research paper ijpsn-6-6-13-kishan drug-excipient ... · drug-excipient interaction during...

13
Research Paper Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery System for Nizatidine Ramireddy Amarnath Reddy, Bomma Ramesh and Veerabrahma Kishan* Department of Pharmaceutics, University College of Pharmaceutical Sciences, Kakatiya University, Warangal-506 009, Andhra Pradesh, India. Received June 6, 2013; accepted July 6, 2013 ABSTRACT The present investigation dealswith the development and evaluation of floating tablets of nizatidine to prolong the gastric residence time, increase local delivery of drug to the H 2 -receptor of the parietal cell wall to reduce stomach acid secretion. The drug-excipient compatibility studies were conducted by using FTIR, DSC and visual observations. Citric acid inclusion in formulations resulted in incompatibility and the composition was modified to eliminate the problem of incompatibility. Floating matrix tablets of nizatidine were developed by direct compression method using hydroxypropyl methylcellulose (HPMC K4M) and polyox WSR 1105 alone as release retardants and sodium bicarbonate as a gas-generating agent. Alleleven formulations exhibited satisfactory physicochemical characteristics andin vitro buoyancy. Formulations F6 and F10 exhibited controlled and prolonged drug release for 10 h with zero order release. Formulation (F10) was selected as optimized formulation based on physicochemical properties and in vitro drug release and was used inradiographic studies by incorporating BaSO 4 . The radiographic studies were conducted in comparison with plain controlled release tablets. These studies revealed that gastric retention time of floating and plain controlled release tablets in fasting state were 2 ± 0.86 h and 0.5 h respectively in human volunteers. Gastric retention time of floating and plain controlled release tablets in fed state were 5.33 ± 0.57 h and 1.66 ± 0.28 h respectively in human volunteers. In conclusion, optimal floating matrix tablet for nizatidine with desired in vitro buoyancy, in vivo gastric retention time and prolonged release could be prepared. KEYWORDS: Floating tablets; drug-excipients compatibility; gastric residence time; nizatidine. Introduction Oral drug delivery is the preferred route for drug administration because of its convenience, low cost, and high patient compliance when compared with several other routes. About 90 percent of drug products are administered via the oral route (Kokate et al., 2006).Controlled-release (CR) pharmaceutical dosage forms may offer one or more advantages over immediate release dosage forms of the same drug, including a reduced dosing frequency, a decreased incidence and/or intensity of adverse effects, a greater selectivity of pharmacologic activity, and a reduction in drug plasma fluctuation resulting in a more constant or prolonged therapeutic effect (Malinowski and Marroum, 1999).However, this CR approach is facing problems with several physiological difficulties such as inability to restrain and locate controlled drug delivery system within the desired region of gastrointestinal tract (GIT), due to variable gastric emptying and motility.Control of placement of drug delivery system in specific region of GIT offers advantage for variety of important drugs characterized by narrow absorption window in GIT or drugs with stability problem (Garg and Gupta, 2008). Various approaches that have been proposed to control the gastric residence time of drug delivery systems in the upper part of the GIT include floating drug delivery systems (FDDS) (Brahma and Kim, 2000), high-density DDS (Caldwell et al., 1998), mucoadhesive systems (Lehr, 1994), swelling and expanding DDS (Urquhart and Theeuwes, 1984; Mamajek and Moyer, 1980), modified shape systems(Fix et al., 1993), and other delayed gastric devices (Gröning and Heun, 1989). FDDS have a bulk density lower than the gastric fluid and thus remain buoyant in the stomach, without affecting the gastric emptying rate for a prolonged period of time. While the systemis floating on the gastric contents, the druggets released slowly (Arora et al., 2005). Based on the mechanism of buoyancy, two distinctly different technologies, i.e., non-effervescent and effervescent system, have been utilized in the development of FDDS. The effervescent system utilizes matrices prepared with swellable polymers and effervescent components, e.g., sodium bicarbonate and International Journal of Pharmaceutical Sciences and Nanotechnology Volume 6 Issue 4December 2013 (extra issue) IJPSN-6-6-13-KISHAN 2281

Upload: others

Post on 27-May-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

Amarnath Reddy et al: Drug-Excipient Interaction during Formulation Development of Gastroretentive Drug Delivery… 2281 Research Paper

Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery System for Nizatidine

Ramireddy Amarnath Reddy, Bomma Ramesh and Veerabrahma Kishan* Department of Pharmaceutics, University College of Pharmaceutical Sciences, Kakatiya University, Warangal-506 009, Andhra Pradesh, India.

Received June 6, 2013; accepted July 6, 2013 ABSTRACT

The present investigation dealswith the development and evaluation of floating tablets of nizatidine to prolong the gastric residence time, increase local delivery of drug to the H2-receptor of the parietal cell wall to reduce stomach acid secretion. The drug-excipient compatibility studies were conducted by using FTIR, DSC and visual observations. Citric acid inclusion in formulations resulted in incompatibility and the composition was modified to eliminate the problem of incompatibility. Floating matrix tablets of nizatidine were developed by direct compression method using hydroxypropyl methylcellulose (HPMC K4M) and polyox WSR 1105 alone as release retardants and sodium bicarbonate as a gas-generating agent. Alleleven formulations exhibited satisfactory physicochemical characteristics andin vitro buoyancy. Formulations F6 and F10 exhibited controlled and prolonged drug release for 10

h with zero order release. Formulation (F10) was selected as optimized formulation based on physicochemical properties and in vitro drug release and was used inradiographic studies by incorporating BaSO4. The radiographic studies were conducted in comparison with plain controlled release tablets. These studies revealed that gastric retention time of floating and plain controlled release tablets in fasting state were 2 ± 0.86 h and ≤ 0.5 h respectively in human volunteers. Gastric retention time of floating and plain controlled release tablets in fed state were 5.33 ± 0.57 h and 1.66 ± 0.28 h respectively in human volunteers. In conclusion, optimal floating matrix tablet for nizatidine with desired in vitro buoyancy, in vivo gastric retention time and prolonged release could be prepared.

KEYWORDS: Floating tablets; drug-excipients compatibility; gastric residence time; nizatidine.

Introduction Oral drug delivery is the preferred route for drug

administration because of its convenience, low cost, and high patient compliance when compared with several other routes. About 90 percent of drug products are administered via the oral route (Kokate et al., 2006).Controlled-release (CR) pharmaceutical dosage forms may offer one or more advantages over immediate release dosage forms of the same drug, including a reduced dosing frequency, a decreased incidence and/or intensity of adverse effects, a greater selectivity of pharmacologic activity, and a reduction in drug plasma fluctuation resulting in a more constant or prolonged therapeutic effect (Malinowski and Marroum, 1999).However, this CR approach is facing problems with several physiological difficulties such as inability to restrain and locate controlled drug delivery system within the desired region of gastrointestinal tract (GIT), due to variable gastric emptying and motility.Control of placement of drug delivery system in specific region of GIT offers advantage for variety of important drugs

characterized by narrow absorption window in GIT or drugs with stability problem (Garg and Gupta, 2008). Various approaches that have been proposed to control the gastric residence time of drug delivery systems in the upper part of the GIT include floating drug delivery systems (FDDS) (Brahma and Kim, 2000), high-density DDS (Caldwell et al., 1998), mucoadhesive systems (Lehr, 1994), swelling and expanding DDS (Urquhart and Theeuwes, 1984; Mamajek and Moyer, 1980), modified shape systems(Fix et al., 1993), and other delayed gastric devices (Gröning and Heun, 1989).

FDDS have a bulk density lower than the gastric fluid and thus remain buoyant in the stomach, without affecting the gastric emptying rate for a prolonged period of time. While the systemis floating on the gastric contents, the druggets released slowly (Arora et al., 2005). Based on the mechanism of buoyancy, two distinctly different technologies, i.e., non-effervescent and effervescent system, have been utilized in the development of FDDS. The effervescent system utilizes matrices prepared with swellable polymers and effervescent components, e.g., sodium bicarbonate and

 

International Journal of Pharmaceutical Sciences and Nanotechnology

Volume 6 • Issue 4• December 2013 (extra issue) IJPSN-6-6-13-KISHAN

2281

Page 2: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

2282 Int J Pharm Sci Nanotech Vol6;Issue 4•December 2013 (extra issue) citric acid or adipic acid. The matrices are fabricated such that in the stomach carbon dioxide is liberated by the acidity of the gastric contents and is entrapped in the swollen gel of hydrocolloid. This produces an upward motion of the dosage form and maintains its buoyancy (Rubinstein and Friend, 1994). In non-effervescent FDDS, the drug is mixed with a gel-forming hydrocolloid, which swells on contact with the gastric fluid after oral administration and maintains relative integrity of shape and a bulk density of less than unity within an outer gelatinous barrier. The air trapped by the swollen polymer confers buoyancy to these dosage forms (Sheth and Tossounian, 1979).

Ranitidine HCl was formulated as FDDS by lowering the density. The local delivery of Ranitidine HCl increased the stomach wall receptor site bioavailability to reduce stomach acid secretion (Raval et al., 2007). Similarly, nizatidine is also an histamine H2-receptor antagonist that inhibits stomach acid production and commonly used in the treatment of peptic ulcer and Gastro Esophageal Reflux disease.It reduces the acid secretion by directly acting on parietal cell wall (Davis, 1997). It is having an oral bioavailability of 70% with a biological half-life of 1-2 h. Due to the above mentioned characteristics it becomes a suitable candidate for the development of gastroretentive delivery system.

The present investigation was concerned with the development and evaluation of floating tablets of nizatidine by using hydroxyl-propyl-methyl cellulose (HPMC K4M) or polyethylene oxide (Polyox WSR 1105) as sole release retardant, and sodium bicarbonate and citric acid were incorporated as gas-generating agents in the formulations to prolong the gastric residence time, increase local delivery of drug to the H2-receptor of the parietal cell wall. Initially, drug-excipients compatibility problemswere studied by using Fourier transform infrared (FTIR), differential scanning calorimetry (DSC) and visual observation of the prepared tablets for one month. The optimized formulation possessed good floating, extended and controlled drug release properties and was chosen for further in vivo radiological investigation to determine the mean gastric retention time.

Materials and Methods

Materials Nizatidine was obtained as a generous gift sample

from Dr. Reddy’s laboratories, Pydibheemavaram, India.Hydroxy propyl methyl cellulose (HPMC) K4M and polyethylene oxide (PEO) WSR 1105 were obtained as gift samples from M/s Orchid Chemicals and Pharmaceuticals Ltd., Chennai, India. Microcrystalline cellulose (Avicel pH102), Sodium bicarbonate were purchased from M/s S.D Fine chemicals Ltd., Mumbai, India. All other ingredients used were of laboratory grade.

Methods Drug-excipient compatibility studies

(i) Fourier transform infrared (FTIR) spectroscopy The FTIR spectra of samples were obtained

using FTIR spectrophotometer (BX I, Perkin Elmer, USA). Pure drug, individual polymers and optimised formulations were subjected to FTIR study. About 2–3 mg of sample was mixed with dried potassium bromide (KBr) of equal weight and compressed to form a KBr disk. The samples were scanned from 400 to 4000 cm−1.

(ii) Differential scanning calorimetry (DSC) The DSC experiments were carried out to find

out the presence of any interaction among drug and the excipients. Pure drug, individual polymers and optimised formulation were subjected to the study. About 5-15 mg of sample to be analysed was taken in the pierced DSC aluminium pan and scanned in the temperature range of 50–250 °C. The heating rate was 10°C/min; nitrogen served as purged gas and the system was cooled down by liquid nitrogen. The differential thermal analyser (DSC 822 e/200, Mettler Toledo, Switzerland) was used for this purpose.

(iii) Visual observation Initially, the tablets of nizatidine were prepared

by using either HPMC K4M or Polyox WSR 1105 as release retardant, sodium bicarbonate and citric acid as gas generating agents, Avicel pH102 as bulking agent, magnesium stearate as lubricant and talc as glidant in the formulations. After compression of the mixture into tablets, the change in physical appearance was observed during storage at room temperature. The visual observation was carried out for one month by excluding one of the ingredients from the composition. Finally, the composition was modified to eliminate the problem of incompatibility due to citric acid.

Development of tablets

Accurately weighed quantities (Table 1) of polymer and Avicel pH102 were taken in a mortar and mixed geometrically. To this required quantity of nizatidine was added and mixed slightly with pestle. Weighed quantity of sodium bicarbonate was taken separately in a mortar and powdered with pestle. The powder was passed through sieve no. 40 and mixed with the drug blend which was also passed through sieve no. 40. The whole mixture was collected in a plastic bag and mixed for 3 min. To this talc was added and mixed for 2 minutes, later magnesium stearate was added and mixed for 3 min. The mixture equivalent to 500 mg was compressed into tablets using rotary tablet machine (Riddhi, Model- RDD3, Ahmedabad) with 10 mm round concave punches at a hardness of 6 kg/cm2.

Page 3: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

Amarnath Reddy et al: Drug-Excipient Interaction during Formulation Development of Gastroretentive Drug Delivery… 2283 TABLE 1

Composition of various formulations of nizatidine floating tablets.

Name of ingredients Formulation codes and ingredients (mg)

F1 F2 F3 F4 F5 F6 F7 F8 F9 F10 F11 Nizatidine 270 270 270 270 270 270 270 270 270 270 270 HPMC K4M 100 80 70 60 75 75 80 - - - - Polyox WSR 1105 - - - - - - - 80 100 110 115 Sodium bicarbonate 50 50 50 50 50 60 60 60 60 60 60 Citric acid anhydrous 10 10 10 10 10 - - - - - - Avicel PH 102 56 76 86 96 81 81 76 76 56 46 41 Talc 7 7 7 7 7 7 7 7 7 7 7 Magnesium stearate 7 7 7 7 7 7 7 7 7 7 7 Total tablet weight 500 500 500 500 500 500 500 500 500 500 500

TABLE 2

Physical parameters of nizatidine floating tablets.

Formulation Code

Weight(mg) Hardness(kg/cm2) Thickness (mm) Friability(%) Floating lag time (s) Assay(%)

F1 506 ±9.06 5.1 ± 0.2 6.13 ± 0.05 0.27 50±3 100.85 F2 506 ± 8.43 5 ± 0.3 6.19 ± 0.03 0.35 38±2 99.03 F3 502.5 ± 9.78 6.1 ± 0.3 6.16 ± 0.04 0.24 46±3 99.21 F4 501 ± 11 5.8 ± 0.5 6.11 ± 0.06 0.30 44±5 99.91 F5 501.5 ± 6.25 6 ± 0.3 6.13 ± 0.05 0.32 36±3 100.7 F6 501.5 ± 7.83 6.1 ± 0.2 6.13 ± 0.04 0.18 85±5 98.3 F7 500.5 ± 7.24 5.9 ± 0.4 6.16 ± 0.04 0.19 92±4 98.9 *F8 507 ± 8.56 6.1 ± 0.3 6.30 ± 0.03 0.18 65±3 99.28 *F9 506.5 ± 10.28 6.2 ± 0.2 6.34 ± 0.04 0.20 83±4 97.48 F10 504.5 ± 10.91 5.9 ± 0.3 6.32 ± 0.04 0.17 114±5 98.96 F11 504.5 ± 8.64 6.1 ± 0.2 6.33 ± 0.04 0.18 171±6 98.29

Note: All the formulations floated for more than 10 h. Formulations *F8 and *F9 floated for 6 h and 8 h respectively.

Physical evaluation of floating tablets of nizatidine

The prepared tablets were evaluated in terms of their weight variation, hardness, thickness, friability and drug content (Table 2).

In vitro buoyancy studies

The in vitro buoyancy studies were carried out for the prepared floating tablets as per published method (Rosa et al., 1994). The tablets were placed in a 100 mL beaker containing 0.1 N HCl. The time required for a tablet to rise to the surface for floating was determined as floating lag time. The duration of time for which the dosage form constantly remained on the surface of medium was determined as the total floating time.

In vitro drug release studies

In vitro dissolution studies were carried out using a USP 29 type II (paddle type) dissolution apparatus (Electro Lab, TDT 06P). A floating tablet of nizatidine was dropped into 900 mL of 0.1 N HCl (pH 1.2), maintained at 37 ± 0.5°C and stirred at a speed of 50 rpm. At different times, 5 ml aliquots were withdrawn and replaced with an equivalent amount of plain dissolution medium kept at 37°C. The collected samples were filtered and analysed at λmax 314 nm using a UV–Visible spectrophotometer (Elico Pvt. Ltd., SL 159). The cumulative percentage of drug released was calculated using standard calibration curve of nizatidine.

Analysis of in vitro drug release data

Model dependent methods

To analyze the mechanism of drug release from the tablets, the in vitro dissolution data was subjected to the zero-order (Chen and Hao, 1998), first-order (Shah et al., 1987), Higuchi (Higuchi, 1961), and Korsmeyer-Peppas kinetic models (Ritger and Peppas, 1987; Korsmeyer et al., 1983). The equations for the kinetic models are given below.

Zero-order: Qt = Q0 + K0t…..(1) First order: logC = logC0 – k1t/2.303 …..(2) Higuchi: Qt = K2t1/2 …..(3) Qt/Q∞ = kptn …..(4)

Where Q0, Qt and Q∞ are the amounts of drug dissolved initially, at any time t and at ∞ time. C0 and C are the concentration of the drug initially and at any time t, and k0, k1, k2 and kp are the rate constants obtained from the linear curves of the respective models.

Model independent methods

For the comparison of release profiles of reference and test products, “difference factor” f1 and “similarity factor” f2, were calculated (Moore and Flanner, 1996). The difference factor (f1) measures the percent error between the two curves over all time points and is calculated by the following equation:

   Σ

Σ

nj=1 j j

1 nj=1 j

R – Tf = ×100

R

Page 4: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

2284 Int J Pharm Sci Nanotech Vol6;Issue 4•December 2013 (extra issue)

where n is the number of sampling points, and Rj and Tj are the percent dissolved of the reference and test products at each time point j. The two release profiles are considered to be similar if f1 value is lower than 15 (between 0 and 15). The similarity factor (f2) is a logarithmic transformation of the sum of squared error of differences between the test Tj and the reference products Rj over all time points. It is calculated by using following equation:

 n 2

j j jj 1

1[1 W R T ]n

=

⎧ ⎫⎛ ⎞+ Σ −⎨ ⎬⎜ ⎟⎝ ⎠⎩ ⎭

-0.5

2f = 50 log ×100

Where Wj is an optional weight factor and other terms are as defined earlier. The two dissolution profiles are considered to be similar if f2 value is more than 50 (between 50 and 100).

Tablet preparation for in vivo radiographic studies

Radiological (X-Ray)study was used for determination of the anatomical location of nizatidine floating tablets in the GI tract. To make the tablets X-ray opaque, the incorporation of BaSO4 was necessary. For this purpose, 65 mg of the drug was replaced with BaSO4 (65 mg BaSO4 + 205 mg nizatidine) and all other ingredients were kept constant. Plain controlled release tablets of nizatidine were used as a control. The optimized formulation F10 was selected for the radiological study, which contained 41% drug, 13% BaSO4 as radio opaque agent, 22% polyox WSR 1105 as polymer and 12% NaHCO3 as gas generating agent, 9.2% Avicel pH 102, 1.4% magnesium stearate and 1.4% talc. The plain controlled release tablets were prepared with same composition, in which gas generating agent NaHCO3 was replaced with Avicel pH 102.

In vivo radiographic studies

Six healthy male volunteers participated in the studies after giving informed written consent. The studies were approved by the Ethical Committee of University College of Pharmaceutical Sciences (UCPSc), Kakatiya University, Warangal, India. The study was conducted by administering to each subject one floating/plain controlled release tablet in one session. The healthy male volunteers, weighing between 65±10 kg and in the age group of 25±2 years were included in the study. Study was conducted in fasted and fed state, 3 volunteers each. (a) Fasting state: The BaSO4-loaded floating/plain

controlled release tablet was administered orally to each volunteer with 200 mL of water. During the study they were not allowed to eat but the water was made available ad libitum. The first X-ray photograph was taken 30 min after administration of tablet. Further, the next pictures were taken at different time intervals viz., 1.5, 3, and 5 h in a standing position under the supervision of an expert

radiologist. The source of X-rays and the subject was kept constant for all images. Thus, the observation of the tablet movements could be easily noticed. The mean gastric residence time was calculated.

(b) Fed state: After overnight fasting, the volunteers were fed with a low calorie food. Half an hour later, a BaSO4-loaded floating/plain controlled release tablet was administered orally to each volunteer with 200 mL of water. At different time intervals (0.5, 1.5, 3, 5 and 7 h post administration of tablet), the volunteers were exposed to abdominal X-ray imaging and the mean gastric residence time was calculated.

Results and Discussion Drug-excipient compatibility studies

(i) Fourier Transform Infrared (FTIR) Spectroscopy Chemical interaction between drug and polymer

could change the therapeutic efficacy of the drug. To investigate the possibility of chemical interaction between drug and excipients, samples were analyzed over the range 400 – 4000 cm−1.

The FTIR spectra of pure drug, drug-HPMCK4M and drug-Polyox WSR 1105 were obtained and shown in Figure 1. The major peaks were observed at 754.5, 1017.8 and 1229.8 cm-1 for pure drug. These results suggested the compatibility between nizatidine, HPMC K4M and polyox WSR1105, because FTIR spectra of nizatidine-HPMC K4M and nizatidine-Polyox WSR 1105 mixture displayed all the characteristic bands of drug, without any significant spectral shift.

Figure 1d suggested that there was incompatibility between nizatidine and citric acid because FTIR spectrum of nizatidine and citric acid mixture (1:1), which was immediately analyzed after preparation, showed an extra peak at 1700 - 1800 cm-1 indicated by an arrow other than characteristic bands of drug. This was confirmed further byFT-IR spectrum of nizatidine and citric acid mixture (Figure 1e), which was preserved for one month and analyzed, showed entirely different peaks and lacking characteristic bands of drug. But, in contrast the Figure 1f and 1g showing FTIR spectra of the formulation F5 containing citric acid displayed all the characteristic bands of drug, without any significant spectral shift. This suggested absence of chemical interaction between the components of the formulation and probably, due to very low concentration of citric acid in formulation (0.02 % w/w).

Page 5: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

Amarnath

Fig. 1.Comd) citric acimonth pre

(ii) D T

miif(ecb171ctFaewoteto

Reddy et al: D

parison of FT-IRid anhydrous anservation at RT

Differential ScThe thermal mixture of druinterest sinceinteraction amformulations. (NIZ-1) showendothermic correspondingbeing started 1:1 mixture of7) was showin139.79°C whicchange in tetowards the Figure 3 (obtaand polyox Wendothermic was well presof broadeningtemperature. endotherm of the drug and of each compo

Drug-Excipient I

R spectra of a) pund nizatidine mixT, f) formulation

canning Calorproperties o

ug and excipie this can mong differen The DSC cur

wed in Figurpeak at 136

g to its meltin at 132°C andf nizatidine ang an endothech was well erms of bro

higher temained from 1:1WSR 1105, peak of drugerved with slig or shiftingThe minor ch

drug could bepolymer, whi

onent in the m

Interaction duri

ure nizatidine, bxture (1:1) immen F5 immediate

rimetry (DSCof the drug ients are of imhelp to ass

nt componentrve of pure ne 2, a singl6.32°C (−130ng point (130

d ended at 13nd HPMC K4ermic peak ofpreserved wit

oadening or mperature. S1 mixture of n

NIZ 8) shog at 141.60°Cight changes

g towards thehange in thee due to the mich lowered thmixture and i

ing Formulation

b) HPMC K4M aediately after prely after prepara

) and the

mportant sess the ts of the izatidine le sharp 0.50 J/g) 0–134°C) 8°C. The 4M (NIZ-f drug at th slight shifting

Similarly, izatidine

owed an C, which in terms e higher melting

mixing of he purity indicated

n Development

and nizatidine meparation, e) citration, and g) af

no pconcluHPMCthe for

The Danhydendothits meorganimeltinthe bashift. anhydpoint gravimdetectpeak depenNIZ-3disappbetwenizati

Furththat N

of Gastroreten

mixture, c) Polyoxric acid anhydrofter one month

otential incouded that theC K4M and pormulation.

DSC thermogdrous (NIZ-2hermic peak elting point (ic acid starng event sinceaseline level,

This thermdrous at tem

was consmetric analystable in all th

around 190o

ndent intensit and NIZ-4 peared, indicaeen nizatidinedine was inco

her in Figure NIZ-9 (Formu

ntive Drug Deliv

x WSR 1105 andous and nizatidinpreservation at

ompatibility. e drug had coolyox WSR 11

gram (Figure 2) showed aat 162.76°C c155°C). Deco

rted immediae the signal d but continue

mal instabilitymperatures absistent withsis (TGA) finhe physical bloC exhibitingty (Schilling endothermicating the potee and citric acompatible with5, the DSC culation, F5 c

very… 22

d nizatidine mixtne mixture after room temperat

Thus, it wompatibility w105 when used

4) of citric aa single shcorrespondingmposition of ately after did not returned its downwy of citric above its melth the therndings and wlends as a brg concentratiet al., 2008).c peak of dential interactcid consequenh citric acid. curves suggescontaining ci

285

ture, r one ture.

was with d in

acid harp g to the the

n to ward acid ting rmo was

road ion-. In

drug tion

ntly,

sted itric

Page 6: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

2286

a1ctew

Fig. 2. DSC

Fig. 3.DSC(NIZ-8).

acid) showed 140.01°C andchanges in ttowards the hendothermic pwell preserve

C curves of pure

curves of nizati

an endotherd was well pterms of bro

higher temperpeak of drug d with slight

drug nizatidine

idine pure drug

rmic peak of preserved witoadening or

rature. NIZ-10at 134.58°C

t changes in

(NIZ-1), pure po

(NIZ-1), pure po

Int J Pha

drug at th slight

shifting 0 showed and was terms of

olymer HPMC K4

olymer polyox W

arm Sci Nanote

shiftinboth Npeak indicaintera

4M (NIZ-5), and

WSR 1105 (NIZ-6

ech Vol6;Issu

ng towards thNIZ-9 and N

(NIZ-2, 1ated that taction between

d 1:1 mixture of n

), and 1:1 mixtu

ue 4•December

he lower tempIZ-10 citric a62°C) disapthere mightn nizatidine a

nizatidine & HPM

re of nizatidine

r 2013 (extra iss

perature. Butacid endotherppeared, wht be potenand citric acid.

MC K4M (NIZ-7

& polyox WSR 1

ssue)

t in mic

hich ntial .

7).

1105

Page 7: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

Amarnath

(iii) V F

ccpFasibiccb

Reddy et al: D

Visual observaFrom both Fconcluded thacitric acid. Ipunched tableFigure 6. Whacid were prestarted. Browimmediately abut graduallyingredients, ccoloration wasclearly provebetween nizat

Drug-Excipient I

ation FTIR and D

at nizatidine wt was also ets with citrichenever both esent the scopwn color waafter preparaty developed. itric acid or ds noticed (Figed the existtidine and citr

Interaction duri

DSC studies was incompativisually obsec acid and shnizatidine an

pe of brown coas not cleartion and after

When eitherdrug was exclgure 6 m and tence of intric acid.

ing Formulation

it was ible with erved in howed in nd citric oloration rlyvisible one day, r of the luded no n). This teraction

ind5foto±00mfoop

n Development

Physical ev

The physicn Table 2. T

different formu500.5 ± 7.24ormulations Fo6.2 ± 0.2 kg/c

± 0.3 to 5.1 ± 00.06to 6.34 ± 0.17 to0.35% mechanical resormulations gf all the formu

properties a

of Gastroreten

valuation of fl

co-chemical chThe average wulations (F1 – 4to 507 ± 8F3 - F11 was focm2, except for0.2 kg/cm2. Th0.04mm and for all the

sistance. The gave a value ofulations showand compli

ntive Drug Deliv

Fn(Na(Noaa(Nopte

Figniz(NIF5 acipreandpretem

oating tablets

haracters of taweight of all F11) was fou8.56 mg. Thound to be in br F1 and F2 w

he thickness vafriability wasformulations assay of the f 97.48 % to 1

wed acceptableied with

very… 22

Fig. 4.DSC curvenizatidine pure dNIZ-1), pure c

acid anhydrNIZ-2), 1:1 mixf nizatidine & c

acid immediaafter preparaNIZ-3), and ane mo

preservation at remperature (NIZ

g. 5.DSC curvezatidine pure dIZ-1), formula containing cid immediately aeparation (NIZd after one moeservation at rmperature (NIZ-

s of nizatidine

ablets are shol the tablets

und to range frhe hardness between 5.8 ±

with hardness aried from 6.1s in the range

indicating gtablets of all

100.85 %. Tabe physicochem

pharmacopo

287

es of drug citric rous

xture citric ately ation after onth room Z-4).

s of drug ation citric after Z-9), onth room 10).

e

own for

rom of 0.5 of 5

11 ± e of

good the

blets mical

eial

Page 8: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

2288 specificatfriability.

In vitrThe in

as gas-forin vitro most of than 10 hlayers fentrapmeincrease dissolutioThe formthe stomathe aciditgel of hydthe dosag2000). Thstomach

tions for wei

tro buoyancy s

nvestigated flrming agent dbuoyancy exthe formulat

h except F8 anformed by tent of gener

in the tableon medium (0

mulations wereach, the carboty of gastric drocolloid. A dge form to floahe extended rcould increas

ight variation

studies

loating systemdispersed in aperiments reions to rema

nd F9. It was sthe polymerrated gas buet porosity m.1 N HCl) for e developed soon dioxide gascontents and

decrease in that on the chymresidence timse local delive

n, drug cont

ms employed Na hydrogel maevealed the aain buoyant fsuggested tha

rs, enabled ubbles. The made it to longer periodo that upon as could be libe got entrappe

he density woume (Brahma a

me of this druery of the dru

Int J Pha

tent and

NaHCO3 trix. The

ability of for more at the gel

efficient possible float on

d of time. arrival in erated by ed in the uld cause and Kim, ug in the ug to H2-

reFb(R

thsh(FciocislfopN1in1

arm Sci Nanote

eceptor of thFDDS and rebioavailabilityRaval et al., 2

One of thehe effervescehown in TablF1-F5), constaitric acid exhif formulationitric acid werlightly longormulations

prepared withNaHCO3 exhib171 s and in ncreased with

1105.

Fig. 6. spots) dwhen ein).

 

 

 

 

 

ech Vol6;Issu

he parietal csult in the i

y of nizatidin2007). e parameters nt systems ise 2, the tableant NaHCO3 ribited short fl

ns F6 and F7 re 85 and 92 ger than t

(F1-F5). Th Polyox WSRbited floating these formuh increasing c

Tablets showidue to the presenither of these is

ue 4•December

ell just like improvement ne to reduce

influencing ts their floatits prepared wratio, 10% w/w

floating lag tim containing Ns respectivelyhat obtaine

The formulaR 1105 contain lag time in thlations the flconcentration

ing the interactnce of citric acid absent no inter

r 2013 (extra iss

Ranitidine H of receptor e acid secret

the behaviouring lag time. with HPMC Kw and 2 % w/wme. The lag tiNaHCO3 withy. This time wed with otations (F8-Fning 12% w/whe range of 65floating lag tin of Polyox W

tion (brown colo and nizatidine (raction is seen (m

ssue)

HCl site tion

r of As

K4M w of ime

hout was ther F11) w of 5 to ime

WSR

ored (a-l), m &

Page 9: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

Amarnath

In vitr

Initialprofile ofon the re(PDR, 2formulatiprofile as46±3% in10 h.

From polymer Hof drug frpercentagF4 and F100.61 ± these fivFormulatrelease adrug withwere unaprofile budifferenceformulaticoncentradrug reledrug witwere contnizatidineby exclud

Fig. 7. In v

Reddy et al: D

tro drug releas

lly, we calcuf nizatidine foeported pharm011). It waion should hs 20±3% in 1

n 4 h, 64±3% i

the Figure HPMC K4M hfrom the floatge of drug relF5 was 81.18 4.0 and 99.1

ve formulatiotions F1 and F

as theoretical hin the desirable to sustaiut released thee in the dions was duations of polymease as theorethin the desirtaining citric e. So the for

ding citric acid

vitro dissolution p

Drug-Excipient I

se studies

lated the theor 10 h. The cmacokinetic paas expected have the theo1h, 28±3% inin 6 h, 82±3%

7, it could bhad sustaininting matrix taease from for

8 ± 0.4, 87.5612 ± 2.3 in 1ns (F1-F5) fF2 were unabprofile and a

red time. Formin the drug re drug withinrug release

ue to the pmer. Formulaetical profile red time. Bu acid, which wrmulations wed.

profiles of nizati

Interaction duri

eoretical drugcalculation warameters of that the d

oretical drugn 2 h, 37±3% in 8 h and 10

be observed ng effect on thablet. The cummulations F16 ± 2.7, 94.010 h respectifloated for 10ble to sustain also didn’t relmulations F3release as th

n the desired tprofiles of

presence of ation F5 sustaand also rele

ut these formwas incompatiere further d

idine floating tab

ing Formulation

g release as based the drug

developed g release

% in 3 h, 00±3% in

that the e release mulative

1, F2, F3, 08 ± 1.4, vely. All 0 hours. the drug lease the 3 and F4 heoretical ime. The

various different

ained the eased the mulations ible with

developed

foinflthdththcowpo

WpreCF±resuFrewrepFfogp

blets prepared w

n Development

The cumuormulations Fn 10 hours rloated for 10 he drug relea

drug within thhe drug releahe drug withionsidered as

with HPMC Kprofiles of varif different con

The formulWSR 1105. Frpolymer polyoelease of d

Cumulative peF8, F9, F10 an± 1.4 and 9espectively. Fustain the

Formulations elease as the

within the deelease profile

presence of Formulation ormulation am

good sustainedprofile.

with HPMC K4M

of Gastroreten

ulative percenF6 and F7 warespectively. h. Formulatiase as theorehe desired timase as theoretin the desiredbest formulatK4M. The diious formulatncentrations olations F8-F1rom Figure 8 x WSR 1105

drug from tercentage of dnd F11 were 97.19 ± 0.4 Formulations

drug releaF10 and F11 oretical profilesired time. es of various

different cF10 was co

mong all the fd release ver

M (n = 3).

ntive Drug Deliv

ntage of druas 97.82 ± 1.4These two foion F7 was uetical profile bme. Formulatitical profile ad time. So formtion among alifference in ttions was dueof polymer. 11 were prepa it could be ob had sustainithe floating drug release fr99.14 ± 1.7, 10in 6, 8, 10 F8 and F9 se for the were able to le and also reThe differenformulations

concentrationnsidered as

four formulatiry near to th

very… 22

ug release fr4 and 92.72 ± ormulations a

unable to sustbut released ion F6 sustain

and also releamulation F6 wll these prepathe drug relee to the prese

ared with Polbserved that ing effect on

matrix tabrom formulati00.39 ± 1.0, 98 and 10 howere unable

desired tisustain the deleased the d

nce in the d was due to s of polymbest optimi

ions as it showheoretical rele

289

rom 1.4 also tain the ned

ased was

ared ease ence

lyox the the

blet. ions 8.01 ours e to me.

drug drug drug

the mer. ized wed ease

Page 10: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

2290

Fig. 8. In v

Analy

The dwasanalyfirst ordevalues ofmechanisFickian d0.45 to 0.If the n vSuper cathe releaclear thabecause tzero-ordeF1-F5 anfollowed F6 and formulatibetween 0

For thtest produf2, were optimizedrespectivrespectiv

vitro dissolution p

ysis of in vitro

data obtainedysed by usinger, Higuchi af Korsmeyer-Psms of drug rdiffusion is ap.89 it represevalue reachesse II transpo

ase would follat F6, F7, F10they showed er when compnd F8 followethe Higuchi m

F10 followions (except F0.45 and 0.89 he comparisonucts, “differen calculated. d formulationely. Similarlyely.

profiles of nizati

o drug release

d from in vitrg different moand Peppas ePeppas equatrelease. If thepparent, if thents anomalous 0.89 and abort are indicalowzero order0 and F11 fohigh regressipared to otheed the Peppasmodel. The op

wed zero-ordeF3, F4, F8 a indicating an

n of release prnce factor” f1 a

The similans (F6 and Fy, the differen

idine floating tab

data

tro dissolutionodels viz., zerequation. Diftion indicate e n value is 0e n value ranus diffusion trbove then casated, which shr. From table

ollowed the zeion coefficienters. The forms model. Wheptimized former release.

and F9) have nomalous tranrofiles of referand “similaritarity factor F10) were 78nce factor was

Int J Pha

blets prepared w

n studies ro order, fferent n different

0.45 then nges from ransport. se II and how that e 3 it is ero-order t (R2) for

mulations ereas F9

mulations All the

n value nsport. ence and ty factor”

for the 8 and 86 s 4 and 3

flcoemFdoa1thflScoo

dostthwmre1rest

arm Sci Nanote

with Polyox WSR

In vivo rad

The in vitloating tabletompared to thxpected bec

manufacturer,Figure 9 shodifferent time ne volunteer

appeared at th1.5 h. Later, the intestine. loating tablet

Similarly, meontrolled relenly ≤ 0.5 h, s

Figure 11 different time ne volunteer tomach for thhe tablet sli

within the stommoved from stetention time

12) was 5.33 etention timetate was 1.66

ech Vol6;Issu

R 1105 (n = 3).

iographic stud

itro buoyancys were 240 ±3he original forcause BaSO has a higherows the rad periods of Bar under fasthe same positthe tablet mo The mean gt in fasting ean gastric rease tablet (Cshown in Figushows the r

periods of Baunder fed sta

he first 3 h atightly movedmach till 5 h. omach into th

e of the floatin ± 0.57 h (n of the plain c±0.28 h (n=3)

ue 4•December

dies

y lag time o35 s. The incrrmula of F10 O4, as repr relative den

diographic imaSO4-loaded fting state. Htion in the stoved down frogastric retentstate was 2.retention timCRDDS) in faure 10. adiographic iaSO4-loaded fate.The tablet t different posd downwards In the next phe intestine. Tng tablet in f

n=3). Whereacontrolled rele.

r 2013 (extra iss

of BaSO4-loarease in lag tim (105 ±15 s), wported by nsity (4.5 g/cmmages taken floating tablet

Here, the tabtomach for abom stomach ition time of .0±0.86 h (n=

me of the plasting state w

images takenfloating tablet remained in sitions. Later s, yet remainicture, the tab

The mean gasfed state (Fig

as, mean gasease tablet in

ssue)

ded me, was

its m3). at t in blet

bout into the =3). lain was

n at t in the on, ned blet

stric gure stric fed

Page 11: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

Amarnath

TABLE 3

Regression

Formulatio

F1 F2

F3 F4

F5 F6

F7 F8

F9 F10

F11

Fig. 9. X-ra

Fig. 10. X-

Reddy et al: D

coefficient (R2) v

on Zero-order

R2

0.936 0.896

0.904 0.818

0.980 0.979

0.980 0.820

0.944 0.987

0.995

ay photographs

ray photographs

Drug-Excipient I

values of nizatid

First-order

R2

0.996 0.987

0.924 0.921

0.789 0.804

0.789 0.893

0.831 0.834

0.830

of GRDDS table

s of CRDDS tabl

Interaction duri

dine floating tabl

Higuchi

R2

0.995 0.999

0.992 0.976

0.974 0.958

0.955 0.974

0.985 0.954

0.928

t containing BaS

et (shown by an

ing Formulation

lets for different

Korsmeyer – Pe

R2

0.997 00.999 0

0.994 00.984 0

0.988 00.971 0

0.968 00.992 0

0.965 00.941 0

0.962 0

SO4 (shown by a

arrow) in fastin

n Development

t kinetic models.

ppas

n

.609

.461

.377

.252

.501

.472

.473

.255

.393

.470

.655

n arrow) in fasti

ng state at differe

of Gastroreten

ing state at diffe

ent time points.

ntive Drug Deliv

erent time points

very… 22

s.

291

Page 12: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

2292

Fig. 11. X-

Fig. 12. X-

Conclu

The cwere sucusing HPand NaHexcipientnizatidineincompatexcipientsubjectedF10 wereas optimiselected fThese stucontrolledretentionrelease taand ≤ 0floating tstate werespectivconfirmedretention

ray photographs

ray photographs

usions

controlled releccessfully devPMC K4M anHCO3 as effes compatibilie had an inttibility existes. Drug rel

d to curve fitte best fits for ized. One of thfor the radioudies were cod release tab

n time of floaablets in fasti.5 hours resp

tablets and plaere 5.33 ±0ely in healthyd the superio

n over plain or

s of GRDDS tabl

s of CRDDS tabl

ease floating veloped by efnd Polyox WServescent ageity study, itteraction wit

ed between lease data oting analysis.F zero-order rehe optimized flogical study

onducted in cblets. This rating tablets ing state werepectively. Gasain controlled

0.57 h and y human voluority of GRDr controlled re

let (shown by an

et (shown by an

tablets of nffervescent teSR 1105 as pent. From tht was observth citric acidthe drug anof formulatioFormulationselease and coformulations, by includingomparison wirevealed that

and plain coe 2 ± 0.86 houstric retentiond release table

1.66±0.28 unteers. These

DDS design inlease tablets.

Int J Pha

arrow) in fed st

arrow) in fed st

izatidine echnique polymers he drug-ved that and no

nd other ons was , F6 and

onsidered F10 was g BaSO4. ith plain t gastric ontrolled urs (n=3) n time of ets in fed h (n=3) e studies n gastric

in

aARpPH

A

B

C

arm Sci Nanote

ate at differentt

ate at different t

Conflict of

The authonterest.

Acknowled

One of acknowledges AICTE, New DReddy’s laborproviding nizPharmaceuticaHPMC K4M an

References Arora S, Ali J, A

Drug Delive6(3):E372-E3

Brahma NS and approach to oControl Relea

Caldwell LJ, Garwhich can betime.US Pate

ech Vol6;Issu

ime points.

time points.

interest

rs declare th

gements

the authorthe financ

Delhi, India. Tratories, Pyzatidine andals Ltd., Chnd Polyox WS

Ahuja A, Khar ery Systems: A90. Kim KH (2000).oral controlled dase63:235–259. rdner RC and Cae retained in theent 4735804.

ue 4•December

hat they hav

rs, R. Amcial support The authors dibheemavarad Orchid

hennai, IndiaSR 1105 as gif

RK andBabootA Review. AA

. Floating drug ddrug delivery via

argill RC(1988).De stomach for a

r 2013 (extra iss

ve no conflict

marnath Rereceived fr

acknowledge am, India, Chemicals a

a, for providft samples.

ta S (2005).FloaAPS PharmSciT

delivery systemsa gastric retentio

Drug delivery de controlled perio

ssue)

t of

ddy rom Dr. for

and ding

ating Tech

s: an on. J

evice od of

Page 13: Research Paper IJPSN-6-6-13-KISHAN Drug-Excipient ... · Drug-Excipient Interaction during Formulation Development, in vitro andin vivo Evaluation of Gastroretentive Drug Delivery

Amarnath Reddy et al: Drug-Excipient Interaction during Formulation Development of Gastroretentive Drug Delivery… 2293 Chen GL andHao WH (1998).In vitro performance of floating

sustained release capsule of verapamil. Drug DevInd Pharm24(11):1067-1072.

Davis AF (1997).Compositions containing histamine-H2-receptor antagonists at low dosage.US Patent 5 656 652.

Fix JA, Cargill R and Engle K (1993). Controlled gastric emptying. III. Gastric residence time of a non-disintegrating geometric shape in human volunteers. Pharm Res10(7):1087–1089.

GargR,Gupta GD (2008). Progress in Controlled Gastroretentive Delivery Systems.Trop J Pharm Res7(3):1055-1066.

Gröning R and Heun G (1989).Dosage forms with controlled gastrointestinal passage - studies on the absorption of nitrofurantoin. Int J Pharm56:111-116.

Higuchi T (1961). Rate of release of medicaments from ointment bases containing drugs in suspension. J Pharm Sci50(10):874-875.

Kokate A, Marasanapalle VP, Jasti BR andXiaoling Li (2006).Physiological and biochemical barriers to the drug delivery. In: Design of Controlled Release Drug Delivery Systems (Jasti BR, Ed.).McGraw-Hill, New York, pp. 41-74.

Korsmeyer RW, Gurny R, Doelker E, Buri P andPeppas NA (1983). Mechanisms of solute release from porous hydrophilic polymers. Int J Pharm15(1):25-35.

Lehr CM (1994). Bioadhesion technologies for the delivery of peptide and protein drugs to the gastrointestinal tract.Crit Rev Ther Drug Carrier Syst11(2-3):119-160.

Malinowski HJ andMarroum PJ (1999).Food and drug administration requirements for controlled release products. In: Encyclopedia of controlled drug delivery, Vol. 1 (Mathiowitz E, Ed.).John Wiley and Sons Inc. 381-395.

Mamajek RC, Moyer ES(1980).Drug-dispensing device and method. US Patent 4 207 890.

Moore JW and Flanner HH (1996). Mathematical comparison of curves with an emphasis on in vitro dissolution profiles. Pharm Tech20(6):64–74.

Physicians’ Desk Reference (2011). 65th ed. PDR Network, Montvale, pp. 1298-3000.

Raval JA, Patel JK, Naihong Li and Patel MM (2007). Ranitidine hydrochloride floating matrix tablets based on low density powder: effects of formulation and processing parameters on drug release. Asian JPharm Sci2(4):130-142.

Ritger PL, Peppas NA (1987). A simple equation for description of solute release I: Fickian and non-fickian release from non-swellable devices in the form of slabs, spheres, cylinder or discs. J Control Release5(1):23-36.

Rosa MJC, Ziaa H and Rhodes CT (1994).Design and testing in-vitro of a bioadhesive and floating drug delivery system for oral application.Int J Pharm105(1):65–70.

Rubinstein A and Friend DR (1994).Specific delivery to the gastrointestinal tract. In: Polymeric Site-Specific Pharmacotherapy (Domb AJ, Ed.).Wiley, Chichester pp.282–283.

Schilling SU, Bruce CD, Shah NH,Malick AW andMcGinity JW (2008). Citric acid monohydrate as a release-modifying agent in melt extrudedmatrix tablets. Int J Pharm361:158–168.

Shah MV, De Gennaro MD andSuryakasuma H (1987). An evaluation of albumin microcapsules prepared using a multiple emulsion technique. J Microencapsul.4(3):223-238.

Sheth PR andTossounian JL (1979). Novel sustained release tablet formulations. US Patent 4 167 558.

Urquhart J and Theeuwes F (1984).Drug delivery system comprising a reservoir containing a plurality of tiny pills.US Patent 4 434 153.

Address correspondence to:Prof. V. Kishan, Departmentof Pharmaceutics, University College of PharmaceuticalSciences, Kakatiya University, Warangal-506009, AP, India. Tel: +91 870 2438844; E-mail: [email protected]