1
|
Šoltésová M, Pinon AC, Aussenac F, Schlagnitweit J, Reiter C, Purea A, Melzi R, Engelke F, Martin D, Krambeck S, Biscans A, Kay E, Emsley L, Schantz S. 1H- 19F cross-polarization magic angle spinning dynamic nuclear polarization NMR investigation of advanced pharmaceutical formulations. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2025; 371:107827. [PMID: 39793183 DOI: 10.1016/j.jmr.2024.107827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025]
Abstract
A new 3.2 mm 1H-19F-X magic angle spinning dynamic nuclear polarization NMR (MAS DNP-NMR) probe was developed with a unique coil design with separate radiofrequency channels for 1H excitation and 13C or 19F detection to enable acquisition of 1H-19F cross-polarization (CP) MAS experiments, direct-detected 19F spectra with proton decoupling, and acquisition on 13C with simultaneous double decoupling on the 1H and 19F channels as well as 1H-19F-13C double-CP experiments under low temperature MAS DNP conditions. We use these sequences to study AZD2811, which is an active pharmaceutical ingredient (API), in its pure dry state as well as in its corresponding drug delivery formulation consisting of drug-loaded polymeric nanoparticles (PNPs). Included in this study are also small interfering RNAs (siRNAs) for therapeutic targeting of peptidyl-prolyl cis-trans isomerase B (Ppib) mRNA. We demonstrate that 1H-19F CP MAS experiments performed on the new HFX probe represent a notable advantage over usually acquired direct-detected 19F experiments. The indirect 19F DNP enhancement εon/off(19F) = 26 was obtained via 1H-19F CP for the pure API impregnated with DNP solution, with an overall 30-fold sensitivity gain compared to the direct-detected 19F experiment under similar conditions. DNP enhancement value of εon/off(19F) = 42 was obtained via 1H-19F CP for the polymeric nanoparticle suspension and εon/off(19F) ≈ 150 were obtained for two different siRNAs in frozen DNP solution.
Collapse
Affiliation(s)
- Mária Šoltésová
- Swedish NMR Centre, University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Arthur C Pinon
- Swedish NMR Centre, University of Gothenburg, 413 90 Gothenburg, Sweden.
| | | | - Judith Schlagnitweit
- Centre de RMN à Très Hauts Champs de Lyon, UMR5082 CNRS/ENS-Lyon/Université Claude Bernard Lyon 1, Villeurbanne, France
| | | | | | | | | | - Dave Martin
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Stefanie Krambeck
- Cell, Gene and RNA Therapy, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Sweden
| | - Annabelle Biscans
- Cell, Gene and RNA Therapy, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Sweden
| | - Emma Kay
- Assays, Profiling and Cell Sciences, Discovery Sciences, AstraZeneca, Gothenburg, Sweden
| | - Lyndon Emsley
- Institut des Sciences et Ingénierie Chimiques, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Staffan Schantz
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
2
|
Isaev N, Lo Presti K, Frieß W. Insights into Folding and Molecular Environment of Lyophilized Proteins Using Pulsed Electron Paramagnetic Resonance Spectroscopy. Mol Pharm 2025; 22:424-432. [PMID: 39718349 DOI: 10.1021/acs.molpharmaceut.4c01008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
There is still an insufficient understanding of how the characteristics of protein drugs are maintained in the solid state of lyophilizates, including aspects such as protein distances, local environment, and structural preservation. To this end, we evaluated protein folding and the molecules' nearest environment by electron paramagnetic resonance (EPR) spectroscopy. Double electron-electron resonance (DEER) probe distances of up to approximately 200 Å and is suitable to investigate protein folding, local concentration, and aggregation, whereas electron spin echo envelope modulation (ESEEM) allows the study of the near environment within approximately 10 Å of the spin label. We spin-labeled human serum albumin (HSA) and freeze-dried different concentrations with 100 g/L deuterated sucrose. DEER showed distinct local concentration behaviors for two folding states, directly correlating folding percentage with the interprotein distance, reaching 2 nm at an HSA concentration of 84 g/L. Interestingly, 50% of the HSA molecules showed partial structural perturbation already at 2.6 g/L, which corresponds to a molar ratio Suc/HSA of 7469. This percentage increased to 97% with an increase in the HSA concentration to 84 g/L. The degree of protein perturbation cannot be told, and no signs of unfolding are found after reconstitution. ESEEM demonstrated a higher sucrose concentration around the protein label compared to the HSA environment in highly concentrated sucrose solutions. The partial unfolding detected in DEER could lead to label exposure and explain the enhanced sucrose detection in the intimate shell. Our work provides new insights regarding sucrose enrichment in the nearest shell of proteins upon lyophilization. In addition, the results indicate substantial partial structural perturbation, even in the presence of enormous supplies of stabilizing sugars. Thus, pulse EPR spectroscopy allows additional understanding of the solid state of protein lyophilizates, which is complementary to SANS, FTIR, or ssNMR.
Collapse
Affiliation(s)
- Nikolay Isaev
- Voevodsky Institute of Chemical Kinetics and Combustion SB RAS, Institutskaya Str. 3, Novosibirsk 630090, Russia
| | - Ken Lo Presti
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Wolfgang Frieß
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| |
Collapse
|
3
|
Hau J, DiPasquale A, Lubach JW, Chakravarty P. Investigation of the State of Hydration of a Non-Stoichiometric Hydrate in a Low Dose Formulation Using 19F Solid-State NMR. J Pharm Sci 2024; 113:3065-3077. [PMID: 39122184 DOI: 10.1016/j.xphs.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
A variable or non-stoichiometric hydrate of GDC-4379 was developed into a formulated capsule with a 1% drug loading. The water content of this hydrate varied from 0-0.7 moles over the relative humidity (RH) range of 0-98% (25°C). Since a variable state of hydration coupled with rapid equilibration of lattice water with the environmental RH can lead to challenges in formulation development, an analytical method to directly and accurately determine the state of hydration of the active in such a low dose formulation was deemed necessary. Owing to its high selectivity and fast acquisition times, 19F solid-state NMR was effectively utilized to directly determine the lattice water content of the active in the formulated capsule. By correlating Δδ, the chemical shift difference between the isotropic peaks, with the relative humidity and ultimately the lattice water content, the state of hydration of GDC-4379 in the formulated capsule was experimentally determined as 0.63 moles of water/mole of anhydrate.
Collapse
Affiliation(s)
- Jonathan Hau
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, CA 94080, United States
| | - Antonio DiPasquale
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, CA 94080, United States
| | - Joseph W Lubach
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, CA 94080, United States.
| | - Paroma Chakravarty
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, CA 94080, United States.
| |
Collapse
|
4
|
Ko CH, Tadesse AB, Kabiso AC. Spectrochip-based Calibration Curve Modeling (CCM) for Rapid and Accurate Multiple Analytes Quantification in Urinalysis. Heliyon 2024; 10:e37722. [PMID: 39328528 PMCID: PMC11425109 DOI: 10.1016/j.heliyon.2024.e37722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Most urine test strips are intended to enable the general population to rapidly and easily diagnose potential renal disorders. It is semi-quantitative in nature, and although the procedure is straightforward, certain factors will affect the judgmental outcomes. This study describes rapid and accurate quantification of twelve urine test strip parameters: leukocytes, nitrite, urobilinogen, protein, pH, occult blood, specific gravity, ketone, bilirubin, glucose, microalbumin, and creatinine using a micro-electromechanical system (MEMS)-based spectrophotometer, known as a spectrochip. For each parameter, absorption spectra were measured three times independently at eight different concentration levels of diluted standard solutions, and the average spectral intensities were calculated to establish the calibration curve under the characteristic wavelength (λ c ). Then, regression analysis on the calibration curve was performed with GraphPad Prism software, which revealed that the coefficient of determination (R 2 ) of the modeled calibration curves was greater than 0.95. This result illustrates that the measurements exceed standard levels, confirming the importance of a spectrochip for routine multi-parameter urine analysis. Thus, it is possible to obtain the spectral signal strength for each parameter at its characteristic wavelength in order to compare directly with the calibration curves in the future, even in situations when sample concentration is unknown. Additionally, the use of large testing machines can be reduced in terms of cost, time, and space by adopting a micro urine testing platform based on spectrochip, which also improves operational convenience and effectively enables point-of-care (POC) testing in urinalysis.
Collapse
Affiliation(s)
- Cheng-Hao Ko
- Graduate Institute of Automation and Control, National Taiwan University of Science and Technology, Taipei, Taiwan
- Spectrochip Inc., Hsinchu, Taiwan
| | - Ashenafi Belihu Tadesse
- Graduate Institute of Automation and Control, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Abel Chernet Kabiso
- Graduate Institute of Automation and Control, National Taiwan University of Science and Technology, Taipei, Taiwan
| |
Collapse
|
5
|
Mi J, Chen Y, Atterberry BA, Nordstrom FL, Hirsh DA, Rossini AJ. Probing the Molecular and Macroscopic Structure of Solid Solutions by Dynamic Nuclear Polarization (DNP) Enhanced 13C and 15N Solid-State NMR Spectroscopy. Mol Pharm 2024; 21:2949-2959. [PMID: 38685852 DOI: 10.1021/acs.molpharmaceut.4c00083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Crystallization is a widely used purification technique in the manufacture of active pharmaceutical ingredients (APIs) and precursor molecules. However, when impurities and desired compounds have similar molecular structures, separation by crystallization may become challenging. In such cases, some impurities may form crystalline solid solutions with the desired product during recrystallization. Understanding the molecular structure of these recrystallized solid solutions is crucial to devise methods for effective purification. Unfortunately, there are limited analytical techniques that provide insights into the molecular structure or spatial distribution of impurities that are incorporated within recrystallized products. In this study, we investigated model solid solutions formed by recrystallizing salicylic acid (SA) in the presence of anthranilic acid (AA). These two molecules are known to form crystalline solid solutions due to their similar molecular structures. To overcome challenges associated with the long 1H longitudinal relaxation times (T1(1H)) of SA and AA, we employed dynamic nuclear polarization (DNP) and 15N isotope enrichment to enable solid-state NMR experiments. Results of solid-state NMR experiments and DFT calculations revealed that SA and AA are homogeneously alloyed as a solid solution. Heteronuclear correlation (HETCOR) experiments and plane-wave DFT structural models provide further evidence of the molecular-level interactions between SA and AA. This research provides valuable insights into the molecular structure of recrystallized solid solutions, contributing to the development of effective purification strategies and an understanding of the physicochemical properties of solid solutions.
Collapse
Affiliation(s)
- Jiashan Mi
- Department of Chemistry, Iowa State University, Ames, Iowa 50010, United States
| | - Yunhua Chen
- Department of Chemistry, Iowa State University, Ames, Iowa 50010, United States
| | | | - Fredrik L Nordstrom
- Material & Analytical Sciences, Boehringer-Ingelheim, Ridgefield, Connecticut 06877, United States
| | - David A Hirsh
- Material & Analytical Sciences, Boehringer-Ingelheim, Ridgefield, Connecticut 06877, United States
| | - Aaron J Rossini
- Department of Chemistry, Iowa State University, Ames, Iowa 50010, United States
| |
Collapse
|
6
|
Rahman M, Dannatt HRW, Blundell CD, Hughes LP, Blade H, Carson J, Tatman BP, Johnston ST, Brown SP. Polymorph Identification for Flexible Molecules: Linear Regression Analysis of Experimental and Calculated Solution- and Solid-State NMR Data. J Phys Chem A 2024; 128:1793-1816. [PMID: 38427685 PMCID: PMC10945485 DOI: 10.1021/acs.jpca.3c07732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 03/03/2024]
Abstract
The Δδ regression approach of Blade et al. [ J. Phys. Chem. A 2020, 124(43), 8959-8977] for accurately discriminating between solid forms using a combination of experimental solution- and solid-state NMR data with density functional theory (DFT) calculation is here extended to molecules with multiple conformational degrees of freedom, using furosemide polymorphs as an exemplar. As before, the differences in measured 1H and 13C chemical shifts between solution-state NMR and solid-state magic-angle spinning (MAS) NMR (Δδexperimental) are compared to those determined by gauge-including projector augmented wave (GIPAW) calculations (Δδcalculated) by regression analysis and a t-test, allowing the correct furosemide polymorph to be precisely identified. Monte Carlo random sampling is used to calculate solution-state NMR chemical shifts, reducing computation times by avoiding the need to systematically sample the multidimensional conformational landscape that furosemide occupies in solution. The solvent conditions should be chosen to match the molecule's charge state between the solution and solid states. The Δδ regression approach indicates whether or not correlations between Δδexperimental and Δδcalculated are statistically significant; the approach is differently sensitive to the popular root mean squared error (RMSE) method, being shown to exhibit a much greater dynamic range. An alternative method for estimating solution-state NMR chemical shifts by approximating the measured solution-state dynamic 3D behavior with an ensemble of 54 furosemide crystal structures (polymorphs and cocrystals) from the Cambridge Structural Database (CSD) was also successful in this case, suggesting new avenues for this method that may overcome its current dependency on the prior determination of solution dynamic 3D structures.
Collapse
Affiliation(s)
- Mohammed Rahman
- Department
of Physics, University of Warwick, Coventry CV4 7AL, U.K.
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| | | | | | - Leslie P. Hughes
- Oral
Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield SK10 2NA, U.K.
| | - Helen Blade
- Oral
Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield SK10 2NA, U.K.
| | - Jake Carson
- Mathematics
Institute at Warwick, University of Warwick, Coventry CV4 7AL, U.K.
| | - Ben P. Tatman
- Department
of Physics, University of Warwick, Coventry CV4 7AL, U.K.
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| | | | - Steven P. Brown
- Department
of Physics, University of Warwick, Coventry CV4 7AL, U.K.
| |
Collapse
|
7
|
Stirk AJ, Holmes ST, Souza FES, Hung I, Gan Z, Britten JF, Rey AW, Schurko RW. An unusual ionic cocrystal of ponatinib hydrochloride: characterization by single-crystal X-ray diffraction and ultra-high field NMR spectroscopy. CrystEngComm 2024; 26:1219-1233. [PMID: 38419975 PMCID: PMC10897533 DOI: 10.1039/d3ce01062g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/26/2024] [Indexed: 03/02/2024]
Abstract
This study describes the discovery of a unique ionic cocrystal of the active pharmaceutical ingredient (API) ponatinib hydrochloride (pon·HCl), and characterization using single-crystal X-ray diffraction (SCXRD) and solid-state NMR (SSNMR) spectroscopy. Pon·HCl is a multicomponent crystal that features an unusual stoichiometry, with an asymmetric unit containing both monocations and dications of the ponatinib molecule, three water molecules, and three chloride ions. Structural features include (i) a charged imidazopyridazine moiety that forms a hydrogen bond between the ponatinib monocations and dications and (ii) a chloride ion that does not feature hydrogen bonds involving any organic moiety, instead being situated in a "square" arrangement with three water molecules. Multinuclear SSNMR, featuring high and ultra-high fields up to 35.2 T, provides the groundwork for structural interpretation of complex multicomponent crystals in the absence of diffraction data. A 13C CP/MAS spectrum confirms the presence of two crystallographically distinct ponatinib molecules, whereas 1D 1H and 2D 1H-1H DQ-SQ spectra identify and assign the unusually deshielded imidazopyridazine proton. 1D 35Cl spectra obtained at multiple fields confirm the presence of three distinct chloride ions, with density functional theory calculations providing key relationships between the SSNMR spectra and H⋯Cl- hydrogen bonding arrangements. A 2D 35Cl → 1H D-RINEPT spectrum confirms the spatial proximities between the chloride ions, water molecules, and amine moieties. This all suggests future application of multinuclear SSNMR at high and ultra-high fields to the study of complex API solid forms for which SCXRD data are unavailable, with potential application to heterogeneous mixtures or amorphous solid dispersions.
Collapse
Affiliation(s)
| | - Sean T Holmes
- Department of Chemistry & Biochemistry, Florida State University Tallahassee FL 32306 USA
- National High Magnetic Field Laboratory Tallahassee FL 32310 USA
| | | | - Ivan Hung
- National High Magnetic Field Laboratory Tallahassee FL 32310 USA
| | - Zhehong Gan
- National High Magnetic Field Laboratory Tallahassee FL 32310 USA
| | - James F Britten
- MAX Diffraction Facility, McMaster University Hamilton ON L8S 4M1 Canada
| | - Allan W Rey
- Apotex Pharmachem Inc. Brantford ON N3T 6B8 Canada
| | - Robert W Schurko
- Department of Chemistry & Biochemistry, Florida State University Tallahassee FL 32306 USA
- National High Magnetic Field Laboratory Tallahassee FL 32310 USA
| |
Collapse
|
8
|
Zheng Z, Su Y, Schmidt-Rohr K. Corrected solid-state 13 C nuclear magnetic resonance peak assignment and side-group quantification of hydroxypropyl methylcellulose acetyl succinate pharmaceutical excipients. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2023; 61:595-605. [PMID: 37649159 DOI: 10.1002/mrc.5390] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023]
Abstract
Hydroxypropyl methylcellulose acetyl succinate (HPMCAS) is widely used as a pharmaceutical excipient, making a detailed understanding of its tunable structure important for formulation design. Several recently reported peak assignments in the solid-state 13 C NMR spectrum of HPMCAS have been corrected here using peak integrals in quantitative spectra, spectral editing, empirical chemical-shift predictions based on solution NMR, and full spectrum simulation analogous to deconvolution. Unlike in cellulose, the strong peak at 84 ppm must be assigned to C2 and C3 methyl ethers, instead of regular C4 of cellulose. The proposed assignment of signals at <65 ppm to OCH sites, including C5 of cellulose, could not be confirmed. CH2 spectral editing showed two resolved OCH2 bands, a more intense one from O-CH2 ethers of C6 at >69 ppm and a smaller one from its esters and possibly residual CH2 -OH groups, near 63 ppm. The strong intensities of resolved signals of acetyl, succinoyl, and oxypropyl substituents indicated the substitution of >85% of the OH groups in HPMCAS. The side-group concentrations in three different grades of HPMCAS were quantified.
Collapse
Affiliation(s)
- Zhaoxi Zheng
- Department of Chemistry, Brandeis University, Waltham, Massachusetts, USA
| | - Yongchao Su
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Klaus Schmidt-Rohr
- Department of Chemistry, Brandeis University, Waltham, Massachusetts, USA
| |
Collapse
|
9
|
Chen Y, Mi J, Rossini AJ. A focus on detection of polymorphs by dynamic nuclear polarization solid-state nuclear magnetic resonance spectroscopy. Chem Sci 2023; 14:11296-11299. [PMID: 37886103 PMCID: PMC10599483 DOI: 10.1039/d3sc90177g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023] Open
Abstract
Solid-state nuclear magnetic resonance (ssNMR) spectroscopy has found increasing application as a method for quantification and structure determination of solid forms (polymorphs) of organic solids and active pharmaceutical ingredients (APIs). However, ssNMR spectroscopy suffers from low sensitivity and resolution, making it challenging to detect dilute solid forms that may be present after recrystallization or reaction with co-formers. Cousin et al. (S. F. Cousin et al., Chem. Sci., 2023, https://doi.org/10.1039/D3SC02063K) have demonstrated that dynamic nuclear polarization (DNP) enhanced 13C cross-polarization (CP) saturation recovery experiments can be used to detect dilute polymorphic forms that are present within a mixture of solid forms. Enhancement of the NMR signal by DNP and differences in signal build-up rates for different polymorphs provide the sensitivity and contrast needed to resolve NMR signals from minor polymorphic forms. This method demonstrated by Cousin et al. should aid the discovery of solid drug forms.
Collapse
Affiliation(s)
- Yunhua Chen
- Analytical Research & Development, AbbVie, Inc. North Chicago Illinois 60064 USA
| | - Jiashan Mi
- Department of Chemistry, Iowa State University Ames IA 50011 USA
| | - Aaron J Rossini
- Department of Chemistry, Iowa State University Ames IA 50011 USA
| |
Collapse
|
10
|
Rehman Z, Franks WT, Nguyen B, Schmidt HF, Scrivens G, Brown SP. Discovering the Solid-State Secrets of Lorlatinib by NMR Crystallography: To Hydrogen Bond or not to Hydrogen Bond. J Pharm Sci 2023; 112:1915-1928. [PMID: 36868358 DOI: 10.1016/j.xphs.2023.02.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023]
Abstract
Lorlatinib is an active pharmaceutical ingredient (API) used in the treatment of lung cancer. Here, an NMR crystallography analysis is presented whereby the single-crystal X-ray diffraction structure (CSD: 2205098) determination is complemented by multinuclear (1H, 13C, 14/15N, 19F) magic-angle spinning (MAS) solid-state NMR and gauge-including projector augmented wave (GIPAW) calculation of NMR chemical shifts. Lorlatinib crystallises in the P21 space group, with two distinct molecules in the asymmetric unit cell, Z' = 2. Three of the four NH2 hydrogen atoms form intermolecular hydrogen bonds, N30-H…N15 between the two distinct molecules and N30-H…O2 between two equivalent molecules. This is reflected in one of the NH21H chemical shifts being significantly lower, 4.0 ppm compared to 7.0 ppm. Two-dimensional 1H-13C, 14N-1H and 1H (double-quantum, DQ)-1H (single-quantum, SQ) MAS NMR spectra are presented. The 1H resonances are assigned and specific HH proximities corresponding to the observed DQ peaks are identified. The resolution enhancement at a 1H Larmor frequency of 1 GHz as compared to 500 or 600 MHz is demonstrated.
Collapse
Affiliation(s)
- Zainab Rehman
- Department of Physics, University of Warwick, Coventry, CV4 7AL, UK
| | - W Trent Franks
- Department of Physics, University of Warwick, Coventry, CV4 7AL, UK
| | | | | | | | - Steven P Brown
- Department of Physics, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
11
|
Hung I, Gan Z. Pushing the limit of MQMAS for low-γ quadrupolar nuclei in pharmaceutical hydrochlorides. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2023; 350:107423. [PMID: 36966726 DOI: 10.1016/j.jmr.2023.107423] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 05/10/2023]
Abstract
Solid-state NMR of quadrupolar nuclei such as 35Cl has become a useful tool to characterize polymorphism in pharmaceutical hydrochlorides. The two-dimensional multiple-quantum magic-angle spinning (MQMAS) experiment can achieve isotropic resolution, and separate quadrupolar line shapes for samples with multiple sites but the pulse sequence efficiency is often low, limiting applications due to the intrinsically low NMR signals and rf field from the low gyromagnetic ratios γ. The use of cosine low-power MQMAS pulse sequences and high magnetic fields is presented to push the limit of MQMAS for insensitive low-γ quadrupolar nuclei. The improved efficiency and fields up to 35.2 T enable the acquisition of MQMAS spectra for pharmaceutical samples with multiple 35Cl sites, large quadrupolar couplings and/or in diluted dosage forms.
Collapse
Affiliation(s)
- Ivan Hung
- National High Magnetic Field Laboratory, 1800 East Paul Dirac Drive, Tallahassee, FL 32310, USA
| | - Zhehong Gan
- National High Magnetic Field Laboratory, 1800 East Paul Dirac Drive, Tallahassee, FL 32310, USA.
| |
Collapse
|
12
|
Kestur U, Patel A, Badawy S, Mathias N, Zhang L. Strategies for Managing Solid Form Transformation Risk in Drug Product. J Pharm Sci 2023; 112:909-921. [PMID: 36513146 DOI: 10.1016/j.xphs.2022.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/03/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022]
Abstract
The International Conference of Harmonization (ICH) Q6A document provides guidance on setting specifications for new drug substances and drug products.1 In this paper we focus on decision trees 4 (#1) to (#3) in the guidance related to solid-state form transformation. Form transformation could occur from use of high energy forms to overcome solubility challenges or stresses from manufacturing processes. The decision trees provide guidance on when and how polymorphic form changes should be monitored and controlled. However, guidance is high level and does not capture aspects related to assessments needed to understand if there is a risk of transformation or tools that can be integrated to understand the severity of bioavailability impact at different stages of development. The objective of this paper is therefore to provide comprehensive chemistry manufacturing and controls (CMC) and regulatory strategies to manage the risk of form transformation. This includes practical workflows for form transformation risk assessment, analytical tools to detect and quantify the transformation including their shortcomings, biopharmaceutical tools to understand the severity of transformation risk and if needed justify the limits based on clinical relevance. Finally, a few case studies are discussed that capture how the workflow can be used to manage transformation risk.
Collapse
Affiliation(s)
- Umesh Kestur
- Drug Product Development, Bristol Myers Squibb, One Squibb Drive, New Brunswick, NJ 08903, USA.
| | - Anisha Patel
- Drug Product Development, Bristol Myers Squibb, One Squibb Drive, New Brunswick, NJ 08903, USA
| | - Sherif Badawy
- Drug Product Development, Bristol Myers Squibb, One Squibb Drive, New Brunswick, NJ 08903, USA
| | - Neil Mathias
- Drug Product Development, Bristol Myers Squibb, One Squibb Drive, New Brunswick, NJ 08903, USA
| | - Limin Zhang
- Analytical Strategy & Operations, Bristol Myers Squibb, One Squibb Drive, New Brunswick, NJ 08903, USA
| |
Collapse
|
13
|
Ogaeri Y, Suzuki N, Fukami T, Nishiyama Y. Internuclear distance measurements between 1H and 14N in multi-component rigid solids at fast MAS. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2023; 348:107378. [PMID: 36702044 DOI: 10.1016/j.jmr.2023.107378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/30/2022] [Accepted: 01/13/2023] [Indexed: 06/18/2023]
Abstract
1H-14N internuclear distances are readily and accurately measured using the symmetry-based phase-modulated resonance-echo saturation-pulse double-resonance (PM-S-RESPDOR) method in rigid solids. The fraction curve, (S0 - S')/S0, is represented by a single variable of a 1H-14N heteronuclear dipolar coupling, where S0 and S' are the PM-S-RESPDOR signal intensity with and without 14N PM saturation pulse, respectively. Analytical equation of the fraction curve easily provides 1H-14N couplings. This treatment is only applicable when NH proton resonance is well separated from the other proton peaks. With the limited 1H resolution even at fast MAS > 60 kHz, unfortunately, this condition is not necessarily satisfied especially in multi-component systems which often appear in pharmaceutical applications. To overcome this problem, T-HMQC filtering is applied to suppress the 1H signals other than NH proton prior to the PM-S-RESPDOR experiments. The method is well demonstrated on two components acetaminophen-oxalic acid (APAP-OXA) systems. Further analysis of orientation dependence of T-HMQC and PM-S-RESPDOR shows that the analytical equation can be safely applied in the analysis of T-HMQC filtered PM-S-RESPDOR experiments.
Collapse
Affiliation(s)
- Yutaro Ogaeri
- JEOL Ltd., Akishima, Tokyo 196-8558, Japan; RIKEN-JEOL Collaboration Center, RIKEN, Yokohama, Kanagawa 230-0045, Japan
| | - Naoto Suzuki
- Laboratory of Pharmaceutics, School of Pharmacy, Nihon University, Chiba, Japan
| | - Toshiro Fukami
- Department of Molecular Pharmaceutics, Meiji Pharmaceutical University, Tokyo, Japan
| | - Yusuke Nishiyama
- JEOL Ltd., Akishima, Tokyo 196-8558, Japan; RIKEN-JEOL Collaboration Center, RIKEN, Yokohama, Kanagawa 230-0045, Japan.
| |
Collapse
|
14
|
Du Y, Frank D, Chen Z, Struppe J, Su Y. Ultrafast magic angle spinning NMR characterization of pharmaceutical solid polymorphism: A posaconazole example. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2023; 346:107352. [PMID: 36535214 DOI: 10.1016/j.jmr.2022.107352] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 06/17/2023]
Abstract
Protons represent the most NMR-sensitive nucleus in pharmaceutical compounds. Therefore, proton-detected solid-state NMR techniques under fast magic angle spinning are among the few solutions to overcome the challenge of low sensitivity to analyze natural abundant drug substances and products. In this study, we report the structural characterization of crystal polymorphs of a commercial drug molecule, posaconazole, with a relatively large molecular weight of 700.8 g·mol-1 and at the natural abundance. The enhanced sensitivity and resolution at 100 kHz MAS enables the exploration of the distinct intermolecular packing in posaconazole forms I, III, and γ. These results demonstrate that proton-detected homo- and heteronuclear correlation methods can probe the structural details of pharmaceutical polymorphism.
Collapse
Affiliation(s)
- Yong Du
- Analytical Research & Development, Merck & Co., Inc, Rahway, NJ 07065, USA
| | - Derek Frank
- Process Research & Development, Merck & Co., Inc, Rahway, NJ 07065, USA
| | - Zhenxuan Chen
- Analytical Research & Development, Merck & Co., Inc, Rahway, NJ 07065, USA
| | | | - Yongchao Su
- Analytical Research & Development, Merck & Co., Inc, Rahway, NJ 07065, USA.
| |
Collapse
|
15
|
Nugrahani I, Susanti E, Adawiyah T, Santosa S, Laksana AN. Non-Covalent Reactions Supporting Antiviral Development. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27249051. [PMID: 36558183 PMCID: PMC9783875 DOI: 10.3390/molecules27249051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/08/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
Viruses are the current big enemy of the world's healthcare systems. As the small infector causes various deadly diseases, from influenza and HIV to COVID-19, the virus continues to evolve from one type to its mutants. Therefore, the development of antivirals demands tremendous attention and resources for drug researchers around the world. Active pharmaceutical ingredients (API) development includes discovering new drug compounds and developing existing ones. However, to innovate a new antiviral takes a very long time to test its safety and effectiveness, from structure modeling to synthesis, and then requires various stages of clinical trials. Meanwhile, developing the existing API can be more efficient because it reduces many development stages. One approach in this effort is to modify the solid structures to improve their physicochemical properties and enhance their activity. This review discusses antiviral multicomponent systems under the research phase and has been marketed. The discussion includes the types of antivirals, their counterpart compound, screening, manufacturing methods, multicomponent systems yielded, characterization methods, physicochemical properties, and their effects on their pharmacological activities. It is hoped that the opportunities and challenges of solid antiviral drug modifications can be drawn in this review as important information for further antiviral development.
Collapse
|
16
|
Du Y, Su Y. 19F Solid-state NMR characterization of pharmaceutical solids. SOLID STATE NUCLEAR MAGNETIC RESONANCE 2022; 120:101796. [PMID: 35688018 DOI: 10.1016/j.ssnmr.2022.101796] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Solid-state NMR has been increasingly recognized as a high-resolution and versatile spectroscopic tool to characterize drug substances and products. However, the analysis of pharmaceutical materials is often carried out at natural isotopic abundance and a relatively low drug loading in multi-component systems and therefore suffers from challenges of low sensitivity. The fact that fluorinated therapeutics are well represented in pipeline drugs and commercial products offers an excellent opportunity to utilize fluorine as a molecular probe for pharmaceutical analysis. We aim to review recent advancements of 19F magic angle spinning NMR methods in modern drug research and development. Applications to polymorph screening at the micromolar level, structural elucidation, and investigation of molecular interactions at the Ångström to submicron resolution in drug delivery, stability, and quality will be discussed.
Collapse
Affiliation(s)
- Yong Du
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, 07065, United States
| | - Yongchao Su
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, 07065, United States; Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, IN, 47907, United States; Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, United States; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT, 06269, United States.
| |
Collapse
|
17
|
Research Progress of NMR in Natural Product Quantification. Molecules 2021; 26:molecules26206308. [PMID: 34684890 PMCID: PMC8541192 DOI: 10.3390/molecules26206308] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 12/17/2022] Open
Abstract
In the fields of medicine and health, traditional high-performance liquid chromatography or UV-visible spectrophotometry is generally used for substance quantification. However, over time, nuclear magnetic resonance spectroscopy (NMR) has gradually become more mature. Nuclear magnetic resonance spectroscopy has certain advantages in the quantitative analysis of substances, such as being nondestructive, having a high flux and short analysis time. Nuclear magnetic resonance spectroscopy has been included in the pharmacopoeiae of various countries. In this paper, the principle of nuclear magnetic resonance spectroscopy and the recent progress in the quantitative study of natural products by NMR are reviewed, and its application in the quantitative study of natural products is proposed. At the same time, the problems of using NMR alone to quantify natural products are summarized and corresponding suggestions are put forward.
Collapse
|
18
|
Nugrahani I. Sustainable Pharmaceutical Preparation Methods and Solid-state Analysis Supporting Green Pharmacy. CURR PHARM ANAL 2021. [DOI: 10.2174/1573412916999200711150729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Every "entity" or compound has physical and chemical properties as references for the synthesis
and determination of the entity's structure. Thermodynamically, solid-state is the most stable
matter in the universe and to be the ideal form in structure elucidation of pharmaceutical. The dry
treatments, such as mechanochemistry, microwave heating, and the using of deep eutectic agent are
becoming popular. These techniques are viewed as futuristic methods for reducing environmental damage,
in line with "green pharmacy" concept. On the other hand, solid-state analysis methods from the
simplest to the most sophisticated one have been used in the long decades, but most are for qualitative
purposes. Recently many reports have proven that solid-state analysis instruments are reliable and prospective
for implementing in the quantitative measurement. Infrared spectroscopy, powder x-ray diffraction,
and differential scanning calorimetry have been employed in various kinetics and content determination
studies. A revolutionary method developed for structural elucidation is single-crystal diffraction,
which is capable of rapidly and accurately determining a three-dimensional chemical structure.
Hereby it is shown that the accurate, precise, economic, ease, rapid-speed, and reliability of solidstate
analysis methods are eco-benefits by reducing the reagent, catalyst, and organic solvent.
Collapse
Affiliation(s)
- Ilma Nugrahani
- Pharmacochemistry Department, School of Pharmacy, Bandung Institute of Technology, Bandung, Indonesia
| |
Collapse
|
19
|
Thakore SD, Akhtar J, Jain R, Paudel A, Bansal AK. Analytical and Computational Methods for the Determination of Drug-Polymer Solubility and Miscibility. Mol Pharm 2021; 18:2835-2866. [PMID: 34041914 DOI: 10.1021/acs.molpharmaceut.1c00141] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In the pharmaceutical industry, poorly water-soluble drugs require enabling technologies to increase apparent solubility in the biological environment. Amorphous solid dispersion (ASD) has emerged as an attractive strategy that has been used to market more than 20 oral pharmaceutical products. The amorphous form is inherently unstable and exhibits phase separation and crystallization during shelf life storage. Polymers stabilize the amorphous drug by antiplasticization, reducing molecular mobility, reducing chemical potential of drug, and increasing glass transition temperature in ASD. Here, drug-polymer miscibility is an important contributor to the physical stability of ASDs. The current Review discusses the basics of drug-polymer interactions with the major focus on the methods for the evaluation of solubility and miscibility of the drug in the polymer. Methods for the evaluation of drug-polymer solubility and miscibility have been classified as thermal, spectroscopic, microscopic, solid-liquid equilibrium-based, rheological, and computational methods. Thermal methods have been commonly used to determine the solubility of the drug in the polymer, while other methods provide qualitative information about drug-polymer miscibility. Despite advancements, the majority of these methods are still inadequate to provide the value of drug-polymer miscibility at room temperature. There is still a need for methods that can accurately determine drug-polymer miscibility at pharmaceutically relevant temperatures.
Collapse
Affiliation(s)
- Samarth D Thakore
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Junia Akhtar
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Ranjna Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Amrit Paudel
- Research Center Pharmaceutical Engineering (RCPE) GmbH, Inffeldgasse 13, 8010 Graz, Austria.,Institute for Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13, 8010 Graz, Austria
| | - Arvind K Bansal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| |
Collapse
|
20
|
Chen Y, Mutukuri TT, Wilson NE, Zhou QT. Pharmaceutical protein solids: Drying technology, solid-state characterization and stability. Adv Drug Deliv Rev 2021; 172:211-233. [PMID: 33705880 PMCID: PMC8107147 DOI: 10.1016/j.addr.2021.02.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/18/2021] [Accepted: 02/22/2021] [Indexed: 01/30/2023]
Abstract
Despite the boom in biologics over the past decade, the intrinsic instability of these large molecules poses significant challenges to formulation development. Almost half of all pharmaceutical protein products are formulated in the solid form to preserve protein native structure and extend product shelf-life. In this review, both traditional and emerging drying techniques for producing protein solids will be discussed. During the drying process, various stresses can impact the stability of protein solids. However, understanding the impact of stress on protein product quality can be challenging due to the lack of reliable characterization techniques for biological solids. Both conventional and advanced characterization techniques are discussed including differential scanning calorimetry (DSC), solid-state Fourier transform infrared spectrometry (ssFTIR), solid-state fluorescence spectrometry, solid-state hydrogen deuterium exchange (ssHDX), solid-state nuclear magnetic resonance (ssNMR) and solid-state photolytic labeling (ssPL). Advanced characterization tools may offer mechanistic investigations into local structural changes and interactions at higher resolutions. The continuous exploration of new drying techniques, as well as a better understanding of the effects caused by different drying techniques in solid state, would advance the formulation development of biological products with superior quality.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Tarun Tejasvi Mutukuri
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Nathan E Wilson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA.
| |
Collapse
|
21
|
|
22
|
Tackling quantitative polymorphic analysis through fixed-dose combination tablets production. Pyrazinamide polymorphic assessment. J Pharm Biomed Anal 2020; 194:113786. [PMID: 33281002 DOI: 10.1016/j.jpba.2020.113786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 11/23/2022]
Abstract
Pyrazinamide (PZA), Rifampicin (RIF), Isoniazid (ISH) and Ethambutol (ETB) form the core for the treatment of Tuberculosis, today a devastating disease in low-income populations around the world. These drugs are usually administrated by fixed-dose combination (FDC) products, to favour the patient compliance and prevent bacterial resistance. PZA exists in four enantiotropically-related polymorphs (Forms α, δ, β and γ), but only Form α is considered suitable for pharmaceutical products due to its stability and bioavailability properties. The classical approaches to address solid-state (microscopy, X-ray diffraction and calorimetry) shows limitations for quantification of polymorphs in the presence of excipients and other active components, as in the case of FDC tablets. In this work, an overall strategy was developed using near infrared spectroscopy (NIR) coupled to partial least squares regression (PLS) to quantify Form α of PZA in drug substance (raw material) and PZA/RIF/ISH-FDC tablets. For this purpose, two PLS models were constructed, one for drug substance preparing training (n = 30) and validation (n = 18) samples with a ternary composition (Form α/Form δ/Form γ), and other for FDC drug products, also including the appropriate amount of RIF, ISH and the matrix of excipients in order to simulate the environment of PZA/RIF/ISH association. The NIR-PLS models were optimized using a novel smart approach based on radial optimization (full range, 3 L V and MSC-D' and SNV-D' as pre-treatment, for raw material and FDC tablets, respectively). During the validation step, both methods showed no bias or systematic errors and yielded satisfactory recoveries (102.5 ± 3.1 % for drug substance and 98.7 ± 1.5 % for FDC tablets). When commercial drug substance was tested, NIR-PLS was able to predict the content of Form α (0.98 ± 0.01 w/w). The model for FDC tablets allowed estimating polymorphic purity in intact (0.984 ± 0.003 w/w), sectioned (0.986 ± 0.002 w/w), and powered (0.985 ± 0.004 w/w) tablets, showing the methodology could be applied to a different stage of the process (i.e premixed-powders or granulates). The suitability of the method was also verified when Form α was satisfactorily analysed in FDC fortified with Form δ and Form γ to reach 0.78, 0.88 and 0.98 w/w, Form α. This strategy results in an excellent alternative to ensure the polymorphic purity of PZA throughout the overall pharmaceutical manufacturing process.
Collapse
|
23
|
Kestur U, Desai D, Zong Z, Abraham A, Fiske J. Effect of coating excipients on chemical stability of active coated tablets. Pharm Dev Technol 2020; 26:41-47. [PMID: 33021427 DOI: 10.1080/10837450.2020.1832520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The objective of this study was to understand the impact of coating excipients on the chemical stability of active pan coated peliglitazar, which was prone to acid as well as base-catalyzed degradation. Four different coating formulations containing either polyvinyl alcohol (PVA) or hydroxypropyl methylcellulose (HPMC) as a coating polymer and triacetin (glycerol triacetate) or polyethylene glycol (PEG) as a plasticizer/detackifier were used for coating of peliglitazar in a perforated pan coater. Tablets of one-milligram strength were manufactured by suspending the drug in the coating suspension and spray coating onto inert core tablets. The active coated tablets were placed on stability (40 °C/75% RH) in high-density polyethylene (HDPE) bottles in closed condition with desiccants or in open condition. Tablet samples were withdrawn and analyzed for degradants using a stability-indicating HPLC method. The overall stability for the film-forming polymer-plasticizer/detackifier combination showed the rank order: HPMC-triacetin > PVA-triacetin > HPMC-PEG > PVA-PEG. Higher stability of triacetin systems over PEG systems was attributed to lower solubility of peliglitazar in triacetin coating systems. For the same plasticizer/detackifier, higher stability of HPMC over PVA-based formulations was attributed to lower solubility and mobility of peliglitazar in HPMC compared with the PVA-based coating.
Collapse
Affiliation(s)
- Umesh Kestur
- Drug Product Science and Technology, Bristol-Myers Squibb Company, New Brunswick, NJ, USA
| | - Divyakant Desai
- Drug Product Science and Technology, Bristol-Myers Squibb Company, New Brunswick, NJ, USA
| | - Zhixin Zong
- Drug Product Science and Technology, Bristol-Myers Squibb Company, New Brunswick, NJ, USA
| | - Anuji Abraham
- Drug Product Science and Technology, Bristol-Myers Squibb Company, New Brunswick, NJ, USA
| | - John Fiske
- Drug Product Science and Technology, Bristol-Myers Squibb Company, New Brunswick, NJ, USA
| |
Collapse
|
24
|
Emwas AH, Szczepski K, Poulson BG, Chandra K, McKay RT, Dhahri M, Alahmari F, Jaremko L, Lachowicz JI, Jaremko M. NMR as a "Gold Standard" Method in Drug Design and Discovery. Molecules 2020; 25:E4597. [PMID: 33050240 PMCID: PMC7594251 DOI: 10.3390/molecules25204597] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/11/2022] Open
Abstract
Studying disease models at the molecular level is vital for drug development in order to improve treatment and prevent a wide range of human pathologies. Microbial infections are still a major challenge because pathogens rapidly and continually evolve developing drug resistance. Cancer cells also change genetically, and current therapeutic techniques may be (or may become) ineffective in many cases. The pathology of many neurological diseases remains an enigma, and the exact etiology and underlying mechanisms are still largely unknown. Viral infections spread and develop much more quickly than does the corresponding research needed to prevent and combat these infections; the present and most relevant outbreak of SARS-CoV-2, which originated in Wuhan, China, illustrates the critical and immediate need to improve drug design and development techniques. Modern day drug discovery is a time-consuming, expensive process. Each new drug takes in excess of 10 years to develop and costs on average more than a billion US dollars. This demonstrates the need of a complete redesign or novel strategies. Nuclear Magnetic Resonance (NMR) has played a critical role in drug discovery ever since its introduction several decades ago. In just three decades, NMR has become a "gold standard" platform technology in medical and pharmacology studies. In this review, we present the major applications of NMR spectroscopy in medical drug discovery and development. The basic concepts, theories, and applications of the most commonly used NMR techniques are presented. We also summarize the advantages and limitations of the primary NMR methods in drug development.
Collapse
Affiliation(s)
- Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology, Thuwal 23955-6900, Saudi Arabia
| | - Kacper Szczepski
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (K.S.); (B.G.P.); (K.C.); (L.J.)
| | - Benjamin Gabriel Poulson
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (K.S.); (B.G.P.); (K.C.); (L.J.)
| | - Kousik Chandra
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (K.S.); (B.G.P.); (K.C.); (L.J.)
| | - Ryan T. McKay
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2W2, Canada;
| | - Manel Dhahri
- Biology Department, Faculty of Science, Taibah University, Yanbu El-Bahr 46423, Saudi Arabia;
| | - Fatimah Alahmari
- Nanomedicine Department, Institute for Research and Medical, Consultations (IRMC), Imam Abdulrahman Bin Faisal University (IAU), Dammam 31441, Saudi Arabia;
| | - Lukasz Jaremko
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (K.S.); (B.G.P.); (K.C.); (L.J.)
| | - Joanna Izabela Lachowicz
- Department of Medical Sciences and Public Health, Università di Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy
| | - Mariusz Jaremko
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (K.S.); (B.G.P.); (K.C.); (L.J.)
| |
Collapse
|
25
|
Li M, Lu X, Xu W, Troup GM, McNevin MJ, Nie H, Su Y. Quantifying Pharmaceutical Formulations from Proton Detected Solid-State NMR under Ultrafast Magic Angle Spinning. J Pharm Sci 2020; 109:3045-3053. [PMID: 32679211 DOI: 10.1016/j.xphs.2020.06.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/20/2022]
Abstract
Probing form conversions of active pharmaceutical ingredients in solid dosages is critical for understanding the physicochemical stability of drug substances in formulations. The multicomponent and low drug loading nature of drug products often results in challenges to quantify the phase stability, at a low detection limit and with the chemical resolution that differentiate drug molecules and excipients, for routine laboratory techniques. Recent advancement of ultrafast magic angle spinning (UF-MAS) enables proton-detected solid-state nuclear magnetic resonance (ssNMR) techniques to characterize pharmaceutical materials with enhanced resolution and sensitivity. This study demonstrates one of the first documented cases implementing 60 kHz UF-MAS techniques to quantify the minor content of pioglitazone free base (PIO-FB) in a binary system with its hydrochloride salt (PIO-HCl) and a multicomponent formulation with typical excipients. One-dimensional 1H methods can unambiguously differentiate the two forms and exhibit a limit of detection at 1.77% (w/w). Moreover, we extended it to a two-dimensional 1H-1H correlation for minimizing peak overlap and successfully quantifying approximately 2.0% (w/w) PIO-FB in a multicomponent formulation. These results have demonstrated that 1H ssNMR as a novel method to quantify solid dosages at a higher resolution and faster acquisition than conventional 13C techniques.
Collapse
Affiliation(s)
- Mingyue Li
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Xingyu Lu
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Wei Xu
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Gregory M Troup
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Michael J McNevin
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Haichen Nie
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Yongchao Su
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA; Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, IN 47907, USA; Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
26
|
Das PP, Guzzinati G, Coll C, Gomez Perez A, Nicolopoulos S, Estrade S, Peiro F, Verbeeck J, Zompra AA, Galanis AS. Reliable Characterization of Organic & Pharmaceutical Compounds with High Resolution Monochromated EEL Spectroscopy. Polymers (Basel) 2020; 12:polym12071434. [PMID: 32605004 PMCID: PMC7408036 DOI: 10.3390/polym12071434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 11/16/2022] Open
Abstract
Organic and biological compounds (especially those related to the pharmaceutical industry) have always been of great interest for researchers due to their importance for the development of new drugs to diagnose, cure, treat or prevent disease. As many new API (active pharmaceutical ingredients) and their polymorphs are in nanocrystalline or in amorphous form blended with amorphous polymeric matrix (known as amorphous solid dispersion—ASD), their structural identification and characterization at nm scale with conventional X-Ray/Raman/IR techniques becomes difficult. During any API synthesis/production or in the formulated drug product, impurities must be identified and characterized. Electron energy loss spectroscopy (EELS) at high energy resolution by transmission electron microscope (TEM) is expected to be a promising technique to screen and identify the different (organic) compounds used in a typical pharmaceutical or biological system and to detect any impurities present, if any, during the synthesis or formulation process. In this work, we propose the use of monochromated TEM-EELS, to analyze selected peptides and organic compounds and their polymorphs. In order to validate EELS for fingerprinting (in low loss/optical region) and by further correlation with advanced DFT, simulations were utilized.
Collapse
Affiliation(s)
- Partha Pratim Das
- NanoMegas SPRL, Boulevard Edmond Machtens 79, B1080 Brussels, Belgium; (A.G.P.); (A.S.G.)
- Electron Crystallography Solutions SL, Calle Orense 8, 28020 Madrid, Spain
- Correspondence: (P.P.D.); (S.N.)
| | - Giulio Guzzinati
- EMAT, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium; (G.G.); (J.V.)
| | - Catalina Coll
- LENS-MIND, Department of Electronics and Biomedical Engineering, Universitat de Barcelona, 08028 Barcelona, Spain; (C.C.); (S.E.); (F.P.)
- Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Alejandro Gomez Perez
- NanoMegas SPRL, Boulevard Edmond Machtens 79, B1080 Brussels, Belgium; (A.G.P.); (A.S.G.)
| | - Stavros Nicolopoulos
- NanoMegas SPRL, Boulevard Edmond Machtens 79, B1080 Brussels, Belgium; (A.G.P.); (A.S.G.)
- Correspondence: (P.P.D.); (S.N.)
| | - Sonia Estrade
- LENS-MIND, Department of Electronics and Biomedical Engineering, Universitat de Barcelona, 08028 Barcelona, Spain; (C.C.); (S.E.); (F.P.)
- Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Francesca Peiro
- LENS-MIND, Department of Electronics and Biomedical Engineering, Universitat de Barcelona, 08028 Barcelona, Spain; (C.C.); (S.E.); (F.P.)
- Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Johan Verbeeck
- EMAT, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium; (G.G.); (J.V.)
| | | | - Athanassios S. Galanis
- NanoMegas SPRL, Boulevard Edmond Machtens 79, B1080 Brussels, Belgium; (A.G.P.); (A.S.G.)
| |
Collapse
|
27
|
Terban MW, Russo L, Pham TN, Barich DH, Sun YT, Burke MD, Brum J, Billinge SJL. Local Structural Effects Due to Micronization and Amorphization on an HIV Treatment Active Pharmaceutical Ingredient. Mol Pharm 2020; 17:2370-2389. [PMID: 32293895 DOI: 10.1021/acs.molpharmaceut.0c00122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Processing procedures for inducing domain size reduction and/or amorphous phase generation can be crucial for enhancing the bioavailability of active pharmaceutical ingredients (APIs). It is important to quantify these reduced coherence phases and to detect and characterize associated structural changes, to ensure that no deleterious effects on safety, function, or stability occur. Here, X-ray powder diffraction (XRPD), total scattering pair distribution function (TSPDF) analysis, and solid-state nuclear magnetic resonance spectroscopy (SSNMR) have been performed on samples of GSK2838232B, an investigational drug for the treatment of human immunodeficiency virus (HIV). Preparations were obtained through different mechanical treatments resulting in varying extents of domain size reduction and amorphous phase generation. Completely amorphous formulations could be prepared by milling and microfluidic injection processes. Microfluidic injection was shown to result in a different local structure due to dispersion with dichloromethane (DCM). Implications of combined TSPDF and SSNMR studies to characterize molecular compounds are also discussed, in particular, the possibility to obtain a thorough structural understanding of disordered samples from different processes.
Collapse
Affiliation(s)
- Maxwell W Terban
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, New York 10027, United States
| | - Luca Russo
- GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Tran N Pham
- GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Dewey H Barich
- GlaxoSmithKline R&D, Collegeville, Pennsylvania 19426, United States
| | - Yan T Sun
- GlaxoSmithKline R&D, Collegeville, Pennsylvania 19426, United States
| | - Matthew D Burke
- GlaxoSmithKline R&D, King of Prussia, Pennsylvania 19406, United States
| | - Jeffrey Brum
- GlaxoSmithKline R&D, Collegeville, Pennsylvania 19426, United States
| | - Simon J L Billinge
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, New York 10027, United States.,Condensed Matter Physics and Materials Science Department, Brookhaven National Laboratory, Upton, New York 11973, United States
| |
Collapse
|
28
|
Probing the Molecular-Level Interactions in an Active Pharmaceutical Ingredient (API) - Polymer Dispersion and the Resulting Impact on Drug Product Formulation. Pharm Res 2020; 37:94. [PMID: 32405662 DOI: 10.1007/s11095-020-02813-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/02/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE An investigation of underlying mechanisms of API-polymer interaction patterns has the potential to provide valuable insights for selecting appropriate formulations with superior physical stability and processability. MATERIALS AND METHODS In this study, copovidone was used as a polymeric carrier for several model compounds including clotrimazole, nifedipine, and posaconazole. The varied chemical structures conferred the ability for the model compounds to form distinct interactions with copovidone. Rheology and nuclear magnetic resonance (NMR) were combined to investigate the molecular pattern and relative strength of active pharmaceutical ingredient (API)-polymer interactions. In addition, the impact of the interactions on formulation processability via hot melt extrusion (HME) and physical stability were evaluated. RESULTS The rheological response of an API-polymer system was found to be highly sensitive to API-polymer interaction, depending both on API chemistry and API-polymer miscibility. In the systems studied, dispersed API induced a stronger plasticizer effect on the polymer matrix compared to crystalline/aggregated API. Correspondingly, the processing torque via HME showed a proportional relationship with the maximum complex viscosity of the API-polymer system. In order to quantitatively evaluate the relative strength of the API-polymer interaction, homogeneously dispersed API-polymer amorphous samples were prepared by HME at an elevated temperature. DSC, XRD, and rheology were employed to confirm the amorphous integrity and homogeneity of the resultant extrudates. Subsequently, the homogeneously dispersed API-polymer amorphous dispersions were interrogated by rheology and NMR to provide a qualitative and quantitative assessment of the nature of the API-polymer interaction, both macroscopically and microscopically. Rheological master curves of frequency sweeps of the extrudates exhibited a strong dependence on the API chemistry and revealed a rank ordering of the relative strength of API-copovidone interactions, in the order of posaconazole > nifedipine > clotrimazole. NMR data provided the means to precisely map the API-polymer interaction pattern and identify the specific sites of interaction from a molecular perspective. Finally, the impact of API-polymer interactions on the physical stability of the resultant extrudates was studied. CONCLUSION Qualitative and quantitative evaluation of the relative strength of the API-polymer interaction was successfully accomplished by utilizing combined rheology and NMR. Graphical Abstract.
Collapse
|
29
|
Lu X, Li M, Huang C, Lowinger MB, Xu W, Yu L, Byrn SR, Templeton AC, Su Y. Atomic-Level Drug Substance and Polymer Interaction in Posaconazole Amorphous Solid Dispersion from Solid-State NMR. Mol Pharm 2020; 17:2585-2598. [DOI: 10.1021/acs.molpharmaceut.0c00268] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Xingyu Lu
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Mingyue Li
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Chengbin Huang
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Michael B. Lowinger
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Wei Xu
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Lian Yu
- School of Pharmacy and Department of Chemistry, University of Wisconsin−Madison, Madison, Wisconsin 53705, United States
| | - Stephen R. Byrn
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, Indiana 47907, United States
| | - Allen C. Templeton
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Yongchao Su
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, Indiana 47907, United States
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
30
|
Su M, Xia Y, Shen Y, Heng W, Wei Y, Zhang L, Gao Y, Zhang J, Qian S. A novel drug–drug coamorphous system without molecular interactions: improve the physicochemical properties of tadalafil and repaglinide. RSC Adv 2020; 10:565-583. [PMID: 35492562 PMCID: PMC9048229 DOI: 10.1039/c9ra07149k] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/17/2019] [Indexed: 01/24/2023] Open
Abstract
Tadalafil and repaglinide, categorized as BCS class II drugs, have low oral bioavailabilities due to their poorly aqueous solubilities and dissolutions. The aim of this study was to enhance the dissolution of tadalafil and repaglinide by co-amorphization technology and evaluate the storage and compression stability of such coamorphous system. Based on Flory–Huggins interaction parameter (χ ≤ 0) and Hansen solubility parameter (δt ≤ 7 MPa0.5) calculations, tadalafil and repaglinide was predicted to be well miscible with each other. Coamorphous tadalafil–repaglinide (molar ratio, 1 : 1) was prepared by solvent-evaporation method and characterized with respect to its thermal properties, possible molecular interactions. A single Tg (73.1 °C) observed in DSC and disappearance of crystallinity in PXRD indicated the formation of coamorphous system. Principal component analysis of FTIR in combination with Raman spectroscopy and Ss 13C NMR suggested the absence of intermolecular interactions in coamorphous tadalafil–repaglinide. In comparison to pure crystalline forms and their physical mixtures, both drugs in coamorphous system exhibited significant increases in intrinsic dissolution rate (1.5–3-fold) and could maintain supersaturated level for at least 4 hours in non-sink dissolution. In addition, the coamorphous tadalafil–repaglinide showed improved stability compared to the pure amorphous forms under long-term stability and accelerated storage conditions as well as under high compressing pressure. In conclusion, this study showed that co-amorphization technique is a promising approach for improving the dissolution rate of poorly water-soluble drugs and for stabilizing amorphous drugs. The coamorphous tadalafil–repaglinide (molar ratio, 1 : 1) prepared by solvent-evaporation method significantly improve the physicochemical properties of tadalafil and repaglinide.![]()
Collapse
Affiliation(s)
- Meiling Su
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- P. R. China
| | - Yanming Xia
- School of Traditional Chinese Pharmacy
- China Pharmaceutical University
- Nanjing
- P. R. China
| | - Yajing Shen
- School of Traditional Chinese Pharmacy
- China Pharmaceutical University
- Nanjing
- P. R. China
| | - Weili Heng
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- P. R. China
| | - Yuanfeng Wei
- School of Traditional Chinese Pharmacy
- China Pharmaceutical University
- Nanjing
- P. R. China
| | - Linghe Zhang
- Department of Chemistry
- Smith College
- Northampton
- USA
| | - Yuan Gao
- School of Traditional Chinese Pharmacy
- China Pharmaceutical University
- Nanjing
- P. R. China
| | - Jianjun Zhang
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- P. R. China
| | - Shuai Qian
- School of Traditional Chinese Pharmacy
- China Pharmaceutical University
- Nanjing
- P. R. China
| |
Collapse
|
31
|
Eedara BB, Tucker IG, Zujovic ZD, Rades T, Price JR, Das SC. Crystalline adduct of moxifloxacin with trans-cinnamic acid to reduce the aqueous solubility and dissolution rate for improved residence time in the lungs. Eur J Pharm Sci 2019; 136:104961. [PMID: 31220546 DOI: 10.1016/j.ejps.2019.104961] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/31/2019] [Accepted: 06/14/2019] [Indexed: 10/26/2022]
Abstract
A crystalline adduct of the anti-tubercular drug, moxifloxacin and trans-cinnamic acid (1:1 molar ratio (MCA1:1)) was prepared to prolong the residence time of the drug in the lungs by reducing its solubility and dissolution rate. Whether the adduct is a salt or cocrystal has not been unequivocally determined. Equilibrium solubility and intrinsic dissolution rate measurements for the adduct (MCA1:1) in phosphate buffered saline (PBS, pH 7.4) revealed a significant decrease in the solubility of moxifloxacin (from 17.68 ± 0.85 mg mL-1 to 6.10 ± 0.05 mg mL-1) and intrinsic dissolution rate (from 0.47 ± 0.04 mg cm-2 min-1 to 0.14 ± 0.03 mg cm-2 min-1) compared to the supplied moxifloxacin. The aerosolization behaviour of the adduct from an inhaler device, Aerolizer®, using a Next Generation Impactor showed a fine particle fraction of 30.4 ± 1.2%. The dissolution behaviour of the fine particle dose of respirable particles collected was assessed in a small volume of stationary mucus fluid using a custom-made dissolution apparatus. The respirable adduct particles showed a lower dissolution (microscopic observation) and permeation compared to the supplied moxifloxacin. The crystalline adduct MCA1:1 has a lower solubility and dissolution rate than moxifloxacin and could improve the local residence time and therapeutic action of moxifloxacin in the lungs.
Collapse
Affiliation(s)
| | - Ian G Tucker
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Zoran D Zujovic
- Department of Chemistry, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Thomas Rades
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jason R Price
- ANSTO - Australian Synchrotron, 800 Blackburn Rd, Clayton, 3168 Victoria, Australia
| | - Shyamal C Das
- School of Pharmacy, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
32
|
Hirsh DA, Wijesekara AV, Carnahan SL, Hung I, Lubach JW, Nagapudi K, Rossini AJ. Rapid Characterization of Formulated Pharmaceuticals Using Fast MAS 1H Solid-State NMR Spectroscopy. Mol Pharm 2019; 16:3121-3132. [DOI: 10.1021/acs.molpharmaceut.9b00343] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- David A. Hirsh
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, United States
| | | | - Scott L. Carnahan
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, United States
| | - Ivan Hung
- Center of Interdisciplinary Magnetic Resonance, National High Magnetic Field Laboratory, 1800 East Paul Dirac Drive, Tallahassee, Florida 32310, United States
| | - Joseph W. Lubach
- Genentech Inc., South San Francisco, California 94080, United States
| | - Karthik Nagapudi
- Genentech Inc., South San Francisco, California 94080, United States
| | - Aaron J. Rossini
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|
33
|
Lu X, Huang C, Lowinger MB, Yang F, Xu W, Brown CD, Hesk D, Koynov A, Schenck L, Su Y. Molecular Interactions in Posaconazole Amorphous Solid Dispersions from Two-Dimensional Solid-State NMR Spectroscopy. Mol Pharm 2019; 16:2579-2589. [DOI: 10.1021/acs.molpharmaceut.9b00174] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Xingyu Lu
- Merck Research Laboratories (MRLs), Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Chengbin Huang
- Merck Research Laboratories (MRLs), Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
- School of Pharmacy, University of Wisconsin−Madison, Madison, Wisconsin 53705, United States
| | - Michael B. Lowinger
- Merck Research Laboratories (MRLs), Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Fengyuan Yang
- Merck Research Laboratories (MRLs), Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
- Ashland Inc., Wilmington, Delaware 19808, United States
| | - Wei Xu
- Merck Research Laboratories (MRLs), Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Chad D. Brown
- Merck Research Laboratories (MRLs), Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - David Hesk
- Merck Research Laboratories (MRLs), Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Athanas Koynov
- Merck Research Laboratories (MRLs), Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Luke Schenck
- Merck Research Laboratories (MRLs), Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Yongchao Su
- Merck Research Laboratories (MRLs), Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| |
Collapse
|
34
|
Smith CJ, Dinh J, Schmitt PD, Stroud PA, Hinds J, Johnson MJ, Simpson GJ. Calibration-Free Second Harmonic Generation (SHG) Image Analysis for Quantification of Trace Crystallinity Within Final Dosage Forms of Amorphous Solid Dispersions. APPLIED SPECTROSCOPY 2018; 72:1594-1605. [PMID: 29896972 DOI: 10.1177/0003702818786506] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
A statistical model enables auto-calibration of second harmonic generation (SHG) images for quantifying trace crystallinity within amorphous solid dispersions (ASDs) over a wide dynamic range of crystallinity. In this paper, we demonstrate particle-counting approaches for quantifying trace crystallinity, combined with analytical expressions correcting for particle overlap bias in higher crystallinity regimes to extend the continuous dynamic range of standard particle-counting algorithms through to the signal averaging regime. The reliability of the values recovered by these expressions was demonstrated with simulated data as well as experimental data obtained for an amorphous solid dispersion formulation containing evacetrapib, an Eli Lilly and Company compound. Since particle counting independently recovers the crystalline volume and the SHG intensity, the average SHG intensity per unit volume can be used as an internal calibrant for quantifying crystallinity at higher volume fractions, for which particle counting is no longer applicable.
Collapse
Affiliation(s)
- Casey J Smith
- 1 Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Janny Dinh
- 1 Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Paul D Schmitt
- 3 Department of Chemistry, Wabash College, Crawfordsville, IN, USA
| | | | | | | | - Garth J Simpson
- 1 Department of Chemistry, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
35
|
Lou X, Shen M, Li C, Chen Q, Hu B. Reduction of the 13C cross-polarization experimental time for pharmaceutical samples with long T 1 by ball milling in solid-state NMR. SOLID STATE NUCLEAR MAGNETIC RESONANCE 2018; 94:20-25. [PMID: 30125796 DOI: 10.1016/j.ssnmr.2018.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 08/02/2018] [Accepted: 08/02/2018] [Indexed: 06/08/2023]
Abstract
Many pharmaceutical samples have notably long 1H T1 (proton spin-lattice relaxation time), leading to lengthy experiments lasting several days in solid-state NMR studies. In this work, we propose the use of ball milling on the pharmaceutical samples to reduce the 1H T1, which also leads to enhanced sensitivity in {1H}-13C Cross-Polarization (CP) experiments due to reduced particle sizes and increased surface areas of the samples. Experimentally, we determined that depending on the substrates and milling time, the signal-to-noise ratio (S/N) of a 1D 13C CP spectrum can be increased by a factor of 3-6, which means that the experimental time can be shortened by a factor of 9-36. Furthermore, the application of simple ball-milling within a short time avoids the amorphization of the studied samples such that no signal due to amorphous state is observed in the 13C CP spectrum. This simple ball milling method used for sensitivity enhancement can be further applied in the SS-NMR studies of pharmaceutical samples.
Collapse
Affiliation(s)
- Xiaobing Lou
- State Key Laboratory of Precision Spectroscopy & Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Materials Science, East China Normal University, Shanghai, 200062, China
| | - Ming Shen
- State Key Laboratory of Precision Spectroscopy & Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Materials Science, East China Normal University, Shanghai, 200062, China
| | - Chao Li
- State Key Laboratory of Precision Spectroscopy & Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Materials Science, East China Normal University, Shanghai, 200062, China
| | - Qun Chen
- State Key Laboratory of Precision Spectroscopy & Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Materials Science, East China Normal University, Shanghai, 200062, China
| | - Bingwen Hu
- State Key Laboratory of Precision Spectroscopy & Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Materials Science, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
36
|
Kumar M, Bhatia R, Rawal RK. Applications of various analytical techniques in quality control of pharmaceutical excipients. J Pharm Biomed Anal 2018; 157:122-136. [DOI: 10.1016/j.jpba.2018.05.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 11/29/2022]
|
37
|
Zhao L, Pinon AC, Emsley L, Rossini AJ. DNP-enhanced solid-state NMR spectroscopy of active pharmaceutical ingredients. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2018; 56:583-609. [PMID: 29193278 DOI: 10.1002/mrc.4688] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 11/15/2017] [Accepted: 11/19/2017] [Indexed: 06/07/2023]
Abstract
Solid-state NMR spectroscopy has become a valuable tool for the characterization of both pure and formulated active pharmaceutical ingredients (APIs). However, NMR generally suffers from poor sensitivity that often restricts NMR experiments to nuclei with favorable properties, concentrated samples, and acquisition of one-dimensional (1D) NMR spectra. Here, we review how dynamic nuclear polarization (DNP) can be applied to routinely enhance the sensitivity of solid-state NMR experiments by one to two orders of magnitude for both pure and formulated APIs. Sample preparation protocols for relayed DNP experiments and experiments on directly doped APIs are detailed. Numerical spin diffusion models illustrate the dependence of relayed DNP enhancements on the relaxation properties and particle size of the solids and can be used for particle size determination when the other factors are known. We then describe the advanced solid-state NMR experiments that have been enabled by DNP and how they provide unique insight into the molecular and macroscopic structure of APIs. For example, with large sensitivity gains provided by DNP, natural isotopic abundance, 13 C-13 C double-quantum single-quantum homonuclear correlation NMR spectra of pure APIs can be routinely acquired. DNP also enables solid-state NMR experiments with unreceptive quadrupolar nuclei such as 2 H, 14 N, and 35 Cl that are commonly found in APIs. Applications of DNP-enhanced solid-state NMR spectroscopy for the molecular level characterization of low API load formulations such as commercial tablets and amorphous solid dispersions are described. Future perspectives for DNP-enhanced solid-state NMR experiments on APIs are briefly discussed.
Collapse
Affiliation(s)
- Li Zhao
- Department of Chemistry, Iowa State University, Ames, IA, USA
- US DOE Ames Laboratory, Ames, IA, USA
| | - Arthur C Pinon
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Lyndon Emsley
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Aaron J Rossini
- Department of Chemistry, Iowa State University, Ames, IA, USA
- US DOE Ames Laboratory, Ames, IA, USA
| |
Collapse
|
38
|
Song Z, Sarkar S, Vogt AD, Danzer GD, Smith CJ, Gualtieri EJ, Simpson GJ. Kinetic Modeling of Accelerated Stability Testing Enabled by Second Harmonic Generation Microscopy. Anal Chem 2018; 90:4406-4413. [DOI: 10.1021/acs.analchem.7b04260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Zhengtian Song
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47906, United States
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Sreya Sarkar
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47906, United States
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Andrew D. Vogt
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Gerald D. Danzer
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Casey J. Smith
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47906, United States
| | - Ellen J. Gualtieri
- Formulatrix, Inc., 10 DeAngelo Drive, Bedford, Massachusetts 01730, United States
| | - Garth J. Simpson
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47906, United States
| |
Collapse
|
39
|
Ueda K, Kanaya H, Higashi K, Yamamoto K, Moribe K. Molecular-level elucidation of saccharin-assisted rapid dissolution and high supersaturation level of drug from Eudragit® E solid dispersion. Int J Pharm 2018; 538:57-64. [DOI: 10.1016/j.ijpharm.2017.12.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 12/18/2017] [Accepted: 12/30/2017] [Indexed: 10/18/2022]
|
40
|
Hanrahan MP, Venkatesh A, Carnahan SL, Calahan JL, Lubach JW, Munson EJ, Rossini AJ. Enhancing the resolution of 1H and 13C solid-state NMR spectra by reduction of anisotropic bulk magnetic susceptibility broadening. Phys Chem Chem Phys 2018; 19:28153-28162. [PMID: 29022618 DOI: 10.1039/c7cp04223j] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We demonstrate that natural isotopic abundance 2D heteronuclear correlation (HETCOR) solid-state NMR spectra can be used to significantly reduce or eliminate the broadening of 1H and 13C solid-state NMR spectra of organic solids due to anisotropic bulk magnetic susceptibility (ABMS). ABMS often manifests in solids with aromatic groups, such as active pharmaceutical ingredients (APIs), and inhomogeneously broadens the NMR peaks of all nuclei in the sample. Inhomogeneous peaks with full widths at half maximum (FWHM) of ∼1 ppm typically result from ABMS broadening and the low spectral resolution impedes the analysis of solid-state NMR spectra. ABMS broadening of solid-state NMR spectra has previously been eliminated using 2D multiple-quantum correlation experiments, or by performing NMR experiments on diluted materials or single crystals. However, these experiments are often infeasible due to their poor sensitivity and/or provide limited gains in resolution. 2D 1H-13C HETCOR experiments have previously been applied to reduce susceptibility broadening in paramagnetic solids and we show that this strategy can significantly reduce ABMS broadening in diamagnetic organic solids. Comparisons of 1D solid-state NMR spectra and 1H and 13C solid-state NMR spectra obtained from 2D 1H-13C HETCOR NMR spectra show that the HETCOR spectrum directly increases resolution by a factor of 1.5 to 8. The direct gain in resolution is determined by the ratio of the inhomogeneous 13C/1H linewidth to the homogeneous 1H linewidth, with the former depending on the magnitude of the ABMS broadening and the strength of the applied field and the latter on the efficiency of homonuclear decoupling. The direct gains in resolution obtained using the 2D HETCOR experiments are better than that obtained by dilution. For solids with long proton longitudinal relaxation times, dynamic nuclear polarization (DNP) was applied to enhance sensitivity and enable the acquisition of 2D 1H-13C HETCOR NMR spectra. 2D 1H-13C HETCOR experiments were applied to resolve and partially assign the NMR signals of the form I and form II polymorphs of aspirin in a sample containing both forms. These findings have important implications for ultra-high field NMR experiments, optimization of decoupling schemes and assessment of the fundamental limits on the resolution of solid-state NMR spectra.
Collapse
|
41
|
Crini G, Fourmentin S, Fenyvesi É, Torri G, Fourmentin M, Morin-Crini N. Fundamentals and Applications of Cyclodextrins. ENVIRONMENTAL CHEMISTRY FOR A SUSTAINABLE WORLD 2018. [DOI: 10.1007/978-3-319-76159-6_1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
42
|
Gandhi AV, Thipsay P, Kirthivasan B, Squillante E. Adsorption onto Mesoporous Silica Using Supercritical Fluid Technology Improves Dissolution Rate of Carbamazepine-a Poorly Soluble Compound. AAPS PharmSciTech 2017; 18:3140-3150. [PMID: 28534299 DOI: 10.1208/s12249-017-0784-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/13/2017] [Indexed: 11/30/2022] Open
Abstract
The purpose of this research was to design and characterize an immediate-release formulation of carbamazepine (CBZ), a poorly soluble anti-epileptic drug, using a porous silica carrier. Carbon dioxide in its supercritical state (2000 psi, 30-35°C) was used as an anti-solvent to precipitate CBZ onto two particle size variants of silica. Adsorption isotherms were used as a pre-formulation strategy to select optimum ratios of silica and CBZ. The obtained drug-silica formulations were characterized by dissolution studies, differential scanning calorimetry (DSC), powder X-ray diffraction (PXRD), and scanning electron microscopy (SEM). This formulation strategy resulted in a 2.4-fold improvement in dissolution rate when compared to pure drug after 30 min of dissolution testing. PXRD and DSC confirmed the amorphous nature of CBZ in the formulations as well as the differences in polymorphic forms of commercial and supercritical fluid-processed CBZ. Additionally, solid-state NMR spectroscopy showed that the spin-lattice relaxation time for bulk drug (without silica) was ∼7.5 times greater than that for silica-confined CBZ, implying that when CBZ was adsorbed onto mesoporous silica, it is structurally disordered and had higher structural mobility, a characteristic of amorphous solids. The mesoporous silica matrix prevented CBZ crystal growth by imposing spatial constraint on CBZ nuclei and hence resulted in faster dissolution compared to bulk solid drug. Adsorption onto mesoporous silica using supercritical fluid technology may be used as a novel formulation strategy for amorphization of poorly soluble compounds, in turn improving their dissolution rate.
Collapse
|
43
|
Assessing the Detection Limit of a Minority Solid-State Form of a Pharmaceutical by 1H Double-Quantum Magic-Angle Spinning Nuclear Magnetic Resonance Spectroscopy. J Pharm Sci 2017; 106:3372-3377. [DOI: 10.1016/j.xphs.2017.07.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/05/2017] [Accepted: 07/18/2017] [Indexed: 01/20/2023]
|
44
|
Insights into Nano- and Micron-Scale Phase Separation in Amorphous Solid Dispersions Using Fluorescence-Based Techniques in Combination with Solid State Nuclear Magnetic Resonance Spectroscopy. Pharm Res 2017; 34:1364-1377. [PMID: 28455777 DOI: 10.1007/s11095-017-2145-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/15/2017] [Indexed: 11/26/2022]
Abstract
PURPOSE Miscibility between the drug and the polymer in an amorphous solid dispersion (ASD) is considered to be one of the most important factors impacting the solid state stability and dissolution performance of the active pharmaceutical ingredient (API). The research described herein utilizes emerging fluorescence-based methodologies to probe (im)miscibility of itraconazole (ITZ)-hydroxypropyl methylcellulose (HPMC) ASDs. METHODS The ASDs were prepared by solvent evaporation with varying evaporation rates and were characterized by steady-state fluorescence spectroscopy, confocal imaging, differential scanning calorimetry (DSC), and solid state nuclear magnetic resonance (ssNMR) spectroscopy. RESULTS The size of the phase separated domains for the ITZ-HPMC ASDs was affected by the solvent evaporation rate. Smaller domains (<10 nm) were observed in spray-dried ASDs, whereas larger domains (>30 nm) were found in ASDs prepared using slower evaporation rates. Confocal imaging provided visual confirmation of phase separation along with chemical specificity, achieved by selectively staining drug-rich and polymer-rich phases. ssNMR confirmed the results of fluorescence-based techniques and provided information on the size of phase separated domains. CONCLUSIONS The fluorescence-based methodologies proved to be sensitive and rapid in detecting phase separation, even at the nanoscale, in the ITZ-HPMC ASDs. Fluorescence-based methods thus show promise for miscibility evaluation of spray-dried ASDs.
Collapse
|
45
|
Ho TM, Truong T, Bhandari BR. Methods to characterize the structure of food powders – a review. Biosci Biotechnol Biochem 2017; 81:651-671. [PMID: 28077060 DOI: 10.1080/09168451.2016.1274643] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Abstract
Food powders can exist in amorphous, crystalline or mixed structure depending on the order of molecular arrangement in the powder particle matrices. In food production, the structure of powders has a greatly effect on their stability, functionality, and applicability. The undesirable structure of powders can be accidentally formed during production. Therefore, characterization of powder structure as well as quantification of amorphous–crystalline proportions presenting in the powders are essential to control the quality of products during storage and further processing. For these purposes, many analytical techniques with large differences in the degree of selectivity and sensitivity have been developed. In this review, differences in the structure of food powders are described with a focus being placed on applications of amorphous powders. Essentially, applicability of common analytical techniques including X-ray, microscopic, vapor adsorption, thermal, and spectroscopic approaches for quantitative and qualitative structural characterization of food powders is also discussed.
Collapse
Affiliation(s)
- Thao M Ho
- School of Agriculture and Food Sciences, The University of Queensland, Brisbane, Australia
| | - Tuyen Truong
- School of Agriculture and Food Sciences, The University of Queensland, Brisbane, Australia
| | - Bhesh R Bhandari
- School of Agriculture and Food Sciences, The University of Queensland, Brisbane, Australia
| |
Collapse
|
46
|
Marchetti A, Chen J, Pang Z, Li S, Ling D, Deng F, Kong X. Understanding Surface and Interfacial Chemistry in Functional Nanomaterials via Solid-State NMR. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1605895. [PMID: 28247966 DOI: 10.1002/adma.201605895] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/26/2016] [Indexed: 05/24/2023]
Abstract
Surface and interfacial chemistry is of fundamental importance in functional nanomaterials applied in catalysis, energy storage and conversion, medicine, and other nanotechnologies. It has been a perpetual challenge for the scientific community to get an accurate and comprehensive picture of the structures, dynamics, and interactions at interfaces. Here, some recent examples in the major disciplines of nanomaterials are selected (e.g., nanoporous materials, battery materials, nanocrystals and quantum dots, supramolecular assemblies, drug-delivery systems, ionomers, and graphite oxides) and it is shown how interfacial chemistry can be addressed through the perspective of solid-state NMR characterization techniques.
Collapse
Affiliation(s)
- Alessandro Marchetti
- Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Juner Chen
- Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Zhenfeng Pang
- Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Shenhui Li
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China
| | - Daishun Ling
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, P. R. China
| | - Feng Deng
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China
| | - Xueqian Kong
- Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou, 310027, P. R. China
| |
Collapse
|
47
|
Ohyagi N, Ueda K, Higashi K, Yamamoto K, Kawakami K, Moribe K. Synergetic Role of Hypromellose and Methacrylic Acid Copolymer in the Dissolution Improvement of Amorphous Solid Dispersions. J Pharm Sci 2017; 106:1042-1050. [DOI: 10.1016/j.xphs.2016.12.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 10/10/2016] [Accepted: 12/07/2016] [Indexed: 11/16/2022]
|
48
|
Schmitt PD. Recent Advances in Nonlinear Optical Analyses of Pharmaceutical Materials in the Solid State. Mol Pharm 2017; 14:555-565. [PMID: 28125239 DOI: 10.1021/acs.molpharmaceut.6b00809] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The past decade has seen an increase in the use of nonlinear optical (NLO) techniques such as second harmonic generation, coherent antistokes Raman scattering, stimulated Raman scattering, and two-photon fluorescence for the solid-state characterization of pharmaceutical materials. These combined techniques offer several advantages (e.g., speed, selectivity, quantitation) of potential interest to the pharmaceutical community, as decreased characterization times in formulation development and testing could help decrease the time required to bring new, higher quality drugs to market. The large body of literature recently published in this field merits a review. Literature will be discussed in order of drug development, starting with applications in initial therapeutic molecule crystallization and polymorphic analysis, followed by final dosage form characterization, and ending with drug product performance testing.
Collapse
Affiliation(s)
- Paul D Schmitt
- Department of Chemistry, Wabash College , Crawfordsville, Indiana 47933, United States
| |
Collapse
|
49
|
Nie H, Su Y, Zhang M, Song Y, Leone A, Taylor LS, Marsac PJ, Li T, Byrn SR. Solid-State Spectroscopic Investigation of Molecular Interactions between Clofazimine and Hypromellose Phthalate in Amorphous Solid Dispersions. Mol Pharm 2016; 13:3964-3975. [PMID: 27653759 DOI: 10.1021/acs.molpharmaceut.6b00740] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Haichen Nie
- Department
of Industrial and Physical Pharmacy, Purdue University, 575 Stadium
Mall Drive, West Lafayette, Indiana 47907, United States
- Formulation
Sciences, Teva Pharmaceuticals, 145 Brandywine Parkway, West Chester, Pennsylvania 19380, United States
| | - Yongchao Su
- Merck Research Laboratories, 770 Sumneytown Pike, West
Point, Pennsylvania 19486, United States
| | - Mingtao Zhang
- Department
of Industrial and Physical Pharmacy, Purdue University, 575 Stadium
Mall Drive, West Lafayette, Indiana 47907, United States
| | - Yang Song
- Department
of Industrial and Physical Pharmacy, Purdue University, 575 Stadium
Mall Drive, West Lafayette, Indiana 47907, United States
- Global
DMPK, Takeda Pharmaceutical Inc., 10410 Science Center Drive, San Diego, California 92121, United States
| | - Anthony Leone
- Merck Research Laboratories, 770 Sumneytown Pike, West
Point, Pennsylvania 19486, United States
| | - Lynne S. Taylor
- Department
of Industrial and Physical Pharmacy, Purdue University, 575 Stadium
Mall Drive, West Lafayette, Indiana 47907, United States
| | - Patrick J. Marsac
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536, United States
| | - Tonglei Li
- Department
of Industrial and Physical Pharmacy, Purdue University, 575 Stadium
Mall Drive, West Lafayette, Indiana 47907, United States
| | - Stephen R. Byrn
- Department
of Industrial and Physical Pharmacy, Purdue University, 575 Stadium
Mall Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
50
|
Yang F, Su Y, Zhang J, DiNunzio J, Leone A, Huang C, Brown CD. Rheology Guided Rational Selection of Processing Temperature To Prepare Copovidone–Nifedipine Amorphous Solid Dispersions via Hot Melt Extrusion (HME). Mol Pharm 2016; 13:3494-3505. [PMID: 27602878 DOI: 10.1021/acs.molpharmaceut.6b00516] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Fengyuan Yang
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Yongchao Su
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jingtao Zhang
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - James DiNunzio
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Anthony Leone
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Chengbin Huang
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
- School
of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Chad D. Brown
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|