1
|
Kapourani A, Pantazos I, Valkanioti V, Chatzitheodoridou M, Kalogeri C, Barmpalexis P. Unveiling the impact of preparation methods, matrix/carrier type selection and drug loading on the supersaturation performance of amorphous solid dispersions. Int J Pharm 2025; 671:125242. [PMID: 39842744 DOI: 10.1016/j.ijpharm.2025.125242] [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: 12/09/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
Amorphous solid dispersions (ASDs) are widely recognized for their potential to enhance the solubility of poorly water-soluble drugs, with factors such as molecular mobility, intermolecular interactions, and storage conditions playing critical roles in their performance. However, the influence of preparation methods on their performance remains underexplored, especially regarding their supersaturation . To address this gap, the present study systematically investigates ASDs of ibuprofen (IBU, used as a model drug) prepared using two widely utilized techniques (solvent evaporation, SE, and melt-quench cooling, M-QC). Three different matrices/carriers (Soluplus®, SOL, povidone, PVP, and copovidone, PVPVA) were employed to evaluate the combined influence of preparation method, matrix/carrier type, and drug loading on ASD performance. Supersaturation behavior during dissolution, particularly its dependence on the Sink Index (SI), was a key focus. All ASDs showed successful amorphization, but molecular near-order structures differed based on the preparation method. ATR-FTIR spectroscopy revealed stronger molecular interactions in M-QC ASDs (compared to SE). Dissolution studies under supersaturation conditions (SI = 0.1 and SI = 0.2) highlighted significant performance differences. M-QC ASDs consistently exhibited higher in vitro AUC(0→t) values under non-sink conditions compared to crystalline IBU. Conversely, SE ASDs showed improved supersaturation primarily under low SI conditions, especially with SOL at low drug loadings. The findings underscore the need for a systematic approach in developing ASDs, considering preparation method, matrix/carrier type, drug loading and dissolution study conditions collectively. These factors significantly influence dissolution behavior and supersaturation, emphasizing that they should not be independently studied but evaluated comprehensively to optimize ASD performance.
Collapse
Affiliation(s)
- Afroditi Kapourani
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Ioannis Pantazos
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Vasiliki Valkanioti
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Melina Chatzitheodoridou
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Christina Kalogeri
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Panagiotis Barmpalexis
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; Natural Products Research Centre of Excellence-AUTH (NatPro-AUTH), Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Thessaloniki 57001, Greece.
| |
Collapse
|
2
|
Martynek D, Ridvan L, Sivén M, Šoóš M. Stability and recrystallization of amorphous solid dispersions prepared by hot-melt extrusion and spray drying. Int J Pharm 2025; 672:125331. [PMID: 39933609 DOI: 10.1016/j.ijpharm.2025.125331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
Spray drying and hot-melt extrusion are among the most prevalent preparation techniques used in the pharmaceutical industry to produce amorphous solid dispersions (ASDs). This study advances previous research by integrating sample production, comprehensive analytical characterization, intrinsic dissolution rate measurements, and assessments of the behavior of ASDs under elevated temperature and humidity conditions. The study focuses on indomethacin, a widely used model for poorly soluble drugs, processed with PVP K30 or HPMC E5, both commonly used polymers. The findings demonstrate that hot-melt extruded samples exhibit superior stability against recrystallization, whereas spray dried samples achieve higher intrinsic dissolution rates. Furthermore, PVP K30 significantly outperforms HPMC E5 in the co-processing of indomethacin, enhancing both the intrinsic dissolution rate and the stability.
Collapse
Affiliation(s)
- Dominik Martynek
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic; Zentiva, k.s., Ukabelovny 130, 10237 Prague 10, Czech Republic
| | - Luděk Ridvan
- Zentiva, k.s., Ukabelovny 130, 10237 Prague 10, Czech Republic
| | - Mia Sivén
- Division of Pharmaceutical Chemistry and Technology, University of Helsinki, Viikinkaari 5 E, 00014, Helsinki, Finland; Helsinki Institute of Sustainability Science, HELSUS, Finland
| | - Miroslav Šoóš
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic.
| |
Collapse
|
3
|
Li S, Zhang Z, Gu W, Gallas M, Jones D, Boulet P, Johnson LM, de Margerie V, Andrews GP. Hot Melt Extruded High-Dose Amorphous Solid Dispersions Containing Lumefantrine and Soluplus. Int J Pharm 2024; 665:124676. [PMID: 39255876 DOI: 10.1016/j.ijpharm.2024.124676] [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: 03/20/2024] [Revised: 08/16/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
Over the last 15 years, a small number of paediatric artemisinin-based combination therapy products have been marketed. These included Riamet® and Coartem® dispersible tablets, a combination of artemether and lumefantrine, co-developed by the Medicines for Malaria Venture and Novartis. Disappointingly, patient compliance, requirement for high-fat meal, and sporadic drug dissolution behaviours following administration still result in considerable challenges for these products. The first and foremost barrier that needs addressed for successful delivery of the artemether/lumefantrine combination is the poor solubility of lumefantrine within the gastrointestinal fluids. In this work, amorphous solid dispersions of lumefantrine within Soluplus®-based matrices have been manufactured using hot melt extrusion as a potential formulation strategy to achieve enhanced dissolution and apparent solubility. The drug loading capacity of Soluplus® to accommodate amorphous lumefantrine, whilst ensuring improved in-vitro dissolution performance, was investigated. The extrusion process employed a variety of processing parameters, including various temperature profiles and different production scales. The influence of variation in extrusion conditions upon the physical stability of manufactured amorphous solid dispersions was also examined. This allowed for a greater understanding of the role of extrusion processing conditions on the performance of supersaturated amorphous solid dispersions during dissolution and storage. This may allow for the design and manufacture of drug enabled formulations with lower drug dosing and thus a lower risk of adverse effects.
Collapse
Affiliation(s)
- Shu Li
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, NI UK
| | - Zi'an Zhang
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, NI UK
| | - Wenjie Gu
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, NI UK
| | - Maël Gallas
- Institut Jean Lamour, 2 allée André Guinier 54011 Nancy, France; Rondol Industrie, 2 allée André Guinier 54011 Nancy, France
| | - David Jones
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, NI UK
| | - Pascal Boulet
- Institut Jean Lamour, 2 allée André Guinier 54011 Nancy, France
| | | | | | - Gavin P Andrews
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, NI UK.
| |
Collapse
|
4
|
Kushwah V, Succhielli C, Saraf I, Paudel A. Amorphous Solid Dispersions: Implication of Method of Preparation and Physicochemical Properties of API and Excipients. Pharmaceutics 2024; 16:1035. [PMID: 39204380 PMCID: PMC11360349 DOI: 10.3390/pharmaceutics16081035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/14/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
The present study investigated the effect of different polymers and manufacturing methods (hot melt extrusion, HME, and spray drying, SD) on the solid state, stability and pharmaceutical performance of amorphous solid dispersions. In the present manuscript, a combination of different binary amorphous solid dispersions containing 20% and 30% of drug loadings were prepared using SD and HME. The developed solid-state properties of the dispersions were evaluated using small- and wide-angle X-ray scattering (WAXS) and modulated differential scanning calorimetry (mDSC). The molecular interaction between the active pharmaceutical ingredients (APIs) and polymers were investigated via infrared (IR) and Raman spectroscopy. The in vitro release profile of the solid dispersions was also evaluated to compare the rate and extend of drug dissolution as a function of method of preparation. Thereafter, the effect of accelerated stability conditions on the physicochemical properties of the solid dispersions were also evaluated. The results demonstrated higher stability of Soluplus® (SOL) polymer-based solid dispersions as compared to hydroxypropyl methylcellulose (HPMC)-based solid dispersions. Moreover, the stability of the solid dispersions was found to be higher in the case of API having high glass transition temperature (Tg) and demonstrated higher interaction with the polymeric groups. Interestingly, the stability of the melt-extruded dispersions was found to be slightly higher as compared to the SD formulations. However, the down-processing of melt-extruded strands plays critical role in inducing the API crystal nuclei formation. In summary, the findings strongly indicate that the particulate properties significantly influence the performance of the product.
Collapse
Affiliation(s)
- Varun Kushwah
- Research Center Pharmaceutical Engineering GmbH, 8010 Graz, Austria (I.S.)
| | - Cecilia Succhielli
- Research Center Pharmaceutical Engineering GmbH, 8010 Graz, Austria (I.S.)
| | - Isha Saraf
- Research Center Pharmaceutical Engineering GmbH, 8010 Graz, Austria (I.S.)
| | - Amrit Paudel
- Research Center Pharmaceutical Engineering GmbH, 8010 Graz, Austria (I.S.)
- Institute of Pharmaceutical Science, Department of Pharmaceutical Technology, University of Graz, 8010 Graz, Austria
| |
Collapse
|
5
|
Li J, Wang X, Yu D, Zhoujin Y, Wang K. Molecular complexes of drug combinations: A review of cocrystals, salts, coamorphous systems and amorphous solid dispersions. Int J Pharm 2023; 648:123555. [PMID: 37890646 DOI: 10.1016/j.ijpharm.2023.123555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023]
Abstract
As the advancements in the medical technology and healthcare develop through the years, combinational therapy has evolved to be an important treatment modality in many disease settings, including cancer, cardiovascular disease and infectious diseases. In an effort to alleviate "pill burden" and improve patient compliance, fixed dose combinations (FDCs) have been developed to be used as effective therapeutics. Among all FDCs, the category of drug-drug molecular complexes has been proven an efficient methodology in designing and treating diseases, with many drugs being approved. Among all drug-drug molecular complexes, drug-drug cocrystals, salts, coamorphous systems and solid dispersions have been successfully developed and many have been approved by the FDA. In this review, we dwell deeply into the molecular mechanisms behind the different types of drug-drug molecular complexes, including the key functional groups involved in the intermolecular interactions, the applications of each category of molecular complexes, as well as the advantages and challenges thereof. This comprehensive review provides useful insights into the practical design and manufacture of drug-drug molecular complexes and points out the future direction for the development of new advantageous combinational therapies that benefit more patients.
Collapse
Affiliation(s)
- Jinghan Li
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Xiyan Wang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Dongyue Yu
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, NJ 08540, United States
| | - Yunping Zhoujin
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Kunlin Wang
- BeBetter Med Inc., Guangzhou, 510663, PR China; College of Pharmacy, Jinan University, Guangzhou, 510006, PR China.
| |
Collapse
|
6
|
Khalid GM, Billa N. Drug-Eluting Sutures by Hot-Melt Extrusion: Current Trends and Future Potentials. MATERIALS (BASEL, SWITZERLAND) 2023; 16:7245. [PMID: 38005174 PMCID: PMC10672932 DOI: 10.3390/ma16227245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/06/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023]
Abstract
Surgical site infections (SSIs) may result from surgical procedures requiring a secondary administration of drugs at site or systemically in treating the infection. Drug-eluting sutures containing antimicrobial agents symbolise a latent strategy that precludes a secondary drug administration. It also offers the possibility of delivering a myriad of therapeutic agents to a localised wound site to effect analgesia, anti-inflammation, or the deployment of proteins useful for wound healing. Further, the use of biodegradable drug-eluting sutures eliminates the need for implanting foreign material into the wound, which needs to be removed after healing. In this review, we expound on recent trends in the manufacture of drug-eluting sutures with a focus on the hot-melt extrusion (HME) technique. HME provides a solvent-free, continuous one-step manufacturing conduit for drug-eluting sutures, hence, there is no drying step, which can be detrimental to the drug or suture threads and, thus, environmentally friendly. There is the possibility of combining the technology with additive manufacturing platforms to generate personalised drug-loaded implantable devices through prototyping and scalability. The review also highlights key material requirements for fabricating drug-eluting sutures by HME, as well as quality attributes. Finally, a preview of emerging drug-eluting sutures and advocacy for harmonisation of quality assurance by regulatory authorities that permits quality evaluation of novelty sutures is presented.
Collapse
Affiliation(s)
- Garba M. Khalid
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK;
- FabRx Ltd., Henwood House, Henwood, Asford TN24 8DH, UK
| | - Nashiru Billa
- Pharmaceutical Sciences Department, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
7
|
Shao S, Stocker MW, Zarrella S, Korter TM, Singh A, Healy AM. In Situ Cocrystallization via Spray Drying with Polymer as a Strategy to Prevent Cocrystal Dissociation. Mol Pharm 2023; 20:4770-4785. [PMID: 37595572 PMCID: PMC10481393 DOI: 10.1021/acs.molpharmaceut.3c00564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
The aim of the present study was to investigate how different polymers affect the dissociation of cocrystals prepared by co-spray-drying active pharmaceutical ingredient (API), coformer, and polymer. Diclofenac acid-l-proline cocrystal (DPCC) was selected in this study as a model cocrystal due to its previously reported poor physical stability in a high-humidity environment. Polymers investigated include polyvinylpyrrolidone (PVP), poly(1-vinylpyrrolidone-co-vinyl acetate) (PVPVA), hydroxypropyl methyl cellulose, hydroxypropylmethylcellulose acetate succinate, ethyl cellulose, and Eudragit L-100. Terahertz Raman spectroscopy (THz Raman) and powder X-ray diffraction (PXRD) were used to monitor the cocrystal dissociation rate in a high-humidity environment. A Raman probe was used in situ to monitor the extent of the dissociation of DPCC and DPCC in crystalline solid dispersions (CSDs) with polymer when exposed to pH 6.8 phosphate buffer and water. The solubility of DPCC and solid dispersions of DPCC in pH 6.8 phosphate buffer and water was also measured. The dissociation of DPCC was water-mediated, and more than 60% of DPCC dissociated in 18 h at 40 °C and 95% RH. Interestingly, the physical stability of the cocrystal was effectively improved by producing CSDs with polymers. The inclusion of just 1 wt % polymer in a CSD with DPCC protected the cocrystal from dissociation over 18 h under the same conditions. Furthermore, the CSD with PVPVA was still partially stable, and the CSD with PVP was stable (undissociated) after 7 days. The superior stability of DPCC in CSDs with PVP and PVPVA was also demonstrated when systems were exposed to water or pH 6.8 phosphate buffer and resulted in higher dynamic solubility of the CSDs compared to DPCC alone. The improvement in physical stability of the cocrystal in CSDs was thought to be due to an efficient mixing between polymer and cocrystal at the molecular level provided by spray drying and in situ gelling of polymer. It is hypothesized that polymer chains could undergo gelling in situ and form a physical barrier, preventing cocrystal interaction with water, which contributes to slowing down the water-mediated dissociation.
Collapse
Affiliation(s)
- ShiZhe Shao
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin D02 PN40, Ireland
- SSPC,
the Science Foundation Ireland Research Centre for Pharmaceuticals, Trinity College Dublin, Dublin D02 PN40, Ireland
| | - Michael W. Stocker
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin D02 PN40, Ireland
- SSPC,
the Science Foundation Ireland Research Centre for Pharmaceuticals, Trinity College Dublin, Dublin D02 PN40, Ireland
- School
of Chemical and Bioprocess Engineering, University College Dublin, Dublin D04 V1W8, Ireland
| | - Salvatore Zarrella
- Department
of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, New York 13244, United States
| | - Timothy M. Korter
- Department
of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, New York 13244, United States
| | | | - Anne Marie Healy
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin D02 PN40, Ireland
- SSPC,
the Science Foundation Ireland Research Centre for Pharmaceuticals, Trinity College Dublin, Dublin D02 PN40, Ireland
| |
Collapse
|
8
|
Macedo J, Vanhoorne V, Vervaet C, Pinto JF. Influence of formulation variables on the processability and properties of tablets manufactured by fused deposition modelling. Int J Pharm 2023; 637:122854. [PMID: 36948473 DOI: 10.1016/j.ijpharm.2023.122854] [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: 12/01/2022] [Revised: 03/07/2023] [Accepted: 03/12/2023] [Indexed: 03/24/2023]
Abstract
The present work studied the influence of different formulation variables (defined also as factors), namely, different polymers (HPC EF, PVA and HPMC-AS LG), drugs with different water solubilities (paracetamol, hydrochlorothiazide and celecoxib) and drug loads (10 or 30 %) on their processability by HME and FDM. Both filaments and tablets were characterized for physic and chemical properties (DSC, XRPD, FTIR) and performance properties (drug content, in vitro drug release). Experiments were designed to highlight relationships between the 3 factors selected and the mechanical properties of filaments, tablet mass and dissolution profiles of the model drugs from printed tablets. While the combination of hydrochlorothiazide and HPMC-AS LG could not be extruded, the combination of paracetamol with HPC EF turned the filaments too ductile and not stiff enough hampering the process of printing. All other polymer and drug combinations could be successfully extruded and printed. Models reflected the influence of the solubility of the drug considered but not the drug load in formulations. The ranking of the drug release rates was in good agreement with their solubilities. Furthermore, PVA presenting the fastest swelling rate, promoted the fastest drugs' releases in comparison with the other polymers studied. Overall, the study enabled the identification of the key factors affecting the properties of printed tablets, with the proposal of a model that has valued the relative contribution of each factor to the overall performance of tablets.
Collapse
Affiliation(s)
- Joana Macedo
- iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - Valérie Vanhoorne
- Laboratory of Pharmaceutical Technology, Ghent University, Ghent, Belgium
| | - Chris Vervaet
- Laboratory of Pharmaceutical Technology, Ghent University, Ghent, Belgium
| | - João F Pinto
- iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
9
|
Myślińska M, Stocker MW, Ferguson S, Healy AM. A Comparison of Spray-Drying and Co-Precipitation for the Generation of Amorphous Solid Dispersions (ASDs) of Hydrochlorothiazide and Simvastatin. J Pharm Sci 2023:S0022-3549(23)00064-3. [PMID: 36805392 DOI: 10.1016/j.xphs.2023.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023]
Abstract
Co-processing of APIs, the practice of creating multi-component APIs directly in chemical processing facilities used to make drug substance, is gaining increased attention with a view to streamlining manufacturing, improving supply chain robustness and accessing enhanced product attributes in terms of stability and bioavailability. Direct co-precipitation of amorphous solid dispersions (ASDs) at the final step of chemical processing is one such example of co-processing. The purpose of this work was to investigate the application of different advanced solvent-based processing techniques - direct co-precipitation (CP) and the benchmark well-established spray-drying (SD) process - to the production of ASDs comprised of a drug with a high Tg (hydrochlorothiazide, HCTZ) or a low Tg (simvastatin, SIM) molecularly dispersed in a PVP/VA 64 or Soluplus® matrix. ASDs of the same composition were manufactured by the two different methods and were characterised using powder X-ray diffraction (PXRD), modulated differential scanning calorimetry (mDSC), attenuated total reflectance Fourier transform infrared spectroscopy (ATR-FTIR) and scanning electron microscopy (SEM). Both methods produced ASDs that were PXRD amorphous, with some differences, depending on the process used, in glass transition temperature and particle size distribution. Irrespective of manufacturing method used, all ASDs remained PXRD amorphous when subjected to high relative humidity conditions (75% RH, 25°C) for four weeks, although changes in the colour and physical characteristics were observed on storage for spray-dried systems with SIM and PVP/VA 64 copolymer. The particle morphology differed for co-precipitated compared to spray dried systems, with powder generated by the former process being comprised of more irregularly shaped particles of larger particle size when compared to the equivalent spray-dried systems which may enable more streamlined drug product processes to be used for CP materials. These differences may have implications in downstream drug product processing. A limitation identified when applying the solvent/anti-solvent co-precipitation method to SIM was the high antisolvent to solvent ratios required to effect the precipitation process. Thus, while similar outcomes may arise for both co-precipitation and spray drying processes in terms of ASD critical quality attributes, practical implications of applying the co-precipitation method and downstream processability of the resulting ASDs should be considered when choosing one solvent-based ASD production process over another.
Collapse
Affiliation(s)
- Monika Myślińska
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, Dublin 2, Ireland; SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland; EPSRC-SFI Centre for Doctoral Training in Transformative Pharmaceutical Technologies, Ireland
| | - Michael W Stocker
- School of Chemical and Bioprocess Engineering, University College Dublin, Dublin 4, Ireland; SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland
| | - Steven Ferguson
- School of Chemical and Bioprocess Engineering, University College Dublin, Dublin 4, Ireland; SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland; EPSRC-SFI Centre for Doctoral Training in Transformative Pharmaceutical Technologies, Ireland; I-Form, The SFI Research Centre for Advanced Manufacturing, School of Chemical and Bioprocess Engineering, University College Dublin, Dublin 4, Ireland; National Institute for Bioprocess Research and Training, Dublin, Ireland
| | - Anne Marie Healy
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, Dublin 2, Ireland; SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland; EPSRC-SFI Centre for Doctoral Training in Transformative Pharmaceutical Technologies, Ireland.
| |
Collapse
|
10
|
Hiew TN, Saboo S, Zemlyanov DY, Punia A, Wang M, Smith D, Lowinger M, Solomos MA, Schenck L, Taylor LS. Improving Dissolution Performance and Drug Loading of Amorphous Dispersions Through a Hierarchical Particle Approach. J Pharm Sci 2022:S0022-3549(22)00583-4. [PMID: 36574837 DOI: 10.1016/j.xphs.2022.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/01/2022] [Accepted: 12/18/2022] [Indexed: 12/26/2022]
Abstract
Co-precipitation is an emerging manufacturing strategy for amorphous solid dispersions (ASDs). Herein, the interplay between processing conditions, surface composition, and release performance was evaluated using grazoprevir and hypromellose acetate succinate as the model drug and polymer, respectively. Co-precipitated amorphous dispersion (cPAD) particles were produced in the presence and absence of an additional polymer that was either dissolved or dispersed in the anti-solvent. This additional polymer in the anti-solvent was deposited on the surfaces of the cPAD particles during isolation and drying to create hierarchical particles, which we define here as a core ASD particle with an additional water soluble component that is coating the particle surfaces. The resultant hierarchical particles were characterized using X-ray powder diffraction, differential scanning calorimetry, scanning electron microscopy, and X-ray photoelectron spectroscopy (XPS). Release performance was evaluated using a two-stage dissolution test. XPS analysis revealed a trend whereby cPAD particles with a lower surface drug concentration showed improved release relative to particles with a higher surface drug concentration, for nominally similar drug loadings. This surface drug concentration could be impacted by whether the secondary polymer was dissolved in the anti-solvent or dispersed in the anti-solvent prior to isolating final dried hierarchical cPAD powders. Grazoprevir exposure in dogs was higher when the hierarchical cPAD was dosed, with ∼1.8 fold increase in AUC compared to the binary cPAD. These observations highlight the important interplay between processing conditions and ASD performance in the context of cPAD particles and illustrate a hierarchical particle design as a successful approach to alter ASD surface chemistry to improve dissolution performance.
Collapse
Affiliation(s)
- Tze Ning Hiew
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Sugandha Saboo
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Dmitry Y Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, United States
| | - Ashish Punia
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Michael Wang
- Biopharmaceutics, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Daniel Smith
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Michael Lowinger
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Marina A Solomos
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Luke Schenck
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
11
|
Gottschalk T, Özbay C, Feuerbach T, Thommes M. Predicting Throughput and Melt Temperature in Pharmaceutical Hot Melt Extrusion. Pharmaceutics 2022; 14:pharmaceutics14091757. [PMID: 36145505 PMCID: PMC9502425 DOI: 10.3390/pharmaceutics14091757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Even though hot melt extrusion (HME) is a commonly applied process in the pharmaceutical area, determination of the optimal process parameters is demanding. The goal of this study was to find a rational approach for predetermining suitable extrusion parameters, with a focus on material temperature and throughput. A two-step optimization procedure, called scale-independent optimization strategy (SIOS), was applied and developed further, including the use of an autogenic extrusion mode. Three different polymers (Plasdone S-630, Soluplus, and Eudragit EPO) were considered, and different optimal process parameters were assessed. The maximum barrel load was dependent on the polymers’ bulk density and the extruder size. The melt temperature was influenced by the screw speed and the rheological behavior of the polymer. The melt viscosity depended mainly on the screw speed and was self-adjusted in the autogenic extrusion. A new approach, called SIOS 2.0, was suggested for calculating the extrusion process parameters (screw speed, melt temperature and throughput) based on the material data and a few extrusion experiments.
Collapse
Affiliation(s)
- Tobias Gottschalk
- Laboratory of Solids Process Engineering, Department of Biochemical and Chemical Engineering, TU Dortmund University, Emil-Figge-Str. 68, 44227 Dortmund, Germany
- INVITE GmbH, Drug Delivery Innovation Center, Chempark Building W32, 51368 Leverkusen, Germany
| | - Cihangir Özbay
- Laboratory of Solids Process Engineering, Department of Biochemical and Chemical Engineering, TU Dortmund University, Emil-Figge-Str. 68, 44227 Dortmund, Germany
| | - Tim Feuerbach
- INVITE GmbH, Drug Delivery Innovation Center, Chempark Building W32, 51368 Leverkusen, Germany
| | - Markus Thommes
- Laboratory of Solids Process Engineering, Department of Biochemical and Chemical Engineering, TU Dortmund University, Emil-Figge-Str. 68, 44227 Dortmund, Germany
- Correspondence: or
| |
Collapse
|
12
|
Factors Influencing the Crystallization-Onset Time of Metastable ASDs. Pharmaceutics 2022; 14:pharmaceutics14020269. [PMID: 35214002 PMCID: PMC8879851 DOI: 10.3390/pharmaceutics14020269] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 02/04/2023] Open
Abstract
In formulation development, amorphous solid dispersions (ASD) are considered to improve the bioavailability of poorly water-soluble active pharmaceutical ingredients (APIs). However, the crystallization of APIs often limits long-term stability and thus the shelf life of ASDs. It has already been shown earlier that the long-term stability of ASDs strongly depends on the storage conditions (relative humidity, temperature), the manufacturing methods, and the resulting particle sizes. In this work, ASDs composed of the model APIs Griseofulvin (GRI) or Itraconazole (ITR) and the polymers poly (vinylpyrrolidone-co-vinyl acetate) (PVPVA) or Soluplus® were manufactured via spray drying and hot-melt extrusion. Each API/polymer combination was manufactured using the two manufacturing methods with at least two different API loads and two particle-size distributions. It was a priori known that these ASDs were metastable and would crystallize over time, even in the dry stage. The amount of water absorbed by the ASD from humid air (40 °C/75% relative humidity), the solubility of the API in the ASD at humid conditions, and the resulting glass-transition temperature were predicted using the Perturbed-Chain Statistical Associating Fluid Theory (PC-SAFT) and the Gordon–Taylor approach, respectively. The onset of crystallization was determined via periodic powder X-ray diffraction (PXRD) measurements. It was shown that simple heuristics such as “larger particles always crystallize later than smaller particles” are correct within one manufacturing method but cannot be transferred from one manufacturing method to another. Moreover, amorphous phase separation in the ASDs was shown to also influence their crystallization kinetics. Counterintuitively, phase separation accelerated the crystallization time, which could be explained by the glass-transition temperatures of the evolving phases.
Collapse
|
13
|
Lagan C, Huckle JE, Katz JM, Khorsand B, Daurio D, Andrews GP, Chung J, Alvarez-Nunez F. Solvent-Assisted Hot Melt Extrusion of a Thermally Labile, High Melting Point Compound. AAPS PharmSciTech 2021; 22:235. [PMID: 34498147 DOI: 10.1208/s12249-021-02122-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 08/18/2021] [Indexed: 11/30/2022] Open
Abstract
Molecular dispersions are a highly effective method of increasing bioavailability for a poorly soluble active pharmaceutical ingredient (API) and can be prepared on a large scale by hot melt extrusion (HME). Processing thermally labile active pharmaceutical ingredients (APIs) via HME is generally more difficult, with operating temperatures limited to below that of the API melting point. API melting is considered essential to facilitate the formation of a fully homogeneous amorphous system. Processing below the melting point renders the system much more susceptible to residual crystalline content; hence, HME is not suitable for APIs which degrade upon melting. In the following work, meloxicam (MEL) was used as a model API, possessing properties of high melting temperature and thermal lability. In this proof of concept work, a modified HME method, termed solvent-assisted HME, was used to overcome this issue and prepare an amorphous solid dispersion using HME, wherein a solvent was incorporated in the formulation blend during extrusion and removed post-processing. Formulations containing 10%wt meloxicam (MEL) and 90%wt polyvinylpyrrolidone vinyl acetate (PVPVA) copolymer were extruded using a twin-screw extruder at temperatures below the melting point of MEL. Dimethylformamide (DMF) solvent was added directly into the extruder barrel through a liquid addition port, resulting in extrudate products having a higher conversion of API to the amorphous form. The incorporation of solvent allowed a significant reduction in processing temperatures due to its increased mobility, while also driving the conversion of the API to its amorphous form. The solvent was successfully reduced through a secondary drying step using a vacuum oven. This advancement has demonstrated the potential for thermally labile APIs to be processed via HME expanding the applications of this technology.
Collapse
|
14
|
Kim DW, Weon KY. Pharmaceutical application and development of fixed-dose combination: dosage form review. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2021. [DOI: 10.1007/s40005-021-00543-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
15
|
Supersaturating drug delivery systems containing fixed-dose combination of two antihypertensive drugs: Formulation, in vitro evaluation and molecular metadynamics simulations. Eur J Pharm Sci 2021; 163:105860. [PMID: 33901683 DOI: 10.1016/j.ejps.2021.105860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/26/2021] [Accepted: 04/19/2021] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to associate the poorly water-soluble antihypertensive drugs candesartan cilexetil (CC) and hydrochlorothiazide (HCTZ) as fixed-dose combination, in the form of ternary Amorphous Solid Dispersions (ASD), using hydroxypropylmethylcellulose acetate succinate (HPMCAS) type M as polymeric carrier. The potential of the system to generate and to maintain supersaturation of both drugs was also evaluated. The ASDs were prepared by ball milling technique and solid-state characterization was performed by differential scanning calorimetry (DSC), Fourier transformed infrared spectroscopy (FTIR) and X-ray powder diffraction (XRPD). Interaction between drugs and polymer in solid-state was evaluated by molecular metadynamics simulations. In vitro supersaturation profiles were determined in biorelevant medium. Physicochemical stability of ASDs was also evaluated under different storage conditions. Amorphization of both drugs was confirmed by solid-state characterization techniques. Molecular metadynamics simulations indicated that CC has stronger interaction with HMPCAS than HCTZ. In vitro supersaturation studies have shown that ternary ASDs could generate and maintain supersaturation of both drugs in biorelevant medium. The polymer reduced the desupersaturation of both drugs. Ternary ASDs also showed physicochemical stability over a period of 90 days, demonstrating the potential of the polymer in reducing the drugs recrystallization over the time. Ternary ASDs of CC, HCTZ and HPMCAS can be considered a promising system to associate the drugs as fixed-dose combinations. Also, these systems generate and maintain supersaturation of both drugs in biorelevant medium, with great storage stability. HPMCAS M was a good carrier for reducing the desupersaturation of associated HCTZ and CC.
Collapse
|
16
|
Schenck L, Boyce C, Frank D, Koranne S, Ferguson HM, Strotman N. Hierarchical Particle Approach for Co-Precipitated Amorphous Solid Dispersions for Use in Preclinical In Vivo Studies. Pharmaceutics 2021; 13:pharmaceutics13071034. [PMID: 34371726 PMCID: PMC8308979 DOI: 10.3390/pharmaceutics13071034] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 11/23/2022] Open
Abstract
Amorphous solid dispersions (ASD) have become a well-established strategy to improve exposure for compounds with insufficient aqueous solubility. Of methods to generate ASDs, spray drying is a leading route due to its relative simplicity, availability of equipment, and commercial scale capacity. However, the broader industry adoption of spray drying has revealed potential limitations, including the inability to process compounds with low solubility in volatile solvents, inconsistent molecular uniformity of spray dried amorphous dispersions, variable physical properties across batches and scales, and challenges containing potent compounds. In contrast, generating ASDs via co-precipitation to yield co-precipitated amorphous dispersions (cPAD) offers solutions to many of those challenges and has been shown to achieve ASDs comparable to those manufactured via spray drying. This manuscript applies co-precipitation for early safety studies, developing a streamlined process to achieve material suitable for dosing as a suspension in conventional toxicity studies. Development targets involved achieving a rapid, safely contained process for generating ASDs with high recovery yields. Furthermore, a hierarchical particle approach was used to generate composite particles where the cPAD material is incorporated in a matrix of water-soluble excipients to allow for rapid re-dispersibility in the safety study vehicle to achieve a uniform suspension for consistent dosing. Adopting such an approach yielded a co-precipitated amorphous dispersion with comparable stability, thermal properties, and in vivo pharmacokinetics to spray dried amorphous materials of the same composition.
Collapse
Affiliation(s)
- Luke Schenck
- Process Research & Development, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (D.F.); (N.S.)
- Correspondence:
| | - Christopher Boyce
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (C.B.); (H.M.F.)
| | - Derek Frank
- Process Research & Development, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (D.F.); (N.S.)
| | - Sampada Koranne
- Preformulation, Merck & Co., Inc., Kenilworth, NJ 07033, USA;
| | - Heidi M. Ferguson
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (C.B.); (H.M.F.)
| | - Neil Strotman
- Process Research & Development, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (D.F.); (N.S.)
| |
Collapse
|
17
|
de Margerie V, McConville C, Dadou SM, Li S, Boulet P, Aranda L, Walker A, Mohylyuk V, Jones DS, Murray B, Andrews GP. Continuous manufacture of hydroxychloroquine sulfate drug products via hot melt extrusion technology to meet increased demand during a global pandemic: From bench to pilot scale. Int J Pharm 2021; 605:120818. [PMID: 34174359 DOI: 10.1016/j.ijpharm.2021.120818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 11/26/2022]
Abstract
During pandemics and global crises, drug shortages become critical as a result of increased demand, shortages in personnel and lockdown restrictions that disrupt the supply chain. The pharmaceutical industry is therefore moving towards continuous manufacturing instead of conventional batch manufacturing involving numerous steps, that normally occur at different sites. In order to validate the use of large-scale industrial processes, feasibility studies need to be performed using small-scale laboratory equipment. To that end, the scale-up of a continuous process and its effect on the critical quality attributes (CQAs) of the end product were investigated in this work. Hydroxychloroquine Sulphate (HCQS) was used as the model drug, Soluplus® as a model polymeric carrier and both horizontal and vertical twin screw extruders used to undertake this hot melt extrusion (HME) study. Seven formulations were processed using a small-scale horizontal extruder and a pilot-scale vertical extruder at various drug loadings, temperature profiles and screw speeds. When utilising a horizontal extruder, formulations with the highest drug load and processed at the lowest screw speed and temperature had the highest crystallinity with higher drug release rates. Upon scale-up to a vertical extruder, the crystallinity of the HCQS was significantly reduced, with less variation in both crystallinity and release profile across the different extrudates. This study demonstrates improved robustness with the pilot-scale vertical extruder compared to lab-scale horizontal extruder. The reduced variation with the vertical extruder will allow for short increases in production rate, with minimum impact on the CQAs of the final product enabling high-performance continuous manufacturing with minimum waste of raw materials. Finally, this research provides valuable information for the pharmaceutical industry in accessing continuous technologies for the manufacture of pharmaceutical products, allowing for efficient utilisation of resources upon scale-up and mass production during global pandemics and drug shortages.
Collapse
Affiliation(s)
| | - Christopher McConville
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Suha M Dadou
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom; China Medical University - Queen's University Belfast joint College (CQC), No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Shu Li
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom; China Medical University - Queen's University Belfast joint College (CQC), No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Pascal Boulet
- Institut Jean Lamour, UMR 7198 CNRS-Université de Lorraine, 2 Allée André Guinier, 54000 Nancy, France
| | - Lionel Aranda
- Institut Jean Lamour, UMR 7198 CNRS-Université de Lorraine, 2 Allée André Guinier, 54000 Nancy, France
| | - Andrew Walker
- Rondol Industrie, 2 Allée André Guinier, 54000 Nancy, France
| | - Valentyn Mohylyuk
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom; China Medical University - Queen's University Belfast joint College (CQC), No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - David S Jones
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Brian Murray
- Rondol Industrie, 2 Allée André Guinier, 54000 Nancy, France
| | - Gavin P Andrews
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom; China Medical University - Queen's University Belfast joint College (CQC), No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
18
|
Browne E, Quinn S, Cheyne S, Healy AM. Design and characterisation of an amorphous formulation of nifedipine for the treatment of autonomic dysreflexia. J Pharm Pharmacol 2021; 73:928-936. [PMID: 33749786 DOI: 10.1093/jpp/rgab034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 02/13/2021] [Indexed: 01/23/2023]
Abstract
OBJECTIVES Current treatment for autonomic dysreflexia (AD) involves rupturing a liquid-filled soft capsule of nifedipine to aid rapid drug release and absorption, however, this application is not covered under the manufacturer's license. The objective of the current work was to design a rapidly dissolving solid dosage formulation for the treatment of AD as an alternative to the off-license "bite and swallow" use of currently available commercial products. METHODS Amorphous solid dispersions (ASDs) of nifedipine were prepared by spray-drying using three different polymers: hydroxypropyl methyl cellulose (HPMC), polyvinyl pyrrolidone (PVP) and polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol (Soluplus), at a 15% w/w drug loading and were formulated and compressed into tablets. Dissolution testing was performed in the paddle dissolution apparatus using either a monophasic or biphasic medium. KEY FINDINGS The PVP-nifedipine ASD tablets exhibited rapid dissolution, with 35% of the total nifedipine dose dissolving within 15 min in the monophasic dissolution medium. The HPMC-nifedipine ASD exhibited a very slow dissolution, while the Solupus-nifedipine system exhibited no nifedipine release over 120 min. When tested in the biphasic dissolution medium, the PVP-nifedipine ASD tablets exhibited a release profile comparable to that of the pre-split/ruptured nifedipine soft capsule product. CONCLUSIONS This study demonstrates that a nifedipine-PVP ASD is a promising formulation strategy in the treatment of AD.
Collapse
Affiliation(s)
- Emer Browne
- SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Sian Quinn
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Sheena Cheyne
- National Rehabilitation Hospital, Dun Laoghaire, County Dublin, Ireland
| | - Anne Marie Healy
- SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
19
|
Yu J, Yu D, Lane S, McConnachie L, Ho RJY. Controlled Solvent Removal from Antiviral Drugs and Excipients in Solution Enables the Formation of Novel Combination Multi-Drug-Motifs in Pharmaceutical Powders Composed of Lopinavir, Ritonavir and Tenofovir. J Pharm Sci 2020; 109:3480-3489. [PMID: 32791073 PMCID: PMC8986323 DOI: 10.1016/j.xphs.2020.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 01/15/2023]
Abstract
Diverging physicochemical properties of HIV drug combinations are challenging to formulate as a single dosage form. We have found that 2-to-4 hydrophilic and hydrophobic HIV drugs in combination can be stabilized with lipid excipients under a controlled solvent removal process to form a novel pharmaceutical powder distinct from typical amorphous material. This discovery has enabled production of a drug combination nanoparticle (DcNP) powder composed of 3 HIV drugs-water-insoluble lopinavir (LogP = 4.7) and ritonavir (LogP = 5.6) and water-soluble tenofovir (LogP = -1.6). DcNP powder, exhibiting repeating units of multi-drug-motifs (referred to as MDM), is made by dissolving all constituents in ethanolic solution, followed by controlled solvent removal. The DcNP powder intersperses chemically diverse drug molecules with lipid excipients to form repeating MDM units. The proposed MDM structure is consistent with data collected with X-ray diffraction, differential calorimetry, and time-of-flight secondary ion mass spectrometry. The successful assembly of chemically diverse drugs in MDM structure is likely due to a novel process of making drug combination powders. The method described here has successfully extended to formulating other clinically prescribed antiviral drug combinations, and thus may serve as a platform technology for developing drug combination nanoparticles for treating a wide range of chronic diseases.
Collapse
Affiliation(s)
- Jesse Yu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195-7610, USA
| | - Danni Yu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195-7610, USA
| | - Sarah Lane
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195-7610, USA
| | - Lisa McConnachie
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195-7610, USA
| | - Rodney J Y Ho
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195-7610, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195-7610, USA.
| |
Collapse
|
20
|
Dadou SM, Tian Y, Li S, Jones DS, Andrews GP. The optimization of process analytical technology for the inline quantification of multiple drugs in fixed dose combinations during continuous processing. Int J Pharm 2020; 592:120024. [PMID: 33130221 DOI: 10.1016/j.ijpharm.2020.120024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 10/23/2022]
Abstract
Complications associated with uncontrolled hypertension are considered the major cause of premature death worldwide. Fixed-dose combinations (FDCs) offer an alternative approach to polypharmacy with the aim to improve patient compliance. Process Analytical Technology (PAT) is gaining momentum as a non-invasive, predictive tool to control the quality of drugs during continuous processing. PAT offers real-time quality control that can be built into the production line. However, the vast majority of studies reported in the literature have focused on quantifying a single drug during continuous processing. The aim of this study was to develop non-destructive, predictive inline PAT tools allowing for the simultaneous quantification of two antihypertensive drugs, Hydrochlorothiazide (HCTZ) and Ramipril (RMP), during the continuous manufacture of FDCs. A calibration set composed of HCTZ and RMP at concentration ranges of 6.5 to 40 and 2.5-15 (% w/w), respectively, were manufactured using hot melt extrusion. The extrudates were analysed during the process using inline Raman spectroscopy. Optimum wavenumber regions were observed at 200-400 and 630-730 cm-1 for HCTZ, and 980-1100 cm-1 for RMP using principal component analysis. Partial least squares (PLS) regression was performed to establish the predictive calibration models. The PLS developed models showed excellent linearity (R2 = 0.986 and 0.974), selectivity (PC1 = 98.6% and 91.9%) and accuracy (RMSEcv = 1.586 and 0.645%) for HCTZ and RMP, respectively. Additionally, RMSEP values were reported as 1.237 and 1.007% for HCTZ and RMP, respectively, depicting good predictability for drug content in the validation set. The output of this study demonstrated that utilisation of the full potential of chemometrics, Raman spectroscopy can be used for the simultaneous inline quantification of multiple drugs in complex formulations. This facilitates the in-process quality control of FDCs and other multicomponent systems during continuous pharmaceutical production.
Collapse
Affiliation(s)
- Suha M Dadou
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom; China Medical University - Queen's University Belfast joint College (CQC), No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Yiwei Tian
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Shu Li
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom; China Medical University - Queen's University Belfast joint College (CQC), No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - David S Jones
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Gavin P Andrews
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom; China Medical University - Queen's University Belfast joint College (CQC), No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
21
|
Characterization of ternary amorphous solid dispersion containing hypromellose phthalate and erythritol prepared by hot melt extrusion using melting point depression. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
22
|
Oral Fixed-Dose Combination Pharmaceutical Products: Industrial Manufacturing Versus Personalized 3D Printing. Pharm Res 2020; 37:132. [PMID: 32556831 DOI: 10.1007/s11095-020-02847-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022]
Abstract
Fixed-dose combination (FDC) products containing at least two different active pharmaceutical ingredients are designed to treat more effectively different pathologies as they have demonstrated to enhance patient compliance. However, the combination of multiple drugs within the same dosage form can bring many physicochemical and pharmacodynamic interactions. The manufacturing process of FDC products can be challenging, especially when it is required to achieve different drug release profiles within the same dosage form to overcome physicochemical drug interactions. Monolithic, multiple-layer, and multiparticulate systems are the most common type of FDCs. Currently, the main manufacturing techniques utilized in industrial pharmaceutical companies rely on the use of combined wet and dry granulation, hot-melt extrusion coupled with spray coating, and compression of bilayered tablets. Nowadays, personalized medicines are gaining importance in clinical settings and 3D printing is taking a highlighted role in the manufacturing of complex and personalized 3D solid dosage forms that could not be manufactured using conventional techniques. In this review, it will be discussed in detail current marketed FDC products and their application in several diseases with an especial focus on antimicrobial drugs. Current industrial conventional techniques will be compared with 3D printing manufacturing of FDCs. Graphical Abstract.
Collapse
|
23
|
The development and validation of a quality by design based process analytical tool for the inline quantification of Ramipril during hot-melt extrusion. Int J Pharm 2020; 584:119382. [DOI: 10.1016/j.ijpharm.2020.119382] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 11/24/2022]
|
24
|
Leyva-Porras C, Cruz-Alcantar P, Espinosa-Solís V, Martínez-Guerra E, Piñón-Balderrama CI, Compean Martínez I, Saavedra-Leos MZ. Application of Differential Scanning Calorimetry (DSC) and Modulated Differential Scanning Calorimetry (MDSC) in Food and Drug Industries. Polymers (Basel) 2019; 12:polym12010005. [PMID: 31861423 PMCID: PMC7023573 DOI: 10.3390/polym12010005] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 02/07/2023] Open
Abstract
Phase transition issues in the field of foods and drugs have significantly influenced these industries and consequently attracted the attention of scientists and engineers. The study of thermodynamic parameters such as the glass transition temperature (Tg), melting temperature (Tm), crystallization temperature (Tc), enthalpy (H), and heat capacity (Cp) may provide important information that can be used in the development of new products and improvement of those already in the market. The techniques most commonly employed for characterizing phase transitions are thermogravimetric analysis (TGA), dynamic mechanical analysis (DMA), thermomechanical analysis (TMA), and differential scanning calorimetry (DSC). Among these techniques, DSC is preferred because it allows the detection of transitions in a wide range of temperatures (−90 to 550 °C) and ease in the quantitative and qualitative analysis of the transitions. However, the standard DSC still presents some limitations that may reduce the accuracy and precision of measurements. The modulated differential scanning calorimetry (MDSC) has overcome some of these issues by employing sinusoidally modulated heating rates, which are used to determine the heat capacity. Another variant of the MDSC is the supercooling MDSC (SMDSC). SMDSC allows the detection of more complex thermal events such as solid–solid (Ts-s) transitions, liquid–liquid (Tl-l) transitions, and vitrification and devitrification temperatures (Tv and Tdv, respectively), which are typically found at the supercooling temperatures (Tco). The main advantage of MDSC relies on the accurate detection of complex transitions and the possibility of distinguishing reversible events (dependent on the heat capacity) from non-reversible events (dependent on kinetics).
Collapse
Affiliation(s)
- César Leyva-Porras
- Centro de Investigación en Materiales Avanzados S.C. (CIMAV), Miguel de Cervantes # 120, Complejo Industrial Chihuahua, Chihuahua 31136, CHIH, Mexico;
| | - Pedro Cruz-Alcantar
- Coordinación Académica Región Altiplano, Universidad Autónoma de San Luis Potosí, Carretera Cedral Km, 5+600, Ejido San José de las Trojes Matehuala, San Luis Potosi 78700, SLP, Mexico; (P.C.-A.); (I.C.M.)
| | - Vicente Espinosa-Solís
- Coordinación Académica Región Huasteca Sur de la UASLP, Universidad Autónoma de San Luís Potosí, km. 5, Carretera Tamazunchale-San Martín, Tamazunchale 79960, SLP, Mexico;
| | - Eduardo Martínez-Guerra
- Centro de Investigación en Materiales Avanzados S.C. (CIMAV), Alianza Norte No. 202, Autopista Monterrey-Aeropuerto Km 10, Parque de Investigación e Innovación Tecnológica (PIIT), Apodaca 66600, NL, Mexico;
| | - Claudia I. Piñón-Balderrama
- Coordinación Académica Región Altiplano, Universidad Autónoma de San Luis Potosí, Carretera Cedral Km, 5+600, Ejido San José de las Trojes Matehuala, San Luis Potosi 78700, SLP, Mexico; (P.C.-A.); (I.C.M.)
- Centro de Investigación en Materiales Avanzados S.C. (CIMAV), Alianza Norte No. 202, Autopista Monterrey-Aeropuerto Km 10, Parque de Investigación e Innovación Tecnológica (PIIT), Apodaca 66600, NL, Mexico;
| | - Isaac Compean Martínez
- Coordinación Académica Región Altiplano, Universidad Autónoma de San Luis Potosí, Carretera Cedral Km, 5+600, Ejido San José de las Trojes Matehuala, San Luis Potosi 78700, SLP, Mexico; (P.C.-A.); (I.C.M.)
| | - María Z. Saavedra-Leos
- Coordinación Académica Región Altiplano, Universidad Autónoma de San Luis Potosí, Carretera Cedral Km, 5+600, Ejido San José de las Trojes Matehuala, San Luis Potosi 78700, SLP, Mexico; (P.C.-A.); (I.C.M.)
- Centro de Investigación en Materiales Avanzados S.C. (CIMAV), Alianza Norte No. 202, Autopista Monterrey-Aeropuerto Km 10, Parque de Investigación e Innovación Tecnológica (PIIT), Apodaca 66600, NL, Mexico;
- Correspondence:
| |
Collapse
|
25
|
Kelleher JF, Madi AM, Gilvary GC, Tian YW, Li S, Almajaan A, Loys ZS, Jones DS, Andrews GP, Healy AM. Metformin Hydrochloride and Sitagliptin Phosphate Fixed-Dose Combination Product Prepared Using Melt Granulation Continuous Processing Technology. AAPS PharmSciTech 2019; 21:23. [PMID: 31832799 DOI: 10.1208/s12249-019-1553-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022] Open
Abstract
The development of oral solid dosage forms, such as tablets that contain a high dose of drug(s), requires polymers and other additives to be incorporated at low levels as possible, to keep the final tablet weight low, and, correspondingly, the dosage form size small enough to be acceptable from a patient perspective. Additionally, a multi-step batch-based manufacturing process is usually required for production of solid dosage forms. This study presents the development and production, by twin-screw melt granulation technology, of a high-dose immediate-release fixed-dose combination (FDC) product of metformin hydrochloride (MET) and sitagliptin phosphate (SIT), with drug loads of 80% w/w and 6% w/w, respectively. For an 850/63 mg dose of MET/SIT, the final weight of the caplets was approximately 1063 mg compared with 1143 mg for the equivalent dose in Janumet®, the marketed product. Mixtures of the two drugs and polymers were melt-granulated at temperatures below the individual melting temperatures of MET and SIT (231.65 and 213.89°C, respectively) but above the glass transition temperature or melting temperature of the binder(s) used. By careful selection of binders, and processing conditions, direct compressed immediate-release caplets with desired product profiles were successfully produced. The melt granule formulations before compression showed good flow properties, were larger in particle size than individual starting API materials and were easily compressible. Melt granulation is a suitable platform for developing direct compressible high-dose immediate-release solid dosage forms of FDC products.
Collapse
|
26
|
Kojima H, Nakamura S, Haraguchi T, Yoshida M, Habara M, Ikezaki H, Uchida T. A new strategy for taste masking on bitter drug by other combined drug in fixed-dose combination: bitterness of Amlodipine besylate could be masked efficiently by Valsartan. J Pharm Pharmacol 2019; 71:1384-1392. [DOI: 10.1111/jphp.13134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/15/2019] [Indexed: 11/29/2022]
Abstract
Abstract
Objectives
The aim of this study was to evaluate the bitterness of amlodipine besylate (AML) combined with other five antihypertensive drugs: alacepril, benazepril, hydrochlorothiazide, telmisartan (TEL) and valsartan (VAL), which have possibility of usage as a fixed-dose combination (FDC) drugs.
Methods
The bitterness of individual six drugs and AML combined with each of the five drugs was evaluated using taste sensor SA402B (Intelligent Sensor Technology Inc.). AML combined with TEL or VAL was evaluated by taste sensor and human gustatory sensation tests. The interaction between AML with TEL or VAL was evaluated by 1H-NMR.
Key findings
The bitterness of AML was significantly decreased by addition of VAL, whereas it remained unchanged by the addition of TEL in taste sensor and human gustatory sensation test. In the 1H-NMR spectrum of AML with VAL, signal shifts of protons in AML were observed compared to that in AML alone. On the other hand, in the 1H-NMR spectrum of AML with TEL, signal shifts of protons in AML were not observed.
Conclusions
It was suggested that when VAL was mixed with AML, the electrostatic interactions between positive charged amino group of AML and negative charged tetrazole group of VAL were caused, and thereby led the suppression the bitterness of AML.
Collapse
Affiliation(s)
- Honami Kojima
- Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya City, Japan
| | - Saki Nakamura
- Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya City, Japan
| | - Tamami Haraguchi
- Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya City, Japan
| | - Miyako Yoshida
- Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya City, Japan
| | | | | | - Takahiro Uchida
- Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya City, Japan
| |
Collapse
|
27
|
Browne E, Charifou R, Worku ZA, Babu RP, Healy AM. Amorphous solid dispersions of ketoprofen and poly-vinyl polymers prepared via electrospraying and spray drying: A comparison of particle characteristics and performance. Int J Pharm 2019; 566:173-184. [DOI: 10.1016/j.ijpharm.2019.05.062] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 11/25/2022]
|
28
|
Kallakunta VR, Sarabu S, Bandari S, Tiwari R, Patil H, Repka MA. An update on the contribution of hot-melt extrusion technology to novel drug delivery in the twenty-first century: part I. Expert Opin Drug Deliv 2019; 16:539-550. [PMID: 31007090 PMCID: PMC6791722 DOI: 10.1080/17425247.2019.1609448] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/16/2019] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Currently, hot melt extrusion (HME) is a promising technology in the pharmaceutical industry, as evidenced by its application to manufacture various FDA-approved commercial products in the market. HME is extensively researched for enhancing the solubility and bioavailability of poor water-soluble drugs, taste masking, and modifying release in drug delivery systems. Additionally, its other novel opportunities or pharmaceutical applications, and capability for continuous manufacturing are being investigated. This efficient, industrially scalable, solvent-free, continuous process can be easily automated and coupled with other novel platforms for continuous manufacturing of pharmaceutical products. AREAS COVERED This review focuses on updates on solubility enhancement of poorly water-soluble drugs and process analytical tools such as UV/visible spectrophotometry; near-infrared spectroscopy; Raman spectroscopy; and rheometry for continuous manufacturing, with a special emphasis on fused deposition modeling 3D printing. EXPERT OPINION The strengths, weakness, opportunities, threats (SWOT) and availability of commercial products confirmed wide HME applicability in pharmaceutical research. Increased interest in continuous manufacturing processes makes HME a promising strategy for this application. However, there is a need for extensive research using process analytical tools to establish HME as a dependable continuous manufacturing process.
Collapse
Affiliation(s)
- Venkata Raman Kallakunta
- Department of Pharmaceutics and Drug Delivery, The University of Mississippi, University, MS 38677
| | - Sandeep Sarabu
- Department of Pharmaceutics and Drug Delivery, The University of Mississippi, University, MS 38677
| | - Suresh Bandari
- Department of Pharmaceutics and Drug Delivery, The University of Mississippi, University, MS 38677
| | - Roshan Tiwari
- Department of Pharmaceutics and Drug Delivery, The University of Mississippi, University, MS 38677
| | - Hemlata Patil
- Department of Pharmaceutics and Drug Delivery, The University of Mississippi, University, MS 38677
| | - Michael A Repka
- Department of Pharmaceutics and Drug Delivery, The University of Mississippi, University, MS 38677
- Pii Center for Pharmaceutical Technology, The University of Mississippi, University, MS 38677
| |
Collapse
|
29
|
Schenck L, Koynov A, Cote A. Particle engineering at the drug substance, drug product interface: a comprehensive platform approach to enabling continuous drug substance to drug product processing with differentiated material properties. Drug Dev Ind Pharm 2019; 45:521-531. [DOI: 10.1080/03639045.2018.1562467] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Luke Schenck
- Chemical Engineering R&D, Merck & Co., Inc., Rahway, NJ, USA
| | - Athanas Koynov
- Preclinical Development, Merck & Co., Inc., Rahway, NJ, USA
| | - Aaron Cote
- Chemical Engineering R&D, Merck & Co., Inc., Rahway, NJ, USA
| |
Collapse
|
30
|
Schenck L, Mann AKP, Liu Z, Milewski M, Zhang S, Ren J, Dewitt K, Hermans A, Cote A. Building a better particle: Leveraging physicochemical understanding of amorphous solid dispersions and a hierarchical particle approach for improved delivery at high drug loadings. Int J Pharm 2019; 559:147-155. [PMID: 30654058 DOI: 10.1016/j.ijpharm.2019.01.009] [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: 11/13/2018] [Revised: 01/03/2019] [Accepted: 01/07/2019] [Indexed: 11/28/2022]
Abstract
Amorphous solid dispersions are a promising option for managing compounds with poor aqueous solubility. However, for compounds with high melting points, thermal stability limitations, or poor solubility in volatile solvents, conventional routes of hot melt extrusion or spray drying may not be viable. Co-precipitated amorphous dispersions (cPAD) can provide a solution. For the material studied in this paper, the cPAD material that was seemingly identical to spray dried material in terms of being single phase amorphous (as measured by DSC and XRD ) but showed slower dissolution behavior. It was identified that physical properties of the cPAD material could be improved to enhance wettability and improve dissolution performance. This was achieved by incorporating the cPAD material into a matrix of water soluble excipients generated via evaporative isolation routes. Importantly, this approach appears to offer another route to further increase the drug load in final dosage units and is significant as increased drug loading generally results in slower or incomplete release. Results showed successful proof of concept via in vitro biorelevant dissolution and confirmatory canine pharmacokinetic studies yielding comparable exposure for capsules comprised of conventional spray dried material as well as capsules with elevated drug load comprised of cPAD hierarchical particles.
Collapse
Affiliation(s)
- Luke Schenck
- Particle Engineering Labs, Chemical Engineering R&D, Merck & Co., Inc, Rahway, NJ 07065, USA.
| | - Amanda K P Mann
- Department of Analytical Sciences, Pharmaceutical Sciences, Merck & Co., Inc, Rahway, NJ 07065, USA.
| | - Zhen Liu
- Preformulation, Pharmaceutical Sciences Merck & Co., Inc, West Point, PA 19486, USA
| | - Mikolaj Milewski
- Biopharmaceutics and Specialty Dosage Forms, Pharmaceutical Sciences, Merck & Co., Inc, West Point, PA 19486, USA
| | - Siwei Zhang
- MMC, Process Research and Design, Merck & Co., Inc, Rahway, NJ 07065, USA
| | - Jie Ren
- OFST, Pharmaceutical Sciences, Merck & Co., Inc, West Point, PA 19486, USA
| | - Kristel Dewitt
- Department of Analytical Sciences, Pharmaceutical Sciences, Merck & Co., Inc, Rahway, NJ 07065, USA
| | - Andre Hermans
- Department of Analytical Sciences, Pharmaceutical Sciences, Merck & Co., Inc, Rahway, NJ 07065, USA
| | - Aaron Cote
- Technology Labs, Chemical Engineering R&D, Merck & Co., Inc, Rahway, NJ 07065, USA
| |
Collapse
|
31
|
Abdelquader MM, Essa EA, El Maghraby GM. Inhibition of Co-Crystallization of Olmesartan Medoxomil and Hydrochlorothiazide for Enhanced Dissolution Rate in Their Fixed Dose Combination. AAPS PharmSciTech 2018; 20:3. [PMID: 30560314 DOI: 10.1208/s12249-018-1207-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/01/2018] [Indexed: 12/21/2022] Open
Abstract
Olmesartan medoxomil (Olm) and hydrochlorothiazide (HCTZ) are fixed dose combination (FDC) for treatment of hypertension. They have hydrogen bonding sites and may interact during co-processing. The consequences of such interaction are not clear. This study investigated the possibility of this interaction during co-processing. The research was extended to inhibit deleterious interactions. The drugs were co-evaporated from ethanolic solution to maximize the chance of interaction. This was performed in the absence and presence of hydroxypropyl methylcellulose (HPMC) and/or aerosil. The products were characterized using Fourier transform infrared spectroscopy (FTIR), differential thermal analysis, and powder X-ray diffraction (PXRD) in addition to dissolution studies. Co-evaporation of Olm with HCTZ in the absence of excipients produced crystalline material with FTIR spectrum showing intermolecular hydrogen bonding. This material showed thermal pattern of new crystalline species. This was identified as Olm/HCTZ co-crystal by PXRD. This co-crystallization reduced the dissolution rate of both drugs. This co-crystallization was inhibited in the presence of HPMC, but the dissolution rate was not significantly enhanced accordingly. Co-processing in the presence of both HPMC and aerosil eliminated the co-crystallization and minimized the intermolecular drug-drug interaction with subsequent dissolution enhancement. The study introduced a composition for fixed dose combination of Olm and HCTZ with enhanced dissolution.
Collapse
|
32
|
Walsh D, Serrano DR, Worku ZA, Madi AM, O'Connell P, Twamley B, Healy AM. Engineering of pharmaceutical cocrystals in an excipient matrix: Spray drying versus hot melt extrusion. Int J Pharm 2018; 551:241-256. [PMID: 30223079 DOI: 10.1016/j.ijpharm.2018.09.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/28/2018] [Accepted: 09/12/2018] [Indexed: 10/28/2022]
Abstract
The comparison of spray drying versus hot melt extrusion (HME) in order to formulate amorphous solid dispersions has been widely studied. However, to the best of our knowledge, the use of both techniques to form cocrystals within a carrier excipient has not previously been compared. The combination of ibuprofen (IBU) and isonicotinamide (INA) in a 1:1 M ratio was used as a model cocrystal. A range of pharmaceutical excipients was selected for processing - mannitol, xylitol, Soluplus and PVP K15. The ratio of cocrystal components to excipient was altered to assess the ratios at which cocrystal formation occurs during spray drying and HME. Hansen Solubility Parameter (HSP) and the difference in HSP between the cocrystal and excipient (ΔHSP) was employed as a tool to predict cocrystal formation. During spray drying, when the difference in HSP between the cocrystal and the excipient was large, as in the case of mannitol (ΔHSP of 18.3 MPa0.5), a large amount of excipient (up to 50%) could be incorporated without altering the integrity of the cocrystal, whereas for Soluplus and PVP K15, where the ΔHSP was 2.1 and 1.6 MPa0.5 respectively, the IBU:INA cocrystal alone was only formed at a very low weight ratio of excipient, i.e. cocrystal:excipient 90:10. Remarkably different results were obtained in HME. In the case of Soluplus and PVP K15, a mixture of cocrystal with single components (IBU and INA) was obtained even when only 10% excipient was included. In conclusion, in order to reduce the number of unit operations required to produce a final pharmaceutical product, spray drying showed higher feasibility over HME to produce cocrystals within a carrier excipient.
Collapse
Affiliation(s)
- David Walsh
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland; SSPC, Synthesis and Solid State Pharmaceutical Centre, Ireland
| | - Dolores R Serrano
- School of Pharmacy, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Zelalem Ayenew Worku
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland; SSPC, Synthesis and Solid State Pharmaceutical Centre, Ireland
| | - Atif M Madi
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland; SSPC, Synthesis and Solid State Pharmaceutical Centre, Ireland
| | - Peter O'Connell
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Anne Marie Healy
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland; SSPC, Synthesis and Solid State Pharmaceutical Centre, Ireland.
| |
Collapse
|
33
|
Kavanagh ON, Albadarin AB, Croker DM, Healy AM, Walker GM. Maximising success in multidrug formulation development: A review. J Control Release 2018; 283:1-19. [DOI: 10.1016/j.jconrel.2018.05.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/18/2018] [Accepted: 05/19/2018] [Indexed: 12/20/2022]
|