Int J App Pharm, Vol 16, Issue 2, 2024, 132-138Original Article

CHARACTERISATION, EVALUATION AND DENSITY FUNCTIONAL ANALYSIS OF CILNIDIPINE-NICOTINAMIDE COCRYSTALS DEVELOPED BY LIQUID ASSISTED GRINDING TECHNIQUE: A SUSTAINABLE APPROACH FOR ENHANCED SOLUBILITY

RENJISH CHERUKKOTH1*, SIBI P. ITTIYAVIRAH2, JYOTI HARINDRAN3, SUDHAKARAN NAIR C. R.4

1*College of Pharmaceutical Sciences, Govt. T D Medical College, Alappuzha, Kerala, India. 2Department of Pharmaceutical Sciences, Centre for Professional and Advanced Studies, RIMSR, Kottayam, Kerala, India. 3Department of Pharmaceutical Sciences, Cheruvandoor Campus, Centre for Professional and Advanced Studies, Kottayam, Kerala, India. 4College of Pharmaceutical Sciences, Govt Medical College, Thiruvananthapuram, Kerala, India
*Corresponding author: Renjish Cherukkoth; *Email: renjishc@yahoo.co.in

Received: 13 Nov 2023, Revised and Accepted: 19 Dec 2023


ABSTRACT

Objective: Improving the solubility of poorly water-soluble drugs has always been a challenge in drug development. This study aimed to enhance the aqueous solubility of a poorly water-soluble drug, Cilnidipine, by cocrystallisation method using liquid-assisted grinding (LAG) technique with Nicotinamide as the coformer. The study also aimed to understand the mechanism of cocrystal formation by quantum mechanical calculations.

Methods: The Cilnidipine-Nicotinamide cocrystals were prepared in various stoichiometric ratios using the liquid-assisted grinding (LAG) technique. The cocrystals obtained were characterised by vibrational spectroscopy, thermal methods such as differential scanning calorimetry (DSC), powder X-ray diffraction (PXRD) and surface morphology by field emission scanning electron microscopy (FE-SEM). The cocrystals were evaluated for saturation solubility, and the mechanistic study of cocrystal formation was performed using the Gaussian 09 software package.

Results: FT-IR spectra of the formulated cocrystal indicated the intermolecular hydrogen bond formation between-N-H of Nicotinamide and the nitro group of Cilnidipine. DSC analysis showed a single endotherm at 96.76 °C, PXRD patterns were different from that of the reactants, and FE-SEM analysis revealed the changes in the surface morphology of the obtained cocrystal. The prepared cocrystal showed a 26.36-fold enhancement in the aqueous solubility of Cilnidipine. The DFT study demonstrated the formation of a strong intermolecular hydrogen bonding between the nitro-oxygen atom of Cilnidipine and the amide hydrogen atom of Nicotinamide in cocrystal formed.

Conclusion: This study highlights the potential of the liquid-assisted grinding method for preparing cocrystals as a sustainable and reliable approach to address the challenges posed by poorly water-soluble drugs.

Keywords: Cocrystals, Cocrystals Cilnidipine, Cilnidipine Nicotinamide, Liquid-assisted grinding, Solubility enhancement, Green, Sustainable


INTRODUCTION

Several factors are involved in determining the absorption of a drug from an oral solid dosage form. These include the release of the drug from the dosage form, its ability to dissolve in gastrointestinal (GI) fluids, and its permeability through the gastrointestinal tract (GIT). Over the past few years, many newly discovered drugs have been classified as BCS class II (highly lipophilic and poor aqueous solubility). For these drugs, the ability to dissolve in the GI fluids is critical in determining their bioavailability [1]. There are various methods that formulation scientists can utilise to address this issue. In recent years, cocrystallisation has emerged as a promising approach for enhancing the aqueous solubility of such active pharmaceutical ingredients (API) without altering their pharmacological activities [2]. Cocrystals offer several potential benefits, such as improved aqueous solubility and consequent increase in bioavailability, better mechanical properties, and stability [3]. Pharmaceutical cocrystals are crystalline materials of two or more different molecules in a specific stoichiometric ratio. One of the components is an API, while the other is a coformer held together by nonionic and non-covalent bonds [4].

Poorly controlled hypertension increases the risk of cardiovascular, renal, and endocrine disorders. To manage hypertension, dihydropyridine class of calcium channel blockers (CCBs) such as Cilnidipine are commonly used as first-line agents, alone or in combination with other antihypertensive drugs [5]. Cilnidipine blocks both L-type and N-type calcium channels and is the recommended medication for treating hypertension in patients with chronic renal diseases and diabetes mellitus [6–8]. Cilnidipine is a BCS class II drug, insoluble in water and with a reported bioavailability of less than 13% [9]. The pKa value of Cilnidipine is 11.39. The N-H of the dihydropyridine ring can serve as a hydrogen bond donor, and eight electronegative atoms, such as the oxygen atom of the NO2 group and the ester functional group, serve as hydrogen bond acceptors [10]. Because of these properties, Cilnidipine is an ideal candidate for developing cocrystals with compatible coformers having complementary hydrogen bonding sites [11].

This study aimed to enhance the aqueous solubility of Cilnidipine by utilising the cocrystallisation technique with Nicotinamide (pyridine-3-carboxamide) as the coformer. Nicotinamide is a valuable coformer in the pharmaceutical industry as it has multiple hydrogen bond acceptor and donor sites [12]. ΔpKa value for the Cilnidipine-Nicotinamide system is less than one, favouring the formation of cocrystals [13, 14]. Moreover, Nicotinamide is a water-soluble vitamin (vitamin B3) used in foods and nutrient supplements and is included in the GRAS list (generally regarded as safe) [15-17]. This study adopted the liquid-assisted grinding (LAG) technique to prepare Cilnidipine-Nicotinamide cocrystals. The LAG method involves grinding solid reactants while incorporating a minimal amount of solvent (micro-liter), making it a swift and dependable method for discovering cocrystals [18]. This process is considered eco-friendly since it uses minimal organic solvents, making it a sustainable option for cocrystal preparation [19]. Methanol was used as the solvent in this study, as it can modify the wettability of the solid surface of Cilnidipine and facilitate cocrystal formation [20]. The resulting cocrystals were characterised using Fourier-transform infrared spectroscopy (FT-IR), differential scanning calorimetry (DSC), powder X-ray diffraction (PXRD) and field emission scanning electron microscopy (FE-SEM). A computational technique based on Density Functional Theory (DFT) using the Gaussian approach was used to study the mechanism of intermolecular and intramolecular interactions within the Cilnidipine-Nicotinamide cocrystal system [21]. Ultimately, the prepared cocrystals were assessed for saturation solubility to ascertain the improvement in aqueous solubility of Cilnidipine achieved through the prepared cocrystals.

MATERIALS AND METHODS

The materials used were Cilnidipine (PubChem: 5282138), Nicotinamide (PubChem: 936) and Methanol (PubChem: 887). Cilnidipine was obtained from Pure Chem Pvt. Ltd., Gujarat, while Nicotinamide was purchased from Merck and Methanol (HPLC grade) was sourced from SD Fine-Chem Limited in Mumbai. The ultra-pure grade water collected from the Milli-Q® water purification system (Millipore, Germany) was used for the study. All other chemicals used in the study were analytical grade (AR).

Preparation of cocrystals of cilnidipine with nicotinamide as coformer by LAG

The LAG method was adopted to prepare cocrystals of Cilnidipine with Nicotinamide as the coformer. Cilnidipine and Nicotinamide in the stoichiometric ratio, as shown in table 1, were taken in an agate mortar and pestle and manually ground at room temperature with the addition of 100 µl (2 drops) of methanol at 10 min intervals for 45 min [22]. The obtained products were dried at room temperature in a desiccator and used as such for further characterisation and evaluation [18]. The physical mixtures (CNPM) were prepared by physically mixing the components in the same molar ratio used to prepare the respective formulation.

Table 1: Ratio of cilnidipine and nicotinamide in the cocrystals

Ingredients CN1 (1:0.5) CN2 (1:1) CN3 (1:2)
Cilnidipine (g) 0.4952 0.4952 0.4952
Nicotinamide (g) 0.0611 0.1221 0.2442

Fourier-transform infrared spectroscopy analysis

The FT-IR analysis of Cilnidipine, Nicotinamide, their physical mixtures and prepared products was carried out by KBr pelleting technique [23], using Nicolet iS10 FT-IR Spectrometer, Themofisher, USA. The spectra were recorded from 4000 to 400 cm-1.

Differential scanning calorimetric analysis

The DSC analyses were conducted in DSC Q20V24.11, TA Instruments, USA. About 5 mg of the sample was accurately weighed and transferred to an aluminium pan. The pan was crimped using a crimping machine and placed in the sample chamber, and an empty aluminium pan was kept in the blank chamber. Dry nitrogen gas was continuously purged into the chamber, and the spectra were recorded from 30 °C to 300 °C at a heating rate of 10 °C/min.

Powder X-ray diffraction (PXRD) measurements

The powder X-ray diffractometer, Ultima IV (Rigaku, Japan), with Cu-Kα-X radiation as source (λ = 1.5405 Å) at 40 kV/30 mA, was used to record powder X-ray diffractograms of samples. 2θ angle was measured from 5°–80° at a step size of 0.02°/0.5 s using Nickel as the filter. The diffraction pattern was obtained by plotting 2θ against the intensity.

Saturation solubility analysis of cilnidipine-nicotinamide cocrystals

Accurately weighed and transferred 100 mg each of pure Cilnidipine and CN2 to vials containing 10 ml of ultrapure water. The vials were shaken continuously at 100±10 agitations/min in a shaker water bath at room temperature for 24 h. It was then centrifuged, the supernatant was separated and filtered using Whatman filter paper No: 10, and the drug content was estimated using an ultraviolet (UV) spectrophotometer.

Field emission scanning electron microscopy analysis

FE-SEM analyses of the samples were carried out in Gemini 300 (Carl Zeiss, Germany). The gold-sputtered samples were analysed under an inert argon atmosphere at an accelerating voltage of 20 kV.

Computational method

Density functional theory calculations of the Cilnidipine–Nicotinamide (1:1) cocrystal system were performed using the Gaussian 09W program to study the possible mechanism of cocrystal formation [24]. The structures of Cilnidipine and Nicotinamide, downloaded from the PubChem database, were converted from the standard data format (SDF) to Gaussian Job File (GJF) using the Open Babel application [25, 26]. The optimised structures of the cocrystal and its components were obtained by DFT calculations using the B3LYP/6-311++G (d,p) level of theory [27].

RESULTS AND DISCUSSION

Preparation of cocrystals of cilnidipine with nicotinamide as coformer by LAG

The LAG technique using trace amounts of methanol as solvent was used to prepare cocrystals of Cilnidipine with Nicotinamide. Grinding the components using micro-liters of methanol facilitated the formation of cocrystals with Nicotinamide [19].

Fourier-transform infrared spectroscopic analysis

FT-IR spectroscopy is a valuable technique used to characterise the cocrystals. It provides helpful information about the intermolecular and intramolecular interactions, especially hydrogen bond formation in a cocrystal [20]. The FT-IR spectra of Cilnidipine and Nicotinamide are given in fig. 1.

Fig. 1: FT-IR spectra of (A) Cilnidipine (Cil) and (B) Nicotinamide (NA)

The Cilnidipine showed the characteristic peaks at 3281 cm-1 (N-H stretching), 1693 cm-1 (-C=O stretching) and 1525 cm-1 (N-O symmetrical stretching) and 1343 cm-1 (N-O asymmetrical stretching) and that of Nicotinamide at 3355.53 cm-1 (-N-H asymmetrical stretching), 3147.48 cm-1 (-N-H symmetrical stretching), 1676.73 cm-1 (-C = O stretching) and 1608.76 cm-1 (-N-H bending) [9].

Fig. 2: FT-IR Spectra of (A) Physical mixture of cilnidipine and nicotinamide (CNPM), formulations of cilnidipine-nicotinamide in the molar ratio: (B) 1:0.5-CN1, (C) 1:1-CN2, (D) 1:2–CN3

The FT-IR spectra of the physical mixture and various formulations (CN1, CN2 and CN3) are shown in fig. 2. The spectra of the physical mixture (CNPM) showed all the characteristic peaks of its components, thus indicating the absence of any chemical or physical interactions between Cilnidipine and Nicotinamide. The FT-IR spectra of CN1 and CN3 were similar to that of the physical mixture, indicating that the cocrystal formation has not occurred in these two formulations. In the formulation CN2, containing equimolar amounts of Cilnidipine and Nicotinamide, a considerable broadening of peaks and shift in the positions of the characteristic peaks of Cilnidipine and Nicotinamide were observed. A shift in the position and broadening of the –N-H stretching peak of Nicotinamide to 3900–3400 cm-1 indicated the formation of hydrogen bonds. A band broadening from 1550 cm-1 to 1450 cm-1 suggested the involvement of the nitro group of Cilnidipine in hydrogen bonding. FT-IR study indicated the formation of cocrystals in formulation CN2 [28].

Differential scanning calorimetric analysis

DSC is a rapid and sensitive technique commonly employed to identify, characterise and screen cocrystals [29]. DSC thermograms of Cilnidipine, Nicotinamide, their physical mixture (CNPM) and cocrystal -CN2 are shown in fig. 3.

Fig. 3: Overlain DSC thermograms of cilnidipine (Cil), Nicotinamide (NA), their physical mixture (CNPM) and cilnidipine-nicotinamide cocrystal (CN2)

The thermogram of Cilnidipine showed a single sharp melting endotherm at 109.69 °C and Nicotinamide at 129.24 °C, corresponding to their respective melting points. During DSC analysis of Cilnidipine and Nicotinamide, a single endothermic peak indicated that they melted without decomposing, making it possible to use this method for cocrystal screening [29]. An equimolar physical mixture of Cilnidipine and Nicotinamide (CNPM) showed endothermic peaks at 99.69 °C, 103.87 °C, 108.49 °C and 124.68 °C corresponding to the melting endotherm of cocrystal, eutectic, Cilnidipine and Nicotinamide respectively. Yamashita et al. showed that these thermal events are typical of mixtures capable of forming cocrystals [30]. Cilnidipine-Nicotinamide cocrystal (CN2) gave a distinct endothermic peak at 96.76 °C, distinguishable from those of its components. The appearance of a sharp melting endotherm and a shift in the peak's position in CN2 indicated the formation of a new crystalline phase. The lower melting point of CN2 than its components contributed to its enhanced water solubility [31].

Powder X-ray diffraction analysis

PXRD analysis is used to study changes in the crystal lattice of molecules, making it a valuable tool for characterising cocrystals. When there is a crystal lattice change, a molecule's unique PXRD pattern is altered [32]. The PXRD pattern of Cilnidipine (CIL), Nicotinamide (NA), and the cocrystal CN2 are given in fig. 4. The characteristic 2θ positions of Cilnidipine are 5.86°, 11.74°, 12.3°, 14.26°, 16.52°, 18.74°, 19.96°, 21.8°, 25.0°, 26.12°, while the peaks for Nicotinamide are 13.14°, 14.64°, 22.12°, 25.26°, 25.74°, and 27.22°. The CN2 exhibited peaks at 5.80°, 11.82°, 14.72°, 16.44°, 21.7°, 23.24° and 25.74°. The 2θ positions of CN2 are more intense, unique and different from its components, which confirm the formation of a new crystalline phase.

Saturation solubility measurements of cilnidipine-nicotinamide cocrystals

The saturation solubility analysis of Cilnidipine and formulation CN2 were determined in water, and the results are tabulated in table 2. The results showed that pure Cilnidipine is practically insoluble in water with an aqueous solubility of 5.53±0.26 µg/ml. The aqueous saturation solubility of the formulation CN2 was 145.77±0.021 µg/ml and thus exhibited 26.36 folds improvement in aqueous solubility compared to the pure Cilnidipine. This enhancement in aqueous solubility is due to the ability of the coformer Nicotinamide to decrease the solvation barrier of the cocrystal of Cilnidipine [33].

Fig. 4: PXRD pattern of cilnidipine (Cil), nicotinamide (NA) and cilnidipine-nicotinamide cocrystal (CN2)

Table 2: Saturation solubility studies of cilnidipine-nicotinamide cocrystals in water

Sample (n=3) Solubility in water (µg/ml) No: of folds increase in solubility
Cilnidipine 5.53±0.26 -
CN2 145.77±0.021 26.36

The value given is the mean±SD (n = 3).

Fig. 5: FESEM images of (A) Cilnidipine (B) Nicotinamide (C) Physical mixture (CNPM) and (D) Cocrystal formulation (CN2). (Magnification: 5.00K X; resolution: 2 µm)

Field emission-scanning electron microscopy

FE-SEM images of Cilnidipine, Nicotinamide, physical mixture (CNPM) and cocrystal formulation CN2 are given in fig. 5.

The FE-SEM analysis revealed that Cilnidipine, in its pure form, appeared as smooth, flat, and rectangular crystalline plates, while Nicotinamide was in the form of tubular solids. The physical mixture of the two substances did not show any noticeable change in the surface morphology of either Cilnidipine or Nicotinamide. However, in CN2, there was a significant reduction in the size of the Cilnidipine particle with a change in surface morphology.

Computational analysis-DFT studies

The optimised structure of cilnidipine, nicotinamide and cilnidipine-nicotinamide cocrystal (CN2) are shown in fig. 6 to 8.

Fig. 6: Optimized structure of cilnidipine

Fig. 7: Optimized structure of nicotinamide

Fig. 8: Optimized structure of cilnidipine-nicotinamide cocrystals with intermolecular hydrogen bonding

The geometrical parameters of Cilnidipine, Nicotinamide and Cilnidipine-Nicotinamide cocrystal (CN2) were calculated, and the details of bonds involved in intermolecular and intramolecular hydrogen bonds are tabulated in table 5. Considerable changes in the geometrical parameters of CN2 from that of its components were noted, including the formation of two intermolecular hydrogen bonds between Cilnidipine and Nicotinamide.

Table 3: Intermolecular and intramolecular hydrogen bonding in cilnidipine, nicotinamide and its cocrystal with their bond length in Å

Definition Bond involved Bond length (Å)
Cilnidipinea Nicotinamideb CN2
R (6-9) O6–N9 1.282 - 1.301
R (7-9) O7–N9 1.295 - 1.280
R (7-39)# O7­­­H39 2.341 - -
R (7-60)# O7H60 2.073 - -
R (6-48)# N6H48 - - 2.365
R (3-14) N2-H5 - 1.010 1.019
R (3-15) N2-H6 - 1.014 1.014
R (3-8) N2-C5 - 1.365 1.359
R (1-8) C5=O1 - 1.244 1.247
R (2-6) N1-C3 - 1.348 1.353
R (1-10)/R (65-74)# O65H74 - 2.363 2.345
R (6-75)* O6aH75b - - 2.322
R (6-78)* O6aH75b - - 1.898

#intramolecular hydrogen bonding; *intermolecular hydrogen bonding. †Numbering of atoms of Nicotinamide changed in CN2

In the optimised structure of Cilnidipine, two intramolecular hydrogen bonds (R(7-39); bond length-2.073 Å and R(7-60); bond length of 2.341 Å) were observed, both involving oxygen atom of the nitro group and hydrogen atom of the phenyl ring present in the side chain of dihydropyridine nucleus of Cilnidipine. However, these two intramolecular hydrogen bonds disappeared in the Cilnidipine-Nicotinamide cocrystal. Instead, two new intermolecular hydrogen bonds were formed, namely R(6-75) and R(6-78), with bond lengths of 2.322 Å and 1.898 Å, respectively. The R(6-75) bond was formed between the nitro oxygen of Cilnidipine and the C2 hydrogen atom of the pyridine nucleus of Nicotinamide(-N-O···H-C=), while the second intermolecular hydrogen bond, R(6-78), formed between the nitro oxygen of Cilnidipine and the amide hydrogen of Nicotinamide (-N-O···H-N-). The hydrogen bond between the oxygen (O6) of the nitro group and the amide hydrogen (-N-O···H-N-) had a bond length of 1.898 Å, comparable to the strong hydrogen bonds of a carboxylic acid with a hydroxyl group (–C=O···H–O-; bond length-1.884 Å) and between a carboxylic acid and an amino group (-C=O···H–N-; bond length-2.011 Å). This indicated that the hydrogen bond in CN2 was also strong [34]. As a result of intermolecular hydrogen bonding, the length of N-H and C=O bonds were increased in Nicotinamide by 0.009 Å and 0.003 Å and the N2-C5 bond in pyridine nucleus of Nicotinamide was decreased by 0.006 Å, which was consistent with many reported studies [35]. This confirmed the involvement of the oxygen atom of the nitro group of Cilnidipine and the amide hydrogen atom of Nicotinamide in forming intermolecular hydrogen bonding in CN2.

CONCLUSION

An eco-friendly approach, liquid-assisted grinding, was adopted to prepare the Cilnidipine-Nicotinamide cocrystals to enhance the poor solubility of Cilnidipine. The FT-IR, DSC, PXRD and FE-SEM studies confirmed that the cocrystals had been successfully formed. The aqueous solubility of Cilnidipine in CN2 was increased by 26-fold compared to the pure drug. Additionally, DFT analysis revealed that two strong intermolecular hydrogen bonds are formed between the nitro group of Cilnidipine and hydrogen atoms of Nicotinamide. Thus, the successful formation of Cilnidipine–Nicotinamide cocrystals by liquid-assisted grinding method demonstrates the potential application of this technique as a sustainable and promising approach to tackle the challenges posed by poorly water-soluble drugs.

ACKNOWLEDGMENT

The authors gratefully acknowledge the support of COE-AMGT, Dept of Chemical Engineering and Materials Science, Amrita Vishwa Vidyapeetham, Coimbatore, Tamilnadu, for the instrumentation support.

FUNDING

The authors thank the State Board of Medical Research, Govt. of Kerala, for the research grant (A1/8301/2016/GTDMCA).

AUTHORS CONTRIBUTIONS

All the authors have contributed equally.

CONFLICT OF INTERESTS

The authors declare that they have no conflict of interest.

REFERENCES

  1. Amidon GL, Lennernas H, Shah VP, Crison JR. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm Res. 1995;12(3):413-20. doi: 10.1023/a:1016212804288, PMID 7617530.

  2. Tharik AAMS, Meyyanathan SN. Recent patents of pharmaceutical co-crystals: product development on anti-cancer drugs and beyond. Recent Pat Anticancer Drug Discov. 2023;18(3):246-67. doi: 10.2174/1574892817666220913151252, PMID 36100994.

  3. Schultheiss N, Newman A. Pharmaceutical cocrystals and their physicochemical properties. Cryst Growth Des. 2009;9(6):2950-67. doi: 10.1021/cg900129f, PMID 19503732.

  4. Aitipamula S, Banerjee R, Bansal AK, Biradha K, Cheney ML, Choudhury AR. Polymorphs, salts, and cocrystals: what’s in a name? Cryst Growth Des. 2012;12(5):2147-52. doi: 10.1021/cg3002948.

  5. Hatta T, Takeda K, Shiotsu Y, Sugishita C, Adachi T, Kimura T. Switching to an L/N-type calcium channel blocker shows renoprotective effects in patients with chronic kidney disease: the kyoto cilnidipine study. J Int Med Res. 2012 Aug;40(4):1417-28. doi: 10.1177/147323001204000420, PMID 22971493.

  6. Kumar V, Agarwal S, Saboo B, Makkar B. RSSDI guidelines for the management of hypertension in patients with diabetes mellitus. Int J Diabetes Dev Ctries. 2022;42(4)Suppl 1:1-30. doi: 10.1007/s13410-022-01143-7, PMID 36536953.

  7. Kauser AS, Unnisa H, Namreen A, Saba A, Ansari JA. The impact of antihypertensive drug therapy in patients with chronic kidney disease: a prospective observational cohort study. Int J Pharm Pharm Sci. 2019 Nov;11(12):10-5. doi: 10.22159/ijpps.2019v11i12.35659.

  8. Ramya R, Kaladharan A. Evaluation of renoprotective effect of cilnidipine in patients with mild to moderate hypertension and type 2 diabetes mellitus-a prospective study. Asian J Pharm Clin Res. 2021 Jan 5:144-6.

  9. Pn R, ND. Formulation, development, and characterisation of cilnidipine-loaded solid lipid nanoparticles. Asian J Pharm Clin Res. 2018 Sep;11(9):120. doi: 10.22159/ajpcr.2018.v11i9.24666.

  10. PubChem. PubChem compound summary for CID 5282138, Cilnidipine. Bethesda: National Library of Medicine (US). National Center for Biotechnology Information; 2004. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/Cilnidipine. [Last accessed on 11 Nov 2023]

  11. Chadha R, Sharma M, Haneef J. Multicomponent solid forms of felodipine: preparation, characterization, physicochemical and in vivo studies. J Pharm Pharmacol. 2017 Feb 20;69(3):254-64. doi: 10.1111/jphp.12685, PMID 28134976.

  12. Miroshnyk I, Mirza S, Sandler N. Pharmaceutical co-crystals–an opportunity for drug product enhancement. Expert Opin Drug Deliv. 2009 Apr;6(4):333-41. doi: 10.1517/17425240902828304, PMID 19348603.

  13. Cruz Cabeza AJ. Acid–base crystalline complexes and the pKa rule. Cryst Eng Comm. 2012;14(20):6362. doi: 10.1039/c2ce26055g.

  14. Saraf GJ, Burade KKB, Gonjari ID, Hosmani AH, Pawar AA. A review on advances in pharmaceutical co-crystal preparation routes, intellectual property perspective and regulatory aspects. Int J Curr Pharm Sci 2022;14(5):4-12. doi: 10.22159/ijcpr.2022v14i5.2038.

  15. Singh M, Barua H, Jyothi VGSS, Dhondale MR, Nambiar AG, Agrawal AK. Cocrystals by design: a rational coformer selection approach for tackling the API problems. Pharmaceutics. 2023 Apr;15(4):1161. doi: 10.3390/pharmaceutics15041161, PMID 37111646.

  16. Raghuram M, Alam MS, Prasad M, Khanduri CH. Pharmaceutical cocrystal of prulifloxacin with nicotinamide. Int J Pharm Pharm Sci. 2014 Oct 1;6(10):180-4.

  17. Fukte S, Wagh PM, Rawat S. Conformer selection: an important tool in cocrystal formation. Int J Pharm Pharm Sci. 2014 Jul;6(7):9-14.

  18. Bidhuri N, Padhi S. Review on comprehensive description of development and assessment of co-crystal drug delivery system. Int J App Pharm. 2023 Sep;15(5):10-6. doi: 10.22159/ijap.2023v15i5.48579.

  19. Trask AV, Jones W. Crystal engineering of organic cocrystals by the solid-state grinding approach. In: Berlin, Heidelberg: Springer; 2005. p. 41-70. doi: 10.1007/b100995.

  20. Kumar Bandaru R, Rout SR, Kenguva G, Gorain B, Alhakamy NA, Kesharwani P. Recent advances in pharmaceutical cocrystals: from bench to market. Front Pharmacol. 2021 Nov;12:780582. doi: 10.3389/fphar.2021.780582, PMID 34858194.

  21. Li F, Zheng Z, Xia S, Yu L. Synthesis, co-crystal structure, and DFT calculations of a multicomponent co-crystal constructed from 1H-Benzotriazole and Tetrafluoroterephthalic acid. J Mol Struct. 2020 Nov;1219:128480. doi: 10.1016/j.molstruc.2020.128480.

  22. Sopyan I, Gozali D, Megantara S, Wahyuningrum R, Sunan Ks I. Review: an effort to increase the solubility and dissolution of active pharmaceutical ingredients. Int J App Pharm. 2022 Jan 7;14(1):22-7. doi: 10.22159/ijap.2022v14i1.43431.

  23. Olori A, Di Pietro P, Campopiano A. Preparation of UltraPure KBr pellet: new method for FTIR quantitative analysis. Int J Sci Acad Res. 2021 Feb;2(2):1015-20.

  24. Frisch MJ, Trucks GW, Schlegel HB, Scuseria GE, Robb MA, Cheeseman JR. Gaussian 09. Gaussian, Inc. Wallingford CT; 2016.

  25. Bolton EE, Wang Y, Thiessen PA, Bryant SH. PubChem: integrated platform of small molecules and biological activities. Elsevier; 2008. p. 217-41.

  26. O’Boyle NM, Banck M, James CA, Morley C, Vandermeersch T, Hutchison GR. Open babel: an open chemical toolbox. J Cheminform. 2011 Dec;3(1):33. doi: 10.1186/1758-2946-3-33, PMID 21982300.

  27. Weinhold F, Landis CR. Natural bond orbitals and extensions of localized bonding concepts. Chem Educ Res Pract. 2001;2(2):91-104. doi: 10.1039/B1RP90011K.

  28. Silverstein RM, Webster FX, Kiemle DJ. Spectrometric identification of organic compounds. 7th ed. NY: John Wiley & Sons, Inc; 2005. p. 80-107.

  29. Lu E, Rodriguez Hornedo N, Suryanarayanan R. A rapid thermal method for cocrystal screening. Cryst Eng Comm. 2008;10(6):665. doi: 10.1039/b801713c.

  30. Yamashita H, Hirakura Y, Yuda M, Teramura T, Terada K. Detection of cocrystal formation based on binary phase diagrams using thermal analysis. Pharm Res. 2013 Jan;30(1):70-80. doi: 10.1007/s11095-012-0850-1, PMID 22907418.

  31. Maddileti D, Jayabun SK, Nangia A. Soluble cocrystals of the xanthine oxidase inhibitor febuxostat. Cryst Growth Des. 2013 Jul;13(7):3188-96. doi: 10.1021/cg400583z.

  32. Padrela L, De Azevedo EG, Velaga SP. Powder X-ray diffraction method for the quantification of cocrystals in the crystallization mixture. Drug Dev Ind Pharm. 2012 Aug;38(8):923-9. doi: 10.3109/03639045.2011.633263, PMID 22092083.

  33. Kuminek G, Cao F, Bahia De Oliveira Da Rocha A, Gonçalves Cardoso S, Rodriguez Hornedo N. Cocrystals to facilitate delivery of poorly soluble compounds beyond-rule-of-5. Adv Drug Deliv Rev. 2016 Jun;101:143-66. doi: 10.1016/j.addr.2016.04.022, PMID 27137109.

  34. Allen FH, Baalham CA, Lommerse JPM, Raithby PR, Sparr E. Hydrogen-bond acceptor properties of nitro-O atoms: a combined crystallographic database and ab initio molecular orbital study. Acta Crystallogr B Struct Sci. 1997 Dec;53(6):1017-24. doi: 10.1107/S0108768197010239.

  35. Myshakina NS, Ahmed Z, Asher SA. Dependence of amide vibrations on hydrogen bonding. J Phys Chem B. 2008 Sep;112(38):11873-7. doi: 10.1021/jp8057355, PMID 18754632.