You are on page 1of 22

REVIEW STUDIES ON SOLVENT DROP SYNTHESIS OF PHARMACEUTICAL COCRYSTAL Tella A.C1, Ameen O.A2* 1.

Department of Chemistry, University of Ilorin, 2. Department of Chemistry, Kwara State Polytechnic Ilorin. ABSTRACT: An efficient and environmental friendly synthesis of some

pharmaceutical co-crystal has been achieved. The combination of active pharmaceutical ingredient (api) and co-crystal former was studied and discovered that this enhance pharmaceutical properties by modification of chemical stability, moisture uptake, mechanical behaviour, solubility, dissolution rate and bioavailability. Keywords: co-crystal, active pharmaceutical ingredient (API), hydrogen bond,
crystalline complex.

Introduction The design of pharmaceutical crystals that possess different molecular components is valuable to control pharmaceutical properties of solids without changing the covalent bonds. These multiple component crystals, crystalline complexes or co-crystal often rely on hydrogenbonded assemblies between neutral molecules of the active pharmaceutical ingredient (API) and other components. As a consequence, co-crystals increase the diversity of solid-state forms of an API, even for non-ionisable APIs and enhance pharmaceutical properties by modification of chemical stability, moisture uptake, mechanical behaviour, solubility, dissolution rate and bioavailability. Co-crystals can be defined as crystalline complexes of two or more neutral molecular constituents bond together in the crystal lattice through noncovalent interaction (primarily hydrogen bond). However pharmaceutical co-crystal is a single crystalline solid that incorporates

two neutral molecules, one being an active pharmaceutical ingredient (API) and the other a co-crystal former. Co-crystal former may be an excipient or another drug. Pharmaceutical co-crystal technology is used to identify and develop new proprietary forms of widely prescribed drugs and offer a chance to increase the number of forms of an API. Scientists showed that modifying the physical properties of a pharmaceutical compound through pharmaceutical co-crystal fotmation improved the performance of a drug know to have poor solubility. Pharmaceutical co-crystallization is a reliable method to modify physical and technical properties of drugs such as solubility, dissolution rate, stability hygroscopisity, and compressibility without alternating their pharmacological behaviour. The use of co-crystals in drug design and delivery and functional materials with potential applications as pharmaceutical has recently attracted considerable interest. Pharmaceutical co-crystal have been described for many drugs such as acetoaminophen, aspirin, ibuprofen, fluriprofen etc.
ACTIVE PHARMACEUTICAL INGREDIENT CLASSIFICATION

The Classification of Active Pharmaceutical Ingredient (API) solid form based on structure and composition showed with the schematic diagram below.

AMORPHOUS

SOLID

CRYSTALLINE

SINGLE COMPONENT

MULTIPLE COMPONENT

POLYMORPHS HYDRATE SALTS SOLVATES COCRYSTAL

COCRYSTAL VERSUS SOLVATES The main difference between solvates and co-crystals is the physical state of the isolated pure components: if one component is a liquid at room temperature, the crystals are designated as solvates; if both components are solids at room temperature, the crystals are designated as co-crystals. SALT VERSUS CO-CRYSTAL FORMATION Co-crystal and Salts may sometimes be confused. The understanding of the fundamental difference between a salt formation and a co-crystal is very important to both pre-formulation activities and

chemical/pharmaceutical development aspects. Indeed, salts and cocrystals can be considered as opposite ends of multi-component structures. Salt are often chosen instead of the free acid or base as these can improve crystallinity, solubility and stability of a pharmaceutical compound. Co-crystals are an alternative to salts when these do not have the appropriate solid state properties or cannot be formed due to the absence of ionisable sites in the API. Salt formation is an acid-base reaction between the API and an acidic or basic substance. The widespread use of salt formation is evidence by the large number of marketed crystalline salts of APIs. salt formation is a three component system having an acid (A), a base (B) and one or more solvents. A salt is formed by transfer of a proton (H +) from an acid (A) to base (B) A-H + B (A-)(B+-H)

Proton transter is thought to mainly depend on the pKa values of the components. The general rules for the packing of hydrogen bonded molecules in crystals were developed by etter. When there is no such transfer and the components are instead present in the crystal as neutral entities, the product is generally defined as co-crytal. In other words. A co-crystal is an A-B composite in which no proton transfer occurred. The formation of a salt or co-crystal can be predicted from pKa value of acid (A) and a base (B). salt formation generally requires a difference of about 2.7 pKa units between the conjugate base and the conjugate acid (A) i.e [pKa (base) - pKa (acid) 2.7]. for example, succinic acid having pKa 4.2 form co-crystal with urea base pKa values are not sufficiently high to allow proton transfer when co-crystal is

formed. Co-crystal of succinic acid- urea has two hydrogen bonds i.e. the oxygen atom in urea molecule is bonded to hydrogen atom in succinic acid molecule while oxygen atom from succinic acid molecule is bonded to hydrogen atom in urea molecule as show below.

OH

C
O

O ............ H

N OH
.............

H
H

SCREENING OF COCRYSTALS Co-crystals can be prepared from two molecules of any shape or size having complementary hydrogen bond functionalities. The ability of an API to form cocrystal is dependent on a range of variables, including the types of co-former, the API co-former ratio, the solvent, the temperature, the pressure, the crystallization technique, etc. experimental screening for cocrystal former is not trivial. Synthesis/processing of cocrystals can be accomplished via a number of methods, including slow solvent evaporation crystallization from solution, solvent-reduced (e.g slurrying, solvent-drop grinding) and solvent-free (e.g grinding, melt [hot stage microscopy]), high throughput crystallization and co-sublimation techniques. Solution co-crystallization For solution co-crystallization, the two components must have similar solubility; otherwise the least soluble component will precipitate our exclusively. However similar solubility alone will not guarantee success. It has been suggested that it may be useful to consider

polymorphic compounds, which exist in more than one crystalline form as co-crystallizing components. If a molecular compound exist in several polymorphic forms it has demonstrated a structural flexibility and is not locked into a single type of crystalline lattice or packing mode. Thus, the chance of bringing such a molecule into a different packing arrangement in coexistence with another molecule is increased. Clearly polymorphism alone does not guarantee the functionality of a compound to act as a cocrystallizing agent, whilst the ability of a molecule to participate in intermolecular interactions obviously plays a critical role. Slurry Conversion Slurry conversion experiments were conducted in different organic solvents and water. Solvent (100 or 200ml) was added to the co-crystal (20mg) and the resulting suspension was stirred at room temperature for some days. After some days, the solvent was decanted and the solid material was dried under a flow of nitrogen for 5min. the remaining solids were then characterized using PXRD. Melt Method Melts method have also generated an interest in co-crystal formation. By simply melting two co-crystal formers together and cooling, a co-crystal may be formed. If a co-crystal is not form from a melt, a seed from melt may be used in a crystallization solution in order to form a co-crystal. Another phase change in order to form co-crystal is that of sublimation. Sublimation may more often than not form hydrate. Grinding Method (Mechanical Method) Grinding has attracted interest into the formation of co-crystals. Both neat and liquid assisted grinding are techniques employed in order to produce these materials. In neat (dry) grinding, co-crsytal formers are ground together manually using a mortar and pestle, using a ball mill, or using a vibratory mill, in liquid-assisted grinding, or kneading, a small or

substoichiometric amount of liquid (solvent) is added to the grinding mixture. The method was develop in order to increase the rate of cocrystal formation, but had advantages over neat grinding such as increased yield, ability to control polymorph production, better product crystallinity, and applies to a significantly larger scope of co-crystal formers. Pharmaceutical co-crystals can also be formed by use of supercritical fluids. Supercritical fluids act as a new media for the generation of co-crystals. Supercritical fluid technology offer a new platform that allows a single-step generation of particles that are difficult or even impossible to obtain by traditional techniques. The generation of pure and dried new co-crystals (crystalline molecular complexes comprising the API and one or more co-formers in the crystal lattice) can be achieved due to unique properties of supercritical fluids (SCFS) by using different supercritical fluid properties; supercritical CO2 solvent power, anti-solvent effect and tits atomization enhancement. Using intermediate phases to synthesis these solid-state compound are also employed. Through the use of a hydrate or an amorphous phase as an intermediate during synthesis is a solid-state route has proven successful in forming a co-crystal. Also, the used of a metal stable polymorphic from of one co-crystal former can be employed. In this method, the metal stable form acts as an unstable intermediate on the nucleation pathway to a co-crystal. As always, a clear connection between pair wise components of the co-crystal are needed inn addition to the thermodynamic requirement in order to form these compounds.

The table below selected examples of pharmaceutical co-crystals and their method of preparation.
API Aspirin Caffeine Co-crystal former 4,4-Dipyridil Oxalic acid Preparation method Slurry conversion Solven-assisted grinding Cooling crystallization Enhanced property Physical stability Physical stability, dissolution rate and Fluoxetine hydrochloride Indomethacin Benzoic acid Saccharin Solvent evaporation Solvent-assisted evaporation oral Childs et al, 2004 Basavoju et al 2008 bioavailability Intrinsic dissolution rate Physical and dissolution rate Walsh et al 2003 Trask et al 2005 Hickey et al 2007 Reference

Glutaric acid Carbamazepine Nicotinamide

grinding or solvent stability

CHARACTERIZATION OF CO-CRYSTAL Characterization of co-crystals involves both structure (infrared spectroscopy, single crystal x-ray crystallography and powder x-ray diffraction) and physical properties (e.g melting point apparatus, differential scanning calorimetry, thermogravimetric analysis). Single crystal x-ray diffraction is the preferred characterization technique in determining whether a co-crystalline material has been generated; how ever, suitable x-ray quality crystals cannot always be produced. Additionally, even if single crystal can be grown of sufficient size and quality, the exact location of the hydrogen atom (determination if proton transfer has occurred from the acid to the base or not) may be ambiguous. Thus, it is advantageous to utilize a variety of solid-state spectroscopic techniques (Raman, infrared, and solid-state NMR). When attempting to characterize potentially new co-crystalline materials.

Infrared spectroscopy can also be a very powerful tool in detecting co-crystal formation, especially when a carboxylic acid is used as a coformer. PROPERTIES OF COCRYSTALS Co-crystal Structures exhibit long-range order and the Components interact via non-covalent interactions such as hydrogen bonding, ionic interactions, vander waals interactions and resulting crystal structures can generate physical and chemical properties that differ from the properties of the individual components such properties include melting point, solubility, chemical stability, and mechanical properties. Some cocrystals have been observed to exist. As polymorphs, which may display different physical properties depending on the form of the crystals. Phase diagram determined from the contact method of thermal microscopy proved valuable in the discovery of new co-crystals. The construction of these phase diagrams is made possible due to the change in melting point upon co-crystallization. Two crystalline substances are deposited on either side of a microscope slide and are sequentially melted and resolidified. This process creates thin films of each substance with a contact zone in the middle. A melting point phase diagram may be constructed by slow heating of the slide under a microscope and observation of the melting points of the various portions of the slide. For a simple binary phase diagram, if one eutectic point is observed then the substances do not form a co-crystal. If two eutectic points are observed, then the composition between these two points corresponds to the cocrystal as shown by the graph below

A schematic for the determination of melting point binary phase diagrams from thermal microscopy.

CASE STUDIES OF SOME PHARMACEUTICAL COCRYSTAL

The Pharmaceutical Co-crystal studied focus on the formation of pharmaceutical Co-crystal with altered Physical Properties of Clinical relevance. Pharmaceutical cocrystal of carbamazepine (tegretol) Carbamazapine (CBZ) is an important antiepileptic drug that has been in use for over three decades. Oral administration of (CBZ) encounters multiple challenges, including low water solubility with high

dosage required for therapeutic effect (i.e > 100mg/day); dissolution, limited bioavailability and auto induction for metabolism. The formation of CBZ:Saccharin co-crystal appears to be superior to existing crystal from of CBZ in the following respect; stability relative to the anhydrous polymorph of CBZ; favourable dissolution and suspension stability, favourable oral absorption profile in dogs. Cocrystal of theophylline: Theophyline is useful in treatment respiratory disease such as asthma. From the physiochemical standpoint, theophylline represents challenge to formulators in that it is known to interconvert between crystalline anhydrate and monohydrate forms as a function of relative humidity (RH). The possibility of crystalline hydrate formation complicates design of a consistent, reproducible for an API in the drug development process. Reversible hydrate formation is particularly problematic, as it indicates that neither the anhydrate nor the hydrate is fully stable across the range of common processing condition, theophyline is stuctural analogue of caffeine. The cocrystals of the theophylline were prepared with oxalic acid, malonic acid, maleic acid, glutaric acid by solvent evaporation technique. The relative humidity stability comprised of the storage and subsequent PXRD analysis at four specific RH levels (0%, 43%, 75%, and 98%) across four different time points ( 1day, 3days, 1 and 7 weeks). Over the course of 7weeks study it was found that, at 75% RH and below, theophylline anhydrate converted into theophylline monohydrate. No formation of theophylline hydrate was found in any case.

The observed RH stability of theophylline co-crystal demonstrates the physical stabillity improvement, specifically avoidance of hydrate formation. The co-crystals formed by oxalic acid found to be more stable. This study demonstrate use of cocrystals in physical property improvement. Cocrystals of aceclofenac: Aceclofenae is an orally effective nonsteoidal antiinflammatory drug of phenyl acetic acid group, which possesses remarkable antiinflamatory, analgesic and antipyretic properties. Aceclofenac exhibits slight solubility in water and a consequence it exhibit low bioavailability after oral administration. Mutalic prepared cocrystals of aceclofenac by simple solvent change approach by using chitosan. Chitosan has been considered to be one of the most promising biopolymer for drug delivery purpose. Chitosan is a linear hyrophilic polysaccharide polymer of D-glucosammine. It is non-toxic natural polcationic polymer that is degraded by the microflora in the colon. It is abundant in nature and is present in the exoskeleton of crustaceans such as crabs and shrimps. Chitosan has been demonstrated to a be good vehicle for enhancing the dissolution properties and bioavailability of a number of poorly water soluble drugs. Chitosan was precipitated on acceclofenac crystal using sodium citrate as a salting out agent. The pure drug and prepared cocrystals with different concentrations (0.05 to 0.6%) were characterized in terms of solubility, drug content, particle size, thermal behaviour (differential scanning calotimetry, DSC), X-ray diffration (X-RD), morphology, in vivo drug release, stability and phamacokinetic study.

It was observed that particle size of cocrystals was drastically reduced during the formulation process. The DSC showed a decrease in melting enthalpy indicating disorder in crystallinity. XRD also reveealed disorder in crystallinity. The dissolution study showed that marked increase in dissolution rate in comparison to pure drug. The considerable dissolution rate of aceclofenac from optimized crystal formulation was attributed to the welting effect of chitosan, decreased drug crystallinity, altered morphology and micronization. The optimized crystals showed excellent stability on storage at accelerated conditions. In vivo study revealed that the crystals provided a rapid phamacological response in mice and rat; beside improve in phamacokinetic parameters in rats Co-crystal of 5-nitrouracil Co-crystals of 5-nitroracil with solvent molecules, dioxane, pyridine, DMSO, formamide and ethanol as well as with piperazine, N,Ndimethlpiperazine, 3-aminopyridine and diazabicyclooctane obtained by deliberate inclution,have been examined by X-ray crystallography. The tape structure found in the parent centric form of nitrouracil is retained with some modifications in the co-crystals with dioxane, piperazine, diazabicyclo-octane, N,N-dimethylpiperazine, pyridine and DMSO, with the guest molecular tapes exhibit mixed compositions. The observed bonding patterns have been classified into six schemes, interestingly, quadruple type of hydrogen bonding patterns are seen in co-crysals containing 3-aminopyridine or ethanol and water, while a network of acyclic tetrahedral pentamers of water is found in the cocrystal containing diazabicyclo-octane and water. This case study reveals that hydrogen acceptors and donors are necessary to form cocrystal. Co-crystal of indomethacin

Indomethacin, a non-steroidal antiinflammatory drug (NSAID), is widely prescribed for patients with athritis. It exist as , y and amorphous forms, in the solid state -form being most stable form at room temperature. Indomethacin is practically insoluble in water (2.5-4 g/ml; it belongs to BCS class II) and poses sever chalenges in the formulation development. Various cocrystal formers, including saccharin, were used in the screening for indomethacin co-crystals in a series of solvents. Solution evaporation method was used in the screening phase. DSC, TGA, IR, Raman and PXRD techniques characterized the potential new phases. The indomethacin saccharin co-crystal (IND-SAC co-crystal) structure was determined from single crystal X- ray diffraction data. Pharmaceutically relevant properties such as the dissolution rate and dynamic vapour sorption (DVS) of the IND-SAC cocrystal were evaluated. Solid-state and solvent-drop co-grinding methods were also applied to indomethacin and saccharin The IND-SAC co-crystals were obtained from ethl acetate. Physical characterization showed that the IND.SAC co-crystal is unique vis-^-vis thermal, spectroscopic and X-ray diffraction properties. The cocrystals were obtained in a 1:1 ratio with a carboxylic acid and imide dimer synthons. The dissolution rate of IND-SAC was considerably faster than that of the stable indomethacin a-form. DVS studies indicated that the co-crystals gained less than 0.05% in weight at 98% RH. IND-SAC cocrystals were also obtained by solid state and solvent drop cocrystal methods. The IND-SAC cocrystal was formed with a unique and interesting carboxylic acid and amide dimer synthons interconnected by weak N-H-----O hydrogen bonds. The co-crystals were associated with significantly faster dissolution rate than indomethacin (a-form) in phosphate buffer pH 7.4 and were non-hygroscopic.

APPLICATION OF PHARMACEUTICAL CO-CRYSTAL


Compared to other solid-state modification techniques employed by Pharmaceutiical Industry, Co-crystal Formation appears to be an advantageous alternative for drug discovery (e.g new molecule synthesis, nutraceutical co-crystals), drug delivery (solubility, bioavailability) and chiral resolution. Experts are of the opinion that pharmaceutical intellectual property landscape may benefit through co-crystallization.

LITERATURE REVIEW According to Lemmere et al (2010), the active pharmaceutical ingredient 2-chloro-4-nitrobenzoic acid is a potentially novel therapy fro immunodeficiency diseases as an anti-viral and anti-cancer agent, and exists as a dimorph in the solid state. Hot stage contacte method was employed to investigate the potential of preparing a co-crystal with nicotinamide GRAS compound. The 1:1 co-crystal 1 was made using liquid-assisted grinding via a carboxylic acid-pyriding hydrogen bond, while the nic form a centrosymmetric R2(2)(8) dimer to ultimately form a ribbon architecture compared to other known co-crystals of nic. The melting point of the cocrystal is higher than the melting of the pure components, indicating that the pharmaceutical co-crystal is thermally more stable than the pure pharmaceutical compound. The relative stability of the interactions in the cocrystal over the pure compounds supported by molecular modelling calculation. Also, Andrew et al (2005) studied screening for crystalline salt via mechanochemistry and found that neat grinding and solvent drop grinding methods are efffective screening tools for indicating the potential for crystalline salt formation involving a given acid- base pair Two structurally similar APIs, the antibacterial drug trimethoprim (T) and the antimalaria drug pyrimethamine (P) were used with 7

different salt former formate, acetate, malate, fumarate, succinate, glutarate, salicylate. Results of the mechanochemical salt screening experiments, as determined by PXRD analysis are summarized in table below.
NH2 N NH2 N OMe Counterio n Neat grindingb Solventdrop grinding b Neat grinding b Solventdrop grindingb OMe OMe

formate acetate maleate fumarate succinat

+ + X X X

+ X + + +

+ X X X + X

++ + + +

e Glutarate salicylate x

The result shows that neat grinding provide 40% overall screening efficiency, measured as the percentage of experiment that generate crystalline products distinct from physical mixtures of starting materials. Upon performing the grinding salt screen with methanol addition, overall screening efficiency increased from 40% for neat grinding to a remarkable 100% for solvent-drop grinding. Likewise, Wenbing et al (2010) examined the high reactivity of metal-organic frameworks under grinding conditions: parallel with organic molecular material. These known MOFs were prepared by grinding 1,4-benzen dicarboxylic acid with zinc oxide (ZnO) or basic zinc carbonate [ZnCO3]2.[Zn(OH)2]3 in a ball mill in the presence of a small amount of added liquid (100L of H2O, MeOH, or DMF) for 20 minutes. The nature of the added liquid determined the product. This shown according to the diagram below.

The findings improve our insight into the possibilities of grindinginduced transformations and extend the application of grinding as a convenient solvent-free or minimal-solvent method. In accordance with Andrew et al (2004) reported solvent-drop grinding: green polymorph control of cocrytallization. By grinding with a minimal addition of solvent of appropriate polarity, control over the polymorphic outcome of novel cocrystallization involving the model pharmaceutical compound caffeine maybe achieved. Equimolar quantities of anhydrous caffeine and glutaric acid were combined in a stainless grinding jar with two grinding balls and the material was ground together either with or without the addition of a few drop of solvents. The resulting material was then characterized by PXRD. When caffeine and G.A are ground together in the absence of solvent, co-crystal form I predominantly results. Similarly, the addition of four drops from a pipette of a non-polar solvent. Such as n-hexane, cyclohexane, or heptane also produces form I. conversely upon addition of four drops of a more polar solvent, including chloroform, dichloromethane, acetonitrile, and water, the grinding experiment result in predominantly form II

CHAPTER FOUR CONCLUSION Pharmaceutical co-crystals represent a advantageous class of crystal form in the context of pharmaceuticals. Co-crystals of drugs and drug candidates represent a new type of material for pharmaceutical development. Co-crystals are relatively new to pharmaceutical industry and pharmaceutical co-crystals have given a new direction to deal with problems of poorly soluble drugs. Co-crystals have potential to be much more useful in pharmaceutical products than solvates or hydrates. The relevance of co-crystals in API formulation includes the ability of fine-tune physical properties. Characterization of API, identify and develop new, proprietary forms of prescribed drugs and the opportunity to generate intellectual property. Further research is desirable in order to scale up co-crystal systems and implement manufacturing of final dosage forms on commercial scale. Screening for solid forms is important to guarantee that the optimum form is carried forward in development and to minimize the likelihood of unexpected form conversion. Co-crystals- High throughput gives vital information on relationship between formation and chemical structure of the API and co-former. Screening of APIs with library of co-crystal formaers requires further investigations to include all possible coformers. Studies regarding polymorphism of co-crystals should be strengthened in order to accelerate the development of new pharmaceuticals. Additional developments in screening methodology will further elevate the profile of co-crystals on the pharmaceutical and intellectual property landscapes.

Reference: 1.shan N, Zaworotko MJ. The role of co-crysstals in pharmaceutical science. Drug Discovery today 2008; 13:440-446 2. Trask AV, Motherwell WDS, Jones W. physical stability enhancement of theophylline via co-crystallization. Int. J pharm 2006; 320: 114-123. 3. Jones W, Motherwell WDS, Trask AV. Pharmaceutical co-crystals: an emerging approach to physical property enhancement. MRs Bull 2006; 31:875-879. 4. Zaworotko M. Crystal engineering of co-crystals and their relevance to pharmaceuticals and solid state chemistry. Acta cryst 2008; a64: C11-C12. 5. Sun CC, Hou H.

You might also like