Abstract
Drugs with low water solubility are predisposed to low and variable oral bioavailability and, therefore, to variability in clinical response. Despite significant efforts to “design in” acceptable developability properties (including aqueous solubility) during lead optimization, approximately 40% of currently marketed compounds and most current drug development candidates remain poorly water-soluble. The fact that so many drug candidates of this type are advanced into development and clinical assessment is testament to an increasingly sophisticated understanding of the approaches that can be taken to promote apparent solubility in the gastrointestinal tract and to support drug exposure after oral administration. Here we provide a detailed commentary on the major challenges to the progression of a poorly water-soluble lead or development candidate and review the approaches and strategies that can be taken to facilitate compound progression. In particular, we address the fundamental principles that underpin the use of strategies, including pH adjustment and salt-form selection, polymorphs, cocrystals, cosolvents, surfactants, cyclodextrins, particle size reduction, amorphous solid dispersions, and lipid-based formulations. In each case, the theoretical basis for utility is described along with a detailed review of recent advances in the field. The article provides an integrated and contemporary discussion of current approaches to solubility and dissolution enhancement but has been deliberately structured as a series of stand-alone sections to allow also directed access to a specific technology (e.g., solid dispersions, lipid-based formulations, or salt forms) where required.
I. Introduction
High hydrophobicity and intrinsically low water solubility are increasingly common characteristics of hits, leads, development candidates, and ultimately marketed drugs. Many hypotheses have been put forward as to why these trends have emerged, and the true explanation is clearly multifaceted. The application of combinatorial chemistries to generate large chemical libraries and the common application of high-throughput screening modalities, often in nonaqueous media (or mixed-solvent media), have probably played a role. The desire for increased potency, coupled with the realization that receptor binding is mediated, at least in part, by hydrophobic interactions, further magnifies the likelihood that drug candidates will have limited aqueous solubility. Finally, the quest to explore unprecedented drug targets, some of which are associated with intracellular signaling pathways, lipid processing architecture, or highly lipophilic endogenous ligands, only amplifies the requirement for highly lipophilic, poorly water-soluble drug candidates to access and interact with the target.
These drivers ultimately bias the identification of poorly water-soluble “hits” during early drug screens. Poor water solubility is a significant risk factor in low oral absorption because drug molecules must, in most cases, be in solution to be absorbed, and oral bioavailability is usually a required characteristic in a target product profile of an orally administered medicine. As such, medicinal chemistry strategies during lead optimization typically seek to modify physicochemical properties (including solubility) such that drug leads have more developable characteristics. Many decision gates, or idealized character panels, are used to identify and reject drug candidates with inappropriate developability properties and, subsequently, to synthetically modify structures to improve physicochemical characteristics. Perhaps the best known of these is Chris Lipinski’s “rule of 5” (Lipinski et al., 1997), but there are many others. In all cases, however, at least moderate water solubility is usually a focus.
Nonetheless, even with contemporary medicinal chemistry programs and increasingly sophisticated lead optimization strategies, it is apparent that for some targets, reducing lipophilicity and increasing water solubility will result in an unacceptable reduction in potency. In spite of attempts to circumvent solubility problems, approximately 40% of currently marketed drugs (Fig. 1) and up to 75% of compounds currently under development have been suggested to be poorly water-soluble (Di et al., 2009, 2011). Furthermore, the problems of low water solubility do not seem to be diminishing and may well be increasing (Takagi et al., 2006). Low water solubility therefore continues to be a challenge to successful drug development.
A comparison of the distribution of solubilities for the top 200 oral drug products in the United States (US), Great Britain (GB), Spain, and Japan and from the World Health Organization (WHO) Essential Drug List. Very soluble drugs: over 1000 mg/ml; freely soluble drugs: 100–1000 mg/ml; soluble drugs: 33–100 mg/ml; sparingly soluble drugs: 10–33 mg/ml; slightly soluble drugs: 1–10 mg/ml; very slightly soluble drugs: 0.1–1 mg/ml; practically insoluble drugs: <0.1 mg/ml. Adapted from Takagi et al. (2006).
This review seeks to provide an overview of the strategies that may be taken to address the problems of low solubility during drug discovery and development and includes a comprehensive review of formulation approaches to support the clinical development of oral and parenteral drug products for poorly water-soluble drugs. In addition, even when lead optimization is successful in increasing aqueous solubility, early preclinical studies are still required with less than optimal leads to provide the data sets to allow informed progression or rejection; therefore, we also address the strategies that might be used when dealing with the complexities of low solubility during the discovery phase. In the former case, at least for oral drug products, the market typically dictates the need for traditional solid dosage forms (e.g., capsules, tablets). In the latter, where studies are usually conducted in small animals (rodents), liquid formulations are often used to allow oral dosing, and therefore a slightly different approach must be taken.
This review is structured to provide an initial and relatively brief introduction to the determinants of low water solubility to provide the theoretical basis for the approaches that might be taken to address solubility challenges. We subsequently present summary sections that outline potential strategies for addressing solubility issues, first in vitro and second in vivo, with the latter section addressing both parenteral and oral administration. Subsequently, we provide a comprehensive review of the technologies that can be used to promote solubility or dissolution. These latter sections are necessarily dense and are intentionally separated from the higher-level strategy summaries provided in the introduction. The technology overviews provide a reference source for the summaries that precede them. A schematic representation of each of these formulation approaches is provided in Fig. 2 and is intended to highlight the variety of proven strategies available to those working with poorly water-soluble drugs.
Schematic diagram illustrating the common strategies currently used (and discussed in this review) to address low drug solubility in drug discovery and development.
A. What Is Low Drug Solubility?
Although low water solubility of drug candidates presents varied and significant challenges throughout drug discovery and development, the greatest concern is generally the risk of reduced and variable absorption after oral administration. The value at which limited solubility begins to impact absorption is difficult to state definitively since it is dependent on a number of other system variables, including drug permeation, dose, and the environment present within the gastrointestinal (GI) tract. To understand these variables, and therefore, the factors that impact the required solubility for a drug candidate, it is instructive first to appreciate the drivers of flux across an absorptive membrane since these in turn will determine whether the drug dose can be absorbed over the timescale available.
Assuming appropriate chemical and metabolic stability and an absence of transporter or carrier-mediated processes, flux (F) across an absorptive membrane is the product of the concentration gradient and the permeability across the membrane and is described as follows:(1)where Cm is the drug concentration immediately adjacent to the membrane, C0 is the drug concentration on the abluminal side of the membrane, K is the partition coefficient between the aqueous solution overlaying the membrane and the membrane itself, h is the width of the diffusion layer, D is the diffusion coefficient of the drug in the membrane, and A is the surface area. Assuming that the drug concentration on the abluminal side of the absorptive membrane is low relative to the concentration at the absorptive site (i.e., Cm > > C0), eq. 1 can be simplified to the following:
(2)
The values for D, K, and h are typically fixed for a particular system and are used to define the permeability coefficient (P) where(3)
In the absence of supersaturation, the maximum concentration that can be attained at the surface of an absorptive membrane is equivalent to the equilibrium solubility (Cs) of the drug, and therefore the maximum flux (per unit area) (F’) is the product of solubility and permeability:(4)
Appreciation of this relationship illustrates that knowledge of the solubility alone is insufficient to anticipate whether solubility will limit flux (or absorption) since flux is also a function of permeability. To some extent, therefore, low solubility can be offset by high permeability; similarly, if permeability is low, the requirements for solubility to generate appropriate flux increase.
A well recognized approach applied in early drug discovery for estimating the required solubility and permeability needed to achieve good oral absorption is the concept of a maximum absorbable dose (MAD), originally derived by Johnson and Swindell (1996) and further applied by Curatolo (1998) and Lipinski (2000):(5)where Cs is the solubility (mg/ml) at pH 6.5 (representing the pH of the small intestine); kabs is the rate constant (h−1) for intestinal absorption (which is related to the permeability); SIWV is the small intestinal water volume (in milliliters), which is typically assumed to be ∼250 ml (the volume of fluid assumed to be present in the fasted GI tract after a glass of water has been drunk when taking medication orally); and SITT is the small intestinal transit time (min) of ∼270 min (4.5 h). Rearranging this relationship provides an expression for the necessary or target solubility for a given dose and kabs (or permeability) and provides an initial indication as to whether solubility is likely to limit oral absorption. This concept is shown graphically in Fig. 3, the data from which are taken from a seminal review that shows the theoretical required solubility to provide good oral absorption for drugs with projected doses ranging from 0.1 to 10 mg/kg and permeabilities ranging from low to high. At one end of the spectrum, highly potent drugs for which the dose is low and the membrane permeability is high have relatively low solubility requirements to achieve good oral absorption. At the other extreme, low-potency drugs for which the dose is high and the permeability is low need considerably higher solubility for good oral absorption (by several orders of magnitude in this example). As a broad initial estimation, and assuming moderate potency (1.0 mg/kg) and moderate permeability (kabs = 1.0 h−1), this approach indicates that where aqueous solubilities are <50 µg/ml, problems associated with low water solubility might be anticipated. It is clear, however, that the required solubility to support drug absorption must be evaluated in light of both the potency (or dose) and the permeability characteristics. It is also clear that at stages during the development pathway, in particular during preclinical toxicity testing, exposure at doses considerably in excess of the predicted clinical dose will be required, magnifying the need for solubility support.
The relationship between the projected dose and the required aqueous solubility for low, medium, and high permeability compounds. Permeability is indicated by the magnitude of the absorption rate constant (kabs), where low permeability, kabs = 0.1; medium = 1.0, high = 10 h−1. From this analysis, moderately permeable compounds (kabs = 1.0 h−1), with projected potencies of 1.0 mg/kg, require aqueous solubilities of ∼52 μg/ml. Adapted from Lipinski (2000).
A further application of the solubility-permeability relationship to oral drug absorption is the Biopharmaceutics Classification System (BCS) (Fig. 4), originally developed by Amidon et al. (1995), with subsequent variations by others (Wu and Benet, 2005; Butler and Dressman, 2010; Chen et al., 2011). The principles of the BCS are well described elsewhere (Amidon et al., 1995; Yu et al., 2002; Dahan et al., 2009), but in brief, the BCS allows classification of drug molecules as a function of their solubility and permeability properties. Originally proposed to provide a scientific basis for biowaivers based on a correlation of in vitro drug dissolution and in vivo drug absorption, this classification system has found much broader applicability across many areas of drug discovery and development. According to the BCS, class I molecules are those having both high solubility and high permeability (and therefore likely few problems with oral absorption); class II compounds are those that have low solubility and high permeability (where solubility is the primary limitation to absorption); class III compounds have high solubility but low permeability (where absorption is limited by membrane permeation and not solubility); and class IV compounds are those in which both poor solubility and poor permeability limit drug absorption. The focus of the current review is therefore BCS class II compounds, which often exhibit solubility-limited absorption. Class IV compounds are also relevant, although they have additional problems associated with low permeability.
Drug dose is also an important factor in the BCS because highly soluble drugs are defined as those in which the highest dose will dissolve in 250 ml over the pH range of the GI tract (i.e., pH 1–6.8). Several examples of dose, solubility, and volume requirements (taken from Amidon et al., 1995) are shown in Table 1. As with the MAD, these calculations are not designed to be definitive but rather illustrate that low-dose compounds, such as digoxin, can have good absorption and bioavailability, even when GI solubility is low, whereas the absorption of high-dose compounds, such as griseofulvin, is more often low, variable, and highly formulation dependent as a result of their solubility limitations.
Examples of dose, solubility, and volume requirements for a range of poorly water-soluble drugs
A complication of the BCS definition of high solubility is that the highest strength dose must be soluble in 250 ml of water at all pH values that might be encountered in the GI tract. Therefore, drugs may be classified as class II even though they have good solubility at one end of this pH range. For example, many weak acids have low solubility at pH 1 and are strictly classified as BCS class II compounds but are quite soluble at intestinal pH (pH 6–7) and in many cases do not exhibit solubility-limited absorption. As such, a BCS class II designation does not always dictate that solubility will be a limitation to absorption; rather, compounds in this BCS class are more likely to be solubility limited than are those in class I. Indeed, a more practical description of BCS class II may be drugs that do not have high solubility rather than those that have low solubility since the classification system specifically identifies compounds that fall into class I and all those with solubilities below this fall inherently into class II (or class IV if permeability is also low) (Fig. 4).
Diagrammatic representation of the BCS, which classifies drugs according to their permeability and solubility properties. Compounds are defined as high solubility when the quantity of drug that is present in the highest strength immediate release dose form is soluble in 250 ml of water across the likely range of gastrointestinal pH (1.2–7.5). Highly permeable compounds are defined as those that are >90% absorbed or where permeability as assessed by in vitro or in vivo methods is equivalent to or higher than that of a reference compound that is 90% absorbed. Adapted from Amidon et al. (1995).
The preceding discussion serves to illustrate further that low solubility is a somewhat arbitrary concept when assessing the likelihood that solubility will limit drug absorption and that additional knowledge of the likely dose and membrane permeability is inevitably required to put a solubility value into an appropriate context.
B. Determinants of Aqueous Solubility
A detailed description of the thermodynamic determinants of drug solubility in aqueous media is beyond the scope of the current discussion; for more information, the interested reader is directed to the following references: Grant and Higuchi (1990), Yalkowsky (1999), and Murdande et al. (2010a). An overview of the broad physicochemical drivers of drug solubility is warranted, however, since these drivers underpin the approaches that can be taken to enhance solubility. Simplistically, the potential for a drug (solute) to pass into solution in an aqueous fluid (solvent) is dictated by three separate events, as shown in Fig. 5. First, solute molecules must be abstracted from the solid state, a process that involves breaking solute-solute bonds. The strength of solute-solute attractive forces in different solids varies significantly and is typically higher for electrolytes than for nonelectrolytes (since ionic attractive forces in the solid state are stronger than nonionic forces), for crystalline solids compared with amorphous materials, and for planar nonelectrolytes (which pack more effectively in the solid state) compared with nonplanar molecules. The melting point provides a reasonable indication of the strength of intermolecular solute-solute interactions in the solid material. Second, a void must be created in the solvent that is sufficient to accommodate the abstracted solute molecule. Since intermolecular forces in the liquid state are much lower than those in the solid state, the energy required to create a void in the solvent is low and is usually ignored when assessing energy changes during dissolution. Finally, the solute molecule is inserted into the solvent void. For molecules with some affinity for a polar solvent such as water, this process is energetically favorable and therefore drives drug solubility. This last concept is the rationale behind the often quoted maxim “like dissolves like.” This is largely true, but it captures only half the story since it ignores the impact of changes to solid-state properties on solubility. Since the energy transitions associated with changes to solvent structure are small, it is apparent that there are two primary determinants of drug solubility: 1) the energy required to overcome the strength of intermolecular forces in the solute solid state and 2) the energy generated on the interaction of solute and solvent molecules in solution (solvation)
Three essential steps are required for a solute drug molecule to be displaced from a solid particle and to enter solution. Step 1: A single solute molecule is removed from the crystal lattice; energy is required in this step to overcome solute-solute interactions in the solid state. Step 2: A void is created within the solvent to accommodate the solute molecule. Although this step also requires energy, it is likely to be considerably lower than the energy required in step 1. Step 3: The solute molecule inserts into the solvent, forming solute-solvent interactions. Simplistically, if the energy released from the solute-solvent interactions (i.e., step 3) is greater than the energy required for steps 1 and 2, solubility is favored.
For an analogous structural series, solubility is therefore generally lower for molecules that have higher melting points (stronger attractive forces) and lower affinity for water (poor solvation).
1. Ideal Versus Nonideal Solubility
Solubility theory defines an idealized condition or an ideal solution, where the intermolecular forces between solute and solvent are equivalent to those between solute and solute and those between solvent and solvent. Under these circumstances, the mixing of solute and solvent molecules in the liquid state results in no net energy change. Where this is the case, solubility is dependent on the strength of the solute crystal lattice and may be defined by(6)where X is the ideal mole fraction solubility of solute, ΔHf is the enthalpy of fusion of solute, Tm is solute melting point, T is the absolute temperature, and R is the gas constant.
In reality, ideal solutions are highly unusual, and solution conditions close to ideality exist pharmaceutically only in solutions of highly lipophilic drugs in nonaqueous (lipidic) solutions. In contrast, in aqueous solution, the differences between solute-solute interactions and solute-solvent interactions are highly significant, leading to nonideal solution behavior and much lower solubility than would otherwise be predicted. In this case, the difference between ideal and nonideal behavior is captured by a correction factor termed the activity coefficient (γ), where(7)
In turn, the activity coefficient is a function of the molar volume of the solvent (Vs), the volume fraction of the solute (ϕ1), and the difference in the solubility parameters for the solvent (δ1) and solute (δ2):(8)
The solubility parameters provide a measure of the cohesive forces in either solute or solvent, and from eq. 8, it is evident that smaller differences between the solubility parameters of the solvent and solute give rise to lower activity coefficients and, therefore, an increase in solubility toward ideality (i.e., δ1 = δ2) (Rubino, 2002).
The solubility equation (eq. 7) captures the determinants of solubility as dictated by changes to the solid-state properties of a drug (and usually manifest by changes to melting point) and the activity coefficient (reflecting differences between the solubility parameters), and it reiterates the importance of solute-solute interactions in the solid state and the need to minimize differences between solute and solvent properties to maximize solvation (i.e., like dissolves like). These principles underpin all the approaches to solubility enhancement that are subsequently described in this review, as each of these approaches either change solid-state properties or change the nature of the interaction between drug (solute) and solvent molecules.
C. Hydrophobic or Lipophilic Drug Candidates?
An understanding of the primary drivers of drug solubility allows an important distinction to be made between poorly water-soluble drugs that are limited by solid-state properties (e.g., the strength of the crystal lattice), those that are limited by solvation (i.e., solute-solvent interactions in solution), and those that are limited by both. In practice, most compounds fall into two categories since almost all poorly water-soluble compounds have limited affinity for water (i.e., they are hydrophobic) and are therefore solvation-hydration limited. Where compounds are hydrophobic and also show strong intermolecular forces in the solid state, they are typically poorly soluble in both aqueous and nonaqueous solvents. In contrast, hydrophobic molecules, in which solubility is not limited by solid-state properties, often show varying degrees of solubility in nonaqueous solvents such as lipids (since the molecular properties that reduce hydration in aqueous media often promote solvation in nonaqueous media). The latter compounds are therefore both hydrophobic and lipophilic, with the former simply being hydrophobic. The difference between these two types of molecules can be illustrated using the analogies of “brick dust” for hydrophobic molecules with poor solubility in all solvents and “grease balls” for compounds that are hydrophobic and lipophilic and show reasonable solubility in lipids (Stella and Nti-Addae, 2007). This distinction is important since the formulation options available for either hydrophobic or lipophilic compounds differ considerably. Simplistically, the increased lipid solubility of lipophilic drug candidates allows access to liquid surfactants and lipid-based delivery technologies that can be filled into soft gelatin capsules (or sealed hard gelatin capsules), whereas the lack of solubility for hydrophobic molecules in almost all vehicles precludes formulation in anything other than modified solid dosage forms.
D. Solubility of Electrolytes, Weak Electrolytes, and Nonelectrolytes
The solvation properties of drug molecules, and therefore a significant part of the driving force for drug solubility in aqueous media, are highly dependent on the extent of ionization. The presence of a charged functional group provides an opportunity for favorable ion-dipole interactions with polar solvents such as water, which directly enhance hydration and water solubility. Strong electrolytes (such as NaCl) that completely dissociate in water are generally highly water soluble. This is not always the case, however, as solubility remains a composite function of the energy required to break the crystal lattice versus the energy liberated on hydration of the ions formed. In some extreme cases, for example, an inorganic salt such as AgCl, the crystal lattice energy is sufficiently high that aqueous solubility remains low. As a strong electrolyte, therefore, dissociation of AgCl molecules in solution is complete, but as a poorly water-soluble strong electrolyte, solubility is limited.
In reality, most drugs are organic materials that are either nonelectrolytes (which do not dissociate to form ionic species in water) or weak electrolytes that dissociate only partially in water such that both un-ionized solute and the dissociated ions are present in solution.
The solubility of weak electrolytes is highly dependent on the degree of ionization (dissociation) since the affinity of the ionized species for water is markedly higher than that of the un-ionized species. The degree of dissociation is in turn dependent on the pKa of the weak electrolyte and the pH of the solution into which it is dissolving. Simplistically, at pH values above the pKa of a weak acid and below the pKa of a weak base, solubility increases significantly as a result of ionization. For weak acids and bases with a single ionizable group, solubility increases by a factor of 10 for every pH unit away from the pKa (although this trend does not continue indefinitely and solubility is ultimately limited by the solubility product of the salts that are formed in situ with available counterion; see Section IV). Nonetheless, optimization of solution pH is an effective means by which solubility can be enhanced and is commonly used to enhance the solubility properties of solution formulations for weak electrolytes. For solid dosage forms, the principles of pH-dependent solubility may be manipulated by the isolation of a drug (or drug candidate) as an appropriate salt form. The use of pH and salt selection to enhance solubility and the dissolution rate is described in more detail in Section IV. For nonelectrolytes, solubility behavior is not complicated by the effects of ionization and remains a function of hydration and the strength of the crystal lattice.
E. Solubility and Dissolution Rate
The solubility of a drug in aqueous solution is a fundamentally important property that affects not only the potential for drug absorption after oral administration and the ability to administer the drug parenterally but also the ease of manipulation and testing in the laboratory and during manufacture. However, drug solubility is an equilibrium measure and the rate at which solid drug or drug in a formulation passes into solution (i.e., the dissolution rate) is also critically important when the time available for dissolution is limited. This rate is particularly relevant after oral administration since intestinal transit time is finite and the rate of drug dissolution must significantly exceed the rate of transit for absorption to be maximized. For example, the absorption of a drug with reasonable solubility may still be poor if the rate of dissolution is low since the solubility limit may never be reached during the intestinal transit time. Similarly, even where the rate of dissolution is relatively rapid, if the equilibrium solubility is low, the quantity of drug available in solution for absorption is unlikely to support rates of flux across the intestine that are sufficient to absorb the entire drug dose in the time available. For different drugs, and under different circumstances, either solubility or dissolution rate (or both) may be the limiting feature.
The process of drug dissolution from the solid state is summarized in Fig. 6. An unstirred water layer is present on the surface of every dissolving solid and provides the barrier to drug equilibration with the well stirred bulk solution. The dissolution rate is therefore defined by the rate at which drug diffuses across the unstirred water layer, and the equations that describe drug dissolution (i.e., the Noyes Whitney Equation, eq. 9) are analogous to simple diffusion equations. The rate of transfer across the unstirred layer is a function of the concentration gradient across the unstirred layer, the width of the diffusion layer (h), the surface area of contact of the solid with the dissolution fluid (A), and the diffusion rate of the drug in water (D). The concentration gradient in turn is a function of the maximum drug concentration at the surface of the dissolving solid (drug solubility Cs) and the concentration in the well-stirred bulk (C) (Noyes and Whitney, 1897):(9)
Graphic depicting the process of drug dissolution from a solid drug particle. An unstirred water layer of width (h) is present on the surface of the dissolving solid. A concentration gradient is established across the unstirred water layer that drives dissolution and results from the difference in drug concentration between that on the surface of the dissolving solid (usually assumed to be the equilibrium solubility of the drug, Cs) and the concentration in the well stirred bulk (C).
If we assume sink conditions where the concentration of drug in bulk solution (C) is low relative to the concentration on the surface of the dissolving solid (Cs), then this relationship collapses to(10)
For most small molecules, the diffusion rate constant in water (D) is relatively high and manipulation of drug structure does not typically affect D to the extent that it has a significant impact on dissolution rate. Similarly, whereas the width of the diffusion layer can be altered by agitation or stirring in vitro, this aspect cannot be easily manipulated in vivo.
The major determinants of in vivo drug dissolution rate are therefore solubility and surface area. Since solubility is a function of the strength of the crystal lattice and the affinity of the solute (drug) for the aqueous environment, three major strategies can be defined to increase the solubility or dissolution rate (realizing that the dependence of dissolution on solubility dictates that increases in solubility inherently increase the dissolution rate):
Reducing intermolecular forces in the solid state (increases solubility and dissolution rate)
Increasing the strength of solute-solvent interactions in solution (increases solubility and dissolution rate)
Increasing the surface area available for dissolution (increases dissolution rate, potential to moderately increase solubility at very small particle sizes (<1 µm)
F. Summary
Low aqueous solubility and reduced dissolution rates are a common property of many new drug candidates, and these properties create a number of challenges during drug discovery and development. An understanding of the determinants of solubility and dissolution provides a framework from which approaches to enhance solubilization may be developed. In subsequent sections of this review, we first address the complexities of working with poorly water-soluble drugs in vitro and subsequently summarize the approaches that can be taken to assist in the development of both parenteral and oral formulations. The main body of the review follows and provides a detailed account of the technological approaches that can be taken to support the development of effective formulations for poorly water-soluble drugs. Comment is made as to the many different approaches that might be taken during lead optimization and preclinical development and also those strategies that are also appropriate for extension into clinical development and ultimately to the market. To constrain the scope of this review, synthetic medicinal chemistry approaches to solubility manipulation are not addressed and the discussion is limited to approaches that do not result in the generation of a fundamentally new chemical entity. For more information on approaches to solubility manipulation via structural modification, the interested reader is directed to the following reviews: Fleisher et al. (1996), Stella and Nti-Addae (2007), Di et al. (2009), Keseru and Makara (2009), and Ishikawa and Hashimoto (2011).
II. In Vitro Complexities of Working with Poorly Water-Soluble Drugs
Despite the drive to identify “drug-like” leads with acceptable physicochemical properties, lead series are often plagued by poor aqueous solubility. The basis for this trend was discussed earlier in this introduction, but regardless of the source, working with inherently poorly water-soluble compounds creates a number of challenges throughout drug discovery, beginning with primary activity screens and progressing through to secondary in vitro assays and in vivo assessment.
In many in vitro assays, there is little scope for improving solubility given the poor tolerability of many in vitro biologic test systems for solubilizing components. In this instance, the focus must be on understanding the consequences of simple solution preparation and manipulation (e.g., dilution), appreciating the potentially confounding effects of compound precipitation on assay results, and grasping the impact of commonly used buffer components in dictating “free” compound concentrations in solution. The sections that follow highlight some of the common problems associated with the in vitro testing of poorly water-soluble compounds and, where available, approaches that can be taken to overcome, or at least minimize, these issues.
A. Drug Precipitation, Adsorption, Binding, and Complexation in In Vitro Assays
Beyond the realms of chemical synthesis, most in vitro evaluations of compound performance are conducted using aqueous-based biologic buffers and related media. The range of in vitro testing protocols is, of course, broad but might include binding or displacement assays, enzyme inhibition studies, activity screening in cell culture, traditional organ-bath pharmacology, assessment of uptake and transport in cell culture systems, or excised tissues and metabolic stability studies using microsomes or hepatocytes. In all cases, an accurate knowledge of the concentration of drug in solution is required as it is critical to the determination of the experimental endpoint. For example, most in vitro assays of drug activity are based on a concentration-response relationship, with the endpoint being some measure of the inhibitory or effective concentration (e.g., IC50 or EC50). In other assays, changes to drug concentrations in solution during the assay are used as an indicator of cellular uptake or transport, metabolism, or binding, all of which rely on a known concentration of compound in solution.
In most drug discovery settings, moderate- to high-throughput assay formats (i.e., 96-, 384-, or 1536-well plates) are used, and compounds are introduced into aqueous biologic media via dilution of concentrated stock solutions prepared in water miscible organic solvents, the most common being dimethyl sulfoxide (DMSO). DMSO is an excellent solvent for many poorly soluble compounds, including those with diverse chemical structures, and allows for the preparation of highly concentrated stock solutions for subsequent dilution for most compounds. However, compound precipitation after dilution of concentrated cosolvent solutions is common (see Section VI) and can lead to variable responses depending on how the dilution is conducted and the composition of the final assay media. This in turn can lead to a high degree of variability and inconsistent results between assays where these variables may be different. Different biologic assays also vary in their tolerance to the final DMSO (or other cosolvent) concentrations, making it difficult to standardize experimental conditions and dilution procedures across different assay formats.
For many in vitro assays (with the exception of high-throughput assays specifically designed to assess solubility), it may be difficult, if not impossible, to detect the presence of finely precipitated material on dilution into aqueous media, and measuring the final concentration of drug may be impractical. Working with compounds with inherently low aqueous solubilities generally limits the available range of drug concentrations that can be used to define concentration-dependent processes. Under these circumstances, complete binding or inhibition profiles may be difficult to define since the solubility limit is reached before saturation of binding sites or approach to maximal inhibition.
Furthermore, the physicochemical properties that predispose compounds to low aqueous solubility can also lead to an association of drug molecule with hydrophobic environments and surfaces. In fact, this phenomenon is often a driving force for biologic potency since partitioning into and across cell membranes and interaction with cellular and molecular targets are thermodynamically favored compared with residence within an aqueous solution. However, these characteristics also lead to an inherent propensity for nonspecific adsorption to surfaces such as tubes, pipette tips, filters, syringes, multiwell plates and cellular supports. Under these circumstances, a decrease in drug concentration in solution may be reflective of nonspecific adsorption rather than binding, uptake, or transport, artificially reducing the concentration of drug in solution and leading to an erroneous endpoint determination if concentrations are assumed on the basis of only the dilution factor. Clearly, there are advantages to obtaining measured concentrations to provide confidence in the experimental results when practical and also to provide an assurance of mass balance, which is a critical control measurement in any mass transport experiment.
The adsorption of drug to filter membranes requires special mention since separation of free drug from bound, complexed, or precipitated drug is a common procedure during the conduct of many in vitro assays. Assessment of the potential for adsorption during filtration is an important validation step but is particularly critical for poorly water-soluble drugs. Where significant adsorption is evident, and unavoidable, then it may be necessary to avoid the process of filtration. Equilibrium dialysis may provide an alternative under such circumstances, but the potential for adsorption to the dialysis membrane is also high. A final approach is to use ultracentrifugation to separate larger drug complexes from free drug in solution, but these measurements are time consuming, require specialized equipment and a significant density difference between species, and ultimately still carry the risk of drug adsorption to centrifuge tubes or plates.
Several different approaches can be taken to overcome issues of adsorption. The first is the choice of tube, filter, culture flask, multiwell plate, filter or pipette tip, as many manufacturers supply materials with modified surface properties to reduce nonspecific adsorption. It is important to appreciate the consequences of using different surfaces as those specifically designed to, for example, promote cell adhesion by making them more hydrophobic, will also increase the likelihood of nonspecific drug adsorption. In contrast, more hydrophilic surfaces will generally reduce adsorption by reducing the thermodynamic favorability of drug leaving the largely aqueous solution environment. Another approach involves pre-exposing potential adsorption sites to drug solution with a view to saturating adsorption before conduct of the experiment. Finally, alteration of the solution properties can reduce adsorption by increasing drug affinity for the bulk solution and reducing the effective partition coefficient between solution and drug adsorption sites. Most commonly, this is achieved via the inclusion of small quantities of a cosolvent (depending on the sensitivity of the particular assay) or by manipulating solution pH to increase solute-solvent interactions in solution. The use of pH and cosolvents to enhance solubility is described in more detail in Sections IV and VI, respectively.
Adsorption may also be reduced via the addition of a complexation or solubilization agent such as a surfactant (see Section VII), cyclodextrin (see Section VIII), or protein. Although these approaches are often highly effective, they introduce a further complexity, namely, changes to the chemical potential or thermodynamic activity of free (unbound) drug in solution as discussed in the following section.
B. Changes to Thermodynamic Activity Resulting from Complexation, Binding, or Solubilization
The effective concentration of a species in solution is most accurately defined by its thermodynamic activity (a), which is related to the concentration (C) and the activity coefficient (γ):(11)
A detailed evaluation of thermodynamic activity and chemical potential is beyond the scope of this review, but in simple terms, the effective concentration or thermodynamic activity can be considered as the concentration of drug in solution that is unconstrained by interaction with any other molecular species and is therefore available to exert its effect, regardless of whether the effect is to bind to a receptor or to diffuse across a membrane. In concentrated solutions, for example, the close molecular proximity of drug molecules to each other may promote solute-solute interactions (i.e., enhance intramolecular association), thereby reducing thermodynamic activity. Under these circumstances, the activity coefficient is less than unity, and the effective concentration or activity is less than the measured concentration. In typical dilute solutions, the degree of dilution is expected to reduce solute-solute intermolecular interactions in solution such that each molecule effectively acts independently, and under these circumstances, the activity coefficient is unity and activity is equal to concentration. This allows the definition of equilibrium constants, for example, in dilute solution using drug concentration instead of the thermodynamically correct use of activities.
The relevance of this discussion to the in vitro testing of poorly water-soluble drugs is that many strategies that promote drug solubility in aqueous solution and assay media can change the thermodynamic activity of the drug in solution. This is most readily illustrated by considering the impact of the addition of a surfactant to an aqueous solution of drug. Surfactants are amphiphilic molecules, and in aqueous solutions, they can exist as either monomers or as micellar structures. As surfactant concentrations are increased above the critical micelle concentration (CMC), the concentration of monomeric surfactant remains constant while the concentration of surfactant present as micelles in solution increases. Surfactant micelles typically enhance drug solubility by providing a hydrophobic environment in the micellar core to solubilize poorly water-soluble drugs as they have a greater affinity for this hydrophobic environment than for the surrounding aqueous environment (see Section VII). However, under this circumstance, the thermodynamic activity of the drug is much lower than the total concentration as the activity coefficient of the drug is lower than unity. In the case of drug solubilized in a surfactant micelle, drug can be considered as existing in equilibrium between solubilized drug within the micelle and free drug in an intermicellar phase or bulk solution phase. Here, solubilized drug is highly constrained by the surrounding micellar structure, and the effective concentration (i.e., the thermodynamic activity) is more accurately represented by the free (nonmicellar) concentration of drug.
In addition to the use of surfactants, other common circumstances where the thermodynamic activity of a compound might be reduced include the introduction of a complexation agent, such as a cyclodextrin, or the addition of plasma or serum proteins to cell-based assay media. For the latter situation, the so-called serum shift phenomenon is widely recognized where in vitro activity changes in response to changing concentrations of serum present in the media. For a more comprehensive review of the effect of protein binding on the pharmacological activity of drugs and the complexities of interpreting static (in vitro) versus dynamic (in vivo) situations, the reader is referred to the excellent review by Smith et al. (2010). In each of these cases, the total concentration of drug is significantly higher than the effective or free concentration, and the situation is further confounded by the difficulty in accurately quantifying free drug concentrations. In contrast, solubility enhancement through pH manipulation or cosolvency will typically have limited impact on thermodynamic activity of drug in solution.
III. In Vivo Assessment of Poorly Water-Soluble Compounds
In vivo assessment of new drug candidates begins with early in vivo efficacy studies in animal models and continues through pharmacokinetic and dose-limiting toxicology studies in various preclinical species and ultimately into human clinical trials. Preclinical pharmacokinetic studies are heavily used to support human dose and pharmacokinetic predictions and, along with results from toxicology studies, are used to select a safe starting dose in humans. For in vivo evaluations in animals, there are multiple approaches that can be used to facilitate i.v. administration and to improve exposure after oral dosing by the use of enabling formulations. However, there is always a risk that early incorporation of an enabling formulation approach may shift the focus away from structural optimization. Even in circumstances where lead optimization strategies are successful in identifying drug candidates with improved solubility properties, it is likely that at some stage during drug discovery there will be the need to assess less soluble early leads for intrinsic activity and proof of concept efficacy to justify compound or series progression.
A complication of poor aqueous solubility in the in vivo assessment of compounds throughout drug discovery and development is that compound supply is frequently limited (particularly in early discovery), and material that is available most likely will not have the final, or optimal, solid-state properties. As compounds progress through discovery and into development, the solid-state properties will almost certainly change; crystal forms will become better defined, particle size reduction may be introduced, and salt forms will likely become available for compounds with ionizable functional groups. The timing at which these changes occur during development will impact early preclinical data, and this needs to be appreciated and factored into study design and data interpretation.
A. Parenteral Administration
1. Complexities with Parenteral Administration of Poorly Water-Soluble Drugs
In comparison with the large number of drugs intended for oral administration, parenteral formulations constitute a more limited proportion of marketed products. However, the generation of useful parenteral formulations remains a key component of drug discovery and drug development for almost all drug molecules (regardless of the intended route of clinical administration) since data obtained after i.v. administration is necessary to generate fundamental pharmacokinetic parameters (volume of distribution, clearance, half-life, absolute bioavailability) to support compound optimization and progression, and such data are also useful to support early stage pharmacodynamic assessment of drugs intended for acute administration.
Low aqueous solubility significantly complicates the development of i.v. formulations since, in almost all cases, simple solution formulations are required. The following section describes several different approaches that can be used as i.v. solution formulations, depending on the physicochemical properties of the drug and the intended use (e.g., animal or humans). Depending on the formulation strategy adopted, increases in solubility of several orders of magnitude are possible. The risks associated with i.v. administration of poorly water-soluble drugs include, first, the potential for drug precipitation on administration and, second, the potential for formulation components to cause pain, phlebitis, inflammation, hemolysis, or unacceptable toxicology. Precipitation is more likely for formulations where pH adjustment or cosolvents form the basis for drug solubilization since both are likely to be altered significantly on dilution (see Sections IV and VI). In contrast, surfactant solubilization, complexation, and i.v. emulsions are far less sensitive to dilution (see Sections VII, VIII, and XI). Where dilution-mediated precipitation is possible, slow administration into large veins, where the blood flow is higher (and therefore the supply of plasma proteins and lipoproteins to provide in vivo binding is higher) is preferred, and this can also reduce the possibility of pain on injection, hemolysis, and phlebitis since the irritant component is maximally diluted. For studies in rats and dogs, this can be accomplished by implanting a dosing cannula into the jugular vein (or another large vein) with administration over a period of 5 to 10 min for rats and up to 30 min or longer in dogs. In contrast, administration into small veins in the extremities (where blood flow is much slower), such as the tail vein in rodent models, and rapid bolus injection, should be avoided with poorly water-soluble drugs, if possible, because of the potential for precipitation. Addition of small quantities of surfactants (<0.5%) or polymers may assist in preventing precipitation at the injection site.
Drug administration via other parenteral routes, including s.c., i.m., and i.p. administration, is on occasion warranted, particularly in proof-of-concept biology studies where there is evidence of poor oral exposure resulting from solubility limitations. With non-i.v. parenteral routes, the potential for the route and the formulation vehicle to have an effect (either desirable or undesirable) on the absorption profile is greater, and depot effects are often seen. For these administration routes, solution formulations are still preferred since the volume of fluid at the injection site is low, and, therefore, dissolution of suspension formulations may be limited. The lack of formulation dilution at the injection site dictates that the range of excipients and conditions to promote drug solubilization is even more limited than it is after i.v. administration. Ideally, non-i.v. parenteral formulations should be as close as possible to physiologic pH, and concentrations of cosolvents should be minimized.
2. Parenteral Formulation Approaches for Poorly Water-Soluble Drugs
The most common approaches for the development of parenteral formulations for poorly water-soluble drugs are summarized in the following discussion, and strategies for rapid identification of the most appropriate approach are discussed. The acceptable limits for each of these will depend on many factors including the species (animal or human), dose volume, rate of administration (e.g., infusion or bolus), parenteral route (i.v., i.m., s.c., i.p.), and duration of treatment (Gad et al., 2006; Li and Zhao, 2007). A summary of parenteral formulation approaches is given in Table 2, and these are briefly described as follows.
Potential hierarchy of formulation approaches for the development of parenteral formulations for poorly water-soluble drugs
Previously published suggestions for maximum allowable quantities provided in parentheses.a,b,c Further details are provided in individual sections. Note that common examples are provided, and this is not intended to represent an exhaustive list.
pH adjustment (see Section IV) is a powerful mechanism by which the solubility of weak acids and weak bases may be enhanced, but the approach is limited by the pKa of the compound and the acceptable pH range of the formulation. In this regard, acidic solutions are generally more readily tolerated than are basic solutions, and working pH ranges of 2–9 (Li and Zhao, 2007) or 4–9 (Lee et al., 2003) have been suggested. The pH of parenteral solutions can be manipulated via the addition of small quantities of strong acids or bases, such as HCl or NaOH but is more usefully achieved via the use of a buffer, generally with as low a buffer concentration as possible while still maintaining the desired pH. Common buffer systems used in parenteral formulations include phosphate and bicarbonate at neutral or slightly basic pH and citrate, lactate, tartrate, acetate, and maleate for more acidic solutions (Flynn, 1980; Kipp, 2007).
Cosolvents (see Section VI) are also commonly used to promote solubility in parenteral formulations, and ethanol, propylene glycol (PG), low-molecular-weight polyethylene glycol (PEG 300 or 400), dimethylacetamide (DMA), and DMSO have been used. Combinations of cosolvents provide benefits since cosolvency is additive, whereas toxicity is often dependent on the particular solvent.
In many cases, judicious manipulation of solution pH in combination with cosolvents is sufficient to provide solubilization and is typically viewed as the first-line approach. For example, in a review of 317 discovery compounds, Lee et al., (2003) reported that >90% of compounds could be formulated using pH and cosolvents and that >60% of these could be formulated using cosolvent levels of less than 50% v/v.
However, where pH adjustment and cosolvents fail to provide the required solubility, alternative approaches must be used. Second-tier approaches include the use of cyclodextrins, surfactants, and lipid emulsions and, ultimately, more complex formulations such as liposomes, microemulsions, or nanosuspensions. For more complex systems, however, the potential effect, either positive or negative, on systemic clearance and distribution (and also absorption for non-i.v. parenteral formulations) should be carefully considered.
Surfactants (see Section VII) can also be used to enhance drug solubility via micellar solubilization. Surfactants are generally limited to those that are nonionic because of their more favorable safety profile and may include polysorbates (e.g., Tween 80), Cremophors (including Cremophor EL and RH40), Solutol HS-15 (BASF Corporation, Washington, DC), vitamin E TPGS, and poloxamers (Pluronics, including Pluronic F68; BASF Corporation). Whereas surfactants provide for relatively robust solubilization, a complexity is the growing realization that many surfactants can affect transport processes, such as cellular efflux, and may also result in significant adverse effects, including hypersensitivity reactions (Gelderblom et al., 2001; ten Tije et al., 2003). Cremophor EL, in particular, seems to promote an immune reaction in some species (most notably dogs) when administered parenterally. Surfactants bring additional complexities to data interpretation and, where possible, might usefully be avoided in favor of pH changes, cosolvents, or the use of cyclodextrins.
Complexation agents (see Section VIII), such as cyclodextrins, provide a mechanism for enhancing solubility for many drugs and have been increasingly used as modified cyclodextrins, such as hydroxypropyl-β-cyclodextrin (HP-β-CD) and sulfobutylether-β-cyclodextrin (SBE-β-CD), have become more cost effective. HP-β-CD and SBE-β-CD have higher aqueous solubilities than do unmodified cyclodextrins (therefore allowing the support of higher concentrations of complexed drug) and, importantly, are becoming well characterized in terms of their safety profile. In rare situations where the binding constant between drug and cyclodextrin is particularly high (e.g., ∼106 M−1), there is the potential for alteration of the pharmacokinetic distribution and elimination processes (Charman et al., 2006), but most drugs exhibit significantly lower binding constants (<104 M−1) and are efficiently “released” from the complex via dilution and displacement by endogenous species (Stella and Rajewski, 1997; Stella et al., 1999; Stella and He, 2008). HP-β-CD and SBE-β-CD are commonly used in parenteral formulations in concentration ranges of up to 20 to 40% w/v in both humans and animals, and combining complexation with pH adjustment can, in some cases, provide additional solubility enhancement.
Intravenous emulsion formulations (see Section XI ) can be used for lipophilic, poorly water-soluble drugs that have high solubility in lipids. Typically, lipid emulsion formulations contain between 10 and 20% lipid, which limits drug load for compounds that do not have high lipid solubility. Intravenous emulsion formulations have been used for many years as nutritional supplements (e.g., Intralipid; Baxter Healthcare, Deerfield, IL) and are usually formulated using soybean oil as the dispersed phase and phospholipids as the emulsifier. Emulsions can be formulated de novo by dissolving drug into the oil phase of the emulsion before emulsification; however, this requires the use of specialized high-pressure homogenization equipment to provide emulsions with submicron particle sizes. Alternatively, drug may be incorporated into preformed commercial emulsions by dissolving the drug in a small volume of cosolvent with subsequent slow incorporation using sonication (El-Sayed and Repta, 1983).
More complex formulation approaches, such as the use of nanosuspensions (see Section IX), liposomes, or microemulsions, have all been used to facilitate parenteral administration but can substantially affect drug disposition. These approaches are better avoided when the intent of the formulation is to define fundamental pharmacokinetic parameters of a drug.
In many cases, combination approaches to solubilization in parenteral formulations are beneficial. The overarching premise for this approach is that the benefits of solubilization are often additive, whereas the effects on toxicity are typically a function of reaching a critical level for a single component. Maintaining concentration of one solubilizer below that threshold, but combining multiple approaches, therefore, may have benefit. In some cases, however, combination approaches add complexity without generating benefit or, indeed, are detrimental. Combination approaches are summarized in Table 3 and are further described in detail in each individual section of this review.
Summary effect of combining two common solubilization approaches for parenteral formulations
B. Oral Administration
In contrast to parenteral formulations, the approaches that are commonly used to support the oral delivery of poorly water-soluble drugs vary significantly depending on the purpose of the in vivo study, the species in which studies are conducted, the quantity of material available, and the level of exposure required. Distinction between the approaches taken at different stages of the discovery-development continuum is difficult, and different organizations will have different approaches depending on in-house experience and expertise. Nonetheless, the formulation strategies that are commonly adopted during discovery and development are summarized as follows.
1. Formulations to Support Drug Discovery
In proof-of-concept pharmacology studies, the focus is generally placed on generating sufficient exposure to allow early indication of the efficacy of a particular pharmacophore or compound series. At this stage, only small quantities of compound are usually available, and control of crystallinity is limited, with early batches often including a significant proportion of amorphous material. Before a salt screening program, ionizable compounds may also be available only as the free acid or base or potentially as a non-optimal salt form. Under these circumstances, simple solution formulations are preferred since they circumvent the complexities of dissolution and, therefore, obviate any variability in exposure resulting from differences in solid-state characteristics. Solution formulations facilitate ease and reproducibility of administration via oral gavage, enable ready conduct of single and multiple dose studies, and support dosing strategies such as cassette dosing. The approaches taken to develop oral solution formations are similar to those used for parenteral formulations, however, the acceptable limits on formulation components are typically wider. A useful order of potential formulation approaches to achieve simple oral solution formulations is provided in Table 4. In early stage proof-of-concept studies, relatively aggressive absorption enabling formulations may be used to overcome solubility and dissolution rate limitations, depending on the frequency and length of dosing; however, caution should be used to ensure that the formulation components do not confound the efficacy readout, and vehicle controls are essential.
Possible hierarchy of formulation approaches for the development of oral solution formulations
Previously published suggestions for maximum allowable quantities of formulation excipients provided in parentheses.a,b,c Exemplar formulations—for example, cosolvent combinations, surfactant formulations, and lipid-based formulations—are provided, realizing that they represent a limited “snapshot” of possible formulations, concentration ranges, etc. (Further details are provided in the sections of this review that describe each technology.)
Oral delivery during lead optimization is typically focused on prioritizing lead compounds from within a series with more favorable absorption properties and on identifying the key structural and physicochemical factors that contribute to poor oral bioavailability, in an effort to guide subsequent structural modification. Understanding the basis for poor oral absorption is fundamental to designing a strategy to overcome this limitation. The approaches used for oral formulations of poorly water-soluble compounds in lead optimization are therefore usually less complex than those incorporated, for example, during discovery biology, and are designed to discriminate between compounds in the absence of enabling formulation effects. Consistency in formulation is important to distinguish between potential effects resulting from the formulation and intrinsic differences in structural and physicochemical properties of the lead.
Simple suspension formulations containing a suspending agent (e.g., cellulose derivatives) along with a low concentration of surfactant to reduce particle agglomeration are often used in early bioavailability studies (see Table 5). These formulations provide little in the way of “assisted” dissolution and therefore tend to discriminate between different compounds. A further advantage is that they can be readily extended into animal toxicology studies, provided sufficient compound exposure can be achieved. A complexity in the use of suspension formulations is the potential impact of particle size on dissolution rate and the possibility of batch-to-batch variability given that solid-state properties and particle size are not likely to be optimized at the time the initial pharmacokinetic and bioavailability studies are conducted. Assessment and monitoring of particle size are beneficial, and normalization of particle size across batches via laboratory scale milling may provide greater consistency.
Typical suspension formulations
Another approach is to use simple solution formulations, typically prepared using cosolvents with or without pH manipulation. These formulations have the advantage of being independent of solid-state properties since compounds are in solution but have the disadvantage of likely precipitation after administration, particularly if the formulations prepared using cosolvents are near the saturation solubility limit. Generally, more specialized enabling formulations, such as surfactant-, cyclodextrin- and lipid-based formulations, are not ideal for lead optimization in that they may mask intrinsically poor physicochemical properties and take the focus away from structural optimization to improve solubility. An early reliance on the use of enabling formulations also complicates subsequent developmental activities.
As larger quantities of drug become available, more definitive information regarding solid-state properties is generated and salt screening programs are initiated for weak acids and bases (see Section IV). Isolation as a salt form can provide for significant enhancements in dissolution rate. The availability of larger quantities of material also allows more complex particle size reduction strategies to be explored including micronized formulations and nanosuspensions (see Section IX).
2. Use of Enabling Formulations to Promote Oral Absorption
In situations where attempts to increase polarity and water solubility lead to significant decreases in potency, and where solubility or dissolution has been identified as the rate-limiting process for absorption, a strategic decision may be made to progress a compound series via the use of enabling formulations to improve bioavailability and exposure. This approach may be taken in an effort to obtain rapid proof-of-concept data in humans against a new target or simply to enable early pharmacokinetic and safety data to be obtained for a novel compound series.
The preclinical assessment of more specialized oral formulations will depend heavily on the type of formulation being assessed. Generally, rodents require liquid formulations that can be easily administered via a gavage tube. Recent developments in the availability of minicapsules and tablets allows dosing of tablets or encapsulated materials to rodents; however, the requirement for specialized equipment to prepare these dosage forms and the low liquid volumes in the rodent GI tract (which may limit the rate of capsule/tablet disintegration) make this approach less common.
Alternate crystal forms such as cocrystals, polymorphs, or amorphous drug may explored, although the inherent instability of metastable crystal and amorphous forms dictates the need for close monitoring of crystallization properties, and formulation approaches to stabilize against solid-state transitions may be necessary, depending on the required stability period. Cocrystals provide some advantage in this respect as their crystal lattice energy is significantly higher than amorphous drug, allowing the generation of formulations with improved stability. Stabilization of polymorphs or amorphous drug may be achieved by the use of (usually polymeric) solid dispersion formulations, where reduced molecular mobility, often achieved via intermolecular interactions between carrier and drug, reduces the risk of changes to the amorphous-crystal structure during storage. Solid dispersions require relatively complex equipment to provide clinical formulations, such as spray drying or hot melt extrusion; however, trial batches can be formulated using simple benchtop equipment such as a rotary evaporator or freeze-driers.
As drug candidates progress through development, preclinical investigations in nonrodent species will be conducted, usually in dogs or nonhuman primates. The physical form of materials used in these studies will vary, and simple solutions or suspensions will often still be used. Larger species generally provide the first opportunity to assess the performance of prototype tablet and capsule formulations that might ultimately progress to the clinic. For ease of preparation on a relatively small scale, hand filling of dry powder blends or granules into hard gelatin capsules or liquid filling into preformed soft gelatin capsules can be used. Sealed hard-gelatin capsules provide an alternative for liquid fills and may be sealed by hand or, more effectively using specialized capsule-sealing equipment, realizing that such equipment may not be readily available at this stage. For poorly water-soluble drugs, the shift to encapsulated and solid dosage forms dictates renewed concentration on solid-state formulation strategies such as salts, cocrystals, and particle size reduction alongside more complex formulation approaches, including solid dispersions and lipid-based formulations.
3. Preclinical Toxicology Formulations
Preclinical toxicology evaluation is a particular challenge for poorly water-soluble drugs. The need to escalate doses to the point where in vivo exposure is sufficient to demonstrate limiting toxicity generally requires absorbed doses (in milligram per kilogram terms) 30 to 100 times greater than might be envisaged with a clinically relevant dose and formulation. Where water solubility is low, this is often impossible with solution or suspension formulations, since beyond a certain point, drug exposure remains constant regardless of increases in dose, dictating the need for more complex formulation approaches; however, this requirement is tempered by potential concerns regarding the toxicology of the large quantities of excipient often required to afford the higher exposure. Relatively little information is publically available that describes the quantities of different combinations of excipients that do not lead to toxic endpoints over a range of dosing periods. One notable exception is an excellent review by Gad et al. (2006), which provides a summary of the effects of a range of excipients in toxicity testing protocols of different lengths. Neervannan (2006) also provide some data regarding the maximum quantities of common excipients that might be safely used in acute and subchronic dosing protocols for up to 7 days. However, these communications provide data on a limited number of surfactants, lipids, and polymers and do not address the potential toxicity of the combinations of components often used.
Toxicity concerns for many of the excipients used in more complex formulations are attenuated by the realization that many polymers and surfactants have high molecular weights and are relatively polar, or amphiphilic. As such, the extent of absorption of these excipients after oral administration is low. Many surfactants are also digestible (Cuiné et al., 2008). Under these circumstances, the risks of systemic toxicity are low, and the oral LD50s of most hydrophilic polymers and nonionic surfactants are high (Rowe et al., 2009). The major concerns therefore revolve around the possibility of GI side effects, including loose stools and emesis (Reppas et al., 1991; Andersen, 1994; Schulze et al., 2005; Ruble et al., 2006). By incorporation of vehicle controls, many of these effects can be accounted for during data analysis. Confounding data interpretations issues arise, however, where GI symptoms manifest as a consequence of the candidate drug’s desired biologic activity or toxicity profile.
Cosolvents, in particular ethanol, typically exhibit more significant toxicity profiles and, hence, cosolvent combinations are commonly used to maintain the concentration of one single component at a relatively low level. Finally, several surfactants have also been shown to inhibit efflux transporters (Batrakova et al., 1999; Yamagata et al., 2007a) and GI cytochrome P450 enzymes (Wandel et al., 2003; Bravo González et al., 2004; Christiansen et al., 2011) and may therefore affect drug bioavailability after oral administration. Evidence of systemic effects, however, is limited except after parenteral administration. After parenteral administration, surfactants such as Tween 80 and Cremophor EL may stimulate hypersensitivity reactions (Weiss et al., 1990; Gelderblom et al., 2001; ten Tije et al., 2003), especially in dogs (Lorenz et al., 1982).
4. Development of Clinical Formulations for Poorly Water-Soluble Drugs
The strategies used when developing clinical formulations typically fall into two categories. The first strategy is the development of simple formulations that maximize the speed to first-in-human studies but provide no functional link to the likely formulations used in later stage clinical trials and that might be intended for commercialization. Second is the development of mature formulations, with the expectation of progressing essentially similar formulations through clinical development to commercialization. Where possible, the former approach uses nonformulated drug to simplify dosage form development, and, where solubility permits, simple direct filling of drug in capsules or drug-in-bottle approaches can be used. The relatively recent application of direct capsule-filling instruments such as Xcelodose (Capsugel, Greenwood, SC) that facilitate accurate filling of doses ranging from 100 μg to 100 mg have greatly simplified this process.
For many poorly water-soluble compounds, however, low solubility limits the possibility of adopting an accelerated route to first-in-human studies, and in these instances, more sophisticated formulations are required to ensure exposure. Formulations with the potential to progress through clinical trials, thereby circumventing the need for repeated bioequivalence bridging studies, are typically evaluated. The escalation of formulation complexity in this case (Table 6) commonly follows a pathway similar to that previously described for preclinical formulations, although in this case, the potential to progress through to full scale tableting or encapsulation methods and the use of Good Manufacturing Practice processing equipment becomes more important. Where simple solid-state or particle size approaches to enhanced solubility such as salt formation, cocrystals, or nanocrystals fail to provide sufficient exposure, the major routes of progression are the use of lipid-based formulations or solid dispersions. The choice of approach is dictated largely by the physicochemical properties of the drug candidate, where lipid-based formulations lend themselves to poorly water-soluble, lipophilic drugs (i.e., those with sufficient solubility in lipids, surfactants, and cosolvents to allow the target dose to be dissolved in the capsule fill material), whereas solid dispersions are commonly explored for drug candidates with insufficient lipophilicity to allow the use of lipid-based formulations.
Formulation approaches for the development of solid oral formulations
In conclusion, the general strategies that may be taken to identify and overcome the challenges of low water solubility in the drug discovery and development environment are summarized in the proceeding discussion. The tools that have been suggested as approaches to overcome the limitations of poor solubility are described in detail in following sections.
IV. Buffers and Salt Formation
Adjusting solution pH, usually via the use of a buffer, provides an effective means of increasing the proportion of a weakly acidic or basic drug that is present in the ionized form. In turn, increasing ionization increases polarity and therefore increases drug solubility in polar (aqueous) solutions. Salt formation results in a similar endpoint, and in many ways salts of weak acids and bases are the solid-state equivalent of pH adjustment. Salts are formed via an ionic interaction between weakly acidic or basic drugs and an oppositely charged basic or acidic counterion. When salts are subsequently allowed to dissolve and dissociate in water, the acidic or basic counterion from which they were derived causes a shift in the pH of the solution to provide an analogous endpoint to that obtained by pH adjustment. Isolation of drugs as a particular salt form also changes the nature of the crystal lattice, resulting in differences in dissolution rate from alterations in solid-state properties compared with the free acid or free base (Huang and Tong, 2004; Corrigan, 2007).
Diclofenac provides a good example of the potential advantages of salt formation. In the un-ionized form, diclofenac has a low aqueous solubility (<0.02 mg/ml) (Fini et al., 1999), whereas the sodium or potassium salt forms of diclofenac are >400 times more water soluble (Fini et al., 1996). Similar trends are also evident with weak bases (which make up the larger proportion of ionizable drug candidates); for example, the aqueous solubility of enalapril maleate is 25 mg/ml (Kasim et al., 2004), whereas that of the free base is ∼0.21 mg/ml. In unbuffered solutions, salts therefore provide an opportunity for significant increases in dissolution and solubility. In contrast, in buffered solutions that favor the un-ionized free acid or free base, the aqueous solubility advantages of pharmaceutical salts are attenuated, although significant increases in dissolution rate are still evident since the pH in the unstirred water layer reflects the pH of the dissolving salt rather than the bulk (buffered) pH (Serajuddin and Jarowski, 1985b; Li et al., 2005b; Serajuddin, 2007).
Exploiting the enhanced solubility and dissolution rate of the ionized form of a drug, either through pH adjustment or salt formation, is therefore a common approach for oral and parenteral routes of drug delivery and should be regarded as a primary strategy for addressing low aqueous solubility (Berge et al., 1977; Stahl and Wermuth, 2002; Tao et al., 2009). Although this approach is applicable only to ionizable compounds, weak acids and weak bases account for up to two-thirds of marketed drugs and drugs in development (Stahl and Wermuth, 2002); consequently, the strategies described here are relevant across a wide range of drug candidates.
In this section, the fundamental principles of ionization potential and the solubility of weak electrolytes and their respective salts are described. Subsequently, the formulation considerations when using buffered systems and salts for parenteral delivery and oral delivery are addressed before finally providing an overview of common salt-screening strategies.
A. Solution Behavior of Weak Electrolytes and Their Salts
1. Ionic Equilibria
On addition to water, strong electrolytes such as hydrochloric acid and sodium hydroxide dissociate almost completely. In contrast, weak electrolytes, such as the weakly acidic and weakly basic molecules that constitute most ionizable drugs, dissociate only partially at most pH values. For a weak acid or weak base, the total concentration in solution at a given pH is therefore the sum of the concentrations of the ionized and un-ionized species. The degree of dissociation into the ionized and un-ionized form is described by the dissociation constant (Ka) for the equilibrium reactions shown in eq. 12 for a monoprotic weak acid (HA) (left equation) and weak base (BH+) (right equation) in water, with the associated expressions for Ka shown in eq. 13.(12)
(13)
As the dissociation constant may vary by several orders of magnitude, the pKa or negative logarithm of Ka (pKa = −log Ka) is more typically used to compare acids and bases (Table 7). Decreasing pKa denotes stronger acids (i.e., species that more readily donate a proton), whereas increasing pKa reflects stronger bases (i.e., species that more readily accept a proton).
Relative acidity/basicity of molecules with varying pKa values, and approximate pKa values for common functional groups
The Henderson-Hasselbalch equation (eq. 14) further describes the pH dependency of the ionization properties of weak acids (left equation) or bases (right equation) and relates the degree of ionization to the pH of a solution and the pKa of the drug.(14)
From eq. 14, it is apparent that under conditions where the pH of a solution is the same as the pKa of a dissolved weak acid or base, the concentrations of ionized and un-ionized species are equivalent. A change in pH by one log unit away from the pKa subsequently results in a 10-fold change in the degree of ionization.
2. pH Solubility Relationships for Weak Electrolytes and Salts of Weak Electrolytes
The intrinsic solubility of a nonelectrolyte is governed by the relative strengths of the intermolecular forces in the solute (i.e., its solid-state properties) and the affinity of the solute for the solvent (i.e., the strength of solute-solvent interactions) (see Section I.B). This is also the case for a weak electrolyte but is complicated by differences in ionization of the solute with changes in pH that, in turn, alter polarity and the affinity of the solute for the solvent. Thus, a weak electrolyte will initially dissolve to an extent governed by the intrinsic solubility of the un-ionized form. Depending on the pH, a proportion of the dissolved species subsequently ionizes in accordance with the Henderson-Hasselbalch equation (eq. 14), leading to further dissolution of solid drug to maintain a constant concentration (i.e., the saturated solubility) of the un-ionized form. This will continue within an unbuffered solution until the saturated solubility of both un-ionized and ionized species is reached. As the ionized species is able to form favorable ion-dipole interactions with polar solvents, it is more soluble than the un-ionized species, and therefore, drugs are more soluble in polar solvents at pH values that favor the ionized form. The total solubility (S) of a weak acid (HA) or weak base (B) at a given pH is therefore the sum of the solubility of the ionized and un-ionized forms (Yalkowsky, 1999).(15)
Combinations of eq. 14 and eq. 15 generate the following solubility equations for weak acids (left, eq. 16) and weak bases (right, eq. 16) where total drug solubility at a particular pH is a function of the solubility of the un-ionized form (S0) and the pKa of the weak acid or base.(16)
From eq. 16, it is evident that for a monoprotic weak acid or base, changes in pH of 1 unit away from the pKa lead to 10-fold (1 log unit) differences in solubility. For diprotic species, similar relationships may be developed (eq. 17) where total solubility is dependent on the pKa of two ionization centers (Yalkowsky, 1999). Accordingly, diprotic species are expected to show a 100-fold (2-log unit) change in solubility with a change in pH of one unit (Garren and Pyter, 1990; Venkatesh et al., 1996). The top equation is for a weak diprotic acid, and the bottom equation is for a weak diprotic base:(17)
The solubility equations for weak electrolytes shown in the preceding should be viewed with two significant caveats. First, eq. 16 and eq. 17 suggest that drug solubility changes indefinitely with changing pH. Ultimately, however, the achievable solubility is regulated in most cases by the presence of counterions that may be intentionally present (in the case of a salt) or unintentionally present (in the case of ions present in blood, GI fluids, buffers, and so forth). In this case, the total solubility is regulated by the limit of solubility of the ionized weak acid or base in the presence of the corresponding basic or acidic counterion, ultimately resulting in precipitation of the solid salt of the acid-base pairs. The solubility of the salt is therefore the limiting condition to the aqueous solubility of drug in the ionized form when a counterion is present. Where multiple counterions are present, the limiting salt solubility typically reflects the salt with the lowest solubility, or the salt formed with the counterion in highest concentration if common ion effects are significant. Salt solubility, including aspects of common ion effects, is described in more detail in Section IV.4. Second, classic ionization texts usually ignore the possibility of ion-pair formation or self-association. Where ion-pair formation or self-association leads to the formation of supramolecular complexes in solution (including multimers, micelles, and liquid crystalline species), apparent solubility values may be much greater than anticipated (Serajuddin et al., 1987; King et al., 1989; Kriwet and Muller-Goyman, 1993; Fini et al., 1995).
Exemplar pH solubility profiles for a weak acid and a weak base, and the salts formed by association with a corresponding basic or acidic counterion, are presented in Fig. 7. The total drug solubility (S in eq. 16) of a weak base is low at high pH where the un-ionized form (B) predominates (region 1 of the pH solubility profile for a weak base). Where the solubility of the ionized drug species (BH+) is significantly in excess of that of the un-ionized form, B (which is typical), decreasing pH results in an increase in the proportion of BH+ and an increase in total solubility. The increase in solubility becomes particularly significant from pH values of approximately 1 pH unit above the pKa (i.e., where the degree of dissociation increases beyond ∼10%) to pH values below the pKa where, as described previously, solubility increases 10-fold for every unit pH decrease (for a weak base). At a critical point (and critical pH; denoted pHmax), the equilibrium solubility of the ionized species (as the salt) is reached, and no further increases in solubility are seen with decreasing pH. This limiting solubility is set by the solubility product of the salt (Ksp):(18)
When the concentration of solid phase (i.e., salt) is in excess and, therefore, approximately constant:(19)
In region 2 of Fig. 7 (i.e., at the solubility limit), excess solid phase is present as the salt, and the saturated species in solution is largely the ionized form. At pH values <pHmax, the total solubility of a weak base is given by [BH+] plus a small (and decreasing) quantity of B (free base) (Kramer and Flynn, 1972; Serajuddin, 2007). The total solubility in region 2 of Fig. 7 is given by eq. 20. for weak acids (top equation) and weak bases (bottom equation).(20)
The pH solubility profiles for (A) a weakly basic and (B) a weakly acidic compound. In region 1, the un-ionized free acid or free base is present as the solid phase and as the saturation or equilibrium species in solution. Total solubility is a function of both un-ionized and ionized species (eq. 16), and at pH values greater than (for a weak acid) and less than (for a weak base) pKa, solubility increases because of increased quantities of the ionized species in solution. Solubility is ultimately limited in the “salt plateau” region (region 2) by Ksp (eq. 19). In region 2, the salt is present as the solid phase, and the ionized form is the saturated or equilibrium species in solution. At pH values greater than (for a weak acid) and lower than (for a weak base) pHmax, the total solubility decreases slightly because of a reduction in the quantity of free acid or free base in solution (eq. 20). Where pH change continues as a result of the further addition of acidic or basic counterion, salt solubility in region 2 may decrease much further (dotted line) as a result of the common ion effect.
Scheme 1 summarizes the solid-solution equilibria for a weak base (B), where the counterion is provided by a strong acid (HX) to form a salt (BH+X-)solid. This scheme brings together the relationships between dissociation (described by Ka and the dominant factor in region 1 of Fig. 7) and the equilibrium solubility of the salt (described by Ksp and the dominant factor in region 2 of Fig. 7). In some cases, additional equilibria, including the potential for hydrate formation or self-association, may be present but have been omitted here for the sake of clarity.
Phase solubility scheme for salt formation between a weak base (B) with a strong acid (X-). Adapted from Pudipeddi et al. (2002).
In a solution of a monovalent salt, dissociation leads to equal concentrations of [BH+] and [X−]. From eq. 19, the maximum solubility of [BH+] (i.e., the limiting value for salt solubility) is equal to the square root of the solubility product (√Ksp). Ksp is a constant for a particular salt and defines the maximum concentration of drug ions and corresponding counterions that a solvent can maintain in solution (Bhattachar et al., 2006). Essentially, the higher the Ksp value, the greater the solubility of the salt. Ksp is therefore a critical parameter in understanding the solubility of salts or the solubility of ionized drugs where salt formation in situ is likely. Unfortunately, techniques that accurately predict Ksp do not exist, largely as a result of the unpredictability of the nature of the crystal lattice. Screening programs that are able to rationally and rapidly assess the limit of solubility of different salt forms are therefore critical to support effective development programs, and are described in more detail in Section IV.E.2.
3. Factors Affecting Salt Formation at pHmax
The pHmax defines the limiting pH above which (for a weak acid) and below which (for a weak base) the solid phase constitutes the salt rather than the free acid or free base. To facilitate salt formation, the bases or acids that are used to provide the counterion must be sufficiently strong to shift solution pH past pHmax (into region 2 of Fig. 7). In general, salts of weak bases will form more readily with more strongly acidic counterions such as hydrochloride compared with more weakly acidic counterions such as citrate. As a general rule, salt formation will occur only between a weak acid or weak base and counterion if the difference in pKa for the two is greater than 2 (Pudipeddi et al., 2002; Stahl and Wermuth, 2002; Black et al., 2007). It is also evident that conditions that cause a shift in pHmax will change the ease of salt formulation. Thus, conditions that result in an increase in pHmax for a weak base and a decrease in pHmax for a weak acid will assist in salt formation by reducing the pH shift that must be generated by the counterion to move into region 2 of Fig. 7. For example, decreasing the pHmax for a weak acid will allow salt formation with a more weakly basic counterion, thereby increasing the possible choices of counterion.
At the pHmax value, the salt and the free acid or free base are present in the solid state, and this situation is unique to this transition point in the pH-solubility profile. Under these circumstances, eq. 16 and eq. 20 are both applicable. Setting eq. 16 equal to eq. 20 and solving for pH (i.e., pHmax) results in eq. 21 and a relationship that provides an indication of the dependence of pHmax on pKa, the intrinsic solubility of the un-ionized free acid or free base (S0) and Ksp (Bogardus and Blackwood, 1979; Serajuddin, 2007). Note that the equation on the left applies for weak acids while the equation on the right applies for weak bases:(21)
From eq. 21, it is clear that the value for pHmax is dependent on the properties of the drug namely, intrinsic solubility (S0), relative acidity/basicity (pKa) and the solubility of the salt (Ksp). The effect of changing these three terms on pHmax is illustrated for a typical weak base in Fig. 8. Increasing the pKa (i.e., increasing basicity) (Fig. 8A), increasing S0 (i.e., increasing intrinsic solubility) (Fig. 8B), and decreasing Ksp (i.e., forming salts with lower solubilities), all assist salt formation by increasing pHmax (realizing that lower solubility salts may not be preferred in many cases).
The effect of (A) pKa, (B) intrinsic drug solubility (S0) and (c) salt solubility (Ksp) on the position of the pHmax of a weak base. Salt formation of a weak base becomes more attainable (an increase in pHmax) with increasing pKa of the base, an increase in intrinsic solubility, or a decrease in salt solubility. Adapted from Pudipeddi et al. (2002).
4. Determinants of Salt Solubility
The maximum solubility of a salt is the boundary condition for in vivo solubility and dissolution and, therefore, the parameter against which alternate salts are screened (at least for their impact on solubility). The accurate determination of salt solubility, however, is not a trivial task due to the potential for interconversion between the salt form and the free base or free acid (see Section IV. D.1.c), and the impact of a number of system variables. These are described below.
a. pHmax
To ensure that solubility data reflect the equilibrium solubility of the salt, measurements must be taken at pH values that ensure that the solid phase in equilibrium with solution is the salt and not the free acid or base. As such, solubility measurement should be taken at pH values above (for weak acids) or below (weak bases) the corresponding pHmax. If this condition is not met, the solution is not saturated with respect to drug in the ionized form, leading to an underestimation of the saturated salt solubility. It is advisable during solubility measurements to determine both the pH of the solution and to characterize the nature of the solid phase (e.g., using XRPD). The presence of salt alone in the analyzed solid provides evidence of location within region 2 of the pH-solubility phase diagram (Fig. 7) and, therefore, that the data obtained reflect the true solubility of the salt. Conversely, the presence of free acid or free base suggests that insufficient salt has been added to reach the pHmax and that the system has equilibrated in region 1 (Alsenz and Kansy, 2007; Hasa et al., 2011). Under these circumstances, the addition of larger quantities of salt is required to provide further counterion, which in turn results in a pH change (Pudipeddi et al., 2002). Typically, a relatively high excess of salt is required to ensure that the pH approaches pHmax. Determination of the solubility of a salt in a buffered solution will reveal only the solubility of the un-ionized plus ionized form of the drug at that particular pH, and where the pH is above pHmax (for a weak base) or below pHmax (for a weak acid), the solubility of the drug is dependent only on pH and the solubility of the ionized form (eq. 16) and is independent of the counterion. This provides no indication of Ksp, and therefore, no indication of the solubility limit for the salt. The reader is directed to a recent article by Murdande et al. (2011a)for a more detailed description of the factors to be considered when measuring the equilibrium solubility of crystalline solids (and applicable here to salts).
b. Choice of Counterion to Maximize Salt Solubility
Table 8 provides a list of common counterions that have been used to isolate salts of weakly acidic and weakly basic drugs for the development of oral and parenteral drug products. An indication of the frequency of use is also provided (Paulekuhn et al., 2007). As Ksp is counterion specific, the solubilities of salts formed with a variety of different counterions can vary significantly. For example, solubility differences of up to 100-fold have been described for a series of amine salts of diclofenac (O’Connor and Corrigan, 2001), and differences in solubility of up to four orders of magnitude have been described for salts of ibuprofen (David et al., 2012). Similarly, for the weak base enalapril, salts formed with a range of aliphatic or aromatic carboxylic acids exhibit solubilities ranging from 22 mg/ml up to 350 mg/ml (Kumar et al., 2008a).
List of acidic and basic counterions commonly used in pharmaceutical formulations
A better understanding of the factors that affect salt solubility (and therefore counterion selection) may be gained by examination of the determinants of the molar free energy of solution (ΔGsoln) (Chowhan, 1978):(22)
(23)
Solubility is a function of the free energy gain achieved by solvation of the anion and cation (eq. 23) offset by the free energy of the crystal lattice (eq. 22). Salt formation has the capacity to alter both the free energy of the crystal lattice (ΔGlattice) (by changing the nature of packing and the strength of intermolecular interactions in the solid state) and the degree of solvation (ΔGsolvation). Solvation is typically enhanced by salt formation since dissociation into ions facilities the formation of ion-dipole interactions with water molecules that are more energetically favorable than, for example, hydrogen bond interactions between water and un-ionized drug. In contrast, the crystal lattice energy is typically increased by salt formation via the generation of stronger intramolecular ionic interactions, and this is usually evident in an increase in melting point (Corrigan, 2007; David et al., 2012). From eq. 22, the increase in (ΔGlattice) is expected to be detrimental to solubility. However, the higher solvation energies resulting from ionization typically offset the energy cost of breaking ionic interactions in the crystal lattice, leading to a net increase in solubility for salts compared with the free acid or base.
The importance of solvation of both anion and cation to the overall solvation energy dictates that hydrophilic counterions, by virtue of higher solvation energies, might be expected to form more soluble salts than hydrophobic counterions (Menegon et al., 2011; David et al., 2012), and indeed this is often the case. However, the choice of counterion affects both solvation and lattice energy, and as such, definitive structure-solubility relationships for differing counterions do not exist (Stahl, 2003; Corrigan, 2007; Serajuddin, 2007). Thus, monovalent and divalent alkali metals (K+, Na+, Mg2+, Ca2+) are widely used counterions for salt formation for weakly acidic drugs (Table 8), and increasing valency and decreasing counterion ionic radius might be expected to increase hydrophilicity and facilitate solvation. However, salts of monovalent and larger cations (of equivalent valency) are typically the most soluble (Chowhan, 1978; Anderson and Conradi, 1985; Forbes et al., 1995). For example, Fig. 9 depicts the solubility of metal salts of 7-methylsulfinyl-2-xanthonecarboxylic as a function of pH (Chowhan, 1978) and illustrates that maximum salt solubility is evident for larger monovalent counterions [i.e., K+ (most soluble) > Na+, > Ca2+ > Mg2+]. These trends suggest that counterion effects on lattice energy (where larger, less highly charged species reduce intermolecular attractive forces and typically result in lower crystal lattice energies) are more critical determinants of the solubility of a salt than corresponding effects on solvation (where higher-charge densities are expected to increase solvation). Hydrophilic counterions may therefore show higher solvation energies but result in lower solubility (compared with a less hydrophilic counterion) by virtue of higher lattice energies. For example, the lactate and mesylate salts of the experimental antimalarial drug [α-(2-piperidyl)-3,6-bis(trifluoromethyl)-9-phenanthrenemethanol] are more soluble than salts formed with the freely soluble hydrochloride counterion (Agharkar et al., 1976), and saccharin salts of lamotrigine are more soluble than those formed with the more hydrophilic succinic acid and fumaric acid counterions (Galcera and Molins, 2009). Kumar et al. (2008a) have also shown that malate salts of enalapril are more than 10 times more soluble than maleate, besylate, and tosylate salts, despite each of the counterions showing very high aqueous solubilities. Finally, the solubilities of 22 salts formed between various p-substituted benzoic acid derivatives and benzylamine have been shown to vary significantly, despite broadly similar energies of solvation (Parshad et al., 2002). Across different salt series, lattice energy effects on solubility therefore appear to dominate over solvation effects in many cases.
The pH-solubility profiles for 7-methylsulfinyl-2-xanthonecarboxylic acid and its salts at 25°C. Symbols represent experimental data. The point at which horizontal lines cross the upward solubility curve represent the pHmax values for respective salts. Solid symbols indicate that the starting material was the acid, and open symbols indicate that the starting material was the salt. Adapted from Chowhan (1978).
Consistent with the potential importance of differences in lattice energy on salt solubility, several authors have described correlations between the melting point of pharmaceutical salts (as a measure of lattice energy) and saturated aqueous solubility (Anderson and Conradi, 1985; Gould, 1986; Thomas and Rubino, 1996; Ledwidge and Corrigan, 1998; O’Connor and Corrigan, 2001; Guerrieri et al., 2010). Figure 10, for example, shows the relationship between melting point and solubility for a range of flurbiprofen salts formed with different organic and inorganic amine cations and provides good evidence of a relationship between decreasing melting point and increasing solubility (Anderson and Conradi, 1985). The properties of the counterion that affect packing within the salt lattice, and therefore the lattice energy, include symmetry and size, the ability to form hydrogen-bonds with the drug (in addition to ionic interactions), and the capacity to delocalize charge (Chowhan, 1978; Gould, 1986; Parshad et al., 2004). For example, salts of the weakly basic drug candidate (UK-47880) formed with planar and aromatic acids were found to be highly crystalline and to have higher melting temperatures compared with an oily, low melting salt formed with two aliphatic and more flexible carboxylic acids (citric acid and dodecyl benzene sulfonic acid) (Gould, 1986). Malate and maleate salts of ephedrine also show higher melting temperatures compared with fumarate and tartrate salts (Black et al., 2007). This has been suggested to reflect the small size of the malate counterion and the symmetrical nature of the maleate counterion, both factors that favor hydrogen bonding within the salt lattice such that the lattice energy is increased (Gould, 1986). Similarly, diclofenac salts formed with the freely soluble tris(hydroxymethyl)aminomethane (TRIS) counterion were found to be less soluble than expected, a situation later ascribed to the ability of TRIS to hydrogen bond within the salt lattice (Fini et al., 1996; O’Connor and Corrigan, 2001). In this example, the TRIS salt had the highest melting temperature and lowest solubility of a range of diclofenac salts formed with organic amine counterions and therefore provides another example of the potential for more polar counterions to result in lower salt solubilities via stronger packing within the crystal lattice (Corrigan, 2007). In contrast, increasing the alkyl chain length of amine counterions led to a decrease in the solubility of salts of several weak acids (ibuprofen, gemfibrozil, etodolac), although increasing the alkyl chain length on the counterion lowered the salt melting temperature (David et al., 2012). The overall effect of counterion polarity on solubility therefore reflects the net effect of changes to both lattice energy (where more polar counterions typically increase lattice energy, decreasing solubility) and hydration (where increasing counterion polarity typically increases hydration and increases solubility).
The relationship between log Ksp and the melting temperature of 1:1 amine salts of flurbiprofen. Basic counterions were 1) = 2-amino-2-methyl-1,3-propanediol, 2) = ammonium, 3) = tromethamine, 4) 2-amino-2-methylpropanol, 5) = tert-butylamine, and 6) = adamantanamine. Adapted from Anderson and Conradi (1985).
Interestingly, mesylate (methanesulfonate) salts appear to be finding increasing favor for the delivery of weak bases both orally and parenterally (Paulekuhn et al., 2007; Elder et al., 2010; Branham et al., 2012). The wider use of the mesylate counterion in salt selection strategies for solubility enhancement may be in part attributed to its high-charge density but also to its retention of a degree of hydrophobicity, which appears to affect molecular packing in the crystal lattice. As a result, salts formed with the mesylate counterion are likely to show high solvation energies compared with salts formed with other organic counterions (because of the high-charge density) but lower lattice energies compared with salts formed with inorganic counterions (which also show a high charge density). For example, mesylate salts of α-(2-piperidyl)-3,6-bis(trifluoromethyl-)9-phenanthrenemethanol(Agharkar et al., 1976), RPR200765 (Bastin et al., 2000), and haloperidol (Li et al., 2005b) all showed lower melting temperatures compared with equivalent hydrochloride salt forms. The mesylate ion also has a very low pKa (the lowest of all common organic acidic counterions) (Table 8), in turn facilitating salt formation with weak and very weak bases (Stahl, 2003). Mesylate salts are also less susceptible to the common ion effect in vivo, and this is described in more detail in Section IV.D.1.d.
Finally, the choice of counterion requires appreciation of the impact of the counterion on a range of pharmaceutical factors, including solubility, but also on other properties, such as flow, compression, and stability. Currently, the ability to predict differences in the solid-state properties of salts isolated with differing counterions is limited by a lack of understanding of the factors that control packing arrangements during crystallization and the potential for ionized drug molecules to self-associate in solution. This in turn limits accurate de novo prediction of counterion effects on solubility. Consequently, it is more typical to select the most appropriate salt via a salt-screening process. Salt screens are described in more detail in Section IV.E.2.
c. The Effect of Common Ions on Salt Solubility
The solubility of a salt is highly sensitive to the presence of common ions. This effect can be rationalized according to the theoretical descriptions of salt solubility as described in Section IV.A.3, in particular eq. 19 and Scheme 1, where it is apparent that excess counterion will result in suppression of salt solubility to maintain Ksp (Berge et al., 1977; Streng et al., 1984; Serajuddin, 2007). The presence of common ions has frequently been shown to reduce salt solubility in vitro (Miyazaki et al., 1980; Streng et al., 1984; Fini et al., 1995; Wang et al., 2002; Bergstrom et al., 2004; Li et al., 2005a; Larsen et al., 2007), and in some cases common ions can suppress the solubility of a salt below that of the free base/acid (Li et al., 2005a). The common ion effect also has potential implications for parenteral formulations where ions present in various additives (e.g., buffers, salts) are common to those in the salt. Common ion effects are more critical for drugs of low solubility where small changes to common ion concentrations can lead to relatively large compensatory changes to drug concentrations (Serajuddin, 2007).
Effects in vivo are also evident where salts come into contact with high concentrations of common ions. The most familiar example is that of changes to the solubility and dissolution rate of hydrochloride salts in the regions of the GI tract that contain endogenous chloride ions, such as the stomach (from acid secretion) and the intestine (due to the presence of sodium chloride) (Miyazaki et al., 1981; McConnell et al., 2008), but this effect is also evident for sodium salts of weak acids where concentrations of endogenous sodium ion are high. The use of counterions other than hydrochloride or sodium (i.e., counterions not found in high concentration in vivo) might be expected to reduce the possibility of common ion effects. However, interconversion of salt forms during dissolution is possible. For example, Li et al. (2005a) have described a situation where mesylate and phosphate salts of haloperidol that were exposed to high concentrations of free chloride subsequently converted to the hydrochloride salt, which showed a slower rate of dissolution resulting from the presence of the common ions.
In vivo salt conversion (most commonly to the hydrochloride or sodium salt) therefore runs the risk of a decrease in solubility due to a change in salt form, and may also increase susceptibility to common ion effects. These possibilities dictate that the Ksp for chloride and sodium salts is typically measured, regardless of the salt form that is ultimately developed, to provide an indication of the potential ramification of in vivo conversion.
Interestingly, although salts formed with hydrochloride and sodium ions are perhaps the most susceptible to common ion effects, they are also by far the most common counterions for weak bases and acids, respectively (see Table 8). This situation may be attributed to the high acidity/basicity of these counterions and, therefore, the potential to form salts with even weakly ionizable drug compounds (Bastin et al., 2000; Pudipeddi et al., 2002) and the realization that hydrochloride and sodium salts are often the default option during drug crystallization and purification. In the absence of obvious problems with factors such as solubility, hygroscopicity, stability, and crystallinity, these salt forms are often progressed, and as such, their high prevalence does not necessarily provide any indication of advantage with respect to solubility.
d. Effect of Organic Solvents on Salt Solubility
As a culmination of the synthetic steps required to obtain a new chemical entity, organic solvents or cosolvent-water mixtures are commonly used to facilitate isolation of a crystalline solid. Isolation as a crystalline solid assists in purification, and the choice of the initial salt form often reflects ease of purification rather than intrinsic downstream pharmaceutical or biopharmaceutical properties. Nonetheless, almost all salt forms are commonly isolated from organic solvents, mixtures of organic solvents or cosolvent/water mixtures (Serajuddin and Pudipeddi et al., 2002; Kumar et al., 2007). The organic solvents (water miscible and immiscible) that may be used for developing drug products are restricted by safety and toxicology issues; a comprehensive review of the use of nonaqueous solvents for oral drug products was recently published (Pole, 2008).
In addition to changing bulk solubility properties (and promoting crystallization), the presence of cosolvent can significantly affect the process of salt formation. For example, in less polar environments (e.g., in the presence of a cosolvent), the ionized form of the drug and counterion is less favored. As such, weak acids and bases become weaker, increasing pKa in the case of weakly acidic drugs and acidic counterions or decreasing pKa in the case of weakly basic drugs and basic counterions. Under these conditions, the use of solvents or cosolvents disfavors salt formation by reducing the ability of the acidic or basic counterion to shift the pH to pHmax (Tarsa et al., 2010). In contrast, where drug and counterion pKa remain largely unchanged, solvents and cosolvents promote salt formation via increases in the solubility of the un-ionized form (Fig. 8C) (Kramer and Flynn, 1972). Ideal solvents and cosolvents for salt formation have good solvent properties for un-ionized drug, but they have limited effects on acidity/basicity (Rubino and Thomas, 1990). Despite the frequency of use of cosolvents during purification and isolation of salt forms, Serajuddin and Pudipeddi (2002) caution that cosolvent use may result in the isolation of salts that would not ordinarily form in water. Such salts therefore are at high risk of conversion to the free acid/base on contact with aqueous fluids and moisture (Serajuddin and Pudipeddi, 2002).
B. pH adjustment Strategies for Addressing Low Drug Solubility
Solution formulations can be readily adjusted to a target pH such that a drug candidate is ionized to a degree that affords solvation of the target drug dose. This approach applies equally to solutions of salts or the corresponding free acid or free base since the solubility is the same at an equivalent pH and counterion concentration (assuming the solubility limit of the salt has not been reached).
1. Buffered Systems Used in Parenteral Formulations
The most commonly used pharmaceutical buffers comprise weak acids (HA) and the conjugate base (A−). Since physiological pH covers mainly neutral and acidic pH, buffers of weak bases (B) and their conjugate acids (BH+) are less frequently used. To exemplify, the acid-conjugate base pair of acetate buffer is illustrated in Scheme 2.
Dissociation of a commonly used (acetate) buffer.
In this system, an increase in hydrogen ion concentration is buffered by the conjugate base (CH3COO−), and the addition of hydroxide ion is similarly neutralized by the acid (CH3COOH). Buffer capacity is widely used to measure the ability of a buffer to resist a change in pH and is a measure of the quantity of acid/base required to change the pH of a buffered solution by 1 pH unit. Increasing buffer concentrations leads to increases in buffer capacity, and at a constant buffer concentration, maximum buffering capacity is achieved when the solution pH is close to the pKa of the buffer.
Details of a wide selection of buffers have been previously published (Flynn, 1980; Merck-Index, 2006). Buffer systems most commonly applied to parenteral pharmaceutical formulations and their effective buffer pH range are listed in Table 9 (Lee et al., 2003; Li and Zhao, 2007; Nema and Brendel, 2011). The most effective buffer range is usually limited to one pH unit either side of the pKa, although buffer combinations may be used to extend the buffer range.
Buffers widely used in parenteral formulations, including details of their buffering range and concentration used in some example marketed formulations
During parenteral administration, pain on administration is likely when using solutions buffered to high or low pH. Formulations are therefore typically limited to a working pH range of 4–9, although a wider pH range (2–9) may be used acutely in a preclinical setting (Li and Zhao, 2007). Extremes of pH may also lead to chemical instability, and in these cases, drug stability should be monitored in the formulation for the intended period of use (Pasini and Indelicato, 1992; Islam and Narurkar, 1993; Carstensen, 1995; Zhu et al., 2002; Chen et al., 2006). Buffer concentrations normally exceed 0.05 M and are generally less than 0.5 M (Wang and Kowal, 1980; Sinko, 2006). Increasing buffer capacity does not increase drug solubility but can prove more effective in maintaining drug in solution on dilution or with the addition of a formulation additive. High buffer concentrations, however, increase the risk of pain on injection and in high volume may alter physiological levels of biologically important buffer components such as lactate (Nema and Brendel, 2011). The buffer will also contribute to the overall ionic strength of the formulation, and high buffer concentrations can promote salting out of drug from solution (McDevit and Long, 1952; Raghavan et al., 1996; Yalkowsky, 1999).
A pH adjustment is a simple and powerful mechanism for solubility adjustment and, as such, is typically the first approach for solution formulations of ionizable drugs. For example, naproxen is a weak acid with a pKa of 4.57 and intrinsic solubility (of the free acid) of 16 μg/ml (Fini et al., 1995). As a comparatively “strong” weak acid, the low solubility of naproxen can be increased dramatically in a pH range acceptable for parenteral administration. Indeed, solution concentrations of ∼45 mg/ml might be expected in pH 8 phosphate buffer and ∼450 mg/ml in pH 9 sodium bicarbonate buffer. Under these circumstances, pH adjustment could be used to deliver a wide range of doses (Lee et al., 2003). In some cases, and in particular for weak(er) acids of higher pKa and weak(er) bases with lower pKa values, pH adjustment alone may be insufficient to allow the administration of an appropriate dose. For example, attempts to formulate a drug candidate that was a weaker acid than naproxen (e.g., a compound with pKa = 7), but of equivalent intrinsic solubility, would provide a maximum concentration of only ∼1.6 mg/ml in bicarbonate buffer (pH 9). In this case, pH adjustment alone is unlikely to offer a viable solubilization strategy, and a combination of strategies would need to be explored, such as the addition of a cosolvent, surfactant, or cyclodextrin. The advantages (and complications) associated with these combination approaches are discussed in the following section, and the benefits of cosolvents, surfactants; cyclodextrins as individual solubility enhancers are described in more detail in Sections VI, VII, and VIII, respectively.
2. Impact of Cosolubilizers and Electrolytes on pH-Mediated Solubilization
a. pH Adjustment and Cosolvents
Cosolvents—including propylene glycol, ethanol, PEG, and DMSO—increase the solubility of nonpolar molecules in polar solvents (such as water) by reducing solvent polarity (see Sections VI and VI.D.1). The presence of a cosolvent may therefore provide additional solubilization for solution formulations where pH manipulation is insufficient. However, cosolvents also alter the strength of the buffers that are included in a pH-cosolvent combination formulation, potentially altering their application (Narazaki et al., 2007b). For example, acidic buffers are less acidic in the presence of cosolvents since the less polar conditions favor the un-ionized form of the buffering species, resulting in an increase in the apparent pKa (Rubino, 1987; Yalkowsky, 1999). Similarly, decreasing concentrations of the ionized species in basic buffers leads to a decrease in pKa and a reduction in basicity. As acids donate protons, they increase the number of free ions in solution; therefore, the pKa of acidic species is more sensitive to changes in solvent polarity compared with bases, which have no effect on the concentration of ions in the system (Rubino, 1987; Yalkowsky, 1999; Narazaki et al., 2007b). The effect of cosolvents on the pKa of both acids and bases is therefore predictably more enhanced with decreasing polarity of the cosolvent (i.e., reduced dielectric capacity). The potential outcome of this effect is that the addition of a cosolvent to a buffer system may alter its capacity to maintain pH by causing a shift in pKa of the buffering species. The effect is further complicated by the ability of cosolvents to shift the pKa of weakly acidic and weakly basic drugs such that there is a reduced fraction of ionized drug at a given pH (Kramer and Flynn, 1972; Narazaki et al., 2007b).
Importantly, however, the net effect of using a cosolvent with pH manipulation is usually an increase in total drug solubility. This reflects the fact that changes in dissociation are usually offset by cosolvent-mediated enhancements in the solubility of the un-ionized form. Cosolvents may in some cases also increase the solubility of the ionized species. Table 10 describes the effect of the addition of a range of cosolvents on the solubility of a weak base across a wide pH range (1–7). In all cases, cosolvent addition increases solubility, and the maximum solubility is evident at the lowest pH (reflecting the largest ionized fraction) (Jain et al., 2001). However, the relative gain that can be attributed to the cosolvent progressively decreases with decreasing pH. This reflects the decreased solubility advantage provided by cosolvents for increasingly ionized species.
The effect of cosolvent and surfactant addition on the solubility of a weak base at differing pH
Since the addition of a cosolvent to a pH-buffered solution leads to a net increase in drug solubility, this approach has been widely implemented in marketed parenteral formulations, including chlordiazepoxide [Librium; Hoffmann-LaRoche, Inc., Nutley, NJ (propylene glycol 20%, pH 3)], dihydroergotamine [DHE-45; Novartis AG, Basel, Switzerland (15% glycerin, 6.1% ethanol, pH 3.6)], fenoldopam [Corlopam; Ben Venue Laboratories, Inc., Bedford, OH (propylene glycol 50%, pH 3)], oxytetracycline [Terramycin; Pfizer Labs, NY (propylene glycol 65–75%, pH 3)], pentobarbital [Nembutal; Lundbeck, Inc., Deerfield, IL (propylene glycol 40%, ethanol 10%, pH 9.5)], and phenytoin [Dilantin; Park-Davis, NY (propylene glycol 40%, ethanol 10%, pH 10–12.3)] (Strickley, 2004).
One concern with the use of pH-cosolvent combinations is the potential for an increased risk of drug precipitation on dilution resulting from a reduction in buffer capacity. This is well exemplified by a relatively recent study of the solubility properties of the weak acid phenytoin in a formulation containing 50 mM carbonate buffer (pH ∼9.8) with and without 15% ethanol. After dilution with phosphate buffer (pH 7.4), formulations containing ethanol showed a greater shift (decrease) in pH as a result of the increased pKa of the buffer in the presence of the cosolvent, in turn leading to increased drug precipitation in comparison with ethanol-free formulations (Narazaki et al., 2007b).
b. pH Adjustment and Strong Electrolytes
Strong electrolytes, such as sodium chloride, are often added to parenteral formulations to ensure isotonicity.