Abstract
This review article is a current survey of excipients used in approved injectable products. Information provided includes concentration ranges, function, frequency of use, and role in dosage form. This article is an update of a paper published more than a decade ago (reference 11). Since then many new products have been approved. Safety concerning excipients has evolved as the scientific community continues to learn about their usage. New excipients are being used in early phases of clinical trials to support novel therapeutic entities like RNAi, aptamers, anti-sense, fusion proteins, monoclonal antibodies, and variant scaffolds. Because these excipients are not inert, various pharmacopoeias are responding with monographs or informational chapters addressing excipient functionality. The final sections of this article discuss new excipients, serving specific needs that traditional excipients are unable to provide and for which safety studies are necessary to support a novel excipient for marketing applications.
LAY ABSTRACT: Excipients are added to parenteral dosage forms to serve a variety of functions including stabilization and as vehicles. This review article is a survey of excipients used in approved injectable products. Information provided includes excipient concentrations, functional roles, and frequency of use. This article is an update of an article originally published over a decade ago. Since then new products have been approved and safety concerns have evolved as the scientific community has learned about the usage of excipients. In addition, new excipients are being used in early phases of clinical trials to support novel therapeutic entities such as RNAi, aptamers, anti-sense, fusion proteins, monoclonal antibodies, and variant scaffolds. Because these excipients are not inert, various pharmacopoeias are responding with monographs or informational chapters addressing excipient functionality. The final sections of this article discuss new excipients, serving specific needs that traditional excipients are unable to provide and for which safety studies are necessary to support a novel excipient for marketing applications.
I. Introduction
Excipients are typically the major components in a drug product. The active drug molecule comprises only a small percentage of the total composition. Excipients are traditionally referred to as inactive or inert ingredients to distinguish them from active pharmaceutical ingredients. Pharmaceutical excipients or additives are compounds added to the finished drug products to serve a specific function. They are added to increase bulk, aid manufacturing, improve stability, enhance drug delivery and targeting, and modify drug safety or pharmacokinetic profile. Ingredients that are used during drug product manufacturing but may not be present in the finished drug product are also considered excipients (examples include water for lyophilized products, and inert gases in the head space of containers) (1). In recent years the “functionality” of excipients in a dosage form (similar to pharmacological activity of an active pharmaceutical ingredient or drug substance) has been recognized by various pharmacopoeias (2). Many excipient monographs do not address this aspect of excipient functionality or its control.
Recent emphasis on quality by design (QbD) has led pharmaceutical companies to better understand how excipient functionality and performance influence a drug product (3). It has been recognized for many years that pharmacopoeial excipient monographs do not address excipient functionality per se. To overcome this deficiency, the USP general chapter on the functionality of excipients has been proposed; the European Pharmacopeia (Ph. Eur.) has introduced a non-mandatory Functionality Related Characteristics section in monographs of some excipients. A QbD approach will enable better understanding of excipient variability and its potential impact on product quality. International Conference on Harmonization (ICH) Q8 and QbD acknowledge excipient variability. Design space studies of excipient variability and functional performance are needed to assure that excipient standards accurately reflect excipient quality (4).
Excipients may not be as inert as the term inactive suggests. Due to safety issues, several countries have restrictions on the type or amount of excipient that can be included in the formulation of parenteral drug product. For example, in Japan, the U.S., and the E.U., amino mercuric chloride or thimerosal use is prohibited, despite the presence of these excipients in products in other regions.
As defined in Ph. Eur. and the British Pharmacopeia (BP), “parenteral preparations are sterile preparations intended for administration by injection, infusion, or implantation into the human or animal body.” In the present article, only sterile preparations for administration by injection or infusion into the human body will be surveyed (5, 6). Injectable products require a unique formulation strategy. The formulated product must be sterile, pyrogen-free, and, in the case of solution, free of particulate matter. No coloring agent may be added solely for the purpose of coloring the parenteral preparation. The formulation should preferably be isotonic, and depending on the route of administration, certain excipients are not allowed. The injected drug by-passes natural defense barriers; hence, for any given drug, the risk of an adverse event may be greater or the effects difficult to reverse if administered as an injection rather than a non-parenteral route. Sterility requirements demand that an excipient is able to withstand terminal sterilization or aseptic processing. These factors limit the choice of excipients available.
Knowledge of excipients that have been deemed safe by the Food and Drug Administration (FDA) or are already present in a marketed product provides increased assurance to a formulator that these excipients will probably be safe for a new drug product. But safety is not guaranteed when excipients are combined with other additives and/or with a new drug molecule creating unforeseen potentiation or synergistic toxic effects. Regulatory bodies may favorably view an excipient previously approved in an injectable dosage form, and will require less safety data. A new additive in a formulated product always requires additional studies, adding to the cost and timeline of product development. Importantly, inclusion of an excipient in the GRAS (Generally Recognized as Safe) list or pharmacopoeia does not mean that the excipient has been deemed safe by the FDA for use in parenteral products.
In Japan if a drug product contains an excipient with no precedence of use, the quality and safety attributes of the excipient must be evaluated by the Subcommittee on Pharmaceutical Excipients of the Central Pharmaceutical Affairs Council concurrently with the evaluation of the drug product application (7). Precedence of use means the excipient has been used in a drug product in Japan which is administered via the same route and in a dose level equal to or greater than the excipient in question in the new application.
This article is a comprehensive review of excipients that have been included in injectable products marketed in the USA, Europe, and Japan. A literature review indicates that few articles have been published dealing specifically with selection of parenteral excipients (8–17). This review does not cover excipients included in other sterile dosage forms not administered parenterally, such as solution for irrigation, ophthalmic, or otic drops and ointments.
Several sources were used to summarize the information compiled in this paper (9–22). Formulation information on commercially available injectable products was entered in a worksheet. Tables presented have been condensed from this worksheet. Each table has been categorized based on an excipient's primary function in a formulation. For example, citrates are classified as buffers, not as chelating agents. Ascorbates are categorized as antioxidants, although they can also serve as buffers. This classification system minimizes redundancy and provides a reader-friendly format. Concentration of excipients is listed as percent weight by volume (w/v) or volume by volume (v/v). If the product was listed as lyophilized or powder, percentages were derived based on reconstitution volume commonly used. The tables list the concentration range and examples of products containing the excipient, especially those using an extremely low or high concentration.
II. Types of Excipients
II.A. Solvents and Co-Solvents
Table I lists solvents and co-solvents used in parenteral products. Water for injection (WFI) is the most common solvent, but there are differences in the pharmacopeial specifications. Ph. Eur. does not recognize WFI produced by reverse osmosis even though the finished excipient will meet all specifications (23, 24). WFI can be combined or substituted with a co-solvent to improve drug solubility or stability (25, 26). Dielectric constant and solubility parameters are among the most commonly used polarity indices for solvent blending (27, 28). Ethanol and propylene glycol are used alone or in combination with other solvents in more than 50% of parenteral co-solvent systems. Surprisingly, propylene glycol is used more often than polyethylene glycols (PEGs) despite its higher myotoxicity and hemolyzing effects (29–32). A toxicity review of commonly used parenteral co-solvents is summarized in an article by Mottu (33). The hemolytic potential of co-solvents is as follows (31): dimethyl acetamide < PEG 400 < Ethanol < propylene glycol < dimethylsulfoxide.
Solvents and Co-Solvents
It is possible that the presence of residual peroxide from bleaching of PEG or generation of peroxides in PEG may result in degradation of drug in a co-solvent system. It is important to use unbleached and/or peroxide-free PEGs in a formulation.
Oils such as safflower and soybean are used in total parenteral nutrition products where they serve as a fat source and as carriers for fat-soluble vitamins. The USP requirements (34) for injectable oils are as follows:
Fixed oils (of vegetable origin)
-
Saponification value (185–200)
-
Iodine Number (79–141); the Japanese Pharmacopoeia (JP) recommends value between 79–137
-
Test for unsaponifiable matter
-
Test for free fatty acid
-
Solid paraffin test at 10 °C
-
Acid value not more than 0.56 (JP only) (35)
Synthetic mono- and di-glycerides of fatty acids (which are liquid and remain so when cooled to 10 °C)
-
Iodine Number (<140)
-
Solid paraffin test at 10 °C
Oils are used to dissolve drugs with low aqueous solubility and provide a mechanism for slow drug release over a long period of time. Deterioration of fixed oils, leading to rancidity and production of free fatty acids, must be avoided in injectable products. Fixed oils or fatty acid esters must not contain mineral oil or paraffin that the body cannot metabolize.
II.B. Polymeric and Surface Active Compounds
Table II includes a broad category of excipients whose function in a formulation could be
-
To impart viscosity or act as suspending agents: carboxy methyl cellulose, sodium carboxy methyl cellulose, acacia, Povidone, hydrolyzed gelatin, sorbitol
-
To act as solubilizing, wetting or emulsifying agents: Cremophore EL, sodium desoxycholate, Polysorbate 20 or 80, PEG 40 Castor oil, PEG 60 castor oil, sodium dodecyl sulfate, lecithin or egg yolk phospholipid
-
To form gels, as when aluminum monostearate is added to fixed oil, forming a viscous or gel-like suspension medium
Solubilizing, Wetting, Suspending, Emulsifying, or Thickening Agents
Polysorbate 80 is the most common and versatile solubilizing, wetting, and emulsifying agent. Again, one must be concerned with the level of residual peroxides present in polysorbates and protecting them from air to prevent further oxidation (36). Polysorbate 80 is a polyoxyethylene sorbitan ester of oleic acid (unsaturated fatty acid) while polyoxyethylene Polysorbate 20 is a sorbitan ester of lauric acid (saturated fatty acid). Thus, there could be stability differences in a drug product formulated with Polysorbate 80 versus Polysorbate 20. One example is Neupogen® which, when exposed to a high concentration of Polysorbate 20, exhibited substantially less oxidation than when exposed to a similar concentration of Polysorbate 80 (37).
II.C. Chelating Agents
A limited number of chelating agents are used in parenteral products (Table III). They serve to complex heavy metals and therefore improve the efficacy of antioxidants or preservatives. Citric acid, tartaric acid, and some amino acids act as chelating agents. There has been misunderstanding that ethylenediaminetetraacetic acid (EDTA) (as calcium or sodium salt) has not been used in an approved injectable product in Japan. Some currently marketed drug products do contain calcium disodium EDTA in Japan. This excipient is listed as an official excipient in Japan (refer to section II.H. for details). One possible advantage of calcium EDTA over tetrasodium salt is that it does contribute little sodium and does not chelate calcium from the blood.
Chelating Agents
A complexing agent should not be used in metallo-protein formulations, where protein sub-units are held by the metal (38). EDTA, in rare instances, can increase oxidation rate due to binding of EDTA-metal complex to protein, resulting in site-specific generation of radicals (39).
II.D. Antioxidants
Antioxidants are used to prevent oxidation of active substances and excipients in the finished product. Antioxidant can be classified into three main types:
-
True antioxidants: those that act via a chain termination mechanism by reacting with free radicals (e.g. butylated hydroxytoluene)
-
Reducing agents: those that have a lower redox potential than the drug and are preferentially oxidized (e.g., ascorbic acid). Consequently, they can be consumed during the shelf life of the product.
-
Antioxidant synergists: those that enhance the effect of antioxidants (e.g., EDTA)
Table IV summarizes antioxidants, their frequency of use, concentration range, and examples of products containing them. Sulfite, bisulfite, and metabisulfite constitute the majority of antioxidants used in parenteral products despite several reports of incompatibility and toxicity (40, 41). Butylated hydroxy anisole, butylated hydroxy toluene, alpha tocopherol, and propyl gallate are primarily used in semi/non-aqueous vehicles because of their low aqueous solubility (42). Ascorbic acid/sodium ascorbate may serve as an antioxidant, buffer, and chelating agent in the same formulation. Some amino acids (cysteine) function as effective antioxidants.
Antioxidants and Reducing Agents
The Committee for Medicinal Products for Human Use (CHMP, which replaced the Committee for Proprietary Medicinal Products) guideline requires a full explanation and justification for including antioxidants in a formulation (43, 44). It further states that antioxidants should be included in a formulation only if it has been demonstrated that their use cannot be avoided. Thus, it is imperative to try an inert gas such as nitrogen or argon in the container head space to prevent oxidation. If an antioxidant must be included, its concentration must be justified in terms of efficacy and safety. Antioxidants, like sulfites and metabisulfites, are especially undesirable.
Some antioxidants possess antimicrobial properties. Propyl gallate and butylated hydroxy anisole are somewhat effective against bacteria. Butylated hydroxy toluene has demonstrated some anti-viral activity. Compatibility of antioxidants with the drug, packaging system, and the body should be studied carefully. For example, tocopherols may be absorbed on plastics; ascorbic acid is incompatible with alkalis, heavy metals, and oxidizing materials like phenylephrine and sodium nitrite; propyl gallate forms complexes with metal ions like sodium, potassium, and iron.
II.E. Preservatives
Benzyl alcohol is the most common antimicrobial preservative present in parenteral formulations (Table V). This observation is consistent with other surveys (12, 45). Parabens are the second most common preservatives. Surprisingly, thimerosal is also common, especially in vaccines, even though some individuals are sensitive to mercurics. Several preservatives can volatilize easily (benzyl alcohol, phenol) and should not be used for a lyophilized dosage form. Chlorocresol is purported to be a good preservative for parenterals, but our survey did not find any examples of commercial products containing chlorocresol. The British Pharmaceutical Codex and Martindale lists chlorocresol as a preservative to be used in multidose aqueous injections at a concentration of 0.1%, but no examples of injectable products were provided (46, 47).
Antimicrobial Preservatives
Antimicrobial preservatives are allowed in multi-dose injections to prevent growth of microorganisms that may accidentally enter the container during withdrawal of the dose. In the U.S., preservatives are discouraged from being used in single-dose injections, while Ph. Eur. and BP allow aqueous preparations manufactured using aseptic techniques to contain suitable preservatives. Ph. Eur. and BP prohibit antimicrobials for single-dose injections where the dose volume is greater than 15 mL or if the drug product is to be injected intracisternally, or via any route giving access to the cerebrospinal fluid (e.g., retro-ocular). It should be emphasized that preservatives should never be used as a substitute for inadequate cGMP (current good manufacturing practice). Toxicity is the primary reason for minimizing use of antimicrobial preservatives. For example, many individuals are allergic to mercury preservatives and benzyl alcohol is contraindicated in children under the age of 2 years. The USP has placed some restrictions on the maximum concentration of preservatives allowed in a formulation to address toxicity and allergic reactions (Table VI). The World Health Organization has set an estimated total acceptable daily intake for sorbate (as acid, calcium, potassium, and sodium salts) as not more than 25 mg/kg body weight. Recently, concerns have been raised on the safety of parabens in pediatric formulations, based primarily on reports by one Japanese laboratory between 2001 and 2004 indicating effects on reproductive apparatus of juvenile male rats given propyl (48) or butyl paraben (49), but lack of effects for methyl and ethyl parabens (50), though a vast majority of toxicological data suggests otherwise (51). Until a comprehensive assessment is performed, formulators should consider the current view of regional regulatory agencies (e.g., AFSSAPS, Scandinavian, et al.) and may investigate other preservative options for pediatric products.
Maximum Permissible Amount of Preservatives and Antioxidants
The efficacy of a preservative should be assessed during product development using antimicrobial preservative effectiveness testing (PET) (52–54). A preserved aqueous parenteral product can be used up to a maximum of 28 days after the container has been opened (55). This time period must be qualified by performing PET on the finished product in the final package. Conversely, un-preserved product should preferably be used immediately or within a short period of time upon opening, reconstitution, or dilution.
Similar to antioxidants, addition of preservatives in parenteral products requires justification. Their use should be avoided, particularly for pediatric products, but if used, their concentration should be as low as possible (44).
II.F. Buffers
Buffers are added to a formulation to adjust and stabilize pH and optimize drug solubility and stability. For parenteral preparations, it is desirable that the product pH be close to physiologic pH. Selection of a buffer concentration (which contributes to the ionic strength of the formulation) and a buffer species is important. For example, citrate buffers in the range of 5–15 mM are typically used in formulations but increasing the buffer concentration to >50 mM will result in excessive pain on sub-cutaneous injection and toxic effects due to chelation of calcium in the blood.
Buffers have maximum buffer capacities near their pKa. Products which may be subjected to excessive temperature fluctuations during processing (steam sterilization, freeze-thaw or lyophilization), buffers with a small ΔpKa/°C should be selected. For example, Tris, whose ΔpKa/°C is large (−0.028/°C), the pH of buffer made at 25 °C will change from 7.1 to 5.0 at 100 °C. This may dramatically alter stability or solubility of a drug. Similarly, the best buffers for a lyophilized product may be those which show the least pH change upon cooling, do not crystallize out, and remain in the amorphous state, protecting the drug. For example, replacing succinate with glycolate buffer improves the stability of lyophilized interferon-γ (56). During the lyophilization of mannitol containing succinate buffer at pH 5, monosodium succinate crystallizes, reducing the pH and resulting in the unfolding of interferon-γ. This pH shift is not seen with glycolate buffer.
Table VII lists buffers and chemicals used for pH adjustment and maintenance of the drug product pH range. Phosphate, citrate, and acetate are the most common buffers used in parenteral products. Mono- and di-ethanolamines are added to adjust pH and form corresponding salts. Hydrogen bromide, sulfuric acid, benzene sulfonic acid, and methane sulfonic acids are added to drugs that are salts of bromide (Scopolamine HBr, Hyoscine HBr), sulfate (Nebcin, Tobramycin sulfate), besylate (Tracrium Injection, Atracurium besylate), or mesylate (DHE 45 Injection, dihydroergotamine mesylate). Glucono delta lactone is used to adjust the pH of Quinidine gluconate. Benzoate buffer, at a concentration of 5%, is used in Valium Injection. Citrates are common buffers that serve a dual role as chelating agent. Amino acids—histidine, arginine, and glycine—function as buffers and stabilize proteins and peptide formulations. These amino acids are also used as lyo-additives and may prevent cold denaturation. Lactate and tartrate are occasionally used as buffer systems. Acetates are good buffers at low pH, but they are not frequently used for lyophilization because of the potential sublimation of acetates.
Buffers and pH Adjusting Agents
II.G. Bulking Agents, Protectants, and Tonicity Adjusters
Table VIII lists additives used as bulking or lyo/cryo-protective agents and to modify osmolality. Dextrose and sodium chloride are the most commonly used tonicity adjusters. Some amino acids such as glycine, alanine, histidine, imidazole, arginine, asparagine, and aspartic acid are used as bulking agents for lyophilization and may also serve as stabilizers and/or as buffers. Monosaccharides (dextrose, glucose, maltose, lactose), disaccharides (sucrose, trehalose), polyhydric alcohols (inositol, mannitol, sorbitol), glycols (PEG 3350), Povidone (polyvinylpyrrolidone, PVP), and proteins (albumin, gelatin) are commonly used as lyo-additives. Hydroxyethyl starch (hetastarch) and pentastarch, currently used as plasma expanders in commercial injectable products like Hespan and Pentaspan, are being evaluated as protectants during freeze-drying of proteins.
Bulking Agents, Protectants, and Tonicity Adjusters
PVP has been used in injectable products (except in Japan) as a solubilizing agent, protectant, and bulking agent. Only pyrogen-free grade, with low molecular weight (K value less than 18) should be used in parenteral products to allow for rapid renal elimination. PVP not only solubilizes drugs like rifampicin, but it also reduces local toxicity, as seen in the case of Oxytetracycline Injection.
Many proteins can be stabilized in a lyophilized state if the stabilizer and protein do not phase separate during freezing or if the stabilizer does not crystallize. In the case of Neupogen® (GCSF), the original formulation was modified by replacing mannitol with sorbitol to prevent loss of activity of the liquid formulation if accidentally frozen (37). Mannitol crystallizes if the solution freezes, while sorbitol remains in an amorphous state, protecting GCSF. A recent report suggests that sorbitol (and even trehalose) can also crystallize under certain conditions (57). Similarly, it is useful that the drug remains dispersed in the stabilizer upon freezing of the solution. Consequently, Cefoxitin, a cephalosporin, is more stable when freeze-dried with sucrose rather than with trehalose, although the glass transition temperature and structural relaxation time is much greater for trehalose than sucrose (58). Fourier transform infrared spectroscopy data indicated that the trehalose-cefoxitin system phase-separated into two nearly pure components resulting in no protection (stability). Similarly, dextran was not found to be as useful a cryoprotectant for protein as sucrose because dextran and protein underwent phase segregation as the solution began to freeze. The mechanism of cryoprotection in the solution has been explained by the preferential exclusion hypothesis (59).
Trehalose is a non-reducing disaccharide composed of two D-glucose monomers. It is found in several animals that can withstand dehydration and therefore had been suggested to stabilize drugs that undergo denaturation during spray or freeze-drying (60). Herceptin® (Trastuzumab) is a recombinant DNA-derived monoclonal antibody (MAb) used for treating metastatic breast cancer. The MAb has been stabilized in the lyophilized formulation using α,α-trehalose dihydrate. Trehalose has also been used as a cryoprotectant to prevent liposomal aggregation and leakage. In the dried state, carbohydrates like trehalose and inositol exert their protective effect by acting as a water substitute (61).
Additives may be included in a formulation to adjust the specific gravity. This is important for drugs which, upon administration, may come in contact with cerebrospinal fluid (CSF). CSF has a specific gravity of 1.0059 at 37 °C. Solutions having the same specific gravity as that of CSF are termed isobaric, while those solutions which have specific gravity greater than that of CSF are called hyperbaric. Upon administration of a hyperbaric solution in the spinal cord, the injected solution will settle and affect spinal nerves at the end of the spinal cord. For example, Dibucaine hydrochloride solution (Nupercaine® 1:200) is isobaric, while Nupercaine® 1:500 is hypobaric (specific gravity of 1.0036 at 37 °C). Nupercaine® heavy solution is made hyperbaric by addition of 5% dextrose solution. This solution will block (anesthetize) the lower spinal nerves as it will settle in the lower spinal cord.
II.H. Special Additives
These additives have been included in pharmaceutical formulations to serve specific functions (Table IX). The following is a summary of some special additives along with their intended use:
-
Calcium gluconate injection (American Regent) is a saturated solution of 10 w/v; calcium d-saccharate tetrahydrate 0.46% w/v is added to prevent crystallization during temperature fluctuations.
-
Cipro IV® (ciprofloxacin, Bayer) contains lactic acid as a solubilizing agent for the antibiotic.
-
Premarin Injection® (conjugated estrogens, Wyeth-Ayerst Labs) is a lyophilized product that contains simethicone to prevent formation of foam during reconstitution.
-
Dexamethasone acetate (Dalalone DP, Forest, Decadron-LA, Merck) and dexamethasone sodium phosphate (Merck) are available as a suspension or as a solution. These dexamethasone formulations contain creatine or creatinine as additives.
-
Adriamycin RDF® (doxorubicin hydrochloride, Pharmacia-Upjohn) contains methyl paraben, 0.2 mg/mL to increase dissolution (62).
-
Ergotrate maleate (ergonovine maleate, Lilly) contains 0.1% ethyl lactate as a solubilizing agent.
-
Estradurin Injection® (polyestradiol phosphate, Wyeth-Ayerst Labs) uses Niacinamide (12.5 mg/mL) as a solubilizing agent. Hydeltrasol® also contains niacinamide. The concept of hydrotropic agents to increase water solubility has been tried on several compounds, including proteins (63, 64).
-
Aluminum, in the form of aluminum hydroxide, aluminum phosphate, or aluminum potassium sulfate, is used as adjuvant in vaccine formulations to elicit an increased immunogenic response.
-
Lupron Depot Injection® is lyophilized microspheres of gelatin and glycolic-lactic acid for intra-muscular injection. Nutropin Depot consists of polylactate-glycolate microspheres.
-
Sodium caprylate (sodium octoate) has antifungal properties, but is also used to improve stability of albumin solution against the effects of heat. Albumin solution can be heat-pasteurized by heating at 60 °C for 10 h in the presence of sodium caprylate. Acetyl tryptophanate sodium is also added to albumin formulations.
-
Meglumine (N-methylglucamine) is used to form in-situ salt. For example, diatrizoic acid, an X-ray contrast agent, is more stable when autoclaved as meglumine salt than as sodium salt (65). Meglumine is also added to Magnevist®, a magnetic resonance contrast agent.
-
Tri-n-butyl phosphate is present as an excipient in human immune globulin solution (Venoglobulin®). Its exact function in the formulation is not known, but it may serve as a scavenging agent.
-
von Willebrand factor is used to stabilize recombinant antihemophilic factor (Bioclate®).
-
Maltose serves as a tonicity adjuster and stabilizer in immune globulin formulation (Gamimune N®).
-
Epsilon amino caproic acid (6-amino hexanoic acid) is used as a stabilizer in anistreplase (Eminase Injection®).
-
Zinc and protamine have been added to insulin to form complexes and control the duration of action.
Special Additives
The FDA has published the “Inactive Ingredient Guide,” which lists excipients in alphabetical order (20). The Inactive Ingredient Guide is reasonably comprehensive but does not include several excipients used in recently approved drug products. Each ingredient is followed by the route of administration, and in some cases, the concentration range used in the approved drug product. This list does not provide the commercial product name(s) corresponding to each excipient. Table X is a summary of all parenteral excipients included in the “Inactive Ingredient Guide.” A majority of these parenteral excipients are covered in our survey (as highlighted in the table). There are excipients, particularly those in radio-labeled products, that are not included and some on the list have questionable function as excipients (e.g., insulin, denatured alcohol). Overall, there is a 70% overlap between our survey and the FDA inactive ingredient list for parenterals.
Excipients Listed in the FDA Inactive Ingredient Guide*
Similarly, in Japan the Japanese Pharmaceutical Excipients Directory (JPED) is published by the Japanese Pharmaceutical Excipients Council with the cooperation and guidance of the Ministry of Health, Labour and Welfare (66). This directory divides excipients into
-
Official Excipients: These include excipients that have been recognized in the JP, Japanese Pharmaceutical Codex (JPC), and Japanese Pharmaceutical Excipients (JPE), and for which testing methods and standards have been determined. Table XI summarizes official excipients that have been used in parenteral products and are described in JPE.
Japanese Pharmaceutical Excipients
-
2. Non-Official Excipients: These excipients are not listed in JP, JPC, or JPE but still have precedence of use in pharmaceutical products. The non-official excipients, which have been used in parenteral products, are listed in Table XII.
List of Excipients in JPED 2007 and Maximum Amount Specified per Route
The JPED lists the excipient, route of administration, and maximum amount or concentration that has been approved. If the excipient falls within these limits it is considered to be “precedented” and no additional data is needed. If it falls outside this range, additional safety information is needed (experimental or published literature). These unprecedented or novel excipients may have to be placed on stability. If the excipient is listed in JP, JPE, or JPC then it must meet the specifications listed in the monograph. If the excipient is not listed in any of the above three books, then in-house specifications must be established. In-house specifications can be based on USP, Ph. Eur., or other pharmacopoeial standards if the excipient is listed there. Proposed in-house specifications are in included in the drug product submission where the excipient is reviewed by the Pharmaceuticals and Medical Devices Agency.
III. Regulatory Perspective
Based on available safety testing information, the International Pharmaceutical Excipients Council (IPEC) has classified excipients into four classes (67):
-
New chemical excipients: These excipients require a full safety evaluation program. It is estimated that the cost of safety studies for a new chemical excipient is about $35 million over 4–5 years. E.U. directive 75/318/EEC states that new chemical excipients will be treated in the same way as new actives. In the U.S. a new excipient requires a Drug Master File (DMF) to be filed with the FDA. Similarly, in Europe a dossier needs to be established. Both the DMF and dossier contain relevant safety information. IPEC Europe has issued a guideline (Compilation of Excipient Master Files Guidelines) providing guidance to excipient producers on constructing a dossier to support MAA (Marketing Authorization Application) while maintaining confidentiality of the data.
-
Existing chemical excipient—first use in man: This class implies that animal safety data exists and that the excipient may have been used in some other application. Additional safety information may be necessary to justify use in humans.
-
Existing chemical excipient: This class designates that the excipient has been used in humans but change in route of administration (e.g., from oral to parenteral), new dosage form, higher dose, etc. may require additional safety information.
-
New modifications or combinations of existing excipients: These excipients indicate a physical interaction, not a chemical reaction. No safety evaluation is necessary in this case.
Simply because an excipient is listed as GRAS (Generally Recognized As Safe), does not mean it can be used in a parenteral dosage form. The GRAS list may include materials proven safe for food (oral administration) but not deemed safe for use in an injectable product, making it difficult for a formulation development scientist to choose additives during dosage form development.
Many pharmacopeial monographs for identical excipients differ considerably regarding specifications, test criteria, and analytical methods. When a pharmaceutical manufacturer plans to supply a product throughout the world, repeat testing on the same excipient will be needed to meet USP, JP, Ph. Eur., BP, and other pharmacopoeias. There is ongoing harmonization of excipient monographs under the auspices of the Pharmacopoeial Discussion Group (PDG). The PDG has been working on several of the commonly used excipients in order to achieve a single monograph for each excipient. At present, 26 general chapters and 40 excipient monographs have been harmonized (Stage 6 of the process). For example, benzyl alcohol undergoes degradation by a free radical mechanism to form benzaldehyde and hydrogen peroxide. The degradation products are much more toxic than the parent molecule. The USP, JP and Ph. Eur. require three different chromatographic systems to test for organic impurity (mainly benzaldehyde). The harmonized monograph of benzyl alcohol has eliminated unnecessary repetition, which does not contribute to the overall quality of the product (68). The following 11 pharmacopoeial tests can be substituted by a single gas chromatography (GC) method:
Regulatory bodies are focusing their attention on the manufacturing process used to produce excipients. Major initiatives by the IPEC to improve the quality of additives has resulted in a publication guide titled Good Manufacturing Practices Guide for Bulk Pharmaceutical Excipients (69). Excipients may be manufactured for food, cosmetic, chemical, agriculture, or pharmaceutical industry; the requirements differ for each area. This guide proposes to develop a quality system framework to be used for suppliers of excipients, one that is acceptable to the pharmaceutical industry and will harmonize the requirements in the U.S., Europe, and Japan.
The United States and Europe require all excipients to be declared, along with their quantity, on the label (what is put on the immediate container) if the product is an injectable preparation. In Japan only the excipient names are required in the labeling (information that is included with the product, such as a package insert); E.U. Article 54(c) requires that all excipients must be declared on the labeling if the medicinal product is an injectable, a topical, or an eye preparation. The European guide for the label and package leaflet also lists excipients with special issues and are addressed in an Annex (70). Table XIII is a summary of some of these ingredients commonly used as parenteral excipients and the corresponding safety information that should be included in a leaflet. A package leaflet must include a list of information on those excipients, knowledge of which is important for the safe and effective use of the medicinal product. Similarly, 21 CFR 201.22 requires prescription drugs containing sulfites to be labeled with a warning statement about possible hypersensitivity. An informational chapter in USP, 〈1091〉 Labeling of Inactive Ingredients, provides guidelines for labeling of inactive ingredients present in dosage forms.
Excipients for Label and Corresponding Information for the Package Leaflet*
According to the Notes for Guidance on Pharmaceutical Development (CHMP/ICH/167068/04), the choice of excipients, their grade, compatibility, concentration, and function should be described in the P2 section of the Common Technical Document. It is necessary to justify inclusion of all ingredients in the drug product and describe their intended function. A specification of ±10% at the end of shelf-life is acceptable except for antioxidants and preservatives where performance data from PET or stability data may justify broader limits.
Bioburden and endotoxin limits of excipients used in the manufacture of sterile medical products shall be stated. Individual testing of excipients may be omitted if bioburden and endotoxin testing of the solution is checked prior to sterilization.
If an excipient is present in Ph. Eur or other major pharmacopoeia, the monograph specifications are usually acceptable in the registration file. For excipients not described in any pharmacopoeia, specifications should include physical characterization, identification tests, purity test, assay, and impurity tests. A certification must be included to confirm that excipients are of non-animal (specifically non-ruminate) origin. If this is not the case, a regulatory agency will require documentation to demonstrate freedom from viral and transmissible spongiform encephalopathies (TSE) risks (71).
IV. Criteria for the Selection of Excipient and Supplier
During the development of parenteral dosage forms, a formulator selects excipients to provide a stable, efficacious, and functional product. The choice and the characteristics of excipients should be appropriate for the intended purpose.
“An explanation should be provided with regard to the function of all constituents in the formulation, with justification for their inclusion. In some cases, experimental data may be necessary to justify such inclusion, e.g., preservatives. The choice of the quality of the excipient should be guided by its role in the formulation and by the proposed manufacturing process. In some cases it may be necessary to address and justify the quality of certain excipients in the formulation” (72).
Normally in the U.S. a pharmaceutical development report is written and should be available at the time of a pre-approval inspection. A development report captures the choice of excipients; their purpose and level in the drug product; their compatibility with other excipients, drug, or package system; and how they may influence the stability and efficacy of the finished product.
The following key points should be considered in selecting an excipient and its supplier for parenteral products:
-
Influence of excipient on overall quality, stability, and effectiveness of drug product.
-
Physical, chemical, and biological compatibility of excipient with drug and packaging system (73).
-
Compatibility of excipient with manufacturing process. For example, preservatives may be adsorbed by rubber tubes or filters; acetate buffers will be lost during lyophilization process, etc.
-
Amount or percentage of excipients that can be added to drug product. Table VI summarizes the maximum amounts of preservatives and antioxidants allowed by various pharmacopoeias.
-
Route of administration. USP, Ph. Eur., and BP do not allow preservatives to be present in injections intended to come in contact with brain tissues or cerebrospinal fluid. Accordingly, intracisternal, epidural, and intradural injections should be preservative free. It is preferred for an intravenously administered drug product to be a solution. If the size of the particle is well controlled, and below a certain maximum size (e.g., in a fat emulsion or a colloidal albumin or amphotericin B dispersion) then the drug can be administered by intravenous route.
-
Dose volume. All large volume parenterals (LVPs) and those vmall volume parenterals (SVPs) in which a single dose injection volume can be greater than 15 mL are required by Ph. Eur./BP to be preservative-free unless otherwise justified. The USP recommends that added substances are carefully chosen for use in injections that are administered in volumes exceeding 5 mL (34).
-
Whether a product is intended for single or multiple dose use. According to the USP, single dose injections should be preservative-free. The FDA's position for single dose injections, which have to be aseptically processed, is that manufacturers should not use a preservative to prevent microbial growth. European agencies have taken a less stringent stance on this subject.
-
The length or duration of time that a drug product will be used once the multi-dose injection is opened.
-
How safe is the excipient? Does it cause tissue irritation, hemolysis, or other toxic effects on cells, tissues, or organs?
-
Does the parenteral excipient contain very low levels of lead, aluminum, or other heavy metals?
-
Does a dossier or DMF exist for the excipient?
-
Has the excipient been used in humans? Has it been used via a parenteral route and in the amount and concentration that is being planned?
-
Has the drug product containing this excipient been approved throughout the world?
-
What is the cost of the excipient; is it readily available?
-
Is the excipient vendor following the GMP guide? Is the vendor ISO 9000 certified?
-
Will the excipient supplier certify the excipient to meet USP, BP, Ph. Eur., JP and other pharmacopoeias?
-
Has the supplier been audited by the FDA or the company's audit group? How did they fare?
Impurities in excipients can have a dramatic influence on the safety, efficacy, or stability of a drug product. Monomers or metal catalysts used during a polymerization process are toxic and also destabilize a drug product if present in trace amounts. Due to safety concerns, the limit of vinyl chloride (monomer) in polyvinyl pyrrolidone is ≤10 ppm and for hydrazine (a side product of polymerization reaction) ≤1ppm. Monomeric ethylene oxide is highly toxic and can be present in ethoxylated excipients like PEGs, ethoxylated fatty acids, etc.
The FDA has issued a guidance suggesting that animal-derived materials (e.g., egg yolk lecithin, egg phospholipid) used in drug products, originating from Belgium, France, and the Netherlands between January and June 1999 should be investigated for the presence of dioxin and polychlorinated biphenyls. Contamination of the animal-derived product was probably caused by consumption of contaminated animal feed.
Excipients manufactured by fermentation processes, such as dextrose, citric acid, mannitol, and trehalose, should be specially controlled for endotoxin levels. Mycotoxins (highly toxic metabolic products of certain fungi species) contamination of an excipient derived from natural material has not been specifically addressed by regulatory authorities. The German health authority issued a draft guideline in 1997 wherein a limit was specified for Aflotoxins M1, B1, and the sum of B1, B2, G1, and G2 in the starting material for pharmaceutical products.
Heavy metal contamination of an excipient is a concern, especially for sugars, phosphate, and citrate. Several rules have been proposed or established. For example, Ph. Eur. sets a limit of ≤1 ppm of nickel in polyols. California Proposition 65 specifies a limit of ≤0.5 μg of lead per day per product (74). Similarly, the USP and FDA limit aluminum content for all LVPs used in TPN therapy to 25 μg per liter (75). Further, it would require that the maximum level of aluminum in SVPs intended to be added to LVPs and pharmacy bulk packages, at expiration date, be stated on the immediate container label.
Physical and chemical stability of an excipient should be considered in assigning a re-evaluation date. Many drug products have a small amount of active drug and a comparatively high amount of excipients. Degradation of even a small percentage of excipient can lead to levels of impurities sufficient to react with or degrade a large percentage of active material. For example, benzyl alcohol decomposes via a free radical mechanism, in the presence of light and oxygen, to form benzaldehyde (x% of benzaldehyde is approximately equivalent to 1/3 x% of hydrogen peroxide). Hydrogen peroxide can rapidly oxidize sulfhydryl groups of amino acids like cysteine present in peptides or proteins.
Adequate research and thought is essential in the selection of a pharmaceutical excipient supplier. It is not uncommon for a supplier to change its manufacturing process to make it more efficient (less costly). Frequently, excipients are commodity (high volume/low value) products which are used by several industries. The pharmaceutical industry is often not a major customer in terms of volume of material purchased. For example, the pharmaceutical industry uses approximately 20% of gelatin produced. Of this 20%, most is used for production of oral dosage forms. The parenteral portion is approximately 5% of the 20%. It is extremely important that a drug manufacturer has a contract with an excipient supplier, prohibiting the supplier from making any change in the process or quality of the material without informing their customer well in advance. Pharmaceutical manufacturers should investigate all alternate sources that could be used in an emergency. A change in supplier should not be made without consulting the pertinent regulatory bodies because such an event may require prior regulatory approval.
A pharmaceutical manufacturer should have an active Vendor Certification Program. It should assure that vendor is ISO 9000 certified. An audit of an excipient manufacturer is essential because the pharmaceutical manufacturer is ultimately responsible for the quality of the drug product that includes the excipient(s) as one of its components. The IPEC GMP guide may be used as an audit tool because it is written in an ISO 9000 format using identical nomenclature and paragraph numbering. An audit ensures that quality is being built into the excipient, which may be difficult to measure later by quality control upon receiving the material. This is especially true for parenteral excipients where not only chemical but also microbiological attributes are critical. Bioburden and endotoxin limits may be required for each of the excipients and several guidelines are available to establish specifications (76, 77).
There are no legal requirements for excipient GMPs in Europe (78). A qualified person is responsible to assure that excipient quality is appropriate based on pharmacopeial specifications or a company's quality systems.
Events in Haiti highlight the importance of assuring excipient quality to the same degree that one normally does for active ingredients. From November 1995 through June 1996, acute anuric renal failure was diagnosed in 86 children. This was associated with the use of diethylene glycol contaminated glycerin used to manufacture acetaminophen syrup (79). The FDA is advising pharmaceutical companies to test for melamine down to a 2.5 ppm level in certain nitrogen-rich drug ingredients (raw materials that contain more than 2.5% nitrogen and those for which purity or strength is determined on the basis of nitrogen content) (80). The list of excipients includes albumin, amino acids derived from casein protein hydrolysates, ammonium salts, protamine sulfate, povidone, lactose, gelatin, etc. This guidance is in response to the incidents of pet food and Chinese milk doped with melamine.
The FDA recognizes the importance of excipients in a drug product for performance and safety. An injectable generic product should have identical non-exceptional excipients (qualitatively and quantitatively) as that of reference listed drug if the generic drug product is to follow the simplest path of registration; otherwise additional data must be submitted to demonstrate that the differences do not affect safety or performance (81). For parenteral products, non-exceptional excipients are ingredients other than preservatives, pH adjuster, antioxidant, and buffers.
V. Safety Issues
Handbook of Excipients edited by Bernstein (82) is an excellent reference on the safety and adverse reaction to several excipients. Sensitization reactions have been reported for parabens, thimerosal, and propyl gallate. Sorbitol, which metabolizes to fructose, can be dangerous when administered to fructose-intolerant patients. Table XIII lists safety concerns that must be included in labeling.
Progress in drug delivery systems and new proteins/peptides being developed for parenteral administration has created a need to expand the list of excipients that can be safely used. An informational chapter in the USP presents a scientifically based approach for safety assessment of new pharmaceutical excipients (83). A new or novel excipient is defined as one not previously used in a pharmaceutical preparation or not fully qualified by existing safety data with respect to the proposed level and duration of exposure or route of administration (84). Table XIV summarizes the approach in developing a new excipient. Besides the baseline toxicity data (either through literature or experimentation), if the drug (excipient) will be administered short-term (<14 days of consecutive days per treatment episode), intermediate-term (14–90 days), or long-term (>3 months) then additional safety information on the excipient is needed (84).
Summary of Safety Evaluation of a New Excipient Used in Injection
Currently, there are concerns regarding TSE via animal-derived excipients such as gelatin (85). TSEs are caused by prions that are extremely resistant to heat and normal sterilization processes. TSEs have a very long incubation time with no cure and include diseases such as
-
Scrapies in sheep and goat
-
Bovine spongiform encephalopathy (BSE), otherwise known as Mad Cow Disease, in cattle
-
Kuru disease in humans
-
Creutzfeldt-Jacob disease in humans, which has been attributed to repeated parenteral administration of growth hormone and gonadotropin derived from human pituitary glands
Several guidelines have been issued that address the issue of animal-derived excipients and scientific principles to minimize the possible transmission of TSE via medicinal products (86, ). The current situation indicates that there are negligible concerns for lactose, glycerol, fatty acids and their esters, but the situation is less clear for gelatin. Gelatin is still a necessary ingredient for some medicinal products and EMA (European Medicine Agency, formerly known as EMEA) has updated its guidance to allow gelatin from Category I and II countries if gelatin is produced by the acid process and Category I, II, and III countries if produced by the alkali process (88). Additional information on risk assessment of ruminate materials originating from USA, Canada, and other countries can be found in the references (87, 89, 90).
In the current regulatory environment, if given a choice, it is beneficial to select non-animal-derived excipient. Concerns about bovine serum albumin or human serum albumin (HSA) because of possible derivation from virus-contaminated blood remain. Recombinant human erythropoietin and darbepoetin alfa formulations were changed to replace albumin with polysorbate 80. Presently, recombinant HSA is available from several companies; this reduces probability of TSE (91). The risk of TSE from excipients can be greatly reduced by controlling the following parameters:
-
Sourcing of animal from countries where BSE has not been reported.
-
Using young animals
-
Category III or IV animal tissue used in manufacture (86)
-
Production process which is likely to destroy TSE agents
European Commission directive EMEA/410/01/rev 2 requires manufacturers to provide a “Certificate of Suitability” or the underlying “scientific information” to attest that their pharmaceuticals are free of TSEs.
Vegetable origin polysorbate should be used. If older products contain animal-sourced polysorbate then a switch should be made. EMA has stated that a change of a polysorbate 80 source will not result in re-performing viral inactivation studies (92). The vegetable source (e.g., trehalose from corn or tapioca) must be known as well as if any processing aids (e.g., enzymes during production of lactose) used during manufacturing of the excipient are derived from an animal source.
VI. Future Directions
Several new excipients, such as cyclodextrins, are being evaluated to improve solubility or stability of parenteral drugs. Currently, there are two FDA-approved parenteral products that utilize alpha and gamma cyclodextrins. Beta-cyclodextrin is unsuitable for parenteral administration because it causes necrosis of the proximal kidney tubules upon intravenous and subcutaneous administration (93). Hydroxypropyl beta-cyclodextrin (HPβCD) and sulfobutylether β-cyclodextrin (SBE-7-β-CD) have shown the most promise. Captisol™, the trade name of SBE-7-β-CD, is anionic. Currently, three Captisol™-based drug formulations have been approved in the U.S. One parenteral formulation is in Phase II/III clinical trials that utilize HPβCD (Cavitron®), and another (Sporanox) has been approved by the FDA. Manufacturers of HPβCD and SBE-7-β-CD have established a DMF with the FDA. Detailed reviews of cyclodextrins have been recently published (94, 95). One caution in using cyclodextrin is that they can (in rare cases) accelerate drug product degradation (96) and can sequester preservatives, rendering them ineffective (97).
Chitosan, β-1,4-linked glucosamine, a naturally occurring, biodegradable, nontoxic polycationic biopolymer, is being investigated for its potential as a cross-linked microsphere matrix to deliver antineoplastic drugs. Because of its charge, it can trap several drugs and can bind strongly with cancer cells, minimizing drug toxicity and enhancing therapeutic efficacy (98). Chitosan has also been shown to stabilize liposomes.
Biodegradable polymeric materials (polylactic acid, polyglycolic acid, and other poly-alpha-hydroxy acids) have been used as medical devices and as biodegradable sutures since the 1960s (99). Currently, the FDA has approved for marketing only devices made from homopolymers or co-polymers of glycolide, lactide, caprolactone, p-dioxanone, and trimethylene carbonate (100). Such bio-polymers are finding increased application as a matrix to deliver parenteral drugs for prolonged delivery (101). At least four drug products—Lupron Depot®, Decapeptyl®, Nutropin Depot®, and Zoladex®—have been approved. All four drug products are microspheres in Polyglycolic acid (PGA), Polylactic acid (PLA), or Polylacic-glycolic acid (PLGA) matrix. PGA is highly crystalline (approximately 50%) with a high melting point (220–225 °C). PLA can be produced by polymerization of L-lactic acid (LPLA), D-lactic acid (DPLA), or a blend of D&L-lactic acid (DLPLA). LPLA is 37% crystalline while DLPLA is amorphous. The degradation time of LPLA is much slower than that of DPLA. By copolymerizing lactic and glycolic acid polymeric matrices, a wide range of properties (tensile strength, crystallinity, and degradation rate) can be obtained. Decapeptyl®, a microsphere for intramuscular administration, is approved in France. It contains drug in a matrix of PLGA and Carboxymethyl cellulose with mannitol and polysorbate 80.
Polyanhydrides degrade primarily by surface erosion and possess excellent in vivo compatibility. In 1996 the FDA approved a polyanhydride-based drug implantable delivery system to the brain for the chemotherapeutic agent 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU).
Several phospholipid-based excipients are finding increased application as solubilizing agents, emulsifying agents, or as components of liposomal formulations. The phospholipids occur naturally and are biocompatible and biodegradable, for example, egg phosphatidylcholine, soybean phosphatidylcholine, hydrogenated soybean phosphatidylcholine (HSPC), dimyristoyl phosphatidylcholine (DMPC), distearoyl phosphatidylcholine (DSPC), 1,2 dioleoyl-sn-glycero-3-phosphocholine (DOPC), distearoyl phosphoethanolamine (DSPE), L-alpha-dimyristoylphosphatidylglycerol (DMPG), 1,2-dipalmitoyl-sn-glycero-3-phosho-rac-(1-glycerol) (DPPG), and distearoylphosphatidylglycerol (DSPG). Spartaject™ technology uses a mixture of phospholipids to encapsulate poorly water-soluble drug, forming a micro-suspension that can be injected intravenously. Busulfan drug product uses this technology and is currently undergoing Phase I clinical trials. Many liposomal and liposomal-like formulations (DepoFoam®) are either approved (DepoCyt®) or are undergoing clinical trials to reduce drug toxicity, improve drug stability, prolong the duration of action, or to deliver drug to the central nervous system (102). Two amphotericin formulations, a liposomal, or lipid complex between the antifungal drug and the positively charged lipid, have been approved in the U.S. Amphotec® is a 1:1 molar ratio complex of amphotericin B and cholesteryl sulfate while Abelcet® is a 1:1 molar complex of amphotericin B with phospholipids (7 parts of L-α-dimyristoylphosphatidylcholine and L-α-dimyristoylphosphatidyl glycerol).
Poloxamer or pluronic are block copolymers comprised of polyoxyethylene and polyoxypropylene segments. They exhibit reverse thermal gelation and are being tried as solubilizing, emulsifying, and stabilizing agents. A depot drug delivery system can be created using pluronics whereby the product is a viscous injection that gels upon intramuscular injection (103). Pluronics can prevent protein aggregation or ad/absorption and help in the reconstitution of lyophilized products. Pluronic F68 (Polaxamer-188), F38 (Poloxamer-108), and F127 (Poloxamer-407) are the most commonly used pluronics. For example, a liquid formulation of human growth hormone and Factor VIII can be stabilized using pluronics. Fluosol® is a complex mixture of perfluorocarbons with a high oxygen carrying capacity emulsified with Pluronic F-68 and various lipids. It was recently approved by the FDA for adjuvant therapy to reduce myocardial ischemia during coronary angioplasty. A highly purified form of Ploxamer 188 (Flocor™), intended for intravenous administration, is undergoing Phase III clinical trials for various cardiovascular diseases. Purification of Poloxamer 188 reduces nephrotoxicity. Another non-ionic surfactant, Solutol HS 15 (Macrogol-15-Hydroxystearate), has been approved by the Health Canada in a vitamin K1 formulation for human application.
Poloxamer and other polymeric materials like albumin may coat micro- or nanoparticles, alter their surface characteristics, and reduce their phagocytosis and opsonization by reticulo-endothelial system following intravenous injection. Such surface modifications often result in prolongation in the circulation time of intravenously injected colloidal dispersions (104). Poloxamers have also been used to stabilize suspensions (105).
The first successful development of an injectable perfluorocarbons-based commercial product was achieved by the Green Cross Corporation in Japan. They made Fluosol-DA®, a dilute (20% w/v) emulsion based on perfluorodecalin and perflurotripropylamine emulsified with potassium oleate, Pluronic F-68, and egg yolk lecithin. These perfluorocarbons are inert and can also be used to formulate non-aqueous preparations of insoluble proteins and small molecules (106). Perfluorocarbons have been approved by the FDA in one ultrasound contrast agent, Optison®, administered via the intravenous route. Optison® is a suspension of microspheres of human serum albumin with octafluoropropane. Heat treatment and sonication of appropriately diluted human albumin, in the presence of octafluoropropane gas, is used to manufacture microspheres in Optison® injection. The protein in the microsphere shell makes up approximately 5–7% (w/w) of the total protein in the liquid. The microspheres have a mean diameter range of 2.0–4.5 μm with 93% of the microspheres being less than 10 μm.
Sucrose acetate isobutyrate (SAIB), a high viscosity liquid system, converts into free flowing liquid when mixed with 10–15% ethanol (107). Upon subcutaneous or intramuscular injection, the matrix rapidly converts to a water-insoluble semi-solid capable of delivering proteins and small molecules for a prolonged period. SAIB is biocompatible, and it biodegrades to natural metabolites.
Several other biodegradable, biocompatible, injectable polymers are being investigated for drug delivery systems. They include polyvinyl alcohol, block copolymer of PLA-PEG, polycyanoacrylate, polyanhydrides, cellulose, alginate, collagen, modified HSA, albumin, starches, dextrans, hyaluronic acid and its derivatives, and hydroxyapatite (108). Efficient parenteral delivery of genes, immunomodulators, RNAi, anti-sense, aptamers, and other novel therapeutic agents will invariably require new excipients, with several excipients in early pre-clinical or clinical use (109).
Recent concerns about swine flu and other infectious diseases have regenerated interest in new vaccines and adjuvants to improve immune response. Some of the adjuvants included in marketed vaccines in Europe but still being evaluated include AS03 and AS04 (GSK) and MF-59 (Novartis). AS04 is composed of 3-O-desacyl-4′-monophosphoryl lipid A. It is present in Cervarix, which was approved in 2010. Newer adjuvants being tested include saponin-based (QS-21, Quil A, ISCOM) or emulsion-based (SA03).
Conflict of Interest Declaration
The authors declare that they have no competing interests.
Acknowledgements
Authors thank Hatsuki Asahara for providing comments, especially relating to the status of excipients in Japan, and Karen Busch for editing and proof-reading.
- © PDA, Inc. 2011
References
- 1.↵
- 2.↵
- 3.↵
- 4.↵
- 5.↵
- 6.↵
- 7.↵
- 8.↵
- 9.↵
- 10.↵
- 11.↵
- 12.↵
- 13.↵
- 14.↵
- 15.↵
- 16.↵
- 17.↵
- 18.↵
- 19.↵
- 20.↵
- 21.↵
- 22.↵
- 23.↵
- 24.↵
- 25.↵
- 26.↵
- 27.↵
- 28.↵
- 29.↵
- 30.↵
- 31.↵
- 32.↵
- 33.↵
- 34.↵
- 35.↵
- 36.↵
- 37.↵
- 38.↵
- 39.↵
- 40.↵
- 41.↵
- 42.↵
- 43.↵
- 44.↵
- 45.↵
- 46.↵
- 47.↵
- 48.↵
- 49.↵
- 50.↵
- 51.↵
- 52.↵
- 53.↵
- 54.↵
- 55.↵
- 56.↵
- 57.↵
- 58.↵
- 59.↵
- 60.↵
- 61.↵
- 62.↵
- 63.↵
- 64.↵
- 65.↵
- 66.↵
- 67.↵
- 68.↵
- 69.↵
- 70.↵
- 71.↵
- 72.↵
- 73.↵
- 74.↵
- 75.↵
- 76.↵
- 77.↵
- 78.↵
- 79.↵
- 80.↵
- 81.↵
- 82.↵
- 83.↵
- 84.↵
- 85.↵
- 86.↵
- 87.↵
- 88.↵
- 89.↵
- 90.↵
- 91.↵
- 92.↵
- 93.↵
- 94.↵
- 95.↵
- 96.↵
- 97.↵
- 98.↵
- 99.↵
- 100.↵
- 101.↵
- 102.↵
- 103.↵
- 104.↵
- 105.↵
- 106.↵
- 107.↵
- 108.↵
- 109.↵