Pharmaceutical development

Chapter 16 Pharmaceutical development





Preformulation studies


As a starting point to developing dosage forms, various physical and chemical properties of the drug substance need to be documented. These investigations are termed preformulation studies. Most synthetic drugs are either weak bases (~75%) or weak acids (~20%), and will generally need to be formulated as salts. Salts of a range of acceptable conjugate acids or bases therefore need to be prepared and tested. Intravenous formulations of relatively insoluble compounds may need to include non-aqueous solvents or emulsifying agents, and the compatibility of the test substance with these additives, as well as with the commonly used excipients that are included in tablets or capsules, will need to be assessed.


The main components of preformulation studies are:



Theoretical treatments of these molecular properties, and laboratory methods for measuring them, which are beyond the scope of this book, are described in textbooks such as Burger and Abraham (2003), Aulton (2007) and Allen (2008). Here we consider some issues that commonly arise in drug development.



Solubility and dissolution rate


The question of solubility, already emphasized in Chapters 9 and 10, is particularly important in relation to the development of pharmaceutical formulation. It is measured by standard laboratory procedures and involves determining the concentration of the compound in solution after equilibration – usually after several hours of stirring – with the pure solid. In general, compounds whose aqueous solubility exceeds 10 mg/mL present no problems with formulation (Kaplan, 1972). Compounds with lower solubility are likely to require conversion to salts, or the addition of non-aqueous solvents, in order to achieve satisfactory oral absorption. Because the extreme pH values needed to induce ionization of very weak acids or bases are likely to cause tissue damage, the inclusion of a miscible solvent of relatively low polarity, such as 20% propylene glycol or some other biocompatible solubilizing agent (see below), will often be required for preparing injectable formulations. Complications may arise with oral formulations if the solubility is highly dependent on pH, because of the large pH difference between the stomach and the small intestine. Gastric pH can range from near neutrality in the absence of any food stimulus to acid secretion, to pH 1–2, whereas the intestinal pH is around 8. Basic substances that dissolve readily in the stomach can therefore precipitate in the intestine and fail to be absorbed. Compounds that can exist in more than one crystal form can also show complex behaviours. The different lattice energies of molecules in the different crystal forms mean that the intrinsic solubility of the compound is also different. Different crystal forms may correspond to different hydration states of the compound, so that a solution prepared from the unhydrated solid may gradually precipitate as hydrated crystals. Selecting the best salt form to avoid complications of this sort is an important aspect of preformulation studies.


Compounds that have low intrinsic solubility in aqueous media can often be brought into solution by the addition of a water-miscible solubilizing agent, such as polysorbates, ethanol or polyethylene glycol (PEG). Preformulation studies may, therefore, include the investigation of various solubilizing agents, such as methylcellulose or cyclodextrin, which are known to be relatively free of adverse effects in man.


As well as intrinsic solubility, dissolution rate is important in determining the rate of absorption of an oral drug. The process of dissolution involves two steps: (a) the transfer of molecules from the solid to the immediately adjacent layer of fluid, known as the boundary layer; and (b) escape from the boundary layer into the main reservoir of fluid, which is known as the bulk phase and is assumed to be well stirred so that its concentration is uniform. Step (a) is invariably much faster than step (b), so the boundary layer quickly reaches saturation. The overall rate of dissolution is limited by step (b), and depends on the intrinsic solubility of the compound, the diffusion coefficient of the solute, the surface area of the boundary layer, and the geometry of the path leading from the boundary layer to the bulk phase.


In practice, dissolution rates depend mainly on:



In pharmaceutical development, dissolution rates are often manipulated intentionally by including different polymers, such as methylcellulose into tablets or capsules, to produce ‘slow-release’ formulations of drugs such as diclofenac, allowing once-daily dosage despite the drug’s short plasma half-life.





Routes of administration and dosage forms


With the knowledge of the characteristics of the drug substance from preformulation studies, the administration route has to be selected and the substance to be included into a dosage form which is effective and convenient for the patient. The preferred dosage form for therapeutic agents is almost always an oral tablet or capsule, either taken as needed to control symptoms, or taken regularly once or several times a day. However, there are many alternatives, and Table 16.1 lists some of the main ones. An important consideration is whether it is desirable to achieve systemic exposure (i.e. distribution of the drug to all organs via the bloodstream) or selective local exposure (e.g. to the lungs, skin or rectum) by applying the drug topically. In most cases systemic exposure will be required, and an oral capsule or tablet will be the desired final dosage form. Even so, an intravenous formulation will normally be required for use in safety pharmacology, toxicology and pharmacokinetic studies in man.



The main routes of administration for drugs acting systemically, apart from oral and injectable formulations, are transdermal, intranasal and oromucosal. Rectal, vaginal and pulmonary routes are also used in some cases, though these are used mainly for drugs that act locally.


Transdermal administration of drugs formulated as small adhesive skin patches has considerable market appeal, even though such preparations are much more expensive than conventional formulations. To be administered in this way, drugs must be highly potent, lipid soluble and of low molecular weight. Examples of commercially available patch formulations include nitroglycerin, scopolamine, fentanyl, nicotine, testosterone, estradiol and ketoprofen, and several others in development. The main limitation is the low permeability of the skin to most drugs and the small area covered, which mean that dosage is limited to a few milligrams per day, so only very potent drugs can be given systemically via this route of administration. Variations in skin thickness affect the rate of penetration, and the occurrence of local skin reactions is also a problem with some drugs. Various penetration enhancers, mainly surfactant compounds of the sort discussed above, are used to improve transdermal absorption. The transfer rate can be greatly enhanced by applying a small and painless electric current (about 0.5 mA/cm2), and this is effective in achieving transfer of peptides (e.g. calcitonin) and even insulin through the skin. It also offers promise as a route of administration of oligonucleotides in gene therapy applications (see Chapter 12). These procedures are used for administration of nucleotides and in gene therapy and are being used experimentally in the clinic, but are not yet available as commercial products for routine clinical use. Ultrasonic irradiation is also under investigation as a means of facilitating transdermal delivery. These procedures would also allow the administration to be controlled according to need. Intranasal drug administration (Illium, 2002, 2003) is another route that has been used successfully for a few drugs. The nasal epithelium is much more permeable than skin and allows the transfer of peptide drugs as well as low-molecular-weight substances. Commercially available preparations have been developed for peptide hormones, such as vasopressin analogues, calcitonin, buserelin and others, as well as for conventional drugs such as triptans, opioids, etc. The main disadvantages are that substances are quickly cleared from the nasal epithelium by ciliary action, as well as being metabolized, and the epithelial permeability is not sufficient to allow most proteins to be given in this way. Ciliary clearance can be reduced by the use of gel formulations, and surfactant permeability enhancers can be used to improve the penetration of larger molecules. The possibility of administering insulin, growth factors or vaccines by this route is the subject of active research efforts. Some studies have suggested (see Illium, 2003) that substances absorbed through the nasal epithelium reach the brain more rapidly than if they are given intravenously, possibly bypassing the blood–brain barrier by reaching the CNS directly via the nerves to the olfactory bulb.


Oromucosal delivery (Madhav et al., 2009) and especially utilizing the buccal and sublingual mucosa as the absorption site is a drug delivery route which promotes rapid absorption and almost immediate pharmacological effect. The sublingual mucosa, especially, is highly vascularized and this route bypasses the gastrointestinal tract and, thus, the first-pass metabolism. However, not all drugs can be efficiently absorbed through the oral mucosa because of physicochemical properties or enzymatic breakdown of the drug and the amount of drug that could be absorbed is limited to a few milligrams. The drug has to be soluble, stable and able to easily permeate the mucosal barrier at the administration site. Also some formulation aspects have to be taken into consideration, using a tablet formulation for a rapid onset of effect a prerequisite is a fast disintegration and dissolution in the oral cavity resulting in an optimal exposure of active substance to the small volume of dissolving fluids. Swallowing of the drug could be a potential problem, but can be minimized by using technologies using mucoadhesive components (Bredenberg et al., 2003; Brown and Hamed, 2007). An optimized formulation and using this administration route has the potential for very fast absorption (Kroboth et al., 1995) and obtaining peak blood levels within 10 to 15 minutes. It is thereby potentially a more comfortable and convenient alternative to the intravenous route of administration.



Formulation


Formulation of an active substance into a dosage form, where there are no special requirements for modified release, involves a good deal of engineering. As already mentioned, the ‘ideal’ drug substance, intended for use as an oral preparation, has the following characteristics:



If these conditions are met the formulation of oral formulations presents no special problems, but they rarely are, and it falls to the pharmaceutical development group to develop formulations that successfully overcome the shortcomings of the compound. Firstly, the drug substance must be dried and converted to a powder form that can be precisely dispensed. Further, depending on the dosage form and the desired properties, other substances called excipients, have to be included.


As mentioned above, tablets and capsules are the most common dosage form (probably due to a combination of convenience for the patient and a cost-effective manufacturing process, at least for conventional tablets). Even for a tablet without special requirements for its drug release profile a range of excipients are needed, e.g. the tablet should be sufficiently strong to withstand handling but also has to disintegrate after intake in order to release the drug.


Inert diluents or fillers, such as lactose or starch, are added to produce tablets of a manageable size (generally 50–500 mg). The filler should have good compactability and flow properties, be non-hygroscopic and have acceptable taste. Binders, such as cellulose and other polymeric materials, may be needed to assist compaction into a solid tablet that will not crumble. A binder could be added both in dry form and in the granulation liquid depending on the manufacturing process. Disintegrating agents, such as starch and cellulose, ensure that the tablet disintegrates rapidly in the gastrointestinal tract. For a very fast disintegration, so-called super disintegrants acting to produce extensive swelling could be used. Slippery, non-adherent materials, such as magnesium stearate, may be needed to ensure that the powder runs smoothly in the tablet machine, by reduction of friction between particles and between particles and parts of the machine in contact. The tablet may need to be coated with cellulose or sugar to disguise its taste.


Capsule formulations are often used for initial clinical trials, as they are generally simpler to develop than compressed tablets, but are less suitable for controlled-release formulations (see below). A two-piece gelatin capsule can be used to contain drugs also in semisolid or liquid form. Other advantages are that capsules are easy to swallow and provide effective taste-masking. Drug dissolution rate could, with a fast dissolving capsule, be increased compared to a conventional tablet resulting in improved drug absorption, especially for poorly soluble substances.


The choice of excipients and the manufacturing process is very much dependent on the characteristics of drug substance and the desired properties of the dosage form. Drug release profile is just one aspect, while the homogeneity or uniformity of content is another. If the drug substance is very potent and cohesive then mixing a small amount of drug with a high amount of filler could lead to a product with low homogeneity. This problem is most severe if the drug particles are micronized to improve the dissolution rate. Then it is important to also choose the most appropriate manufacturing process, such as dry mixing with larger filler paticles, so-called ordered or interactive mixing, wet granulation or drug coating of placebo tablets. Different processes used in drug development and manufacturing are described in textbooks such as Aulton (2007).


As mentioned in the preformulation section the shelf-life of the drug product should preferably be at least 3 years. The presence of moisture is the main contributor to the degradation of the drug substance. Tablets normally have a longer shelf-life than other formulations such as oral and parental liquids since they are a dry dosage form. However, it is important to choose excipients which are not hygroscopic since small amounts of moisture could decrease the stability of the drug. Therefore, selection of the packing material is also an important aspect to take into consideration: for example, moisture-sensitive freeze-dried tablets are often packed in almost impenetrable aluminium blisters.


In reality, formulation development has to take into account not only the properties of the drug substance, but also the desired delivery system and the form of the final product. In developing a new nitrate preparation for treating angina, for example, the preferred delivery system might be a skin patch to be packaged in a foil sachet, or a nasal spray to be packaged as a push-button aerosol can. Although a simple oral preparation may be feasible, the medical need and patient convenience might require the development of different dosage forms, and the development plan would have to be directed towards this more demanding task.

Stay updated, free articles. Join our Telegram channel

Oct 1, 2016 | Posted by in GENERAL SURGERY | Comments Off on Pharmaceutical development

Full access? Get Clinical Tree

Get Clinical Tree app for offline access