015) 3879–3886.
time. The proposed system expected to be popular substituted of the
[26] D.J. Begley, Delivery of therapeutic agents to the central nervous system: the pro-
tablets and capsules. Newly developed system would likely to overcome blems and possibilities, Pharmacol. Ther. 104 (2004) 29–45.
all the drawbacks of the presently available therapy of Rivastigmine. [27] P.K. Gour, S. Mishra, S. Purohit, K. Dave, Transdermal drug delivery system- A
review, Asian J. Pharmaceut. Clin. Res. 2 (2009) 14–20.
Thus, developed system can be considered as an effective and con- [28] G. Aggarwal, S. Dhawan, Development, fabrication and evaluation of transdermal
venience drug delivery system for treatment of Alzheimer disease. drug deliverey system - A Review, Pharmainfo.net. 7 (5) (2009).

414

You might also like