Development, Characterization and Evaluation of Ritonavir Nanosuspension Treatment of Antiretroviral Therapy

 

Shital V. Sonawane*, Avish D. Maru, Mitesh P. Sonawane

Loknete Dr. J.D. Pawar College of Pharmacy, Manur, Tal-Kalwan-423501, Dist-Nashik, (MH) India.

*Corresponding Author E-mail: sonawaneshital97@gmail.com

 

ABSTRACT:

Oral nanosuspension of ritonavir was prepared by antisolvent precipitation method using various polymers such as Eudragit RS100, Poloxamer 407, SLS and Methanol.The effect of eudragit RS100 and poloxamer 407 used stabilizer and SLS is surfactant was investigated on particle size and distribution, drug content, entrapment efficiency was observed. Ritonavir is having low solubility and low permeability drug belonging to class-IV according to BCS. Drug-excipient compatibility and amorphous nature of ritonavir drug is prepared nanosuspension was confirmed by FTIR, DSC and Motic microscope studies, respectively. The nanosuspension was further evaluated for drug content, saturation solubility study and entrapment efficiency. The average particle size of ritonavir nanaosuspensions formulas was observed from 0.006 µm to 0.017 µm. The studied in the solubility and dissolution rate there are the increase solubility and dissolution rate of ritonavir nanosuspension.

 

KEYWORDS: Antiretroviral therapy, Antisolvent nanoprecipitation method, Nanoparticle, Solubility enhancement techniques, Molecular level.

 

 


INTRODUCTION:

A nanosuspension is a submicron dispersion of nanosized drug particle which are stabilized by surfactants and polymers. A pharmaceutical nanosuspension is defined as very finely dispersed solid drug particles in an aqueous vehicle for oral, topical, parenteral or pulmonary administration. The particle size distribution of the solid particles in nanosuspensions is usually less than one micron with an average particle size ranging between 200 and 600 nm 1,2.

 

A nanosuspension not only solves the problems like poor solubility and bioavailability but also improve the safety and efficacy of the drug by altering the pharmacokinetics properties of the drug. Nanosuspensions differ from nanoparticles. Oral route is commonly used and most convenient route for the drug delivery. Oral drug delivery system gained more attention in the pharmaceutical field, due to its more flexibility in designing the dosages from than the other drug delivery systems. In recent years novel drug delivery systems like nanosuspension draws a considerable attention in search for improving bioavailability of poorly soluble drugs 3,4.

 

Approximately 40% drug candidate currently available for therapeutic purpose are hydrophobic in nature which affects the bioavailability of such type drug molecules. Many techniques are employed to overcomes is problem, out of which most important are size reduction to nano-size (Formation of Nanocrystals), pH adjustment, use of salt and co-solvents, surfactant, and preparation of lipid formulation 5,6.

 

Crystals are defined as presence of high degree of order with a three dimensional periodicity in position of atoms or ions in molecule, with configurationally periodicity of molecules that form the crystal. All chemical entities including drug molecules are able to possess different crystalline forms called as polymorphic form, which have same molecular formula, but arranged in different configuration and showing different physical            properties7,8. Specific polymorphic forms are often considered as important quality-relevant attributes of a drug substance. Polymorphic forms exhibit different dissolution rate, solubility, flow properties, hygroscopicity and behavior under mechanical stress encountered during milling, tableting, agglomeration, etc. Amorphous molecules shows molecular level disorder in configuration, having high internal energy (Chemical potential) i.e. thermodynamically unstable9,10. These compounds possess higher solubility in aqueous phase as compared to crystalline compounds which possess low internal energy i.e. highly thermodynamically stable with lower solubility. Various techniques are used to enhance the solubility of crystalline drugs which include structural modification by physical and chemical method and other methods like particle size reduction (Micronization/Nanonization), crystal engineering and solid dispersion, use of surfactant, salt formation, and complexation 11,12.

 

The drugs classified under BCS class II and IV reported to have problems in their solubility, stability and compatibility which affects their bioavailability and formulation aspects, which can be overcome by making nanocrystals, nanosuspensions during their formulations. This can be used to formulate different dosages forms such as tablets, capsules, parenteral, ophthalmic and nasal preparations 13.

 

Selection Criteria of Drug for Nanosuspension14, 15:

Nanosuspension can be prepared for the API that is having either of following characteristics:

·      Water insoluble but which are soluble in oil (high log P) OR API is insoluble in both water and oils.

·      Drugs with a reduced tendency of the crystals to dissolve, regardless of the solvent

·      API with a very large dose.

 

ANTIRETROVIRAL THERAPY:

Before people start antiretroviral therapy (ART), health-care provides should initiate a detailed discussion about the redliness of patients to initiate ART, the antiretroviral (ARV) drug regimen, dosages, scheduling, likely benefits, possible adverse effects and the required follow-up and monitoring visits In the case of children with HIV, this conversation should directly involve the caregiver and includes discussion about disclosing their HIV status [16] [17]. Retesting all people living with HIV before initiating ART is recommended to ensure a correct diagnosis of HIV infection. Initiation of ART should always consider nutritional status, any comorbidity and other medications being taken to assess for possible interactions, contraindications and dose adjustment.

 

HIV/AIDS has always been one of the most thoroughly global of diseases. The human immunodeficiency virus (HIV) is a lent virus that causes HIV infection and AIDS.

 

HIV stands for human immunodeficiency virus. AIDS stands for acquired immune deficiency syndrome.

 

Protease inhibitors used in the treatment of AIDS found to influence the glycoprotein synthesis independently in turn inhibits the growth of HIV. Ritonavir one of the potential protease inhibitor could also act as a substrate for efflux pump that is ultimately preventing the elimination of other co-administered antiretroviral drugs18.

 

The purpose of the present study to develop a nanosuspension of ritonavir by the anti-solvent precipitation method with improved the solubility and oral bioavailability. The effect of eudragit RS100, poloxamer 407 (stabilizer) and SLS (surfactant) contents was also studied on the quality attributes of the prepared nanosuspension such as drug content, particle size, entrapment efficiency and stability study 19.

 

MATERIALS AND METHODS:

Materials:

Ritonavir was obtained from Mylan Laboratories Limited, Sinnar, Nashik, Poloxamer 407, Eudragit RS100 obtained from Balaji Drug, Gujarat, SLS (Sodium Lauryl Sulphate) and Methanol was obtained from Research-Lab, Mumbai.

 

Preparation of Nanosuspension method:

The nanoprecipitation method is used for the preparation of ritonavir Nanosuspension. This method is also called as solvent displacement method or anti-solvent precipitation method 20.

 

The most common method of precipitation used in anti-solvent addition method in which drug is dissolved in suitable organic solvent and this solution mixed with a miscible antisolvent. Rapid addition of drug solution in to the anti-solvent leads to the sudden supersaturation of drug in the mixed solution forms ultrafine drug solids. Precipitation method involves two phases-nuclei formation and crystal growth. When preparing a stable suspension with the minimum particle size, a high nucleation rate and but low growth rate is necessary. Both rates are depending on temperature. In this technique the drug needs to be soluble in at least one solvent which is miscible with non-solvent.

 

Experimental Work:

1) Preformulation Studies:

1. Solubility studies:

The solubility of the ritonavir was determined by dissolving excess amount of drug in the solvents (water, methanol and ethanol).

 

2. Physical constant (melting point) determination:

Melting point of ritonavir was measured with the use of Thieles tube apparatus by using paraffin oil, thermometer, thread and burner. The capillary is tight with thermometer and placed in Thieles tube containing paraffin oil, the tube is heated by using burner. The range of temperature when drug just start melting and till it completely melts was noted. Observed values were compared with the reported value.

 

3. In-vitro drug determination by UV Spectrophotometer:

 

1. Construction of calibration curve in methanol:

A stock solution of concentration 1000 µg/ml of drug in methanol was prepared separately by dissolving accurately weighed 10 mg of ritonavir 10 ml methanol from this solutions take 1 ml and diluted up to 10 ml with corresponding solvent (100ppm). Appropriate volumes solution were further diluted to obtained final concentrations in range of 10-100 µg/ml. the spectrum of this solution was recorded using UV-visible spectrophotometer 21.

 

4. Drug–excipient compatibility studies:

The drug excipient interaction study was carried out by using physical observation.

Physical observation: the mixture of a drug and each excipients (1:1) ratio placed in separate vials with rubber closure, in order to make hermetically sealed and kept in the environmental test chamber. (Stability chamber, 40℃ at 75 % RH) for 15 days. During the storage, samples were observed for changes if any, such as caking, liquefaction gas formation, and colour change, odour change etc22.

 

2) Formulation batches of nanosuspensions:

Nine formulation batches were prepared and evaluated for the selection of Best Batch. After the formulation of nanosuspension it was characterized for particle size, Total drug content, entrapment efficiency, and saturation solubility. The low, high and medium concentration of stabilizers selected for the formulation of nanosuspension the final batch subjected to the stability study. The preformulation design of 32 full factorial batches 1 to 9 depicted in table 1.

 

3. Procedure for Formulation Development of nanosuspensions:

The nanoprecipitation technique is used for the preparation of ritonavir nanosuspension. The pure drug of ritonavir (100mg) was dissolve in (10ml) of methanol to produced organic phase. (Solution 1) The Stabilizers was dissolved in water (40ml) to form aqueous phase. (Solution 2) The solution 1 added into solution 2 drop wised with the help of syringe under the magnetic stirrer for 15 min. After the magnetic stirring the formulation is subjected to agitation with the help of lab stirrer (REMI) at 2000 rpm at 2 hr. 23.

 

·      Evaluation of the Prepared Nanosuspension:

1. Particle size analysis:

The average particle size of all NS formulations was determined by the motic digital microscopy (DMWB1-223). The nanosuspension showing the lowest particle size was selected for further studies. The particle size of the formulated batches was measured in micrometers.

 

2. Total drug content:

Prepared Nanosuspensions was analyzed for drug content by UV spectroscopic method. Different batches of nanosuspensions equivalent to 10 mg of drug ritonavir weighed accurately and diluted up to 100 ml with methanol. Stock solutions will be diluted with methanol and analyse by UV spectroscopy (LABINDIA 3000+) at 234 nm 24.


 

Table 1: Batches of Design-Experts for Formulation of for Formulation of Ritonavir Nanosuspension (F1-F9).

 

Ingredients

 Formulation Code

 F1

 F2

 F3

 F4

 F5

 F6

 F7

 F8

 F9

 

Ritonavir (mg)

 100

 100

 100

 100

 100

 100

 100

 100

 100

 

Eudragit RS100 (%)

 200

 300

 300

 400

 300

 200

 400

 200

 400

 

Poloxamer 407 (%)

 400

 400

 500

 600

 600

 500

 400

 600

 600

 

 SLS (mg)

 10

 10

 10

 10

 10

 10

 10

 10

 10

 

Methanol (ml)

 10

 10

 10

 10

 10

 10

 10

 10

 10

 

Water (ml)

 30

 30

 30

 30

 30

 30

 30

 30

 30

 

Stirring Speed(RPM)

 2000

 2000

 2000

 2000

 2000

 2000

 2000

 2000

 2000

 

 


 

3. Entrapment efficiency:

The method suitable for determining entrapment efficiency of nanosuspension when fairly high concentration of free drug is present in the supernatant after centrifugation (REMI 12C). 10 ml portion of the freshly prepared nanosuspension was centrifuged at 1000 rpm for 10 min. using centrifuge the supernatant was removed and the amount of incorporated drug was measured by taking the absorbance of supernatant solution at 234 nm by using UV spectrophotometer. (LABINDIA 3000+).

 

Entrapment efficiency was calculated by following formula:

Entrapment Eficiency (%)

= (Winitial drug – W Free drug / × 100)

 

4. Saturation solubility study:

The saturation solubility of various orally accepted stabilizers was carried out by shake flask method. The solubility of ritonavir in powder form was determined by a shake flask method. Briefly, an excess amount of drug was suspended in 10 ml of water, and the suspensions were shaken and filter through a 0.22µm whatmen filter. The filtered solution was suitably diluted and the ritonavir concentration in the filtrate was analysed by UV analysis method at 234nm.

 

The solubility of best batch of formulation was measured by centrifugal method. Briefly, 10 ml of nanosuspension was loaded into centrifugal tubes. Samples were centrifuged at 10000 rpm for 10 min. The supernatant solutions were analysed using UV spectrophotometer at 234 nm.

 

5. Stability Study:

The final formulations were subjected to stability studies as per ICH guidelines. Various parameters such as drug content, entrapment efficiency were measured before and after 30, days of stability 25.

 

Table 2: Conditions for Stability Study

Duration of study

30 days

Temperature conditions

40º±2ºC

Relative humidity

75± 5 %

 

Drug Excipients compatibility study:

Drugs are administered and often they are combined with excipients to compound them into a convenient dosage form. Excipients, although pharmacologically inert, result in chemical or physical interaction with drug and interfere with quality and stability of drug products.

 

The quality of solid dosage form is dependent on physicochemical properties of drug and the excipients. The evaluation of drug–excipients compatibility is based on inherent properties of the excipient. Assessment of possible incompatibilities between the drug and different excipients is an important part of preformulation.

Thermal analysis (DSC) and FTIR has been used extensively used for stability and compatibility studies.

 

1. FTIR Spectroscopy:

Fourier Transform Infrared (FTIR) (Shimadzu) spectroscopy was performed by placing a Ritonavir with excipient sample in FTIR sample holder. The drug sample was placed and scanned over the range of 4000-400 cm-1. The obtained spectrum was recorded.

 

2. Differential scanning calorimetry (DSC):

DSC (METTLER) can be used to detect the physical compatibility among the drug and polymer, by determining the thermal behaviour of pure ritonavir, eudragit RS100, poloxamer 407, SLS and physical mixture of ritonavir + eudragit RS100 + poloxamer 407 + SLS and selected formulas [26] [27].

 

RESULTS AND DICUSSIONS:

1.    Melting point:

The melting point was determined by capillary method and it was found to be and complies with I.P.2014.

 

Table 3: Melting Point of Ritonavir.

Parameter

Observed

Reported standard

Inference

Melting point

120-125ºC

119-123ºC

Complies with I.P.

 

2. UV- Visible spectrophotometric characterization:

Determine of Calibration Curve ritonavir in methanol:

The Ritonavir with scanning range of 200-400 nm. The maximum wavelength of the drug was found to be 234 nm in methanol.

 

Figure 1: Calibration Curve of Ritonavir in Methanol


 

Table 4: Particle Size Data of NS Formulations:

 Formulation Code

Average Particle Size (µm)

 F1

0.012

 F2

0.015

 F3

0.017

 F4

0.014

 F5

0.013

 F6

0.016

 F7

0.006

 F8

0.007

 F9

0.009

 

3. IR Spectroscopy:

Interpretation IR spectrum of ritonavir:

 

Figure 2: IR Spectrum of Ritonavir

 

·      Drug –Excipient compatibility studies:

 

Figure 3: IR Spectrum of Drug and Other excipient

 

Figure 4: DSC graph for RTV

 

Figure 5: DSC graph for prepared nanosuspension

 


 

Differential scanning calorimetric analysis:

The thermal behaviour of drug was examined by DSC, and RTV shows exothermic peak at the 160°C therefore drug is in pure state.

 

Particle size analysis:

The particle size of the NS produced Systems was analysed by motic digital microscopy. The particle size of each formulation was carried out and a result indicates that all formulations found in the nanosized range. The F7 formulation shows better results because of low were particle size compared to others. The batch F7 had an average particle size of 0.006µm which indicate the particle is in uniform distribution.


 

Figure 6: Particle Size Analysis by Motic Microscope of F7 Formulation.

 


 

Total drug content:

The drug content of all NS formulations was found to be greater than 80%. Indicating suitability of these methods for particle size reduction.

 

Table 5: Total Drug Content of Formulations.

Formulation Code

Total Drug Content (%)

F1

82.64

F2

84.25

F3

80.79

F4

85.27

F5

81.69

F6

87.99

F7

96.90

F8

92.57

F9

94.89

 

Figure 7: Total Drug Content of Formulated Nanosuspension.

 

Entrapment efficiency:

The entrapment efficiency of all formulations was found to be in the range of (80.11-94.29%) the entrapment efficiency of F7 was high when compared to other formulations.

 

Table 6: Entrapment Efficiency Data of Formulations.

Formulation Code

Entrapment Efficiency (%)

F1

82.70

F2

81.56

F3

85.21

F4

81.39

F5

85.57

F6

80.11

F7

94.29

F8

87.49

F9

91.89

 

Figure 8: Entrapment Efficiency of Formulated Nanosuspension.

 

Saturation solubility studies:

Formulations

Saturation Solubility (µm/ml)

Pure drug

13.65

Nanosuspension

66.75

 

Figure 9: Comparison of Solubility of Ritonavir and its Nanosuspension

 

Stability Study:

 

Table 7: Stability Data of F7 Formulation of Nanosuspension.

Time Period

 Entrapment Efficiency

Total Drug Content

Initial (0days)

 94.29%

96.19%

Average Storage

1 month

 93.65%

96.03%

 

CONCLUSION:

The obtained results of the present study demonstrate that nanoprecipitation technique was employed to produce nanoparticles of ritonavir, a poorly water-soluble drug, for the improvement of solubility and bioavilability. In this process, the particle size of ritonavir can be obtained in the nano-size ranges, by adjusting the operation parameters. In conclusion, the appropriate selection of process parameter and formulation parameters we can conclude that nanoprecipitation method is a simple and effective approach to produce nanosized particles of poorly water soluble drugs with enhance solubility.

 

LIST OF ABBREVATIONS:

1.    SLS – Sodium Lauryl Sulphate

2.    ART – Antiretroviral Therapy

3.    HIV – Human immunodeficiency virus

4.    AIDS – Acquired immune deficiency syndrome

5.    UV – Ultra violet

6.    NS – Nanosuspension

7.    ICH – International Conference on Harmonization

8.    DSC – Differential Scanning Calorimeter

9.    FTIR – Fourier Transform Infrared

10. IP – Indian Pharmacopoeia

11. RTV – Ritonavir

12. RPM – Revolution Per Minute

13. TDC – Total Drug Content

14. EE – Entrapment Efficiency

 

CONFLICT OF INTEREST:

The authors have no conflicts of interest regarding this investigation.

 

ACKNOWLEDGEMENT:

We are grateful to the teacher’s and Principal of Loknete Dr. J. D. Pawar College of Pharmacy, Manur, Tal. Kalwan for their helpful guidance.

 

REFERENCES:

1.     Sarika V. Kandbahale. A Review- Nanosuspension Technology in Drug Delivery System. Asian J. Pharm.Res.2019, 9(2):130-138. Doi: 10.5958/2231-5951.2019.00021.2

2.     Arunkumar N, Deecarman M, Rani C; Nanosuspension technology and its application in drug delivery. Asian Journal of Pharmaceutics. 2009; 3:168-173.DOI: 10.4103/0973-8398.56293

3.     Subrahmanyam CVS. Text book of Physical Pharmaceutics. 3rd edition, Vallabh Prakashan, 2015; 411-412.

4.     Hsu A, Granneman G.R, Bertz R.J. Ritonavir: clinical pharmacokinetic and interactions with other anti-HIV agents.Clin. Pharmacokinet.1998 35, 275-291. http://doi.org/10.2165/00003088-199835040-00002.

5.      I Somasundaram, B.V. Nagarjuna Yadav, S. Sathesh Kumar. Formulation of PLGA Polymeric Nanosuspension containing Pramipexole Dihydrochloride for improved treatment of Parkinson’s Diseases.Research J. Pharm. And Tech.2016;9(7):810-816.doi:10.5958/0974-360X.2016.00155.4

6.     C.Rubina Reichal, Christa Roshan Pius, S. Manju, M. Shobana.Formulation and Characterization of Gliclazide Nanosuspension. Research J.Pharm. and Tech.2011;14(2):779-786.doi:10.5958/0974-360X.2021.00136.0

7.     M.Santhosh Raja, K. Venkataramana. Formulation and Evaluation of Stabilized Glipizide Nanosuspension prepared by Precipitation Method.Research J, Pharm. And Tech. 2020; 13(11):5145-5150.doi:10.5958/0974-360X.2020.00900.2

8.     Pawar R.N, Chavan S. N, Menon M.D, Development, Characterization and Evaluation of Tinidazole Nanosuspension for Treatment of Amoebiasis. J Nanomed Nanotechnol 2019.7:413.doi:10.4172/2157-7339.1000413.

9.     Shengokar r, Mullar R. H, Nanocrystals: Industrially feasible multifunctional formulation technology for poorly soluble actives. Int.J.Pharm.Elsevier B. V.; 2010;399:129-39.http://doi.org/10.1016/j.ijpharm.2010.07.044

10.   Amol S. Deshmukh. Solid Lipid Nanoparticles. Res. J. Pharm.Dosages Form. And Tech. 6(4): Oct.-Dec.2014; Page 282-285.

11.   Solid Lipid Nanoparticles: A Promising Nanotechnology. Akshat Sharma, Amit Dubey, Reenu Yadav. Research J. Pharm. Dosages Forms and Tech. 2011; 3(5):167-175.

12.   Pankaj A. Jadhav, Adhikrao V. Yadav.Polymeric Nanosuspension Loaded Oral Thin Flims of Flurbiprofen: Design, Development and In Vitro Evaluation. Research J. Pharm. And Tech. 2020;13(4):1905-1910.

13.   Jessy Shaji, Monika Kumbhar. Linezolid Loaded Biodegradable Polymeric Nanoparticles Formulation and Characterization. Res. J.Pharm. Dosages Form. and Tech. 2018; 10(4): 271-278. Doi: 10.5958/0975-4377.2018.00040.x.

14.   Pramod Salve, Suvarna Pise, Nikhil Bali. Formulation and Evaluation of Solid Lipid Nanoparticles Based Transdermal Drug Delivery System for Alzheimer’s Disease.Res. J. Pharm. Dosages Form. And Tech. 2016: 8(2):73-80. Doi: 10.5958/0975-4377.2016.00011.2

15.   Mistry Khushboo, Kavya Naik, Vasanti, Alicia Menezes, Anup Naha, Koteshwara K. B., Girish Pai K, Formulation and Evaluation of Irbesartan nanosuspension for Dissolution Enhancement. Research J. Pharm.and Tech. 2017; 10(9):3043-3048. doi:10.5958/0974-360X.2017.00540.6

16.   Nawale R. B., Deokate U. A., Shahi S. R., Lokhande P. M., Formulation and Characterization of Efavirenz Nanosuspension by QbD approach. Research J. Pharm. and Tech 2017; 10(9):2960-2972.doi:10.5958/0974-360X.2017.00525.X

17.   Patel H. M, Patel B.B, Shah C. N, Shah D. P, Nanosuspension Technologies for Delivery of Poorly Soluble Drugs-A Review.Research J. Pharm. and Tech. 2016;9(5):625-632.doi:10.5958/0974-360X.2016.00120.7

18.   Sharma S., Issarani R., Nagori B.P., Development of Acaclofenac Nanosuspension Stabilized by Poly vinyl alcohol and Sodium dodecyl sulphate. Research. J. Pharm. and Tech. 8(3): Mar., 2015; Page 235-241. doi:10.5958/0974-360X.2015.00027.X

19.   Nayak S., Panda D., Patnaik A. K., Nanosuspension-Preparation, In Vitro and Ex Vivo Evaluations of Felodipine Hydrochoride.Research J. Pharm.and Tech. 8(1): Jan. 2015; Page 38-43. doi:10.5958/0974-360X.2015.00008.6

20.   D’Souza S. A review of in vitro drug release test methods for nano-sized dosage forms. Advances in Pharmaceutics. 2014; 2014:1-12.DOI: 10.1155/2014/30

21.   Harsh Joshi, Priyanka Ahlawat. Solid Lipid Nanoparticles for nose to Brain delivery: A Review. Res. J. Pharm. Dosages Forms and Tech. 2021; 13(1):57-61.doi:10.5958/09754377.2021.00010.0

22.   Bhalekar M, Upadhya P, Reddy S, Kshirsagar S, Madgulkar A, Formulation and evaluation of acyclovir nanosuspension for enhancement of bioavaibility. Asian J. Pharma 2014;8:110-8

23.   Junyaprasert VB, Morakul B. Nanocrystals for enhancement of oral bioavailability of poorly water soluble drugs. Asian journal of pharmaceutical sciences. 2015 1;10(1):13-23.DOI: 10.1016/j.ajps.2014.08.005

24.   Prabhakar C.H, Krishna B.K. A Review on nanosuspensions in drug delivery. International Journal of Pharma and Bio Sciences, 2011.

25.   Pudipeddi M, Serajuddin A. T. M, Trends in solubility of polymers. J. Pharm. Sci. 2015. http:// doi.org./10.1002/jps.20302.

26.   Atul Phatak, Pallavi Jorwekar, P. D. Chaudhari. Nanosuspension: A Promising Nanocarrier Drug Delivery System. Research J. Pharma.Dosage Forms and Tech.2011; 3(5): 176-182.

27.   Lakavath Sunil k. A Review on Novel vesicular systems for enhanced Oral bioavailability of lipophilic drugs. Research Journal of Pharmaceutical Dosages Forms and Technology.2021; 13(2):139-6. Doi: 10.52711/0975-4377.2021.00025.

 

 

 

Received on 30.05.2021       Modified on 12.06.2021

Accepted on 28.06.2021     ©A&V Publications All Right Reserved

Res. J. Pharma. Dosage Forms and Tech.2021; 13(4):297-304.

DOI: 10.52711/0975-4377.2021.00049