throbber
Water—Inso|ub|e
`Drug
`Formulation
`
`I I I
`
`Edited by
`Rong Liu
`
`‘9 Interpharm/CRC
`
`B<)c;1R;lLm1 London New York
`
`\X/'.1shil1gLnr1.I).C.
`
`MYLAN - EXHIBIT 10
`
`MYLAN - EXHIBIT 1025
`
`

`
`Library ol' Congress Cataloging-in-Publication l)ata
`
`
`Wztter-insoluble drug lormulzltion I Rong l.iu, editor.
`p.
`cm.
`'
`Includes l7ll\ll()gl'11[)l]lC1ll relet‘ei1ccs and index.
`ISBN I—574‘)l-l05~8
`
`()U—()33~lS(l
`
`I. Solutions (|’l1arnr.tcy) 2.
`RSZUI .S()W38
`2()()()
`
`6| 5'42~«dt:2 I
`
`|)rugs'—Soluhility,
`
`l. Liu. Jung.
`
`is quoted with
`This book contains inlorinution obtained from authentic and highly regarded sources. Reprinted ll1L1lL’I'lLtl
`permission, and sources are indicated. A wide vmiety of rctcrences are listed. l{ensonuh|e efforts have been made to publish
`reliable data and intbrmution, but the authors and the publisher cannot ztsstllne responsibility for the validity ofall Intttcriuls
`or for the consequences of their use.
`
`Y
`Y
`-
`an metms clccmmic or Illccllallliclll.
`form or h
`in an
`he re noduecd or lI'L1llSl11lllt.‘Ll
`art mat
`Neither this boo]; nor an
`illcludillg [3h0l0CODyirI:_1. Illltlvlllllllllg. and recording, or hy nny inl'orinnlion storage or retrieval system, \vithoul prior
`permission in writing from the puhlis‘lter.
`
`The consent ol'Cl{C Press LLC does not extend to copying for general distribution, for promotion, for creating new works,
`or for resale. Specific permission must be Ol7lLlll1C(l in writing from CRC I’ress LLC for such copying.
`
`Direct all inquiries to CRC l’rcss l.l.(.‘. 2000 N.W. Corpontle l3lvd., lioeu Raton, liloridu 3343 I.
`
`'l'r:t(lenIark Notice: Product or corporate IMIIIICS may he tI':1t|emnI'ks or rcgis‘tcred trntletnmks, und are used only for
`identilication and explanation. without intent to ittfliitgc.
`
`Visit the CRC Press Web site at www.crcpress.c0in
`
`© 2000 by CRC Press LLC
`lnlerphawm is an imprint of CRC Press l.LC
`
`No claim to original U.S. (iovernrnent works
`ll1lCl‘llilll0i1tll St;1Itdm'd Book Number l~574‘)|-105-8
`Lihrziry o|' (‘ongI'ess Curd Nun1l3er()()-l)334S()
`Printed in the United Slnles of America
`I
`2 3 4 5 6 7 8 9 ()
`Printed on acid-lice paper
`
`

`
`Contents
`
`ix
`
`Preformulation Studies
`
`Formulation Development
`
`Methods of Manufacture
`
`Processing Issues
`
`Product Testing and Specifications
`
`Applications
`
`Future Perspective
`
`References
`
`15. Alteration of the Solid State of the Drug Substance:
`Polymorphs, Solvates, and Amorphous Forms
`
`Michael J. Joz,wiukow.s'ki
`
`Theoretical and Practicai Considerations
`
`Special Considerations for Polymorphs
`
`Solvates and Hydrates of Drugs
`
`The Utility of/\morphous (Noncrystalline) Forms
`
`Strategy forWater-Insoluble Drug Formulation Using Metastable
`Sohds
`
`References
`
`501
`
`502
`
`507
`
`511
`
`513
`
`515
`
`516
`
`517
`
`525
`
`526
`
`546
`
`549
`
`555
`
`561
`
`562
`
`16. Solubilization Systems—The Impact of Percolation Theory and
`Fractal Geometry
`569
`
`Hcms L(’Il(’Il]7(’I'_L{(’I‘
`Silvia Komvu El-A rim’
`
`Introduction to Percolation Theory
`
`References
`
`Experiences with a Novel Fluidized Bed System Operating under
`Vacuum Conditions
`
`Applications
`
`An Alternz-1t'ive Novel Process Technology to Obtain Well—soluble
`Substances: Atmospheric Spray—free7.e Drying
`
`570
`
`589
`
`591
`
`596
`
`597
`
`

`
`_l§_
`
`Alteration of the Solid State of the
`Drug Substance: Polymorphs,
`Solvates, and Amorphous Forms
`
`
`
`Michael I. Iozwiakowski
`3M Plzarmacezmeals
`
`St. Paul, M1Tm1.esoIa
`
`The maximum solubility of a drug substance is a function of the nature of the solid
`phase in equilibrium with a specified solvent system at a given temperature and pres-
`sure. Solubility is an equilibrium constant for the dissolution ofthe solid into the sol-
`vent and thus depends on the competition of solutezsolvent
`interactions and
`solid:solid interactions. Alteration of the solid phase of the drug substance can influ-
`ence its solubility and dissolution properties by affecting the molecular interactions
`in the solid.
`
`A crystal of higher free energy will yield an apparent higher solubility than a lower
`energy stable crystal form of the same molecular structure. In the lowest energy solid
`state, the energetically favorable solidzsolid interactions reduce the escaping tendency of
`the molecules, and thus fewer molecules dissolve in a given solvent under the same set
`of environmental conditions. Crystalliiie polymorphs, solvates and hydrates, and amor-
`phous forms of drug substances have been used to change the thermodynamic driving
`force for dissolution and to increase the apparent solubility of poorly soluble drugs.
`Unlike solul)ilizati()n techniques that change the nature of the solvent environ-
`ment (cosolvent systems, emulsions, micellization) or the chemical identity of the dis-
`solved solute (salt formation, complexation, pro-drugs), manipulation ofthe solid state
`of the drug substance results in only a transient change in the system. Since the solvent
`
`525
`
`

`
`526 Water-Insoluble Drug For/rmlrifiorz
`
`and the chemical form are identical, the system will ultimately revert to the lowest-
`energy solid phase in equilibrium with the solvent, with the lowest solubility. (Irystal
`growth and dissolution have been used to assign relative physical stability for poly-
`morphs by observing the direction of the transformation under a microscope under
`controlled temperature in contact with a solvent. The rate of tt'ansfor1nation in contact
`with a solvent is normally too fast to consider solution or suspension dosage forms of
`metastable solids. Systems with unusually large energy barriers, slow reversion kinet-
`ics, or excipients to retard crystallization can be useful in litnited circumstances.
`The most practical use ofthis technique is to alter the solid phase in dry dosage
`forms where molecular mobility is greatly reduced. Metastable forms of solid drugs
`are often stable to physical transformation in the time context required for marketable
`formulations. Tablets, capsules, lyophilized powders, granules for constitution, and
`other solid dosage forms are ideal systems for incorporation of metastable solid
`phases. In most cases, the brief exposure to gastrointestinal (GI) lluids does not result
`in conversion to the lower solubility form prior to generating the desired enhanced
`effect. Solid-state transformations and transformations induced by adsorbed water
`during long—term storage can still l)e problematic. Any consideration of formulating
`metastable solid phases must balance the expected gain in efficacy with the potential
`for reversion to the less—favorable form prior to patient use. This involves both an
`understanding of the phase diagrams (which forms are physicaily stable under which
`conditions) and the physical principles governing transformation kinetics.
`in this chapter,
`the theoretical and practical considerations for the use of
`metastable solids in formulations to gain a solubility or dissolution-rate advantag ' are
`explored. lixperiments are suggested that identify the potential solid forms of the drug
`and elucidate the potential advantages and disadvantages. Specific examples of the
`degree ofenhancement that can be expected and special considerations for each type
`ofsolid are covered (polymorphs, solvates, and amorphous forms).
`
`THEORETICAL AND PRACTICAL CONSIDERATIONS
`
`Importance of the Solid State of the Drug
`
`()rigz'n of the hjfect ofsolizl State on S0lubz'lity
`
`When a medicinal chemist discovers a new chemical entity (NCIE) with a desired phar-
`macological effect, structure-activity relationships are used to optimize the series for
`activity. Aqueous solubility, partition coefficient, crystallinity, melting point, particle
`size, and hygroscopicity, all of interest to the formulator of this NCIE, will also vary
`within the series ofdrug candidates. Because the biological activity is often estimated
`by target enzyme binding studies in very dilute media, solubility may not be opti-
`mixed simultaneously. If an ioni'/.al)le drug candidate is selected, the choice of free
`acid/base form versus the salt forms again produces a myriad of possible physical
`properties. 'l‘he alteration of solubility by judicious choice of the salt was covered in a
`
`

`
`/llI'emt’iorz oft/re Solid State oftlze Drug Subslarzcc
`
`527
`
`previous chapter. ln 111any cases the chosen salt or acid/base can crystallize in a vari-
`ety of possible arrangements, each of which l1as the possibility of different‘ physical
`properties. This includes the possibility of polymorphs, solvates, or noncrystalline
`(amorphous) forms. Thus, the solid phase chosen for development is the third deci-
`sion tnade by the pharmaceutical scientists that has a major impact on the ultimate
`physical properties ofthe NCIE, including the solubility. ln a typical development pro-
`gram, the number of candidates decreases at each stage:
`
`Stage 1. Selection of best chemical structure (1()0-1000)
`
`Stage II. Selection ofacid/base/salt form (3-25)
`
`Stage lll. Selection ofsolid phase for development (1-3)
`
`Each of these stages produces molecules ofvarying solubility by virtue of a change in
`the crystalline lattice, the last stage being the only one in which the chemical identity
`or counterion identity is unchanged.
`Yalkowsky (1081) has developed equations describing solubility as a function of
`both hydrophobicity and crystal lattice forces. This has led to the observation that
`melting point or heat effusion, both a function of the strength of forces holding mole-
`cules together in the solid state, can correlate with solubility within a homologous
`series. This is because the disruption ofthe crystal forces is a necessary prerequisite to
`the release ofindividual molecules into the solvent for dissolution. Grant and I-Iiguchi
`
`(1990) sunnnari'/.ed correlations in the case of diphenylhydantoin derivatives and
`substituted pteridines in their book on organic compound solubility. Wells (1988) has
`noted that in the series of phenols with hydroxy substituents, the high-melting para-
`form (hydroquinone) has a much lower solubility than the ortho- or meta-derivatives.
`Morelock et al. (1994) have used melting points and retention times to correlate with
`aqueous solubility in a series of reverse transcriptase inhibitors. They have found this
`useful in selecting the drug candidate that can possess optimum biological parame-
`ters and in guiding the further synthetic effort. The same factors can govern the solu-
`bility differences between drug-salt forms or solid phases, since, fundamentally, the
`change is brought about by a difference in solidzsolid forces in each case. Wells (1988)
`has shown that riboflavin polymorphs follow a similar inverse correlation with melt-
`ing point. Form lll melts at 180° to 185°C and is soluble to greater than 1000 mg/ml. in
`water. Forms 1 and ll melt at 270° to 290°C and have solubilities ofless than 100n1g/n1L.
`The success of altering solubility by manipulating the solid phase of the drug
`depends on which factor dominates the aqueous solubility behavior, the hydropho-
`bicity, or the lattice forces. When the molecule is too lipophilic to have adequate
`aqueous solubility, cosolvents, pro-drugs, or emulsions are effective in increasing the
`solubility. Altering the solid state i11 this case may have little effect on it.s solubility,
`since poor aqueous solubility is due to the molecular lipophilicity. In contrast, cosol-
`vents and emulsions do not have much impact if the reason for low solubility is the
`stability of the crystal lattice. When a drug has low solubility and a high melting point
`(>250°C), it is likely that disruption of the lattice is needed to increase the effective
`
`

`
`528 Water- Insoluble Drug I<'orm.ulm.‘iorz
`
`form (polymorphs or
`solubility. This can be done by altering the crystal
`solvates/hydrates) or by producing the amorphous form and stabilizing it to sponta-
`neous crystallization. The effect of altering the salt form of the drug is discussed in a
`separate chapter and has also been reviewed by Berge, Highly, and Monkhouse
`(I977).
`
`Historical Perspective and Defirzirimzs
`
`The effect of the solid state on drug solubility has been known for decades, and pio-
`neer articles by llaleblian and McCrone (1969); Shefter and lliguchi (1963); and
`Higuchi et al. (1963) have formed the basis for furtherstudies in this area. More recent
`reviews by Shefter (1981), /\bdou (I989), Byrn (1982), Wall (1986), Brittain (1995), and
`Fiese and liagen (1986) have summarized the effects ofpolyrnorphism or solvate for-
`mation on drug solubility. The existence ofdifferent internal crystalline arrangements
`for the same chemical structure has been termed polyIIz01'plu'.s‘m. Verma and Krishna
`(1966) have observed that the great majority ofsubstances seem to be capable of1nul-
`tiple solid states. The work of Kuhnert—Brandstatter in steroids and barbiturates (as
`reviewed by llaleblian and McCrone 1969) seems to indicate that simple organic drug
`molecules with multiple functional groups can arrange into numerous crystal-
`packing structures. McCrone, McCrone, and Delly (1987): Carstensen (1993); and Byrn
`(1982) have reviewed the seven different crystal systems, which are uniquely identi-
`fied by the length ofthe axes and the angles between them in the unit cell, Many drugs
`crystallize into multiple polymorphic forms, especially monoclinic,
`triclinic, or
`orthorhombic types (Wall 1986; Borka and llaleblian I990; Giron 1995); this relatively
`common diversity in solid forms gives the formulation scientist variations in physical
`properties to exploit.
`in general, the crystalline form with the closest packing (greatest density) and
`the highest melting point is the stable form. The stable form will have the lowest sol-
`ubility and the lowest free energy of the different solid phases of the drug. All other
`phases with the same composition are termed metastable forms at this temperature
`and pressure. in practical terms, the energy barrier for conversion to the stable form
`can be high enough that the metastable forms can be examined and formulated. This
`is especially true if the free energy difference is small (leading to a small driving force)
`or ifsignificant bond breaking, molecular motion, and bond formation are required
`for transformation.
`
`if the crystals contain solvent molecules within the lattice structure in defined
`locations and stoichiometry, these are referred to as solvates (hydrates if the solvent is
`water). The term psemlopolymorph has been used historically but is not as specific
`and should be avoided if the composition is known. in most cases, the solvated form
`of the drug is the least-soluble form in that solvent (e. g., hydrates are the low-energy
`forms in water). Due to additional mixing terms, solvates are often more soluble in a
`solvent of different composition than the nonsolvated crystal (Shefter and lliguclii
`1963). The types ofsolvates that appear in pharmaceutical systems and their proper-
`ties of interest are covered later in this chapter.
`
`

`
`/lllerarimz ofthe Solid State of the Dmg Sitbslmice 529
`
`Amorphous forms can be made for most pharmacettticals by producing the solid
`form faster than the molecules can arrange into a crystalline lattice. Noncrystalline
`solids may have some short-range order but lack the long-range periodicity and reg-
`ular intertnolecular bonding ofcrystalline solids. Their synthesis and properties differ
`markedly from their crystalline forms, as will be described in detail in the section on
`amorphous drugs. In general, amorphous forms are high-energy, low-density solids
`that can yield transient dissolution rates much greater than crystalline solids.
`Liqnirl crystals are an intermediate state in which the molecules in a crystal can
`undergo a secondary phase transition to a mesophase, which gives them tnobility in
`one or two directions. They are birefringent but possess [low properties like a liquid
`phase.
`l.yotropic liquid crystals form upon uptake of water into a system, which
`increases its tnobility, and thermotropic liquid crystals can be disrupted by heating
`above a transition temperature. Cromolyn sodium (Cox, Woodard, and McCrone
`1971 ), the l lM(}-(Jo/\ reductase inhibitor SQ 33(S()() (Brittain, Ranadive, and Serajuddin
`1995), and the leukotriene 1)‘! antagonist 1. 660,711 (Vadas, ’l‘oma, and Zografi 199])
`are examples ofpharmaceuticals that can form liquid crystals.
`The crystal lmbit or external shape may differ when drugs are recrystallized frotn
`different solvent systems without changing the internal structure. The presence of
`additives, the rate ofcooling, the degree ofagitation, and the degree ofsaturation can
`all affect crystal habit (Byrn 1982). llabit can affect bulk properties such as density and
`flowability ((Iarstensen 1993) or influence the ability to filter crystals during purifica-
`tion. Chow and Grant (1988) have shown that the dissolution rate ofacetaminophen
`can be altered two or three times by modifying the length-to~wid'th ratio through
`incorporation of additives. In general, habit effects on solubility are transient and ofa
`magnitude eqttivalent to particle-size—reduction techniques.
`In summary, most drugs are developed as crystalline forms, which have the
`greatest physical and chemical stability, so that their purity can be increased during
`recrystalli7,ation. Knowledge ofthe potential polymorphic forms may allow the devel-
`opment. scientist
`to find a metastable form with the prerequisite stability and
`increased dissolution rate to make it the desirable marketed form. Anhydrate forms
`usually give faster dissoltttion rates and higher aqueous solubilities than the hydrated
`form. Other solvates are not commonly used in pharmaceutical systems due to poten-
`tial toxicity of the solvent but may provide additional solubility enhancetnent. /\mor—
`phous forms have the highest free energy with the greatest degree of solubility
`enhancement but are the most difficult to stabilr/.e against transformation to the sta-
`
`ble crystal form. The remainder of the chapter describes the advantages and disad-
`vantages ofeach type ofsolid and gives numerous examples from the pharmaceutical
`literature where alteration of the solid form resulted in increased solubility.
`
`Methods to Stmly the Solid State dmingPrefi)rm.uI(1tion Screening
`
`After initial selection of the candidate drug and its salt form (if applicable), it is rec-
`ommended that a purposeful effort be undertaken to examine the solid states avail-
`
`able for development consideration. In the absence ofsuch programs, new crystalline
`
`

`
`530 Water-Insoluble Drug F0mmlcm.'0n
`
`forms may be discovered by accident from precipitation of less-soluble phases or a
`change in appearance of the bulk drug during scale—up. lfsufficient formulation, ana-
`lytical, and toxicology work has been completed and it becomes necessary to change
`the solid state, it may significantly delay the development program. ll‘ tl1e newly (lis-
`covered form is the more stable modification, it may be difficult to reproduce the
`metastable form. Carstensen (1993) noted that the diazepam tablet development pro-
`gram was complicated by the crystallization of a more stable polymorph after clinical
`trials had begun. If both forms can still be synthesized and they prove to be bioequiv-
`alent,
`the consequences of changing the form later in development are greatly
`reduced.
`
`A recent publication by Byrn et al. (1995) suggests a minimal preforrnulation
`program for examining the drug solid state and the regulatory implications ofvarious
`scenarios. The information that may be useful at this early phase includes the
`
`- Number ofsolid phases that exist for this drug
`
`- Relative physical and chemical stability of these phases
`
`0 Solubility ofeach form in relevant media
`
`- Resistance of metastable forms to conversion during processing
`
`0 Possible means to stabilize amorphous or metastable forms, ifneeded
`
`Another major consideration at this point is to decide on the patentability of any
`new crystal forms discovered to have significant practical advantages over those in the
`original patent. Byrn and Pfeiffer (1992) have listed >350 patents on crystal forms in
`the pharmaceutical patent literature granted for showing advantages in terms ofsta-
`bility, formulation, solubility, bioavailability, purification, hygroscopicity, preparationl
`synthesis, recovery, and prevention ofprecipitation.
`‘
`Different crystal forms can be sought by recrystallization experiments varying
`the solvent‘ system, temperature, precipitation method, and level ofsupersaturation.
`Precipitation methods may include slow evaporation of the solvent, addition of anti-
`solvents, or saturation at high temperatures followed by cooling. The solvent systems
`chosen for study are one of the key aspects of these experiments. Water must be
`included because ofits physiological significance, the possibility ofcontamination of
`crystallization solvents, and the possibility of moisture uptake during storage. Cer-
`tainly the usual recrystallization solvents for the drug synthesis and purification
`scheme and any alternate systems that may be used for scale—up of this process
`should also be studied. Mixtures used in the final steps ofthe synthetic process should
`be examined, including azeotropes and solvents with small amounts of miscible
`wat.er. A suggested list for solvents that have been known to produce different crys-
`talline forms is provided in Table 15.1 (Wells 1988; Byrn and Pfeiffer 1992; Byrn et al.
`1995).
`
`Solid forms isolated from these systems should be subjected to characterization
`techniques such as hot-stage or polarized light microscopy, differential scanning
`calorimetry (DSC),
`thermogravimetric analysis (TGA), X—ray diffraction (XRD),
`
`

`
`
`
`/tltemriou ofrhe Solid State ofllie Drug Substrmce 53]
`
`Table 15. 1
`
`Partial List of Solvents for Preformulation Studies Looking
`for Different Crystal Forms
`
`Water
`
`Ethanol
`Acetone
`
`Methanol
`
`lsopropanol
`Acetonitrile
`
`lithyl acetate
`Dimethylformamide
`Diethyl other
`
`llexane
`Methylene chloride
`Glacial acetic acid
`
`Any other solvents used in the last steps ofsynthcsis
`Aqueous mixtures with the above
`
`infrared spectroscopy (IR), t7’l‘-Raman spectroscopy, and solid-state nuclear magnetic
`resonance (NMR). The solubility oleach new form isolated should be studied in water
`or the solvent/solution of pharmaceutical interest. Composition versus vapor pres-
`sure data should be obtained to understand the stability of any hydrated forms with
`respect to the anhydrate and other stoichiometric hydrates. if the transition to the
`stable form is last, intrinsic dissolution measurements can be used to estimate the rel-
`
`ative solubilities of new crystal forms.
`’l‘he method used to identify new solid phases depends in part on the expertise
`of the investigator and the availability olequipment and in part on the properties of
`the solid phases. llaleblian and McCrone (1969) have demonstrated the power of the
`polari7.ed light microscope and hot-stage microscope in studying the phase relation-
`ships between solid forms versus temperature. Byrn (1982), Wall H986), and Surya‘-
`narayanan (1989) have noted that only X-ray tlillraction and single crystal X-ray stud-
`ies uniquely identify a solid phase unambiguously, since other methods depend on
`properties that may or may not change with a change in crystal lattice structure. if the
`new phase is a solvate, additional techniques are needed to identify the solvent of
`crystalli7.ation and its stoichiometry. Differential scanning calorimetry has been used
`extensively in polymorph investigations since the melting point is often the first ll‘t(ll-
`cation of a new crystalline form (Giron 1995). Other phenomena such as solid~st'atC
`transitions and water-loss endotherms a1'e often discernible horn the thertnograni.
`l.indenbaum and Mc(.}raw (1985) have shown how solution calorimetry can be used
`to assign relative enthalpy differences between polymorphs of drugs. It is best to use
`a combination of techniques to elucidate the nature ol‘ the new crystal forms, as one
`teclmique may not be able to dil'l'erentiate the forms. Fol‘ example, the two polymor~
`phic forms ol‘ amiloride IICI dihydrate could not be distinguished by IR spectra or
`microscopic morphology (lo7.wiakowski, Williams, and llathaway 1993) but are easily
`identified by X-ray dit't‘raction.
`ll‘ a metastable form is identilied that may be needed to produce the desired or
`optimal pharmacological effect, further studies should be done to define the relative
`stability of this form and the proper storage conditions to prevent conversion.
`/\
`metastable form should not be used when the more stable crystalline form produces
`
`

`
`532 W(tt'er—1/zsoluble Drug Formulation
`
`the desired effects; in this case the potential benefits probably do not offset the risk of
`conversion between production ofthe drug product and patient use. The stresses dur-
`ing manufacture of the product need to be studied as well, since compression,
`milling, granulation, and lyophilization can all change the solid state of the drug
`between synthesis and formulation. in some cases, appropriate precautions, resistant
`packaging, or stabilizing excipients can keep the drug in the optimum form despite its
`rnetastability. Once some of these properties are known, a rational decision can be
`made on whether to proceed with development ofa metastable solid based on a ther-
`apeutic need.
`
`Properties Dependent on the Solid State
`
`Once the different solid phases have been identified, their physical properties can be
`compared to find the form that is optimum for drug product development. Verma
`(1966) described polymorphic forms ofcommon substances that illustrated how their
`physical properties can be quite different. The cubic form of carbon (di2l1n0l1d) is
`hard, dense (3.5 g/cc), brilliantly clear, and a poor conductor. in contrast, the hexago-
`nal form of the same element (graphite) is soft, less dense (2.2 g/cc), dull in appear-
`ance, and a good conductor. Optical properties of different solid forms can change
`their color; mercuric iodide is red (tetragonal form) or yellow (orthorhombic form).
`Dt'ug substances, which tend to be larger organic molecules with multiple
`hydrogen bonding sites, are especially prone to different crystalline arrangements
`that produce variable physical properties. Table 152 lists physical and chemical prop—
`erties ofpharmaceuticals that have been cited in the literature as depending on the
`solid state of the drug. Some of these may provide significant advantages in drug
`development. Form 1 of celiprol ll(2l is much less hygroscopic than the form ll poly-
`morph (Narurkar et al. 1988). Form A chlorpropamide forms tablets with greater l1ard—
`ness than those ofform C under identical compression forces (Matsumoto et al. 1991).
`Phenobarbitone also has crystalline forms that differ in compressibility profiles (Shell
`1963). The two polymorphs of methylprednisolone have (lifferent chemical stability
`profiles when exposed to identical temperatures and humidities of storage (Munshi
`and Simonelli 1970). Crystal size and shape can alter the filterability or syringeability
`ofsuspensions and affect the weight uniformity of tablets and capsules.
`
`Properties of Drug Substances Dependent on the Solid State
`
`Table 15.2
`
`Solubility
`Dissolution rate
`Bioavailability
`Chemical stability
`lilowability
`Melting point
`Compressibility
`Particle size/shape
`Density
`llygroscopicity
`Suspension viscosity
`llilterability
`(joloy
`Tablet hardness
`:%__:_,
`
`

`
`/lflerarion ofrhe S0lt'(f Stare ofthe Drug SuI)smnce 533
`
`While there are numerous examples of miscellaneous physical property differ-
`ences between different solid phases of the same drug substance, the excess free
`energy of metastable states is the most important. The higher energy state, a conse-
`quence of decreased crystal lattice energy, produces a greater molecular mobility and
`thermodynamic escaping tendency in metastable solids. This leads to faster dissolu-
`tion t'ates and greater solubilities, which can have formulation and therapeutic i1npli-
`cations for pharmaceuticals. Drugs with poor aqueous solubility are more likely to
`show enhanced bioavailability when metastable solids are used, because their oral
`absorption tends to be dissolution-limited.
`
`Advantages of Using Metastable Solids
`
`f)is.s‘0lIl.ti0Iz Rate Irrrpr'or/emertli
`
`The Noyes-Whitney equation for the dissolution ofsolids into a solvent (Noyes and
`Whitney 1897) can be used to calculate the rate ofdrug concentration Cincrease with
`time 1:
`
`(IC
`(it
`
`.
`= K/l C ~— C
`i
`4,
`
`J
`
`(l)
`
`where A is the surface area of the solid exposed to solvent, C‘ is the saturation con-
`centration or solubility, and Kis a constant including the diffusion coefficient of the
`solute, the thickness of the unstirred diffusion layer, and the volume ofsolvent. Rotat-
`ing die methods have been developed, such as the Woods die apparatus, which main-
`tain a constant surface area during the initial phase of dissolution experiments. Under
`sink conditions, where C‘ >> C, and with constant surface area /1, the intrinsic disso-
`lution rate (IDR) can be obtained. The [DR (units mg/cin“ /min) is directly propor-
`tional to solubility and depends on the intrinsic dissolution properties of the drug in
`the media, not the dissolution method:
`
`5
`IDR: K(C)
`
`(2)
`
`Changes in the solid state can influence the dissolution rate through the surface
`area term or the solubility term. Surface area differences can arise from simple particle
`size effects between different crystal fornts and also from shape factors. Different crystal
`habits and shapes can alter the exposed surface area without a change in median parti-
`cle size measurements, since these are often calculated by methods that assume spher-
`ical shapes. Abdou (1989) has reviewed the effect of crystalline state on the dissolution
`rate ofpharmaceuticals and how this contributes to bioinequivalence ofvarious forms.
`l)il’ferences in solubility between different crystal forms alter the driving force for
`dissolution, controlled by the difference between the solution concentration and the
`saturation concentration (C5 — C). Hamlin, Northam, and Wagner (1965) have shown
`that dissolution rate correlates well with solubility for a large number of pharmaceu-
`tical compounds varying in solubility from 0.0] to it) mg/ml. at 37°C. Nicklasson and
`
`

`
`0034
`
`0.032
`
`0.028
`
`
`
`S0/ubi/iiy(molar)
`
`0.024
`
`534 Warer~[rrs0lrrbZc Drug Formulatio/r
`
`
`
`Figure 15.1 Dissolution rates for anhydrous and hydrated [3-cyclodextrin in water at
`40° C. [Source.' Reprinted from C(u'b0hydr(rle Research, vol. 143, by Jozwiakowski and
`(Jonnors, Aqueous Solubility ol"l‘hree (Iyclodextrins, pp. 51-59, Copyright 1985, with
`permission lrorrr Elsevier Science]
`
`r-—-4 Anhydrous
`
`0——O Hydrate
`
`90
`
`Time (min)
`._j.______.
`
`Brodin (1984) have shown that using cosolverrt nrixtures for drugs with poor aqueous
`solubility produces a good correlation between dissolution rate and solubility.
`The dissolution rate improvement for rrrost rrretastable solids is only trarrsient;
`ultimately the excess solid in equilibrium with the solvent converts to the lowest
`energy phase. lligure 15.1 tlozwiakowski and Connors 1985) illustrates a typical disso-
`lution pr'ol“ile for a metastable solid and a stable solid. In this case, [3-cyclodextrin dis-
`solution proliles were measured in 40°C distilled water and plotted on a molar basis
`(where the molecular weight dil‘l'erence is inconsequential). The c.orrcerrtration of the
`stable form in water at room temperature (:1 dodecalrydrate) gradually increases to
`the limit of its solubility (().()298 M).
`The rnetastable form (the arrlrydrate from oven drying) shows a rapid initial dis-
`solrrtiorr rate over the lirst 10 min, peaks at less than 30 rrrirr, and their declines to the
`solubility lirrrit as the excess solid converts to the hydrated l'or'm, as verified by
`microscopy. The same behavior is exhibited by metastable and stable polymorphic
`lorrrrs, e.g., form I and form ll ol’rrreprobanrate (Clernerrts and Popli 1973) and lornr I
`and form lI olgepirorre ll(Il {Behme et al. 1985). Ultimately, the sarrre eqrrilibriunr sol-
`ubility will be reached regardless of the direction of approach, although the kinetics of
`this transition can vary considerably for dr'l‘l‘erent drugs.
`Dissolution rate irnproverrrerrt is often characterized by the intrinsic dissolution
`rates or the change in the initial rate, since in many cases the advantage decreases
`with time. Table 15.3 shows sorrre data from the plrarmaceulical literature that iridi-
`cates the typical dissolrrtion rate increases that can be obtained by altering the solid
`
`

`
`
`
`/llrermion oftlie Solid State of the Drug Su.1)sIancc 535
`
`Table 15.3
`
`Comparison of Initial Dissolution Rates of Drugs upon Alteration
`of the Solid State
`
`Drug
`
`Solid Form
`
`Relative Rate
`
`Reference
`
`Sulphathia7,ole
`
`'l‘egal"ur
`
`Dillunisal
`
`ll
`l
`III
`
`on
`[3
`y
`l
`II

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket