84
LUČKA BIBIČ FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE ORAL ADMINISTRATION IN A SEMI-SOLID CARRIER MASTER’S THESIS Ljubljana, 2014

FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

Page 1: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

LUČKA BIBIČ

FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE ORAL

ADMINISTRATION IN A SEMI-SOLID CARRIER

MASTER’S THESIS

Ljubljana, 2014

Page 2: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

LUČKA BIBIČ

FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE ORAL

ADMINISTRATION IN A SEMI-SOLID CARRIER

RAZVOJ MIKROKAPSUL Z ALBENDAZOLOM V POLTRDNEM VEHIKLU ZA

PERORALNO JEMANJE

Academic supervisor: Prof. Dr. Mirjana Gašperlin

Academic co-supervisor: Assoc. Prof. Dr. Daniel Bar-Shalom

Ljubljana, 2014

Page 3: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 3

This Master’s thesis was done at the Faculty of Pharmaceutical Sciences (Rational Oral Drug Delivery

Group), University of Copenhagen under the academic supervision of Prof. Dr. Mirjana Gašperlin

from the Faculty of pharmacy, University of Ljubljana, Slovenia and co-supervision of Assoc. Prof. Dr.

Daniel Bar-Shalom, from the Faculty of Pharmaceutical Sciences, University of Copenhagen,

Denmark.

Page 4: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 4

Statement

I hereby declare that this Master’s thesis was done by me, Lučka Bibič, under the academic

supervision of Prof. Dr. Mirjana Gašperlin from the Faculty of pharmacy, University of Ljubljana,

Slovenia and co-supervision of Assoc. Prof. Dr. Daniel Bar-Shalom from the Faculty of Pharmaceutical

Sciences, University of Copenhagen, Denmark.

Lučka Bibič

Head of Committee: Prof. Dr. Janko Kos

Member of Committee: Doc. Dr. Jurij Trontelj

Page 5: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 5

Just keep swimming.

[Nemo]

Page 6: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 6

ACKNOWLEDGEMENTS

I’m not sure if anyone reads the acknowledgments, but I hope so because without the following

people, it would have been nearly impossible to create this piece of work.

First of all, I would like to acknowledge my co-supervisor, Assoc. Prof. Daniel Bar-Shalom. Your

brilliant ideas and pearls of wisdom, accompanied with the enormous experience and knowledge

inspired me through my Master thesis’s path and helped me realize the power of critical and creative

thinking. I would like to thank you for general guidance on writing scientific papers and putting up

patiently with my long barrages of e-mails while providing amazingly timely feedback. I really

appreciate your willingness to treat me as a future colleague during my stay at the Faculty of

Pharmaceutical Sciences in Copenhagen and let me experience this research project beyond the

scope of the textbooks.

My next greeting goes to Prof. Mirjana Gašperlin, my supervisor at the Faculty of Pharmacy in

Ljubljana. It was my privilege to have you as my supervisor and I would like to say one big “Thank

you!” - for your patient, care and editorial guidance. Your kindness and reassuring words mean a

world to me.

I am indebted to all the great people working at the Department of Pharmaceutics and Analytical

Chemistry at the Faculty of Pharmaceutical Sciences in Copenhagen. Special thanks go to the RODD

group on the 3rd floor for providing a great working atmosphere with plenty of support, genuine

interest and good company. It would be a lonely lab without all of you.

I also want to express my sincere gratitude to Kaisa, Jette, Charlotte, Tonie, Dorte, Alan, Martin and

Yun. I really appreciate that you always manage to be available when discussions and guidance were

needed and that some of you always found the time for a coffee break, when I was buffeted by the

winds of uncertainty.

I am also very grateful to all the children and their caring parents at the Hvidovre Hospital in

Denmark, Pediatric Department, for letting us close. Thank you kids for the invaluable insights into

this thesis’s topic.

Pharmaceutical study turned out to be a long 5-years battle against the small molecules. Therefore, I

wish to thank to all of my great friends for keeping me sane. I am superlatively thankful that you

understood my mental pauses from time to time. Nina, Rok, Vid, Katarina, Špela, Barbara, Maruša,

Mariša and Maša, thanks for reminding me, what really matters in life.

It is hard to overstate my gratitude to my father Vojko and both my brothers, Klemen and Jošt, who

are probably saying they do not need thanking. I am also grateful to both my grandmothers and

grandfathers – I bet you are proud of me.

Finally, a loving thanks to my mother, Renata, who helped and lead me through my first

pharmaceutical breakthrough and who still managed to save some nerves. Thanks mom for putting

up patiently with my frantic Danish calls at 2am, when I needed to know why my HPLC method is not

working properly.

Page 7: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 7

TABLE OF CONTENTS

ACKNOWLEDGEMENTS ........................................................................................................................... 6

TABLE OF CONTENTS ............................................................................................................................... 7

LIST OF TABLES ........................................................................................................................................ 9

LIST OF FIGURES .................................................................................................................................... 10

LIST OF GRAPHS ..................................................................................................................................... 10

ABSTRACT .............................................................................................................................................. 11

POVZETEK .............................................................................................................................................. 12

LIST OF ABBREVIATIONS ........................................................................................................................ 14

1. BACKGROUND ............................................................................................................................... 15

1.1. Parasitic Helminths in the Third World Countries ................................................................. 15

1.2. Pediatric drug formulation .................................................................................................... 15

1.2.1. UNICEF gel ..................................................................................................................... 16

1.3. Active Pharmaceutical Ingredient (API) ................................................................................. 17

1.3.1. Albendazole (ALB).......................................................................................................... 18

1.3.2. Caffeine (CAF) ................................................................................................................ 18

1.4. Microcapsules as a Drug Delivery System ............................................................................. 19

1.4.1. Definition ....................................................................................................................... 19

1.4.2. Microencapsulation (MEC) operation ........................................................................... 20

1.4.3. Microencapsulation (MEC) materials ............................................................................ 22

1.4.4. Pediatric patients and taste concealing ........................................................................ 23

1.5. Pharmacokinetic barriers in anthelmintic action and the role of dissolution ....................... 25

1.5.1. BCS Class II Drugs and IVIVC .......................................................................................... 26

1.5.2. Solubility ........................................................................................................................ 26

1.5.3. Taste-Concealing drives Dissolution .............................................................................. 27

1.5.4. Dissolution rate ............................................................................................................. 27

1.5.5. In vitro methods ............................................................................................................ 28

1.6. HPLC Analysis ......................................................................................................................... 28

1.6.1. Method Development for RP-HPLC ............................................................................... 29

1.6.2. Method Validation ......................................................................................................... 30

2. AIM ................................................................................................................................................ 30

3. MATERIALS and METHODS ............................................................................................................ 32

3.1. Equipment ............................................................................................................................. 32

3.2. Materials ................................................................................................................................ 33

Page 8: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 8

Methyl Cellulose (MC) Taste-Concealing .............................................................................................. 33

3.3. Methods ................................................................................................................................ 35

3.3.1. Characterization of ALB ................................................................................................. 35

3.3.2. Microencapsulation (MEC) ............................................................................................ 37

3.3.3. Dissolution ..................................................................................................................... 41

3.3.4. HPLC Analysis ................................................................................................................. 43

3.3.5. UV Spectroscopy ............................................................................................................ 47

3.3.6. Student t-test................................................................................................................. 47

4. RESULTS and DISCUSSION ............................................................................................................. 48

4.1. Characterization of ALB ......................................................................................................... 48

4.1.1. Identification of ALB by LC/MS ...................................................................................... 48

4.1.2. Particle size distribution (PSD) ...................................................................................... 48

4.1.3. Determination of bulk, tapped densities and flow properties ...................................... 50

4.1.4. Morphology determination of powders, granules and MIC ......................................... 51

4.2. Microencapsulation ............................................................................................................... 53

4.2.1. Determination of wall thickness of the MICs ................................................................ 53

4.3. Dissolution ............................................................................................................................. 54

4.3.1. Granules ........................................................................................................................ 54

4.3.2. Microcapsules for taste-concealing............................................................................... 58

4.3.3. ALB dissolution studies ................................................................................................. 61

4.3.4. Microcapsules and granules/ MICs in 0,1M HCl and in MilliQ Water ........................... 62

4.4. UV Spectroscopic measurements and t-test ......................................................................... 65

4.5. HPLC Analysis ......................................................................................................................... 65

4.5.1. Analysis for validation ................................................................................................... 65

4.5.2. Analysis for drug content estimation ............................................................................ 70

5. CONCLUSION ................................................................................................................................. 71

6. FUTURE RESEARCH SUGGESTIONS ................................................................................................ 73

7. REFERENCES .................................................................................................................................. 74

8. APPENDICES ................................................................................................................................... 84

Page 9: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 9

LIST OF TABLES

Table 1: Physicochemical properties of ALB and CAF -------------------------------------------------------------- 17

Table 2: The main variables that affect the granule's quality ---------------------------------------------------- 21

Table 3: Apparatuses for manufacturing and analysis of MEC -------------------------------------------------- 32

Table 4: Substances for manufacturing the granules -------------------------------------------------------------- 33

Table 5: Substances for manufacturing the coated granules ---------------------------------------------------- 33

Table 6: Chemicals for the UNICEF gel --------------------------------------------------------------------------------- 34

Table 7: Chemicals for HPLC analysis ---------------------------------------------------------------------------------- 34

Table 8: Chemicals used in the dissolution test --------------------------------------------------------------------- 34

Table 9: Twelve different formulations were prepared in the granulation experiments ----------------- 38

Table 10: the composition of the MC coating suspension -------------------------------------------------------- 39

Table 11: Process conditions for coating in fluid bed -------------------------------------------------------------- 39

Table 12: Excipients used for preparation of UNICEF gel ------------------------------------------------------ 43

Table 13: Calculated Bulk, Tap Densities and flow properties (CI and HR) of ALB, Dextrin powder,

granules and MIC ----------------------------------------------------------------------------------------------------------- 50

Table 14: Measured and calculated variables for the determination of the microcapsules’s wall

thickness ---------------------------------------------------------------------------------------------------------------------- 53

Table 15: Precision results from the nine replicate injections of ALB with their mean tr and Area with

the calculated RSD value -------------------------------------------------------------------------------------------------- 67

Table 16: Precision results from the nine replicate injections of CAF with their mean tr and Area with

the calculated RSD value -------------------------------------------------------------------------------------------------- 68

Table 17: Ruggedness results from the nine replicate injections of ALB with their mean tr and Area

with the calculated RSD value ------------------------------------------------------------------------------------------- 68

Table 18: Rugedness results from the nine replicate injections of CAF with their mean tr and Area

with the calculated RSD value ------------------------------------------------------------------------------------------- 69

Table 19: Results from the six replicate injections of ALB and CAF with their mean Area, LOD and LOQ

---------------------------------------------------------------------------------------------------------------------------------- 69

Table 20: Albendazole encapsulation efficiency for granules and microcapsules -------------------------- 70

Page 10: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 10

LIST OF FIGURES

Figure 1: Granulation process ------------------------------------------------------------------------------------------- 20

Figure 2: The process of coating in the fluid bed with a Wurster partition. ---------------------------------- 22

Figure 3: Modified Kenwood Food Processor -------------------------------------------------------------------- 32

Figure 4: Fluid bed MiniGlatt -------------------------------------------------------------------- 32

Figure 5: The pouch with the UNICEF gel ------------------------------------------------------ 43

Figure 6: Granules FOR6 size distribution with the granules diameter range from 75 μm to 177 μm- 49

Figure 7: MICs size distribution with the diameter range from 134 μm to 340 μm ------------------------ 49

Figure 8: SEM images magnification series (x75and x1000 respectively) of the granules ---------------- 52

Figure 9: SEM images magnification series (x35 and x750 respectively) of the MIC ----------------------- 52

LIST OF GRAPHS

Graph 1: Drug release profiles of ALB formulations FOR1-FOR5 in water within 30 min time range - 54

Graph 2: Drug release profiles of ALB formulations FOR6-FOR12 in water within 30 min time range 54

Graph 3: ALB release from uncoated, coated FOR6 granules and coated FOR6 in UNICEF gel with ALB

pure drug release ----------------------------------------------------------------------------------------------------------- 58

Graph 4: ALB release without and with the incorporation into the UNICEF gel ----------------------------- 61

Graph 5: ALB release from MICs and granules in 0,1M HCL and water --------------------------------------- 62

Graph 6: Standard curve for ALB ---------------------------------------------------------------------------------------- 66

Graph 7: Standard curve for CAF ---------------------------------------------------------------------------------------- 66

Page 11: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 11

ABSTRACT

Albendazole (ALB) is an anthelmintic drug, classified as BCS type II compound, mainly used in the

pediatric population in the third world countries. As the main setback of ALB is its low aqueous

solubility (1 μg/ml), the enhancement of ALB release rate is essential. Another main disadvantage of

oral drug delivery for pediatrics is the children’s inability to swallow. That is why a semi-solid

formulation might be a better oral dosage form. Because the bitter taste of ALB might deter children

from consuming the medicine, a microencapsulation technique would be a valuable achievement in

order to avoid ALB unpleasant taste. Furthermore, it is predicted that semi-solid formulation,

consisting of “UNICEF gel”, an HPMC based gel, with the incorporated taste-concealedmicroparticles,

should overcome this major problems.

In the first part of this thesis, a quantitative particle-size analysis, determination of flow properties

and LC/MS identification of ALB were carried out. Following that step, the granulation process, with

the various compositions of the numerous excipients, was conducted. After the granulation, the

appropriate polymer, dextrin, was chosen, according to the dissolution studies. In order to

quantitatively determine the ALB content and release from granules and microcapsules (MICs), a

rapid RP-HPLC method was developed, optimized and validated. In the second stage, a fluid bed

technique with a Wurster partition was selected for the coating with the Methyl Cellulose (MC) as a

potential taste-concealing polymer. Once the ALB microcapsules were MC-coated, they were

incorporated into UNICEF-gel and further dissolution studies were conducted at the neutral pH and

pH 1.2. Finally, spectroscopy was used in order to evaluate the ALB release in the oral cavity and

stomach from the MICs and the final semi-solid formulation.

The results obtained from our studies, suggest that it is possible to achieve a micro sized product of

poorly soluble drug ALB with the defined and narrow particle size distribution. Furthermore, dextrin

not only gives a good coverage of the core material, resulting in the smooth and uniform surface and

provides high entrapment efficiency, but also greatly improves the ALB release. The ALB dissolution

of the pure drug was 7,80% ± 0,39% in 30 min, whereas 93,15% ± 0,34% and 67,36% ± 1,19% of the

ALB present in uncoated (dextrin granules) and coated MICs, respectively, dissolved within the same

time range. Thus, a remarkably surprising ALB release from the MICs was achieved. It has also been

found that MICs incorporated into UNICEF gel might sufficiently conceal the bitterness of ALB for the

first 2 min.

Keywords:albendazole, microencapsulation, oral drug delivery, fluid bed, coating, granulation,

dissolution, taste-concealing, methyl-cellulose, dextrin, HPLC, semi-solid, pediatric

Page 12: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 12

POVZETEK

Albendazol (ALB) je antihelmintična zdravilna učinkovina, uvrščena v skupino II po biofarmacevtskem

klasifikacijskem sistemu (BCS), ki se pretežno uporablja v nerazvitih državah sveta pri pediatrični

populaciji. Ker je ena izmed glavnih pomanjkljivosti ALB je njegova nizka topnost v vodnem mediju

(manj kot 1 μg/ml), je ključnega pomena povečati sproščanje učinkovine, katere absorpcija je

omejena s hitrostjo raztapljanja. Druga pomanjkljivost peroralne dostave učinkovine pri pediatrični

populaciji pa je nezmožnost požiranja trdnih oblik pri mlajših otrocih. Eno od ustreznih rešitev tega

problema predstavlja poltrden vehikel. Z metodo mikrokapsuliranja sem želela izboljšati grenak okus

ALB, saj ta odločilno vpliva na otrokovo percepcijo jemanja zdravila. Zaradi tega poltrdna formulacija,

sestavljena iz UNICEF gela (HPMC gela), skupaj z vgrajenimi mikrokapsulami ALB, predstavlja možno

rešitev pri premagovanju problema požiranja pri otrocih.

V prvem delu naloge je bila izvedena kvantitativna analiza velikosti delcev, določitev pretočnih

lastnosti in test identifikacije molekule ALB z LC/MS. Sledili so številni procesi granuliranja z različnimi

količinami in vrstami pomožnih snovi. Po granuliranju je bil, glede na rezultate študij raztapljanja,

izbran najprimernejši polimer – dekstrin. Z namenom kvantitativne določitve količine ALB v granulah

in mikrokapsulah ter odstotka sproščene učinkovine iz obeh farmacevtskih oblik smo razviliRP-HPLC

metodo in potrdili njena ustreznost. V drugem delu študije smo izvedli oblaganje zrnc z

vrtinčnoslojno tehnologijo(Wurster). Kot polimer za oblaganje je bila uporabljena metilceluloza (MC)

s potencialno sposobnostjo prekrivanja neželenega okusa. Po oblaganju z MC smo mikrokapsule z

ALB vgradili v UNICEF gel in opravili številne študije raztapljanja pri nevtralnem in kislem pH. Na

koncu smo uporabili UV spektroskopijo za ovrednotenje sproščanja količine ALB iz ustne votline in

želodčnega soka, tako iz samih mikrokapsul kot mikrokapsul, predhodno vgrajenih v poltrdni vehikel.

Rezultati magistrske naloge potrjujejo, da je možno narediti mikrokapsule s težko topno zdravilno

učinkovino ALB z definirano velikostjo in ozko porazdelitvijo delcev. Hkrati je bilo ugotovljeno, da

dekstrin ne samoda omogoča ustrezno pokrivnost jedra mikrokapsul, kar se kaže v gladki in

enakomerni površini, temveč tudi zagotavlja dobro ujetost učinkovine v zrncih ter močno izboljša

količino sproščenega ALB, kar potrjujejo rezultati testa raztapljanja (po 30 min. je bilo količina

sproščenega ALB v le 7,80% ± 0,39%, medtem ko se je zrnc z dekstrinom in obloženih mikrokapsul z

MC sprostilo 93,15% ± 0,34% in 67,36% ± 1,19% v ALB enakem času).

Doseženo je bilo presenetljivo sproščanje ALB iz mikrokapsul in ugotovljeno, da mikrokapsule,

predhodno vgrajene v UNICEF-gel, zadovoljivo prekrijejo grenkobo ALB v prvih dveh minutah

sproščanja učinkovine.

Page 13: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 13

Ključne besede: albendazol, mikrokapsuliranje , peroralna dostava, vrtinčnoslojna tehnologija,

oblaganje, granuliranje, raztapljanje, prekrivanje okusa, metilceluloza, dekstrin, HPLC,poltrdni

vehikel, pediatrija

Page 14: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 14

LIST OF ABBREVIATIONS

AIDS = Acquired Immune Deficiency Syndrome

ALB = Albendazole

API = Active Pharmaceutical Ingredient

ATP = Adenosine Triphosphate

BCS= Biopharmaceutical Classification System

CAF = Caffeine

CCS = Croscarmellose Sodium

CI = Carr’s Compressibility Index

CIKDEX = Cyclodextrin Beta

DEX = Dextrin

EPA = Environmental Protection Agency

ESI = Electro-Spray Ionization

FDA = Food and Drug Administration

FOR = Formulation

GR = Granules

GR FOR6 = Granules of Formulation 6

H= Wall thickness

HAc = Acetic Acid

HIV = Human Immunodeficiency virus

HPMC = Hydroxypropylmethycellulose

HPLC = High-Performance Liquid

Chromatography

HR = Hausner Ratio

ID = Internal Diameter

IR = Immediate Release

ISO = International Standards Organization

IVIVC = In Vitro In Vivo Correlation

LC = Liquid Chromatography

LOD = Limit of Detection

Log P = Partition Coefficient

LOQ = Limit of Quantification

KCl = Potassium Chloride

MEC = Microencapsulation

MC =Methyl Cellulose

MIC = Microcapsule

MICs = Microcapsules

MP = Mobile Phrase

MS = Mass Spectroscopy

NO. = Number

OTC = Over the Counter

PH EUR = European Pharmacopoeia

PIP = Pediatric investigation Plan

PSD = Particle Size Distribution

PVP = Polyvinylpyrrolidone

RP = Reverse Phrase

SA = Surface Area

SD = Standard Deviation

SEM = Scanning Electron Microscope

SP = Stationary Phrase

STH = Soil-Transmitted Helminth

UNICEF = United Nations Children’s Fund

USP = United States Pharmacopoeia

VH20 = Volume of Water

WAR = Water Addition Rate

WHO= World Health Organization

Page 15: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 15

1. BACKGROUND

1.1. Parasitic Helminths in the Third World Countries

Globally, soil-transmitted helminth (STH) infections constitute one of the most significant neglected

infection clusters of our time. (1,2) It is estimated that there are more than two billion cases

worldwide, of which 450 million have attributed a major morbidity due to their infection; the

majority of whom are children between 5-14 years. (3) In addition, the infectious disease is the most

common reason for death of children, which are younger than 5 years, predominantly in the low-

income countries. (4)Black et al.estimated that among the 8.80 million children’s deaths, all under

the age of 5, two-third were due to infectious diseases. (5)

The World Health Organization (WHO) and the United Nations Children's Fund (UNICEF), among

others, suggested deworming programs targeting school-age children to reduce STH burden of

disease. (6)(7)The principal intervention available for controlling helminthes infections is the periodic

administration of one of the four anthelmintic drugs recommended by WHO, of which

benzimidazoles – mebendazole and albendazole are the most regularly used anthelmintics for the

treatment of STH infections. (1)

Furthermore, Bentwichet al. (3) outlined that intestinal helminth infections might have an effect on

the vaccination efficacy for HIV and tuberculosis due to the increased susceptibility of the host’s

immune system and spread of the diseases. Consequently, eradication of helminth infections may

have a major impact on AIDS and tuberculosis in the developing countries and also carry possible

benefits in the disease pathogenesis.

1.2. Pediatric drug formulation

Pediatric drug delivery is a major challenge in the drug development. Focusing on the factors such as

solubility and taste of the formulation, development might be technically demanding and time

consuming. Yet, some drugs compulsory for pediatric patients are not commercially accessible in

dosage forms that should be suitable for use in this population.(4,8)

There is a safety concern when unsuitable medicines are administrated to children.

It is highly improbable that a “one-size-fits-all” solution will provide the best outcome, but with the

current increase in interest and research, an upward trend for better pediatric dosage forms is likely

to follow. (4,9)

Page 16: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 16

A new pediatric regulation came into operation in the European Union on the January 2007 and the

regulatory environment for pediatrics in Europe was dramatically changed. Its objective is to improve

the children’s health by promoting the development of medicines as well as ensuring the high quality

and appropriate authorization. These pediatric trials must be conducted according to a detailed

pediatric investigation plan (PIP), which ought to demonstrate the quality, safety and efficacy of the

therapeutic product for the pediatric population. It is truly optimistic to know that new campaigns

have been launched to accelerate pediatric drug development. For example, “Make medicines child

size” is a worldwide initiative under the leadership of WHO, which is promoting the need for the age-

appropriate formulation developments. To sum up, this initiatives aim at that pediatric drug

development will become an integral part of the development of medicinal products. (4,10,11)

So far, there is a lack of suitable and safe drug formulations for children. (4) Pediatric oral dosage

forms include mostly tablets, emulsions and suspensions. (12) However, with the liquid formulations,

a number of drawbacks have been shown: difficulties in transport, storage and handling, stability

issues and poor dose uniformity when using spoons and dosing cups. (13) In addition, finding non-

toxic excipients and masking the taste is harder to achieve than for solid formulations. (4)

The main disadvantage of oral drug delivery for the young children is difficulty or inability to swallow.

(9) Hence, there is a need for a semi-solid composition that behaves like a liquid when consumed and

yet, acts like a solid in many other ways. In such formulation, chewing and biting would not be

necessary, which explains their ideal nature for being use in the pediatric field. Moreover, semi-solid

formulations enable better taste, mouth feel and storage stability. (12,14)In Krause&Breitkreutz(4)

review it had been noted that small-sized multiparticles can be swallowed together with the liquid

food by very young children, aged from 6 months, who are unable to receive solid oral dosage form.

In this thesis, a semi-solid drug formulation with incorporated microparticles will be developed. The

dosage form would primary be meant for toddlers and children, between two and five years,

regarding that the main disadvantage of oral drug delivery them is difficulty or inability to swallow,

especially in the case of the bitter drugs.

1.2.1. UNICEF gel

It was reasoned before, that children are not fond of syrups as a liquid formulation. (15,16)

In addition, Bar-Shalom (17) proposed to formulate a dosage form that should resemble something

children are used to take, for instance a dry pudding, where the microencapsulated drug would swell

into the pudding-like vehicle - UNICEF gel. (17,18)

Page 17: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 17

Small-sized multiparticles are thus a valuable alternative to the liquid formulations. Since the drug

will be suspended in the pudding, the particles should be microparticles (75 – 200 µm). If smaller, the

particles can be trapped between the taste buds on the tongue, whereas if bigger, a child could find

the grittiness objectionable. (17)

To produce a gel, a fast hydrating quality of Hydroxypropyl methylcellulose (HPMC), as a gelling

agent, was used in the vehicles. The vehicle should be attractive for the target group in terms of

taste, viscosity, texture and appearance, and possess properties like being fast hydrating and

easy to prepare.(18)

1.3. Active Pharmaceutical Ingredient (API)

In this thesis, albendazole (ALB) has been investigated as a main compound and caffeine (CAF) as a

model drug. In Table 1, the physicochemical properties of ALB and CAF are seen.

Table 1: Physicochemical properties of ALB and CAF

Physicochemical property / Drug ALBENDAZOLE(19,20) CAFFEINE (19,21,22)

BCS Class II I

MW 265,3 194,2

Structural formula

Chemical Formula C12H15N3O2S C8H10N4O2

Melting Point 209 °C 238 °C

pKa 2,68 (pKa1)and 11,83 (pKa2) 14,0

Aqueous solubility1in buffer pH 6,0 1 μg/ml 20 mg/ml

Log P 3,5 (predicted) and 2,7

(experimental)

-0,07 (predicted)and

-0,55 (experimental)

Absorption Poorly absorbed from the

gastrointestinal tract

Readily absorbed after

oral administration

1According to Wu, Zimei, et al, water solubility is 0,61 μg/L. (23)Nevertheless, the study from Bassani, V.L., et.al., shows that

water solubility at pH 7,2 is even lower; 0,2 μg/L. (24)

Page 18: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 18

1.3.1. Albendazole (ALB)

ALB is a broad-spectrum anthelmintic which is used to treat infections caused by worms. ALB is a

benzimidazole drug whose selectivity is known to be 250 – 400 times more potent in helminth than

in mammalian tissue.(2)

ALB mechanism of action is to keep the worm from absorbing glucose, which leads to the decreased

production of adenosine triphosphate (ATP). ATP then causes the depletion of vital energy for the

worm’s endurance. Due to diminished energy production, the worm is immobilized and ultimately

dies.(20)

Furthermore, ALB is a potential anticancer agent that is currently under development for the

treatment of cancer. (25)

The majority of the orally administrated drugs taste very bitter and ALB is no exception.

(24,25,26,27) Thus, one of the main drawbacks of ALB is its bitter taste and oral administration of

ALB is a key issue for the formulation scientist since the drug acts on the parasites in the

gastrointestinal lumen.

An additional challenge, when ALB is concerned, includes ALB’s low aqueous solubility. (26)Regarding

low ALB solubility and its high partition, the drug is classified as BCS type II compound. Moreover,

ALB is known to be practically insoluble in both gastric and intestinal fluids. Slow release from the

stomach increases the time for ALB to dissolve before entering the intestine, thus favoring increased

bioavailability. (20,24)Thereby, achieving better dissolution in the stomach would decrease the time

for ALB to dissolve before entering the small intestinal lumen, where the helminth parasites are

presented.

As with other poorly soluble compounds, the dissolution rate is likely to be dependent on the

formulation. (24) One of the aims in this work is to improve the dissolved percentage of ALB in the

stomach, which could consequently lead to better exposure to the parasites in the intestine and

enhanced antiparasitic activity of ALB.

1.3.2. Caffeine (CAF)

In this study, CAF has been chosen as a model drug. This high-water soluble and bitter xantine

derivate was mainly selected to investigate the patency of the taste concealing.

Page 19: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 19

That is to ensure if the taste concealing coat could remain unobstructed and, consequently, if the

drugs release rate from a microencapsulated dosage form, containing ALB and CAF, can be limited for

that reason.

1.4. Microcapsules as a Drug Delivery System

1.4.1. Definition

“Microencapsulation is both an art and a science.” (28)

By the Allen et al. (29), the microencapsulation (MEC) is a drug delivery technique, defined as the

entrapment of solids, liquids or gases in the microscopic particles. This encapsulation is achieved by

the deposition of a thin layer of coat material around the core. Others, Banker et al. (30) define MEC

as a process where the particles of core material, either solid or liquid in nature, are enclosed within

a thin wall of the polymer.

There are numerous reasons for MEC: concealing the bitter taste of drugs; reducing GIT irritation;

easier handling and storage; reducing the hygroscopic properties of core material; reducing the drugs

odor and volatility; protecting the core material against atmospheric effects and separating the

incompatible substances. In addition, MEC can be used as a controlled release delivery system. (28)

In this study, the microcapsules (MICs) have been prepared by two separated production processes:

wet granulation, with a modified Kenwood Food Processor, followed by film coating using fluid bed

equipment.

1.4.1.1. Wet granulation

Benali et al. (31) define the granulation as the process where particles agglomerate into bigger

aggregates (granules) where the initial particles could, yet, be identified. (31,32) With the

granulation, we may: improve flow properties of the powders and hence the uniformity of the dose;

increase the bulk density of a product; decrease dust generation and, last but not least, improve the

product appearance.(29) That is why this process is one of the powerful unit operations in the

production of pharmaceutical oral dosage forms. (33)

Granulation methods are divided into two separate types: dry granulation and wet granulation. (34)

In the wet granulation process, the granulation is typically performed in a tumbling drum, a fluidized

bed, a high shear mixer or similar apparatus.

Page 20: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 20

Wetting and nucleation consolidation and growth attrition and breakage

After the particles are placed in the equipment, the agitation starts and the addition of a liquid,

commonly water, is added in a pre-defined range of time. (31)

The major advantage of wet granulation is, among others, its feature to make hydrophobic surfaces

hydrophilic with increased sphericity and uniformity. (32,34,35)

However, the preferred product’s attributes are not controlled just by choosing the right powder

mass and liquid, since the granulator type and operating parameters, as seen in Table 2, are also of a

great importance.(35)It is fairly common to see granulation as three-stage process, starting with the

wetting and nucleation, followed by the consolidation and growth, and at the end there comes the

attrition and breakage:

Figure 1: Granulation process

1.4.1.1. Coating

Coating is defined as the process of application when a liquid material, which is applied on the

surface, results in either a continuous or discontinuous film after drying. (36,37) This shell-like barrier

structure provides a shelter of a core material that may be enclosed in the particle, its surface or

entrapped within the coating layer. (29,38) Coating of the solid particles is considered to have

countless advantages, among them also the taste concealing that was the main reason for MEC in

this thesis.

1.4.2. Microencapsulation (MEC) operation

1.4.2.1. Granulation in a modified food processor Kenwood

The use of the smaller scale mixer (food processor) was choose according to the time and material

efficiency issues as more experiments can be conducted using smaller quantity of material and less

time.

The modified food processor is, when compared with the high-shear mixers, the equipment without

the chopper and with the different operating impeller.

Page 21: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 21

In recent years, many experiments, for instance from Michaels et al. (39), established a belief that

chopper has no effect on the granule size distribution. On the other hand, Schaefer et al. (40)well –

documented study suggested that chopper’s effect vaguely decreases the mean particle size and

narrows the PSD.

Fu, J. S., et al.(41)has established a belief that when different features of granules, made with high-

shear mixer and food processor, are questioned, a good correlation between the results can be

established.

Therefore it can be suggested that granulation process in the food processor resembles the one use

in the high shear mixer. In high shear granulation, as well as in the food processor, the powder is

constantly agitated by the impeller and a binder solution is sprayed from the top. (42,43,44)The first

nuclei is rapidly made as the liquid droplets are dispersed into the powder, while the growth of the

inseparable, and thus larger, granules (agglomerates) is prevented by the agitation force. When

mixing and spraying persist, the agglomerates develop into harder particles with more adhesive

surface and hence enlarge. (42,45) The principal difficulty, concerning the wet granulation in the

both of the mixers, is the control of the end point. (46) Whilst an excess of liquid can form a slurry or

unmanageable growth of the particles, the granulation process is supposed to be stopped. (47)

The main variables affecting the quality of the granules are seen in Table 2. (31,45,47,48,49,50)

Table 2: The main variables that affect the granule's quality

1.4.2.2. Coating in a fluid bed with a Wurster partition

The requirement for the coated particles is constantly emerging in the pharmaceutical area. (51)

There are few apparatus which can be applied for the coating of pellets, granules and powders, with

the fluidized bed as the most common one. (52)

Material parameters Granulation condition in the

mixer Granule properties

Powder particle size distribution (PSD) Impeller and Chopper Speed PSD

Wettability of the solid by the liquid Massing time Bulk density and

porosity

Binder concentration and viscosity Load of mixer (generally 2/3) Flow properties

Solid solubility and degree of swelling and

binder liquid

Liquid spray rate Flow properties

Quantity of the solvent Drug content

uniformity

Temperature Binder distribution

Page 22: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 22

When spraying of the coating material is concerned, three basic designs are commonly known: top-

spray, side spray and bottom-spray (Wurster). (52) The last configuration (Wurster on Figure 2) has

been extensively studied to be a powerful and most acceptable tool for the coating of small

particles. (33,53) A nozzle and a bottom-spray tube are located right in the middle of a perforated

distributor plate, which allows the granules to flow in a predictable circulating trajectory. (51,54)

During the coating process there is a short distance between the coating materials and particles,

which greatly contributes to the high coating uniformity and coating efficiency. (55)

Numerous investigations, regarding the particle circulation in the Wurster, have been published.

Experiments from Teunouet al. (52), Becheret al. (56), Ström et al.(57), Tzika et al.(58), Arimoto et al.

(59) and Christansenet al. (60) have evaluated the Wurster’s impact of the coating thickness and

quality of the layer.

They all hold a common belief that the

bottom spray design (Figure 2) is the most

proficient fluidized bed configuration for

coating when the high-quality of the coated

layer is essential. Furthermore, Hampel et al.

(51) and Watano et al. (61) have confirmed

that when uniform layer is concerned, the

Wurster equipment surely provides the most

excellent outcome.

All this features determines the highest coating quality, which is mandatory if one requires producing

the defined and reproducible drug delivery profiles. (29)

1.4.3. Microencapsulation (MEC) materials

1.4.3.1. Core materials

The core material, which can be either liquid or solid, is identified as the particular material that

requires to be coated. (28) The solid core should consist either of the active constituents, stabilizers,

diluents or other excipients and release-rate retardants. In order to achieve a certain design of MICs

with the efficient properties, changing the core material’s composition is vital. (36)

Figure 2: The process of coating in the fluid bed with a

Wurster partition.

Page 23: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 23

1.4.3.2. Coating materials (polymers)

Selecting the most suitable coating material is of a key importance as it indicates the physical and

chemical characteristics of the MICs. (54)

The classic feature of the appropriate coating polymer is to be inert and chemically compatible with

the core material as forming a cohesive film with the core material is fundamental. In addition, the

coating material should provide the optimal layer properties such as strength, flexibility,

impermeability, optical properties and stability. (54)

For MEC, hydrophilic or hydrophobic polymers can be used. (33) When MEC process is used, as either

a taste masking or a taste concealing technique, the selection of the appropriate taste concealing

polymer is crucial. Vummaneni et al. (62) state that hydrophobic polymers are more popularly used

for coating of bitter drugs than hydrophilic polymers. Ideally, the taste concealing polymer should be

such that is prevents the release of API in the oral cavity, following per oral intake, but allows its

release in stomach.

Polymers, which are not soluble at salivary pH 6,8 but rapidly dissolve at gastric fluid (pH 1,2), play a

major part in taste concealing’ techniques. (63)

Coating agents used in microencapsulation are gelatin, povidone, HPMC, ethylcellulose, carnauba

wax, acrylics (different grades of Eudragit) and shellac. (63,64) From Ayenew Z. et al. (65),Sharma et

al (63) and Vummaneni et al. (62), it is evident that there have been a few successful studies how to

either mask or conceal a bitter taste of a drug by MEC using a Wurster fluid bed coating. However,

using hydrophilic methyl cellulose (MC), as a potential taste-concealing polymer, is fairly an

unexploded area.

1.4.4. Pediatric patients and taste concealing

Taste is one of the traditional five senses and is the ability to detect the flavor of substances, like

food, drugs etc. (62)Moreover, taste can surely refer to a subjective perception which arises from the

taste buds stimulus on the tongue’s surface. (65)

Human beings are able to differentiate among five types of taste: sourness, saltiness, sweetness,

umami (savory) and bitterness,(66) which is stimulated at the back of the tongue. (65)

In case of pediatrics and geriatrics, taste – masking and taste-concealing of a bitter drug might

represent a key role in the formulation development when unwillingness to swallow solid dosage

forms, is concerned. (62,64)

Page 24: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 24

Bar-Shalom (17) postulated that children have to be taught how to prevail their instinctive reactions,

and their organ development might prevent the swallowing of solid oral dosage formulations.

Moreover, Iwai N. (67) confirmed that more than half of the subjects in his study had certain

complications in taking the medicines.

In addition, the majority of troubles regarded taste, primarily bitter taste, followed by roughness on

the tongue, too large quantity and odor. Bitter taste was a major criticism within all age groups.

(67)It was reasoned that children are more sensitive to the bitter and unfamiliar taste of drugs than

are adults.(17)That is why it is prerequisite for the pharmaceutical industry to find new ways to

soften the bitterness of the drugs as the commercial success of the product depends upon the

patient compliance.

Taste masking is defined as the apparent reduction of an unpleasant taste by using suitable

agents.(68)It is, however, important to note that Bar-Shalom (17)proposed a distinction between

taste masking and taste concealing. According to his definition (17) taste masking represents the

addition of chemicals (sweeteners and flavors) to modify the taste of the medication, whereas taste

concealing approach implicates the coating of the drug to make it unavailable to the taste buds.

The major taste mask/conceal technologies yearn for a reduction of solubility of the drug in the saliva

hence the drug concentration in saliva will remain below taste threshold value. (63) These

technologies include two aspects: selection of suitable taste masking/concealing substance such

polymers, sweeteners, flavors, amino acids etc., as well as selection of suitable taste concealing

techniques, which can powerfully impact the quality of taste concealing and process effectiveness.

(68)

There are many techniques developed for, either taste masking or taste concealing of bitter drug, for

example the addition of flavoring and sweetening agents, ion-exchange complexation, inclusion

complexes, gel formation, emulsion techniques, granulation and microencapsulation or incorporation

of the bitterness inhibitor. (62)

For now, MEC seems to be the one method, among others, to conceal the taste of drug. (69) Fast-

hydrating dry granules which swell into pudding-like carriers have been developed for the

microencapsulated drugs to limit their exposure to the taste receptors. (17,69)That is why MEC has

been chosen as a taste-concealing technique in the present study.

Page 25: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 25

When the small drug particles are encapsulated by using a suitable polymer, which can reduce the

drug solubility in saliva, the core material (API) cannot contract to the taste buds in the mouth and

hence the unpleasant taste of bitter drug is concealed. (63,68) When the distasteful taste is avoided,

this results in the increase of the patient compliance. (64,68)

1.5. Pharmacokinetic barriers in anthelmintic action and the role of

dissolution

It is generally accepted that API has to be initially dissolved in the GIT’s aqueous medium in order to

be effortlessly absorbed. (70)

Within drug delivery, drug formulation and quality control, dissolution testing is an essential device,

primarily chosen for immediate release dosage forms. In addition, when the evaluation of

performance in vivo is questioned, the dissolution testing’s goal is to offer the rapid dissolution in the

test medium. (71)

In this thesis, ALB, an anthelmintic drug, was chosen in order to improve its release and,

consequently, attain elevated ALB concentrations where the intestinal parasites are located. This

should further permit the delivery of effective ALB concentrations in the worm’s cell and adequate

time to provoke its anthelmintic effect.

It is widely accepted that dissolution of the drug in aqueous content of GIT is a limited step which can

further provide absorption through the GIT and thus limit the amount of ALB for efficiently reaching

the bloodstream. (72) After the reversible exchange of ALB between the systemic circulation and

tissues, the entrance of ALB molecules should be able to penetrate into the parasite’s cells.(2)

That is why the drug delivery can only be efficient if the ALB molecule is capable to reach the

parasite’s structure in the anthelmintically effective concentrations. (73)

Therefore, the dissolution as rate-limiting step of ALB’s absorption, greatly influence the

concentration of poor soluble ALB, which enters the parasites.

In the last decade, the improvement of the solubility of poor water soluble drugs has been one of the

main targets of drug development. (74) Because the taste-concealing is a main target of the dosage

form, it is crucial to note that the taste-concealing aspect also greatly influences dissolution method

development, specifications and testing.(75)

The two most widespread dissolution tests (basket and paddle method) are based on the principle of

having a solid dosage form in a vessel filled with the dissolution medium that is agitated by a paddle

or by rotation of the basket. (76)

Page 26: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 26

1.5.1. BCS Class II Drugs and IVIVC

There remains a need of how to correlate in vitro drug release of numerous drug formulations to the

in vivo drug profiles. (70) The dissolution rate for in vivo prediction can only be improved when the in

vitro in vivo correlation (IVIVC) is fully settled. (71)

When it comes to the BCS Class 2 Compounds, as ALB, the drug dissolution could be the rate limiting

step for the drug absorption and IVIVC might be predictable. In this case, the dissolution critically

affects the bioavailability of BCS class 2 drugs which have very low water solubility and high human

intestinal permeability.(33) Consequently, even a diminutive increase in dissolution rate might result

in a great increase in bioavailability.(72,77) For that reason, the enhancement of the dissolution rate

of the drugs is a fundamental aspect when improving the bioavailability of BCS class II drugs is

preferred.

In this thesis, the improvement of ALB release rate on the basis of its encapsulation into polymeric

MIC would be a useful achievement.

1.5.2. Solubility

Solubility is a thermodynamic feature of a chemical substance, existing in a solid state, which takes

contact with a liquid solvent. (78)

As the result of BCS Class 2 drug’s poor water solubility, as in the case of the API - albendazole (ALB),

its oral delivery is a common challenge for pharmaceutical scientists. (79)

One of the aims in this thesis was to improve the drug release rate of ALB by its encapsulation into

polymeric MIC in order to see if it is possible to enhance the initial ALB release.

Several strategies have been considered to be beneficial for the enhancement of ALB aqueous

solubility including complexation with povidone and cyclodextrins (27,80,81) as well as using the

dendrimers as solubility enhancers for ALB. (23)

The use of microencapsulation as a technique to enhance the solubility of ALB is considerably

unexplored area and only a few studies have focused on this aspect; for instance, the preparation of

enteric microparticles of poorly soluble and bitter drugs (79) and hyaluronic microspheres of

cyclosporin A (82), both for the improvement of the ALB oral bioavailability. It is important to note,

however, that micronization in terms of the particle size’s reduction increases the surface area and

dissolution rate, but does not affect the solubility.(20)

Page 27: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 27

1.5.3. Taste-Concealing drives Dissolution

As previously described, taste-concealing of bitter drug substance is important for any orally-

administrated dosage form. However, it is important to note that the dissolution performance of

these dosage forms is influenced by their wettability, particle size determination and surface area.

(71)

Because granulation of the bitter drug, followed by a polymer coating, can achieve taste-concealing,

it is vital to understand that several polymer coatings are known for their pH-dependent solubility,

which is capable to influence the dissolution profiles.(75) In addition, the polymers can swell, dissolve

or become permeable throughout the dissolution; it is important to acknowledge that, however, the

dissolution medium is of imperative importance here. (75)

Because the taste-concealing is an essential element of the microencapsulated dosage form, a multi-

point profile in a neutral pH medium with early points of analysis (e.g. ≤5min) is recommended. Such

a dissolution criterion (typical ≤10% dissolved in 5min) primarily depends on the taste intensity of the

drug, but might, however, allow the in vitro estimation of the taste concealing characteristics. (71)

1.5.4. Dissolution rate

“Dissolution rate may be defined as an amount of drug substance that goes in the solution per unit

time under standard conditions of liquid/solid interface, temperature and solvent composition.”(83)

According to the Noyes-Whitney equation (20,83), the rate of dissolution may be modified primarily

by altering the surface area of the solid with the micronization. (84)

When designing a rapidly-dissolving dosage form, the formulation scientists want to omit even the

slightest possibility of delaying or prolonging the drug’s release value after the application.

It has been generally accepted that for immediate-release (IR) products, more than 85% of the API

must dissolve in half an hour (30 min), using either USP Apparatus I (basket) or USP Apparatus II

(paddle) in 900 ml of aqueous medium. (33,71)

For poorly soluble compounds, solubility in the conventionally used dissolution medium may be

insufficient. It has been suggested that this might be normal; however, if that’s the case, dissolution

tests should be carried out in more robust medium with various testing settings. If that what the

conditions are, the surfactants could be used in order to improve the drug’s solubility. (70)

Page 28: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 28

1.5.5. In vitro methods

According to the European (76) and US (83) Pharmacopoeia it is crucial to determine and measure

the solubility of both pure pharmaceutical ingredient and solid formulation, where the compounds

must be dissolved in a special vessel. The apparatuses that are foremost appropriate for performing

the dissolution test of solid oral dosage forms are Apparatus I (basket) and Apparatus II (paddle).

Both of these apparatuses basic equipments are positioned perpendicularly to the vessel. Dissolution

medium is placed into the vessel and its temperature could be further maintained at the required

level. (76,83)

When Apparatus I is concerned, the dosage form is positioned in a basket and the agitation is

achieved by rotating the basket, whether in Apparatus II, the dosage form is dropped straight into

the vessel. If this is the case, a paddle is used to stimulate the material’s movement with the usual

rotation speed, agitated commonly around 50-100 rpm. If the dosage form may float, Apparatus I is

more suitable. (76)

The pH of the dissolution medium is also an importing factor, considering which in vivo environment

is representing.

Dissolution testing is generally performed within the specific pH range. Normally that is from pH 1.2

(gastric pH) to pH 7.5 (intestinal pH). Even though purified water is commonly acceptable, it has no

buffering capacity that is why it is not an ideal medium. On the other hand, water is believed to be

the optimal medium if pH-sensitivity, either of a drug or excipients, is excluded from the dissolution

testing.(76,83)

1.6. HPLC Analysis

HPL Chromatographic separation is a technique, based on the distribution of a sample between two

phases. One phase is stationary, where the small surface-active solid particles are captured in a

chromatographic column, whilst the second phase – mobile phase, consists of a liquid that passes

through the first phase, as an eluent. (85)

Within the pharmaceuticals, High Performance Liquid Chromatography (HPLC) is one of the most

important analytical tools, frequently used for the quantitative and qualitative analysis of drug

substances and drug products. (85) Since the analytical methods are expected to be accurate and

reliable, it is fundamental to provide the high degree of assurance through the documented

evidence, known as the method validation. (85,86,87)

Page 29: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 29

In practice, scientists are keen on using RP-HPLC since its versatility, reliability and ease to use have

been thoroughly verified.(86) The term “reversed phase” (RP) has been applied as the first HPLC

separation, consisting of a polar stationary phase (SP) and a non-polar mobile phase (MP). In a

common RP-HPLC, the stationary and the mobile phrases are reversed. Normally, SP consists of

several silica particles, previously bonded with alkyl chains (for example C8, C18 columns)or phenyl

rings (known as phenyl columns),representing a non-polar entity. Typical RP-HPLC mobile phases are

water, methanol, or acetonitrile, making a mobile phase a polar constituent in RP-HPLC separation.

(85)

Retention of small and poorly-water soluble molecules by RP-HPLC strictly occurs when the

hydrophobic portion of the molecule reversibly absorbs to the bonded phase of the column and after

an increase in the concentration of organic modifier, the poorly water-soluble molecule is desorbed

and eluted from the column. (86)

1.6.1. Method Development for RP-HPLC

When developing a method, it is one of the main priorities to choose the most suitable stationary

and mobile phase in order to sufficiently retain and separate the molecules.(88) While the majority

of the RP-HPLC stationary phases (C8, C18) can form weak van der Waals interactions with the

molecules in the sample, the other RP-HPLC phases, for instance phenyl columns, are capable of

forming the different ones.

Nevertheless, while in the process of a RP-HPLC method development, it might be useful to try as

many different particle chemistries as possible. By doing so, the RP-HPLC separation can be

effectively optimized. (85,88)The suitable mobile phase also needs to be appropriately chosen. RP-

HPLC mobile phases usually consist of an organic modifier (methanol or acetonitrile) and water,

which usually contains a very small portion of an ionic additive as the acetic acid.

After the suitable stationary phase (column) and a mobile phrase system (organic and ionic modifier)

are reasonably chosen, the elution mode should be determined. This can be either isocratic, where

the composition of the MP is unchanged during the entire elution process or gradient elution with

the increasing strength of MP during the elution. (89,90) This can provide an optimal separation

within an adequate analysis time. In addition, isocratic elution is preferable as the development of

gradient elution can be found difficult. (86,89,90)

Page 30: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 30

1.6.2. Method Validation

In the pharmaceutical field, the analytical methods might be in widely in use; hence they must be

precise, reliable, accurate, specific and robust. To establish that kind of an analytical tool, there is an

important regulatory requirement in pharmaceutical analysis named “method validation” in which

the documented validation ensures that the material produced meets the required specifications.

(85,87,91)

Typically method validation parameters are dependent on the type and applications of the method.

In case of the identification tests, where the aim is to confirm the identity of the API in the samples,

the typically required parameters, according to the USP (83), are specificity, accuracy, precision,

detection limit, quantification limit, linearity and range. Other analytical tests often include other

parameters as system suitability and solution stability. (83)

2. AIM

A considerable number of experiments have proved that producing a pediatric drug formulation is a

unique challenge. The ideal medicine must be effective, stable, affordable, have a good palatability,

acceptable taste and smell.(8) However, some drugs crucial for pediatric patients are not

commercially obtainable in dosage forms suitable for use in this population. (9)

Hence, there are substantial benefits to be gained from developing an age-appropriate dosage form

since there is a growing concern that children are at risk when they are administered unsuitable

medicines.

For patient acceptability, oral drug delivery is the preferred route of administration. Nevertheless,

the main obstacle of oral drug delivery for babies, toddlers and children is difficulty or inability to

swallow.(9) Children do not like novelty in their food as well as its bitterness and, contrary to the

belief of the industry; they are not so fond of syrups. (15,67) It has been reported that the most

accepted consistency for children from 6 months to 5 years, is the semi-solid formulation (16),where

mastication is not necessary. That explains the ideal nature of such formulations for in the pediatric

field. For that vital reason, the potential development of age-adapted dosage form with its beneficial

taste-concealing properties for the oral administration is a redoubtable challenge for the formulation

researchers.(92)

Over the past years, masking unpleasant taste with sweeteners and flavors has been extensively

studied.(62,63,65,93)However, if this is not feasible, more sophisticated formulation approaches

such as encapsulation of drug particles are desirable. (94)

Page 31: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 31

What is more, the microencapsulation of drugs has been widely used in pharmaceuticals, but only a

few studies have used this technique in order to increase the drug release and bioavailability of poor

soluble drugs. (95) Nevertheless, the performance of formulation of poorly soluble drugs depends

not only on the feature of the manufacturing procedure used.

A close attention should be paid to the fact that uniform microparticles with control size distribution

play an essential role in the drug release of poorly soluble drugs. (96)Moreover, the types of

excipients, applied in a manufacturing process, are also of great importance. (24)

In this thesis, a poorly soluble drug, albendazole (ALB), which is very frequently used in the third

world countries as an anthelmintic (24), was used in order to microencapsulate. Therefore, the focus

of this thesis is to microencapsulate ALB the purpose to conceal its unpleasant taste, initially

decrease drug release in the mouth and on the other hand, enhance the drug release in the stomach,

all on the basis of ALB encapsulation into the polymeric microparticles.

The main aims in this formulation study are:

Achieve PSD of the granules in the range of 70-180 μm

Improving the drug release from the granules of poorly soluble drug ALB

Microencapsulating to obtain efficient taste concealing of the ALB

Allowing easy swallowing by integrating microparticles into an UNICEF gel, a HPMC based gel,

resulting in a production of a semi-solid formulation

Developing simple production process for the dosage form (primarily) appropriate for

children in the third world countries

Page 32: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 32

3. MATERIALS and METHODS

3.1. Equipment

Table 3: Apparatuses for manufacturing and analysis of MEC

These apparatus were used in manufacturing and analysis of MEC (Table 3, Figure 3 and Figure 4):

Figure 3: Modified Kenwood Food Processor Figure 4: Fluid bed MiniGlatt

Apparatus Process Manufacturer

Kenwood FP250 food processor Manufacturing the granules KENWOOD

Microscope Characterization Carl Zeiss

Malvern Mastersizer 2000 Particle size distribution Malvern Ins.

HPLC Analysis, separation Dionex

Paddle apparatus USP In vitro dissolution test ERWEKA DT70

Scanning Electron Microscope Characterization LEICA, S340

Ultrasonic Bath (GS13) Dissolving, degassing Qteck GmbH

Weight (AG 204 Delta Range) Weighting Mettler-Toledo

Oven Drying Lytzen

Meterlab pH meter pH measurements Radiometer analytical

Magnetic Stirrer Agitation Ika

Sieve No.75, No.125, No.180 USP Sieving Retsch

Microcentrifuge Centrifuging LabnetInternation

UV Imager Dissolution Test Actipix SD130, Paryatec Ltd.

Mass spectrometer (MS)

SingleQuad Characterization

Agilent Technology LC/MS 6150

MSD module connected to the

1200 series

Fluidized bed (Wurster) Coating Glatt GmbH, Germany

Page 33: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 33

3.2. Materials

These substances were used in manufacturing and analysis of MEC (Table 4 – Table 8):

Chemicals for granulation

Table 4: Substances for manufacturing the granules

Substances Batch number Manufacturer

API

Albendazole 620130311 Hangzhou Dayangchem Com, Ltd.

Caffeine powder 22234.187 VWR Prolabo

Excipiens

Avicel pH 101 61515033 Fagron

Povidone BCBJ4899V Sigma-Aldrich

MilIiQ Water / FARMA

Dextrin from corn SLBD3699V Sigma-Aldrich

Sucrose 22545001 Fagron

Ethanol 70% 1680766 Kemetyl

Maize starch 61515051 Fagron

Tween 80 (Polysorbate 80) BCBG4438V Sigma-Aldrich

Ciklodextrin Beta / Fagron

Lactosum 03455011 Fagron

PEG400 0072500 Fluka Analytical

KCl K42278904121 Merck

Croscarmellose sodium 432801 BASF

Chemicals for coating

Table 5: Substances for manufacturing the coated granules

Substances Batch number Manufacturer

API

Caffeine granules 2377500 Fagron

Excipiens

Methyl Cellulose (MC) Taste-Concealing RAD-27-01-SL-131 Dow Company

Glycerol 00344LH Sigma-Aldrich

Talcum 20365001 Fagron

MilliQ Water / FARMA

Page 34: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 34

Chemicals for the preparation of UNICEF gel

Table 6: Chemicals for the UNICEF gel

Substances Batch number Manufacturer

HPMC K250 RAD – 57-02-LL-555 Dow Company

Sucrose 22545001 Fagron

Vanillin 22529627 Fagron

MilliQ Water / FARMA

Chemicals used for HPLC analysis

Table 7: Chemicals for HPLC analysis

Substances Batch number Manufacturer Quality

Methanol / VWR prolab HPLC isocratic grade

MilliQ Water / FARMA demineralized

Acetic acid (glacial) 100% K42116163112 MERCK Ph. Eur.

Chemicals used for dissolution test

Table 8: Chemicals used in the dissolution test

Substances Batch number Manufacturer Quality

Hydrochloric Acid 37% 12E020010 VWR prolab Ph. Eur.

Na2HPO4 BCBD8825V Sigma-Aldrich Ph. Eur.

NaH2PO4 BCBG8983V Sigma-Aldrich Ph. Eur.

MilliQ Water / FARMA demineralized

Page 35: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 35

3.3. Methods

3.3.1. Characterization of ALB

3.3.1.1. Identification of ALB by LC/MS

In order to confirm that the main API is undoubtedly ALB, the specific identification test for the active

substance is essential.

The LC-MS analysis of ALB was performed as a part of the characterization of API. A mass

spectrometer with electro-spray ionization (ESI) interface was used for MS analysis. The analyte

infusion experiments were performed using a built-in syringe pump with the mass spectrometer.

The ESI parameters were as follows: ionization mode, positive; scan 100-300 m/z; fragmentor 70;

capillary voltage, 4.1 kV; drying gas temperature, 300°C; vaporizer temperature, 200 °C; dissolvation

gas flow rate, 400 L/h and cone gas flow rate, 20 L/h.

A stock solution containing 0,5mg/ml of ALB in methanol was prepared just before the analysis and

was put on the magnetic stirrer for 45min. Standard solution containing ALB was prepared from one

dilution from the stock solution, yielding final concentration of 1,20 µg/ml.

3.3.1.2. Particle size distribution

A qualitative analysis of drug particle size was carried out using optical microscopy, where a small

amount of each sample was placed on a slide.

A more detailed particle size analysis of the samples was performed using a Malvern Mastersizer,

where different pressures (from 0,6 to 1,6 bar) were used in order to classify the potential presence

of primary agglomerates and then break them. A small, but sufficient mass of samples were added

on the measuring cell and then the Mastersizer measured the volume percent of the particles by

laser ensemble light scattering.

The results are given as a volume median diameter D(0,5) which is the diameter where 50% of the

distribution is above; in case D (0,1), 10% of the volume distribution is below and in case of D(0,9),

90% of the volume distribution is below a given value.

In all cases, the obscuration (light intensity absorbed by the sample) was between 0,1-0,3, which is

known as an optimal and reliable interval for the measurements. (97)

Page 36: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 36

3.3.1.3. Determination of bulk, tapped densities and flow properties

Bulk and tapped densities of powders and granules have to be measured as a part of flowability

indicators as the powder and granule flow is one of the most important considerations in the

pharmaceutical industry manufacturing solid dosage forms. (69)(70)(71)(72)

According to ICH Guideline (72) determination of Bulk and Tapped Densities is a method to

determine the bulked densities of the powdered drugs under loose and tapped packing conditions

respectively. The bulk and tapped densities measurements were used to calculate the compressibility

index and Hausner ratio, from which the powder flow was further characterized. (72)(73) In the

equation below, V0 stands for the apparent volume and Vf for the final tapped volume after 1250

taps.

(

)

Both densities were calculated in accordance with the method described in Section 2.9.15. of

European Pharmacopoeia (74) and ICH Guidelines. (72)

100,0g of each powder and finished batch of the granules (50,0g in case of ALB and MC powder,

because 100 g of ALB has not fit into the 250ml cylinder as the powder density of ALB is too low)

were weighted and gently introduced into a graduated cylinder of 250ml. As the bulk density is

determined by measuring the volume of a known powder in a graduated cylinder, the unsettled

apparent volume V0was read and the bulk density was calculated by the formula m/V0.

The cylinder was put into the holder and 250, 500 and 1250 taps were carried out.

The volume after 500 and 1250 taps was read as V500 and V1250. Because difference between these

two measurements was less than 2 ml, more taps were not necessary.

Therefore, the tapped density was obtained after mechanically tapping a cylinder with the powder

sample until the apparent volume of the sample becomes almost constant. The tapped density was

calculated using the formula m/Vf where Vf is the final tapped volume after 1250 taps. The

determinations of bulk and tapped volume were done in triplicate.

The Carr’s compressibility index (CI, %) and Hausner ratio (HR) were calculated based on the

equations above and the ratios were be used to estimate the flow characteristics of the powders and

granules.

Page 37: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 37

3.3.1.4. Morphology determination of powders, granules and MICs

Morphology of all samples, used for produce the MICs, was observed by scanning electron

microscope (SEM) after the samples were mounted directly on to the SEM sample stub, using double

sided sticking tape, and then sputter-coated with gold to create electric conductivity on the surface

on the samples.

3.3.2. Microencapsulation (MEC)

Twelve different formulations (FOR1-FOR12) were prepared by granulation technique. One

formulation was chosen and further coating of MICs was then followed.

3.3.2.1. Granulation

The powders of drugs (ALB and CAF) were mixed together in the modified Kenwood Food Processor

FP250 with Avicel /Maize starch/Lactose/Sucrose/Dextrin/Ciklodextrin, and Croscarmellose sodium

(CCS) for 2 minutes at the lower speed. Then, a granulating liquid composed of PEG400/ Polysorbate

80 (Tween 80)/Potassium Chloride (KCl) or Povidone (PVP) in MilliQ Water (in FOR1-FOR5) or in

Ethanol (FOR6-FOR12) was slowly (with the rate of 1.1 – 3.5 mL/min, depending from formulation to

formulation) added at the same speed until granules were formed.

After all the granulation liquid was added, 7-9 min, powders were forming into granules for

approximately 6-7 minutes on lower speed, which was, for the first minute, manually increased to

the highest speed.

The formulations used in the granulation experiments are listed in Table 9. Each formulation was

granulated using the 1L bowl of the Kenwood at a batch size of 50 g. The polymeric binder was added

dry and its concentration was selected based on previous investigations. Granulation was stopped

several times to mix granules which had been trapped to the Kenwood wall and to collect samples

for the microscope. The samples were collected every 1-3 min to observe them under the

microscope whether the end point of granulation had been reached. Granulation was terminated

and end point was reached when a satisfactory granulation was observed – this was mainly after 1-2

minutes on granulation on the highest speed. The granules were then spread on a tray and left to dry

overnight (25°C for 12h). The granules were automatically sieved with SIEVES NO.180, NO.125 and

NO.75 and these sieve shakers were used to break the agglomerates, obtained after drying, into

smaller particles. The percentage of particles between 75-180 µm was then calculated and it was

almost in all cases above 70%. This fraction was used for the following studies.

Page 38: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 38

Table 9: Twelve different formulations were prepared in the granulation experiments

3.3.2.2. Coating

In the preliminary experiments, trial settings had been investigated and, as a model compound, CAF

granules (instead of ALB granules, previously produced in the granulating process) were used and the

experimental conditions for coating were optimized.

Preparation of coating suspension:

Methyl Cellulose (MC) was dispersed and dissolved in 1/3 of the whole amount of cold water, which

had been pre-heated to 40°C. The dispersion was continuously mixed on the magnetic stirrer till all

the particles were thoroughly wetted. No lumping was observed, so there was no incomplete wetting

of the individual powder particles. The slurry was formed and it was on the magnetic stirrer for 3

hours at 750rpm.

2Dextrin

3Ciklodextrin Beta

Name ALB CAF AVICEL STARCH SUCROSE DEX2 CIKDEX3 PVP TWEEN80 PEG400 KCl CCS V H20

( w / w % ) (ml)

FOR1 35 2 20 20 - - - 3 10 7 - 3 19

FOR2 38 2 25 22 - - - 3 8 - - 3 22

FOR3 38 2 20 20 - - - 7 10 - - 3 24

FOR4 35 2 24 16 - - - 5 10 - 5 3 15

FOR5 35 2 33 - - - - 5 10 7 5 3 10

FOR6 38 2 - - - 40 - - 15 - - 5 19

FOR7 38 2 - - - 35 - - 10 10 - 5 22

FOR8 38 2 15 20 - - - - 10 10 - 5 21

FOR9 33 2 - - 50 - - - 10 - - 5 20

FOR10 38 2 10 30 - - - - 15 - 5 21

FOR11 33 2 - - - - 40 - 10 - - 15 20

FOR12 33 2 - 10 30 - - - 10 10 - 5 21

Page 39: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 39

The rest of the water, previously cooled to 4°C was then added as cold water to lower the

temperature of the dispersion for complete solubilization, improved clarity and reproducible control

of viscosity. Once the dispersion had reached the temperature at which MC became water soluble,

the powder began to hydrate and viscosity increased.

Then Talcum and Glycerol were added and the spray suspension was stirred gently for 1 hour at

750rpm. After preparation, the solution needed to deaerate, so it was put on in the refrigerator

(T=4°C) overnight and it was used the next day. During the coating process, the coating solution was

continuously and gently mixed on the magnetic stirrer.

The composition of the coating suspension can be seen in the Table 10 below:

Table 10: the composition of the MC coating suspension

Ingredient Quantity (w/w %) Function

MC Taste Concealing 5 Polymer

Glycerol 1 Plasticizer

Talcum 2,5 Anti-tacking

Water 91,5 Diluent

The coating procedure:

For coating, 50g quantities of drug-loaded granules were used. Fluid Bed Coating was performed by

bottom spray technique in a Mini-Glatt fluid bed with a 2L chamber fitted with a standard Wurster

partition, using process conditions as seen in Table 11.

Table 11: Process conditions for coating in fluid bed

Before coating, the fluid bed was preheated to a

temperature of 40 °C. Atomization air flow was set to

0,29bar and then liquid feed flow with 0,5 ml/min

started until 10% w/w (solids) coating level was

reached. Bag filters were shaken for 10s every

minute.

There were some minor variations of the inlet air

temperature, but a steady temperature of 34 °C was

maintained after the initial requirement of excess

energy for heating of the substrate.

Process variable Value

Inlet air T (°C) 40

Liquid flow (ml/min) 0,5

Atomization air flow (bar) 0,29

Wurster height (mm) 1,5

Inlet air pressure (bar) 0,26

Batch size (g) 50

Start product T (°C) 22

End product T (°C) 28

Inlet air pressure (bar) 0,26

Page 40: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 40

The granules were coated to a 10% weight gain which is acceptable according to the taste-masking

guidelines in Evonik.(102) According to Alkire et al.(64), the coating should preferably constitute at

least about 10 percent by weight of the microparticle.

After the end point was reached, the MICs were allowed to dry for an additional ten minutes to drive

off any residual solvents. Initial weight of the granules was noted and during different intervals in

between coating, the weight of the coated granules was, yet, acknowledged and the coating

percentage was calculated as:

3.3.2.3. Determination of wall thickness of the MICs

As seen from the SEM Images, the microcapsules are uniform and spherical. Consequently, the wall

thickness of the microcapsules was determined by the method described by Si‐Nang, Luu, et al. (103)

using the equation:

( )

[( ) ( ) ]

Where, h is the wall thickness, is the arithmetic mean radius of the microcapsule, d1is the density of

core material, d2 is the density of the coat material and ‘p’ is the proportion of the medicament in the

microcapsules, which had been determined by Particle Size Mastersizer.

The density (δ in g/m3) of the core (d1) and coat (d2) material was also estimated from

measurements by the Particle Size Mastersizer, which determined the mean diameter (D) and

specific surface area (SA) of the uncoated granules and MC as the coat material. The density was

then calculated using the equitation:

For the mean diameter and mean radius, the values from Volume weight means [D 4.3] was used

according to Loveland et al. (97), when the most common mean value noted when using laser

diffraction is the volume mean or D [4.3]. The surface weighted mean D [3.2] can also be used, but it

is more typically used if the product is an aerosol or spray.

Page 41: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 41

3.3.3. Dissolution

Preparation of sodium phosphate buffer solutions pH 6,80 :

First, stock solutions of disodium hydrogen phosphate (Na2HPO4) and sodium dihydrogen phosphate

(NaH2PO4) were prepared, both at 0, 1 mol/L. 7L of buffer solutions at 0,1 mol/L were then prepared

at the pH 6,80 by mixing together 324,5ml Na2HPO4 and 375,9 ml NaH2PO4 and then diluting to a

final volume of 7L using Milli-Q Water. Buffer solutions were placed on the magnetic stirrer for 15min

and then in an ultrasonic water-bath to remove gases from the solution, because dissolved gases

could form bubbles which may change the results of the test. The pH of dissolution medium solution

was checked with a pH-meter and it was between 6, 75 – 6, 85 for each buffer solution.

Measurement of ALB release from MICs:

Because HPLC method was not suitable for estimation of ALB release from MICs, UV Spectroscopy

was used. In addition, the comparison between ALB release from granules by HPLC and Spectroscopy

was made, using a Student t-test.

3.3.3.1. ALB solubility estimations in the dissolution vessels

ALB powder (m=80mg) was introduced and incorporated into the 2,5 ± 0,1 g UNICEF gel just before

the test. The preparation of UNICEF gel is described below (Section 3.3.3.4.).

The dissolution was performed in 900ml medium consisted of phosphate buffer pH 6.8, using USP

dissolution apparatus I equipped with a basket which was operated at the speed of 75 rpm and

temperature of 37 ± 0,5°C.The amount of ALB dissolved was measured at 5min; 10min; 20min;

30min; 40min, 50min and 60min and was then determined with HPLC.

In further experiments, Tween80 was added into the dissolution medium of phosphate buffer pH 6,8

and ALB was incorporated into the gel. The amount of ALB dissolved was measured at 5min; 10min;

20min; 30min; 40min, 50min, 60min, 120min, 150min and 180min and was then determined with

HPLC. Each dissolution study was performed in triplicate and the following sample preparation

procedure was then the same as the one described in the Section “3.3.3.2.”

3.3.3.2. ALB release from granules and microcapsules

Accurately weighed granules of 200mg (equivalent to a specific amount of ALB and 4mg of CAF in

FOR1-FOR12) were used for dissolution studies.

Page 42: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 42

In case of MICs, accurately weighed MICs of 200mg (equivalent to 80mg of ALB in FOR6) were used

for dissolution studies. The dissolution was performed in 900ml dissolution medium Mili-Q® Water

(with the pre-checked pH of 6,9 ± 0,1) or 0,1M HCl with pH 1,2) using USP dissolution apparatus II

equipped with a paddle which was operated at the speed of 75 rpm and temperature of 37 ± 0,5°C.

The peak vessels were used as they reduce the hydrodynamic problems associated with the

traditional vessels by eliminating the unstirred region underneath the paddle. (104)

The amount of the drug dissolved was measured at the suitable time interval for every 5minutes in

30min time range for granules and at 5min; 10min; 20min; 30min; 40min and 60min for MICs and

was then determined with HPLC. After each sampling, the aliquots were replaced with the equal

volume of fresh dissolution medium to correct the volume change for calculation. The vessels were

covered for the duration of the test and the temperature was checked at suitable times. The volume

of each sample was 5ml and it was diluted with methanol up to 10ml. Before measuring on the HPLC,

the samples were centrifuged for 5 minutes on 13000 rpm (17900 x g) speed in order to eliminate

the undissolved particles present during the sampling.

Each dissolution study was performed in triplicate for granules, whether the ALB release from MICs

was studied in the six peak vessels.

3.3.3.3. ALB release from the granules and MICs in the semi-solid formulation

Accurately weighed granules and MICs of 200mg (equivalent to 80mg of ALB in FOR6) were used for

dissolution studies and incorporated into the 2,5±0,1 g UNICEF gel, just before the test. The

preparation of UNICEF gel is described below (Section 3.3.3.4.)

The dissolution was performed in 900ml of medium consisting of phosphate buffer pH 6.8, using USP

dissolution apparatus I equipped with a basket which was operated at the speed of 75 rpm and

temperature of 37 ± 0,5°C.The amount of the drug dissolved was measured at 5min; 10min; 20min;

30min; 40min, 50min and 60min and was then determined with HPLC. Each dissolution study was

performed in triplicate for granules+gel, whether the ALB release from MICs+gel was studied in the

six peak vessels. The following sample preparation procedure was then the same as the one

described in the Section “3.3.3.2.”

Page 43: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 43

3.3.3.4. Preparation of UNICEF gel

The mixture of 13g HPMC K250, 200g Sucrose and 0.2g of Vanillin was added into a transparent

pouch as seen in Table 12. Dry blending with sweetening agents such as sucrose was used as

dispersion method for the vehicles. Then 1L of Milli-Q Water was slowly added into a pouch, as seen

on Figure 5, which was continuously shaken and after 3 minutes a homogeneous viscous material

was obtained. The air bubbles were removed by a vacuum pump and the gel was prepared for the

further use. The single-dosing vehicle was further optimized by addition of effervescent agents in

order to achieve a fast gelation without shaking.(18)

Table 12: Excipients used for preparation of UNICEF gel Figure 5: The pouch with the UNICEF gel

3.3.4. HPLC Analysis

3.3.4.1. Analysis for method development

Apparatus:

The method development was performed with a Dionex auto sampler HPLC system using 20µL

sample loop. The detector was set at 273nm for CAF and 295nm for ALB and peak areas were

integrated by computer. The maximum absorbance values for CAF and ALB were determined by UV

Spectrum Measurement (Appendix 10).

Separation was carried out at ambient temperature using a Phenylhexyl column. A guard column

was also used to safeguard the analytical column. All the calculations concerning the quantitative

analysis were performed with external standardization by the measurements of peak areas.

Excipient Amount

[g] Purpose

HPMC K250 13,0 Fast hydrating polymer

Sucrose 200,0 Sweetener for taste masking;

separation and hydration of

polymer

Vanillin 0,2 Increased taste perception of

the Sucrose

Water 1000,0 Diluent

Page 44: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 44

Method development:

The mobile phase was chosen after several trials with methanol (+0,1% Acetic Acid [HAc]), methanol,

water, water (+0,1% HAc) and acetonitril in various proportions. A mobile phase consisting of

methanol (+0,1%HAc) and water (+0,1% HAc) (80:20) was selected to achieve maximum separation

and sensitivity of CAF and ALB.

A flow rate of 1.0 ml/min gave an optimal signal to noise ratio with a reasonable separation time.

Various columns are available for RP-HPLC and therefore different columns were studied. Among

reversed-phase C8 and C18 column, the Phenylhexyl column (150 x 4,6 mm) with 5 μm particle size

was chosen. It showed a good resolution, lower back pressure, excellent peak symmetry, sharp

separation peaks of ALB and CAF and the good degree of retention. In addition, retention times were

observed to be less than 4 min for ALB and CAF respectively.

Under these defined conditions, all peaks were well defined, without any tailing observed.

λ(Amaxalbendazol) = 295nm

λ(Amaxcaffeine) = 275nm

Preparation of mobile phases:

Water + 0,1% HAc:1ml of 100% acetic acid are added to 1000ml MilliQ water and then degassed in

ultrasonic bath for 15min

Methanol + 0,1% HAc: 1ml of 100% acetic acid are added to 1000ml Methanol and then degassed in

ultrasonic bath for 15min

3.3.4.2. Analysis for validation

Chromatographic conditions

HPLC analysis was performed by isocratic elution with a flow rate of 1.0 ml/min. The mobile phase

composition was methanol with 0,1% HAc and water with 0,1% HAc (80:20).

Volumes of 20 µL prepared solutions and samples were injected into the column. Quantification was

made by measuring at 273nm for CAF and at 295nm for ALB. The chromatographic run time was 5

min.

Page 45: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 45

Preparation of stock solutions:

A stock solution containing 0,5mg/ml of ALB in methanol was prepared just before the analysis.

100mg of ALB was dissolved in methanol in a 200ml volumetric flask and then the stock solution was

put on the magnetic stirrer for 45min.

A stock solution containing 0,05mg/ml of CAF in methanol was prepared just before the analysis.

5mg of CAF in methanol was dissolved in a 100ml volumetric flask and then the stock solution was

put on the magnetic stiller for 45min.

Linearity

Standard solutions containing ALB and CAF in methanol, yielding final concentrations of 5, 20, 40, 80,

120, 150 and 200 µg/ml for ALB and 1, 2, 4, 6, 8, 10 and 12 µg/ml for CAF were prepared from one

dilution from the stock solution. The samples were injected in triplicate and standard curve was

made.

Accuracy

Standard solutions containing ALB in methanol with concentrations 10, 30, 50, 100,140 and 180

µg/ml and for CAF with concentrations 3, 5, 7, 8, 9 and 11 µg/ml in methanol were prepared from

one dilution from the stock solution. The prepared solutions were injected 3 times. From the

respective area counts, the concentrations of ALB and CAF were then calculated using the equitation

obtained from the standard curve.

Precision

The precision of this method was checked by nine injections of ALB at concentration 25, 50 and 100

µg/ml and nine injection of caffeine at concentration 2, 6 and 10 µg/ml.

Ruggedness

The ruggedness of the HPLC method was evaluated by carrying out the analysis using a stock

solution, standard solutions (same as solutions described in “Precision”), the same chromatographic

system and the same column on two different days. The prepared solutions were injected 9 times at

three different concentrations.

Page 46: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 46

LOD and LOQ estimation

For estimating LOD and LOQ the “from the noise” approach was chosen. (105)

The noise magnitude was taken as an estimate of the blank standard deviation and then the noise

and signal were measured manually on the chromatogram printout.

The seven concentrations of ALB and CAF were prepared in concentration range from 0,5μg/ml –

0,04μg/ml. Based on 6 injection (V = 20 μL) of the same concentration, the average area was then

calculated. The concentration of the prepared sample has corresponded to LOD and the ratio

between the mean area and SD was 3,3.

Then the concentration of c (ALB) = 0,1μg/ml and c(CAF) = 0,5 μg/ml were chosen.

LOD and LOQ were calculated based on standard deviation of the response (SD) and the slope of the

calibration curve (slope) at levels approximating the LOD or LOQ, according to the below formulas:

3.3.4.3. Analysis for drug content estimation

100mg granules were weighted (which is similar to 35mg ALB in FOR1 and FOR4; to 38mg of ALB in

FOR2, FOR3, FOR6 – FOR8 and FOR10; and to 33mg ALB in FOR9, FOR11 and FOR12), crushed with a

mortar and pestle until a fine powder was obtained, then transferred into 20ml volumetric flask,

filled up with methanol to 20ml and put on the magnetic stirrer for 15min. The samples were put on

ultrasonic bath for 10 minutes to achieve breakdown and homogenization. 1ml of this sample

solution was diluted with methanol in a 10ml volumetric flask and the solution was stirred for 15min.

The samples were prepared in five replicates and then averaged. Before measuring on the HPLC, the

samples were centrifuged for 5 minutes on 13000 rpm (17900 x g) speed.

The same procedure was followed when MICs (coated granules) were estimated for ALB content. The

only difference was in number of replicates, which were 5 in case of granules; whereas for MICs they

were 10.

Page 47: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 47

This variation in the number of samples was done intentionally since smaller standard deviations

were needed when MICs were evaluated. Therefore, it was demonstrated that with 10 MICs

replicates, the results were reasonably representative.

3.3.5. UV Spectroscopy

Measurement of ALB absorbance spectrum

ALB and CAF absorbance spectra was measured after the samples were prepared by the same

method procedure as for the standard solution preparation of ALB and CAF, described in HPLC

Methodology Section.

Standard solution preparation and standard curve for ALB

A stock solution containing 0,25mg/ml of ALB in methanol was prepared by dissolving 12,5mg of ALB

in methanol in a 50ml volumetric flask and placed on a magnetic stirrer for 30min. Eight standard

solutions were prepared in concentration range from 45 µg/ml to 10 µg/ml from one dilution of the

stock solution in methanol. Solutions were further diluted because the Beer-Lambert Law is most

accurate between absorbance of 0,05 to 1,00. For example, the diluted (1:2) ALB solution (c=45

μg/ml) had absorbance 1,06, meaning that undiluted solution had an absorbance of 1,06 x 2= 2,12.

The standard curve for the spectrophotometric measurements was linear (R= 0,9985) under work

concentration interval.

The absorbance spectrum and standard curve for UV Spectroscopy measurements can be seen in

Appendix 10.

3.3.6. Student t-test

In order to compare the analytical results obtained with two different methods (HPLC and UV

Spectroscopy), from which HPLC is validated and confirm that UV Spectroscopy provide similar

analytical results, a statistical tool was used, known as a Student t-test. The data were obtained by

the HPLC and Spectroscopic determination of ALB release from uncoated FOR6 granules. The paired

t-test was made with a predefined confidence interval (CI) of 99%, using Microsoft Excel 2007 as an

analytical tool and the null hypothesis being that both methods provide the same analytical results

and that differences (if any) are purely due to random errors.

Page 48: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 48

4. RESULTS and DISCUSSION

4.1. Characterization of ALB

4.1.1. Identification of ALB by LC/MS

Liquid chromatography with mass spectrometer was used to characterize and thus identify the drug

Albendazole (ALB) that has been used throughout this thesis. The identification spectrums of ALB,

obtained by MS can be seen in Appendix 1. The mass spectrum showed a main peak at m/z 266.1

which is in good agreement with the ALB standard spectrum obtained by MassBank’s database. (106)

One can conclude that ALB was therefore determined and it can be further applied in this study.

4.1.2. Particle size distribution (PSD)4

Data obtained in previous studies suggested that reducing the particle size have a positive impact on

the dissolution rate (72,81,84,107), stability in drug formulation (108), efficacy of delivery (109),

children’s projection of texture and feel of the drug (67,110,111), appearance (67), flowability and

handling (70). One of the specific research problems was getting the PSD of the granules between

70-180 μm.

In this study, the evaluation of granule size distribution was accurately measured using Malvern laser

diffraction and results were obtained as the volume percentage of the material.

Both Figure 6 and Figure 7 represent symmetrical and narrow normalized distribution (volume

based) which show that the diameter of the granules and MICs ranged from 75 to 177 μm and from

134 to 340 μm, respectively. The term D(0,9) represents the particle diameter at which 90% of the

distribution is below 177 μm for granules and 340μm for MICs. As noted, there is a significant

difference between their particle sizes, resulting in the size enlargement due to the coat forming. As

the ALB particle size remains unchanged, reduction of the granules and MICs size increases the

specific surface area (and might enhance the drug’s dissolution). However, that does not affect the

solubility. In addition, the narrow distribution of granules is one of the crucial material parameters

for the coating process in the fluidized bed. (54)

However, in Appendix 2 can be seen that ALB particles of pure drug are noticeably small (>90%,

<38μm) and present non-symmetrical bimodal distribution with a wider PSD.

4 The whole analysis report from Malvern Mastersizer of granules, MICs and ALB powder can be seen in

Appendix 2.

Page 49: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 49

Additionally, the specific surface area of ALB particles is forty and seventy-five times larger than the

area of granules and MICs, respectively.

Mosharraf and Nyström(84) have proved that increase in particle size (which resulted in decreased

surface area) enhances the dissolution rate in case of poor-water soluble drug Griseofulvin.

Moreover, from a manufacturing perspective it is preferable to have taste-concealed MICs that are

larger in size (0,25-1 mm) due to the lower cost in the manufacturing process.(112)

In the Figure 6and 7it can be seen that is possible to achieve a micro sized product of poorly soluble

drug ALB with defined and narrow distribution range.

Figure 6: Granules FOR6 size distribution with the granules diameter range from 75 μm to 177 μm

Figure 7: MICs size distribution with the diameter range from 134 μm to 340 μm

Page 50: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 50

4.1.3. Determination of bulk, tapped densities and flow properties

As generally accepted, many powders do not flow well. When the material has poor flowing

characteristics, a greater interparticulate interactions and differences between the bulk and tapped

densities can be inspected. (100)That can be due to their small size, irregular shape or surface

characteristic. For that reason the microencapsulation, consisting of granulation and coating, should

improve the flow properties of powders and granules respectively. For the coating process, a good

product flow is an important characteristic, because it influences on the MIC quality. (51)

Before the flow properties were calculated (based on the equations explained in the Method section

and Appendix 3, where the average results for V0 as unsettled apparent volume (bulk volume) and Vf

as the final tapped volume of ALB, excipients and granules can be seen), the bulk and tapped volume

were measured as shown in Table 13.

Table 13: Calculated Bulk, Tap Densities and flow properties (CI and HR) of ALB, Dextrin powder, granules and MIC

Parameter Albendazole Dextrin MC GR FOR65

Bulk density (g/ml) 0,23 0,59 0,27 0,49

Tapped density (g/ml) 0,30 0,68 0,42 0,55

Carr’s index (%) (CI) 25 13,53 35,68 9,9

Hausner ratio (HR) 1,33 1,16 1,56 1,10

Flow character Passable Good Very poor Excellent

From the same Table 13 it can be demonstrated that the order of decreasing flowability, based on HR

and CI, is:

G r a n u l e s F O R 6 > D e x t r i n > A L B > M C .

While MC powder has very high HR and CI and thus poor flow character, dextrin has a good

flowability. One can claim that FOR6 granules have an excellent flowability as having the lowest CI

and HR. Its free flowing behavior is probably due to spherical particle shape, larger particle size and

narrow PSD.

According to the bulk and tapped values in the Handbook of Ph.Excipiens, (113), MC has HR of 1,68

and Dextrin of 1,14, which is slightly different than in this experiment.

5GR FOR6 = granules of formulation 6

Page 51: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 51

Nevertheless, dextrin flow properties, according to the previous reference, can still be described as

“Good”, where MC properties have superior flow in our experimental.

In literature (113) MC flow properties are described as “very, very poor” and as “very poor” in our

study. This might be due to the different particle size and particle shape of MC used in this report as

the flow is highly dependent on these values. Poor flow properties are caused by smaller particle

size, different particle shapes and its wide PSD. (41)

Despite this, a good approximation of flow properties of excipients was made in this experimental

study. Therefore it can be concluded that granulation greatly improved the flow properties of

powder mix, which should result in better quality of the final product after the fluidized bed coating.

4.1.4. Morphology determination of powders, granules and MIC

The surface of ALB, MC, DEXTRIN, FOR6 granules and coated granules were observed by using

scanning electron microscope (SEM) on two different magnifications.

Granules before and after coating were spherical as shown in Figure 8 and Figure 9, respectively, and

a smooth surface was found for both products. That is strongly related to the fact that children and

infants like more spherical and uniform particles than rough and uneven.(67) Moreover, the

roundness also resulted in the excellent granules flowability, as discussed previously. According to

expectations, the surface texture of the uncoated granules is less spherical than the coated surface

which confirms that the Wurster apparatus is valuable in achieving small spherical particles. (51) The

incomplete spherical shape of uncoated granules can be due to the granulation process or the

surface nature of dextrin powder and its insoluble behavior when adding ethanol as the granulation

liquid. Regarding this, from SEM Images in Appendix 14 can be concluded that dextrin balloon-like

particles have smooth surface which also resulted in their good flowability behavior, indicating that

dextrin is an appropriate material for the granulation process. However, in previous investigations

when using AVICEL pH 101 as a binder, more spherical and smoother granules were observed, which

Law et al. (114)also demonstrated in their study.

SEM Images also confirmed the enlargement of the particles after the coating process and

revealed potential holes on the granule’s surface, which may resulted from the manufacturing

process, either the granulation or coating. The holes on the surface can also indicate that the pores

had been formed throughout the granules. Those crevices could favor ALB release with enhancing

the inward water penetration in the drug release study. In addition, on the surface of the both

products, there are very few irregular particles, which have adhered to the polymer coat.

Page 52: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 52

It is possible, that ALB is not completely trapped into the dextrin/MC polymer which should be

further investigated as this may affect, not just the dissolution rate of ALB, but also the taste-

concealing properties of the formulation. This process of adhesion can also affect the liquid uptake

and therefore the hydration rate, swelling of the MC and finally, the ALB release from the eroded

surface to the dissolution medium.

Nevertheless, it can be concluded that dextrin gives good coverage of the core materials. This

resulted in the smooth and uniform surface morphology which is preferred for a further fluid-bed

coating. In addition, in Julian’s and Radebaugh’s patent (115), the granules with irregular shape and

broad PSD have led to poor taste concealing efficiency and varying dissolution of coated particles.

The SEM Images of powders (ALB, DEXTRIN and MC) are shown In Appendix 14 just for comparison. It

can be concluded that their particle size are smaller and thus their PSD is more heterogeneous as can

be seen in Appendix 2 and discussed before.

Figure 8: SEM images magnification series (x75and x1000 respectively) of the granules

Figure 9: SEM images magnification series (x35 and x750 respectively) of the MIC

Page 53: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 53

4.2. Microencapsulation

4.2.1. Determination of wall thickness of the MICs

Wall thickness (h) of the microcapsules was determined by the equation explained in the Method

Section and then, on the basis of the variables seen in Table 14, calculated as:

( )

[(

) ( )

]

In the coating process, the surface of solid particles is covered by a solid layer, where the coating

thickness is, according to Guignon et al. (38), generally between 0,01 and 0,05 mm with a size of

particles from 0,1 to 10 mm. The study of Pearnchob et al. (116) has demonstrated that the taste-

concealing efficiency is enhanced with increasing coating thickness, but on the other hand this can

significantly alter the ALB release rate.

Therefore, there is a need, in the art of taste-concealing formulation, to develop a coat which is not

too thick and, yet, not too thin. As the more round particles were obtained after MC taste-concealed

coating, this gives a fairly good thin layer around the granules and thus the better coverage of MICs.

Furthermore, a thin layer of MC, 29,5 is confirmed. This should achieve, according to the Guignon

et al. (38), sufficient taste-concealing properties of the microcapsules. Further dissolution studies

would confirm if the MC coat had altered the diffusion rate of ALB and consequently decreased its

release rate.

Material Core material

(uncoated granules)

Coat material (polymer - MC) MICs

Variables SA D d1 SA D d2 p SA

Values 0,055

m2/g

121.562

µm

0,897

g/cm3

0,108

m2/g

128,200

µm

0,433 g/cm3 0,37 113,48

µm

0,03

m2/g

Table 14: Measured and calculated variables for the determination of the microcapsules’s wall thickness

Legend: h = wall thickness, 𝒓 = arithmetic mean radius of the microcapsule, d1 = density of core material, d2 = the density of the coat material, ‘p’ = proportion of the medicament in the microcapsules. D = mean diameter, SA= specific surface area

Page 54: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 54

4.3. Dissolution

In Appendix 4 the numeric values of ALB and CAF release from the different granules’ formulations in

water are fully provided. Furthermore, in Appendix 12, the example of calculation for ALB release can

be seen.

4.3.1. Granules6

Graph 1: Drug release profiles of ALB formulations FOR1-FOR5 in water within 30 min time range

Graph 2: Drug release profiles of ALB formulations FOR6-FOR12 in water within 30 min time range

6In Appendix 4 and Appendix 5 the individual graphs of each formulation can be seen and thus, the ALB and also the CAF release can be

observed in more details.

0%

10%

20%

30%

40%

50%

60%

70%

80%

0 5 10 15 20 25 30 35

% d

rug

rele

ase

time (min)

% ALB release from FOR1-FOR5 in water

ALB in FOR1 ALB in FOR2 ALB in FOR3 ALB in FOR4 ALB in FOR5

0%

10%

20%

30%

40%

50%

60%

70%

80%

90%

100%

0 5 10 15 20 25 30 35

%d

rug

rele

ase

time (min)

% ALB release from FOR6 - FOR12 in water

ALB in FOR6 ALB in FOR7 ALB in FOR8 ALB in FOR9 ALB in FOR10 ALB in FOR11 ALB in FOR12

Page 55: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 55

The development of microparticulate formulation is hampered by the fact that ALB is very sparingly

soluble in water (app. 1μg/ml). It has been suggested that the enhancement of ALB release in the

water can be due to, for instance, the improvement of wetting and solubilization by a hydrophilic

carrier. Gilis et al. (117) showed that the incorporation of a poorly soluble substance into a

hydrophilic polymer results in excellent dissolution properties. In the preliminary investigations, a lot

of different hydrophilic materials have been used in the granulation procedure. Examples of such are

sucrose, dextrin, maize starch and microcrystalline cellulose (Avicel pH101).

Avicel pH101 was used as a polymer in seven different formulations out of twelve: FOR1 – FOR5,

FOR8 and FOR10, where the ALB release was the lowest for FOR5 (37,38% ± 0,80% in 30min) while

the highest release was achieved with FOR10 (88,47% ± 1,27% in 30min). In this study, the

improvement of ALB release rate was found to be greater from granules where Avicel and Maize

starch were used together with superdisintegrant Cross-Carmellose Sodium (CCS) and non-ionic

surfactant Tween80.

It was observed that lower concentration of Avicel and thus higher concentration of Maize starch

resulted in greater ALB release. This was confirmed with formulation FOR5 (having Avicel/Maize

Starch mass ratio of 2,5) from which ALB release was twice less than for FOR10 with the same ratio of

0,33. The reason for this occurrence can be found in different swelling mechanism of Avicel and

Maize starch with starch absorbing water via a capillary system, between and into the particles.(118)

Because Avicel can only absorb water into the capillary system, wetting of ALB is not easy and thus

lower ALB release rate has been noticed. Since the model drug Caffeine (CAF) was almost fully

released (above 91% in all formulations in 30 minutes as seen in Appendix 5) and ALB is not, it can be

suggested that ALB had been entrapped into the microcrystalline cellulose’s water insoluble chain,

which delay the wetting and leads to the poor dissolution. Although Avicel has been found as the

excellent binder, diluent and disintegrant in the wet granulation process (113), researches have also

proved its lack of disintegration. (118,119) They found that Avicel granules do not disintegrate and

drug release occurs by diffusion through an insoluble matrix. In addition, it is generally accepted that

unlike starch, Avicel cannot be wet granulated without losing some of its disintegration properties.

(113) In this dissolution study, this was confirmed with the high soluble drug caffeine (CAF) as its

lowest release of 91% ± 0,76% in 30 min was noted for FOR5. Therefore the lack of disintegration of

Avicel is an important issue for immediate drug release formulation.

The differences in release can be further explained by the diversity in ALB and CAF solubility since the

drug release mechanism depends primarily on the solubility of the drug.

Page 56: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 56

When the uncoated particles were exposed to the water, the liquid penetrated into the granules,

dissolved the both drugs and thus form a saturated solution (as long as undissolved drugs were

present). Therefore it is reasonable to expect a faster CAF release since it is far more soluble.

The other reason might be the difference in the mechanism of diffusion. Because CAF is 20 000x

more soluble in water than ALB (19,22), migration of CAF into the dextrin polymer during granulation

process is possible. Thus, faster dissolution of that drug is probable as the CAF particles might be

caught on the granule surface.

It has to be noted that a majority of studies and patents have confirmed that pellets prepared by

microcrystalline cellulose (MCC) show tendency to have a prolong drug release. (120,121,122,123)

On the other hand, researches have also found that combining MMC, CCS and Tween80 with poor

water soluble drug Curcumin, the disintegration of pellets was noted to be within 4 min.(124)

Moreover, Newton et al. (125) have proved that Tween80 is able to solubilize water-insoluble drugs.

On that basis, the Tween80 was chosen properly as this surfactant can absorbs onto the surface of a

hydrophobic ALB particle with its hydrophobic chain and thus promote ALB release.

Although it was found that release can be increased up to 80% when using Avicel and Maize Starch

with Tween80 and CCS, the ALB release was even higher when using dextrin as a carrier. In addition,

dextrin has the same general formula as starch, although it’s smaller and less complex. (113) On the

Graph 1 (look in Appendix 4 for the more detailed graph values) one can compare the ALB release

from the dextrin granules in FOR6 and FOR7. As it is presented, nearly 88% ALB was released in the

first 10 min, 93,15% ± 1,59% at 30 min and 95,04% ± 0,34% after one hour from FOR6. This

corresponds to the USP Guidelines for Immediate release formulation.(126)On the other hand, ALB

release from FOR7 was 60,00% ± 0,26% at 30 min. In the case of CAF, its release was at 30 min

98,31% ± 1,81% and 99,45% ± 1,57% for FOR6 and FOR7, respectively. Dextrin, together with

Tween80 and CSS, is obviously much more appropriate polymer for improving the ALB release rate

which can be due to the rapid solubilization of the polymer in the water.

Generally, the dissolution is explained by the rate of solid-solvent interaction leading to the

solubilization of the molecule and the diffusion rate of the solvated molecule into the water. Because

of dextrin’s polar O-H groups, water easily forms hydrogen bonds with polar O-H groups in the

polymer. What is more, the strong electrophilic carbamat group in ALB might spontaneously react

with the nucleophilic groups. After the contact, the water molecules solvate the Tween-solubilized

dextrin vehicle (with entrapped ALB) and then break the hydrogen bonds between the ALB-dextrin

complexes. As a result, a layer of dissolved dextrin and ALB is formed, promoting the ALB release.

Page 57: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 57

However, this may not be the case with Avicel, probably due to its poor disintegration properties.

(127)

When Cyclodextrin Beta was used in FOR11, 88,47% ± 1,27% ALB was released at 30 min and 93,06%

± 0,19% in case of CAF. This result can further confirm that dextrin can be a suitable polymer for

improving the apparent solubility of poor water soluble drugs. This occurrence might be interpreted

due to a change in the cyclodextrin confirmation, which may be unfolded and acted as a linear

dextrin. (127) Gharibi et al. (128)proposed that the nonionic surfactant’s head absorbs on the PVP

chain, which leads to an expansion of the polymer and results in its unfolding. However, as Tween80

acts as a nonionic surfactant, the unfolding from the cyclic to dextrin’s linear form should be further

investigated.

In case of FOR9 and FOR12, the sucrose was used as a binder. Nonetheless, sucrose was not found to

be as good excipient as the dextrin. From the Graph 2 (look in the Appendix 4 for more in-depth

view) it can be seen that ALB release at 30 min was 77,90% ± 1,45% and 66,04% ± 0,03% for FOR9

and FOR12, respectively. The CAF release was noted to be above 98% at 30 min in both formulations.

Comparing the ALB release from formulation with and without PEG400 (FOR1 in comparison with

FOR2; FOR8 with FOR3 and FOR10; FOR7 with FOR6 and FOR12 with FOR9), it can be concluded that

with PEG-formulations, ALB release is significantly lower. On both graphs, Graph 1 and Graph 2, it

appears that only 60,83% ± 1,57%; 60,00% ± 0,26%; 70,00% ± 1,25% and 66,04% ± 0,03% ALB release

was noted at 30 min with FOR1, FOR8, FOR7 and FOR12, respectively. In contrast with their parallel

formulations, without PEG, it can be found that ALB release is notably greater: 72,75% ± 2,51%;

71,35% ± 0,82% / 88,47% ± 1,27%; 93,15% ± 1,59% and 77,90% ± 1,45% in FOR2; FOR3/FOR10; FOR6

and FOR9, respectively. Thus, the ALB release rate has been found to be greater when only Tween80

was incorporated into the formulation FOR2, FOR3, FOR6, FOR9 and FOR10, respectively. This may

be due to the findings reported by Newton et al.(119,125) and Ghembremeskel et al. (129), when

incorporation of Tween80 as a non-ionic surfactant has lead to the more porous pellets with

increased drug release rate.

Apart from that, in the Jachowicz’s study (130), PEG400 was found as the carrier for increasing the

dissolution rate of several poorly water-soluble drugs. Moreover, Nicklasson and Alderborn(131)

have found the evidence that the PEG resulting pellets are less rigid than MMC pellets.

Nevertheless, the use of PEG400 in this study was found to decrease the ALB release from granules

and thus was not suitable for immediate release formulations. It may be reasonable to conclude that

the greatest difference has appeared between FOR6 and FOR7, 93,15% ± 1,59% and 60,00% ± 0,26%

at 30 min, where the enhanced ALB release is linked with FOR6 (formulation without PEG).

Page 58: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 58

It can be summed up that ALB granules containing PEG400 diminished the apparent solubility from

the particles. This effect was further investigated as the interaction between ALB and PEG can be

likely a reason since it was found by Cory et al., (132) Carvalho et al.,(133) Vecchio et al.,(134) and

Mura et al. (135) that PEG show interactions with some major APIs on the market.

In conclusion, 12 formulations with different excipients have been developed by the wet granulation

process. The ALB release was greatly improved when using dextrin polymer; nonetheless, the

incorporation of Tween80 and CCS allowed granules to disintegrate within a very short time, while

PEG400 was found to decrease the ALB release in the water. The results suggest that the dextrin

granules containing ALB can be a promising carrier for enhancing drug dissolution of poorly water

soluble drugs.

4.3.2. Microcapsules for taste-concealing7

In Appendix 6, the ALB and CAF release can be observed in more details.

Graph 3: ALB release from uncoated, coated FOR6 granules and coated FOR6 in UNICEF gel with ALB pure drug release

It was reasoned before that with the great improvement in the social standards of living, it is no

longer acceptable for drugs to have bitter taste.

Among various technologies, the easiest method is to add sweeteners, but this may not be effective

enough to conceal the drug’s bitterness. (63,64,65)

7Because the HPLC method was not suitable for ALB release estimation from coated microparticles, UV Spectroscopy was used.

Furthermore, the statistical t-test has been made between ALB release from granules by HPLC and Spectroscopy in order to confirm the

comparability of both methods.

0%

20%

40%

60%

80%

100%

0 10 20 30 40 50 60 70 80

%d

rug

rele

ase

time (min)

% ALB release from uncoated and coated FOR6 granules in comparison with coated FOR6 MICs incorporated in UNICEF gel in water

FOR6 uncoated granules Coated FOR6 Coated FOR6 in UNICEF gel Albendazole powder

Page 59: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 59

Therefore, microencapsulation with the methyl cellulose coating was made in order to avoid

albendazole’s unpleasant taste. In this regard, severe considerations must be made to prevent any

remarkable change in the immediate release kinetics after the formulation dissolves in the saliva and

goes to the stomach.

It is important to reiterate that if taste concealing (using MC as polymer coating in this study) is a key

aspect of the dosage form, a multi-point profile in a neutral pH medium with early point of analysis

(e.g. ≤5min) may be recommended. Dissolution criteria of ≤10% dissolved drug in 5 min may enable

the in vitro evaluation of the taste concealing properties. (71)

In order to evaluate whether the goal of improving the ALB release rate from uncoated and coated

granules was reached, in vitro dissolution profiles were compared with that of pure ALB powder. The

ALB dissolution in Milli-Q® Water (with the pre-checked pH of 6,9 ± 0,1) was very low. As can be seen

in Graph 3, only 7,80% ± 0,39% was dissolved in the first 30 minutes, whereas 93,15% ± 0,34% and

67,36% ± 1,19% of the ALB present in uncoated and coated microparticles, respectively, dissolved

within the same time range. Thus, a major ALB release from the microparticles was achieved.

However, the coating polymer methyl cellulose (MC) had retarded the drug release, resulting in

67,36% ± 1,19% ALB release at 30 min from the coated granules. In addition, ALB release was noted

to be 5,88% ± 0,42% and 15,31% ± 1,99% after initially 2 min and 5 min, respectively. Although this

not follows the taste-masked guidelines (71), the microparticles may sufficiently mask the bitterness

of ALB for the first 2 min.

The aforementioned theory explains that for the therapeutic effect it is essential to deliver a

sufficient amount of drug to the site of action, which can be hampered in the pediatric population

especially if the drug is bitter. (9,92,110,136) After my own observations and reports by the nurses at

the Pediatric Department in the Hvidovre Hospital in Denmark, the majority of parents solve this

problem by mixing the bitter drug with the food. Therefore, there is a need for efficient drug delivery

system as there is no proof whether the medicine interacts with food constituents.

Therefore, the coated MICs were incorporated into the UNICEF gel, a semi-solid carrier, having the

consistency of porridge. In the patent, granted by Cuff (136), it was reported that a drug delivery

system of porridge/pudding consistency can sufficiently help to mask the unpleasant taste. In

addition, the pudding formulation is also more acceptable to the children (136). After the

incorporation, the ALB release was, against our expectations, initially vaguely higher; it has reached

9,01% ± 1,44% and 19,45% ± 1,69% in 2 min and 5 min, respectively.

Page 60: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 60

It can be proposed that, even though this is not in accordance with the taste-masked guidelines (71),

the semi-solid formulation, with the incorporated MICs, may sufficiently mask the bitterness of ALB

for the first 2 min.

Hence, it could be clearly seen that the ALB release from the semi-solid formulation was faster than

from the MICs. The reason for this might be in the linkages between hydrophobic portions of MC

chains between the MICs and UNICEF-gel or in faster solubilization of MICs, initially promoted by the

gel.

However, the ability of the polymer coating to conceal the bitterness depends on various factors,

such as its permeability for ALB and water, mechanical stability, water-solubility and coating

thickness. (116) It was confirmed by Pearnchub et al. (116) that taste-concealing is efficiently

improved with increasing the coating thickness. In this thesis, the coating thickness was confirmed to

be 29, 47μm. For that reason, the future work should include the coat dimensions up to 50 μm in

order to provide better taste concealing.

It might be reasonable to propose that with greater coating levels of MC, the diffusion pathways for

water should also be increased. Once the microparticles come into the contact with the saliva in the

mouth, water leads to the swelling and dissolution of the coating polymer. While CAF, a water

soluble drug, is released by diffusion through a swollen polymer, ALB as a water insoluble drug is

released by erosion of the polymer. (116,137) Finally, ALB releases from MICs and causes the bitter

taste. Therefore, higher coating levels than 10% should be required when coating with MC as this

can result in more sufficient outcome in the taste concealing.

Although hydrophobic polymers have been popularly used for coating bitter drugs to achieve taste-

concealing (65), hydrophilic polymers may also provide good taste concealing. According to Sohi et

al. (93), Ayenew et al. (65), Sharma et al. (63) and Vummaneni et al. (62), there were few polymers

used for MEC in order to conceal the taste. The polymers are mainly used in the combination of

water soluble, like gelatin and water insoluble polymers like Ethyl Cellulose/Hydroxyl Propil Methyl

Cellulose. However, there were no microencapsulation studies which could affirm the sufficient

coating with MC so far.

To sum up, as the ALB release of the pure drug was only 7,80% ± 0,39% at 30 min, ALB presented in

the uncoated and coated MICs was 93,15% ± 0,34% and 69,36% ± 1,19% within the same time range.

Thus, an exceptional increase in ALB release was achieved in the both formulations. It was, however,

found that the coating polymer MC had somehow retarded the ALB release when compared to the

granules.

Page 61: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 61

When MICs were incorporated into the UNICEF gel, it was indicated that MICs would sufficiently

mask the bitterness of ALB in the first 2 minutes. This does not follow the guidelines, but knowing the

fact that poor taste masking is not due irregular shape, nor either broader PSD, the unsuitable factor

may be the MC. In addition, the coating levels should be increased for higher taste-concealing

properties as this might be in correlation with increased coat thickness. In future, care should be

taken when considering the appropriate coating polymer.

4.3.3. ALB dissolution studies 8

In Appendix 7, the ALB and CAF release can be observed in more details.

Graph 4: ALB release without and with the incorporation into the UNICEF gel

The analysis in Rogers et al. reviews (138,139) obviously showed that only 5% of all MICs references

till 2012 reported the use of methylcellulose (MC) to achieve MEC. No references were identified

where MC microcapsules were used for taste concealing, although the numerous preparation

technologies for MEC of core material have been reported. (138) However, there were a few studies

where MC was used as a polymer film for coating, but none of these include fluidized bed

coating.(138)

8In Appendix 8 more detailed information about ALB release in the gel at pH 6,8 can be seen. Moreover, another graph was

made in order to pinpoint the difference between ALB release with and without the UNICEF gel.

0%

5%

10%

15%

20%

25%

30%

35%

40%

45%

50%

0 20 40 60 80 100 120 140 160 180 200

dru

g re

leas

e

time (min)

ALB and ALB incorporated in the gel at pH 6,8 with the addition of Tween80

Albendazole Albendazole in gel Albendazole + Tween80 Albendazole + Tween80 + GEL

Page 62: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 62

Beyond the scope of this study, dissolution of ALB and ALB incorporated into the UNICEF gel can be

seen from the Graph 4. As noted before, the ALB release was higher when MICs were incorporated

into the gel. However, when ALB dissolution of a pure drug was plotted as a reference to the

dissolution of ALB in the UNICEF gel, it was clearly confirmed that UNICEF gel had altered the drug

release. As can be seen that 13,93% ± 1,87% ALB was dissolved in 120 min, whereas only 7,56% ±

1,27% at the same time when ALB was incorporated in the gel. In the later investigations, when ALB

was mixed with Tween80 and then incorporated into the UNICEF gel, it was found that ALB had

dissolved rapidly when Tween80 was added.

The data shows that when ALB+TWEEN80 was incorporated in the gel, just 34,50% ± 1,50% ALB had

dissolved, whereas 41,39% ± 1,46% ALB+TWEEN80 had dissolved without the gel incorporation. As

mentioned previously, Newton et al. (125) have proved that Tween80 is able to solubilize water-

insoluble drugs. This confirms that whilst the specific semi-solid carrier (UNICEF gel) slightly alters the

ALB release, Tween80 is the efficient solubilizer for promoting the drug’s release.

4.3.4. Microcapsules and granules/ MICs in 0,1M HCl and in MilliQ Water

In Appendix 8, the ALB and CAF release can be observed in more details.

Graph 5: ALB release from MICs and granules in 0,1M HCL and water

0%

20%

40%

60%

80%

100%

120%

0 10 20 30 40 50 60 70

% d

rug

rele

ase

time (min)

ALB release from microencapsules and granules in 0,1M HCl and in water

Microencapsules in 0,1M HCl Granules in 0,1M HCl Granules in Milli-Q Water Microencapsules in Milli-Q Water

Page 63: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 63

The objective of the present investigation was to conceal the bitterness of ALB by encapsulation in

microparticles and lower ALB release in neutral medium (in order to emulate the oral cavity) within

the first 5 min. The complete release of ALB should be then achieved in the stomach at much more

lower pH.

As outlined before in the Background section, once the coated microparticles reach the stomach, the

taste-concealed coat, consisting of MC polymer, should be rapidly dissolved or ruptured to assure

immediate release kinetics. However, it is generally accepted that when MC, being a cellulose ether,

is exposed to an aqueous medium, it undergoes fast hydration and chains relaxation to form a gel

layer, which control water uptake and therefore modify the drug release.(140,141) When polymer

begins to swell, the drug starts to dissolve and then diffuse out of the system due to the different

concentration gradients. (141)

Nevertheless, failure to form the gel layer may cause immediate drug release.(142) From the Graph 5

it is visible, that ALB release from MICs after 30 min, under acidic conditions, was only 71,26% ±

0,51%; whether from the granules it was 92,92% ± 1,54% at the same time, respectively. It appears

that when ALB from the coated granules was dissolved under acidic pH in stomach, the release was

slower in comparison with ALB release from granules under the same conditions. Consequently, the

gel layer was formed as the immediate release was not achieved. This might be due poor solubility

of ALB, as poor soluble drugs are primary released thought erosion of the gel layer. (141) In the case

of ALB, the release mechanism is dictated by the low solubility of the drug and thus dissolve and non-

dissolved drug coexist within the polymer matrix, whilst non-dissolve drug is not available for

diffusion/erosion. On the other hand, it could also be due to the inability of ALB to release from

microcapsules in the restricted fluid space. As the coated granules were exposed to the medium, the

water penetrated through MC’s coat and dextrin’s barrier into the microcapsules and dissolved ALB

to form a saturated solution. Therefore the non-dissolve ALB becomes entrapped inside the MC coat

which lowers its release.

Furthermore, the interactions between ALB and methyl cellulose polymer can also alter the barrier

properties of the coat. Nevertheless, this is less probable because the dextrin subcoating around ALB

was sealed before coating with MC.

The release of ALB from FOR6 granules was abundantly achieved within 30 min at acidic (92,92% ±

1,54%) and neutral pH (93,15% ± 0,34%), followed by a plateau as a result of the high porosity of the

granules, the hydrophilic nature of polymer, improved wettability, excellent PSD and spherical shape,

as noted in the previous sections.

Page 64: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 64

Graph 5 shows ALB release from uncoated and coated granules in 0,1M HCl, whilst their release in

water is plotted as a reference. From the results it is evident that after changing a dissolution

medium from water to 0,1M HCl, there were approximately no variations between percent of ALB

release from the uncoated granules. However, a slight difference in the slopes of these two curves

when comparing ALB release from MICs in water and in 0,1M HCl, can be observed in Graph 5.

It is interesting that ALB release from the coated granules in the acidic pH was 54,76% ± 1,71% and

61,02% ± 0,91%; whilst at the neutral pH was noted to be 15,31% ± 1,99% and 27,54% ± 1,36% after

5 and 10 min, respectively. Hence, the initial release after 5 and 10 min was 2-3 folds lower in the

water with the pre-checked pH 6,9 ± 0,1. However, ALB release in the acidic environment at 30 min

was 71,26% ± 0,51%, which is not radically higher than in salivary pH with 67,36% ± 1,19%.

Therefore, ALB release from the coated granules was initially influenced by pH of the release media.

As METHOCEL Dow Handbook suggests, MC is stable between the pH range of 2.0 to 13.0 (143). It

can be speculated that at pH less than 2, acid-catalyzed hydrolysis of the glucose-glucose linkages in

MC occurs, which significantly reduce the viscosity of the polymer. (113) Because the erosion plays

the key role in promoting drug release from poor soluble drugs, MC viscosity is one of the essential

factors in the poor water soluble drug’s release mechanism. (142,144,145) Thus, polymer chain

relaxation occurs rapidly at lower pH, leading to faster ALB release in the acidic environment than in

the neutral dissolution media.

As reported by Dashevskyet al. (146) many drugs, being weak bases (as ALB) demonstrate pH-

dependent solubility from the coated pellets. On the contrary, the release rate of ALB for the

uncoated granules was relatively unaffected by pH of the release media, suggesting that this must be

due to the MC coat which had prolonged ALB release in the water in the first 10 min. However, after

10 min, the coat starts swelling and releases ALB. This occurrence might be directly related to the

lower ALB solubility in water than in 0,1M HCl and, hence it could be explained by the pH-dependant

solubility of MC.

To sum up, it is evident that there were insignificant variations between the percent of ALB release

from the uncoated granules in different medium. However, when coated MICs were exposed to the

neutral medium, the ALB release was decreased. As the MC is expected to behave as insoluble at

salivary pH, it can be suggested that MC coat swells and erodes more slowly in neutral, than in the

acidic, environment. For this reason, the ALB release from the coated MICs was found to be MC

dependent.

Page 65: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 65

4.4. UV Spectroscopic measurements and t-test

Because the HPLC method was not suitable for ALB release estimation from MICs, UV Spectroscopy

was used. When comparing the measurements of the validated HPLC method with the Spectroscopic

method, the issue was if there had been a significant difference between the results of the same

samples. Since Bland and Altman(147) showed that plotting a difference between measurements

obtained by two methods against a validated method is misleading, the more accurate Student t-test

was made.

T-tests are commonly used in health studies to determine differences between two paired groups.

The significance level (or p value) is used to quantify how probable it is that the null hypothesis is

true. (148) In this comparison, a p value of 0,01 was used, indicating that there is 1% chance that the

null hypothesis could be mistakenly rejected.

In this study, the null hypothesis is that both methods provide the same analytical results. As seen

from Appendix 11, the P two sided experimental value is 0,0126, which is more than the tabulated

value of P= 0,0100, indicating that the null hypothesis can be accepted at the 99% confidence

interval. As the null hypothesis is accepted, we can conclude that the both methods provide the

same analytical results with 99% probability that our conclusion is correct.

4.5. HPLC Analysis

4.5.1. Analysis for validation

Linearity

Tables in Appendix 9.1 presents triplicate concentrations of ALB (λ=295 nm) and CAF (λ=273nm) and

their measured areas. Good linearity was obtained for ALB with equitation Y= 0,8248x + 0,785 (r2

=0,9997) with standard deviation (SD) of 0,2% , relative standard deviation (RSD) for slope 0,24% and

for intercept 4,33% and for CAF with equation Y= 1,0648x-0,2223 (r2 =0,9993) with standard

deviation (SD) of 0,72%, relative standard deviation (RSD) for slope 0,68% and for intercept 6,72%.

Therefore, the method was found linear over the concentration range 5 – 200 μg/ml for ALB and 1 –

12 μg/ml for CAF and the regression coefficients are for both drugs ≥0,999, which follows the ICH

guidelines. (149,150)In Appendix 9.2 and Appendix 9.3, the chromatogram peaks for ALB and CAF can

be seen.

Page 66: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 66

y = 0,8248x + 0,785 R² = 0,9997

0

20

40

60

80

100

120

140

160

180

0 50 100 150 200 250

Are

a (m

Au

*min

)

conc. (µg/ml)

Standard curve for albendazole

y = 1,0648x - 0,2223 R² = 0,9993

0

2

4

6

8

10

12

14

0 2 4 6 8 10 12 14

Are

a (m

Au

*min

)

conc. (µg/ml)

Standard curve for caffeine

Accuracy:

Accuracy is given in terms of % deviation of the calculated concentrations, an average-measured

concentration ± SD and RSD, which according to the FDA guidelines is inside the recommended limit

(<2%). However, following ICH Guidelines, the accuracy criterion is the mean recovery of 100 ± 2%

over the range of 80 – 120% of target concentration over three concentration levels at three

replicates. (150) In this study, data were collected over five concentration levels with three replicates

at each level. The % of deviation for ALB and CAF are below 2%, as well as RSD and from this is

evident that the test results are close to the true value. Therefore, the method is accurate within the

desired range.

Graph 7: Standard curve for CAF

Graph 6: Standard curve for ALB

Page 67: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 67

ALBENDAZOLE:

Spiked conc.

(µg/ml) Area 1 Area 2 Area 3

Average

Area

Measured conc.

(Mean ± SD)

RSD

%

Deviation

%

30 26,075 26,166 25,731 25,99 30,56 ± 0,23 0,88% 1,87%

50 43,065 42,837 42,549 42,82 50,96 ± 0,26 0,60% 1,92%

100 83,529 84,237 82,651 83,47 100,25 ± 0,79 0,95% 0,25%

140 118,332 117,883 119,363 118,53 142,75 ± 0,76 0,64% 1,96%

180 149,741 146,986 144,857 147,19 177,51 ± 2,45 1,66% 1,38%

% Deviation= (Spiked Concentration – Mean Measured Concentration) x 100 / Spiked Concentration

CAFFEINE:

Spiked conc.

(µg/ml)

Area

1

Area

2

Area

3

Average

Area

Measured conc.

(Mean ± SD) RSD %

Deviation

%

5 5,203 5,137 5,276 5,21 5,10 ± 0,07 1,34% 1,95%

7 7,149 7,043 7,121 7,10 6,88 ± 0,05 0,77% 1,70%

8 8,105 8,109 8,164 8,13 7,84 ± 0,03 0,41% 2,00%

9 9,299 9,285 9,387 9,32 8,97 ± 0,06 0,59% 0,39%

11 11,325 11,536 11,13 11,33 10,85 ± 0,20 1,80% 1,37%

% Deviation= (Spiked Concentration – Mean Measured Concentration) x 100 / Spiked Concentration

Precision:

The precision is expressed as the RSD % and the results can be seen in tables below. ICH (150)

recommends that the instruments’ precision (RSD) is ≤1%. In this study, precision of the technique

was evaluated through the repeatability by assaying nine replicate injections of ALB and CAF at three

different concentration levels. The RSD value for the retention times and peak areas are found to be

≤0,40% for ALB and CAF, respectively.

9.5.2013 for ALBENDAZOLE

Table 15: Precision results from the nine replicate injections of ALB with their mean tr and Area with the calculated RSD value

conc.ALB[μg/ml] tr 1 tr 2 tr 3 tr 4 tr 5 tr 6 tr 7 tr 8 tr 9 Mean tr RSD

%

25 2,667 2,663 2,655 2,657 2,65 2,645 2,645 2,647 2,643 2,65 0,32%

50 2,685 2,675 2,672 2,673 2,667 2,67 2,663 2,67 2,665 2,67 0,24%

100 2,693 2,695 2,703 2,678 2,697 2,675 2,69 2,678 2,688 2,69 0,36%

conc.

ALB[μg/ml] Area 1 Area 2 Area 3 Area 4 Area 5 Area 6 Area 7 Area 8 Area 9

Mean

Area

RSD

%

25 17,197 17,129 17,082 17,116 17,154 17,134 17,111 17,151 17,196 17,14 0,22%

50 37,461 37,462 37,441 37,464 37,48 37,466 37,518 37,513 37,514 37,48 0,08%

100 78,672 78,677 78,645 78,621 78,54 78,689 78,568 78,658 78,7 78,64 0,07%

Page 68: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 68

9.5.2013 for CAFFEINE

Table 16: Precision results from the nine replicate injections of CAF with their mean tr and Area with the calculated RSD value

conc. CAF

[μg/ml] tr 1 tr 2 tr 3 tr 4 tr 5 tr 6 tr 7 tr 8 tr 9 Mean tr

RSD

%

2 2,003 2 2,012 2,01 2,003 2,015 2,007 2,002 2,003 2,01 0,26%

6 2,007 2 2,017 2,017 2,008 1,998 1,993 2,002 2,005 2,01 0,40%

10 2,012 2,01 2,003 2 2,01 2,007 2,015 2,013 2,01 2,01 0,24%

conc. CAF

[μg/ml] Area 1 Area 2 Area 3

Area

4 Area 5 Area 6 Area 7 Area 8 Area 9

Mean

Area

RSD

%

2 1,985 1,984 2 1,992 1,999 1,999 2,005 1,989 2 1,99 0,37%

6 6,391 6,398 6,399 6,415 6,405 6,405 6,407 6,419 6,414 6,41 0,14%

10 11,139 11,132 11,208 11,21 11,163 11,154 11,159 11,151 11,143 11,16 0,25%

Ruggedness:

Ruggedness, expressed as the intermediate precision (inter-day variation) is the result from within

lab variations, due to different analyzing days. (91) Ruggedness criterion is expressed in RSD and it

should be, according to the ICH, less than 2%. (150)

As seen in the Table 17 and Table 18, an RSD of less than 1,52% for ALB retention times and less than

2,03% for ALB areas were obtained. For CAF, RSD for areas was less than 0,95% and for retention

time less than 0,38%. Nevertheless, the experimental values are not radically higher than

recommending values, which indicates that the method is capable of producing results with high

precision on different days.

10.5.2013 for ALBENDAZOLE

Table 17: Ruggedness results from the nine replicate injections of ALB with their mean tr and Area with the calculated RSD value

conc. ALB[μg/ml]

tr 1 tr 2 tr 3 tr 4 tr 5 tr 6 tr 7 tr 8 tr 9 Mean tr RSD

%

25 2,795 2,715 2,695 2,693 2,677 2,68 2,667 2,663 2,668 2,69 1,52%

50 2,78 2,703 2,702 2,705 2,69 2,673 2,682 2,678 2,673 2,70 1,23%

100 2,773 2,733 2,733 2,738 2,722 2,71 2,717 2,717 2,705 2,73 0,75%

conc. ALB[μg/ml]

Area 1 Area 2 Area 3 Area 4 Area 5 Area 6 Area 7 Area 8 Area 9 Mean Area

RSD %

25 13,568 13,578 13,593 13,197 13,217 13,89 13,854 13,532 13,147 13,51 2,02%

50 30,664 29,796 29,538 29,941 29,18 29,66 29,03 29,329 30,742 29,76 2,03%

100 78,375 77,335 76,318 77,99 75,527 77,634 75,568 75,673 77,862 76,92 1,49%

Page 69: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 69

10.5.2013 for CAFFEINE

LOD and LOQ determination:

The LOD and LOQ of the method were determined by calculating the signal to noise (S/N) ratio of 3:1

and 10:1 respectively. LOD and LOQ values for ALB were estimated to be 0,085 μg/ml and 0,26μg/ml,

respectively and similarly LOD and LOQ values for CAF were estimated to be 0,38 μg/ml and 1,16

μg/ml, respectively.

In Appendix 9.4 – 9.7., the chromatograms with LOD and LOQ for ALB and CAF can be observed.

A simple and rapid isocratic RP-HPLC method was developed for the determination of ALB and CAF.

The method was validated for various statistical parameters, including linearity, accuracy, precision,

and ruggedness and LOD/LOQ estimation. It is found to be linear, precise, accurate, and therefore

reliable for the estimation of ALB and CAF. It has runtime of 5 min which allows analysis of large

number of samples in a short period of time. This method can be used for further determinations.

Table 18: Rugedness results from the nine replicate injections of CAF with their mean tr and Area with the calculated RSD value

conc. CAF [μg/ml]

tr 1 tr 2 tr 3 tr 4 tr 5 tr 6 tr 7 tr 8 tr 9 Mean

tr RSD

%

2 2,02 2,023 2,022 2,015 2,018 2,022 2,008 2,017 2 2,02 0,38%

6 2,022 2,012 2,02 2,017 2,027 2,018 2,015 2,013 2 2,02 0,38%

10 2,017 2,017 2,025 2,013 2,017 2,007 2,017 2,017 2,008 2,02 0,27%

conc. CAF [μg/ml]

Area 1 Area 2 Area 3 Area 4 Area 5 Area 6 Area 7 Area 8 Area 9 Mean Area

RSD %

2 22,757 22,919 22,919 23,393 22,976 22,965 22,931 23,234 23,34 23,05 0,95%

6 70,552 70,738 70,738 71,282 70,811 70,68 70,898 71,159 71,838 70,97 0,56%

10 122,396 122,644 122,644 123,403 122,086 122,188 122,654 122,687 122,9 122,62 0,32%

Table 19: Results from the six replicate injections of ALB and CAF with their mean Area, LOD and LOQ

Area1 Area2 Area3 Area4 Area5 Area6

Average Area

SD LOD

[μg/ml] LOQ

[μg/ml]

ALB 0,863 0,467 0,769 0,861 0,405 0,861 0,704 0,212 0,0848 0,257

CAF 0,567 0,655 0,606 0,298 0,604 0,332 0,510 0,154 0,382 1,16

Page 70: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 70

4.5.2. Analysis for drug content estimation9

Drug content estimation is an important factor when determining the incorporation efficiency and

thus, the MICs quality. From Appendix 13, it can be seen that ALB encapsulation efficiency was in

FOR1 – FOR3, FOR6 and FOR8 – FOR10 above 95%, while in all other was noted to be above 80% with

the exception of FOR12, when the efficiency was above 100%. In addition, the CAF granulation

efficiency was also in most cases above 90% as seen in Appendix 13.

From the results in Table 20, it can be observed that ALB content was 96,92 ± 0,35% and 92,43 ±

0,74% for uncoated and coated granules respectively. It is essential to note that the main purpose of

this estimation study was not investigating the reasons for either low or high encapsulating

efficiency. Therefore, the data obtained from this study was strictly used for the correct estimation

of ALB and CAF release in dissolution studies.

Nevertheless, due to immensely entrapment efficiency, especially in FOR6, it can be suggested that

the granulation and the coating method, done by food processor and fluid bed, respectively, are

suitable for the preparation and estimation of ALB microcapsules. In addition, the high efficiency can

also be due interactions between the core and the coat material, as reported by Boury et al. (151) On

the other hand, Crotts et al. (152)indicated that these interactions can, thus, limit the drug release

from the microparticles. Johansen et al. (153) proposed that a co-encapsulated excipient can

mediate the interactions between the core and coat material. Therefore, this might explain the

slighter lower ALB efficiency value in coated granules when compared to the uncoated ones.

To sum up, the entrapment efficiencies were, in both formulations, unprecedentedly high. Since it is

evident from the dissolution studies that MC delays the ALB release, the interaction between MC

polymer and ALB can be reasonably suggested. However, the variation between the granulation and

coating efficiency is not significant, which can be due to the different excipients used in the both

production processes.

9In Appendix 11, the example of the calculation for the estimation of ALB drug content was made and the efficacy of the

granulation process can be predicted. These calculations were used for the calculations of drug release in dissolution

studies.

Table 20: Albendazole encapsulation efficiency for granules and microcapsules

ALB content EFFICACY SD

FOR6 granules 96,92% 0,35%

Microcapsules (coated granules) 92,43% 0,74%

Page 71: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 71

5. CONCLUSION

The use of microencapsulation as a technique to conceal the albendazole’s bitter taste and enhance

the solubility is a considerably unexploded area. Hence, this study is among the first of its kind. It was

the initial attempt to provide the insights into the improvement of ALB release rate on the basis of its

granulation in dextrin microparticles, further fluid bed coating with methyl cellulose and, finally, their

incorporation into the semi-solid formulation, known as UNICEF gel.

The surprising outcome of the research lends support to the belief that it is achievable to attain the

micro size product of poorly soluble drug ALB with defined and narrow distribution range, where the

granules and MICs size range from 75-174 μm and 134-340 μm, respectively. According to the

measured bulk and tapped density values, granulation in Kenwood Food Processor greatly improved

the flow properties of the powder mix and 12 formulations, all with various excipients, had been

designed.

For the dissolution studies, as well as for the entrapment efficiency studies, it was fundamental to

have a linear, precise, accurate and reliable determination method for the quantification of ALB.

Therefore, a simple and rapid isocratic RP-HPLC method for the estimation of ALB (and a model drug

CAF) was developed and validated.

After the dissolution studies, using USP Apparatus II, the ALB release has been greatly improved

when using a dextrin as a granulation polymer. From the SEM Images, it can be further concluded

that dextrin gives a good coverage of the ALB, resulting in the smooth and uniform surface

morphology which has been preferred for fluid bed coating. What's more, the incorporation of

Tween80 and CCS allow granules to disintegrate within a very short time, while PEG400 has been

found to decrease the ALB release in the water. After the taste-masked MC layer of 29,5μm had been

formed in the fluid bed onto the granules, even more round MICs were obtained. As the ALB release

of the pure drug was only 7,80% ± 0,39% in water at 30 min, ALB presented in uncoated and coated

MICs was 93,15% ± 0,34% and 67,36% ± 1,19% within the same time range. Thus, an exceptional

increase has been achieved in the both formulations. Nonetheless, the coating polymer MC had

slightly retarded the ALB release from MICs when compared to the granules.

When MICs were incorporated into UNICEF gel, it was found that MICs could sufficiently conceal the

bitterness of ALB in the first 2 minutes. This, however, does not follow the taste-masked guidelines,

which suggest a broader time range of 5min. Furthermore, it was confirmed that while UNICEF gel

slightly alters the ALB release, Tween80 is the efficient solubilizer for promoting its release.

Page 72: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 72

It was also demonstrated that with the dissolution media change from the neutral media to 0,1M HCl

(pH 1,2) the ALB release from the MC-coated MICs was initially influenced by the pH environment.

Conversely, this occurrence does not happen with the uncoated granules.

Therefore ALB release was found to be MC dependent. As the immensely high entrapment

efficiencies were confirmed in granules and MICs, 96,92% ± 0,35% and 92,43% ± 0,74%, this might

also offer an understanding why MC delays the ALB release. As reported by Boury et al. (151),

Crotts&Gwan-Park(152) and Johansen et al.(153), the interactions between the core and coat

material could be the reason for the high entrapment efficiency.

To sum up, the novel results provide striking evidence that it is feasible to attain the round and

uniform granules of poorly soluble drug ALB for the immediate release formulation. In addition, the

dextrin granules, containing ALB, with the incorporation of Tween80 and CCS, can be a fruitful and

promising carrier for enhancing the drug dissolution of poorly water soluble drugs. In case of MC,

there is an overwhelming need to optimize the level of coating in order to completely conceal the

taste of a bitter ALB while not adversely affect the immediate drug release profile.

Page 73: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 73

6. FUTURE RESEARCH SUGGESTIONS

A number of open problems need to be solved in order to allow the microencapsulation

development. Hence, some suggestions and a few possible research directions would be made in

order to provide the future steps along the microencapsulation of poor soluble drugs, particularly

albendazole.

One such direction would be to investigate the rupturability of a microparticle, as the SEM Images

clearly suggested there are a few visible cervices on the MIC’s surface. In order to get the

quantitative effect of the above mentioned limitation, the rupturability/mechanical robustness can

be measured on a practical basis, by subjecting the microcapsules to, either a chewing test, or a

texture analyzer.

The idea of albendazole not being completely trapped into the dextrin/MC polymer was addressed in

the Section 4.3. This might affect, not just the dissolution rate of ALB, but also the taste-concealing

properties of the formulation. Therefore, this suggestion is certainly the one that would need further

investigation.

It would also be preferable to optimize the level of coating by coordinating the right type of the

coating material in order to completely conceal the taste of a bitter drug while at the same time, not

adversely affect the immediate drug release profile.

Another possibility would be to form a multiparticulate system, using different anthelmintic drugs as

the resistance to single active antihelmintic is nowadays an arising problem. However, several

stability studies should be done beforehand.

Page 74: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 74

7. REFERENCES

1. Vercruysse, Jozef, et al. Is anthelmintic resistance a concern for the control of human soil-

transmitted helminths? International Journal for Parasitology: Drugs and Drug Resistance. 1.1, 2011,

pp. 14-27.

2. Muller, Ralph.Worms and human disease. Wallingford : CABI publishing, 2002. 0-85199-516-0.

3. Bentwich, Zvi, et al. Can eradication of helminthic infections change the face of AIDS and

tuberculosis? Immunology Today. 20.11, 1999, pp. 485-487.

4. Krause, Julia and Breitkreutz, Jörg. Improving drug delivery in paediatric medicine. Pharmaceutical

Medicine. 22.1, 2008, pp. 41-50.

5. Black, Robert E., et al. Global, regional, and national causes of child mortality in 2008: a systematic

analysis. The Lancet. 2008, pp. 1969-1987.

6. The World Bank.School deworming at a glance. Washington DC : The World Bank, 2003.

7. WHO Deworming: the Millennium Development Goals. 2005. p. 12.

8. Nahata, Milap C; Loyd Vr, Allen Jr. Extemporaneous drug formulation. Clinical therapeutics. 30.11,

2008, pp. 2112-2119.

9. Costello, Ian, ed.Paediatric drug handling. s.l. : Pharmaceutical Press, 2007.

10. WHO.Promoting Safety of Medicines for Children. Geneva : World Heatlh Organization, 2007.

11. Michele, Theresa M., et al. Safety of chewable tablets for children. Journal of asthma. 39.5, 2002,

pp. 391-403.

12. Cherukuri, Subraman Rao.Rapid-melt semi-solid compositions, methods of making same and

methods of using same. No. 6,589,556 Jul 8, 2003. U.S. Patent.

13. Breitkreutz, Jörg and Boos, Joachim. Paediatric and geriatric drug delivery. Expert Opinion on

Drug Delivery 4. 2007, pp. 37-45.

14. Tuleu, Catherine.Paediatric formulations in practice:. 2007. pp. 52-53.

15. Bar-Shalom, Daniel. Pediatric Formulations – Are We Living in the Past? American

Pharmaceutical Review. 14.6, 2001.

16. Bar-Shalom, Daniel. The Future of Oral Paediatric Formulations. 2010, pp. 128-130.

17. Bar-Shalom, Daniel. Necessity of Rethinking Oral Pediatric Formulations. Clinical therapeutics.

2014, Vol. 36.2, pp. 180-183.

Page 75: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 75

18. Bircan, Gökhan and Coban, Cansu.Investigation of non-ionic cellulosics for development and

characterization of semi-solid dosage forms for pediatric patients [thesis]. Copenhagen, Denmark :

Faculty of Pharmaceutical Sciences, University of Copenhagen, 2013.

19. Drug Information Online. Drugs.com. [Online] http://www.drugbank.ca/drugs/DB00518.

20. Rang, H. P., et al.Pharmacology. New York : Churchill Livingstone, 2003.

21. Lovett, R. Coffee: The demon drink? New Scientist. 2009.

22. DrugBank. Open Data Drug & Drug Target Database. [Online]

http://www.drugbank.ca/drugs/DB00201.

23. Fernández, L., et al. Solubility improvement of an anthelmintic benzimidazole carbamate by

association with dendrimers. Brazilian Journal of Chemical Engineering. 28.4, 2011, pp. 679-689.

24. Galia, E., Horton, J. and Dressman, J.B. Albendazole generics—a comparative in vitro study.

Pharmaceutical research. 16.12, 1999, pp. 1871-1875.

25. Bassani, V. L., et al. Enhanced water-solubility of albendazole by hydroxypropyl-β-cyclodextrin

complexation. Journal of inclusion phenomena and molecular recognition in chemistry. 25.1-3, 1996,

pp. 149-152.

26. Jung, H., et al. Absorption Studies of Albendazole and Some Physicochemical Properties of the

Drug and Its Metabolite Albendazole Sulphoxide. Journal of pharmacy and pharmacology

Biopharmaceutics. 50.1, 1998, pp. 43-48.

27. Moriwaki, C., et al. Enhancement of solubility of albendazole by complexation with β-

cyclodextrin. Brazilian Journal of Chemical Engineering. 25.2, 2008, pp. 255-267.

28. Agnihotri, Nitika, et al. Microencapsulation–A Novel Approach in Drug Delivery: A Review. Indo

Global Journal Of Pharmaceutical Sciences. 2.1, 2012, pp. 1-20.

29. C, Ansel, LV, Allen and NG, Popovich.Pharmaceutical dosage form and Drug delivery system. 8.

India : B.I Publications,, 2005. pp. 171-175.

30. Banker GS, Rhodes CT.Modern pharmaceutics. s.l. : In Parma Publication, 2002. pp. 501-527.

31. Benali, Mohammed, Hemati, Mehrdji and Gerbaud, Vincent. Effect of operating conditions and

physico–chemical properties on the wet granulation kinetics in high shear mixer. Powder Technology.

190.1, 2009, pp. 160-169.

32. Parikh, Dilip M., ed.Handbook of pharmaceutical granulation technology. s.l. : CRC Press, 1997.

Vol. 81.

33. Aulton, ME.Pharmaceutics: The Science of Dosage Form Design. 2. Edinburgh : Churchill

Livingstone, 2002.

34. Summers, Malcolm and Aulton, Michael.Pharmaceutics The Science of Dosage Form Design

(Granulation). London : Churchill Livingstone, 2002. pp. 364-378.

Page 76: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 76

35. Iveson, Simon M., et al. Nucleation, growth and breakage phenomena in agitated wet

granulation processes: a review. Powder Technology. 117.1, 2001, pp. 3-39.

36. Wicks, Jr, Z.W., Pappas, S.P and Jones, F.N.Organic Coatings: Science and Technology. New York :

John Wiley & Sons, Inc, 1992. Vol. 1.

37. Gosh, Swapan Kumar, ed.Functional coatings: by polymer microencapsulation. s.l. : John Wiley &

Sons, 2006.

38. Guignon, Bérengère, Albert Duquenoy, and Elisabeth D. Dumoulin. Fluid bed encapsulation of

particles: principles and practice. Drying Technology. 20.2, 2002, pp. 419-447.

39. Michaels, James N., et al. Steady states in granulation of pharmaceutical powders with

application to scale-up. Powder Technology. 189.2, 2009, pp. 295-303.

40. Schæfer, Torben and Mathiesen, Christina. Melt pelletization in a high shear mixer. IX. Effects of

binder particle size. International journal of pharmaceutics. 139.1, 1996, pp. 139-148.

41. Fu, J. S., et al. An experimental study of the variability in the properties and quality of wet

granules. Powder technology. 140.3, 2004, pp. 209-216.

42. Chitu, T.M., Oulahna, D. and Hemati, M. Wet granulation in laboratory-scale high shear mixers:

effect of chopper presence, design and impeller speed. Powder Technology. 206.1, 2011, pp. 34-43.

43. Knight, P. C. An investigation of the kinetics of granulation using a high shear mixer. Powder

technology. 77.2, 1993, pp. 159-169.

44. Saleh, K., L. Vialatte, and P. Guigon. Wet granulation in a batch high shear mixer. Chemical

engineering science. 60.14, 2005, pp. 3763-3775.

45. Knight, P. C., et al. An investigation of the effects on agglomeration of changing the speed of a

mechanical mixer. Powder Technology. 110.3, 2000, pp. 204-209.

46. Mackaplow, Michael B., Lawrence, A. Rosen and Michaels, James N. Effect of primary particle

size on granule growth and endpoint determination in high-shear wet granulation. Powder

technology. 108.1, 2000, pp. 32-45.

47. Faure, A., York, P. and Rowe, R. C. Process control and scale-up of pharmaceutical wet

granulation processes: a review. European Journal of Pharmaceutics and Biopharmaceutics. 53.3,

2001, pp. 269-277.

48. Shah, Rakhi B., Mobin A. Tawakkul, and Mansoor A. Khan. Comparative evaluation of flow for

pharmaceutical powders and granules. AAPS PharmSciTech. 9.1, 2008, pp. 250-258.

49. A. Jægerskou, P. Holm, T. Schæfer, H.G. Kristensen. Granulation in high speed mixers, part 3:

effects of process variables on the intragranular porosity. Pharm. Ind. 46.3, 1984, pp. 310-314.

50. Holm, P., T. Schaefer, and H. G. Kristensen. Granulation in high-speed mixers part VI. Effects of

process conditions on power consumption and granule growth. Powder technology. 43, 1985, Vol. 3,

pp. 225-233.

Page 77: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 77

51. Hampel, N., et al. Continuous pellet coating in a Wurster fluidized bed process. Chemical

Engineering Science. 86, 2013, pp. 87-98.

52. Teunou, E., and D. Poncelet. Batch and continuous fluid bed coating–review and state of the art.

Journal of food engineering. 53.4, 2002, pp. 325-340.

53. KuShaari, KuZilati, et al. Monte Carlo simulations to determine coating uniformity in a Wurster

fluidized bed coating process. Powder technology. 166.2, 2006, pp. 81-90.

54. Srivastava, Saurabh, and Garima Mishra. Fluid bed technology: Overview and parameters for

process selection. International Journal of Pharmaceutical Sciences and Drug Research . 2.4, 2010,

pp. 236-246.

55. Lachman LA, Lieberman HA, Kang JI.The Theory and Practice of Industrial Pharmacy. 3. Mumbai :

Varghese Publishing House, 1987. pp. 293-415.

56. Becher, Rolf-Dieter, and Ernst-Ulrich Schlünder. Fluidized bed granulation: gas flow, particle

motion and moisture distribution. Chemical Engineering and Processing: Process Intensification. 36.4,

1997, pp. 261-269.

57. Ström, Daniel, et al. A new device for coating single particles under controlled conditions.

Chemical engineering science. 60.16, 2005, pp. 4647-4653.

58. Tzika, M., Kiparissides, C. and Alexandridou, S. Evaluation of the morphological and release

characteristic of coated fertilizer granules produced in a Wurster fluidized bed. Powder Techology.

132, 2003, pp. 16-24.

59. Arimoto, Masahiro, Hideki Ichikawa, and Yoshinobu Fukumori. Microencapsulation of water-

soluble macromolecules with acrylic terpolymers by the Wurster coating process for colon-specific

drug delivery. Powder technology. 141.3, 2004, pp. 177-186.

60. Christensen, F. Norring, and P. Bertelsen. Qualitative description of the Wurster-based fluid-bed

coating process. Drug development and industrial pharmacy. 23.5, 1997, pp. 451-463.

61. Watano S, Sato Y, Miyanami K, Murakami T, Oda N. Scale-up ofagitation fluidized bed

granulation. Part 1: preliminary experimental approach for optimization of process variables. Chem

Pharm Bull (Tokyo). 1995, pp. 1212-1216.

62. Vummaneni, Vishnumurthy, and Dheeraj Nagpal. Taste Masking Technologies: An Overview and

Recent Updates. Int. J. Res. Pharm. Biomed. Sci. 3.2, 2012, pp. 510-24.

63. Sharma, Shalini, and Shaila Lewis. Taste masking technologies: a review. International Journal of

Pharmacy and Pharmaceutical Sciences. 2.2, 2010, pp. 6-13.

64. Alkire, Todd G., Ronald A. Sanftleben, and Steven S. Schuehle.Taste masking microparticles for

oral dosage forms. 5,607,697 U.S., Mar 4, 1997.

65. Ayenew, Zelalem, et al. Trends in pharmaceutical taste masking technologies: A patent review.

Recent patents on drug delivery & formulation. 3.1, 2009, pp. 26-39.

Page 78: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 78

66. Zhao, Grace Q., et al. The receptors for mammalian sweet and umami taste. Cell. 115.3, 2003, pp.

255-266.

67. Iwai, Naoichi. Drug compliance of children and infants with oral antibiotics for pediatric use.

Pediatrics International 39.1. 1997, pp. 132-142.

68. Sikandar, M. K., R. Malviya, and K. P. Sharma. Taste masking: an important pharmaceutical

technology for the improvement of organoleptic property of pharmaceutical active agents. Eur. J.

Biol. Sci. 3.3, 2011, pp. 67-71.

69. Bar-Shalom, Daniel. The Challenge of Automated Compounding. [book auth.] Daniel Bar-Shalom

and Klaus Rose. Pediatric Formulations. New York : Springer, 2014, 13, pp. 181-191.

70. Qiu, Yihong, et al.Developing solid oral dosage forms: pharmaceutical theory & practice. s.l. :

Academic Press, 2009.

71. Siewert, Martin, et al.FIP/AAPS guidelines to dissolution/in vitro release testing of novel/special

dosage forms. s.l. : AAPS PharmSciTech, 2003. pp. 43-52.

72. Hörter, D., and J. B. Dressman. Influence of physicochemical properties on dissolution of drugs in

the gastrointestinal tract. Advanced drug delivery reviews. 46.4, 2001, pp. 75-87.

73. Alvarez, Luis I., M. Lourdes Mottier, and Carlos E. Lanusse. Drug transfer into target helminth

parasites. Trends in parasitology. 23.3, 2007, pp. 97-104.

74. Bazzo, Giovana C., et al. Enhancement of felodipine dissolution rate through its incorporation

into eudragit® E–PHB polymeric microparticles: In vitro characterization and investigation of

absorption in rats. Journal of pharmaceutical sciences. 101.4, 2012, pp. 1518-1523.

75. Klancke, James. Dissolution testing of orally disintegrating tablets. Dissolution technologies. 10.2,

2003, pp. 6-9.

76. European pharmacopoeia 7th Ed. Strasbourg : Council of Europe, 2001.

77. Löbenberg, Raimar, and Gordon L. Amidon. Modern bioavailability, bioequivalence and

biopharmaceutics classification system:New scientific approaches to international regulatory

standards. European Journal of Pharmaceutics and Biopharmaceutics. 50.1, 2000.

78. Huuskonen, Jarmo. Estimation of aqueous solubility for a diverse set of organic compounds

based on molecular topology. Journal of chemical information and computer sciences. 40.3, 2000, pp.

773-777.

79. Alhnan, Mohamed A., Sudaxshina Murdan, and Abdul W. Basit. Encapsulation of poorly soluble

basic drugs into enteric microparticles: A novel approach to enhance their oral bioavailability.

International journal of pharmaceutics. 416.1, 2011, pp. 55-60.

80. Pacioni, Natalia L., et al. Spectrofluorimetric determination of benzoimidazolic pesticides: Effect

of< i> p</i>-sulfonatocalix [6] arene and cyclodextrins. Analytica chimica acta. 624.1, 2008, pp. 133-

140.

Page 79: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 79

81. Bassani, V. L., et al. Enhanced water-solubility of albendazole by hydroxypropyl-β-cyclodextrin

complexation. Journal of inclusion phenomena and molecular recognition in chemistry . 25.1-3, 1996,

pp. 149-152.

82. Woo, Jong Soo, et al. Development of cyclosporin A-loaded hyaluronic microsphere with

enhanced oral bioavailability. International journal of pharmaceutics. 345.1, 2007, pp. 134-141.

83. XXIII, US Pharmacopeia.The United States Pharmacopeia: Twentieth Revision. Rockville, MD :

United States Pharmacopeia Convention.

84. Mosharraf, Mitra, and Christer Nyström. The effect of particle size and shape on the surface

specific dissolution rate of microsized practically insoluble drugs. International journal of

pharmaceutics. 122.1, 1995, pp. 35-47.

85. Green, J. Mark. Peer Reviewed: A Practical Guide to Analytical Method Validation. Analytical

Chemistry. 68.9, 1996, pp. 305A-309A.

86. Kazakevich, Yuri V., and Rosario LoBrutto.HPLC for pharmaceutical scientists. s.l. : Wiley-

Interscience, 2007.

87. Shah, Vinod P., et al. Bioanalytical method validation—a revisit with a decade of progress.

Pharmaceutical research. 17.12, 2000, pp. 1551-1557.

88. Meyer, Veronika R.Practical high-performance liquid chromatography. s.l. : Wiley, 2003.

89. Dong, Michael W.Modern HPLC for practicing scientists. s.l. : John Wiley & Sons, 2006.

90. Schellinger, Adam P., and Peter W. Carr. Isocratic and gradient elution chromatography: A

comparison in terms of speed, retention reproducibility and quantitation. Journal of Chromatography

A. 1109.2, 2006, pp. 253-266.

91. Shabir, Ghulam A. Validation of high-performance liquid chromatography methods for

pharmaceutical analysis: Understanding the differences and similarities between validation

requirements of the US Food and Drug Administration, the US Pharmacopeia and the ICH. Journal of

Chromatography A. 987.1, 2003, pp. 57-66.

92. Nunn, Tony and Williams, Julie. Formulation of medicines for children. British journal of clinical

pharmacology. 59.6, 2005, pp. 674-676.

93. Sohi, Harmik, Yasmin Sultana, and Roop K. Khar. Taste masking technologies in oral

pharmaceuticals: recent developments and approaches. Drug development and industrial pharmacy.

30.5, 2004, pp. 429-448.

94. Tan, Elaine. Dosing information for paediatric patients: are they really "therapeutic orphans"?

Medical journal of Australia. 179.4, 2003, pp. 195-198.

95. Bazzo, Giovana C., et al. Enhancement of felodipine dissolution rate through its incorporation

into eudragit® E–PHB polymeric microparticles: In vitro characterization and investigation of

absorption in rats. Journal of pharmaceutical sciences. 101.4, 2012, pp. 1518-1523.

Page 80: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 80

96. Jinno, Jun-ichi, et al. Effect of particle size reduction on dissolution and oral absorption of a

poorly water-soluble drug, cilostazol, in beagle dogs. Journal of controlled release. 111.1, 2006, pp.

56-64.

97. Loveland, Peter J., and W. Richard Whalley. Particle size analysis. [ed.] KA Smith and CE Mullins.

Soil and Environmental analysis. 2nd, 2000, pp. 281-314.

98. J. Prescott, R. Barnum. On powder flowability. Pharmaceutical Technology. 24, 2000, pp. 60-84.

99. Bell, T. A. Industrial needs in solids flow for the 21st century. Powder Handling and Processing.

1999, pp. 3-12.

100. Agency, European Medicines. EMA/CHMP/ICH/405290/2010. ICH guideline Q4B. [Online]

September 2010.

http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2010/09/WC50009

7055.pdf.

101. European Pharmacopoeia. Strasbourg (France) : Council of Europe, 2005. Vol. 5.

102. Skalsky, B., T. Felisiak, and H. Petereit.Eudragit–application Guidelines. Darmstadt : Evonik

Röhm GmbH, 2009.

103. Si‐Nang, Luu, et al. Determination of coating thickness of microcapsules and influence upon

diffusion. Journal of pharmaceutical sciences. 62.3, 1973, pp. 452-455.

104. Beckett, Arnold H., Tinh T. Quach, and G. S. Kurs. Improved hydrodynamics for USP apparatus

2. Dissolut Technol 3. 1996, pp. 7-10.

105Experimental comparison of the different approaches to estimate LOD and LOQ of an HPLC

methodAnalytical Chemistry71.142672-2677

106. MassBank. Albendazole Reference Mass Spectrum. [Online] [Cited: October 24, 2013.]

http://www.massbank.jp/jsp/Dispatcher.jsp?type=disp&id=KOX00902&site=0.

107. Perrut, Michel, Jennifer Jung, and Fabrice Leboeuf. Enhancement of dissolution rate of poorly-

soluble active ingredients by supercritical fluid processes: Part I: Micronization of neat

particlesEnhancement of dissolution rate of poorly-soluble active ingredients by supercritical fluid

processes. International journal of pharmaceutics. 288.1, 2005, pp. 3-10.

108. Kemper, W. D., and R. C. Rosenau.Aggregate stability and size distribution. 1986. pp. 425-442.

109. Yin Win, Khin, and Si-Shen Feng. Effects of particle size and surface coating on cellular uptake of

polymeric nanoparticles for oral delivery of anticancer drugs. 26.15, 2005, pp. 2713-2722.

110. Pawari, Shiwaj; Ashir, Kumar. Issues in the formulation of drugs for oral use in children.

Pediatric Drugs. 4.6, 2002, pp. 370-379.

111. Guinard, Jean-Xavier, and Rossella Mazzucchelli. The sensory perception of texture and

mouthfee. Trends in Food Science & Technology. 7.7, 1996, pp. 213-219.

Page 81: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 81

112. Wehling, Fred, Steve Schuehle, and Navayanarao Madamala.Effervescent dosage form with

microparticles. No. 5,178,878 U.S.A, January 12, 1993.

113. Rowe, Raymond C., Paul J. Sheskey, and Siân C. Owen, eds.Handbook of pharmaceutical

excipients. London : Pharmaceutical press, 2006.

114. Law, M. F. L., et al. Comparison of two commercial brands of microcrystalline cellulose for

extrusion-spheronization. Journal of microencapsulation. 14.6, 1997, pp. 713-723.

115. Julian, Thomas N., and Galen W. Radebaugh.Chewable medicament tablet containing means

for taste masking. No. 4,851,226 U.S.A, July 25, 1989.

116. Pearnchob, N., J. Siepmann, and R. Bodmeier. Pharmaceutical applications of shellac: moisture-

protective and taste-masking coatings and extended-release matrix tablets. Drug development and

industrial pharmacy. 29.8, 2003, pp. 925-938.

117. Gilis, Paul MV, Valentin FV De Conde, and Roger PG Vandecruys.Beads having a core coated

with an antifungal and a polymer. No. 5,633,015 USA, May 27, 1997.

118. Chamsai, Benchawan, and Pornsak Sriamornsak. Novel disintegrating microcrystalline cellulose

pellets with improved drug dissolution performance. Powder Technology. 2012.

119. Newton, Michael, et al. The influence of formulation variables on the properties of pellets

containing a self‐emulsifying mixture. Journal of pharmaceutical sciences. 90.8, 2001, pp. 987-995.

120. Dempski, Robert E., Gunvant N. Mehta, and Joseph C. Saboe.Sustained release indomethacin.

No. 4,173,626 USA, Nov 6, 1979.

121. DeNeale, Richard J., Paul C. Guley, and George Milosovich.Sustained release pharmaceutical

compositions. No. 4,309,404 Jan 5, 1982.

122. Dukić-Ott, A., et al. Production of pellets via extrusion–spheronisation without the

incorporation of microcrystalline cellulose: a critical review. European Journal of Pharmaceutics and

Biopharmaceutics. 71.1, 2009, pp. 38-46.

123. Nigalaye, Ashok G., P. Adusumilli, and S. Bolton. Investigation of prolonged drug release from

matrix formulations of chitosan. Drug Development and Industrial Pharmacy. 1990, pp. 449-467.

124. Setthacheewakul, Saipin, et al. Development and evaluation of self-microemulsifying liquid and

pellet formulations of curcumin, and absorption studies in rats. European Journal of Pharmaceutics

and Biopharmaceutics. 76.3, 2010, pp. 475-485.

125. Newton, John Michael, Mariana Reis Pinto, and Fridrun Podczeck. The preparation of pellets

containing a surfactant or a mixture of mono-and di-gylcerides by extrusion/spheronization.

European journal of pharmaceutical sciences. 30.3, 2007, pp. 333-342.

126. (FDA), Food and Drug Administration.Guidance for Industry: Dissolution Testing of Immediate

Release Solid Oral Dosage Forms. U.S. Department of Health and Human Services. s.l. : (CDER), Center

for Drug Evaluation and Research, 1997.

Page 82: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 82

127. Valizadeh, Hadi, et al. Physicochemical characterization of solid dispersions of indomethacin

with PEG 6000, Myrj 52, lactose, sorbitol, dextrin, and Eudragit® E100. Drug development and

industrial pharmacy. 30.3, 2004, pp. 303-317.

128. Gharibi, H., et al. Thermodynamic studies of interaction between cationic surfactants and

polyvinyl pyrrolidone using potentiometric techniques. Colloids and Surfaces A: Physicochemical and

Engineering Aspects. 145.1, 1998, pp. 47-60.

129. Ghebremeskel, Alazar N., Chandra Vemavarapu, and Mayur Lodaya. Use of surfactants as

plasticizers in preparing solid dispersions of poorly soluble API: selection of polymer–surfactant

combinations using solubility parameters and testing the processability. International journal of

pharmaceutics. 328.2, 2007, pp. 119-129.

130. Jachowicz, R. E. B. B. A., et al. Solid dispersion of ketoprofen in pellets. International journal of

pharmaceutics . 206.1, 2000, pp. 13-21.

131. Nicklasson, Fredrik and Alderborn, Göran. Modulation of the tabletting behaviour of

microcrystalline cellulose pellets by the incorporation of polyethylene glycol. European journal of

pharmaceutical sciences. 9.1, 1999, pp. 57-65.

132. Cory, Wendy Clevenger, Harris, Corbyn and Martinez, Sabrina. Accelerated degradation of

ibuprofen in tablets. Pharmaceutical Development and Technology. 15.6, 2010, pp. 636-643.

133. Carvalho, L. A. E., M. Paula M. Marques, and John Tomkinson. Drug-excipient interactions in

ketoprofen: A vibrational spectroscopy study. Biopolymers. 82.5, 2006, pp. 420-424.

134. Vecchio, S., F. Rodante, and M. Tomassetti. Thermal stability of disodium and calcium

phosphomycin and the effects of the excipients evaluated by thermal analysis. Journal of

pharmaceutical and biomedical analysis. 24.5, 2001, pp. 1111-1123.

135. Mura, P., et al. Compatibility study between ibuproxam and pharmaceutical excipients using

differential scanning calorimetry, hot-stage microscopy and scanning electron microscopy. Journal of

pharmaceutical and biomedical analysis. 18.1, 1998, pp. 151-163.

136. Cuff, George William.Novel drug delivery system. No. EP 0422290 April 17, 1991. European

Patent.

137. Sriwongjanya, Mongkol, and Roland Bodmeier. Effect of ion exchange resins on the drug

release from matrix tablets. European journal of pharmaceutics and biopharmaceutics. 46.3, 1998,

pp. 321-327.

138. Rogers, True L., and Dave Wallick. Reviewing the use of ethylcellulose, methylcellulose and

hypromellose in microencapsulation. Part 2: Techniques used to make microcapsules. Drug

Development and Industrial Pharmacy. 37.11, 2011, pp. 1259-1271.

139. —. Reviewing the use of ethylcellulose, methylcellulose and hypromellose in

microencapsulation. Part 1: materials used to formulate microcapsules. Drug Development and

Industrial Pharmacy. 38.2, 2012, pp. 129-157.

Page 83: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 83

140. Colombo, Paolo. Swelling-controlled release in hydrogel matrices for oral route. Advanced drug

delivery reviews. 11.1, 1993, pp. 37-57.

141. Siepmann, J., and N. A. Peppas. Modeling of drug release from delivery systems based on

hydroxypropyl methylcellulose (HPMC). Advanced drug delivery reviews. 2012.

142. Ford, James L. Thermal analysis of hydroxypropylmethylcellulose and methylcellulose: powders,

gels and matrix tablets. International Journal of Pharmaceutics. 179.2, 1999, pp. 209-228.

143. Company, Dow Chemical.Ethers, METHOCEL Cellulose. "Technical Handbook". Midland : Dow

Chemical Company, 1997.

144. Mitchell, K., et al. The influence of drugs on the properties of gels and swelling characteristics of

matrices containing methylcellulose or hydroxypropylmethylcellulose. International journal of

pharmaceutics. 100.1, 1993, pp. 165-173.

145. Gao, Ping, Phillip R. Nixon, and John W. Skoug. Diffusion in HPMC gels. II. Prediction of drug

release rates from hydrophilic matrix extended-release dosage forms. Pharmaceutical research. 12.7,

1995, pp. 965-971.

146. Dashevsky, A., K. Kolter, and R. Bodmeier. pH-independent release of a basic drug from pellets

coated with the extended release polymer dispersion Kollicoat® SR 30 D and the enteric polymer

dispersion Kollicoat MAE 30 DP. European journal of pharmaceutics and biopharmaceutics. 58.1,

2004, pp. 45-49.

147. Bland, J. Martin, and Douglas G. Altman. Comparing methods of measurement: why plotting

difference against standard method is misleading. The Lancet. 1995, pp. 1085-1087.

148. Howard, Seltman. Carnegie Mellon's Department of Statistics. The t-test and Basic Inference

Principles. [Online] [Cited: October 22, 2013.]

http://www.stat.cmu.edu/~hseltman/309/Book/chapter6.pdf.

149. ICH. ICH Validation of Analytical Procedures: Q2(R1). [Online] [Cited: November 22, 2013.]

http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q2_R1/Step4/Q2_

R1__Guideline.pdf.

150. ICH.Proceedings of the International Conference on Harmonisation of Technical Requirement of

Registration of Pharmaceuticals for Human Use (ICH): Validation of Analytical Procedures:

Methodology, Q2B. Geneva, Switzerland : s.n., 1996.

151. Boury, F., et al. Bovine serum albumin release from poly (α-hydroxy acid) microspheres: effects

of polymer molecular weight and surface properties. Journal of controlled release. 45.1, 1997, pp. 75-

86.

152. Crotts, G., and Tae Gwan Park. Protein delivery from poly (lactic-co-glycolic acid) biodegradable

microspheres: release kinetics and stability issues. Journal of microencapsulation. 15.6, 1998, pp.

699-713.

153. Johansen, Pål, et al. Improving stability and release kinetics of microencapsulated tetanus

toxoid by co-encapsulation of additives. Pharmaceutical research. 15.7, 1998, pp. 1103-1110.

Page 84: FORMULATION OF ALBENDAZOLE MICROCAPSULES FOR THE …

P a g e | 84

8. APPENDICES