Sunteți pe pagina 1din 17

Novel Furosemide Cocrystals and Selection of High Solubility

Drug Forms
N. RAJESH GOUD,1 SWARUPA GANGAVARAM,2 KUTHURU SURESH,1 SHARMISTHA PAL,3 SULUR G. MANJUNATHA,3
SUDHIR NAMBIAR,3 ASHWINI NANGIA1,2
1
School of Chemistry, University of Hyderabad, Prof. C.R. Rao Road, Central University PO, Gachibowli,
Hyderabad 500 046, India
2
Technology Business Incubator, University of Hyderabad, Prof. C. R. Rao Road, Central University PO, Gachibowli,
Hyderabad 500 046, India
3
Pharmaceutical Development, AstraZeneca India Pvt. Ltd., Bellary Road, Hebbal, Bangalore 560 024, India

Received 21 July 2011; revised 24 September 2011; accepted 13 October 2011


Published online 11 November 2011 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.22805

ABSTRACT: Furosemide was screened in cocrystallization experiments with pharmaceutically


acceptable coformer molecules to discover cocrystals of improved physicochemical properties,
that is high solubility and good stability. Eight novel equimolar cocrystals of furosemide were
obtained by liquid-assisted grinding with (i) caffeine, (ii) urea, (iii) p-aminobenzoic acid, (iv)
acetamide, (v) nicotinamide, (vi) isonicotinamide, (vii) adenine, and (viii) cytosine. The prod-
uct crystalline phases were characterized by powder x-ray diffraction, differential scanning
calorimetry, infrared, Raman, near IR, and 13 C solid-state NMR spectroscopy. Furosemide–
caffeine was characterized as a neutral cocrystal and furosemide–cytosine an ionic salt by
single crystal x-ray diffraction. The stability of furosemide–caffeine, furosemide–adenine, and
furosemide–cytosine was comparable to the reference drug in 10% ethanol–water slurry; there
was no evidence of dissociation of the cocrystal to furosemide for up to 48 h. The other five
cocrystals transformed to furosemide within 24 h. The solubility order for the stable forms is
furosemide–cytosine > furosemide–adenine > furosemide–caffeine, and their solubilities are
approximately 11-, 7-, and 6-fold higher than furosemide. The dissolution rates of furosemide
cocrystals were about two times faster than the pure drug. Three novel furosemide compounds
of higher solubility and good phase stability were identified in a solid form screen. © 2011 Wiley
Periodicals, Inc. and the American Pharmacists Association J Pharm Sci 101:664–680, 2012
Keywords: bioavailability; co-crystal; crystallography; dissolution; furosemide; solid dosage
form; solubility; stability; thermal analysis; x-ray diffractometry

INTRODUCTION are classified into four categories in the Biopharma-


ceutics Classification System3,4 (BCS): class I (high
Poor aqueous solubility is a major bottleneck in the
solubility, high permeability), class II (low solubility,
development of new drug molecules as pharmaceuti-
high permeability), class III (high solubility, low per-
cal formulations.1 More than 80% of marketed drugs
meability), and class IV (low solubility, low permeabil-
are sold as tablets; 40% drugs in the marketplace have
ity). Low solubility drugs are those with a concentra-
poor solubility and, more alarmingly, almost 80–90%
tion of <20 mg/L in water. A well-accepted parameter
of drug molecules that are at advanced stages of drug
for solubility is the dimensionless quantity D0 , or
development will pose a solubility problem.2 Drugs
dose number. D0 is the ratio of the highest drug
dose strength in the administered volume (taken as
Additional Supporting Information may be found in the online
version of this article. Supporting Information 250 mL = a glass of water) to the saturation solubil-
Correspondence to: A. Nangia (Telephone: +91-40-23134854; ity of that drug in water (measured in mg/L). D0 < 1
Fax: +91-40-23012460; E-mail: ashwini.nangia@gmail.com); for high solubility drugs and D0 is 25 to 100 for low
Sharmistha Pal (Telephone: +91-80-66213000; Fax: +91-80-
23621214; E-mail: sharmistha.pal@astrazeneca.com)
solubility drugs. This value can go as high as 1000
Journal of Pharmaceutical Sciences, Vol. 101, 664–680 (2012)
or even 10,000. In addition to good oral absorption,
© 2011 Wiley Periodicals, Inc. and the American Pharmacists Association permeability in the gastrointestinal tract is equally

664 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012


NOVEL FUROSEMIDE COCRYSTALS AND SELECTION OF HIGH SOLUBILITY DRUG FORMS 665

important so that the drug can bind to the recep- conditions.36 Even though there was good success in
tor target. The reference standard for defining high achieving faster dissolution rates with furosemide
or low permeability boundary is the n-octanol/water polymorphs, observations such as the transformation
partition coefficient for metoprolol, which has log of these metastable forms to the stable modification
Pow = 1.72. during dissolution experiments or on storage made
Furosemide (Lasix) is a loop diuretic drug com- these results less attractive for further drug devel-
monly used in the treatment of hypertension and opment. The fact remains that furosemide is still
edema. It is a BCS class IV drug,5,6 that is of low marketed in its stable crystalline modification (Form
solubility (6 mg/L in water) and low permeability (log 1)13 despite its poor aqueous solubility. In this back-
Pow 1.4). The highest dose strength of furosemide ground, we carried out a study on pharmaceutical
is 80 mg, which means that it has a D0 number cocrystals37,38 of furosemide with the idea that being
of 53. The practiced approaches to improve the crystalline in nature they are relatively more stable
solubility of furosemide may be classified into two and less prone to accidental phase transformations.
broad categories: (1) altering the physicochemical A pharmaceutical cocrystal is defined as a stoichio-
property of the drug substance and (2) improving metric hydrogen-bonded complex in the solid state
the way in which the drug is processed or formu- between the active drug species and a suitable co-
lated. The cocrystals strategy described in this former molecule that is safe for human consumption
work is currently a popular approach to modify the (selected from the generally recognized as safe (GRAS)
physicochemical properties of drugs.7,8 A closely list39 of U.S. Food and Drug Administration).
related method for solid form modification is that
of fast dissolving furosemide polymorphs,9–13 which
RESULTS AND DISCUSSION
were first characterized more than 20 years ago.
The stabilization of nanoparticles14 and amor- Furosemide was cocrystallized with several coform-
phous phases15 to achieve high dissolution rates ers containing CONH and COOH functional groups
by particle size reduction, cogrinding, and copre- with the intent of making cocrystals. The coformer
cipitation of the drug with crospovidone,16 solid selection was kept as broad as possible to widen the
dispersions with polymers,17 complexation with $ possibility for functional group pairing through hy-
cyclodextrins,18–20 calix[n]arenes,21 emulsification,22 drogen bonds. Non-GRAS molecules were excluded
micelle formation,23 SEDDS (self-emulsifying because our end goal was to make cocrystals for
drug delivery systems),24 micronization and spray pharmaceutical form development. Thus even though
drying,25 etc. have also been reported for furosemide. pyridine-N-oxides are known to form strong and re-
The use of pharmaceutical cocrystals has gained liable heterosynthons with the sulfonamide group,34
increasing popularity in the past decade26–28 as a they were excluded because there is no pyridine-N-
supramolecular approach to improve the physico- oxide compound of the GRAS status.39 We obtained
chemical and pharmacokinetic behavior of drug sub- the following new crystalline phases (Scheme 1) in
stances. Furosemide has three functional groups solution crystallization, manual grinding, and slurry
for hydrogen bonding to make cocrystals: COOH, crystallization screen for novel furosemide cocrys-
SO2 NH2 , and NH. Of these, the carboxylic acid and tals: (i) furosemide –caffeine (FUROS–CAF), (ii)
sulfonamide functional groups are well studied and furosemide –urea (FUROS–UREA), (iii) furosemide-
known to give robust supramolecular synthons29,30 p-aminobenzoic acid (FUROS–PABA), (iv) furosemide
via O H···O and N H···O hydrogen bonds in cocrys- –acetamide (FUROS–ACT), (v) furosemide –nicoti-
tals. Our objective was to use COOH and SO2 NH2 namide (FUROS–NIC), (vi) furosemide –isonicoti-
functional groups in heterosynthons with comple- namide (FUROS–INIC), (vii) furosemide –adenine
mentary coformer molecules, such as pyridine,31 (FUROS–ADEN), and (viii) furosemide –cytosine
CONH2 ,32 NH2 ,33 and pyridine-N-oxide34 , etc. with (FUROS–CYT). The solvents used for cocrystalliza-
the idea that a multicomponent crystalline adduct tion are given in the Experimental section. A com-
will be produced by self-assembly in the solid state. plete list of all coformers attempted with furosemide
Within the domain of solid form modification strate- under different cocrystallization conditions is given
gies, we noted that apart from the metastable in the Supporting Information (Table S1).
polymorphs of furosemide with higher dissolution
Characterization of Cocrystals
rates,9,10 a crystal engineering approach to tune the
physicochemical characteristics of furosemide has not All the above-mentioned crystalline phases were char-
been reported in the literature. Furosemide salts acterized by powder x-ray diffraction (PXRD), in-
with amino acids and their solubility characteristics frared, near infrared and Raman spectroscopy (IR,
were reported in a U.S. patent 5182300.35 Furosemide NIR, Raman), solid-state NMR spectroscopy (ss-
is relatively stable to photodegradation in alkaline NMR), and differential scanning calorimetry (DSC)
medium, but the molecule rapidly degrades in acidic techniques. Microcrystalline powders were obtained

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012
666 GOUD ET AL.

Scheme 1. Furosemide and coformers discussed in this study. The stoichiometry of


furosemide–coformer cocrystal is indicated in each case along with the analytical method used.
Compound abbreviations are used throughout the paper.

in all cases. Diffraction quality single crystals could mated by using 1 H NMR spectroscopy. Finally, solid-
be grown for FUROS–CAF and FUROS–CYT. The state 13 C NMR spectra were recorded to character-
molecular composition and stoichiometry of these two ize all novel cocrystals. PXRD patterns, DSC ther-
cocrystals was unambiguously confirmed from their mograms, and ss-NMR spectra are presented in the
x-ray crystal structures. The nature of the cocrys- paper, whereas IR, NIR, and Raman spectra are dis-
tal–salt continuum,40 or the exact position of the H played in the Supporting Information. The structures
atom in an acid–base crystal structure, is sometimes of FUROS–CAF and FUROS–CYT determined to be
difficult to ascertain except by accurate x-ray diffrac- cocrystal and salt, respectively, by x-ray crystallog-
tion. Solid-state NMR41 and XPS (x-ray photoelec- raphy are discussed first, followed by the remaining
tron spectroscopy)42 are complementary techniques six cocrystals for which single crystal x-ray data are
to study the cocrystal/ salt state. FUROS–CAF was unavailable at the present time.
confirmed to be a cocrystal (neutral complex) and
FUROS–CYT a salt (ionic structure) by x-ray crys- FUROS–CAF
tallography. The exact location of the acidic hydrogen
atom in other furosemide cocrystals could not be con- Crystallization of a ground mixture of furosemide
clusively established due to nonavailability of single and caffeine from a MeOH–MeCN solvent mixture af-
crystals for x-ray diffraction. forded diffraction quality single crystals, which solved
The structures of new multicomponent solid phases and refined in the triclinic space group P-1 (Table 1).
(i–viii) were confirmed by IR, NIR and Raman spec- The crystal structure contains one molecule each
tral analysis of the product cocrystals. This was fol- of furosemide and caffeine in the asymmetric unit
lowed by comparison of fingerprint lines in the PXRD (Fig. S1), which confirms the cocrystal composition
patterns, which showed clear differences in 2θ values. and stoichiometry. The most acidic COOH donor of
A homogeneous cocrystal phase chemically different furosemide is hydrogen bonded to the most basic N3
from the drug and the coformer was indicated by a acceptor of caffeine via an O H···N hydrogen bond
sharp melting endotherm in DSC. The cocrystal com- (Fig. 1a). The primary sulfonamide NH donor hydro-
position was established, and stoichiometry was esti- gen bonds to different caffeine C O acceptor groups
(Fig. 1b), and the secondary NH is bonded to one of the

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012 DOI 10.1002/jps
NOVEL FUROSEMIDE COCRYSTALS AND SELECTION OF HIGH SOLUBILITY DRUG FORMS 667

Table 1. X-Ray Crystal Structure Data of Furosemide–Caffeine electron density maps. The purity of the bulk cocrys-
Cocrystal and Furosemide–Cytosine Salt tal phase was confirmed by an excellent overlay of
FUROS–CAF FUROS–CYT the experimental PXRD pattern with the calculated
lines from the x-ray crystal structure (Fig. 2). Thus a
Empirical formula C20 H18 Cl N6 O7 S C16 H16 Cl N5 O6 S
Formula weight 521.91 441.85 novel cocrystal of furosemide was obtained by liquid-
Crystal system Triclinic Triclinic assisted grinding,43 and solution crystallization gave
Space group P-1 P-1 diffraction quality single crystals of the same com-
T (K) 100(2) 298(2) pound. The furan moiety of furosemide was disor-
a (Å) 7.512(2) 7.909(4) dered in the crystal structure, determined at 100 K
b (Å) 9.462(3) 9.467(5)
(Low Temperature (LT) structure). Since the phar-
c (Å) 17.198(5) 12.484(7)
α (◦ ) 95.387(5) 100.151(8) maceutical cocrystal composition is relevant at room
β (◦ ) 102.271(5) 95.950(8) temperature, we redetermined the structure at 298
γ (◦ ) 110.101(5) 97.644(9) K (Room Temperature (RT) structure). There was no
Z 2 2 change in the proton state (neutral O H···N hydro-
V (Å3 ) 1103.0(6) 904.2(8) gen bond) except that the disorder in the furan ring
Reflections collected 8290 9245
Unique reflections 4045 3489
portion was too severe to assign partial occupancies
Observed reflections 3388 3153 in structure solution. The more accurate LT crystal
Parameters 363 290 structure is reported in this paper.
R1 0.0735 0.0390 The carbonyl peak of furosemide occurs at
wR2 0.1874 0.1070 1673 ncm−1 in the IR spectrum and that of caffeine
GOF 1.065 1.060
CCDC No. 833554 833555
at 1658 and 1700 cm−1 ; the peaks are shifted to 1699
and 1650 cm−1 in the cocrystal adduct. There are dif-
ferences in the N H stretching region between 3200
and 2500 cm−1 . The C N stretch is shifted from 1658
sulfonyl O acceptors (Fig. 1c). Hydrogen bond metrics cm−1 in caffeine to 1650 cm−1 in the cocrystal and the
are listed in Table 2. The COOH group is in a neutral N H bending vibration from 1592 cm−1 in furosemide
state (C O 1.221(5) Å, C O 1.315(5) Å), and the H to 1595 cm−1 in the product. IR, NIR, and Raman
atom is covalently bonded to the OH group and hy- spectra details are presented in the Supporting In-
drogen bonded to the N acceptor (1.66 Å, 177◦ ). The formation (Tables S2, S3, and S4 and Figs. S2, S3,
position of the acidic H atom was located in difference and S4).

Table 2. Hydrogen Bonds in Furosemide–Caffeine Cocrystal and Furosemide–Cytosine Salt Crystal Structures

Interaction H···A (Å) D···A (Å) ∠D–H···A (◦ ) Symmetry Code


FUROS–CAF
O3 H3A···N3 1.66 2.642(3) 177 2–x, 1–y, 1–z
N1 H4A···O1 2.33 3.001(4) 139 x,–1+y, z
N1 H4A···O4 1.82 2.669(7) 123 Intramolecular
N2 H5A···O6 1.87 2.849(3) 164 1 + x, y, z
N2 H5B···O7 1.91 2.899(6) 166 1 + x, 1+ y, z
C1 H1B···N3 2.48 2.937(6) 104 Intramolecular
C1 H1B···O3 2.43 3.484(6) 165 2–x, 1–y, 1–z
C2 H2B···O6 2.31 2.726(5) 100 Intramolecular
C7 H7···O2 2.29 3.270(7) 149 2–x, 2–y, 1–z
C8 H8C···O1 2.30 3.190(7) 138 1+x, y, z
C11 H11···O5A 2.44 3.343(6) 140 Intramolecular
C14 H14···O2 2.32 2.790(7) 104 Intramolecular
FUROS–CYT
N1 H1···O2 1.80 2.605(3) 134 Intramolecular
N2 H2A···Cl1 2.66 3.257(2) 118 Intramolecular
N2 H2A···O6 2.37 3.016(2) 121 1+x, y, z
N2 H2A···O5 2.44 3.349(9) 149 2–x,–y,–z
N5 H4···O3 1.83 2.828(8) 173 1–x,–y,–z
N5 H5···O3 1.89 2.849(2) 157 x, 1+y, z
N4 H6···O2 1.63 2.641(1) 178 1–x,–y,1–z
N3 H7···O6 1.85 2.857(8) 172 1–x,–y,–z
C2 H2···O4 2.38 2.842(7) 104 Intramolecular
C12 H12···O4 2.46 3.413(2) 146 x, 1+y, 1+z
C15 H15···O4 2.45 3.244(6) 129 x, 1+y, z

Neutron-normalized distances are used in the paper.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012
668 GOUD ET AL.

caffeine. Melting data from DSC thermograms are


summarized in Table 3.
The solid-state 13 C NMR spectrum of the cocrystal
showed peaks for furosemide plus caffeine, but the
chemical shifts were moved upfield/downfield relative
to the pure components. Since short-range aggrega-
tion and shielding/deshielding will be different in the
starting components and product species, there are
small but discernible chemical shift differences in the
ss-NMR spectra (Fig. 4; see Table S5 for chemical shift
values). The equimolar stoichiometry of FUROS—
CAF cocrystal was confirmed by 1 H NMR integration
(Fig. S5).
FUROS–CYT
Single crystals of furosemide–cytosine complex were
obtained from MeOH solution. The x-ray crystal
structure of FUROS–CYT was solved and refined in
the triclinic space group P-1 (Table 1). The ORTEP
diagram shows both the molecular components in
the crystal lattice (Fig. S6). The main difference be-
tween this crystal structure and that of FUROS—
CAF is that the proton is transferred from the COOH
group of the drug to the basic N4 nitrogen of cy-
tosine to make an ionic N H+ ···O− hydrogen bond
(1.63 Å, 178◦ ). FUROS–CYT is a salt. The protonation
state of the carboxylic acid and the basic nitrogen in
Figure 1. Hydrogen bonding in FUROS–CAF crys- acid–base structures can be difficult to predict a pri-
tal structure. (a) COOH···N, (b) SO2 NH···O C, and (c) ori in cocrystal structures. There are several cases
NH···O2 S. The terminal furan ring is disordered over two of borderline proton location and even a continuum
sites with 0.65: 0.35 site occupancy factor of C18, C20, and
of O···H···N hydrogen bond states was noted.40,42,45
O5 atoms in the low-temperature crystal structure data col-
The electron density maps of x-ray diffraction showed
lected at 100 K.
that the acidic H atom is transferred and covalently
bonded to the cytosine N and makes a hydrogen
The DSC thermogram (Fig. 3) of the cocrystal bond with the COO− acceptor. The two C–O dis-
showed a single endotherm at a temperature different tances are nearly equal (1.247(2) Å, 1.262(2) Å) in
from that of the components (Tpeak 225◦ C). The post- the carboxylate group. Hydrogen bonding is mediated
melting exotherm above 230◦ C is due to decomposi- by the two-point carboxylate···aminopyridine synthon
tion of furosemide to 4-chloro-5-sulfamoylanthranilic of R2 2 (8) geometry.46 Two furosemide molecules are
acid (saluamine) and other by-products upon heating hydrogen-bonded to two cytosine coformers via the
(Scheme 2).44 The single endotherm of the cocrystal is carboxylate···amine R2 4 (8) motif (Fig. 5). The cytosine
at a higher temperature than that of furosemide but molecules pair up via the carboxamide N H···O dimer
lower than caffeine melting point. Furthermore, the synthon. The bulk crystalline material prepared by
DSC of the cocrystal did not exhibit small thermal liquid-assisted grinding matched with the single crys-
events (e.g., polymorphic phase transitions), which tal phase purity (Fig. 6).
were observed in DSC of commercial furosemide and IR, Raman and NIR spectra (Figures S7, S8, and
S9), DSC (Fig. 7), and NMR spectra (solid state in
Fig. 8 and solution spectrum in Fig. S10) were satis-
factory and consistent with the x-ray crystal structure
analysis. DSC endotherms for the salt appeared at a
different temperature from that for furosemide and
the coformer (Table 3).
FUROS–UREA, FUROS–PABA, FUROS–ACT,
FUROS–NIC, FUROS–INIC, and FUROS–ADEN
Scheme 2. Thermal degradation of furosemide to salu-
amine, furfuryl alcohol, and levulinic acid. This transfor- The next six cocrystal structures are described
mation occurs after melting of furosemide. briefly because the same techniques were used

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012 DOI 10.1002/jps
NOVEL FUROSEMIDE COCRYSTALS AND SELECTION OF HIGH SOLUBILITY DRUG FORMS 669

Figure 2. The experimental powder x-ray diffraction pattern of FUROS–CAF (black) and
calculated lines from the crystal structure (red) show an excellent overlay in 2θ and reasonably
good match of peak intensity. The small difference in peak position could be due to x-ray
reflections being collected at 100 K and the PXRD was recorded at 300 K. Reitveld refinement
gave Rp = 0.1953, Rwp = 0.2500, and Rexp = 0.0257.

as described above. That the structure and com- matched solubility of the components, whereas mi-
position of furosemide–caffeine and furosemide– crocrystalline powders are relatively easy to prepare
cytosine determined by several analytical methods47 by neat or liquid-assisted grinding.48 The IR spectra
matched with the x-ray crystal structure in two cases of these six cocrystals (Fig. S11) showed significant
gave us the confidence to characterize novel cocrystal differences compared to those of the drug and the co-
phases, even in the absence of suitable single crys- former, providing evidence for new crystalline phases.
tal data. Diffraction quality single crystals can be Shifts in the CONH fragment of cocrystal spectra
difficult to obtain for cocrystals, often due to mis- compared with components were observed in NH

Table 3. Melting Points of Furosemide Cocrystals Compared with Those of the Drug and Coformers Used

Drug/Coformer Melting Point of Coformera (◦ C) Cocrystal/Salt Melting Point of New Phase (◦ C)


FUROS 203 — —
CAFb 227 FUROS–CAF 225
UREA 133 FUROS–UREA 156
PABA 187 FUROS–PABA 200
ACT (form 1, 2) 71, 81 FUROS–ACT 123
NICc (form 1, 2) 106, 125 FUROS–NIC 150, 166
INIC (form 1, 2, 3) 161 FUROS–INIC 154, 196
ADEN 360 FUROS–ADEN 218
CYT 320 FUROS–CYT 232
a Multiple melting points are given for polymorphic compounds.
b Melting point of caffeine monohydrate is 234◦ C.
c At least six polymorphic forms of nicotinamide are reported by hot stage microscopy in Kofler L, Kolfer A. 1943., Chem Ber 76:246—248,
having melting points (I) 129, (II) 110, (III) 113, (IV) 111, (V) 110, and (VI) 105◦ C. Melting points quoted in the table are taken from Li et al.50

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012
670 GOUD ET AL.

Figure 3. DSC thermogram of furosemide–caffeine cocrystal. The compound melts at 225◦ C


(Tpeak ) followed by decomposition of furosemide between 227 and 240◦ C.Melting point of
furosemide is 203–205◦ C and of caffeine is 227–228◦ C.

stretching and bending regions as well as for the car- 1


H NMR integration (Fig. S15). The analysis of nicoti-
bonyl stretch peak, for example, in FUROS–UREA, namide and isonicotinamide cocrystals is complicated
FUROS–ACT, and FUROS–INIC. Their Raman and by the possibility of polymorphs and multiple stoi-
NIR spectra are compared in Figures S12 and S13 chiometry. The 1:1 predominant cocrystal composition
(in the Supporting Information). The melting point proposed is consistent with the available data.
of the cocrystal was determined, and the phase pu- Powder x-ray diffraction is a fingerprint technique
rity was ascertained in DSC thermograms (Table 3; to characterize the solid state. This is the most quanti-
Fig. S14). All cocrystals exhibited sharp melting en- tative method for identifying novel crystalline phases
dotherms at temperatures significantly different from when the x-ray crystal structure is not feasible due to
those for the drug and the coformer. The cocrystal microcrystalline nature of the sample. We observed
melting point was lower than that of furosemide for new diffraction peaks in the powder XRD patterns of
urea, PABA (p-aminobenzoic acid), acetamide, nicoti- cocrystals compared with pure furosemide and the co-
namide, and isonicotinamide cocrystals but higher former (Fig. 9a). The calculated diffraction lines from
than the drug melting point for caffeine, adenine, x-ray crystal structures (Fig. 9b) are shown to detect
and cytosine compounds. The intermediate melting the starting materials or polymorphic phases in the
point of furosemide cocrystals, that is in-between the product cocrystal.
drug and the coformer, is consistent with the general To confirm the cocrystal structure and composi-
trend in cocrystals.49 Multiple endotherms were ob- tion, 13 C ss-NMR spectra41 were recorded to discern
served for nicotinamide and isonicotinamide cocrys- short-range order differences and molecular mobil-
tals, and a reason could be that polymorphs of the ity changes. The chemical shift values (δ scale) of ss-
coformer are produced in the cocrystallization ex- NMR spectra (Fig. 10) recorded at cross-polarization
periment by grinding. Polymorphs of nicotinamide magic-angle spinning setting (CP-MAS) are convinc-
and isonicotinamide50 and multiple stoichiometries of ing to confirm cocrystal formation. The δ values are
their cocrystals were reported in recent studies.51,52 presented in Table S5 for a detailed peak-by-peak
The molecular composition of all the multicomponent comparison. The homogeneity of cocrystals and the
phases and their stoichiometry were ascertained by absence of starting materials were established by 13 C

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012 DOI 10.1002/jps
NOVEL FUROSEMIDE COCRYSTALS AND SELECTION OF HIGH SOLUBILITY DRUG FORMS 671

Figure 4. 13 C ss-NMR spectrum of FUROS–CAF cocrystal compared with the individual


components.

ss-NMR spectra. A difference in the chemical shift nent systems are also multifunctional (COOH/COO− ,
peak positions of the cocrystal with respect to the CONH) makes it difficult to assign the carbonyl peaks
individual components was considered as evidence individually. Whereas normally it is relatively easy to
for the formation of a new phase. For example, the differentiate between COOH and COO− by IR spec-
carbonyl group of furosemide moved downfield in troscopy, it would have been difficult to make the
FUROS–UREA (δ 176.9–178.6) whereas the carbonyl same assignment for the above-discussed cocrystal/
group of urea moved upfield (δ 168.2–166.6), indicat- salt without knowledge of the x-ray crystal structures.
ing a hydrogen-bonding interaction between the com- The carbonyl resonance occurred between δ 160 and
ponents. Similarly, carbonyl groups of furosemide and 170 ppm in the NMR spectra for furosemide whether
PABA were shifted in the cocrystal relative to the pure the molecule is in the neutral (COOH) or ionized
components. The carbonyl peaks in FUROS–ACT at (COO− ) state. 15 N ss-NMR41 and XPS42 should un-
(δ 174.7, 180.4) are shifted compared with individual equivocally clarify the cocrystal/salt nature of all the
resonances (δ 176.9, 181.5). Cocrystals of furosemide furosemide adducts, and these measurements will be
with nicotinamide, isonicotinamide, and adenine in- carried out to clarify the ionization state. The main
dicated new cocrystal phases by NMR. objective of this preliminary study was to search for
Of eight molecular complexes prepared in this novel solid-state forms of furosemide with GRAS co-
work, single crystals were obtained for two com- formers to discover crystalline materials of improved
pounds. Furosemide–caffeine is a cocrystal and solubility and stability.
furosemide–cytosine is a salt. The structural details The melting endotherms in DSC for cocrystals
of the neutral or ionic state were known after sin- are sharp and occur in-between the furosemide
gle crystal x-ray diffraction. Surprisingly, the IR spec- and the coformer (Table 3). This indicated cocrys-
trum of furosemide–cytosine did not exhibit a broad tal formation.49 We believe that the examples dis-
carboxylate band at 1600–1650 cm−1 and a sharper cussed in this paper are cocrystals, not eutectic com-
peak for the carboxylic acid group at 1700 cm−1 for positions. Generally, the melting point of eutectic is
furosemide–caffeine. The fact that the multicompo- lower than either of the components and, moreover,

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012
672 GOUD ET AL.

that are metastable and undergo phase transforma-


tion during the slurry conditions of dissolution. The
rate at which the equilibrium solubility is reached is
the dissolution rate, which is a kinetic parameter. The
IDR method overcomes the problems of drug particle
size and morphology effects. Dissolution rate stud-
ies rely on the supersaturation phenomenon, that is
the peak concentration of the drug delivered through
a suitable carrier (e.g., amorphous, salt, cocrystal,
nanoparticle)55 in a short period of time (say 2–4–6
h) depending on the stability of the drug form being
tested. The survival of the starting drug form during
the course of the test conditions and until the end of
the experiment must be verified independently, usu-
ally by PXRD or video microscopy. IDR measures the
rate of drug dissolution from a constant surface area
of the disk in a unit time. Because of the very low
solubility of furosemide in aqueous medium, all dis-
solution and solubility experiments were carried out
in 10% ethanol–water (Fig. 11).
The drug concentration was determined by UV–vis
spectroscopy from a calibrated concentration–inten-
sity curve. Furosemide shows maxima of decreasing
intensities at 230 nm, 275 nm, and a third shallow,
broad maximum at 335 nm. Among the coformers
in this study, caffeine, cytosine, and p-aminobenzoic
acid absorb between 240 and 300 nm but they exhibit
Figure 5. X-ray crystal structure of FUROS–CYT salt. no maxima beyond this region even at the highest
(a) R2 2 (8) and R2 4 (8) N H···O hydrogen bond ring mo-
concentration considered. There is a slight up-curve
tifs. (b) The SO2 NH2 group donors engage in N H···O and
above the base line at 230–240 nm due to the coformer
N H···B(furan) hydrogen bonds.
peaks, but this small contribution is no more than
10% (Fig. S16). This artifact can be corrected using
it does not depend on the stoichiometry in which the the drug:coformer concentration calibration curves.
components were mixed or reacted. This was not the To minimize possible errors due to overlapping peaks,
case for furosemide cocrystals. Another indication for the maximum in furosemide UV–vis spectrum at 335
cocrystal instead of the eutectic phase is the stabil- nm was used to calculate drug concentrations without
ity experiments (discussed next). The PXRD of the any interference from coformer peaks.
stable cocrystals in this study, that is furosemide– The solubility of the drug and cocrystal pow-
caffeine, furosemide–adenine, and furosemide–cyto- ders was determined at 24 h. Apart from
sine, matched with those of the product solid forms, furosemide, which was stable to the slurry condi-
but not with the individual components. This implies tions, furosemide–caffeine, furosemide–adenine, and
that a novel crystalline adduct is present, not a eu- furosemide–cytosine were stable for the same pe-
tectic phase. A physical mixture would have shown riod (Fig. S17); there was no perceptible change in
PXRD lines matching with those of the starting ma- the PXRD pattern even up to 48 h. The solubil-
terials after the slurry experiment. ity of all cocrystals is superior to that of the pure
drug. The other five cocrystals with urea, PABA,
Solubility and Dissolution
acetamide, nicotinamide, and isonicotinamide con-
Solubility and intrinsic dissolution rate (IDR) were verted to furosemide within 24 h. The solubility order
determined on a U.S. Pharmacopeia (USP) approved for the stable cocrystals (no phase transformation ob-
dissolution tester.53,54 A typical measurement is de- served) is furosemide–cytosine > furosemide–adenine
scribed in the Experimental section. Solubility is the > furosemide–caffeine > furosemide, which have 11,
concentration of a substance in the solvent when the 7, and 6 times higher solubility than furosemide. The
dissolved and undissolved particles are in a state of dissolution rate advantage is modest (about twofold
dynamic equilibrium. Solubility is a thermodynamic faster). Since there is no evidence of the solid-form
quantity and usually taken as the concentration of the conversion for extended periods of time, the solubil-
solute at 24 or 48 h after mixing in a solvent. The sol- ity numbers may be used as a safe guide to estimate
ubility measurement is unsuited for those drug forms the higher aqueous exposure and bioavailability of

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012 DOI 10.1002/jps
NOVEL FUROSEMIDE COCRYSTALS AND SELECTION OF HIGH SOLUBILITY DRUG FORMS 673

Figure 6. Experimental PXRD of FUROS–CYT (black trace) overlaid on the calculated diffrac-
tion lines from the x-ray crystal structure (red). The matching of the bulk material with the
single crystal phase is excellent. Reitveld refinement gave Rp = 0.1833, Rwp = 0.2514, and Rexp
= 0.0270.

cocrystals compared with the pure drug. The equilib- the highest solubility of FUROS–CYT does not corre-
rium solubility at 24 h and the dissolution rate from late with the low solubility of cytosine. On the other
the linear region of the IDR curve are presented in hand, FUROS–CAF has lower solubility even though
Table 4. caffeine is more soluble. A reason for these observa-
The reasons for solubility enhancement may be tions is that there are drastic changes in the crys-
described on the basis of the solubility of the co- tal structure and hydrogen bonding from neutral to
former in water (Table 4) and from crystal struc- ionic in FUROS–CAF and FUROS–CYT. We believe
ture analysis (Figs. 1 and 5). The high solubility of that the salt nature of the latter compound guides
FUROS–NIC cocrystal is not surprising56 because its highest aqueous solubility. The solubility data of
nicotinamide is one of the highest soluble coform- Table 4 indicate that FUROS–CAF, FUROS–ADEN,
ers that is used in pharmaceutical cocrystallization. and FUROS–CYT are congruently dissolving systems
Hence the highly soluble coformer guides the high because they are pairs of similar solubility. The other
concentration of the drug cocrystal.57,58 However, this systems are incongruently dissolving due to the huge
explanation, which is based on the linear relation- difference in their components solubilities.59,60 Their
ship between the solubility ratio of the components stabilities are consequently related. Congruent sys-
plotted against the solubility of the former cocrystal tems tend to be more stable in the slurry medium.
divided by the solubility of the API (Active Pharma- In incongruent systems, however, the highly solu-
ceutical Ingredient),57 has its limitations. The above ble coformer causes rapid dissolution and dissoci-
model is faithfully realized only when the cocrystals ation of the cocrystal to its components. FUROS–
whose solubility is being compared have similar hy- PABA is an exception to the above-mentioned
drogen bonding and molecular packing. For example, analysis.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012
674 GOUD ET AL.

Figure 7. DSC of FUROS–CYT. The salt exhibits a melting endotherm at 232◦ C (Tpeak ) fol-
lowed by decomposition of furosemide between 240 and 260◦ C. Melting point of furosemide
is203–205◦ C and of cytosine is 320–322◦ C.

CONCLUSIONS ture and the neutral/ionic state of the product are


difficult to know without a crystal structure. Struc-
Solid-state crystalline forms of an insoluble anti-
ture solution from powder diffraction data is still not
hypertensive drug furosemide were prepared using
a routine method for large and flexible molecules.
liquid-assisted grinding and slurry methods. Single
Furosemide–caffeine is a cocrystal, and furosemide–
crystal x-ray structure validation was performed on
cytosine is a salt as revealed by x-ray crystal struc-
two of eight materials. All the products were charac-
ture analysis. A definitive answer about the neu-
terized by DSC, ss-NMR, PXRD, IR, NIR, and Raman
tral/ionic state of other six furosemide compounds
measurements. Even as crystal engineering princi-
is pending upto the availability of XPS/NMR mea-
ples offer guidelines for the rational selection of co-
surements or good quality single crystals. A practi-
formers for a particular API, the exact cocrystal struc-
cal advantage that cocrystals offer over metastable

Table 4. Solubility and IDR in 10% Ethanol–Water

Solubility at 24 h in Solubility in Water Intrinsic Dissolution Rate in Stability in 10% EtOH–Water


Compound 10% EtOH–Water (mg/L) (mg/mL) 10% EtOH–Water (mg/cm2 )/min Slurry Medium
FUROS 118 0.006 44 × 10−3 Stable after 24, 48 h
FUROS–CAF 720 (×6.1) 22 87 × 10−3 (×1.9) Stable after 24, 48 h
FUROS–UREA 632 (×5.3) 1000 83 × 10−3 (×1.9) Converted to furosemide within 24 h
FUROS–PABA 370 (×3.1) 5 53 × 10−3 (×1.2) Converted to furosemide within 24 h
FUROS–ACT 812 (×6.9) 2000 94 × 10−3 (×2.1) Converted to furosemide within 24 h
FUROS–NIC 1040 (×8.8) 1000 111 × 10−3 (×2.5) Converted to furosemide within 24 h
FUROS–INIC 856 (×7.2) 192 102 × 10−3 (×2.3) Converted to furosemide within 24 h
FUROS–ADEN 787 (×6.7) 9 91 × 10−3 (×2.1) Stable after 24, 48 h
FUROS–CYT 1260 (×10.7) 8 116 × 10−3 (×2.6) Stable after 24, 48 h

Numbers in parentheses give the number of times solubility is higher compared to the pure drug.
Solubility of the coformer is given in water.
The stable cocrystal entries are highlighted in bold.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012 DOI 10.1002/jps
NOVEL FUROSEMIDE COCRYSTALS AND SELECTION OF HIGH SOLUBILITY DRUG FORMS 675

Figure 8. 13 C ss-NMR of FUROS–CYT and the individual components show that a novel
solid-state complex was formed upon grinding.

polymorphs or amorphous drug forms is that they are Preparation of Furosemide Cocrystals
relatively more stable owing to their crystalline na-
Furosemide (34 mg) and caffeine (20 mg) (1:1 molar
ture. Thus cocrystals can combine the twin aspects
ratio) were ground and mixed in a mortar–pestle for
of improved solubility and good stability for opti-
20 min after adding 5–6 drops of acetonitrile, a liquid-
mal drug delivery. Furosemide–caffeine, furosemide–
assisted grinding procedure. Cocrystal formation was
adenine, and furosemide–cytosine exhibited com-
confirmed by changes in the signature peaks of FT-IR
parable stability to the commercial drug in 10%
and PXRD. Forty milligram of the ground material
ethanol–water slurry medium up to 48 h. More-
was dissolved in 5 mL of MeOH–CH3 CN solvent mix-
over, their solubility is 6–11-fold higher relative to
ture and left aside for evaporation at ambient con-
furosemide. Thus cocrystals could offer a superior
ditions. Single crystals suitable for x-ray diffraction
strategy to improve the solubility of BCS class II/IV
appeared after 4–5 days.
drugs compared to metastable polymorphs and amor-
Furosemide (68 mg) and urea (12 mg) (1:1 molar
phous dispersions.
ratio) were ground and mixed together in a mortar–
pestle for 20 min after adding 5–6 drops of acetone, a
liquid-assisted grinding procedure. Cocrystal forma-
EXPERIMENTAL
tion was confirmed by changes in the signature peaks
Furosemide (purity >99.8%) was supplied by As- of FT-IR and PXRD.
traZeneca India Pvt. Ltd (Bangalore, India). Coform- Furosemide (34 mg) and p-aminobenzoic acid
ers (purity >99.8%) were purchased from Sigma- (14 mg) (1:1 molar ratio) were ground and mixed
Aldrich (Hyderabad, India). Solvents (purity >99%) in a mortar–pestle for 20 min after adding 5–
were purchased from Hychem Laboratories (Hyder- 6 drops of acetone, a liquid-assisted grinding proce-
abad, India). Water filtered through a double deion- dure. Cocrystal formation was confirmed by changes
ized purification system (AquaDM, Bhanu, Hyder- in the signature peaks of FT-IR and PXRD. The
abad, India) was used in all experiments. same cocrystal was also obtained by using the slurry

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012
676 GOUD ET AL.

Figure 9. (a) Experimental PXRD plots of furosemide, cocrystal, and coformer to compare 2θ
values in the new crystalline phases. (b) Calculated PXRD lines form the x-ray crystal struc-
ture for furosemide, coformer polymorphs, and cocrystals for fingerprint matching of starting
materials in the product phases of (a).

crystallization method. PABA (14 mg) was dissolved Furosemide (34 mg) and nicotinamide (13 mg) (1:1
in 1.5-mL MeOH and 3.5 mL of H2 O, and furosemide molar ratio) were ground and mixed in a mortar–pes-
(34 mg) was added with stirring. The formation of tle for 20 min after adding 5–6 drops of acetone, a
cocrystal was completed after 30 min. liquid-assisted grinding procedure. Cocrystal forma-
Furosemide (68 mg) and acetamide (12 mg) (1:1 mo- tion was confirmed by changes in the signature peaks
lar ratio) were ground and mixed in a mortar–pestle of FT-IR and PXRD.
for 20 min after adding 5–6 drops of acetonitrile, a Furosemide (34 mg) and isonicotinamide (13 mg)
liquid-assisted grinding procedure. Cocrystal forma- (1:1 molar ratio) were ground and mixed in a mor-
tion was confirmed by changes in the signature peaks tar–pestle for 20 min after adding 5–6 drops of ace-
of FT-IR and PXRD. tone, a liquid-assisted grinding procedure. Cocrystal

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012 DOI 10.1002/jps
NOVEL FUROSEMIDE COCRYSTALS AND SELECTION OF HIGH SOLUBILITY DRUG FORMS 677

Figure 11. Dissolution curve of furosemide and its cocrys-


tals for up to 6 h.

formation was confirmed by changes in the signature


peaks of FT-IR and PXRD.
Furosemide (34 mg) and adenine (14 mg) (1:1 molar
ratio) were ground and mixed in a mortar–pestle for
20 min after adding 5–6 drops of acetone, a liquid-
assisted grinding procedure. Cocrystal formation was
confirmed by changes in the signature peaks of FT-IR
and PXRD. The same cocrystal was also obtained by
the slurry crystallization method. Furosemide (110
mg) and adenine (40 mg) was added to 8-mL MeOH
with stirring. The formation of cocrystal was complete
after 7–8 h.
Furosemide (34 mg) and cytosine (12 mg) (1:1 mo-
lar ratio) were ground and mixed in a mortar–pestle
for 20 min after adding 5–6 drops of acetonitrile, a
liquid-assisted grinding procedure. Cocrystal forma-
tion was confirmed by changes in the signature peaks
of FT-IR and PXRD. Fifty milligram of the ground ma-
terial was dissolved in 6 mL of MeOH and left aside
for evaporation at ambient conditions. Single crystals
suitable for x-ray diffraction appeared after 5–6 days.

Vibrational Spectroscopy
A Thermo-Nicolet 6700 Fourier transform infrared
spectrophotometer with an NXR-Fourier transform
Raman module (Thermo Scientific, Waltham, MA)
was used to record IR, Raman, and NIR spectra. IR
and NIR spectra were recorded on samples dispersed
in KBr pellets. Raman spectra were recorded on sam-
ples contained in standard NMR diameter tubes or on
compressed samples contained in a gold-coated sam-
13
Figure 10. C ss-NMR spectra of cocrystals, drug, and ple holder. Data were analyzed using the Omnic soft-
coformers. ware (Thermo Scientific, Waltham, MA).

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012
678 GOUD ET AL.

Solid-State NMR Spectroscopy tally located in difference electron density maps. All
C H atoms were fixed geometrically using HFIX com-
Solid-state 13 C NMR spectra were obtained on a
mand in SHELX-TL (Bruker-AXS). A check of the fi-
Bruker Avance 400 MHz spectrometer (Bruker-
nal CIF file using PLATON66 did not show any missed
Biospin, Karlsruhe, Germany). Ss-NMR measure-
symmetry. Hydrogen bond distances shown in Table 2
ments were carried out on Bruker 4-mm double reso-
are neutron normalized to fix the D–H distance to its
nance CP-MAS probe in zirconia rotors with a Kel-F
accurate neutron value in the x-ray crystal structures
cap at 5.0-kHz spinning rate with a cross-polarization
(O–H 0.983 Å, N–H 1.009 Å, and C–H 1.083 Å). X-
contact time of 2.5 ms and a delay of 8 s. 13 C NMR
Seed67 was used to prepare packing diagrams. Crys-
spectra were recorded at 100 MHz and referenced to
tallographic.cif files (CCDC Nos. 833554–833555) are
the methylene carbon of glycine (δglycine = 43.3 ppm).
available at www.ccdc.cam.ac.uk/data or as part of
Thermal Analysis Supporting Information.

Differential scanning calorimetry was performed on a Dissolution and Solubility Measurements


Mettler-Toledo DSC 822e module, and TGA was per- The solubility of furosemide and its cocrystals were
formed on a Mettler Toledo TGA/SDTA 851e mod- determined according to the Higuchi and Connor
ule (Mettler-Toledo, Columbus, OH). Samples were method68 in 10% ethanol–water medium at 30◦ C.
placed in sealed pin-pricked alumina pans for TG ex- First, the absorbance of a known concentration of
periments and in crimped but vented aluminum pans the cocrystal/drug was measured at the respective
for DSC experiments. The typical sample size is 3–5 λmax (furosemide 334 nm, furosemide–caffeine 330
mg for DSC and 8–12 mg for TGA. The temperature nm, furosemide–urea 331 nm, furosemide–PABA 331
range for the thermogram was 30–300◦ C, and the nm, furosemide–acetamide 330 nm, furosemide–ni-
sample was heated at a rate of 5◦ C/ min. Samples cotinamide 330 nm, furosemide–isonicotinamide 329
were purged in a stream of dry nitrogen flowing at 80 nm, furosemide–adenine 330 nm) in 10% ethanol–wa-
mL/min for DSC and 50 mL/min for TGA. The TGA of ter medium on a Thermo Scientific Evolution 300
FUROS–UREA and FUROS–ACT is shown as repre- UV–vis spectrometer (Thermo Scientific, Waltham,
sentative cases of cocrystals (Fig. S18) to monitor the MA). These absorbance values were plotted against
decomposition of furosemide in the postmelting stage. several known concentrations to prepare the concen-
Powder X-Ray Diffraction tration versus intensity calibration curve. From the
slope of the calibration curves, molar extinction co-
Powder x-ray diffraction of all the samples was efficients (Table S6) for the cocrystal/drug were cal-
recorded on a Bruker D8 advance diffractometer culated. An excess amount of the sample was added
(Bruker-AXS, Karlsruhe, Germany) using Cu K" x- to 6 mL of 10% ethanol–water medium. The super-
radiation (8 = 1.5406 Å) at 40 kV and 30 mA power. saturated solutions were stirred at 300 rpm using a
X-ray diffraction patterns were collected over the 22 magnetic stirrer at 30◦ C. After 24 h, the suspension
range 5–50◦ at a scan rate of 1◦ /min. Powder Cell 2.461 was filtered through Whatman’s filter paper No. 1.
(Federal Institute of Materials Research and Testing, The filtered aliquots were diluted sufficiently, and the
Berlin, Germany) was used for Rietveld refinement absorbance was measured at the respective λmax . Fi-
of experimental PXRD and calculated lines from the nally, the concentrations of furosemide and its cocrys-
x-ray crystal structure. The calculated crystal struc- tals were calculated at regular time intervals of 24
tures of coformer polymorphs not determined in this and 48 h using the relevant calibration curve. IDR
study were taken from the literature50 or extracted experiments were carried out on a USP-certified Elec-
from the CCDC (Cambridge Crystallographic Data trolab TDT-08L dissolution tester (Mumbai, India).
Centre).62 Dissolution experiments were performed for 6 h in
10% ethanol–water medium at 37◦ C. Prior to IDR
X-Ray Crystallography
estimation, standard curves for all the compounds
X-ray reflections were collected on a Bruker Smart- were obtained spectrophotometrically at their respec-
Apex CCD diffractometer (Bruker-AXS, Karlsruhe, tive λmax . The respective molar extinction coefficients
Germany). Mo K" x-radiation (λ = 0.71073 Å) was were used to determine the IDR values. For IDR mea-
used to collect x-ray reflections on the single crys- surements, 100 mg of the compound was taken in
tal. Data reduction was performed using the Bruker the intrinsic attachment and compressed to a 0.5-cm2
SAINT software.63 Intensities for absorption were cor- disk using a hydraulic press at pressure of 4.0 ton/
rected using SADABS,64 the Siemens area detector in.2 for 5 min. The intrinsic attachment was placed
absorption correction program (Bruker-AXS). Crystal in a jar of 500-mL medium preheated to 37◦ C and
structures were solved and refined using SHELX-9765 rotated at 150 rpm. Five milliliter of aliquot was col-
with anisotropic displacement parameters for non-H lected at specific time intervals, and the concentra-
atoms. Hydrogen atoms on O and N were experimen- tion of the aliquots was determined with appropriate

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012 DOI 10.1002/jps
NOVEL FUROSEMIDE COCRYSTALS AND SELECTION OF HIGH SOLUBILITY DRUG FORMS 679

dilutions from the predetermined standard curves of 12. Karami S, Li Y, Hughes DS, Hursthouse MB, Russell AE,
the respective compounds. The IDR of the compound Threlfall TL, Claybourn M, Roberts R. 2006. Further errors
in polymorph identification: Furosemide and finasteride. Acta
was calculated in the linear region of the dissolu-
Crystallogr B 62:689–691.
tion curve (which is the slope of the curve or amount 13. Babu NJ, Cherukuvada S, Thakuria R, Nangia A. 2010. Con-
of drug dissolved/surface area of the disk) per unit formational and synthon polymorphism in furosemide (Lasix).
time. The identity of the undissolved material after Cryst Growth Des 10:1979–1989.
the dissolution experiment was ascertained by PXRD. 14. Ai H, Jones SA, deVilliers MM, Lvov YM. 2003. Nano encapsu-
lation of furosemide microcrystals for controlled drug release.
The stability of the solid samples after disk compres-
J Control Release 86:59–68.
sion and solubility measurements was confirmed by 15. Matsuda Y, Otsuka M, Onoe M, Tatsumi E. 1992. Amor-
PXRD. phism and Physicochemical stability of spray-dried frusemide.
J Pharm Pharmacol 44:627–633.
16. Shin SC, Oh IJ, Lee YB, Choi HK, Choi JS. 1998. Enhanced
ACKNOWLEDGMENTS dissolution of furosemide by coprecipitating or cogrinding with
crospovidone. Int J Pharm 175:17–24.
AN thanks the Department of Science and Technol- 17. Shin SC, Kim J. 2003. Physicochemical characterization of
ogy, Government of India (DST) for research fund- solid dispersion of furosemide with TPGS. Int J Pharm
251:79–84.
ing (SR/S1/OC-67/2006) and J.C. Bose fellowship (SR/
18. Özdemir N, Ordu S. 1998. Improvement of dissolution proper-
S2/JCB-06/2009) and Council of Scientific and In- ties of furosemide by complexation with $-cyclodextrin. Drug
dustrial Research (CSIR) project (01/2410)/10/EMR- Dev Ind Pharm 24:19–25.
II). DST (IRPHA) and University Grants Commis- 19. Kreaz RMA, Abu-Eida EY, Erős I, Kata M. 1999. Freeze dried
sion (UGC) (PURSE grant) are thanked for providing complexes of furosemide with $-cyclodextrin derivatives. J Incl
Phenom Macrocycl Chem 34:39–48.
instrumentation and infrastructure facilities at Uni-
20. Wongmekiat A, Yoshimatsu S, Tozuka Y, Moribe K,
versity of Hyderabad (UOH). NRG thanks CSIR, and Yamamoto K. 2006. Investigation of drug nanoparticle for-
KS thanks UGC for a fellowship. Crystalin Research mation by cogrinding with cyclodextrins: studies for In-
Pvt. Ltd. (Hyderabad, India) thanks AstraZeneca domethacin, Furosemide and Naproxen. J Incl Phenom Macro-
Pvt. Ltd. (Bangalore, India) for funding a research cycl Chem 56:29–32.
21. Yang W, de Villiers MM. 2004. Aqueous solubilization of
project. We thank Dr. Mike Quayle and Dr. David
furosemide by supramolecular complexation with 4-sulphonic
Berry (AstraZeneca PLC) for their critical comments calix[n]arenes. J Pharm Pharmacol 56:703–708.
and helpful suggestions. This publication is assigned 22. Zvonar A, Berginc K, Kristl A, Gašperlin M. 2010. Microen-
AstraZeneca ATP No. 11/1199. capsulation of self-microemulsifying system: Improving solu-
bility and permeability of furosemide. Int J Pharm 388:151–
158.
23. Latere Dwan’lsa JP, Rouxhet L, Brewster ME, Préat
REFERENCES V, Ariën A. 2008. Spontaneously self-assembled mi-
1. Qiu Y, Chen Y, Zhang GGZ., Eds. 2009. Developing solid oral celles from poly(ethylene glycol)-b-poly(g-caprolactone-
dosage forms: Pharmaceutical theory and practice, 1st ed. co-trimethylene carbonate) for drug solubilization. Pharmazie
New York: Academic Press. 63:235–240.
2. Thayer AM. 2010. Finding solutions. Chem Eng News 24. Shaji J, Jadhav D. 2010. Newer approaches to self emulsifying
88:13–18. drug delivery system. Int J Pharm Pharm Sci 2:37–42.
3. Amidon GL, Lennernas H, Shah VP, Crison JR. 1995. Theo- 25. Otsuka M, Onoe M, Matsuda M. 1993. Hygroscopic stability
retical basis for a biopharmaceutical drug classification: corre- and dissolution properties of spray-dried solid dispersions of
lation of in vitro drug product dissolution and in vivo bioavail- furosemide with eudragit. J Pharm Sci 82:32–38
ability. Pharm Res 12:413–420. 26. Brittain HG. 2010. Cocrystal systems of pharmaceutical inter-
4. Dahan A, Miller JM, Amidon GL. 2009. Prediction of solubility est: 2007-2008. Profiles Drug Substances, Excipients Related
and permeability class membership: Provisional BCS classifi- Methodology 35:373–390.
cation of world’s top oral drugs. AAPS J 11:740–746. 27. Vishweshwar P, McMahon JA, Bis JA, Zaworotko MJ. 2006.
5. Friedman PA, Berndt WO. 1997. In Modern pharmacology Pharmaceutical Co-Crystals. J Pharm Sci 95:499–516.
with clinical applications; Craig CR, Stitzel RE, Eds. 5th ed. 28. Friščić T, Jones W. 2010. Benefits of cocrystallisation in phar-
Boston, MA: Little Brown and Company, pp 239–255. maceutical materials science: an update. J Pharm Pharmacol
6. Khan MG. 2006. Encyclopedia of heart diseases. 15th ed.New 62:1547–1559.
York: Elsevier, pp 486. 29. Desiraju GR. 1995. Supramolecular synthons in crystal engi-
7. Blagden N, de Matas M, Gavan PT, York P. 2007. Crystal neering–A new organic synthesis. Angew Chem, Int Ed Engl
engineering of active pharmaceutical ingredients to improve 34:2311–2327.
solubility and dissolution rates. Adv Drug Del Rev 59:617–630. 30. Desiraju GR. 2007. Crystal Engineering: A holistic view.
8. Shan N, Zaworotko MJ. 2008. The role of cocrystals in phar- Angew Chem, Int Ed 46: 8342–8356.
maceutical science. Drug Discov Today 13:440–446. 31. Bhogala BR, Nangia A. 2003. Cocrystals of 1,3,5-
9. Doherty C, York P. 1988. Furosemide crystal forms: Solid state cyclohexanetricarboxylic acid with 4,4’ bipyridine homologues:
and physicochemical analyses. Int J Pharm 47:141–155. Acid–pyridine hydrogen bonding in neutral and ionic com-
10. Matsuda Y, Tatsumi E. 1990. Physicochemical characteriza- plexes. Cryst Growth Des 3:547–554.
tion of furosemide modifications. Int J Pharm 60:11–26. 32. Caira MR. 2007. Sulfa drugs as model cocrystal formers. Mol
11. Ge M, Liu G, Ma S, Wang W. 2009. Polymorphic forms of Pharmaceutics 4:310–316 .
furosemide characterized by THz time domain spectroscopy. 33. Ishida T, In Y, Doi M, Inoue M, Yanagisawa I. 1989.
Bull Korean Chem Soc 30:2265–2268. Structural study of Histamine H2 –receptor antagonists. Five

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012
680 GOUD ET AL.

3-[2-(diaminomethyleneamino)-4-thiazolylmethylthio] propi- namide cocrystals with alkanediacids HO2 C(CH2 )n–2 CO2 H,
onamidine and amide derivatives. Acta Crystallogr B n = 7–9. CrystEngComm 13:4188–4195.
45:505–512. 52. Vishweshwar P, Nangia A, Lynch VM. 2003. Molecular com-
34. Goud NR, Babu NJ, Nangia A. 2011. Sulfonamide–pyridine- plexes of homologous alkanedicarboxylic acids with isonicoti-
N-oxide cocrystals. Cryst Growth Des 11:1930–1939. namide: X-ray crystal structures, hydrogen bond synthons and
35. Pellegata RL. 1990. Furosemide salts. Patent US 5182300. melting point alternation. Cryst Growth Des 3:783–790.
36. Carda-Broch S, Esteve-Romero J, Garcia-Alvarez-Coque MC. 53. Cherukuvada S, Babu NJ, Nangia A. Nitrofurantoin-p-
2000. Furosemide assay in pharmaceuticals by micellar liquid aminobenzoic acid cocrystal: Hydration stability and dissolu-
chromatography: Study of the stability of the drug. J Pharm tion rate studies. J Pharm Sci 100:3233–3244.
Biomed Anal 23:803–817. 54. Childs SL, Chyall LJ, Dunlap JT, Smolenskaya VN, Stahly
37. Trask AV. 2007. An overview of Pharmaceutical cocrystals as BC, Stahly GP. 2004. Crystal engineering approach to forming
Intellectual property. Mol Pharmaceutics 4:301–309 . cocrystals of amine hydrochlorides with organic acids. Molec-
38. Babu NJ, Nangia A. 2011. Solubility Advantage of Amorphous ular complexes of fluoxetine hydrochloride with benzoic, suc-
Drugs and Pharmaceutical Cocrystals. Cryst Growth Des 11: cinic and fumaric acids. J Am Chem Soc 126:13335–13342.
2662–2679. 55. Fahr A, Liu X. 2007. Drug-delivery strategies for poorly water
39. GRAS chemical list. Accessed at www.fda.gov/Food/ soluble drugs. Expert Opin Drug Deliv 4:403–416.
FoodIngredientsPackaging/GenerallyRecognizedasSafeGRAS/ 56. Cheney ML, Shan N, Healey ER, Hanna M, Wojtas L,
GRASSubstancesSCOGSDatabase/default.htm . Zaworotko MJ, Sasa V, Song S, Sanchez-Ramos JR. 2010.
40. Childs SL, Stahly GP, Park A. 2007. The salt-cocrystal con- Effects of crystal form on solubility and pharmacokinetics:
tinuum: the influence of crystal structure on ionization state. a crystal engineering case study of lamotrigine. Cryst Growth
Mol Pharmaceutics 4:323–328 . Des 10:394–405.
41. Vogt FG, Clawson JS, Strohmeier M, Edwards AJ, Pham TN, 57. Good D, Rodrı́guez-Hornedo N. 2009. Solubility advantage of
Watson SA. 2009. Solid state NMR analysis of organic cocrys- pharmaceutical cocrystals. Cryst Growth Des 9:2252–2264.
tals and complexes. Cryst Growth Des 9:921–937. 58. Bethune SJ, Huang N, Jayasankar A, Rodrı́guez-Hornedo N.
42. Stevens JS, Byard SJ, Schroeder SLM. 2010. Salt or co-crystal? 2009. Understanding and predicting the effect of cocrystal
Determination of protonation state by x-ray photoelectron components and pH on cocrystal solubility. Cryst Growth Des
spectroscopy (XPS). J Pharm Sci 99:4453–4457. 9:3976–3988.
43. Shan N, Toda F, Jones W. 2002. Mechanochemistry and co- 59. Jayasankar A, Reddy LS, Bethune SJ, Rodrı́guez-Hornedo N.
crystal formation: Effect of solvent on reaction kinetics. Chem 2009. Role of cocrystal and solution chemistry on the formation
Commun 20:2372–2373. and stability of cocrystals with different stoichiometry. Cryst
44. Beyers H, Malan SF, van der Watt JG, de Villiers MM. 2000. Growth Des 9:889–897.
Structure-solubility relationship and thermal decomposition 60. Guo K, Sadiq G, Seaton C, Davey R, Yin Q. 2010. Co-
of furosemide. Drug Dev Ind Pharm 26:1077–1083. crystallization in the caffeine/maleic acid system: Lessons
45. Li ZJ, Abramov Y, Bordner J, Leonard J, Medek A, Trask AV. from phase equilibria. Cryst Growth Des 10:268–273.
2006. Solid-state acid-base interactions in complexes of hete- 61. Powder Cell, a program for structure visualization, powder
rocyclic bases with dicarboxylic acids: Crystallography, hydro- pattern calculation and profile fitting. Accessed at www.ccp14.
gen bond analysis and 15 N NMR spectroscopy. J Am Chem Soc ac.uk/tutorial/powdercell.
128:8199–8210. 62. Cambridge Crystallographic Data Center, Cambridge Struc-
46. Bernstein J, Davis RE, Shimoni L, Chang NL. 1995. Patterns tural Database, version 5.32, November 2010, August 2011
in hydrogen bonding: Functionality and graph set analysis in update Available at www.ccdc.cam.ac.uk/products/csd.
crystals. Angew Chem, Int Ed Engl 34:1555–1573. 63. SAINT-Plus, version 6.45. 2003. Madison, WI: Bruker AXS
47. Chieng N, Rades T, Aaltonen J. 2011. An overview of re- Inc.
cent studies on the analysis of pharmaceutical polymorphs. 64. Sheldrick GM. 1997. SADABS, program for empirical absorp-
J Pharm Biomed Anal 55:618–644. tion correction of area detector data. Göttingen, Germany:
48. Trask AV, Jones W. 2005. Crystal Engineering of organic University of Göttingen.
cocrystals by the solid-state grinding approach. Top Curr 65. Sheldrick GM. 1997. SHELXS-97 and SHELXL-97, Göttingen,
Chem 254:41–70. Germany: University of Göttingen.
49. Schultheiss N, Newman A. 2009. Pharmaceutical cocrys- 66. Spek AL. 2002. PLATON, a multipurpose crystallographic tool.
tals and their physicochemical properties. Cryst Growth Des Utrecht, The Netherlands: Utrecht University.
9:2950–2967. 67. Barbour LJ. 1999. X-Seed, graphical interface to SHELX-
50. Li J, Bourne SA, Caira MR. 2011. New polymorphs of isoni- 97 and POV-Ray. Columbia, MO: University of Missouri–
cotinamide and nicotinamide. Chem Commun 47:1530–1532. Columbia.
51. Thompson LJ, Voguri RS, Male L, Tremayne M. 2011. The 68. Higuchi T, Connors KA. 1965. Phase-solubility techniques.
crystal structures and melting point properties of isonicoti- Adv Anal Chem Instrum 4:117–212.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 2, FEBRUARY 2012 DOI 10.1002/jps

S-ar putea să vă placă și