Salt Selection in Drug Development

Published on: 
, ,
Pharmaceutical Technology, Pharmaceutical Technology-03-02-2008, Volume 32, Issue 3

The selection of an appropriate salt form for a potential drug candidate is an opportunity to modulate its characteristics to improve bioavailability, stability, manufacturability, and patient compliance.

An estimated 50% of all drug molecules used in medicinal therapy are administered as salts. This fact indicates that the salification, or salt formation, of a drug substance is a critical step in drug development (1, 2). A drug substance often has certain suboptimal physicochemical or biopharmaceutical properties that can be overcome by pairing a basic or acidic drug molecule with a counterion to create a salt version of the drug (3). The process is a simple way to modify the properties of a drug with ionizable functional groups to overcome undesirable features of the parent drug (4). This fact underlines the importance of salt formation for drugs that are designed, developed, and marketed after a rigorous research and development program (1).

Basic concepts in salt formation

Salts are formed when a compound that is ionized in solution forms a strong ionic interaction with an oppositely charged counterion, leading to crystallization of the salt form (5). In the aqueous or organic phase, the drug and counterion are ionized according to the dielectric constant of the liquid medium. The charged groups in the drug's structure and the counterion are attracted by an intermolecular coulombic force. During favorable conditions, this force crystallizes the salt form (see Figure 1). All acidic and basic compounds can participate in salt formation (4). However, the success and stability of salt formation depends upon the relative strength of the acid or base or the acidity or basicity constants of the species involved (6).

Figure 1: Diagrammatic representation of salt formation.

The salt form is separated into individual entities (i.e., the ionized drug and the counterion) in liquid medium, and its solubility depends upon the solvation energy in the solvent. The solvent must overcome the crystal lattice energy of the solid salt and create space for the solute. Thus, the solubility of a salt depends on its polarity, lipophilicity, ionization potential, and size. A salt's solubility also depends on the properties of solvent and solid such as the crystal packing and presence of solvates (7).

Table Ia: Advantages of salt formation for drug properties.

The importance of salt formation

Salt forms of drugs have a large effect on the drugs' quality, safety, and performance. The properties of salt-forming species (i.e., counterions) significantly affect the pharmaceutical properties of a drug (see Tables Ia and Ib) and can greatly benefit chemists and formulators in various facets of drug discovery and development (6).

Table Ib: Disadvantages of salt formation for drug properties.

Salt-selection strategy

The stage of salt selection in drug development. Pharmaceutical companies previously selected salts at various stages in drug development. However, companies now tend to move the salt-selection process to the research phase to make the process more foolproof (25). Ideally, the salt form should be chosen before long-term toxicology studies are performed (i.e., at the start of Phase I clinical trials) (24). This timing is an important factor in the early stages of new-drug development because changing the salt form at a later stage may force a repetition of toxicological, formulation, and stability studies, thus increasing development time and cost (26). A new salt form introduced at a late stage must also be evaluated for potential impurity changes, and its bioequivalence (bio-bridge), pharmacokinetic equivalence (PK-bridge), and toxicity equivalence (tox-bridge) to the previous salt form must be proven.

Objectives of salt selection. Innumerable salt forms are available to pharmaceutical scientists. The selection process must therefore be rational and streamlined. A lack of proper planning may lead to the synthesis of several salt forms of the drug candidate for preformulation testing. Moreover, this hit-or-miss approach results in many failures and may cause the loss of test substance and time. These considerations underscore the need for a well-formatted decision tree to help scientists choose a suitable salt form in an efficient and timely manner, depending upon the intended use, with a minimum number of failures and expended resources.

The main objective of a salt-selection study is to identify the salt form most suitable for development. The following four parameters are often considered primary or essential criteria for the selection of a particular form:

  • Aqueous solubility measured at various pH values, depending upon the intended pharmaceutical profile

  • High degree of crystallinity

  • Low hygroscopicity (i.e., water absorption versus relative humidity), which gives consistent performance

  • Optimal chemical and solid-state stability under accelerated conditions (i.e., minimal chemical degradation or solid-state changes when stored at 40 °C and 75% relative humidity).

A serious deficiency in any of these characteristics should exclude that form from further development. In addition to these essential criteria, the following desirable criteria also influence salt-form selection:

  • Limited number of polymorphs or absence of variability because of polymorphism

  • Ease of synthesis, handling, or formulation development (27).

A single salt form generally cannot satisfy all the requirements for developing drug-substance dosage forms. However, introducing a second or third salt form consumes additional developmental resources and increases the cost of manufacturing, handling, storing, and characterizing the additional salt forms. Therefore, the dosage form is developed with a single salt form whenever possible (9). The major drug-development issues are addressed by choosing the appropriate salt form. Minor issues can be addressed using other development tools. Decreasing development timelines intensify the pressure to select the right salt form the first time. Salt selection is a critical step in the preformulation stage of drug development. Gould says that "the balance required in assessing the correct salt from to progress into drug development makes it a difficult semiempirical exercise" (8). This statement emphasizes the need to prioritize the salt-selection process so that various development issues are addressed as early as possible.

Potential candidate for salt formation. The decision about whether salt or free acid or base should be developed depends on these forms' relative pharmaceutical and commercial merits. If the active compound is a liquid, a solid dosage form is usually preferred because oil is difficult to purify, characterize, and maintain in its effective form. Oil is also difficult to transport, sensitive to oxygen, and susceptible to batch-to-batch variations. If the free acid or base is a water-soluble solid with a high melting point, preparing a salt form is generally unnecessary. Alternatively, several useful properties of salt forms may be explored (6).

In spite of the numerous advantages associated with salt forms, developing them is not always feasible. The preparation of a stable salt may not be possible for some drugs. The salt may have certain undesirable properties compared with the free acid or base, and it would thus be appropriate to develop the free acid or base (28). In a salt-screening study of RPR111423, a pyridine base, hydrochloride and mesylate salts were formed. The hydrochloride salt showed a loss of hydrogen chloride at high temperatures (110–120 °C) and precipitation at an acidic pH because of the common-ion effect. The mesylate salt also showed precipitation at acidic pH. The two salts were polymorphic and hygroscopic in comparison with free base, which was nonpolymorphic and nonhygroscopic. These results proved the free base to be a better candidate than the salt forms (29).

Pharmacological indications also help determine whether the salt form or the free acid or base should be pursued. For example, when a slow onset or a constant plasma level is required, a highly ionized salt form may be inappropriate if the free acid or base provides a sufficient plasma level. Tolbutamide sodium, an antihyperglycemic agent, causes a rapid fall in blood glucose levels because it is highly ionized. This characteristic causes hypoglycemia in patients with normal insulinomas. Therefore, tolbutamide sodium's corresponding free acid was preferred for oral administration. The salt form's only application is the diagnosis of pancreatic adenomas (30).

Pharmaceutical considerations. The choice of salt is governed largely by the acidity or basicity of the ionizable group, the safety of the counterion, the drug indications, the route of administration, and the intended dosage form. The expectations of the salt form must be outlined as a desirable pharmaceutical profile that guides the synthesis of the salt forms (see Table II).

Table II: The selection of potential salt forms based on intended pharmaceutical profile.

Ionic considerations. The degree of ionization is a critical parameter for the physiological performance of the drug and for its formulation development (25). The pKa of the drug and counterion is important for successful salt formation as well. For the preparation of salt forms of basic drugs, the pKa of the counterion should be at least 2 pH lower than the pKa of the drug (34). Similarly, for the preparation of salt forms of acidic drugs, the pKa of the counterion should be at least 2 pH higher than the pKa of the drug. These specifications are required because the counterion must bring the solution's pH to a level lower than the pHmax (see Figure 2) to reach the salt plateau, at which the solubility of salt prevails over the solubility of free acid or base (5). The generalized rule of difference in pKa units of the ionizable group in the active pharmaceutical ingredient (API) and in the acid or base is meant to make the proton transfer energetically favorable. When the pKa of the API and counterion are not significantly different, a solid complex may form but may rapidly disproportionate (i.e., break down into the individual entities of drug and counterion) in an aqueous environment (35).

Generation of salt forms. Salts can be prepared on a small scale using various methods. Forming salts from free acid or base is the most common method. The free acid or base of the drug substance is combined with the counterion base or acid in specific molar ratios in a suitable solvent system. The salt form is then isolated, and the solid precipitate is recrystallized. A less common method is to form salts through salt exchange. In this method, a counterion salt is treated with a free acid or base in a specific molar concentration in a suitable solvent. The solid is then isolated and recrystallized. The sulfate salt of methyl pyridinium-2-aldoxime was prepared using silver sulfate as a counterion. The unwanted silver ions were removed as insoluble iodide salt, and the desired sulfate salt was precipitated by adding antisolvent (36). A wide range of salts are generally prepared for each new substance. Their properties are compared during a preformulation program that improves the chances of selecting the optimal salt form (29). However, a balanced approach should be adopted because limited resources are available at this early stage of drug development. Commonly used salts such as hydrochlorides and sodium have advantages over other salt-forming moieties. For example, they have low molecular weight and low toxicity. However, other salt forms such as mesylate may sometimes offer advantages such as higher solubility and bioavailability (37).

Advertisement

High-throughput synthesis has gained greater importance in the salt-selection process. This technique allows many counterions and crystallization solvents to be evaluated using as little as 50 mg of drug substance. After the optimum drug-substance form is selected at the microlevel, the synthesis of the compound can be scaled up to several hundred grams to test for other stages of preformulation (27). In situ salt screening also offers a viable alternative to traditional salt screening (34). This method has a special relevance for poorly soluble compounds because it can rapidly rank compounds based on their solubility, effectively screening out insoluble compounds immediately (13). During in situ salt screening, a known concentration of drug is added to a concentrated counterion solution sufficient to obtain the pHmax (see Figure 2) for successful salt formation that may be subjected to solubility screening. Tong applied in situ salt screening to GW1818, an alpha 1A adrenergic receptor antagonist, and short-listed four salts—the phosphate, succinate, mesylate, and hydrochloride—for further development. These salts were found to be crystalline with adequate solubility (comparable with the authentic salts later prepared by the traditional method) and were selected for further evaluation (34).

Figure 2: The pH-solubility profile for a compound with a single, basic pKa value of 5. The four regions of pH-dependent solubility are: salt plateau, pHmax, ionized compound, and unionized compound.

In spite of the abundance of available counterions, few are used frequently. The preference for pharmaceutical counter-ions is explained by studying the distribution of different counterions of medicinal compounds in USP 2006 (38) (see Table III). The table shows that salt forms of drugs (56.15%) are preferred over free forms (43.85%). Hydrochloride and sodium remain the favorite counterions for the salt formation of medicinal compounds. However, the availability of many pharmaceutically acceptable counterions makes the salt-selection process difficult and cumbersome.

Table III: Salt forms in USP 2006.

Salt-form selection. The generated salt forms are compared for the desired physico-chemical and biopharmaceutical properties, which guide the final selection of an optimal salt form.

Techniques for the characterization of salts. After synthesis, the formation of the salt forms must be confirmed. Next, their pharmaceutical properties must be assessed. Several characterization techniques provide valuable information for salt screening (see Table IV).

Table IV: Techniques for the characterization of salt forms.

Salt-selection studies. Morris et al. adopted a multitiered approach to screen salts for their optimal physical forms (39). In this approach, physicochemical tests are conducted in several tiers, and a go–no-go decision is made after each tier. Only appropriate salts, free acids, or bases are tested further, thus avoiding the generation of extensive data about each salt form generated. The studies can be planned so that the least time-consuming experiments that could still prompt a go–no-go decision are conducted in the first tier. Experiments that are more time consuming and labor intensive can be conducted at later tiers. In this way, many salt forms can be screened with a minimum of experimental effort. If the tiered approach eliminates all the candidates, additional salts must be considered before reevaluating any salt rejected in an earlier tier.

For a rational approach to salt screening, the tiered approach should be combined with a goal-oriented approach in which the main problems associated with the free acid or base are handled first, followed by secondary problems. For example, ranitidine hydrochloride is hygroscopic with a critical relative humidity of approximately 67% (40). However, the hydrochloride salt of ranitidine has better absorption properties compared with the free base and is one of the most successful drugs ever marketed. In a multitiered approach, the hydrochloride salt would have been rejected after hygroscopicity testing, in spite of its better absorption profile (41). High hygroscopicity could be mitigated by developing proper packaging. Similarly, the hydrochloride form of sertaline (i.e., Pfizer's Zoloft) might have been rejected because of its reported 28 polymorphic forms (42). This fact underlines the importance of a goal-oriented approach that addresses the most critical problems first. Less critical problems could be overcome by a proper development strategy. The final salt form selected should have a fine balance of the optimal physicochemical and biopharmaceutical properties. Each stage of salt selection (see Figure 3) is relevant and contributes to the selection of the optimal salt form. However, salt selection can be a difficult task because each salt imparts unique properties to the parent compound.

Figure 3: Flow diagram for selecting the optimal salt form of a drug.

Stages of salt selection. Salt screening starts with the characterization of free acid or base, followed by the identification of possible counterions. The acid or base characterization provides information for potential counterion selection and for planning relevant crystallization experiments. This stage is followed by a screening of crystallization conditions for the desired salts, salt formation and its confirmation, and finally the preformulation characterization of generated salts (20).

During salt-form selection, the determination of pKa and corresponding ionizable groups gives an idea of the feasibility of salt formation. This information is the basis for selecting suitable counterions and a preliminary synthesis of salt forms, preferably at the microlevel, coupled with characterization for salt formation. After the confirmation of salt formation, the prepared salts are screened for various biopharmaceutical properties with a view to selecting the optimal salt form.

Assessment of crystallinity is the first stage of salt selection. The salt form should preferably be crystalline so that its properties remain constant during pharmaceutical handling, transportation, and use. However, the amorphous form may have advantages (e.g., solubility) that can be harnessed by proper formulation development. On the other hand, stabilizing the amorphous form for devitrification to crystalline form may lead to the loss of these advantages. Atorvastatin calcium was originally developed in an amorphous form. During Phase III clinical trials, it reverted to crystalline form, and the final product was developed using a crystalline form (43).

After crystallinity assessment, the salt form's hygroscopicity profile is assessed to find a salt form that retains its properties in the varying humidity conditions of pharmaceutical operations. This assessment can be performed using methods such as traditional saturated salt solutions in a desiccator or more advanced dynamic vapor sorption methods. The salt forms with acceptable hygroscopicity profiles are then evaluated for their solubility. The salts with adequate solubility are tested for their physicochemical stability, including polymorphic stability and excipient compatibility. These tests are especially relevant in combination formulations such as aspirin–propoxyphene. Aspirin–propoxyphene hydrochloride is unstable, but aspirin–propoxyphene napsylate is stable (8).

Salt forms having adequate stability are assessed for variability in their properties resulting from polymorphism. Compounds with a limited number of polymorphs are preferred because their performance during pharmaceutical operations and performance is predictable. The salt forms that qualify the stage of polymorphism are tested for process control, economic feasibility, and processability (including parameters such as corrosiveness, taste, wettability, and flowability). These criteria are generally evaluated at a small scale by a medicinal chemist, who narrows the choice to a particular salt form. However, after a particular salt form is selected, these parameters are evaluated at a larger scale so that the selected salt form has properties that are easily controlled batchwise and over time.

The selected salt form is subjected to pharmacological testing for drug release as per the requirements of onset and the duration of activity. Pharmacological safety studies are also performed. For example, epinephrine borate causes occasional mild stinging in the eye, compared with hydrochloride and bitartrate salt, which cause moderate to severe stinging (44). The selected salt form may then be subjected to extensive long-term toxicology studies in Phase I clinical trials of drug development.

Patent aspects of salt forms

Salt-selection studies provide a viable extension of a drug's patent because salts with superior properties can be patent-protected. New salt forms often have novel physical properties related to processability (e.g., crystallization, morphology, and filtration) and formulation (e.g., stability) (45). They may also result in the detection of new polymorphs (1).

A new salt form may have a profile that makes it suitable for a new route of administration. For example, diclofenac sodium salt (Ciba-Geigy) was marketed as Voltaren. Before the Voltaren patent expired, other salts (e.g., diclofenac diethylamine) with substantially better skin-penetration properties were discovered and patented. These salts, in corresponding formulations, were particularly suitable for topical applications. Patenting new salts, therefore secures an exclusive position in the market (20).

Selecting an appropriate salt form of an API may also play a role in blocking the development of generic drug products. Dr. Reddy's Laboratories tried to obtain marketing authorization for amlodipine maleate, a different salt version of amlodipine besylate. However, the US Court of Appeals for the Federal Circuit concluded that the basic patent for amlodipine covers other salt forms of the drug, including its maleate salt. The verdict against Dr. Reddy's Laboratories effectively prevented the generic version from entering the market (19).

Regulatory aspects of salt forms

Salt-selection studies should consider the regulatory aspects of introducing a new salt form. A new salt form of an approved drug substance is considered a new chemical entity, thus requiring a full dossier to be submitted for marketing approval (1). For regulatory purposes, a new salt form is designated a "pharmaceutical alternative" to the original form (46, 47). However, the approval process for a new salt may use some of the details already known about the active entity of a related, previously accepted salt (1). Therefore, when scientists change the active moiety of a salt that is already marketed, they may usually submit an abbreviated application, popularly known as the 505 b(2) filings (or the hybrid NDA), if they can prove that the new salt form's active moiety has the same pharmacokinetics, pharmacodynamic, and toxicity characteristics as the original (19). A generic version of a drug based on an alternative salt form may also be approved in a similar way. However, the benefits expected from the introduction of a new salt form must be weighed against the cost and time involved in the studies required for regulatory approval.

Conclusion

Selecting an optimal salt form for development is a critical step in ensuring the efficient and successful development of a robust product. Salt selection requires a well designed screening strategy that fulfills the essential and desirable criteria that set the standard for salt screening. In addition, salt selection procedures must also assess the regulatory, intellectual, and marketing considerations to balance the drug's physicochemical and biopharmaceutical properties against commercial considerations.

Lokesh Kumar is a student, Aeshna Amin is a student, and Arvind K. Bansal* is an associate professor at the National Institute of Pharmaceutical Education and Research, Sector-67, Phase-X, S.A.S. Nagar, Punjab 160062, India, tel +91 0 172 2214682 87, fax +91 0 172 2214692, akbansal@niper.ac.in

*To whom all correspondence should be addressed.

Submitted: Apr. 2, 2007. Accepted: Apr. 5, 2007.

References

1. C.G. Wermuth and P.H. Stahl, "Introduction," in Handbook of Pharmaceutical Salts: Properties, Selection and Use, P.H. Stahl and C.G. Wermuth, Eds. (Wiley–VCH, Weinheim, Germany, 2002), pp. 1–7.

2. S.M. Berge, L.M. Bighley, and D.C. Monkhouse, "Pharmaceutical Salts," J. Pharm. Sci. 66 (1), 1–19 (1977).

3. Tessella Scientific Software Solutions, "Automated Salts and Polymorph Screening," www.tessella.com/Services/CaseStudies/pdfs/e_GSK_ASAP.pdf, accessed Dec. 15, 2006.

4. B.D. Anderson and R.A. Conradi, "Predictive Relationships in the Water Solubility of Salts of a Nonsteroidal Anti-inflammatory Drug," J. Pharm. Sci. 74 (8), 815–820 (1985).

5. S.N. Bhattachar, L.A. Deschenes, and J.A. Wesley, "Solubility: It's Not Just for Physical Chemists," Drug Discov. Today 11 (21/22), 1012–1018 (2006).

6. L.D. Bighley, S.M. Berge, and D.C. Monkhouse, "Salt Forms of Drugs and Absorption," in Encyclopedia of Pharmaceutical Technology, J. Swarbrick and J.C. Boylan, Eds. (Marcel Dekker, New York, 1996), pp. 453–499.

7. "Properties of the Solid State," in Physicochemical Principles of Pharmacy, A.T. Florence and D. Attwood, Eds. (Macmillan Press Ltd., London, 3rd ed., 1998), pp. 5–34.

8. P. L. Gould, "Salt Selection for Basic Drugs," Int. J. Pharm. 33 (1–3), 201–217 (1986).

9. P.H. Stahl and M. Nakano, "Pharmaceutical Aspects of the Drug Salt Form," in Handbook of Pharmaceutical Salts—Properties, Selection, and Use, P.H. Stahl and C.G. Wermuth, Eds. (Wiley–VCH, Weinheim, Germany, 2002), pp. 83–116.

10. M.K. Chourasia and S.K. Jain, "Pharmaceutical Approaches to Colon Targeted Drug Delivery Systems," J. Pharm. Pharm. Sci. 6 (1), 33–66 (2003).

11. A. Leone-Bay, D. Moy-Sherman, and B. R. Wilson. (Mankind Corp.), "Diketopiperazine Salts, Diketomorpholine Salts or Diketodioxane Salts for Drug Delivery," PCT Patent WO 06/23943 (2006).

12. S.I.F. Badawy, "Effect of Salt Form on Chemical Stability of an Ester Prodrug of a Glycoprotein IIb/IIIa Receptor Antagonist in Solid Dosage Forms," Int. J. Pharm. 223 (1–2), 81–87 (2001).

13. L.F. Huang and W.Q. Tong, "Impact of Solid State Properties on Developability Assessment of Drug Candidates," Adv. Drug Deliv. Rev. 56 (3), 321–334 (2004).

14. W.D. Walking et al., "Xilobam: Effect of Salt Form on Pharmaceutical Properties," Drug Dev. Ind. Pharm. 9 (5), 809–819 (1983).

15. F. Pfannkuch, H. Rettig, and P.H. Stahl, "Biological Effects of the Drug Salt Form," in Handbook of Pharmaceutical Salts—Properties, Selection, and Use, P.H. Stahl and C.G. Wermuth, Eds. (Wiley–VCH, Weinheim, Germany, 2002), pp. 117–134.

16. K.M.O. Connor and O.I. Corrigan, "Preparation and Characterisation of a Range of Diclofenac Salts," Int. J. Pharm. 226 (1–2), 163–169 (2001).

17. G.M. Dull et al. (Targacept Inc.), "Hydroxybenzoate Salts of Metanicotine Compounds," PCT Patent WO 06/53039 (2006).

18. C. Shaopei et al. (Chiron Corp.), "Pharmaceutical Acceptable Salts of Quinoline Compounds Having Improved Pharmaceutical Properties," PCT Patent WO 05/46589 (2005).

19. R.K. Verbeeck, L. Kanfer, and R.B. Walker, "Generic Substitution: The Use of Medicinal Products Containing Different Salts and Implications for Safety and Efficacy," Eur. J. Pharm. Sci. 28 (6), 1–6 (2006).

20. V.M. Raumer, J. Dannappel, and R. Hilfiker, "Polymorphism, Salts, and Crystallization: The Relevance of Solid-State Development," Chem. Today 24 (1), 41–44 (2006).

21. M.J. Bowker, "A Procedure for Salt Selection and Optimization," in Handbook of Pharmaceutical Salts—Properties, Selection and Use, P.H. Stahl and C.G. Wermuth, Eds. (Wiley–VCH, Weinheim, Germany, 2002), pp. 161–189.

22. U.J. Griesser and D.E. Braun, "Crystal Polymorphism in Pharmaceuticals: A Statistical Approach," www.eurostar-science.org/conferences/abst9/Griesser.pdf, accessed Dec. 15, 2006.

23. W.Q. Tong, "Salt Screening and Selection: New Challenges and Considerations in the Modern Pharmaceutical R&D Paradigm,"

, accessed Dec. 20, 2006.

24. R. Hilfiker, F. Blatter, and M.V. Raumer, "Relevance of Solid-State Properties for Pharmaceutical Products," in Polymorphism in the Pharmaceutical Industry, R. Hilfiker, Ed. (Wiley–VCH, Weinheim, Germany, 2006), pp. 1–19.

25. S. Balbach and C. Korn, "Pharmaceutical Evaluation of Early Development Candidates—the 100-mg Approach," Int. J. Pharm. 275 (1–2), 1–12 (2004).

26. G. Davies, "Changing the Salt, Changing the Drug," Pharm. J. 266 (7138), 322–323 (2001).

27. J.D. Higgins and W.L. Rocco, "Pharma Preformulation: A Stop Along the Drug Development Highway," Today's Chemist at Work 12 (7), 22–26 (2003).

28. A.T.M. Serajuddin et al., "Preformulation Study of a Poorly Water Soluble Drug α-pentyl-3-(2-quinolinylmethoxy) benzenemethanol: Selection of the Base for Dosage Form Design," J. Pharm. Sci.75 (5), 492–496 (1986).

29. R.J. Bastin, M.J. Bowker, and B.J. Slater, "Salt Selection and Optimisation Procedures for Pharmaceutical New Chemical Entities," Org. Proc. Res. Dev. 4 (5), 427–435 (2000).

30. "Orinase Diagnostic-Product Information," www.pfizer.com/pfizer/download/uspi_orinase.pdf, accessed Jan. 21, 2006.

31. R.M. Everett et al., "Nephrotoxicity of Pravadoline Maleate (WIN 48098-6) in Dogs: Evidence of Maleic Acid-Induced Acute Tubular Necrosis," Fundam. Appl. Toxicol. 21 (1), 59–65 (1993).

32. D. J. Snodin, "Residues of Genotoxic Alkyl Mesylates in Mesylate Salt Drug Substances: Real or Imaginary Problems?," Regul.Toxicol. Pharmacol. 45 (1), 79–90 (2006).

33. A. Allerman, "New Drug Watch," www.pec.ha.osd.mil/Updates/0307web/Aug-Sep_03_Update_Page_5.htm, accessed Dec. 15, 2006.

34. W.Q. Tong and G. Whitesell, "In situ Salt Screening—A Useful Technique for Discovery Support and Preformulation Studies," Pharm. Dev. Technol. 3 (2), 215–223 (1998).

35. C.R. Gardner et al., "Application of High Throughput Technologies to Drug Substance and Drug Product Development," Comput. Chem. Eng. 28 (6–7), 943–953 (2004).

36. A.W. Newman and G.P. Stahly, "Form Selection of Pharmaceutical Compounds," in Handbook of Pharmaceutical Analysis, vol. 117, L. Ohannesian and A.J. Streeter, Eds. (Marcel Dekker, New York, 2002), pp. 1–58.

37. G.L. Engel et al., "Salt Form Selection and Characterization of LY333531 Mesylate Monohydrate," Int. J. Pharm. 198 (2), 239–247 (2000).

38. USP, USP 29–NF 24 (US Pharmacopeial Convention, Rockville, MD, 2006).

39. K.R. Morris et al., "An Integrated Approach to the Selection of Optimal Salt Form for a New Drug Candidate," Int. J. Pharm. 105 (3), 209–217 (1994).

40. R. Teraoka, M. Otsuka, and Y. Matsuda, "Effects of Temperature and Relative Humidity on the Solid-State Chemical Stability of Ranitidine Hydrochloride," J. Pharm. Sci. 82 (6), 601–604 (1993).

41. H.Y. Ando and G.W. Radebaugh, "Preformulation," in Remington: The Science and Practice of Pharmacy, II, A.R. Gennaro, Ed. (Lippincott Williams and Wilkins, Baltimore, MD, 20th ed., 2002), pp. 700–720.

42. J.F. Remenar et al., "Salt Selection and Simultaneous Polymorphism Assessment via High-Throughput Crystallization: The Case of Sertraline," Org. Proc. Res. Dev. 7 (6), 990–996 (2003).

43. C.R. Gardner, C.T. Walsh, and ö. Almarsson, "Drugs as Materials: Valuing Physical Form in Drug Discovery," Nat. Rev. Drug. Discov. 3 (11), 926–934 (2004).

44. "Ophthalmic/Otic Dosage Forms," www.cop.ufl.edu/safezone/prokai/pha5100/eye.htm, accessed Dec. 20, 2006.

45. Solvias, "Introduction: Solid-State Development," www.solvias.com/documents/Dokumente/Downloads_news/Solvias_Salt_and_Polymorphism_Programs.pdf, accessed Dec. 30, 2006.

46. European Agency for the Evaluation of Medicinal Products, "Note for Guidance on the Investigation of Bioavailability and Bioequivalence" (EMEA, London, UK, 2001).

47. FDA, Guidance for Industry: Bioavailability and Bioequivalence Studies for Orally Administered Drug Products—General Considerations (FDA, Rockville, MD, 2000).