Anda di halaman 1dari 26

Journal of Controlled Release 114 (2006) 15 – 40

www.elsevier.com/locate/jconrel

Review
Buccal bioadhesive drug delivery — A promising option
for orally less efficient drugs
Yajaman Sudhakar, Ketousetuo Kuotsu, A.K. Bandyopadhyay ⁎
Buccal Adhesive Research Laboratory, Division of Pharmaceutics, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
Received 4 December 2005; accepted 26 April 2006
Available online 7 July 2006

Abstract

Rapid developments in the field of molecular biology and gene technology resulted in generation of many macromolecular drugs including
peptides, proteins, polysaccharides and nucleic acids in great number possessing superior pharmacological efficacy with site specificity and devoid
of untoward and toxic effects. However, the main impediment for the oral delivery of these drugs as potential therapeutic agents is their extensive
presystemic metabolism, instability in acidic environment resulting into inadequate and erratic oral absorption. Parentral route of administration is
the only established route that overcomes all these drawbacks associated with these orally less/inefficient drugs. But, these formulations are costly,
have least patient compliance, require repeated administration, in addition to the other hazardous effects associated with this route. Over the last
few decades' pharmaceutical scientists throughout the world are trying to explore transdermal and transmucosal routes as an alternative to
injections. Among the various transmucosal sites available, mucosa of the buccal cavity was found to be the most convenient and easily accessible
site for the delivery of therapeutic agents for both local and systemic delivery as retentive dosage forms, because it has expanse of smooth muscle
which is relatively immobile, abundant vascularization, rapid recovery time after exposure to stress and the near absence of langerhans cells.
Direct access to the systemic circulation through the internal jugular vein bypasses drugs from the hepatic first pass metabolism leading to high
bioavailability. Further, these dosage forms are self-administrable, cheap and have superior patient compliance. Developing a dosage form with the
optimum pharmacokinetics is a promising area for continued research as it is enormously important and intellectually challenging. With the right
dosage form design, local environment of the mucosa can be controlled and manipulated in order to optimize the rate of drug dissolution and
permeation. A rational approach to dosage form design requires a complete understanding of the physicochemical and biopharmaceutical
properties of the drug and excipients. Advances in experimental and computational methodologies will be helpful in shortening the processing
time from formulation design to clinical use. This paper aims to review the developments in the buccal adhesive drug delivery systems to provide
basic principles to the young scientists, which will be useful to circumvent the difficulties associated with the formulation design.
© 2006 Elsevier B.V. All rights reserved.

Keywords: Buccal delivery; Bioadhesive; Polymers; Formulation; Permeation enhancers; Evaluation

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
1.1. Buccal mucosal structure and its suitability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
1.2. Absorption pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
1.3. Barriers to penetration across buccal mucosa . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
1.3.1. Membrane coating granules or cored granules . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
1.3.2. Basement membrane . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
1.3.3. Mucus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
1.3.4. Saliva. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19

⁎ Corresponding author. Tel.: +91 9831261813; fax: +91 033 30940712.


E-mail addresses: yajaman_pharma@yahoo.com (Y. Sudhakar), akbju@yahoo.com (A.K. Bandyopadhyay).

0168-3659/$ - see front matter © 2006 Elsevier B.V. All rights reserved.
doi:10.1016/j.jconrel.2006.04.012
16 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

2. Formulation design . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
2.1. Pharmaceutical considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
2.1.1. Buccal adhesive polymers. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
2.2. Physiological considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
2.3. Pharmacological considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
2.4. Permeation enhancers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
2.4.1. Mechanisms of action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
3. Muco/bioadhesion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
3.1. Bio/mucoadhesive forces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
3.1.1. Van der Waal's forces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
3.1.2. Hydrogen bonding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
3.1.3. Disulphide bridging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
3.1.4. Hydration forces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
3.1.5. Electrostatic double-layer forces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
3.1.6. Hydrophobic interactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
3.1.7. Steric forces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
3.1.8. Covalent bonds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
3.2. Methods for measuring mucoadhesion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
3.2.1. Quantitative methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
3.2.2. Qualitative methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
3.3. Factors affecting bio/mucoadhesion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
4. Developments in buccal adhesive drug delivery. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
4.1. Commercial buccal adhesive drug delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
4.2. Research on buccal adhesive drug delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
4.2.1. Solid buccal adhesive formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
4.2.2. Semi-solid dosage forms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
4.2.3. Liquid dosage forms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
4.3. Delivery of proteins and peptides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
5. Evaluation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
5.1. Determination of the residence time . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
5.1.1. In vitro residence time . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
5.1.2. In vivo residence time test . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
5.2. Permeation studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
5.2.1. In vitro methods. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
5.2.2. Ex vivo methods. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
5.2.3. In vivo methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
6. Conclusion. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36

1. Introduction clearance extensive in liver, which often leads to a lack of sig-


nificant correlation between membrane permeability, absorption,
The main impediment to the use of many hydrophilic macro- and bioavailability [1]. Difficulties associated with parenteral
molecular drugs as potential therapeutic agents is their inadequate delivery and poor oral availability provided the impetus for ex-
and erratic oral absorption. The relatively recent evolution of ploring alternative routes for the delivery of such drugs. These
recombinant DNA research and modern synthetic and biotech- include routes such as pulmonary, ocular, nasal, rectal, buccal,
nological methodologies allow the biochemist and chemist to sublingual, vaginal, and transdermal. In absence of external sti-
produce vast quantities of variety of peptides and proteins pos- muli to facilitate absorption, use of these alternative routes has had
sessing better pharmacological efficacy. However, therapeutic limited success. Various strategies have been implemented to
potential of these compounds lies in our ability to design and promote the bioavailability of these drugs, including supplemen-
achieve effective and stable delivery systems. The future chal- tal administration of enzyme inhibitors, use of absorption en-
lenge of pharmaceutical scientists will not only be polypeptide hancers, novel formulation strategies, and reversible chemical
cloning and synthesis, but also to develop effective nonparenteral modifications [2].
delivery of intact proteins and peptides to the systemic circulation. Among the various transmucosal routes, buccal mucosa has
Based on our current understanding of biochemical and phy- excellent accessibility, an expanse of smooth muscle and relatively
siological aspects of absorption and metabolism of many bio- immobile mucosa, hence suitable for administration of retentive
technologically-produced drugs, they cannot be delivered dosage forms. Direct access to the systemic circulation through the
effectively through the conventional oral route. Because after internal jugular vein bypasses drugs from the hepatic first pass
oral administration many drugs are subjected to presystemic metabolism leading to high bioavailability. Other advantages such
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 17

as low enzymatic activity, suitability for drugs or excipients that


mildly and reversibly damages or irritates the mucosa, painless
administration, easy drug withdrawal, facility to include perme-
ation enhancer/enzyme inhibitor or pH modifier in the formulation
and versatility in designing as multidirectional or unidirectional
release systems for local or systemic actions etc, opts buccal
adhesive drug delivery systems as promising option for continued
research [3].
However, the effect of salivary scavenging and accidental
swallowing of delivery system; barrier property of buccal mu-
cosa stands as the major limitations in the development of
buccal adhesive drug delivery systems.
Our intent, therefore, is to discuss the implication of various
approaches for buccal adhesive delivery strategies applied for
the systemic delivery of orally less/in efficient drugs, in addition
to the widely used local drug delivery.

1.1. Buccal mucosal structure and its suitability


Fig. 1. Cross-section of buccal mucosa.
Buccal region is that part of the mouth bounded anteriorly and
laterally by the lips and the cheeks, posteriorly and medially by 1.2. Absorption pathways
the teeth and/or gums, and above and below by the reflections of
the mucosa from the lips and cheeks to the gums. Numerous Studies with microscopically visible tracers such as small
racemose, mucous, or serous glands are present in the submucous proteins [11] and dextrans [12] suggest that the major pathway
tissue of the cheeks [4]. The buccal glands are placed between the across stratified epithelium of large molecules is via the inter-
mucous membrane and buccinator muscle: they are similar in cellular spaces and that there is a barrier to penetration as a result of
structure to the labial glands, but smaller. About five, of a larger modifications to the intercellular substance in the superficial
size than the rest, are placed between the masseter and buccinator layers. However, rate of penetration varies depending on the
muscles around the distal extremity of the parotid duct; their ducts physicochemical properties of the molecule and the type of tissue
open in the mouth opposite the last molar tooth. They are called being traversed. This has led to the suggestion that materials uses
molar glands [5]. Maxillary artery supplies blood to buccal mu- one or more of the following routes simultaneously to cross the
cosa and blood flow is faster and richer (2.4 ml/min/cm2) than that barrier region in the process of absorption, but one route is pre-
in the sublingual, gingival and palatal regions, thus facilitates dominant over the other depending on the physicochemical pro-
passive diffusion of drug molecules across the mucosa. The perties of the diffusant [13].
thickness of the buccal mucosa is measured to be 500–800 μm
and is rough textured, hence suitable for retentive delivery sys- ➢ Passive diffusion
tems [6]. The turnover time for the buccal epithelium has been ◦ Transcellular or intracellular route (crossing the cell
estimated at 5–6 days [7]. membrane and entering the cell)
Buccal mucosa composed of several layers of different cells as ◦ Paracellular or intercellular route (passing between the
shown in Fig. 1. The epithelium is similar to stratified squamous cells)
epithelia found in rest of the body and is about 40–50 cell layers ➢ Carrier mediated transport
thick [5]. Lining epithelium of buccal mucosa is the nonkeratinized ➢ Endocytosis
stratified squamous epithelium that has thickness of approximately
500–600 μ and surface area of 50.2 cm2. Basement membrane, The flux of drug through the membrane under sink condition
lamina propria followed by the submucosa is present below the for paracellular route can be written as Eq. (1)
epithelial layer [8]. Lamina propria is rich with blood vessels and
Dp e
capillaries that open to the internal jugular vein. Lipid analysis of Jp ¼ Cd ð1Þ
buccal tissues shows the presence of phospholipid 76.3%, glu- hp
cosphingolipid 23.0% and ceramide NS at 0.72%. Other lipids Where, Dp is diffusion coefficient of the permeate in the inter-
such as acyl glucosylated ceramide, and ceramides like Cer AH, cellular spaces, hp is the path length of the paracellular route, ε is the
Cer AP, Cer NH, Cer AS, and EOHP/NP are completely absent [9]. area fraction of the paracellular route and Cd is the donor drug
The primary function of buccal epithelium is the protection of concentration.
the underlying tissue. In nonkeratinized regions, lipid-based Similarly, flux of drug through the membrane under sink
permeability barriers in the outer epithelial layers protect the condition for transcellular route can be written as Eq. (2).
underlying tissues against fluid loss and entry of potentially
harmful environmental agents such as antigens, carcinogens, ð1−eÞDc Kc
Jc ¼ Cd ð2Þ
microbial toxins and enzymes from foods and beverages [10]. hc
18 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

Where, Kc is partition coefficient between lipophilic cell mem- morphology is far less marked than in keratinized epithelia. The
brane and the aqueous phase, Dc is the diffusion coefficient of mature cells in the outer portion of nonkeratinized epithelia be-
the drug in the transcellular spaces and hc is the path length of come large and flat, retain nuclei and other organelles and the
the transcellular route [14]. cytokeratins do not aggregate to form bundles of filaments as seen
In very few cases absorption also takes place by the process of in keratinizing epithelia. As cells reach the upper third to quarter of
endocytosis where the drug molecules were engulfed by the the epithelium, membrane-coating granules become evident at the
cells. It is unlikely that active transport processes operate within superficial aspect of the cells and appear to fuse with the plasma
the oral mucosa; however, it is believed that acidic stimulation of membrane so as to extrude their contents into the intercellular
the salivary glands, with the accompanying vasodilatation, faci- space. The membrane-coating granules found in nonkeratinizing
litates absorption and uptake into the circulatory system [15]. epithelia are spherical in shape, membrane-bounded and measure
The absorption potential of the buccal mucosa is influenced by about 0.2 μm in diameter [25]. Such granules have been observed
the lipid solubility and molecular weight of the diffusant. Ab- in a variety of other human nonkeratinized epithelia, including
sorption of some drugs via the buccal mucosa is found to increase uterine cervix [26] and esophagus [27]. However, current studies
when carrier pH is lowered and decreased with an increase of pH employing ruthenium tetroxide as a post-fixative indicate that in
[16]. However, the pH dependency that is evident in absorption of addition to cored granules, a small proportion of the granules in
ionizable compounds reflects their partitioning into the epithelial nonkeratinized epithelium do contain lamellae, which may be the
cell membrane, so it is likely that such compounds will tend to source of short stacks of lamellar lipid scattered throughout the
penetrate transcellularly [17]. Weak acids and weak bases are intercellular spaces in the outer portion of the epithelium. In
subjected to pH-dependent ionization. It is presumed that ionized contrast to the intercellular spaces of stratum corneum, those of the
species penetrate poorly through the oral mucosa compared with superficial layer of nonkeratinizing epithelia contain electron lu-
non-ionized species. An increase in the amount of non-ionized cent material, which may represent nonlamellar phase lipid, with
drug is likely to increase the permeability of the drug across an only occasional short stacks of lipid lamellae.
epithelial barrier, and this may be achieved by a change of pH of the
drug delivery system. It has been reported that pH has effect on the 1.3.2. Basement membrane
buccal permeation of drug through oral mucosa [18]. The diffusion Although the superficial layers of the oral epithelium represent
of drugs across buccal mucosa was not related to their degree of the primary barrier to the entry of substances from the exterior, it is
ionization as calculated from the Henderson–Hasselbalch equation evident that the basement membrane also plays a role in limiting the
and thus it is not helpful in the prediction of membrane diffusion of passage of materials across the junction between epithelium and
weak acidic and basic drugs [19]. connective tissue. A similar mechanism appears to operate in the
In general, for peptide drugs, permeation across the buccal opposite direction. The charge on the constituents of the basal
epithelium is thought to be through paracellular route by passive lamina may limit the rate of penetration of lipophilic compounds that
diffusion. Recently, it was reported that drugs that have a mono- can traverse the superficial epithelial barrier relatively easily [28].
carboxylic acid residue could be delivered into systemic circulation
from the oral mucosa via its carrier [20]. The permeability of oral 1.3.3. Mucus
mucosa and the efficacy of penetration enhancers have been inves- The epithelial cells of buccal mucosa are surrounded by the
tigated in numerous in vivo and in vitro models. Various kinds of intercellular ground substance called mucus with the thickness
diffusion cells, including continuous flow perfusion chambers, varies from 40 μm to 300 μm [29]. Though the sublingual glands
Ussing chambers, Franz cells and Grass–Sweetana, have been used and minor salivary glands contribute only about 10% of all saliva,
to determine the permeability of oral mucosa [21]. Cultured epi- together they produce the majority of mucus and are critical in
thelial cell lines have also been developed as an in vitro model for maintaining the mucin layer over the oral mucosa [30]. It serves as
studying drug transport and metabolism at biological barriers as an effective delivery vehicle by acting as a lubricant allowing cells
well as to elucidate the possible mechanisms of action of pene- to move relative to one another and is believed to play a major role
tration enhancers [22,23]. Recently, TR146 cell culture model was in adhesion of mucoadhesive drug delivery systems [31]. At buccal
suggested as a valuable in vitro model of human buccal mucosa for pH, mucus can form a strongly cohesive gel structure that binds to
permeability and metabolism studies with enzymatically labile the epithelial cell surface as a gelatinous layer [8]. Mucus mole-
drugs, such as leu-enkefalin, intended for buccal drug delivery [24]. cules are able to join together to make polymers or an extended
three-dimensional network. Different types of mucus are produced,
1.3. Barriers to penetration across buccal mucosa for example G, L, S, P and F mucus, which form different network
of gels. Other substances such as ions, protein chains, and enzymes
The barriers such as saliva, mucus, membrane coating granules, are also able to modify the interaction of the mucus molecules and,
basement membrane etc retard the rate and extent of drug ab- as a consequence, their biophysical properties [32].
sorption through the buccal mucosa. The main penetration barrier Mucus is composed chiefly of mucins and inorganic salts
exists in the outermost quarter to one third of the epithelium [8]. suspended in water. Mucins are a family of large, heavily gly-
cosylated proteins composed of oligosaccharide chains attached
1.3.1. Membrane coating granules or cored granules to a protein core. Three quarters of the protein core are heavily
In nonkeratinized epithelia, the accumulation of lipids and glycosylated and impart a gel like characteristic to mucus. Mucins
cytokeratins in the keratinocytes is less evident and the change in contain approximately 70–80% carbohydrate, 12–25% protein
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 19

and up to 5% ester sulphate [33]. The dense sugar coating of by bathing the mouth in copious neutralizing fluid [44]. With
mucins gives them considerable water-holding capacity and also stimulation of salivary secretion, oxygen is consumed and
makes them resistant to proteolysis, which may be important in vasodilator substances are produced; and the glandular blood
maintaining mucosal barriers [4]. flow increases, due to increased glandular metabolism [45]. Saliva
Mucins are secreted as massive aggregates by prostaglandins is composed of 99.5% water in addition to proteins, glycoproteins
with molecular masses of roughly 1 to 10 million Da. Within these and electrolytes. It is high in potassium (7 × plasma), bicarbonate
aggregates, monomers are linked to one another mostly by non- (3 × plasma), calcium, phosphorous, chloride, thiocyanate and urea
covalent interactions, although intermolecular disulphide bonds and low in sodium (1/10 × plasma). The normal pH of saliva is 5.6–
also play a role in this process. Oligosaccharide side chains con- 7. Saliva contains enzymes namely α-amylase (breaks 1–4
tain an average of about 8–10 monosaccharide residues of five glycosidic bonds), lysozyme (protective, digests bacterial cell
different types namely L-fucose, D-galactose, N-acetyl-D-glucos- walls) and lingual lipase (break down the fats) [46].
amine, N-acetyl-D-galactosamine and sialic acid. Amino acids Saliva serves multiple important functions. It moistens the
present are serine, threonine and proline [34]. Because of the mouth, initiates digestion and protects the teeth from decay. It also
presence of sialic acids and ester sulfates, mucus is negatively controls bacterial flora of the oral cavity. Because saliva is high in
charged at physiological salivary pH of 5.8–7.4 [8]. calcium and phosphate, it plays a role in mineralization of new
At least 19 human mucin genes have been distinguished by teeth repair and precarious enamel lesions. It protects the teeth by
cDNA cloning-MUC1, 2, 3A, 3B, 4,5AC, 5B, 6–9, 11–13, and forming “protective pellicle”. This signifies a saliva protein coat on
15–19. Mucin genes encode mucin monomers that are synthesized the teeth, which contains antibacterial compounds. Thus, problems
as rod-shaped apomucin covers that are post translationally modi- with the salivary glands generally result in rampant dental caries.
fied by exceptionally abundant glycosylation. Two distinctly dif- Lysozyme, secretory IgA, and salivary peroxidase play important
ferent regions are found in mature genes. The amino- and carboxy- roles in saliva's antibacterial actions. Lysozyme agglutinates bac-
terminal regions are very lightly glycosylated, but rich in cysteines, teria and activates autolysins. Ig A interferes with the adherence of
which are likely, involved in establishing disulfide linkages within microorganisms to host tissue. Peroxidase breaks down salivary
and among mucin monomers. A large central region formed of thiocyanate, which in turn, oxidizes the enzymes involved in
multiple tandems repeats of 10 to 80 residue sequences in which up bacterial glycolysis. However, salivary flow rate may play role in
to half of the amino acids are serine or threonine. This area becomes oral hygiene. Intraoral complications of salivary hypofunction may
saturated with hundreds of O-linked oligosaccharides also found cause candidiasis, oral lichen planus, burning mouth syndrome,
on mucins, but much less abundantly [4]. recurrent aphthous ulcers and dental caries. A constant flowing
Mucins are characterized not only by large molecular masses down of saliva within the oral cavity makes it very difficult for
but also by large molecular mass distributions, as seen by analytical drugs to be retained for a significant amount of time in order to
ultra centrifugation, and by the powerful technique of size facilitate absorption in this site [44,45]. The other important factor
exclusion chromatography coupled to multi-angle laser light sca- of great concern is the role of saliva in development of dental
ttering [35,36]. In solution, mucins adopt a random-coil confor- caries. Salivary enzymes act on natural polysaccharidic polymers
mation [37], occupying a time averaged spheroidal domain as that hasten the growth of mutants of streptococci and other plaque
shown by hydrodynamics and critical-point-drying electron mi- bacteria leading to development of dental caries.
croscopy. Mucins, which are different, are the submaxillary mu- In general, intercellular spaces pose as the major barrier to
cins, with a lower carbohydrate content and different structure [38]. permeation of lipophilic compounds, and the cell membrane
which is lipophilic in nature acts as the major transport barrier for
1.3.4. Saliva hydrophilic compounds because it is difficult to permeate through
The mucosal surface has a salivary coating estimated to be the cell membrane due to a low partition coefficient [13].
70 μm thick [39], which act as unstirred layer. Within the saliva Permeabilities between different regions of the oral cavity vary
there is a high molecular weight mucin named MG1 [40] that can greatly because of the diverse structures and functions. In general,
bind to the surface of the oral mucosa so as to maintain hydration, the permeability is based on the relative thickness and degree of
provide lubrication, concentrate protective molecules such as keratinization of these tissues in the order of sublingual > buc-
secretory immunoglobulins, and limit the attachment of micro- cal > palatal. The permeability of the buccal mucosa was estimated
organisms. Several independent lines of evidence suggest that to be 4–4000 times greater than that of the skin [47].
saliva and salivary mucin contribute to the barrier properties of oral
mucosa [41]. The major salivary glands consist of lobules of cells 2. Formulation design
that secrete saliva; parotids through salivary ducts near the upper
teeth, submandibular under the tongue, and the sublingual through Buccal adhesive drug delivery systems with the size 1–3 cm2
many ducts in the floor of the mouth. Besides these glands, there and a daily dose of 25 mg or less are preferable. The maximal
are 600–1000 tiny glands called minor salivary glands located in duration of buccal delivery is approximately 4–6 h [3].
the lips, inner cheek area (buccal mucosa), and extensively in other
linings of the mouth and throat [42]. Total output from the major 2.1. Pharmaceutical considerations
and minor salivary glands is termed as whole saliva, which at
normal conditions has flow rate of 1–2 ml/min [43]. Greater Great care needs to be exercised while developing a safe
salivary output avoids potential harm to acid-sensitive tooth enamel and effective buccal adhesive drug delivery device. Factors
20 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

Table 1
Properties and characteristics of some representative bioadhesive polymers
Bioadhesives Properties Characteristics
Polycarbophil (polyacrylic acid crosslinked with • Mw 2.2 × 105 • Synthesized by lightly crosslinking of 0.5–1%
divinyl glycol) • η 2000–22,500 cps (1% aq. soln.) w/w divinyl glycol
• κ 15–35 mL/g in acidic media (pH 1–3) 100 mL/g in • Swellable depending on pH and ionic strength.
neutral and basic media • Swelling increases as pH increases.
• φ viscous colloid in cold water • At pH 1–3, absorbs 15–35 ml of water per gram but
• Insoluble in water, but swell to varying degrees in absorbs 100 ml per gram at neutral and alkaline pH.
common organic solvents, strong mineral acids, and • Entangle the polymer with mucus on the surface of
bases. the tissue
• Hydrogen bonding between the nonionized
carboxylic acid and mucin.
Carbopol/carbomer (carboxy polymethylene) • Pharmaceutical grades: 934 P, 940 P, 971 P and 974 P. • Synthesized by cross-linker of allyl sucrose or
empirical formula: (C3H4O2)x (C3H5–Sucrose)y • Mw 1 × 106–4 × 106 allyl pentaerythritol
• η 29,400–39,400 cps at 25 °C with 0.5% neutralized • Excellent thickening, emulsifying, suspending,
aqueous solution. gelling agent.
• κ 5 g/cm3 in bulk, 1.4 g/cm3 tapped. • Common component in bioadhesive dosage forms.
• pH 2.5–3.0 • Gel looses viscosity on exposure to sunlight.
• φ water, alcohol, glycerin • Unaffected by temperature variations, hydrolysis,
• White, fluffy, acidic, hygroscopic powder with a slight oxidation and resistant to bacterial growth.
characteristic odour. • It contributes no off-taste and may mask the
undesirable taste of the formulation.
• Incompatible with Phenols, cationic polymers,
high concentrations of electrolytes and resorcinol.
Sodium carboxymethyl cellulose SCMC (cellulose • It is an anionic polymer made by swelling cellulose • Emulsifying, gelling, binding agent
carboxymethyl ether sodium salt) empirical with NaOH and then reacting it with monochloroacetic • Sterilization in dry and solution form, irradiation
formula: [C6H7O2(OH)3x (OCH2–COONa)x]n acid. of solution loses the viscosity.
• Grades H, M, and L • Stable on storage.
• Mw 9 × 104–7 × 105 • Incompatible with strongly acidic solutions
• η 1200 cps with 1.0% soln. • In general, stability with monovalent salts is very
• ρ 0.75 g/cm3 in bulk good; with divalent salts good to marginal; with
• pH 6.5–8.5 trivalent and heavy metal salts poor, resulting in
• φ water gelation or precipitation.
• White to faint yellow, odorless, hygroscopic powder or • CMC solutions offer good tolerance of water
granular material having faint paper-like taste. miscible solvents, good viscosity stability over the
pH 4 to pH 10 range, compatibility with most water
soluble nonionic gums, and synergism with HEC
and HPC.
• Most CMC solutions are thixotropic; some are
strictly pseudoplastic.
• All solutions show a reversible decrease in viscosity at
elevated temperatures. CMC solutions lack yield value.
• Solutions are susceptible to shear, heat, bacterial,
enzyme, and UV degradation.
• Good bioadhesive strength.
• Cell immobilization via a combination of ionotropic
gelation and polyelectrolyte complex formation (e.g.,
with chitosan) in drug delivery systems and dialysis
membranes.
Hydroxypropyl cellulose partially substituted • Grades: Klucel EF, LF, JF, GF, MF and HF • Best pH is between 6.0 and 8.0.
polyhydroxy propylether of cellulose HPC • Mw 6 × 104–1 × 106 • Solutions of HPC are susceptible to shear, heat,
(cellulose 2-hydroxypropyl ether) empirical • η 4–6500 cps with 2.0% aq. soln. bacterial, enzymatic and bacterial degradation.
formula: (C15H28O8)n • pH 5.0–8.0 • It is inert and showed no evidence of skin irritation
• ρ 0.5 g/cm3 in bulk or sensitization.
• Soluble in water below 38 °C, ethanol, propylene • Compatible with most water-soluble gums and resins.
glycol, dioxane, methanol, isopropyl alcohol, dimethyl • Synergistic with CMC and sodium alginate.
sulphoxide, dimethyl formamide etc. • Not metabolized in the body.
• Insoluble in hot water • It may not tolerate high concentrations of
• White to slightly yellowish, odorless powder. dissolved materials and tend to be salting out.
• It is also incompatible with the substituted
phenolic derivatives such as methyl and propyl
parahydroxy benzoate.
• Granulating and film coating agent for tablet
• Thickening agent, emulsion
• Stabilizer, suspending agent in oral and topical
solution or suspension
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 21

Table 1 (continued )
Bioadhesives Properties Characteristics
Hydroxypropylmethyl Cellulose HPMC (cellulose • Methocel E5, E15, E50, E4M, F50, F4M, K100, K4M, • Mixed alkyl hydroxyalkyl cellulosic ether
2-hydroxypropylmethyl ether) empirical formula: K15M, K100M. • Suspending, viscosity-increasing and film-
C8H15O6–(C10H18O6)n–C8H15O5 • Mw 8.6 × 104 forming agent
• η E15–15 cps, E4M–400 cps and K4M–4000 cps • Tablet binder and adhesive ointment ingredient
(2% aqueous solution.) • E grades are generally suitable as film formers
• φ Cold water, mixtures of methylene chloride and while the K grades are used as thickeners.
isopropylalcohol. • Stable when dry.
• Insoluble in alcohol, chloroform and ether. • Solutions are stable at pH 3.0 to 11.0
• Odorless, tasteless, white or creamy white fibrous or • Incompatible to extreme pH conditions and
granular powder. oxidizing materials.
Hydroxyethyl Cellulose non-ionic polymer made • Available in grades ranging from 2 to 8,00,000 cps at 2%. • Solutions are pseudoplastic and show a reversible
by swelling cellulose with NaOH and treating with • Light tan or cream to white powder, odorless and decrease in viscosity at elevated temperatures.
ethylene oxide. tasteless. It may contain suitable anticaking agents. • HEC solutions lack yield value.
• ρ 0.6 g/mL • Solutions show only a fair tolerance with water
• pH 6–8.5 miscible solvents (10 to 30% of solution weight).
• φ in hot or cold water and gives a clear, colorless • Compatible with most water-soluble gums and resins.
solution. • Synergistic with CMC and sodium alginate.
• Susceptible for bacterial and enzymatic degradation.
• Polyvalent inorganic salts will salt out HEC at
lower concentrations than monovalent salts.
• Shows good viscosity stability over the pH 2 to pH
12 ranges.
• Used as suspending or viscosity builder
• Binder, film former.
Xanthan gum xanthan gum is an anionic • It is soluble in hot or cold water and gives visually • Xanthan gum is more tolerant of electrolytes, acids
polysaccharide derived from the fermentation of hazy, neutral pH solutions. and bases than most other organic gums.
the plant bacteria Xanthamonas campestris • It will dissolve in hot glycerin. • It can, nevertheless, be gelled or precipitated with
• Solutions are typically in the 1500 to 2500 cps range at certain polyvalent metal cations under specific
1%; they are pseudoplastic and especially shear-thinning. circumstances.
In the presence of small amounts of salt, solutions shows • Solutions show very good viscosity stability over
good viscosity stability at elevated temperatures. the pH 2 to 12 range and good tolerance of water-
• Solutions possess excellent yield value. miscible solvents.
• It is more compatible with most nonionic and
anionic gums, featuring useful synergism with
galactomannans.
• It is more resistant to shear, heat, bacterial,
enzyme, and UV degradation than most gums.
Guar gum (galactomannan polysaccharide) empirical • Obtained from the ground endosperms of the seeds of • Stable in solution over a pH range of 1.0–10.5.
formula: (C6H12O6)n consists chiefly of a high Cyamposis tetyragonolobus (family leguminosae). • Prolonged heating degrades viscosity.
molecular weight hydrocolloid polysaccharide • MW approx. 220,000 Bacteriological stability can be improved by the
composed of galactan and mannan units combined • η 2000–22500 Cps (1% aqueous solution.) addition of mixture of 0.15% methyl paraben or
through glycosidic linkages • Forms viscous colloidal solution when hydrated in 0.1% benzoic acid.
cold water. The optimum rate of hydration is between pH • The FDA recognizes guar gum as a substance
7.5 and 9.0. added directly to human food and has been affirmed
as generally recognized as safe.
• Incompatible with acetone, tannins, strong acids,
and the alkalis. Borate ions, if present in the
dispersing water, will prevent hydration of guar.
• Used as thickener for lotions and creams, as tablet
binder, and as emulsion stabilizer.
Hydroxypropyl Guar non-ionic derivative of guar. • Φ in hot and cold water • Compatible with high concentration of most salts.
Prepared by reacting guar gum with propylene • Gives high viscosity, pseudoplastic solutions that show • Shows good tolerance of water miscible solvents.
oxide. reversible decrease in viscosity at elevated temperatures. • Better compatibility with minerals than guar gum.
• Lacks yield value. • Good viscosity stability in the pH range of 2 to 13.
• More resistance to bacterial and enzymatic degradation.
Chitosan a linear polysaccharide composed of • Prepared from chitin of crabs and lobsters by N- • Mucoadhesive agent due to either secondary
randomly distributed β-(1-4)-linked D- deacetylation with alkali. chemical bonds such as hydrogen bonds or ionic
glucosamine (deacetylated unit) and N-acetyl-D- • Φ dilute acids to produce a linear polyelectrolyte with interactions between the positively charged amino
glucosamine (acetylated unit). a high positive charge density and forms salts with groups of chitosan and the negatively charged sialic
inorganic and organic acids such as glutamic acid, acid residues of mucus glycoproteins or mucins.
hydrochloric acid, lactic acid, and acetic acid. • Possesses cell-binding activity due to polymer
• The amino group in chitosan has a pKa value of ∼6.5, cationic polyelectrolyte structure and to the negative
thus, chitosan is positively charged and soluble in acidic charge of the cell surface.
to neutral solution with a charge density dependent on • Biocompatible and biodegradable.
pH and the %DA-value. • Excellent gel forming and film forming ability.
(continued on next page)
22 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

Table 1 (continued )
Bioadhesives Properties Characteristics
• Widely used in controlled delivery systems such as
gels, membranes, microspheres.
• Chitosan enhance the transport of polar drugs
across epithelial surfaces. Purified qualities of
chitosans are available for biomedical applications.
Chitosan and its derivatives such as trimethylchitosan
(where the amino group has been trimethylated)
have been used in non-viral gene delivery.
Trimethylchitosan, or quaternised chitosan, has
been shown to transfect breast cancer cells. As the
degree of trimethylation increases the cytotoxicity
of the derivative increases. At approximately 50%
trimethylation the derivative is the most efficient at
gene delivery. Oligomeric derivatives (3–6 kDa) are
relatively non-toxic and have good gene delivery
properties.
Carrageenan an anionic polysaccharide, extracted • Available in sodium, potassium, magnesium, calcium • All solutions are pseudoplastic with some degree
from the red seaweed Chondrus crispus. and mixed cation forms. of yield value. Certain ca-Iota solutions are
• Three structural types exist: Iota, Kappa, and thixotropic. Lambda is non-gelling, Kappa can
Lambda, differing in solubility and rheology. produce brittle gels; Iota can produce elastic gels.
• The sodium form of all three types is soluble in both All solutions show a reversible decrease in viscosity
cold and hot water. at elevated temperatures. Iota and Lambda
• Other cation forms of kappa and Iota are soluble only carrageenan have excellent electrolyte tolerance;
in hot water. kappa's being somewhat less. Electrolytes will
• All forms of lambda are soluble in cold water. however decreases solution viscosity. The best
solution stability occurs in the pH 6 to 10. It is
compatible with most nonionic and anionic water-
soluble thickeners. It is strongly synergistic with
locust bean gum and strongly interactive with
proteins. Solutions are susceptible to shear and
heat degradation.
• Excellent thermoreversible properties.
• Used also for microencapsulation.
Sodium Alginate consists chiefly of the alginic acid, • Purified carbohydrate product extracted from brown • Safe and nonallergenic.
a polyuronic acid composed of β-D-mannuronic seaweed by the use of dilute alkali. • Incompatible with acridine derivatives, crystal
acid residues. empirical formula: (C6H7O6Na)n • Occurs as a white or buff powder, which is odorless violet, phenyl mercuric nitrate and acetate, calcium
anionic polysaccharide extracted principally from and tasteless. salts, alcohol in concentrations greater than 5%, and
the giant kelp Macrocystis Pyrifera as alginic acid • pH 7.2 heavy metals.
and neutralized to sodium salt. • η 20–400 Cps (1% aqueous solution.) • Stabilizer in emulsion, suspending agent, tablet
• φ Water, forming a viscous, colloidal solution. disintegrant, tablet binder.
• Insoluble in other organic solvents and acids where the • It is also used as haemostatic agent in surgical
pH of the resulting solution and acids where the pH of dressings
the resulting solution falls below 3.0. • Excellent gel formation properties
• Biocompatible
• Microstructure and viscosity are dependent on the
chemical composition.
• Used as immobilization matrices for cells
and enzymes, controlled release of bioactive
substances, injectable microcapsules for treating
neurodegenerative and hormone deficiency
diseases.
• Lacks yield value.
• Solutions show fair to good tolerance of water
miscible solvents (10–30% of volatile solvents; 40–
70% of glycols)
• Compatible with most water-soluble thickeners
and resins.
• Its solutions are more resistant to bacterial and
enzymatic degradation than many other organic
thickeners.
Poly (hydroxy butyrate), Poly (e-caprolactone) • Biodegradable • Used as a matrix for drug delivery systems, cell
and copolymers • Properties can be changed by chemical modification, microencapsulation.
copolymerization and blending.
Poly (ortho esters) • Surface eroding polymers. • Application in sustained drug delivery and
ophthalmology.
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 23

Table 1 (continued )
Bioadhesives Properties Characteristics
Poly (cyano acrylates) • Biodegradable depending on the length of the alkyl • Used as surgical adhesives and glues.
chain. • Potentially used in drug delivery.
Polyphosphazenes • Can be tailored with versatile side chain functionality • Can be made into films and hydrogels.
• Applications in drug delivery.
Poly (vinyl alcohol) • Biocompatible • Gels and blended membranes are used in drug
delivery and cell immobilization.
Poly (ethylene oxide) • Highly biocompatible. • Its derivatives and copolymers are used in various
biomedical applications.
Poly (hydroxytheyl methacrylate) • Biocompatible • Hydrogels have been used as soft contact lenses,
for drug delivery, as skin coatings, and for
immunoisolation membranes.
Poly (ethylene oxide-b-propylene oxide) • Surfactants with amphiphilic properties. • Used in protein delivery and skin treatments.

influencing drug release and penetration through buccal ✓ pH should be biocompatible and should possess good
mucosa, organoleptic factors, and effects of additives used viscoelastic properties.
to improve drug release pattern and absorption, the effects of ✓ Should adhere quickly to buccal mucosa and should
local drug irritation caused at the site of application are to be possess sufficient mechanical strength.
considered while designing a formulation. ✓ Should possess peel, tensile and shear strengths at the
bioadhesive range.
2.1.1. Buccal adhesive polymers ✓ Polymer must be easily available and its cost should not
Polymer is a generic term used to describe a very long be high.
molecule consisting of structural units and repeating units ✓ Should show bioadhesive properties in both dry and
connected by covalent chemical bonds. The term is derived liquid state.
from the Greek words: polys meaning many, and meros ✓ Should demonstrate local enzyme inhibition and penetra-
meaning parts [4]. The key feature that distinguishes tion enhancement properties.
polymers from other molecules is the repetition of many ✓ Should demonstrate acceptable shelf life.
identical, similar, or complementary molecular subunits in ✓ Should have optimum molecular weight.
these chains. These subunits, the monomers, are small ✓ Should possess adhesively active groups.
molecules of low to moderate molecular weight, and are ✓ Should have required spatial conformation.
linked to each other during a chemical reaction called ✓ Should be sufficiently cross-linked but not to the degree
polymerization. Instead of being identical, similar monomers of suppression of bond forming groups.
can have varying chemical substituents. The differences ✓ Should not aid in development of secondary infections
between monomers can affect properties such as solubility, such as dental caries.
flexibility, and strength. The term buccal adhesive polymer
covers a large, diverse group of molecules, including 2.1.1.2. Some representative polymers:
substances from natural origin to biodegradable grafted co- 2.1.1.2.1. Hydrogels. Hydrogels, often called as “wet”
polymers and thiolated polymers. adhesives because they require moisture to exhibit the
Bioadhesive formulations use polymers as the adhesive adhesive property. They are usually considered to be cross
component. These formulations are often water soluble and linked water swollen polymers having water content ranging
when in a dry form attract water from the biological surface from 30% to 40% depending on the polymer used. These are
and this water transfer leads to a strong interaction. These hydrophilic matrices that absorb water when placed in an
polymers also form viscous liquids when hydrated with water aqueous media. This may be supplied by the saliva, which
that increases their retention time over mucosal surfaces and may also act as the dissolution medium. They are structured
may lead to adhesive interactions. Bioadhesive polymers in such a manner that the crosslinking fibers present in their
should possess certain physicochemical features including matrix effectively prevent them from being dissolved and thus
hydrophilicity, numerous hydrogen bond-forming groups, help them in retaining water. When drugs are loaded into
flexibility for interpenetration with mucus and epithelial these hydrogels, as water is absorbed into the matrix, chain
tissue, and visco-elastic properties [48]. relaxation occurs and drug molecules are released through the
spaces or channels within the hydrogel network. Polymers
2.1.1.1. Ideal characteristics. such as polyacrylates (carbopol and polycarbophil), ethylene
vinyl alcohol, polyethylene oxide, poly vinyl alcohol, poly
✓ Polymer and its degradation products should be non-toxic, (N-acryloylpyrrolidine), polyoxyethylenes, self cross linked
non-irritant and free from leachable impurities. gelatin, sodium alginate, natural gums like guar gum, karaya
✓ Should have good spreadability, wetting, swelling and gum, xanthan gum, locust bean gum and cellulose ethers like
solubility and biodegradability properties. methyl cellulose, hydroxypropyl cellulose, hydroxy propyl
24 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

methyl cellulose, sodium carboxy methyl cellulose etc. form 2.1.1.2.5. Milk protein. A particular example is a milk
part of the family of hydrogels [49]. protein concentrate containing a minimum of 85% of proteins
2.1.1.2.2. Copolymers. Researchers are currently working such as Prosobel L85, LR85F at concentration of 15% to 50%,
on carrier systems containing block copolymers rather than preferably 20% to 30% in a bioadhesive tablet showed good
using single polymeric system. Copolymerization with two or bioadhesive property [57].
more different monomers results in chains with varied 2.1.1.2.6. In general.
properties. A block copolymer is formed when the reaction is
carried out in a stepwise manner, leading to a structure with long ➢ Cationic and anionic polymers bind more effectively than
sequences or blocks of one monomer alternating with long neutral polymers.
sequences of the other. These networks when composed of ➢ Anionic polymers with sulphate groups bind more
hydrophilic and hydrophobic monomers are called polymer effectively than those with carboxylic groups.
micelle. These micelles are suitable for enclosing individual ➢ Polyanions are better than polycations in terms of binding
drug molecules. Their hydrophilic outer shells help to protect potential and toxicity.
the cores and their contents from chemical attack by aqueous ➢ Water-insoluble polymers give greater flexibility in
medium. Most micelle-based systems are formed from poly dosage form design compared to rapidly or slowly dis-
(ethylene oxide)-b-polypropylene-b-poly (ethylene oxide) tri- solving water-soluble polymers.
block network. ➢ Degree of binding is proportional to the charge density on
There are also graft copolymers, in which entire chains of one the polymer.
kind (e.g., polystyrene) are made to grow out of the sides of
chains of another kind (e.g., polybutadiene), resulting in a Some of the properties and characteristics of buccal adhesive
product that is less brittle and more impact-resistant. Thus, block polymers are listed in Table 1.
and graft copolymers can combine the useful properties of both
constituents and often behave as quasi-two-phase systems [50]. 2.1.1.3. Factors governing drug release from a polymer. For
2.1.1.2.3. Multifunctional polymers. These are the bioad- a given drug the release kinetics from the polymer matrix
hesive polymers having multiple functions. In addition to the could be governed predominantly by the polymer morphology
possession of bioadhesive properties, these polymers will also and excipients present in the system. Drug release from a
serve several other functions such as enzyme inhibition, per- polymeric material takes place either by the diffusion or by
meation enhancing effect etc. Examples are polyacrylates, po- polymer degradation or by a combination of the both.
lycarbophil, chitosan etc. Polymer degradation generally takes place by the enzymes
2.1.1.2.4. Thiolated polymers. These are the special class or hydrolysis either in the form of bulk erosion or surface
of multifunctional polymers also called thiomers. These are erosion [58].
hydrophilic macromolecules exhibiting free thiol groups on the 2.1.1.3.1. Polymer morphology. The polymer matrix
polymeric backbone. Due to these functional groups various could be formulated as macro or nanospheres, gel film or an
features of well established polymeric excipients such as poly extruded shape (cylinder, rod etc). Also the shape of the ex-
(acrylic acid) and chitosan were strongly improved [51]. Thi- truded polymer can be important to the drug release kinetics. It
olated polymers designated thiomers are capable of forming has been shown that zero order release kinetics can be achieved
disulphide bonds with cysteine-rich subdomains of mucus gly- using hemispherical polymer form.
coproteins covering mucosal membranes [52]. Consequently, the 2.1.1.3.2. Excipients. The main objective of incorporating
bridging structure most commonly used in biological systems is excipients in the polymer matrix is to modulate polymer degra-
utilized to bind drug delivery systems on the mucosal membranes. dation kinetics. Studies carried out have shown that by in-
By immobilization of thiol groups the mucoadhesive properties of corporating basic salts as excipients slow down the degradation
poly (acrylicacid) and chitosan, was improved to 100-fold to 250- and increases the stability of protein polymers. Similarly hydro-
fold [53,54]. philic excipients can accelerate the release of drugs although
Thiomers are capable of forming intra- and interchain they may also increase the initial burst effect.
disulphide bonds within the polymeric network leading to strongly
improved cohesive properties and stability of drug delivery sys- 2.2. Physiological considerations
tems such as matrix tablets. Due to the formation of strong covalent
bonds with mucus glycoproteins, thiomers show the strongest Physiological considerations such as texture of buccal
mucoadhesive properties of all so far tested polymeric excipients mucosa, thickness of the mucus layer, its turn over time, effect
via thioldisulphide exchange reaction and an oxidation process. of saliva and other environmental factors are to be considered in
Zinc dependent proteases such as aminopeptidases and carbox- designing the dosage forms [59]. Saliva contains moderate
ypeptidases are inhibited by thiomers. The underlying mechanism levels of esterases, carbohydrases, and phosphatases that may
is based on the capability of thiomers to bind zinc ions and this degrade certain drugs. Although saliva secretion facilitates the
property is highly beneficial for oral administration of protein and dissolution of drug, involuntary swallowing of saliva also
peptide drugs. They also exhibit permeation-enhancing effects for affects its bioavailability. Hence development of unidirectional
the paracellular uptake of drugs based on a glutathione-mediated release systems with backing layer results high drug bioavail-
opening process of the tight junctions [55,56]. ability [60].
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 25

2.3. Pharmacological considerations interactions are structural and functional properties. Penetration
enhancement to the buccal membrane is drug specific [65].
Drug absorption depends on the partition coefficient of the Effective penetration enhancers for transdermal or intestinal
drugs. Generally lipophilic drugs absorb through the transcel- drug delivery may not have similar effects on buccal drug
lular route, where as hydrophilic drugs absorb through the delivery because of structural differences; however, enhancers
paracellular route. Chemical modification may increase drug used to improve drug permeation in other absorptive mucosae
penetration through buccal mucosa. Increasing nonionized improve drug penetration through buccal mucosa. These
fraction of ionizable drugs increases drug penetration through permeation enhancers should be safe and non-toxic, pharma-
transcellular route. In weakly basic drugs, the decrease in pH cologically and chemically inert, non-irritant, and non-aller-
increases the ionic fraction of drug but decreases its genic [66]. However, examination of penetration route for
permeability through buccal mucosa [61]. Electrostatic transbuccal delivery is important because it is fundamental to
interactions of drugs such as tetracycline, hydrogen bonding select the proper penetration enhancer to improve the drug
with drugs like urea and hydrophobic interactions with drugs permeability. The different permeation enhancers available are
like testosterone with mucin will decrease rate of absorption [66–68].
[62]. Residence time and local concentration of the drug in
the mucosa, the amount of drug transported across the ➢ Chelators: EDTA, citric acid, sodium salicylate, methoxy
mucosa into the blood are the responsible factors for local or salicylates.
systemic drug delivery. Optimization by a suitable formula- ➢ Surfactants: sodium lauryl sulphate, polyoxyethylene,
tion design hastens drug release from the dosage form and Polyoxyethylene-9-laurylether, Polyoxythylene-20-cety-
taken up by the oral mucosa. Drugs such as buprenorphine, lether, Benzalkonium chloride, 23-lauryl ether, cetylpyr-
testosterone, fentanyl, nifedipine and several peptides such as idinium chloride, cetyltrimethyl ammonium bromide.
insulin, thyrotropin-releasing hormone, and oxytocin have ➢ Bile salts: sodium glycocholate, sodium deoxycholate,
been tried to deliver via the buccal route. However the sodium taurocholate, sodium glycodeoxycholate, sodium
relative bioavailabilities of peptides by the buccal route were taurodeoxycholate.
still low due to its poor permeation and enzymatic barrier of ➢ Fatty acids: oleic acid, capric acid, lauric acid, lauric acid/
buccal mucosa but can be improved by the incorporation of propylene glycol, methyloleate, lysophosphatidylcholine,
penetration enhancers and/or enzyme inhibitors [63]. Previous phosphatidylcholine.
drug absorption studies have demonstrated that oral mucosal ➢ Non-surfactants: unsaturated cyclic ureas.
absoption of amines and acids at constant concentration are ➢ Inclusion complexes: cyclodextrins.
proportional to their partition coefficients. Similar dependen- ➢ Others: aprotinin, azone, cyclodextrin, dextran sulfate,
cies on partition coefficients were obtained from acyclovir, β- menthol, polysorbate 80, sulfoxides and various alkyl
adrenoreceptor blocking agents, substituted acetanilide, and glycosides.
others [18]. ➢ Thiolated polymers: chitosan-4-thiobutylamide, chitosan-
4-thiobutylamide/GSH, chitosan-cysteine, Poly (acrylic
2.4. Permeation enhancers acid)-homocysteine, polycarbophil-cysteine, polycarbo-
phil-cysteine/GSH, chitosan-4-thioethylamide/GSH, chit-
Membrane permeation is the limiting factor for many drugs osan-4-thioglycholic acid.
in the development of buccal adhesive delivery devices. The
epithelium that lines the buccal mucosa is a very effective 2.4.1. Mechanisms of action
barrier to the absorption of drugs. Substances that facilitate the Mechanisms by which penetration enhancers are thought to
permeation through buccal mucosa are referred as permeation improve mucosal absorption are as follows [69,70]
enhancers [64]. As most of the penetration enhancers were
originally designed for purposes other than absorption ➢ Changing mucus rheology: Mucus forms viscoelastic
enhancement, a systemic search for safe and effective layer of varying thickness that affects drug absorption.
penetration enhancers must be a priority in drug delivery. The Further, saliva covering the mucus layers also hinders the
goal of designing penetration enhancers, with improved absorption. Some permeation enhancers' act by reducing
efficacy and reduced toxicity profile is possible by under- the viscosity of the mucus and saliva overcomes this
standing the relationship between enhancer structure and the barrier.
effect induced in the membrane and of course, the mechanism ➢ Increasing the fluidity of lipid bilayer membrane: The
of action. However, the selection of enhancer and its efficacy most accepted mechanism of drug absorption through
depends on the physicochemical properties of the drug, site of buccal mucosa is intracellular route. Some enhancers
administration, nature of the vehicle and other excipients. In disturb the intracellular lipid packing by interaction with
some cases usage of enhancers in combination has shown either lipid packing by interaction with either lipid or
synergistic effect than the individual enhancers. The efficacy of protein components.
enhancer in one site is not same in the other site because of ➢ Acting on the components at tight junctions: Some
differences in cellular morphology, membrane thickness, enhancers act on desmosomes, a major component at
enzymatic activity, lipid composition and potential protein the tight junctions there by increases drug absorption.
26 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

➢ By overcoming the enzymatic barrier: These act by Long-range forces, or van der Waal's forces as they are also
inhibiting the various peptidases and proteases present called, are attractive and account for a wide range of physical
within buccal mucosa, thereby overcoming the enzymatic phenomena, such as friction, surface tension, adhesion and
barrier. In addition, changes in membrane fluidity also cohesion of liquids and solids, viscosity, and the discrepancies
alter the enzymatic activity indirectly. between the actual behavior of gases and that predicted by the
➢ Increasing the thermodynamic activity of drugs: Some ideal gas law [80].
enhancers increase the solubility of drug there by alters Many theories have been proposed to explain the forces that
the partition coefficient. This leads to increased thermo- underpin bioadhesion. They are
dynamic activity resulting better absorption.
❖ Electronic theory
Surfactants such as anionic, cationic, nonionic and bile ❖ Adsorption theory
salts increases permeability of drugs by perturbation of ❖ Wetting theory
intercellular lipids whereas chelators act by interfering with ❖ Diffusion theory
the calcium ions, fatty acids by increasing fluidity of ❖ Fracture theory, etc. [81].
phospholipids and positively charged polymers by ionic
interaction with negative charge on the mucosal surface However, there is yet to be a clear explanation. As
[71–76]. Chitosan exhibits several favorable properties such bioadhesion occurs between inherently different mucosal
as biodegradability, biocompatibility and antifungal/antimicro- surfaces and formulations that are solid, semisolid and liquid,
bial properties in addition to its potential bioadhesion and it is unlikely that a single, universal theory will account for all
absorption enhancer [77,78]. types of adhesion observed. In biological systems it must be
recognized that, owing to the amphiphilicity of many biological
3. Muco/bioadhesion macromolecules, orientation effects can often occur at inter-
faces. These are crucially important and have in fact been
Bioadhesion is the phenomenon between two materials, reported to be so dramatic as to change overall long-range
which are held together for extended periods of time by interactions from being purely repulsive to their becoming
interfacial forces [79]. It is generally referred as bioadhesion attractive [82]. For any type of charged surface, such as
when interaction occurs between polymer and epithelial surface; biosurfaces, it is common to distinguish between pure
mucoadhesion when occurs with the mucus layer covering a electrostatic repulsive forces, which oppose adhesion, and
tissue. Generally bioadhesion is deeper than the mucoadhesion. attractive forces, which, if the surfaces come close enough,
However, these two terms seem to be used interchangeably. It is will strive to bring the interacting bodies together. This balanced
interesting that the interaction between the layers adsorbed from relationship between repulsive and attractive interactions is
whole saliva resembles the one previously reported between expressed in the DLVO theory [83]. In biological systems,
layers of adsorbed gastric mucins, which points to a strong interactions can be more complex, as they often take place in
contribution to the interaction of high molecular weight high ionic strength aqueous media and in the presence of
glycoproteins. macromolecules. Therefore electrostatic contributions may be
less important, at least at long range, in favor of force
3.1. Bio/mucoadhesive forces components such as steric forces, hydrophobic interactions,
and hydration forces.
The common nature of all adhesive events, interfacial
phenomena and forces that are involved in bioadhesion are 3.1.1. Van der Waal's forces
strongly related to those considered in classical colloid and The attractive forces included in the DLVO theory are nor-
surface science. Intermolecular forces are electromagnetic mally termed van der Waal's forces and will arise in a number of
forces which act between molecules or between widely ways. These may be further divided into the following three
separated regions of a macromolecule. These are fundamen- components [84,85]:
tally electrostatic interactions or electrodynamic interactions.
Such forces may be either attractive or repulsive in nature. (i) London dispersion forces: These are also called as dis-
They are conveniently divided into two classes: short-range persion forces. These originate out of the electronic
forces, which operate when the centers of the molecules are motions in paired molecules and give rise to attractive
separated by 3 angstroms or less and long-range forces, which interactions. These forces involve the attraction between
operate at greater distances. Generally, if molecules do not tend temporarily induced dipoles in nonpolar molecules (often
to interact chemically, the short-range forces between them are disappear within a second) [86]. This polarization can be
repulsive. These forces arise from interactions of the electrons induced either by a polar molecule or by the repulsion of
associated with the molecules and are also known as exchange negatively charged electron clouds in nonpolar molecules.
forces. Molecules that interact chemically have attractive These results when two atoms belonging to different
exchange forces; these are also known as valence forces. molecules are brought sufficiently close together. These
Mechanical rigidity of molecules and effects such as limited interactions involve a force of about 0.5–1 K cal/mole.
compressibility of matter arise from repulsive exchange forces. London Dispersion forces exist between all atoms [87].
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 27

(ii) Dipole–dipole interactions: These are also called Keesom to polar surface sites, has been observed between phospholipids
interactions after Willem Hendrik Keesom who produced [97] and solid surfaces under certain conditions [98]. This
the first mathematical description in 1921, are the forces hydration force is believed to be particularly important in
that occur between two molecules with permanent biological systems, since it prevents contact even in the absence
dipoles. These work in a similar manner to ionic inter- of charge–charge repulsion.
actions, but are weaker because only partial charges are
involved. These are due to attraction between polar 3.1.5. Electrostatic double-layer forces
groups. These have force of 1–7 K cal/mole. Dipole– A charged surface is always surrounded by a cloud of counter
dipole interactions also come from partial charges another ions (double-layer), which balances the surface charge. When
order of magnitude weaker [88]. two surfaces with the same charge approach each other, a
(iii) Debye type forces: These are the interactions between repulsive force will arise due to the overlap of the double layers.
permanent and induced dipoles. Permanent dipoles can This is the origin of the electrostatic double-layer forces, which
induce a transient electric dipole in non-polar molecules can be described by the so-called Poisson–Bolzmann equation.
and produce dipole induced dipole interactions. These These forces decay exponentially with the surface separation,
interactions involve a force of about 1–3 K cal/mole [86]. with a decay length that decreases with increasing ionic
strength in the surrounding medium. It should be noted that
The non-retarded van der Waal's force is inversely pro- specific dispersion force-induced ion adsorption could some-
portional to the square of the distance between two spherical times dominate at charged interfaces, thereby making it
particles, where the proportionality constant is the Hamaker virtually impossible to distinguish between the contributions
constant, which has the dimension of energy, can be used to of electrostatic and dispersion forces [89]. In biological fluids,
describe the strength of the van der Waal's interaction and is which generally carry a large net negative charge, contribute
dependent on the properties of the involved particles and on the significantly to the decay length already at low concentrations
medium where the interaction takes place [89]. [90]. Thus, the decay length in saliva is likely to be less than
the value of approximately 1.0 nm calculated from its salt
3.1.2. Hydrogen bonding composition. Any increase in ionic strength, increases
Hydrogen bonding is basically an electrostatic interaction adhesion to negatively charged surfaces; this was assigned to
that arises when a hydrogen atom bound to an electronegative less repulsion between the surface and the adhering cells [91].
atom, e.g., nitrogen, oxygen, or fluorine, interacts with another
electronegative atom [90]. The result is a dipolar molecule. The 3.1.6. Hydrophobic interactions
hydrogen atom has a partial positive charge and hence can Hydrophobic effect is another particularly important phenom-
interact with another highly electronegative atom in an adjacent enon with respect to bioadhesion related to the presence of water.
molecule. This results in a stabilizing interaction that binds the It is the property that nonpolar molecules like to self-associate in
two molecules together. The force is short range and highly the presence of aqueous solution. It has been assigned to the
directional. In a more hydrophobic environment, hydrogen tendency of water molecules to form ordered structures in
bonds become significant and are essential in the formation of proximity to non-polar molecular domains and may give rise to
stable structures. Bond energy serves as a measure of strength of attractive interactions between non-polar residues such as
bonds. Magnitude of bond energy for hydrogen bond is between hydrocarbon side chains. The hydrophobic effect is usually
10 and 20 kJ/mol [91]. Role of hydrogen bonding in interaction described in the context of protein folding, protein–protein
between mucoadhesive and mucin at gastric pH was studied by interactions, nucleic acid structure, and protein–small molecule
Tobyn et al. [92]. The bonding is stronger and is directional. The interactions. In the case of protein folding, it is used to explain
directional nature of hydrogen bonding requires the two why many proteins have a hydrophobic core which consists of
molecules to adopt a specific relative geometry [93]. hydrophobic amino acids, such as alanine, valine, leucine,
isoleucine, phenylalanine, and methionine grouped together;
3.1.3. Disulphide bridging often coiled-coil structures form around a central hydrophobic
A disulfide bond (SS-bond), also called a disulfide bridge, is axis. The energetics of DNA tertiary structure assembly were
a strong covalent bond between two sulfhydryl (–SH) groups. determined by Eric Kool to be mostly caused by the hydrophobic
Oxidation of the thiol group yields a disulfide (S–S) bond [94]. effect, as opposed to Watson–Crick base pairing [99].
This bond is very important to the folding, structure, and The hydrophobic effect can be nullified to a certain extent by
function of proteins [95]. Due to the formation of strong lowering the temperature of the solution to near zero degrees; at
covalent bonds with mucus glycoproteins, thiomers show the such temperatures, water prefers to be in an ordered structure
strongest mucoadhesive properties of all so far tested polymeric and the order generated by hydrophobic patches is no longer as
excipients via thioldisulphide exchange reaction and an energetically unfavorable. This is neatly demonstrated by the
oxidation process [96] as shown in Fig. 2. increased solubility of benzene in water at temperatures lower
than room temperature. On the macroscopic level, long-range
3.1.4. Hydration forces attractive forces have been observed between hydrophobic
A type of short-range (< 1 nm) repulsive interaction, surfaces formed by adsorption or deposition of amphiphilic
suggested as originating from the binding of water molecules molecules and are believed to be non-equilibrium forces
28 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

Fig. 2. Formation of covalent bonds between thiolated polymers and mucin glycoproteins.

[93,97]. It should be noted that the origin of the long-range 3.2. Methods for measuring mucoadhesion
attractive forces between hydrophobic surfaces is controversial,
but their occurrence has been related to instability of the These tests are important during the design and development
deposited monolayer [100]. Strength of these interactions is of a mucoadhesive release system to study compatibility, sta-
about 0.37 kcal/mol [86]. bility, surface analysis and bioadhesive bond strength. These
tests are broadly classified in to qualitative methods and quan-
3.1.7. Steric forces titative methods.
Repulsive steric interactions or steric forces appear as the
result of the increasing concentration of molecular segments 3.2.1. Quantitative methods
that occurs when surfaces bearing for example bound macro- These are also called macroscopic methods. The majority of the
molecules come close to each other and therefore considered to quantitative bio and/or mucoadhesion measurement methods
be important in biological systems. The maximum possible found in the literature are based on measuring the force required
number of molecular contacts between an adhesive and its to break the adhesive bond between the model membrane and the
substrate may be greatly restricted by the steric aspects of adhesive. Depending on the direction in which the adhesive is
molecular geometry [80]. being separated from the substrate, peel, shear, and tensile forces
can be measured.
3.1.8. Covalent bonds
Like metallic bonds, covalent bonds are characterized by the 3.2.1.1. Determination of peel strength. The peel adhesion
electrons that are shared between the engaged atoms. Covalent tests are mainly used for buccal and transdermal patches [102].
bonds operate only over short interatomic distances (1– The test is based on the calculation of energy required to detach
2 × 10− 1 nm). They tend to decrease in strength with increasing the dosage form from the substrate material (usually excised
bond-length, and are oriented at well-defined angles. Unless buccal mucosa) attached through the bioadhesive material in the
chemical reactions take place, based on the formation or direction as shown in Fig. 3.
breakup of for example disulphide bridges, covalent bonds are Fracture Energy (G)
unlikely to be important in bioadhesion processes under
physiological conditions. Pð1−Cos hÞ
G¼ ¼ W -ð1 þ kÞ
On the basis of molecular interactions, the interaction between w
two molecules is composed of attraction and repulsion. Attractive
interactions arise from weak forces such as van der Waal's forces, Where P is the peel force;
electrostatic attraction, hydrogen bonding, hydrophobic interac- w is the peel width;
tions and/or strong forces, which are covalent in nature. Repulsive W° is the intrinsic work of adhesion and k is the proportionality
interactions occur because of electrostatic and steric repulsion. constant that accounts for hysteretic losses.
For muco/bioadhesion to occur, the attractive interactions should Peel work is the sum of the following components
be larger than nonspecific repulsion [101].
Steps involved in the process of bio/mucoadhesion are ➢ Surface energy that results from the creation of two free
surfaces (energy of dewetting) also referred to as the
(i) Spreading, wetting, swelling and dissolution of bio/ intrinsic work of adhesion (or cohesion)
mucoadhesive polymer at the interface, initiates intimate ➢ Bulk energy that dissipates into the stripping member
molecular contact at the interface between the polymer ➢ Strain energy in the newly detached strip
and the epithelial/mucus layer.
(ii) Interdiffusion and interpenetration between the chains of Intrinsic work of adhesion (or cohesion) is independent of
the adhesive polymer and the mucus/epithelial surface the following:
resulting physical cross links or mechanical interlocking.
(iii) Adsorption: The orientation of the polymers at the interface ◦ Peel rate (speed)
so that adhesive bonding across the interface is possible. ◦ Peel angle
(iv) Formation of secondary chemical bonds between the ◦ Thickness of the adhesive
polymer chains and mucin molecules. ◦ Thickness of the stripping member
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 29

of a material to a force tending to tear it apart, measured as the


maximum tension the material can withstand without tearing.
Tensile strength can be defined as the strength of material
expressed as the greatest longitudinal stress it can bear without
tearing apart. As it is the maximum load applied in breaking a
tensile test piece divided by the original cross-sectional area of the
test piece, it is measured as Newtons/sq.m. Specifically, the tensile
strength of a material is the maximum amount of tensile stress that
it can be subjected to before failure. The definition of failure can
vary according to material type and design methodology.
There are three typical definitions of tensile strength:
Fig. 3. Representation of peel, shear and tensile forces.
▪ Yield Strength — The stress a material can withstand
Values of intrinsic work of adhesion vary from 0.07 J/m squared without permanent deformation.
for hydrocarbon van der Waal's interactions, 2 J/m squared for a ▪ Ultimate Strength — The maximum stress a material can
system with covalent bonding as part of the adhesion. The work of withstand.
fracture can be several orders of magnitude greater than the ▪ Breaking Strength — The stress coordinate on the stress–
intrinsic work of adhesion [4]. strain curve at the point of rupture.

3.2.1.2. Determination of shear strength. Shear stress, τ is Methods using the tensile strength usually measure the force
the force acting tangentially to a surface divided by the area of required to break the adhesive bond between a model membrane
the surface. It is the force per unit area required to sustain a and the test polymers.
constant rate of fluid movement. Mathematically, shear stress Lehr et al. [103] determined tensile strength of flat-faced
can be defined as: buccal adhesive tablets, with a diameter of 5.5 mm containing
s ¼ F=A 50 mg of the mucoadhesive material is to be tested for its shear
stresses by clamping the model mucosal surface between two
where, plates, one having a U-shaped section cut away to expose the test
surface. The tablet was attached to a Perspex disc, and then placed
τ shear stress into contact with the exposed mucosa at the base of the U shaped
F force cut. 1.5 g weight was used to consolidate the adhesive joint for
A area of the surface subjected to the force. 2 min, and the plates were oriented from horizontal to vertical and
Perspex disc attached to the underside of the balance, which was
If a fluid is placed between two parallel plates spaced 1.0 cm linked to a microcomputer for data collection. A shear stress was
apart, and a force of 1.0 dyn is applied to each square centimeter of applied by lowering the plates and model mucosa at a rate of 2 mm
the surface of the upper plate to keep it in motion, the shear stress min− 1 until adhesive joint failure occurred (Fig. 4).
in the fluid is 1 dyn/cm2 at any point between the two plates [4]. Many researchers studied shear strength of polymers such as
Shear stress measures the force that requires causing the polyacrylic acid, hydroxy propylcellulose, carbapol 934,
bioadhesive to slide with respect to the mucus layer in a direction HPMC etc. using buccal mucosa as substrate by using different
parallel to their plane of contact as shown in Fig. 3. instruments such as tensile tester, modified pan balance etc.
Sam et al. studied the mucoadhesiveness of Ca polycarbophil,
sodium CMC, HPMC using homogenized mucus from pig in- 3.2.1.4. Colloidal gold staining method. Park [107] proposed
testine as model substrate by modified wilhelmy plate surface the colloidal gold staining technique for the study of bioadhesion.
tension apparatus [103]. Similarly, Smart et al. studied mucoad- The technique employs red colloidal gold particles, which were
hesive strength of CP 934, Na CMC, HPMC, gelatin, PVP, acacia, adsorbed on mucin molecules to form mucin–gold conjugates,
PEG, pectin, tragacanth and sodium alginate was measured by the which upon interaction with bioadhesive hydrogels develops
force required to pull the plate out of the solution is determined a red color on the surface. This can be quantified by measuring
under constant experimental conditions by using mucus from at 525 nm either the intensity on the hydrogel surface or the
guinea pig intestine as model substrate by Wilhelmy plate method, conjugates.
where a glass plate suspended from a microbalance, which was
dipped in a temperature-controlled mucus sample [104]. Instead 3.2.1.5. Direct staining method. It is a novel technique to
of biological substrates, Ishida et al. [105] used glass plates as evaluate polymer adhesion to human buccal cells following
model substrate by shearing stickiness apparatus and Gurney et al. exposure to aqueous polymer dispersion, both in vitro and in
[106] used polymethylmethacrylate to study shear stress of vivo. Adhering polymer was visualized by staining with 0.1% w/
carbapol and sodium CMC by Instron model 1114, respectively. v of either Alcian blue or Eosin solution; and the uncomplexed
dye was removed by washing with 0.25 M sucrose. The extent of
3.2.1.3. Determination of tensile strength. Tensile stress is also polymer adhesion was quantified by measuring the relative
termed Maximum Stress or Ultimate Tensile Stress. The resistance staining intensity of control and polymer treated cells by image
30 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

Fig. 4. Schematic representation of apparatus for measuring tensile strength.

analysis. Carbopol 974 P, polycarbophil and chitosan were found chitosan–mucin interaction, assessed by means of viscosimetric
to adhere to human buccal cells from 0.10% w/w aqueous dis- measurements. Two hydration media, distilled water and 0.1 M
persions of these polymers. Following in vivo administration as a Hcl were used. Chitosan solutions were prepared at concentra-
mouthwash, these polymers persisted upon the human buccal tions greater than the characteristic entanglement concentration
mucosa for at least one hour. This method is only suitable for and mixed with increasing amounts of porcine gastric mucin.
assessing the liquid dosage forms, which are widely employed to Viscosity measurements were performed on the polymer–mucin
enhance oral hygiene and to treat local disease conditions of the mixtures and on polymer and mucin solutions having the same
mouth such as oral candidacies and dental caries [108]. concentrations as in the mixtures. The formation of chitosan–
mucin interaction products was determined on the basis of the
3.2.2. Qualitative methods changes in low shear viscosity and high shear viscosity of the
These methods are useful for preliminary screening of the mixtures as a function of polymer: mucin weight ratio. Rheo-
respective polymer for its bio or mucoadhesion, compatibility logical synergism parameter was also calculated. The results
and stability. However, these methods are not useful in mea- obtained suggest that two different types of rheological interaction
suring the actual bioadhesive strength of the polymers. They are occur between chitosan and mucin in both media, depending on
polymer concentration and polymer: mucin weight ratio.
3.2.2.1. Viscometric method. Katarina Edsman [109] has
studied the dynamic rheological measurements on gels containing 3.2.2.2. Analytical ultracentrifuge criteria for mucoadhesion.
four different carbopol polymers and the corresponding mixtures These methods are useful in identifying the material that is able to
with porcine gastric mucin and bovine submaxillary mucin. The form complexes with the mucin. The assay can be done for change
method does not give the same ranking order when two different in molecular mass using sedimentation equilibrium, but this has an
comparison strategies were used. The results were contrast to the upper limit of less than 50 MDa. Since complexes can be very large,
results obtained with the tensile strength measurements. a more sensible assay procedure is to use sedimentation velocity
Hassan [110] developed a simple viscometric method to with change in sedimentation coefficient, s, as their marker for
quantify mucin–polymer bioadhesive bond strength. Viscosities mucoadhesion. Where mucin is available in only miniscule
of 15% w/w porcine gastric mucin dispersion were measured amounts, a special procedure known as Sedimentation Fingerprint-
with Brookfield's viscometer. In absence or presence of selected ing can be used for assay of the effect on the mucoadhesive. UV
neutral, anionic and cationic polymer, viscosity components and absorption optics is used as the optical detection system. However,
the forces of bioadhesion were calculated. He observed a in this case the mucoadhesive is invisible, but the pig gastric mucin
positive rheological synergism when chitosan solutions pre- at the concentrations normally employed is visible. The sedimen-
pared in pH 5.5 acetate buffers and in 0.1 M Hcl, were mixed tation ratio (scomplex/smucin), the ratio of the sedimentation coef-
with a fixed amount of porcine gastric mucin. The mixtures with ficient of any complex involving the mucin to that of pure mucin
mucin showed a viscosity greater than the sum of polymer and itself, is used as the measure for mucoadhesion [113].
mucin viscosities.
Mortazavi and Smart [111] investigated the effect of carbopol 3.2.2.3. Atomic force microscopy. This method is based on the
934 P on rheological behavior of mucus gel and role of mucus and changes in surface topography when the polymer bound on to
effect of various factors such as ionic concentration, polymer mo- buccal cell surfaces. Unbound cells shows relatively smooth
lecular weight, its concentration and the introduction of anionic, surface characteristics with many small craters like pits and
cationic and neutral polymers on mucoadhesive mucus interface. indentations spread over cell surfaces, while polymer bound
Carla Caramella et al. [112] investigated the influence of cells will loose crater and indentation characteristics and gained
polymer concentration and polymer: mucin weight ratio on a higher surface roughness.
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 31

3.2.2.4. Electrical conductance. Bremakar used modified Swelling depends both on polymer concentration and the
rotational viscometer to determine electrical conductance of various presence of water. When swelling is too great, a decrease in
semi-solid mucoadhesive ointments and found that the electrical bioadhesion occurs [116].
conductance was low in the presence of adhesive material. pH was found to have significant effect on mucoadhesion as
observed in studies of polyacrylic polymers cross-linked with
3.2.2.5. Fluorescent probe method. In this method the carboxyl groups. pH influences the charge on the surface of both
membrane lipid bilayered and membrane proteins were labeled mucus and the polymers. Mucus will have a different charge
with pyrene and fluorescein isothiocyanate, respectively. The depending on pH because of differences in dissociation of
cells were mixed with the mucoadhesive agents and changes in functional groups on the carbohydrate moiety and amino acids of
fluorescence spectra were monitored. This gave a direct indication the polypeptide backbone. It was observed that the pH of the
of polymer binding and its influence on polymer adhesion [114]. medium was critical for the degree of hydration of highly cross
linked polyacrylic acid polymers, increasing between pH 4 and 5,
3.2.2.6. Lectin binding inhibition technique. The method continuing to increase slightly at pH 6 and 7 and decreasing at
involves an avidin–biotin complex and a colorimetric detection more alkaline pH levels [118]. To place a solid bioadhesive
system to investigate the binding of bioadhesive polymers to system, it is necessary to apply a defined strength. Whatever the
buccal epithelial cells without having to alter their physico- polymer may be, the adhesion strength increases with the applied
chemical properties by the addition of marker entities. The strength and duration of its application, up to an optimum [119].
lectin cancanavalian A has been shown to specifically bind to The initial contact time between mucoadhesive and the mucus
sugar groups present on the surface of buccal cells. If polymers layer determine the extent of swelling and the interpenetration of
bind to buccal cells, they will mask the surface glycoconjugates, polymer chains. Along with the initial pressure, the initial contact
thus reducing or inhibiting cancanavalian A binding [115]. time can dramatically affect the performance of a system. The
mucoadhesive strength increases as the initial contact time
3.2.2.7. Thumb test. This is a very simple test used for the increases [101]. Dehydration of the mucosa, causes by water
qualitative determination of peel adhesive strength of the movement from the mucosa to the dry powder, may have resulted
polymer and is useful tool in the development of buccal in adhesion between the two surfaces [120]. A low interfacial
adhesive delivery systems. The adhesiveness is measured by the tension value for the bioadhesive tissue increases the possibility
difficulty of pulling the thumb from the adhesive as a function of obtaining adhesive bonds [121]. Addition of highly water-
of the pressure and the contact time. Although the thumb test soluble additive reduces the water content when the material
may not be conclusive, it provides useful information on peel dissolves, and thus makes the water unavailable for the
strength of the polymer. bioadhesive material, and subsequently decreases bioadhesion
[122]. The duration of adhesion depends on the amount of water
3.3. Factors affecting bio/mucoadhesion at the interface. Excessive water reduces the duration of adhesion.
However the magnitude of this change is not the same for all the
Numerous studies have indicated that there is a certain materials. It is believed that faster the rate of absorption of water,
molecular weight at which bioadhesion is optimum. The optimum the shorter is the time required for the material to obtain initial
molecular weight for the maximum bioadhesion depends on the adhesion and maximum adhesive strength. But rapid water
type of polymers. It dictates the degree of swelling in water, which absorbency may cause the shortening of the duration of adhesion
in turn determines interpenetration of polymer molecules within [123]. Previous drug absorption studies have demonstrated that
the mucus. It seems that the bioadhesive force increases with the buccal absorption through oral mucosa for drugs such as
molecular weight up to 100,000 and beyond this level there is not morphine sulphate, nicotine, flecainide, sotalol, propanolol and
much effect [106]. For the best bioadhesion to occur, the con- others changed with changing pH [18].
centration of polymer must be at optimum. Flexibility of polymer
chain is also important for interpenetration and entanglement 4. Developments in buccal adhesive drug delivery
[116,117]. As water-soluble polymers become cross-linked, the
mobility of the individual polymer chain decreases. As the cross Retentive buccal mucoadhesive formulations may prove to be
linking density increases, the effective length of the chain, which an alternative to the conventional oral medications as they can be
can penetrate into the mucus layer, decreases even further and readily attached to the buccal cavity retained for a longer period of
mucoadhesive strength is reduced. Besides molecular weight or time and removed at any time. Buccal adhesive drug delivery
chain length, spatial conformation of a molecule is also important. systems using matrix tablets, films, layered systems, discs, micro
Despite a high molecular weight of 19,500,000 for dextrans, they spheres, ointments and hydrogel systems has been studied and
have similar adhesive strength to that of polyethylene glycol with reported by several research groups. However, limited studies exist
a molecular weight of 200,000. The helical conformation of on novel devices that are superior to those of conventional buccal
dextran may shield many adhesively active groups, primarily adhesive systems for the delivery of therapeutic agents through
responsible for adhesion, unlike PEG polymers, which have a buccal mucosa. A number of formulation and processing factors
linear conformation. Swelling is not only related to the polymer can influence properties and release properties of the buccal ad-
itself, and also to its environment. Interpenetration of chains is hesive system. There are numerous important considerations that
easier as polymer chains are disentangled and free of interactions. include biocompatibility (both the drug/device and device/
32 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

environment interfaces), reliability, durability; environmental sta- Table 2


bility, accuracy, delivery scalability and permeability are to be Commercial name Bioadhesive Company Dosage
considered while developing such formulations. While biocom- polymer form
patibility is always an important consideration, other considerations Buccastem PVP, Xanthum gum, Rickitt Tablet
vary in importance depending on the device application. Bioadhe- Locust bean gum Benckiser
sive formulations designed for buccal application should exhibit Suscard HPMC Forest Tablet
Gaviscon liquid Sodium alginate Rickitt Oral liquid
suitable rheological and mechanical properties, including pseudo-
Benckiser
plastic or plastic flow with thixotrophy, ease of application, good Orabase Pectin, gelatin ConvaTech Oral paste
spreadability, appropriate hardness, and prolonged residence time Corcodyl gel HPMC Glaxosmithkline Oromucosal
in the oral cavity. These properties may affect the ultimate perfor- gel
mance of the preparations and their acceptance by patients [124]. Corlan pellets Acacia Celltech Oromucosal
pellets
An ideal buccal adhesive system must have the following
Fentanyl Oralet™ Lexicomp Lozenge
properties: Miconaczole Bioalliance Tablet
Lauriad
✓ Should adhere to the site of attachment for a few hours, EmezineTM BDSI's
✓ Should release the drug in a controlled fashion, BEMA Fentanyl BDSI's
Straint™ SR Ardana
✓ Should provide drug release in an unidirectional way
Zilactin Zila Buccal film
toward the mucosa, Luborant Sodium CMC Antigen Artificial
✓ Should facilitate the rate and extent of drug absorption, saliva
✓ Should not cause any irritation or inconvenience to the Saliveze Sodium CMC Wyvern Artificial
patient and saliva
Tibozole Tibotec Tablet
✓ Should not interfere with the normal functions such as
talking, drinking etc.

4.1. Commercial buccal adhesive drug delivery systems 4.2.1.2. Microparticles. Bioadhesive microparticles offer the
same advantages as tablets but their physical properties enable
Recent reports suggest that the market share of buccal them to make intimate contact with a lager mucosal surface
adhesive drug delivery systems are increasing in the American area. In addition, they can also be delivered to less accessible
and European market with the steady growth rate of above 10%. sites including the GI tract and upper nasal cavity. The small
Some of the commercially available buccal adhesive formula- size of microparticles compared with tablets means that they are
tions are listed in Table 2. less likely to cause local irritation at the site of adhesion and the
uncomfortable sensation of a foreign object within the oral
4.2. Research on buccal adhesive drug delivery systems cavity is reduced.

Several buccal adhesive delivery devices were developed at 4.2.1.3. Wafers. Bromberg et al. [160] described a conceptually
the laboratory scale by many researchers either for local or novel periodontal drug delivery system that is intended for the
systemic actions. They are broadly classified in to treatment of microbial infections associated with peridontitis. The
delivery system is a composite wafer with surface layers posses-
❖ Solid buccal adhesive dosage forms sing adhesive properties, while the bulk layer consists of anti-
❖ Semi-solid buccal adhesive dosage forms microbial agents, biodegradable polymers and matrix polymers.
❖ Liquid buccal adhesive dosage forms
4.2.1.4. Lozenges. Bioadhesive lozenges may be used for the
4.2.1. Solid buccal adhesive formulations delivery of drugs that act topically within the mouth including
Dry formulations achieve bioadhesion via dehydration of the antimicrobials, corticosteroids, local anaesthetics, antibiotics
local mucosal surface. and antifungals. Conventional lozenges produce a high initial
release of drug in the oral cavity, which rapidly declines to
4.2.1.1. Tablets. Several bioadhesive tablet formulations were subtherapeutic levels, thus multiple daily dosing is required. A
developed in recent years either for local or systemic drug slow release bioadhesive lozenge offers the potential for pro-
delivery. Tablets that are placed directly onto the mucosal longed drug release with improved patient compliance. Codd
surface have been demonstrated to be excellent bioadhesive and Deasy investigated bioadhesive lozenges as a means to
formulations. However, size is a limitation for tablets due to the deliver antifungal agents to the oral cavity [161].
requirement for the dosage form to have intimate contact with
the mucosal surface. These tablets adhere to the buccal mucosa 4.2.2. Semi-solid dosage forms
in presence of saliva. They are designed to release the drug
either unidirectionally targeting buccal mucosa or mutidirec- 4.2.2.1. Gels. Gel forming bioadhesive polymers include cross-
tionally in to the saliva. Table 3. represents some of the research linked polyacrylic acid that has been used to adhere to mucosal
done so far in the development of buccal adhesive tablets. surfaces for extended periods of time and provide controlled release
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 33

Table 3 release of drug at the absorption site. A limitation of gel formu-


Buccal adhesive tablets lations lies on their inability to deliver a measured dose of drug to
Drug Bioadhesive polymer Excipients References the site. They are therefore of limited use for drugs with narrow
Ketoprofen Chitosan and sodium [125] therapeutic window. He et al. [162] designed a novel, hydrogel-
alginate based, bioadhesive, intelligent response system for controlled drug
Nifedipine Chitosan, polycarbophil, [126] release. This system combined several desirable facets into a single
sodium alginate, gellan gum
Propranolol CP, HPMC, PC, SCMC, [127]
formulation; a poly (hydroxyethyl methacrylate) layer as barrier,
PAA poly (methacrylic acid-g-ethylene glycol) as a biosensor and poly
Propranolol HPMC, CP 934 [128] (ethyleneoxide) to promote mucoadhesion.
Propranolol HPMC, PC [129]
Diltiazem CP, HPMC, PC, SCMC, [130] 4.2.2.2. Patches/films. Flexible films may be used to deliver
PAA
drugs directly to a mucosal membrane. They also offer advantages
Diltiazem CP 934 and PVP K-30 Citric acid and [81]
PEG 4000 over creams and ointments in that they provide a measured dose of
Metaclopromide CP, HPMC, PC, SCMC, [127] drug to the site. Buccal adhesive films are already in use com-
PAA mercially for example, Zilactin used for the therapy of canker
Nystatin Carbomer, HPMC Lactose [131] sores, cold sores and lip sores. These were represented in Table 4.
Verapamil HPC-M, CP 934 [132,133]
Triamcilone HPC, CP-934 [134]
Triamcilone HPMC, PADH [135] 4.2.3. Liquid dosage forms
Lidocaine CP-934, HPC-H [136] Viscous liquids may be used to coat buccal surface either as
Metronidazole CP-934, HPMC [137] protectants or as drug vehicles for delivery to the mucosal surface.
Sodium fluoride Modified starch, PAA [138] Traditionally, pharmaceutically acceptable polymers were used to
Miconazole Modified starch, CP-934 [139]
enhance the viscosity of products to aid their retention in the oral
Pentazocine CP-934P, HPMC [140]
Chlorpheneramine Hakea gum [141] cavity. Dry mouth is treated with artificial saliva solutions that are
Calcitonin Hakea gum [141] retained on mucosal surfaces to provide lubrication. These
Omeprazole Sodium alginate, Magnesium [142] solutions contain sodium CMC as bioadhesive polymer.
HPMC, CP-934P, PC oxide
Nicotine HPC, CP-934P, PVP [143]
4.3. Delivery of proteins and peptides
Clotrimazole CP 974P, HPMC K4M [144]
Nicotine hydrogen Aionic, cationic and pH increasing [145]
tartrate nonionic additive The buccal mucosa represents a potentially important site for
Citrus oil and Cross linked PAA and [146] controlled delivery of macromolecular therapeutic agents, such
magnesium salt HPC as peptides and protein drugs with some unique advantages such
Buspirone Hcl CP 974 HPMCK4M [147]
Omeprazole Sodium alginate and MgO and [148]
as the avoidance of hepatic first-pass metabolism, acidity and
HPMC croscaramellose protease activity encountered in the gastrointestinal tract.
sodium
Hydrocortisone HPMC (methocelk4m), [149] Table 4
acetate carbapol934P, Buccal adhesive patches/films
polycarbiphyl
Drug Bioadhesive polymer Excipients Reference
Ergotamine PVA Witepsol W-35 [150]
tartrate and cod liver oil Plasmid DNA Noveon, eudragit S-100 [163]
extract B-galactosidase Noveon, eudragit S-100 [163]
Hydralazine Hcl CP 934P and CMC [151] Ipriflavone PLGA, chitosan [164]
Prednisolone Polycarbophil and CP [152] Chlorhexidine Chitosan [165]
934P gluconate
Buprenorphine HEMA and Polymeg [153] Chlorpheneramine Polyoxyethylene [166]
Morphine Carbomer and HPMC [154] maleate
sulphate Protirelin HEC, HPC, PVP, PVA [167,168]
Piribedit [155] Buprenorphine CP-934, PIB and PIP [169]
Nimesulide [156] Isosorbide HPC, HPMCP Glycerrhizinic [170]
Cetylpyridinium [157] dinitrate acid
chloride Lidocaine HPC, CP [171]
Thiocolchicoside [158] Miconazole nitrate SCMC, chitosan, PVP [172]
Propranolol CP-934P, HPMC K4M [159] PVA, HEC and HPMC
Nifedipine Sodium alginate Mannitol, PEG 6000 [173]
Abbrevations: CP: carbapol, HPMC: hydroxypropylmethylcellulose, PC:
Acyclovir [P (AA-co-PEG)] Sodium glycocholate [174]
polycarbophil, SCMC: sodium carboxymethylcellulose, PAA: polyacrylic
acid, HPC: hydroxypropyl cellulose, PVP: poly (vinylpyrrolidone), PADH: Abbrevations: CP: carbapol, HPMC: hydroxypropylmethylcellulose, PC:
poly (acrylicacid-2-5-dimethyl 1-5 hexadiene). polycarbophil, SCMC: sodium carboxymethylcellulose, PVP: poly (vinylpyrro-
lidone), PLGA: poly (D,L-lactide co-glycolide), HPMCP: hydroxypropylmethyl
cellulosephthalate, PIB: polyisobutylene, PIP: polyisoprene, [P (AA-co-PEG)]:
of drugs. Gels have been widely used in the delivery of drugs to the copolymers of polyacrylic acid and polyethylene glycol monomethylether
oral cavity. Advantages of gel formulations include their ability to monomethacrylate, HPC: hydroxypropylcellulose, HEC: hydroxyethylcellulose,
form intimate contact with the mucosal membrane and their rapid PVA: polyvinylalcohol.
34 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

Another interesting advantage is its tolerance (in comparison optimized properties were selected for the in vivo evaluation. The
with the nasal mucosa and skin) to potential sensitizers. A variety bioadhesive tablet was placed on the buccal mucosa between the
of proteins/peptides with or without penetration enhancer were cheek and gingiva in the region of the upper canine and gently
studied by different scientists using different animal models like pressed onto the mucosa for about 30 s. The tablet and the inner
dogs, rabbits, rats, pigs and humans. Some of those develop- upper lip were carefully moistened with saliva to prevent the
ments were represented in Table 5. sticking of the tablet to the lip. The volunteers were asked to
monitor the ease with which the system was retained on the mucosa
5. Evaluation and note any tendency for detachment. The time necessary for
complete erosion of the tablet was simultaneously monitored by
In addition to the routine evaluation tests such as weight carefully observing for residual polymer on the mucosa. In ad-
variation, friability, hardness, content uniformity, in vitro dis- dition, any complaints such as discomfort, bad taste, dry mouth or
solution for tablets; tensile strength, film endurance, hygroscop- increase of salivary flux, difficulty in speaking, irritation or mucosal
icity etc for films and patches; viscosity, effect of aging etc for gels lesions were carefully recorded. Repeated application of the bio-
and ointments; buccal adhesive drug delivery devices are also to adhesive tablets was allowed after a two days period for the same
be evaluated specifically for their mucoadhesive strength and volunteer [189].
permeability.
5.2. Permeation studies
5.1. Determination of the residence time
During the preformulation studies, buccal absorption/perme-
5.1.1. In vitro residence time ation studies must be conducted to determine the feasibility of this
It was determined using a modified USP disintegration appa- route of administration for the candidate drug and to determine the
ratus as shown in Fig. 5. The disintegration medium composed of type of enhancer and its concentration required to control the rate
800 ml isotonic phosphate buffer pH 6.75 maintained at 37 °C. A of permeation of drugs. These studies involve methods that would
segment of rabbit intestinal mucosa, 3 cm long, was glued to the examine in vitro, ex vivo and/or in vivo buccal permeation profile
surface of a glass slab, vertically attached to the apparatus. The and absorption kinetics of the drug.
mucoadhesive tablet was hydrated from one surface using 15 ml
IPB and then the hydrated surface was brought into contact with 5.2.1. In vitro methods
the mucosal membrane. The glass slab was vertically fixed to the Deasy [157] used an apparatus consisting of a water jacket and
apparatus and allowed to move up and down so that the tablet was an internal compartment containing 50 ml of simulated saliva as
completely immersed in the buffer solution at the lowest point and dissolution medium to study the release of cetylpyridinium chlo-
was out at the highest point. The time necessary for complete ride tablet by placing in the metal die sealed at the lower end by
erosion or detachment of the tablet from the mucosal surface was paraffin wax to ensure the drug release from one end alone. The
recorded [189]. medium was stirred with a rotating stirrer at 250 rpm. Ishida [171]
conducted dissolution studies with similar apparatus with slight
5.1.2. In vivo residence time test modification of providing a water jacket for the maintenance
The experiment was conducted on four human healthy of temperature for dosage forms of lidocaine. Nagai [190] used
volunteers of 25–50 years old. Plain bioadhesive tablets with Toyamp-Sangyo TR-553 dissolution tester to measure the

Table 5
Buccal adhesive formulations for proteins/peptides
Protein/peptide drug Dosage form Enhancer Animal model % increase in bioavailability Ref.
Buserelin Patch SGDC Pig, rat 12.7% [175]
Calcitonin Tablet No enhancer Rabbits 37% [176]
Captopril Tablet SGDC Humans [177]
Colony stimulating factor (G-SCF) Patch No enhancer Dogs Two fold increase in [178]
pharmacological action.
Enalapril Solution No enhancer Human No significant increase [179]
Glucagon like peptide Tablet STC Human 4–23% [180]
Gonadotropin releasing hormone Tablet SC, SDC, STC, STDC Dog SDC > SC > STC > STDC [181]
Interferon Solution No enhancer Mice Marked increase [182]
Insulin Liposomes No enhancer Rat No significant increase [183]
Lisinopril Solution No enhancer Human No significant increase [179]
Lutinizing hormone releasing hormone Tablet SDC 5% Dog 237% [184]
Octreotide acetate Azone, SC, EDTA, STC Dog Azone > SC > EDTA > STC [185]
Oxytocin Patch No enhancer Rabbit Slight increase [186]
Protrelin (TRH) Patch Citric acid, Sodium- Human, rats Increase in plasma [187]
5-methoxy salicylate thyrotropin concentration
Recombinant human interferon alpha B/D hybrid Solution No enhancer Rabbit, rat 0.005% [188]
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 35

5.2.3. In vivo methods

5.2.3.1. Selection of animal species. Apart from the specific


methodology used to study buccal drug permeation characteristics,
special attention is warranted to the selection of experimental
animal species for such experiments. Many researchers have used
small animals including rats and hamsters for permeability studies
[196,197]. But unlike humans, most laboratory animals have totally
keratinized oral lining, hence not suitable. The rat has a buccal
mucosa with a very thick, keratinized surface layer. The rabbit is the
only laboratory rodent that has non-keratinized mucosal lining
similar to human tissue. But, the sudden transition to keratinized
tissue at the mucosal margins makes it hard to isolate the desired
Fig. 5. Schematic diagram of the apparatus used for the determination of non-keratinized region [198].
residence time. S: glass slab; D: disintegration apparatus; B: glass beaker; M: Among the larger experimental animals monkeys are practical
mucosal membrane; T: mucoadhesive tablet; IBP: Isotonic phosphate buffer. models because of the difficulties associated with its mainte-
nance. Dogs [199,200] are easy to maintain and less expensive
than monkeys [201] and their buccal mucosa is non-keratinized
dissolution rate of disk like dosage forms by keeping in a rotating and has a close similarity to that of the human buccal mucosa.
basket at 100 rpm in 900 ml of purified water. The same apparatus Pigs also have non-keratinized buccal mucosa similar to that of
was used for the evaluation of oral mucosal dosage forms of human and their inexpensive handling and maintenance costs
insulin. Hughes and Gehris [191] described a novel dissolution make them a highly suitable animal model for buccal drug
testing system that is capable of characterizing buccal dissolution. delivery studies. In fact, the oral mucosa of pigs resembles that of
It comprises of a single, stirred, continuous flow-through filtration human more closely than any other animal in terms of structure
cell that includes a dip tube designed to remove finely divided solid and composition [202].
particles. Filtered solution is removed continuously and used to
analyze for dissolved drug. 5.2.3.2. Buccal absorption test. Beckett and Triggs [203]
developed a method to measure the kinetics of drug absorption.
5.2.2. Ex vivo methods It is carried out by swirling of a 25 ml sample of the test solution
Most of the ex vivo studies examining drug transport across for 15 min by human volunteers followed by the expulsion of the
buccal mucosa uses buccal tissues from animal models. Im- solution. The amount of drug remaining in the expelled volume
mediately after sacrificing the animals the buccal mucosal tissue is then determined to assess the amount of drug absorbed. The
is surgically removed from the oral cavity. The membranes are drawbacks of this method are inability to localize the drug
stored in Krebs buffer at 4 °C until mounted in the diffusion cells solution within a specific site of the oral cavity, accidental
for the ex vivo permeation experiments. Preservation of the swallowing of a portion of the sample solution and the salivary
dissected tissue is an important issue that will affect the studies. dilution of the drug.
There is no standard means by which the viability or the integrity
of the dissected tissue can be assessed. The most meaningful 5.2.3.3. Modified buccal absorption test. Gonzalez-Younes et
method to assess tissue viability is the actual permeation experi- al. [204] developed this method by correcting for salivary
ment itself, if the drug permeability does not change during the dilution and accidental swallowing, but these modifications also
time course of the study under the specific experimental con- suffer from the inability of site localization.
ditions of pH and temperature, then the tissue is considered
viable. 5.2.3.4. Perfusion system. A circulating perfusion chamber
Dowty [192] studied tissue viability by using ATP levels in attached to the upper lip of anesthetized dogs by cyanoacrylate
rabbit buccal mucosa. He reported a 50% drop in the tissue cement and the drug solution is circulated through the device for
ATP concentration during the initial 6 h of the experiment a predetermined period of time. Sample fractions are collected
without a corresponding drop in tissue permeability. Despite from the perfusion chamber and blood samples are drawn at
certain gradual changes, the buccal tissue seems to remain regular intervals [205].
viable for a rather long period of time. Hence, a decrease in
ATP levels does not assure a drop in permeability character- 5.2.3.5. Buccal perfusion cell apparatus. Rathbone [206]
istics of the tissue. developed an apparatus that provides continuous monitoring of
Buccal cell cultures have also been suggested as useful in drug loss as a function of time offers larger area for drug transfer
vitro models for buccal drug permeation and metabolism. and has no leakage problem. He used several methods to study the
However, to utilize these culture cells for buccal drug transport, rate and extent of drug loss from human oral cavity. These include
the number of differentiated cell layers and the lipid composition the buccal absorption test, disk methods and perfusion cells.
of the barrier layers must be well characterized and controlled These methods have provided information on the mechanism by
[193–195]. which drugs are transported across oral cavity membranes and
36 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

suggest that passive diffusion or carrier mediated transport bioavailability of orally less/inefficient drugs by manipulating the
systems may be involved. formulation strategies like inclusion of pH modifiers, enzyme
In vivo buccal permeation of FITC labeled dextran 4400 and inhibitors, permeation enhances etc. Novel buccal adhesive
the peptide drug buserelin was investigated in pigs. The delivery delivery systems, where the drug delivery is directed towards
device consisted of an application chamber with a solution of buccal mucosa by protecting the local environment is also gaining
FD4 or buserelin, and was attached to the buccal mucosa for interest. Currently solid dosage forms, liquids and gels applied to
four hours using an adhesive patch. The randomized crossover oral cavity are commercially successful. The future direction of
study including intravenous administration and buccal delivery buccal adhesive drug delivery lies in vaccine formulations and
without and with 10 mM sodium glycodeoxycholate as an delivery of small proteins/peptides. Microparticulate bioadhesive
absorption enhancer was performed in pigs [207]. systems are particularly interesting as they offer protection to
Tanaka et al. [208] studied the absorption of salicylic acid therapeutic entities as well as the enhanced absorption that result
through the hamster cheek pouch. Ointments containing salicylic from increased contact time provided by the bioadhesive
acid were applied to the cheek pouch of hamster and the influence component. Exciting challenges remain to influence the bioavail-
of the type of base on drug absorption was examined. ability of drugs across the buccal mucosa. Many issues are yet to
be resolved before the safe and effective delivery through buccal
6. Conclusion mucosa. Successfully developing these novel formulations
requires assimilation of a great deal of emerging information
The need for research into drug delivery systems extends about the chemical nature and physical structure of these new
beyond ways to administer new pharmaceutical therapies. The materials.
safety and efficacy of current treatments may be improved if their
delivery rates, biodegradation, and site specific targeting can be References
predicted, monitored and controlled. From both a financial and
global healthcare perspective, finding ways to administer [1] L.M. Sanders, Drug delivery system and routes of administration of
peptide and protein drugs, Eur. J. Drug Metab. Pharmacokinet. 15 (1990)
injectable medications is costly and some time leads to serious
95–102.
hazardous effects. Hence inexpensive multiple dose formulations [2] Y.J. Wang, R. Pearlman, Stability and characterization of protein and
with better bioavailabilities are needed. Improved methods of peptide drugs, case histories, in Pharmaceutical Technology, New York/
drug release through transmucosal and transdermal methods London, vol. 5.
would be of great significance, as by such routes, the pain factor [3] H.H. Alur, T.P. Johnston, A.K. Mitra, Encyclopedia of Pharmaceutical
Technology, in: J. Superbrick, J.C. Boylan (Eds.), Peptides and Proteins:
associated with parenteral routes of drug administration can be
Buccal Absorption, vol. 20 (3), Marcel Dekker Inc., New York,
totally eliminated. Buccal adhesive systems offer innumerable 2001, pp. 193–218.
advantages in terms of accessibility, administration and with- [4] Wikipedia, The free encyclopedia, http://en.wikipedia.org/wiki/.
drawal, retentivity, low enzymatic activity, economy and high [5] The mouth (cavum oris; oral or buccal cavity). XI. Splanchnology, Gray's
patient compliance. Since the introduction of Orabase in 1947, Anatomy of the Human Body.
[6] M.J. Rathbone, G. Ponchel, F.A. Ghazali, Systemic and oral mucosal
when gum tragacanth was mixed with dental adhesive powder to
drug delivery and delivery systems, in: M.J. Rathbone (Ed.), Oral
apply penicillin to the oral mucosa; the market share of bioad- Mucosal Drug Delivery, vol. 74, Marcel Dekker Inc., New York, 1996,
hesive drug delivery systems is increasing. The growth rate for pp. 241–284.
transmucosal drug delivery systems is expected to increase 11% [7] D. Harris, J.R. Robinson, Drug delivery via the mucous membranes of the
annually through 2007. Worldwide market revenues are at $3B oral cavity, J. Pharm. Sci. 81 (1992) 1–10.
[8] R.B. Gandhi, J.R. Robinson, Bioadhesion in drug delivery, Ind. J. Pharm.
with the U.S. at 55%, Europe at 30% and Japan at 10%.
Sci. 50 (3) (1988) 145–152.
Adhesion of buccal adhesive drug delivery devices to [9] P.C. Fox, Acquired salivary dysfunction: drugs and radiation, Ann. N.Y.
mucosal membranes leads to an increased drug concentration Acad. Sci. 842 (1998) 132–137.
gradient at the absorption site and therefore improved bioavail- [10] C.A. Squier, M.W. Finkelstein, in: A.R. Ten Cate (Ed.), Oral Histology,
ability of systemically delivered drugs. In addition, buccal Development, Structure and Function, C.V. Mosby, St. Louis, 1989,
pp. 345–385.
adhesive dosage forms have been used to target local disorders at
[11] C.A. Squier, The permeability of keratinized and nonkeratinized oral
the mucosal surface (e.g., mouth ulcers) to reduce the overall epithelium to horseradish. Peroxidase, J. Ultrastruct. Res. 43 (1973) 160–177.
dosage required and minimize side effects that may be caused by [12] A.J. Hoogstraate, S. Senel, C. Cullander, J. Verhoef, H.E. Junginger, H.E.
systemic administration of drugs. Researchers are now looking Bodde, Effects of bile salts on transport rates and routes of FTIC-labelled
beyond traditional polymer networks to find other innovative compounds across porcine buccal epithelium in vitro, J. Control. Release
40 (1996) 211–221.
drug transport systems. Much of the development of novel
[13] J. Hao, P.W.S. Heng, Buccal delivery systems, Drug Dev. Ind. Pharm. 29
materials in controlled release buccal adhesive drug delivery is (8) (2003) 821–832.
focusing on the preparation and use of responsive polymeric [14] A.H. Shojaei, X. Li, Determination of transport route of acyclovir across
system using copolymer with desirable hydrophilic/hydrophobic buccal mucosa, Proc. Int. Symp. Control. Release Bioact. Mater. 24
interaction, block or graft copolymers, complexation networks (1997) 427–428.
[15] L. Chen, X. Hui-Nan, L. Xiao-Ling, In vitro permeation of tetramethyl-
responding via hydrogen or ionic bonding and new biodegradable
pyrazine across porcine buccal mucosa, Acta Pharmacol. Sin. 23 (2002)
polymers especially from natural edible sources. At the current 792–796.
global scenario, scientists are finding ways to develop buccal [16] H.M. Nielsen, M.R. Rassing, TR146 cells grown on filters as a model of
adhesive systems through various approaches to improve the human buccal epithelium. III. Permeability enhancement by different pH
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 37

value, different osmolarity value, and bile salts, Int. J. Pharm. 185 (1999) second edition, Lippincott_raven publishers, Philadelphia, PA, 1998,
215–225. pp. 531–539.
[17] H. Zhang, J.R. Robinson, In vitro methods for measuring permeability [43] R.D. Mattes, Physiologic responses to sensory stimulation by food:
of the oral mucosa, in: J. Swarbrick, J.C. Boylan (Eds.), Oral Mucosal nutritional implications, J. Am. Diet. Assoc. 97 (1997) 406–410.
Drug Delivery, 1st edition, vol. 74, Marcel Dekker, INC, New York, [44] K.L. Moore, Clinically Oriented Anatomy, Third edition, Williams and
1996, pp. 85–100. Wilkins, MD, Baltimore, 1992, pp. 751–752.
[18] R. Mashru, V. Sutariya, M. Sankalia, J. Sankalia, Transbuccal delivery of [45] U.K. Sinha, M. Ng, Surgery of the salivary glands, Otolaryngol. Clin.
lamotrigine across porcine buccal mucosa: in vitro determination of North Am. 32 (5) (1999) 887–918.
routes of buccal transport, J. Pharm. Pharmaceut. Sci. 8 (1) (2005) 54–62. [46] A.R. Silvers, P.M. Som, Salivary glands, Head Neck Imag. 36 (1998)
[19] M.A. Randhawa, S.A. Malik, M. Javed, Buccal absorption of weak acidic 941–966.
drugs is not related to their degree of ionization as estimated from the [47] W.R. Galey, H.K. Lonsdale, S. Nacht, The in vitro permeability of skin
Henderson–Hasselbalch equation, Pak. J. Med. Res. 42 (2) (2003). and buccal mucosa to selected drugs and tritiated water, J. Invest. Dermat.
[20] N. Utoguchi, Y. Watanabe, T. Suzuki, J. Maeharai, Y. Matsumoto, 67 (1976) 713–717.
M. Matsumoto, Pharm. Res. 14 (1997) 320–324. [48] H. Batchelor, Novel bioadhesive formulations in drug delivery, The Drug
[21] C.A. Squier, M.J. Kremer, P.W. Wertz, J. Pharm. Sci. 86 (1997) 82–84. Delivery Companies Report Autumn/Winter, Pharma Ventures Ltd, 2004.
[22] P.P.H.L. Brun, P.L.A. Fox, M.E.D. Vries, H.E. Bodde, In vitro penetration [49] P.R. Mishra, A. Namdeo, S. Jain, N.K. Jain, Hydrogels as drug delivery
of some β-adrenoreceptor blocking drugs through porcine buccal system, Indian Drugs 33 (5) (1996) 181–186.
mucosa, Int. J. Pharm. 49 (1989) 141–145. [50] A. Lowman, N.A. Peppas, Complexation graft copolymers as oral drug
[23] K.L. Audus, Oral mucosal drug delivery, in: M.J. Rathbone (Ed.), Marcel delivery systems, Polym. Prepr. 38 (2) (1997) 566–567.
Dekker, New York, pp 101–119. [51] M.D. Hornof, W. Weyenberg, A. Ludwig, A. Bernkop-Schnurch,
[24] H.M. Nielsen, M.R. Rassing, TR146 cells grown on filters as a model of J. Control. Release 89 (2003) 419–428.
human buccal epithelium. III. Permeability enhancement by different pH [52] P.L. Soo, L. Luo, D. Maysinger, A. Eisenberg, Incorporation and release
value, different osmolarity value, and bile salts, Int. J. Pharm. 185 (1999) of hydrophobic probes in biocompatible polycaprolactone-block-poly
215–225. (ethylene oxide) micelles: implications for drug delivery, Langmuir 18
[25] C.A. Squier, J. Ultrastruct. Res. 60 (1977) 212–220. (2002) 9996–10004.
[26] C. Grubb, M. Hackemann, K.R. Hill, J. Ultrastruct. Res. 22 (1968) [53] R. Saviae, L.L.A. Eisenberg, D. Maysinger, Micellar nanocontainers distribute
458–468. to defined cytoplasmic organelles, Science 300 (2003) 615–618.
[27] D. Hopwood, K.R. Logan, I.A. Bouchier, D. Virchows, Arch. B. Cell [54] C. Allen, D. Maysinger, A. Eisenberg, Nano-engineering block
Path. 26 (1978) 345–358. copolymer aggregates for drug delivery, Colloids Surf., B. Biointerfaces
[28] K. Wolff, H.J. Honigsmann, J. Ultrastruct. Res. 36 (1971) 176–190. Spec. Issue, Polym. Micelles Biol. Pharma. 16 (1999) 3–27.
[29] A. Allen, in: J.G. Forte (Ed.), Handbook of Physiology — the [55] C.E. Kast, D. Guggi, N. Langoth, A. Bernkop-Schnürch, Pharm. Res. 20
Gastrointestinal Physiology, Salivary, Gastric and Hepatobiliary Secre- (2003) 931–936.
tions, vol. III(6), American Physiological Society, Bethesda, MD, 1989, [56] V.M. Leitner, D. Guggi, A. Bernkop-Schnürch, 5th Central Eur. Symp.
pp. 359–382. Pharm. Technology, Ljubljana, Slovenia, 2003.
[30] F.S. Rosen, B.J. Bailey, Anatomy and physiology of salivary glands, [57] United States Patent: 6,916,485 Title: Prolonged release bioadhesive
Grand Rounds presentation, UTMB, Department of Otolaryngology. therapeutic systems. Inventors: Aiache; Jean-Marc (Paris, FR); Costan-
[31] N.A. Peppas, P.A. Buri, Surface, interfacial and molecular aspects tini; Dominique (Paris, FR); Chaumont; Christine (Paris, FR) Issued: July
of polymer bioadhesion on soft tissues, J. Control. Release 2 (1985) 12, 2005 Assignee: Bioalliance Pharma (Paris, FR).
257–275. [58] J. Heller, D.C. Washington, D.W.H. Penhale, Use of bioerodible polymers in
[32] E. Odeblad, The discovery of different types of cervical mucus, Bull. self-regulated drug delivery systems, in: P.I. Lee, W.R. Good (Eds.),
Ovul. Method Res. Ref. Cent. Aust. 21 (1994) 3–35. Controlled Release Technology, Pharmaceutical Applications, Washing-
[33] S.E. Harding, J.M. Creeth, A.J. Rowe, in: A. Chester, D. Heinegard, A. ton DC, ACS Symposium Series, vol. 76, 1997, pp. 281–282.
Lundblad, S. Svenssion (Eds.), Proceedings of the 7th International [59] J.R. Robinson, X. Yang, Absorption enhancers, in: J. Swarbrick, J.C.
Glucoconjugates Conference Olsson Reklambyra, Sweden, 1983, Boylan (Eds.), Encyclopedia of Technology, vol. 18, Marcel Dekker Inc,
pp. 558–559. New York, 1999, pp. 1–27.
[34] M.R. Jimmenez-Castellanos, H. Zia, C.T. Rhodes, Mucoadhesive drug [60] J.A. Weathercell, C. Robinson, M.J. Rathbone, Site-specific differences
delivery systems, Drug Dev. Ind. Pharm. 19 (1 and 2) (1993) 143–194. in the salivary concentrations of substances in the oral cavity —
[35] S.E. Harding, An analysis of the heterogeneity of mucins: no evidence for implications for the etiology of oral-disease and local drug delivery, Adv.
a self-association, Biochem. J. 219 (1984) 1061–1064. Drug Del. Rev. 130 (1–2) (1994) 24–42.
[36] S.E. Harding, Mucoadhesive interactions, Adv. Carbohydr. Chem. [61] F. Veuillez, Y.N. Kalia, Y. Jacques, J. Deshusses, P. Buri, Factors and
Biochem. 47 (1989) 345–381. strategies for improving buccal absorption of peptides, Eur. J. Pharm.
[37] S. Dodd, G.A. Place, R.L. Hall, S.E. Harding, Hydrodynamic properties Biopharm. 51 (2) (2001) 93–109.
of mucins secreted by primary cultures of Guinea pig tracheal [62] K. Khanvilkar, M.D. Donovan, D.R. Flanagan, Drug transfer through
epithelial cells: determination of diffusion coefficients by analytical mucus, Adv. Drug Del. Rev. 48 (2–3) (2001) 173–193.
ultracentrifugation and kinetic analysis of mucus gel hydration and [63] S. Jay, W. Fountain, Z. Cui, R.J. Mumper, Transmucosal delivery of
dissolution, Eur. Biophys. J. 28 (1998) 38–47. testosterone in rabbits using novel bi-layer mucoadhesive wax-film
[38] P. Hallet, A.J. Rowe, S.E. Harding, A highly expanded spheroidal model composite disks, J. Pharm. Sci. 91 (9) (2002) 2016–2025.
for a mucus glycoprotein from a cystic fibrosis patient: new evidence [64] S.C. Chattarajee, R.B. Walker, Penetration enhancer classification, in:
from electron microscopy, Biochem. Soc. Trans. 12 (1984) 878–879. E.W. Smith, H.I. Maibach (Eds.), Percutaneous Penetration Enhance-
[39] L.M.C. Collins, C. Dawes, J. Dent. Res. 66 (1987) 1300–1302. ment, CRC Press, Boca Raton, FL, 1995, pp. 1–4.
[40] M.J. Levine, P.C. Jones, R.E. Looms, M.S. Reddy, I. Al-Hashimi, E.J. [65] A.H. Shojaei, Buccal mucosa as a route for systemic drug delivery: a
Bergey, in: I.C. Mackenzie, C.A. Squierv, Dablesteen (Eds.), Oral review, J. Pharm. Pharmaceut. Sci. 1 (1) (1998) 15–30.
Mucosal Diseases: Biology, Etiology and Therapy, Laege-foreningens [66] A. Aungst, Permeability and metabolism as barriers to transmucosal delivery
Folag, Copenhagen, 1987, pp. 7–9. of peptides and proteins. in: D.S. Hsieh (Ed.) , Drug Permeation Enhancement.
[41] L. Schenkels, T.L. Gururaja, M.J. Levine, in: M.J. Rathbone (Ed.), Oral Theory and Applications, Marcel Dekker, New York, (1994) 323-343.
Mucosal Drug Delivery, Marcel Dekker, New York, 1996, pp. 191–220. [67] Y. Kurosaki, S. Hisaichi, L. Hong, T. Nakayana, Int. J. Pharm. 51 (1889)
[42] T.C. Kontis, M.E. Johns, Anatomy and physiology of salivary glands, 47–52.
in: Byron J. Bailey (Ed.), Head and Neck surgery–Otolaryngology, [68] V. Lee, Crit. Rev. Ther. Drug Carr. Syst. 8 (1991) 91–92.
38 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

[69] A. Ganem, R. Falson, P. Buri, Eur. J. Drug Metab. Pharmacokinet. 111 [97] P.M. Claesson, H.K. Christensson, Very long-range attractive forces
(1996) 112–123. between uncharged hydrocarbon and fluorocarbon surface in water,
[70] J.A. Siegel, H.P. Gordon, Toxicol. Lett. 26 (1985) 153–157. J. Phys. Chem. 92 (1988) 1650–1655.
[71] C.K. Oh, W.A. Ritschel, Biopharmaceutic aspects of buccal absorption of [98] L.G.T. Erikson, P.M. Claesson, S. Ohnishi, H. Masakatsu, Stability of
insulin, Methods Find. Exp. Clin. Pharmacol. 12 (1990) 205–212. dimethyldioctadecyl-ammonium bromide LB films on mica in aqueous
[72] I.A. Siegel, H.P. Gordon, Effects of surfactants on the permeability of salt solutions, Thin Solid Films 300 (1997) 240–255.
canine oral mucosa in-vitro, Toxicol. Lett. 26 (1985) 153–157. [99] K.A. Dill, Science 250 (1990) 297.
[73] G.J.M. Wolany, J. Munzer, A. Rummelt, H.P. Merkle, Buccal absorption [100] R. Baldwin, Proc. Natl. Acad. Sci. U. S. A. 83 (1986) 8069.
of Sandostatin (octreotide) in conscious beagle dogs, Proc. Int. Symp. [101] K.R. Kamath, K. Park, Mucosal adhesive preparations, in: J. Swabrick,
Control. Release Bioact. Mater. 17 (1990) 224–225. J.C. Boylan (Eds.), Encyclopedia of Pharmaceutical Technology,
[74] A. Steward, D.L. Bayley, C. Howes, The effect of enhancers on the vol. 10, Marcel Dekker, New York, 1994, pp. 133–163.
buccal absorption of hybrid (BDBB) alpha interferon, Int. J. Pharm. 104 [102] J.W. Lee, J.H. Park, J.R. Robinson, Bioadhesive based dosage forms: the
(1994) 145–149. next generation, J. Pharm. Sci. 89 (7) (2000) 850–866.
[75] A.M. Manganaro, P.W. Wertz, The effects of permeabilizers on the in [103] C.M. Lehr, F.G.J. Poelma, H.E. Junginger, J.J. Tucker, An estimate of
vitro penetration of propranolol through porcine buccal epithelium, Mil. turnover time of intestinal mucus gel layer in the rat intestinal in situ loop,
Med. 161 (1996) 669–672. Int. J. Pharm. 70 (1991) 235–240.
[76] S. Senel, M.J. Kremer, S.H. Ka, P.W. Wertz, A.A. Hıncal, C.A. Squier, [104] J.D. Smart, M.E. Johnson, A new technique for assessing mucoadhesion
in: M.G. Peter, R.A.A. Muzzarelli, A. Domard (Eds.), Effect of Chitosan by application of tensile and shear stresses, Eur. J. Pharm. Sci. 4 (1996)
in Enhancing Drug Delivery Across Buccal Mucosa, Advances in S65–S65.
Chitin Science, University of Potsdam, vol. 4, 2000, pp. 254–258. [105] M. Ishida, Y. Machida, N. Nambu, T. Nagai, Chem. Pharm. Bull. 29 (1981)
[77] S. Senel, M.J. Kremer, S.H. Ka, P.W. Wertz, A.A. Hıncal, C.A. Squier, 810–816.
Enhancing effect of chitosan on peptide drug delivery across buccal [106] R. Gurney, J.M. Meyer, N.A. Peppas, Bioadhesive intraoral release
mucosa, Biomaterials 21 (2000) 2067–2071. systems: design, testing and analysis, Biomaterials 5 (1984) 336–340.
[78] N.G.M. Schipper, K.M. Varum, P. Artursson, Chitosans as absorption [107] K. Park, H. Park, Test methods of bioadhesion, Int. J. Pharm. 52 (1989)
enhancers for poorly absorbable drugs: influence of the molecular 265–270.
weight and degree of acetylation on drug transport across human [108] S. Koclkisch, G.D. Rees, S.A. Young, J. Tsibouklis, J.D. Smart, A direct
intestinal epithelium (Caco-2) cells, Pharm. Res. 21 (2004) 344–353. staining method to evaluate the mucoadhesion of polymers from aqueous
[79] M.A. Longer, J.R. Robinson, Fundamental aspects of bioadhesion, dispersion, J. Control. Release 77 (1–2) (2001) 1–6.
Pharm. Int. 7 (1986) 114–117. [109] H. Hagerstorm, K. Edsman, Limitations of the rheological method: the
[80] P. J. Glantz, T. Arnebrant, T. Nylander, R.E. Baier, Bioadhesion—a effect of the choice of conditions and the rheological synergism parameter,
phenomenon with multiple dimensions, Acta Odontol. Scand. 57 (1999) Eur. J. Pharm. Sci. 18 (5) (2003) 349–357.
238–241. [110] E.E. Hassan, J.M. Gallo, Pharm. Res. 7 (1990) 491.
[81] A. Ahuja, R.K. Khar, J. Ali, Mucoadhesive drug delivery systems, Drug [111] S.A. Mortzavi, J.D. Smart, An investigation of some factors influencing the
Dev. Ind. Pharm. 23 (5) (1997) 489–517. in vitro assessment of mucoadhesion, Int. J. Pharm. 116 (1995) 223–230.
[82] T. Nylander, N.M. Wahlgren, Forces between adsorbed layers of β- [112] C. Carmella, S. Rossi, M.C. Bonferoni, F. Ferrari, Characterization of
casein, Langmuir 13 (1997) 6219–6225. chitosan hydrochloride–mucin rheological interaction: influence of polymer
[83] H. Elwing, B. Nilson, K.E. Svensson, A. Askendahl, U.R. Nilson, L. concentration and polymer:mucin weight ratio, Eur. J. Pharm. Sci. 12 (2001)
Lundstrom, Conformational changes of model protein (complement factor 3) 479–485.
adsorbed on hydrophilic and hydrophobic solid surface, J. Colloid Interface [113] S.E. Harding, Mucoadhesive interactions, Biochem. Soc. Trans. 31 (5)
Sci. 125 (1988) 139–145. (2003) 1036–1041.
[84] P.C. Hiemenz, Principles of Colloid and Surface Chemistry, Marcel [114] K. Park, J.R. Robinson, Bioadhesive polymers as platforms for oral
Dekker, New York, 1986. controlled drug delivery, Int. J. Pharm. 19 (1984) 107–127.
[85] J.N. Israelachvili, Intermolecular and Surface Forces, London academic [115] P.K. Nanti, D.J. Cook, D.J. Rogers, J.D. Smart, Lectins for drug delivery
press, 1991. within the oral cavity — investigation of lectin binding to oral mucosa,
[86] A. Martin, P. Bustamante, P. chun, (eds), BI Waverly Pvt ltd. New Delhi, J. Drug Target 5 (1997) 45–55.
4th edition. 1995. [116] H. Park, On the mechanism of bioadhesion. Ph.D. Thesis, Pharmaceutics,
[87] G.M. Whitesides, J.P. Mathias, C.T. Seto, Science 254 (1991) 1312. School of Pharmacy, University of Wisconsin-Madison, 1986.
[88] E.A. Rawlins (Ed.), Bentley's Textbook of Pharmaceutics, 8th edition, [117] R.M. Barrer, J.A. Barrie, P.S.L. Wong, The diffusion and solution of
ELBS publishers, 1984, pp. 32–33. gases in highly cross-linked copolymers, Polymer 9 (1968) 609–627.
[89] B.W. Ninham, V. Yaminsky, Ion binding and ion specificity: the [118] H.S. Ch'ng, H. Park, P. Kelly, J.R. Robinson, Bioadhesive polymers as
Hofmeister effect, Onsager and Lifshitz theories, Langmuir 13 (1997) platforms for oral controlled drug delivery. II. Synthesis and evaluation of
2097–2108. some swelling, water-insoluble bioadhesive polymers, J. Pharm. Sci. 74
[90] T. Nylander, P. Kekicheff, B.W. Ninham, The effect of solution behavior (1985) 399–405.
of insulin on interactions between adsorbed layers of insulin, J. Colloid [119] D. Duchene, F. Touchard, N.A. Peppas, Pharmaceutical and medical
interface Sci. 164 (1994) 136–150. aspects of bioadhesive systems for drug administration, Drug Dev. Ind.
[91] K. Larsson, P.O. Glantz, Microbial adhesion to surfaces with different Pharm. 14 (1988) 283–318.
charges, Acta Odontol. Scand. 39 (1981) 79–82. [120] E. Mathiwitz, D.E. Chickering, C.M. Lehr, Bioadhesive Drug Delivery
[92] M.J Tobyn, J.R. Johnson, S.A.W. Gibson, J. Pharm. Pharmacol. 44 Systems: Fundamentals, Novel Approaches and Development, Marcel
(1992) 1048 (suppl). Dekker, Inc., New York, 1999.
[93] H.K. Christensson, P.M. Claesson, Cavitation and the interaction between [121] M.J. Tobyn, J.N. Staniforth, E. Jorgensen, D. Bhagwat, Use of gel
microscopic hydroscopic surfaces, Science 239 (1988) 390–392. strength analysis to characterize a polysaccharide controlled release
[94] V.M. Leitner, G.F. Walker, A. Bernkop-Schnurch, Eur. J. Pharm. matrix, Controlled Release of Bioactive Materials Conference Paper,
Biopharm. 56 (2) (2003) 207–214. 1997.
[95] K. Kafedjiiski, Multifunctional Polymeric Excipients in Non-Invasive [122] J.L. Chen, G.N. Cyr, in: R.S. Manly (Ed.), Composition Producing
Delivery of Hydrophilic Macromolecular Drugs, The Drug Delivery Adhesion Through Hydration, Adhesion in Biological System, Academic
Companies Report Autumn/Winter, Pharm Ventures Ltd, 2004. press, New York, 1970, pp. 163–181.
[96] D. Guggi, M.K. Marschutz, A. Bernkop-Schnurch, Int. J. Pharm. 274 [123] C.M. Lehr, J.A. Bouwstra, J.J. Tucker, H.E. Junginger, Intestinal transit
(2004) 97–105. of bioadhesive microspheres in an in situ loop in the rat — a comparative
Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40 39

study with copolymers and blends bases on poly (acrylic acid), J. Control. [146] B. Mizrahi, J. Golenser, J.S. Wolnerman, A.J. Dombi, Adhesive tablet
Release 13 (1990) 51–62. effective for treating canker sores in humans, Assoc. J. Pharm. Sci. 93
[124] D.S. Jones, A.D. Woolfson, A.F. Brown, Textural analysis and flow (2004) 2927–2935.
rheometry of novel bioadhesive antimicrobial oral gels, Pharm. Res. 114 [147] Q. Du, Q.N. Ping, G.J. Liu, Preparation of Buspirone hydrochloride
(4) (1997) 450–457. buccal adhesive tablet and study on its drug release mechanism, Yao Xue
[125] S. Miyazaki, A. Nakayama, M. Oda, M. Takada, D. Attwood, Chitosan Xue Bao 37 (8) (2002) 653–656.
and sodium alginate based bioadhesive tablets for intraoral drug delivery, [148] H. Choi, J. Jung, C.S. Yong, C. Rhee, M. Lee, J. Han, K. Park, C. Kim,
Biol. Pharm. Bull. 17 (1994) 745–747. Formulation and in vivo evaluation of Omeprazole buccal adhesive
[126] C. Remunan-Lopez, A. Portero, J.L. Vila-Jato, M.J. Alonso, Design and tablet, J. Control. Release 68 (3) (2000) 405–412.
evaluation of chitosan/ethylcellulose mucoadhesive bilayered devices for [149] G.C. Ceschel, P. Maffei, S.L. Borgia, C. Ronchi, Design and evaluation of
buccal drug delivery, J. Control. Release 55 (1998) 143–152. buccal adhesive hydrocortisone acetate tablets, Int. J. Pharm. 238 (2002)
[127] B. Taylan, Y. Capan, O. Guven, S. Kes, A.A. Hincal, Design and 161–170.
evaluation of sustained release and buccal adhesive propronolol [150] K. Tsutsumi, Y. Obata, T. Nagai, T. Loftsson, K. Takayama, Buccal
hydrochloride tablets, J. Control. Release 38 (1) (1996) 11–20. absorption of ergotamine tartrate using the bioadhesive tablet system in
[128] K.G.H. Desai, T.M.P. Kumar, Preparation and evaluation of a novel guinea-pigs, Int. J. Pharm. 238 (1–2) (2002) 161–170.
buccal adhesive system, AAPS PharmSciTech 5 (2004) 1–9. [151] Dinsheet, S.P. Agarwal, A. Ahuja, Preparation and evaluation of buccal
[129] B. Taylan, Y. Capan, O. Güven, S. Kes, A.A. Hincal, Design and adhesive tablets of Hydralazine hydrochloride, Indian J. Pharm. Sci. 59
evaluation of sustained-release and buccal adhesive propranolol hydro- (3) (1997) 135–141.
chloride tablets, J. Control. Release 38 (1996) 11–20. [152] M. Rafiee-Tehrani, G. Jazayeri, T. Toliyat, K. Bayati, K. Khalkhali, K.
[130] N.A. Nafee, F.A. Ismail, N.A. Boraie, L.M. Mortada, Mucoadhesive Shamimi, A. Miremadi, F.A. Dorkoosh, Development and in-vitro
delivery systems. II. Formulation and in-vitro/in-vivo evaluation of evaluation of novel buccoadhesive tablet formulation of prednisolone,
buccal mucoadhesive tablets containing water-soluble drugs, Drug Dev. Acta Pharm. 52 (2002) 123–130.
Ind. Pharm. 30 (2004) 995–1004. [153] J.P. Cassidy, N.M. Landzert, E. Quadros, Controlled buccal delivery of
[131] J.M. Labot, R.H. Manzo, A. Allemandi, Double layered mucoadhesive buprenorphine, J. Control. Release 25 (1993) 21–29.
tablets containing nystatin, AAPS PharmSciTech 3 (3) (2002) 22. [154] S. Anlar, Y. Capan, O. Guven, A. Gogus, T. Dlakara, A.A. Hincal,
[132] S. Bouckaert, H. Schautteet, R.A. Lefebvre, J.P. Remon, R.V. Clooster, Formulation and in vitro and in vivo evaluation of buccoadhesive
Double-layered mucoadhesive tablets containing nystatin, Eur. J. Clin. morphine sulphate tablets, Pharm. Res. 11 (2) (1994) 231–236.
Pharmacol. 43 (1992) 137. [155] E. Beyssac, J.M. Aiche, C. Chezaubernarul, H. Caplin, M.J. Douin, A.
[133] A. Gupta, S. Garg, R.K. Khar, Interpolymer complexation and its effect Renoux, Proc. Int. Symp. Control. Rel. Biact. Mater., Contr. Rel. Soc. 21
on bioadhesion strength and dissolution characteristics of buccal drug (1984).
delivery systems, Drug Dev. Ind. Pharm. 20 (1994) 315–325. [156] G.C. Ceschel, P. Maffei, S.L. Borgia, Design and evaluation of a new
[134] T. Nagai, Adhesive topical drug delivery system, J. Control. Release 2 mucoadhesive bilayered tablet containing nimesulide for buccal admin-
(1985) 121–134. istration, Drug Deliv. 11 (2004) 225–230.
[135] T. Nagai, R. Konishi, Buccal/gingival drug delivery systems, J. Control. [157] A.E. Collins, P.B. Deasy, Bioadhesive lozenge for the improved delivery
Release 6 (1987) 353–360. of cetylpyridinium chloride, J. Pharm. Sci. 79 (2) (1990) 116.
[136] T. Nagai, Y. Machida, Buccal delivery systems using hydrogels, Adv. [158] M. Artusi, P. Santi, P. Colombo, H.E. Junginger, Buccal delivery of
Drug Del. Rev. 11 (1993) 179–191. thiocolchicoside: in vitro and in vivo permeation studies, Int. J. Pharm.
[137] G. Ponchel, F. Touchard, D. Wouessidjewe, D. Duchëne, N.A. Peppas, 250 (1) (2003) 203–213.
Bioadhesive analysis of controlled release systems. III. Bioadhesive and [159] H.K. Goud, T.M.P. Kumar, Preparation and evaluation of a novel buccal
release behaviour of metronidazole containing poly (acrylic acid)- adhesive systems, AAPS PharmSciTech 5 (3) (2004) 35.
hydoxypropyl methylcellulose systems, Int. J. Pharm. 38 (1987) 65–70. [160] L.E. Bromberg, D.K. Buxton, P.M. Friden, J. Control. Release 71 (2001)
[138] P. Bottenberg, R. Cleymaet, C.D. Mynck, J.P. Remenn, D. Coomans, Y. 251–259.
Michotte, D. Slop, Comparison of salivary fluoride concentration after [161] J.E. Codd, P.B. Deasy, Int. J. Pharm. 173 (1998) 13–24.
administration of a bioadhesive slow-release tablet and a conventional [162] H. He, X. Cao, I.J. Lee, J. Control. Release 95 (2004) 391–402.
fluoride tablet, J. Pharm. Pharmacol. 43 (1991) 457. [163] Z. Cui, R.J. Mumper, Bilayer films for mucosal (genetic) immunization
[139] S. Bouckaert, H. Schautteet, R.A. Lefebvre, J.P. Remon, R. Van Clooster, via the buccal route in rabbits, Pharm. Res. 19 (2002) 947–953.
Comparison of salivary miconazole concentrations after administration of [164] P. Perugini, I. Genta, B. Conti, T. Modena, F. Pavanetto, Periodontal
a bioadhesive slow-release buccal tablet and an oral gel, Eur. J. Clin. delivery of ipriflavone: new chitosan/PLGA film delivery system for a
Pharmacol. 43 (1992) 137–140. lipophilic drug, Int. J. Pharm. 18 (2003) 1–9.
[140] V. Agarwal, B. Mishra, Design, development, and biopharmaceutical [165] S. Senel, G. Ikinci, S. Kas, A. Yousefi-Rad, M.F. Sargon, A.A. Hıncal,
properties of buccoadhesive compacts of pentazocine, Drug Dev. Ind. Chitosan films and hydrogels of chlorhexidine gluconate for oral mucosal
Pharm. 25 (1999) 701–709. delivery, Int. J. Pharm. 193 (2000) 197–203.
[141] H.H. Alur, S.I. Pather, A.K. Mitra, T.P. Johnston, Transmucosal [166] D. Tiwari, D. Goldman, R. Sause, P.L. Madan, Evaluation of
sustained-delivery of chlorpheniramine maleate in rabbits using a novel polyoxyethylene homopolymers for buccal bioadhesive drug delivery
natural mucoadhesive gum as an excipient in buccal tablets, Int. J. Pharm. device formulations, AAPS PharmSci 1 (1999) 1–8.
188 (1999) 1–10. [167] R. Anders, H. Merckle, Evaluation of laminated mucoadhesive patches
[142] H.G. Choi, C.K. Kim, Development of omeprazole buccal adhesive for buccal drug delivery, Int. J. Pharm. 49 (1989) 231–240.
tablets with stability enhancement in human saliva, J. Control. Release 68 [168] R. Anders, H.P. Merckle, W. Schurr, R. Ziegler, Buccal absorption of
(2000) 397–404. Protirelin: an effective way to stimulate Thyrotropin and Prolactin,
[143] C.R. Park, D.L. Munday, Development and evaluation of a biphasic J. Pharm. Sci. 72 (1983) 1481–1483.
buccal adhesive tablet for nicotine replacement therapy, Int. J. Pharm. 237 [169] J.H. Guo, Investigating the surface properties and bioadhesion of buccal
(2002) 215–226. patches, J. Pharm. Pharmacol. 46 (1994) 647–650.
[144] K. Rajesh, S.P. Agarwal, A. Ahuja, Buccoadhesive erodible carriers for [170] K. Danjo, H. Kato, A. Otsuka, K. Ushimaru, Fundamental study on the
local drug delivery: design and standardization, Int. J. Pharm. 138 (1996) evaluation of strength of granular particles, Chem. Pharm. Bull. 42 (1994)
68–73. 2598–2603.
[145] G. İkinci, S. Senel, C.G. Wilson, M. Şumnu, Development of buccal [171] M. Ishida, N. Nambu, T. Nagai, Mucosal dosage form of lidocaine for
bioadhesive nicotine tablet formulation for smoking cessation, Int. J. Pharm. toothache using hydroxypropyl cellulose and carbopol, Chem. Pharm.
277 (1–2) (2004) 173–178. Bull. 30 (1982) 980–984.
40 Y. Sudhakar et al. / Journal of Controlled Release 114 (2006) 15–40

[172] N.A. Nafee, F.A. Ismail, A. Nabila, L.M. Boraie, Mucoadhesive buccal [192] M.E. Dowty, K.E. Knuth, B.K. Irons, J.R. Robinson, Transport of
patches of miconazole nitrate: in vitro/in vivo performance and effect of thyrotropin releasing hormone in rabbit buccal mucosa in vitro, Pharm.
ageing, Int. J. Pharm. 264 (2003) 1–14. Res. 9 (1992) 1113–1122.
[173] T. Save, U.M. Shah, A.R. Ghamande, P. Venkatachalam, Comparative [193] M.W. Hill, C.A. Squier, The permeability of oral palatal mucosa
study of buccoadhesive formulations and sublingual capsules of maintained in organ cell cultures, J. Anat. 128 (1979) 169–178.
nifedipine, J. Pharm. Pharmacol. 46 (3) (1994) 192–195. [194] M.R. Tavakoli-Saberi, K.L. Andus, Cultured buccal epithelium: an in
[174] A.H. Shojaei, X. Li, Mechanisms of buccal mucoadhesion of novel vitro model derived from the hamster pouch for studying transport and
copolymers of acrylic acid and polyethylene glycol monomethylether metabolism, Pharm. Res. 6 (1989) 160–162.
monomethacrylate, J. Control. Release 47 (1997) 151–161. [195] H.R. Leipold, E. Quadros, Nicotine permeation through buccal cultures,
[175] A.J. Hoogstraate, J.C. Verhoef, A. Pijpers, L.A.V. Leengoed, J.H. Proc. Int. Symp. Control. Release Bioact. Mater. 20 (1993) 242–243.
Verheijden, H.E. Junginger, H.E. Bodde, In vivo buccal delivery of the [196] B.J. Angust, N.J. Rogers, Comparison of the effects of various
peptide drug buserelin with glycodeoxycholate as an absorption enhancer transmucosal absorption promoters on buccal insulin delivery, Int. J
in pigs, Pharm. Res. 13 (1996) 1233–1237. Pharm. 53 (1989) 227–235.
[176] H. Alur, J.D. Beal, S.I. Pather, A.K. Mitra, T.P. Johnston, J. Pharm. Sci. [197] I.A. Siegel, H.P. Gordon, Surfactant — induced increase of permeability
88 (12) (1999) 1313–1319. of rat oral mucosa to non-electrolytes in vivo, Arch. Oral Biol. 30 (1985)
[177] Y. Yaziksiz-Iscan, Y. Capan, S. Senel, M.F. Sahin, S. Kes, D. Duchêne, A.A. 43–47.
Hincal, S.T.P. Pharma, Pharm. Sci. 8 (1998) 357–363. [198] C.A. Squier, P.W. Wertz, in: M.J. Rathbone (Ed.), Structure and Function
[178] Y. Ito, Z. Hu, M. Yoshikawa, M. Murakami, K. Takada, Proc. Int. Symp. of the Oral Mucosa and Implications for Drug Delivery, Oral Mucosal
Control. Release Bioact. Mater. 26 (1999) 6303–6304. Drug Delivery, Marcel Dekker Inc., New York, 1996, pp. 1–26.
[179] J.C. Mcelnay, T.A. Furaih, C.M. Hughes, M.G. Scott, J.S. Elborn, D.P. [199] M. Ishida, Y. Machida, N. Nambu, T. Nagai, New mucosal dosage forms
Nicholls, Eur. J. Clin. Pharmacol. 54 (1998) 609–614. of insulin, Chem. Pharm. Bull. 84 (1981) 810–816.
[180] M.K. Gutniak, H. Larsson, S.J. Heiber, O.T. Juneskans, J.J. Holst, B. [200] C.L. Barsuchn, L.S. Olanoff, D.D. Gleason, E.L. Olanoff, D.D. Gleason,
Ahren, Diabetes Care 19 (1996) 843–848. E.L. Adkins, N.F.H. Ho, Human buccal absorption of flubiprofen, Clin.
[181] Y. Chien, S. Nakane, Y. Lee, M. Kakumoto, K. Yukimatsu, Proc. 1st Pharmacol. Ther. 44 (1988) 225–231.
World Meeting APGI/APV, Budapest, 1995, pp. 813–814. [201] M. Mehta, B.W. Kemppainen, R.G. Stafford, In vitro penetration of
[182] M.G. Towey, C. Maury, J. Interferon Cytokine Res. 19 (2) (1999) tritium labeled water (THO) and [3H] Pb Tx-3 (a red tide toxin) through
145–155. monkey buccal mucosa and skin, Toxicol. Lett. 55 (1991) 185–194.
[183] J. Zhang, S. Nui, C. Ebert, T.H. Stanley, Int. J. Pharm. 101 (1994) 15–22. [202] C.A. Squier, P. Cox, P.W. Wertz, Lipid content and water permeability of
[184] S. Nakane, M. Kakumoto, K. Yukimatsu, Y.W. Chien, Pharm. Dev. skin and oral mucosa, J. Invest. Dermat. 96 (1991) 123–126.
Technol. 1 (1996) 251–259. [203] A.H. Beckett, E.J. Triggs, Buccal absorption of basic drugs and its
[185] H.P. Merkle, G.J.M. Wolany, J. Control. Release 21 (1992) 155–164. application as an in vivo model of passive drug transfer through lipid
[186] C. Li, P.B. Padmanabh, T.P. Johnston, Pharm. Dev. Technol. 2 (3) (1997) membranes, J. Pharm. Pharmacol. 19 (1967) 31S–41S.
265–274. [204] I. Gonzalez-Younes, J.G. Wagner, D.A. Gaines, Absorption of flubripro-
[187] R. Anders, H.P. Merkle, W. Schurr, R. Ziegler, J. Pharm. Sci. 72 (1983) fen through human buccal mucosa, J. Pharm. Sci. 80 (1991) 820–823.
1481–1483. [205] H. Yamahara, V.H. Lee, Drug metabolism in the oral cavity, Adv. Drug
[188] D. Bayley, C. Temple, V. Clay, A. Steward, N. Lowther, J. Pharm. Del. Rev. 12 (1993) 25–39.
Pharmacol. 47 (1995) 721–724. [206] M.J. Rathbone, J. Hadgraft, Absorption of drugs from human oral cavity,
[189] N.A. Nafee, F.A. Ismail, N.A. Boraie, L.M. Mortada, Mucoadhesive Int. J. Pharm. 74 (1991) 9–24.
delivery systems. I. Evaluation of mucoadhesive polymers for buccal [207] H.E. Junginger, J.A. Hoogstate, J.C. Verhoef, Recent advances in buccal
tablet formulation, Drug Dev. Ind. Pharm. 30 (9) (2004) 985–993. drug delivery and absorption — in vitro and in vivo studies, J. Control.
[190] Y. Machida, H. Masuda, N. Fujiyama, S. Ito, M. Iwata, T. Nagai, Chem. Release 62 (1–2) (1999) 149–159.
Pharm. Bull. 27 (2) (1990) 116. [208] M. Tanaka, N. Yanagibashi, H. Fukuda, T. Nagai, Absorption of salicylic
[191] D.L. Hughes, A. Gehris, A new method for characterizing the buccal acid through the oral mucous membrane of hamster cheek pouch, Chem.
dissolution of drugs, Rohm and Haas Research laboratories, Spring Pharm. Bull. 28 (1980) 1056–1061.
house, PA, USA.

Anda mungkin juga menyukai