1
|
Appell MB, Malmberg K, Pasupathy A, Josyula A, Ortiz J, McDonnell PJ, Shekhawat N, Parikh KS, Ensign LM. Ion-paired moxifloxacin nanocrystal formulation improves treatment and prevention of ocular infection. J Control Release 2025; 381:113634. [PMID: 40096866 PMCID: PMC11994289 DOI: 10.1016/j.jconrel.2025.113634] [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: 08/27/2024] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
Ocular infections may arise spontaneously or following penetrating globe injury or operation, such as corneal transplant or cataract extraction. Treatment and prophylaxis of bacterial infections using antibiotic eye drops requires a strict dosing regimen to avoid irreversible vision loss. At present, moxifloxacin eye drops are prescribed for use multiple times per day, leading to patient non-adherence, the emergence of bacterial resistance, and infection progression. The desire to avoid sub-lethal antibiotic dosing and visual impairment through inconsistent eye drop application motivates the development of a sustained release injectable formulation. Herein, we report the development of an ion-paired, nanocrystalline moxifloxacin formulation that provided increased intraocular antibiotic accumulation with a single subconjunctival injection compared to 3× daily eye drops. The sustained release functionality further led to improved or non-inferior prevention and treatment of Staphylococcus aureus-induced ocular infection in both rats and rabbits compared to gold standard intracameral moxifloxacin and moxifloxacin eye drops. By achieving therapeutically relevant moxifloxacin accumulation, this nanocrystalline moxifloxacin formulation may be a promising alternative to conventional therapies to achieve improved post-surgical infection prevention and bacterial keratitis treatment outcomes.
Collapse
Affiliation(s)
- Matthew B Appell
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kiersten Malmberg
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ashwin Pasupathy
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Aditya Josyula
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jairo Ortiz
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA
| | - Peter J McDonnell
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD 21231, USA
| | - Nakul Shekhawat
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD 21231, USA
| | - Kunal S Parikh
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Bioengineering Innovation & Design, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Laura M Ensign
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD 21231, USA; Department of Gynecology and Obstetrics and Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
2
|
Shalmani AA, Daware R, Elshafei AS, De Lorenzi F, Moeckel D, Buhl EM, Klinkhammer BM, Boor P, Banala S, Adams A, Kiessling F, Storm G, Metselaar JM, Sofias AM, Shi Y, Lammers T, Peña Q, Ojha T. Hydrophobic ion pairing enables co-loading of water-soluble drugs in polymeric micelles. J Control Release 2025:113748. [PMID: 40252978 DOI: 10.1016/j.jconrel.2025.113748] [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: 10/28/2024] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Polymeric micelles are widely used for the delivery of hydrophobic drugs. However, several highly potent and ubiquitously used anticancer drugs are water-soluble, complicating their (co-) formulation in polymeric micelles without having to chemically modify them. We here explore hydrophobic ion pairing to enable the co-delivery of the water-soluble anthracycline chemotherapy drug doxorubicin and the angiotensin II receptor antagonist telmisartan (a clinically used antihypertensive drug that has shown promising (pre-) clinical outcomes in combination with anthracyclines). We show that hydrophobic ion pairing of doxorubicin and telmisartan promotes the co-encapsulation of both drugs in π electron-stabilized [PEG-b-p(HPMAm-Bz)]-based polymeric micelles. The cytotoxic activity of doxorubicin is retained, and the dual drug-loaded micelles display enhanced antitumor activity in vivo as compared to the combination of the free drugs, while also exhibiting good tolerability. Taken together, this work provides proof-of-concept for hydrophobic ion pairing as a promising formulation strategy to promote multidrug nanomedicine and drug combination therapy.
Collapse
Affiliation(s)
- Armin Azadkhah Shalmani
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Rasika Daware
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Asmaa Said Elshafei
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Federica De Lorenzi
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Diana Moeckel
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Eva M Buhl
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | | | - Peter Boor
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany; Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Srinivas Banala
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Alina Adams
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, 52074 Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584CG Utrecht, the Netherlands; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore
| | - Josbert M Metselaar
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Alexandros Marios Sofias
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Yang Shi
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany.
| | - Quim Peña
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany.
| | - Tarun Ojha
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584CG Utrecht, the Netherlands.
| |
Collapse
|
3
|
Jeon Y, Kim SG, Choi KO, Park JT. Encapsulation of hydrophobically ion-paired teduglutide in nanoemulsions: Effect of anionic counterions. Food Chem 2025; 471:142774. [PMID: 39788016 DOI: 10.1016/j.foodchem.2025.142774] [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: 08/07/2024] [Revised: 12/11/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
This study presents a novel method for encapsulating the bioactive peptide teduglutide to enhance its oral bioavailability using O/W nanoemulsion (NE). Recombinant teduglutide (rTGT), produced in E. coli with 93 % purity, was hydrophobically modified through ion-pairing with phytic acid (PA) and sodium dodecyl sulfate (SDS). This approach increased encapsulation efficiency from 48.5 % to 87.5 % and 88.3 %, respectively. rTGT/SDS was incorporated within the core of lipid particles, whereas rTGT/PA was likely oriented on the surface. rTGT/SDS_NE exhibited smaller particle size, greater stability, and low cytotoxicity across all tested concentrations in HT-29 cells. Additionally, rTGT/SDS_NE achieved the highest upregulation of genes associated with intestinal function (VIL1, SGLT1, and GLUT2), although the differences were not statistically significant. These findings highlight the potential of the hydrophobic ion-pairing of rTGT with SDS and its encapsulation in nanoemulsion for efficient delivery of rTGT, suggesting promise for advancing oral peptide therapeutics.
Collapse
Affiliation(s)
- Youkyung Jeon
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Sung-Gun Kim
- Department of Biomedical Science, U1 University, Chungbuk 29131, Republic of Korea
| | - Kyeong-Ok Choi
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea.
| | - Jong-Tae Park
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
4
|
Shen Y, Tan X, Zhou L, Sun Z, Han M, Liang M, Xu D. An injectable in situ gel formed by ropivacaine with small lipid molecules to achieve long-term postoperative analgesia. Acta Biomater 2025:S1742-7061(25)00275-2. [PMID: 40246260 DOI: 10.1016/j.actbio.2025.04.033] [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: 12/18/2024] [Revised: 03/29/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Although local anesthetics (LA) are non-addictive and effective in managing postoperative pain, their short-term effects limit their clinical utility. In this study, we constructed an in situ gel injection formulation with stearic acid-ropivacaine hydrophobic ion-pair (HIP) and glycerol monoglyceride (SA-ROP-GMS) for multiday postoperative pain management. As an in situ gel, SA-ROP-GMS avoids the burst release effect common to water-soluble small molecule drugs in liposomes and hydrogels. Meanwhile, the hydrophobic ion-pair formed by ropivacaine and stearic acid contributes to the gel's stabilization and slow-release properties. In vivo evaluation of mouse models of pain demonstrated that the formulation provided multiday analgesia. And the absence of systemic toxicity of SA-ROP-GMS was verified by histological and blood biochemical studies. This study demonstrated the safety and efficacy of an injectable ropivacaine formulation based on in situ gel. The materials used in SA-ROP-GMS are safe and easy to obtain, and the preparation process is simple and fast, providing a convenient and effective strategy for the development of single-dose long-acting local anesthetic products, which is of great significance for postoperative pain management. STATEMENT OF SIGNIFICANCE: More than 80% of patients experience severe postoperative pain, and poor pain control reduces recovery satisfaction while increasing the risk of chronic pain. In this study, we constructed an injectable in situ gel using all-small-molecule excipients for slow-release drug delivery for the first time. We designed a method to form a lipid drug coupling between ropivacaine and stearic acid (SA-ROP HIP), which was then combined with glyceryl monostearate (SA-ROP-GMS). SA-ROP-GMS maintained analgesia for nearly 10 days in a single dose, significantly improving ease of administration and patient compliance. In addition, the auxiliary ingredients used in this study are biocompatible and inexpensive. This formulation follows the trend of regional anesthesia and provides a new solution for postoperative pain management.
Collapse
Affiliation(s)
- YiQing Shen
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Xin Tan
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Lin Zhou
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - ZiHan Sun
- Qingyun Experimental Middle School International Department, Suzhou 215235, PR China
| | - Min Han
- Department of Pharmacy, The Second Affifiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, PR China; Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China.
| | - Min Liang
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Department of Anesthesiology, Anesthesiology Research Institute, The first Affiliated Hospital of Fujian Medical University, Fuzhou 350005, PR China.
| | - DongHang Xu
- Department of Pharmacy, The Second Affifiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, PR China.
| |
Collapse
|
5
|
To D, Steinbring C, Weber LI, Ricci F, Polidori I, Postina A, Hartl M, Bernkop-Schnürch A. Design of lipid-based formulations for oral delivery of a BASP1 peptide targeting MYC-dependent gastrointestinal cancer cells. J Control Release 2025; 382:113677. [PMID: 40185335 DOI: 10.1016/j.jconrel.2025.113677] [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: 01/11/2025] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
HYPOTHESIS Oral delivery of the proliferation-inhibiting brain acid-soluble protein 1 effector domain peptide (Myr-NT) towards MYC-dependent gastrointestinal tumors can be achieved by forming hydrophobic ion pairs (HIPs) and incorporating them into lipid-based formulations. EXPERIMENTS Hydrophobic ion pairing of fluorescently-labelled Myr-NT (Myr-NT-TAMRA) was performed, increase in lipophilicity was assessed, and the most promising HIP was subsequently incorporated into a nanoemulsion. Stability of the peptide towards degradation by trypsin was evaluated. Anti-proliferative and anti-invasive measurements were performed upon application of the loaded nanoemulsion on various MYC-dependent human cancer cell lines. Cellular uptake and molecular effect were complementary investigated by confocal laser scanning microscopy (CLSM) and by immunoblot analyses, respectively. FINDINGS HIPs of Myr-NT-TAMRA exhibited up to 10,000-fold increase in lipophilicity, thereby enabling incorporation into a nanoemulsion. The formulation significantly boosted stability of incorporated peptide towards enzymatic degradation by trypsin. Furthermore, anti-proliferative measurements on human cancer cell lines revealed superior biological activity of the loaded nanoemulsion compared to the native peptide particularly in lymphoma cells, but also in colorectal cancer cells. Thereby, a correlation with proliferation inhibition as well as differences in MYC protein expression were observed. Finally, CLSM imaging revealed up to 15-fold increased cellular uptake of Myr-NT-TAMRA from the nanoemulsion confirming efficient intracellular delivery of the peptide. CONCLUSION Myr-NT can be efficiently delivered into intestinal tumor cells using orally administered lipid-based formulations.
Collapse
Affiliation(s)
- Dennis To
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Christian Steinbring
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Leonie I Weber
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Fabrizio Ricci
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; Thiomatrix Forschungs- und Beratungs GmbH, Trientlgasse 65, 6020 Innsbruck, Austria
| | - Ilaria Polidori
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Annika Postina
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Markus Hartl
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Andreas Bernkop-Schnürch
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| |
Collapse
|
6
|
Lee JJ, Choi M, Jeon Y, Khanal D, Lee J, Kim D, Chan HK, Hwang SJ. Physicochemical characterization and nanochemical analysis of ciprofloxacin hydrophobic ion Pairs for enhanced encapsulation in PLGA nanoparticle. Int J Pharm 2025; 672:125314. [PMID: 39909154 DOI: 10.1016/j.ijpharm.2025.125314] [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: 11/22/2024] [Revised: 01/14/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
This study investigates the physicochemical transformation of ciprofloxacin (CIP) through hydrophobic ion pairing with five counter ions-sodium oleate, sodium laurate, sodium caprate, disodium pamoate, and sodium deoxycholate-to enhance compatibility with hydrophobic Poly (lactic-co-glycolic acid) (PLGA) nanoparticles. Complexation efficiencies (CE) reached up to 92.26 %, with ciprofloxacin pamoate (CIP-PAM) achieving over 90 % CE at a 1:0.5 M ratio. Differential scanning calorimetry (DSC) and X-ray diffraction (XRD) analyses showed reduced crystallinity across all complexes, with CIP-PAM exhibiting an amorphous form. Optical photothermal infrared spectroscopy (O-PTIR) confirmed uniform complexation within particles, while CIP-PAM displayed a broad peak and weak intensity in the 900-1300 cm-1 region, supporting its amorphous nature. Log P values demonstrated increased hydrophobicity for all complexes, with ciprofloxacin oleate (CIP-OLE) showing a 93-fold increase (p < 0.001). In vitro dissociation patterns varied: CIP-OLE maintained steady release in DW (49.7 %) and PBS (32.3 %) over 48 h, whereas CIP-PAM exhibited strong stability in DW (25.2 %) and a contrasting 68.1 % release in PBS, highlighting solvent-dependent dissociation behaviors. PLGA nanoparticles prepared via S/O/W achieved particle sizes under 200 nm, with CIP-PAM showing the highest encapsulation efficiency (63.02 % vs 17.21 % (CIP)). These findings underscore the importance of counter ion selection to optimize CIP compatibility with hydrophobic carriers, providing a basis for improved drug loading of hydrophilic antibiotics.
Collapse
Affiliation(s)
- Jong-Ju Lee
- College of Pharmacy & Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Minji Choi
- College of Pharmacy & Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Yuim Jeon
- College of Pharmacy & Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Dipesh Khanal
- Advanced Drug Delivery Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW 2006, Australia
| | - Juseung Lee
- College of Pharmacy & Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Dowoong Kim
- College of Pharmacy & Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Hak-Kim Chan
- Advanced Drug Delivery Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW 2006, Australia.
| | - Sung-Joo Hwang
- College of Pharmacy & Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea.
| |
Collapse
|
7
|
Venkatasubramanian R, Al-Maghrabi PM, Alavi O, Lind T, Sassene PJ, Kirkensgaard JJK, Mota-Santiago P, Rades T, Müllertz A. Design, evaluation, and in vitro-in vivo correlation of self-nanoemulsifying drug delivery systems to improve the oral absorption of exenatide. J Control Release 2025; 379:440-451. [PMID: 39805462 DOI: 10.1016/j.jconrel.2025.01.013] [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: 04/28/2024] [Revised: 12/04/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
The ability to predict the absorption of exenatide (Ex), a GLP-1 analogue, after oral dosing to rats in self-nanoemulsifying drug delivery systems (SNEDDS), using in vitro methods, was assessed. Ex was complexed with soybean phosphatidylcholine (SPC) prior to loading into SNEDDS. A design of experiments (DoE) approach was employed to develop SNEDDS incorporating medium-chain triglycerides (MCT), medium-chain mono- and diglycerides (MGDG), Kolliphor® RH40, and monoacyl phosphatidylcholine. SNEDDS with higher proportions of MGDG and Kolliphor® RH40 demonstrated a 9-fold reduction in droplet size (230 to 26 nm), a 1.5-fold decrease in lipolysis (0.23 to 0.34 mmol of FFA), and a 2-fold enhancement in exenatide protection against proteolysis (73 % to 38 %) compared to those with higher MCT content. Permeability studies in Caco-2 cells showed that SNEDDS with higher proportion of MGDG displayed a 40-fold increase in apparent permeability of FD4, when compared to SNEDDS with higher proportion of MCT. An oral gavage study in rats revealed a 1.8-fold higher absorption of Ex in SNEDDS with a higher proportion of MGDG and Kolliphor®RH40 compared to SNEDDS with higher MCT. These results establish a clear in vitro-in vivo correlation, demonstrating that the selected in vitro methods effectively differentiated formulations with high and low absorption of exenatide after oral dosing in rats.
Collapse
Affiliation(s)
| | - Passant M Al-Maghrabi
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark.
| | - Oscar Alavi
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark.
| | - Tania Lind
- Bioneer A/S, Kogle Allé 2, Hørsholm 2970, Denmark.
| | | | - Jacob J K Kirkensgaard
- Department of Food Science, University of Copenhagen, Frederiksberg C 1958, Denmark; Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, Copenhagen 2100, Denmark.
| | | | - Thomas Rades
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark.
| | - Anette Müllertz
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark; Bioneer A/S, Kogle Allé 2, Hørsholm 2970, Denmark.
| |
Collapse
|
8
|
Jindal A, Kumar Sharma P, Kumar A. Self-nanoemulsifying drug delivery system (SNEDDS) as nano-carrier framework for permeability modulating approaches of BCS class III drug. J Drug Target 2025:1-21. [PMID: 40013328 DOI: 10.1080/1061186x.2025.2469751] [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: 10/30/2024] [Revised: 02/10/2025] [Accepted: 02/15/2025] [Indexed: 02/28/2025]
Abstract
The purpose of this review is to focus on the Self-Nanoemulsifying Drug Delivery System (SNEDDS) as an effective nanocarrier framework for permeability modulating approaches (PMA) of BCS class-III drugs and its challenges. Present review updates the recent trends in the SNEDDS research where it was employed as a cargo carrier for PMA and challenges. Patient compliance, ease of administration and non-invasiveness mode are non-trivial aspects in the oral administration of drugs. However, low aqueous solubility and impaired permeability are two prominent challenges resulting poor absorption of a drug. SNEDDS emerged as a dual nano-carrier system to enable nanodispersion of PMA via e.g. ion-pairing, phospholipid-complex, surfactant-drug interaction, loading of non-ionizable, free drug bases etc. These PMAs are embedded within the lipid phase of SNEDDS to produce nanosizing, enhancing nano-dispersibility via micellization/solubilization mechanism owing to its ternary components. Review highlights different PMAs employed in bioavailability enhancement of BCS class-III. It covers excipients employed in SNEDDS-loaded PMA, strategies for the hydrophobic transformation of water-soluble drugs for BCS class-III drugs. SNEDDS as a nano-cargo system for PMAs significantly modifies the bioavailability of BCS class-III drugs. SNEDDS is an isotropic-mixture of oil, surfactant:co-surfactant offers multipoint access to PMA loading and produces nano-dispersion in aqueous-medium.
Collapse
Affiliation(s)
- Amulya Jindal
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh, India
| | - Pankaj Kumar Sharma
- Department of Pharmacy, Raj Kumar Goel Institute of Technology, Ghaziabad, Uttar Pradesh, India
| | - Anoop Kumar
- Department of Pharmacy, Dr. K. N. Modi Institute of Pharmaceutical Education and Research, Modinagar, Uttar Pradesh, India
| |
Collapse
|
9
|
Haye L, Pini F, Soro LK, Knighton RC, Fayad N, Benard M, Gagliazzo F, Light ME, Natile MM, Charbonnière LJ, Hildebrandt N, Reisch A. Molecular Upconversion Nanoparticles for Live-Cell Imaging. ACS NANO 2025; 19:7178-7187. [PMID: 39937164 DOI: 10.1021/acsnano.4c16762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Precise molecular control has become a highly attractive feature to develop the next generation of upconversion materials for autofluorescence-free deep tissue imaging. However, in aqueous environments, upconversion molecules are orders of magnitude dimmer than inorganic upconversion nanoparticles, thereby strongly limiting their applicability to bioimaging. By encapsulating ca. 1,900 upconversion molecules into sub-40 nm polymer nanoparticles, we show that molecular precision and nanomaterial brightness can be combined into a new type of hybrid nanomaterial. The brightness of these molecular upconversion nanoparticles (UCMol-NPs) is almost on par with widely used inorganic upconversion nanoparticles, permitting the experimental demonstration of live-cell imaging with UCMol-NPs, an important step toward advancing molecular upconversion into the application era. Fabrication, characterization, and modeling of UCMol-NPs with various sizes and loadings reveal that significant brightness enhancement is possible. This will be paramount for advancing upconversion beyond the current limits of inorganic nanoparticles and translating them into clinical applications.
Collapse
Affiliation(s)
- Lucie Haye
- Université de Strasbourg, CNRS, Laboratoire de Bioimagerie et Pathologies UMR 7021, Strasbourg 67000, France
- McMaster University, Department of Engineering Physics, Hamilton, ON M8S 4K1, Canada
| | - Federico Pini
- Istituto di Chimica della Materia Condensata e Tecnologie per l'Energia (ICMATE), Consiglio Nazionale delle Ricerche (CNR), Padova, PD 35131, Italy
- Dipartimento di Scienze Chimiche, Università di Padova, Padova 35131, Italy
- nanoFRET.com, Laboratoire COBRA (Chimie Organique, Bioorganique, Réactivité et Analyse), Université de Rouen Normandie, CNRS, INSA76821, Rouen 76000, France
| | - Lohona Kevin Soro
- Equipe de Synthèse Pour l'Analyse (SynPA), Institut Pluridisciplinaire Hubert Curien (IPHC), UMR 7178, CNRS, Université de Strasbourg, ECPM, Strasbourg 67087, Cedex, France
| | - Richard C Knighton
- Equipe de Synthèse Pour l'Analyse (SynPA), Institut Pluridisciplinaire Hubert Curien (IPHC), UMR 7178, CNRS, Université de Strasbourg, ECPM, Strasbourg 67087, Cedex, France
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, UK
| | - Nour Fayad
- nanoFRET.com, Laboratoire COBRA (Chimie Organique, Bioorganique, Réactivité et Analyse), Université de Rouen Normandie, CNRS, INSA76821, Rouen 76000, France
| | - Magalie Benard
- PRIMACEN, Université de Rouen Normandie, INSERM, CNRS, HeRacLeS US51 UAR2026, Rouen 76000, France
| | - Francesco Gagliazzo
- Istituto di Chimica della Materia Condensata e Tecnologie per l'Energia (ICMATE), Consiglio Nazionale delle Ricerche (CNR), Padova, PD 35131, Italy
- Dipartimento di Scienze Chimiche, Università di Padova, Padova 35131, Italy
| | - Mark E Light
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, UK
| | - Marta Maria Natile
- Istituto di Chimica della Materia Condensata e Tecnologie per l'Energia (ICMATE), Consiglio Nazionale delle Ricerche (CNR), Padova, PD 35131, Italy
- Dipartimento di Scienze Chimiche, Università di Padova, Padova 35131, Italy
| | - Loïc J Charbonnière
- Equipe de Synthèse Pour l'Analyse (SynPA), Institut Pluridisciplinaire Hubert Curien (IPHC), UMR 7178, CNRS, Université de Strasbourg, ECPM, Strasbourg 67087, Cedex, France
| | - Niko Hildebrandt
- McMaster University, Department of Engineering Physics, Hamilton, ON M8S 4K1, Canada
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Andreas Reisch
- Université de Strasbourg, CNRS, Laboratoire de Bioimagerie et Pathologies UMR 7021, Strasbourg 67000, France
- Biomaterials and Bioengineering, Inserm UMRS 1121, CNRS EMR 7003, Centre de Recherche en Biomédecine de Strasbourg, Université de Strasbourg, Strasbourg 67000, France
| |
Collapse
|
10
|
Sim YS, Wong LC, Yeoh SC, Almashhadani A, Alrimawi BH, Goh CF. Skin penetration enhancers: Mechanistic understanding and their selection for formulation and design. Drug Deliv Transl Res 2025:10.1007/s13346-025-01809-9. [PMID: 39982640 DOI: 10.1007/s13346-025-01809-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2025] [Indexed: 02/22/2025]
Abstract
The skin functions as a formidable barrier, particularly the stratum corneum, effectively restricting the penetration of most substances, including therapeutic agents. To circumvent this barrier, skin penetration enhancers (SPEs) are frequently employed to transiently increase skin permeability, facilitating drug absorption without causing irritation or damage. Despite advancements in dermal formulation development, a deeper understanding of the fundamental science underpinning drug delivery via SPEs remains essential. This review delivers a critical update on conventional SPEs, exploring their mechanisms in promoting drug permeation across the skin. In addition to offering an overview of percutaneous drug delivery, we examine the prevailing theories on how SPEs enhance drug transport. Furthermore, we address the intricate interplay between SPEs, drugs and the skin, providing valuable insights into how the molecular properties and permeation behaviours of SPEs influence their efficacy. This comprehensive review aims to support the ongoing development of optimised drug delivery systems for dermal applications by elucidating the complexities and challenges involved in using SPEs effectively.
Collapse
Affiliation(s)
- Yee Shan Sim
- Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Li Ching Wong
- Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Soo Chin Yeoh
- Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Abdulsalam Almashhadani
- Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Bilal Harieth Alrimawi
- Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Choon Fu Goh
- Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia.
| |
Collapse
|
11
|
Elhassan E, Omolo CA, Gafar MA, Ismail EA, Ibrahim UH, Khan R, Lesouhaitier M, Kubes P, Govender T. Multifunctional hyaluronic acid-based biomimetic/pH-responsive hybrid nanostructured lipid carriers for treating bacterial sepsis. J Biomed Sci 2025; 32:19. [PMID: 39930418 PMCID: PMC11812216 DOI: 10.1186/s12929-024-01114-6] [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: 07/12/2024] [Accepted: 12/17/2024] [Indexed: 02/13/2025] Open
Abstract
INTRODUCTION The application of biomimetic and stimuli-responsive nanocarriers displays considerable promise in improving the management of bacterial sepsis and overcoming antimicrobial resistance. Therefore, the study aimed to synthesize a novel hyaluronic acid-lysine conjugate (HA-Lys) and to utilize the attributes of the synthesized HA-Lys with Tocopherol succinate (TS) and Oleylamine (OLA) in the formulation of multifunctional biomimetic pH-responsive HNLCs loaded with vancomycin (VCM-HNLCs), to combat bacterial sepsis. METHODS A novel hyaluronic acid-lysine conjugate (HA-Lys) was synthesized and characterized using FTIR and 1H NMR spectroscopy. Vancomycin-loaded hybrid nanosystems (VCM-HNLCs) were prepared through hot homogenization ultrasonication and evaluated for particle size, polydispersity index (PDI), zeta potential (ZP), and encapsulation efficiency (EE%). In vitro biocompatibility was assessed via MTT assay and red blood cell hemolysis test. The binding affinity to TLR2 and TLR4 was measured using microscale thermophoresis (MST). Drug release was evaluated using the dialysis bag method. Antimicrobial activity against MRSA and efflux pump inhibition were also determined. Efficacy was demonstrated in an MRSA-induced sepsis mice model. RESULTS The VCM-HNLCs, produced via hot homogenization ultrasonication, exhibited particle size (PS), polydispersity index (PDI), zeta potential (ZP), and encapsulation efficiency (EE%) of 110.77 ± 1.692 nm, 0.113 ± 0.022, - 2.92 ± 0.210 mV, and 76.27 ± 1.200%, respectively. In vitro, biocompatibility was proven by hemolysis and cytotoxicity studies. The VCM-HNLCs demonstrated targetability to human Toll-like receptors (TLR 2 and 4) as validated by microscale thermophoresis (MST). VCM-HNLCs showed a twofold reduction in MIC values at physiological pH compared to the bare VCM against S. aureus and MRSA for 48 h. While at pH 6.0, MIC values were reduced by fourfold in the first 24 h and by eightfold in the subsequent 48 and 72 h against tested strains. Furthermore, VCM-HNLCs showed inhibitory effects against MRSA efflux pumps, reactive oxygen species (ROS), and lipopolysaccharide (LPS)-induced hyperinflammation. In an MRSA-induced sepsis mice model, VCM-HNLCs demonstrated superior efficacy compared to free VCM, significantly eliminated bacteria and improved survival rates. CONCLUSIONS Overall, these results highlight the potential of VCM-HNLCs as novel multifunctional nanocarriers to combat antimicrobial resistance (AMR) and enhance sepsis outcomes.
Collapse
Affiliation(s)
- Eman Elhassan
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, P. O. Box 14634-00800, Nairobi, Kenya.
| | - Mohammed A Gafar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
- Department of Pharmaceutics, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Eman A Ismail
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
- Department of Pharmaceutics, Faculty of Pharmacy, University of Gezira, Wad Medani, Sudan
| | - Usri H Ibrahim
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Rene Khan
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Mathieu Lesouhaitier
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AL, Canada
| | - Paul Kubes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AL, Canada
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
| |
Collapse
|
12
|
Subedi L, Bamjan AD, Phuyal S, Shim JH, Cho SS, Seo JB, Chang KY, Byun Y, Kweon S, Park JW. An oral liraglutide nanomicelle formulation conferring reduced insulin-resistance and long-term hypoglycemic and lipid metabolic benefits. J Control Release 2025; 378:637-655. [PMID: 39709071 DOI: 10.1016/j.jconrel.2024.12.039] [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: 08/19/2024] [Revised: 11/20/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Type 2 diabetes is a chronic disease characterized by insulin resistance and often worsened by obesity. Effective management involves the use of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) to assist with glycemic control and weight management. However, these drugs must be administered subcutaneously due to their low oral bioavailability. We developed an oral liraglutide (LRG) formulation by electrostatic complexation of GLP-1 RA with bile acid derivatives and nanomicelle (NM) formation, with non-ionic surfactant n-dodecyl-β-d-maltoside (DDM). The optimized formulation, LDD[1:2:4]-NM, had a mean particle size of 75.9 ± 5.60 nm and a permeability 1347 % higher than that of unformulated LRG when tested in Caco-2/HT29-MTX-E12 cell monolayers. In rats, oral bioavailability was 4.63-fold higher than that of unformulated LRG (1.11 ± 0.20 % vs. 5.14 ± 0.63 %). The absorption mechanism included clathrin-mediated endocytosis, macropinocytosis, and an ASBT-mediated pathway. A 12-week oral treatment consisting of a daily dose of 20 mg LDD[1:2:4]-NM/kg significantly reduced glycohemoglobin levels, a marker of diabetic control, and the HOMA-IR index, a marker of insulin resistance. The weight of epididymal and inguinal white adipose tissue and brown adipose tissue (BAT) was also reduced. Moreover, LDD[1:2:4]-NM had a greater impact on BAT activation, pro-inflammatory gene expression, and lipid metabolism than subcutaneous LRG. This study showed that an oral NM formulation can efficiently deliver LRG. Long-term treatment led to improved hyperglycemic effects, insulin resistance, and modulated lipid metabolism. LDD[1:2:4]-NM is thus a promising oral therapeutic option for the management of type 2 diabetes, potentially transforming treatment paradigms based on the availability of a more convenient administration route.
Collapse
Affiliation(s)
- Laxman Subedi
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Arjun Dhwoj Bamjan
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Susmita Phuyal
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Jung-Hyun Shim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Seung-Sik Cho
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Jong Bae Seo
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | | | - Youngro Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seho Kweon
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Jin Woo Park
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea.
| |
Collapse
|
13
|
Sasse R, Carpenter N, Simpkins CO. Selective nitric oxide redistribution by phospholipid nanoparticles: A novel strategy to mitigate massive nitric oxide release and prevent reperfusion injury in septic shock. Free Radic Biol Med 2025; 227:276-281. [PMID: 39645204 DOI: 10.1016/j.freeradbiomed.2024.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Nitric oxide plays a critical role in regulating vascular tone, but excessive nitric oxide release during septic shock results in hypotension due to excessive vasodilation and the formation of toxic free radicals. VBI-S is a phospholipid nanoparticle based fluid composed of lipid bilayers formed primarily by phosphatidylcholine and micelles of soybean oil encapsulated by a monolayer of phosphatidylcholine. These nanoparticles offer a novel solution by absorbing and redistributing nitric oxide and nitrite, potentially mitigating the harmful effects of excessive nitric oxide in sepsis. This paper proposes a mechanism in which VBI-S not only redistributes nitric oxide but also reduces ischemia-reperfusion injury by limiting the production and availability of reactive species. VBI-S captures nitric oxide and nitrite in areas of high concentration and redistributes them in low-nitric oxide environments, primarily within oxygen-deprived tissues. Nitrite then contributes to nitric oxide regeneration in hypoxic microvasculature via various reduction pathways, thereby improving tissue perfusion and minimizing oxidative stress. Preliminary studies suggest that nitrite may also decrease reactive species production, primarily superoxide, through the inhibition of mitochondrial complex I. Additionally, the lipid composition of VBI-S is rich in poly and monounsaturated fatty acids which allows VBI-S to act as a substrate for peroxidation via peroxynitrite. Therefore, VBI-S acts as a decoy target thereby protecting cellular membranes from oxidative damage caused by reactive species. These findings position VBI-S as a promising therapeutic agent, offering both nitric oxide regulation and protection against hypotension and toxic free radicals in septic shock patients. Further research is necessary to fully elucidate the molecular pathways and optimize its clinical application.
Collapse
Affiliation(s)
- Ryan Sasse
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA.
| | - Nathan Carpenter
- Department of Surgery, University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Cuthbert O Simpkins
- Department of Surgery, University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| |
Collapse
|
14
|
Preedanorawut P, Chatchawankanphanich O, Yiamsawas D, Crespy D. Controlled Release of Hydrophilic Drug from Hollow Nanodots. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409112. [PMID: 39888222 DOI: 10.1002/smll.202409112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/14/2025] [Indexed: 02/01/2025]
Abstract
Here the challenge of limited encapsulation efficiency of ionizable hydrophilic molecules in silica materials is addressed. Two effective strategies are showcased that allow high encapsulation efficiency of salicylic acid, while simultaneously maintaining the morphology and particle size of silica nanocapsules. These promising approaches involve the formation and encapsulation of a prodrug or the complexation of the hydrophilic payload with a hydrophobic moiety to form a complex that is dissociated in acidic conditions. Well-defined core-shell silica nanocapsules with a diameter of 6 nm are obtained and exhibited an encapsulation efficiency of over 90%. High amounts of salicylic acid are released in acidic conditions from silica nanocapsules entrapping the prodrug or the complex, leading to pH-responsive characteristics. This work demonstrates promising strategies for the encapsulation and the controlled release of hydrophilic fertilizers, pesticides or drugs.
Collapse
Affiliation(s)
- Patitta Preedanorawut
- Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, 21210, Thailand
| | - Orawan Chatchawankanphanich
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Doungporn Yiamsawas
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Daniel Crespy
- Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, 21210, Thailand
| |
Collapse
|
15
|
Chen Y, Xu M, Pan J, Liao Y, Na J, Li P, Sun Y, Yu S, Zhao Y, Hu H. Moxifloxacin-Loaded Polymeric Nanoparticles for Overcoming Multidrug Resistance in Chronic Pulmonary Infections Caused by Pseudomonas aeruginosa. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5695-5709. [PMID: 39804842 DOI: 10.1021/acsami.4c14991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) infections are increasingly challenging due to their propensity to form biofilms and low outer membrane permeability, especially in chronically infected patients with thick mucus. P. aeruginosa exhibits multiple drug resistance mechanisms, making it one of the most significant global public health threats. In this study, we found that moxifloxacin (MXC) and antibacterial peptides (ε-poly-l-lysine, ε-PLL) exhibited a synergistic effect against multidrug-resistant P. aeruginosa (MDR-P. aeruginosa). MXC was combined with ε-PLL to prepare lipase-responsive nanoparticles (MCIP/(PEG-PCL)/PLL NPs) with a weakly negative charge. The weakly negatively charged MCIP/(PEG-PCL)/PLL NPs demonstrated remarkable mucus and biofilm penetration capabilities, thereby overcoming one of the adaptive drug resistance mechanisms. MCIP/(PEG-PCL)/PLL NPs improved the outer and inner membrane permeability and inhibited the expression of the efflux pump MexAB-OprM gene in MDR-P. aeruginosa, thereby overcoming mechanisms of both intrinsic and acquired drug resistance. Meanwhile, the nanoparticles demonstrated an ability to reduce repeated infections with MDR-P. aeruginosa. Additionally, the bacterial burden in the lungs of mice treated with MCIP/(PEG-PCL)/PLL NPs was significantly lower than that in the MXC group, resulting in a 99% clearance rate. Notably, MCIP/(PEG-PCL)/PLL NPs showed no toxicity toward BEAS-2B cells or RAW 267.4 cells, nor did they adversely affect pulmonary function or major organs. This study demonstrated the potential of the nanodrug delivery system composed of the antibiotic moxifloxacin and the antibacterial peptide ε-PLL in addressing the clinical challenges of treating chronic pulmonary infections caused by MDR-P. aeruginosa.
Collapse
Affiliation(s)
- Yujun Chen
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Mao Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jieyi Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuan Liao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Pengyu Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yingying Sun
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shihui Yu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yongxiang Zhao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Haiyan Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
16
|
Gray M, de Janon A, Seeler M, Heller WT, Panoskaltsis N, Mantalaris A, Champion JA. Intracellular Biomacromolecule Delivery by Stimuli-Responsive Protein Vesicles Loaded by Hydrophobic Ion Pairing. ACS OMEGA 2025; 10:2628-2639. [PMID: 39895718 PMCID: PMC11780410 DOI: 10.1021/acsomega.4c07666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/05/2024] [Accepted: 12/20/2024] [Indexed: 02/04/2025]
Abstract
Proteins can perform ideal therapeutic functions. However, their large size and significant surface hydrophilicity and charge prohibit them from reaching intracellular targets. These chemical features also render them poorly encapsulated by nanoparticles used for intracellular delivery. In this work, a novel combination of protein vesicles and hydrophobic ion pairing (HIP) was used to load protein cargo and achieve cytosolic delivery to overcome the limitations of previous protein vesicle properties. Protein vesicles are thermally self-assembling nanoparticles made from elastin-like polypeptide (ELP) fused to an arginine-rich leucine zipper and a globular protein fused to a glutamate-rich leucine zipper. To impart stimuli-responsive disassembly, physiological stability, and small size, the ELP sequence was modified to include histidine and tyrosine residues. HIP was used to load and release protein cargo requiring endosomal escape for cytosolic function. HIP vesicles enabled delivery of cytochrome c, a cytosolically active protein, and a significant reduction in viability in both a traditional two-dimensional (2D) human cancer cell line culture and a biomimetic three-dimensional (3D) organoid model of acute myeloid leukemia. By examining the uptake of positively and negatively charged fluorescent protein cargos loaded by HIP, this work revealed the necessity of HIP for cytosolic cargo delivery and how HIP loading influences protein vesicle self-assembly and disassembly using microscopy, small-angle X-ray scattering, and nanoparticle tracking analysis. HIP protein vesicles have the potential to broaden the use of intracellular proteins as therapeutics for various diseases and extend protein vesicles to deliver other biomacromolecules, as the strategy developed here resulted in the first cytosolic protein cargo delivery using protein vesicles.
Collapse
Affiliation(s)
- Mikaela
A. Gray
- Chemical
and Biomolecular Engineering, Georgia Institute
of Technology Atlanta, Georgia 30332-0002, United States
| | - Alejandro de Janon
- Biomedical
Systems Engineering Laboratory, Georgia
Institute of Technology, Atlanta, Georgia 30332-0002, United States
| | - Michelle Seeler
- School
of Biological Sciences, Georgia Institute
of Technology, Atlanta, Georgia 30332-0002, United States
| | - William T. Heller
- Neutron
Scattering Division, Oak Ridge National
Laboratory, Oak Ridge, Tennessee 37831-6473, United States
| | - Nicki Panoskaltsis
- Biomedical
Systems Engineering Laboratory, Georgia
Institute of Technology, Atlanta, Georgia 30332-0002, United States
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
- Department
of Haematology, St. James’s Hospital, Dublin 8, Ireland
| | - Athanasios Mantalaris
- Biomedical
Systems Engineering Laboratory, Georgia
Institute of Technology, Atlanta, Georgia 30332-0002, United States
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
- National
Institute for Bioprocessing Research and Training, Dublin A94 X099, Ireland
| | - Julie A. Champion
- Chemical
and Biomolecular Engineering, Georgia Institute
of Technology Atlanta, Georgia 30332-0002, United States
| |
Collapse
|
17
|
Lindner S, Ricci F, Sandmeier M, Holm R, Michalowski CB, Washburn N, Sun D, Di Pretoro G, Bernkop-Schnürch A. Optimizing hydrophilic drug incorporation into SEDDS using dry reverse micelles: a comparative study of preparation methods. Drug Deliv Transl Res 2025:10.1007/s13346-024-01787-4. [PMID: 39821869 DOI: 10.1007/s13346-024-01787-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
AIM It was the aim of this study to compare two different dry reverse micelle (RM) preparation methods for the incorporation of hydrophilic drugs into oral self-emulsifying drug delivery systems (SEDDS). METHODS Cationic ethacridine lactate, anionic fluorescein sodium salt and the antibiotic peptide bacitracin were solubilized in RM containing sodium docusate, soy phosphatidylcholine and sorbitan monooleate in highly lipophilic oils such as squalane. In the dry addition (DA) method, drugs were directly added to empty RM in their powder form. In the organic solvent-aided (OS) method, drugs were pre-dissolved in ethanol or 2-propanol, which were then evaporated to form loaded dry RM. RESULTS RM with sorbitan monooleate prepared via the DA method yielded up to 2.7-fold higher solubility only for bacitracin compared to the OS method. In contrast, OS-RM with sodium docusate and soy phosphatidylcholine exhibited significantly higher drug solubilities, achieving up to 109-fold, 44-fold and 97-fold increase for ethacridine, fluorescein and bacitracin, respectively. For all model drugs, the logDlipophilic phase/water was highest for RM comprising sorbitan monooleate, with a slight increase for OS-RM. This was consistent with the release profiles from SEDDS, showing an enhanced retention when loaded with OS-RM. While DA-RM showed no significant difference in cellular uptake, it was 1.6-fold higher in OS-RM loaded squalane-based SEDDS. CONCLUSION The DA method is an easier approach for incorporating hydrophilic drugs into dry RM. However, the OS method presents a more promising alternative as it significantly enhanced the solubility and retention of these drugs in highly lipophilic formulations.
Collapse
Affiliation(s)
- Sera Lindner
- Thiomatrix Forschungs- und Beratungs GmbH, Trientlgasse 65, 6020, Innsbruck, Austria
| | - Fabrizio Ricci
- Thiomatrix Forschungs- und Beratungs GmbH, Trientlgasse 65, 6020, Innsbruck, Austria
| | - Matthias Sandmeier
- Thiomatrix Forschungs- und Beratungs GmbH, Trientlgasse 65, 6020, Innsbruck, Austria
| | - René Holm
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
| | - Cecilia Bohns Michalowski
- Pharmaceutical Product Development & Supply, Johnson & Johnson Innovative Medicine, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Nathaniel Washburn
- Immunology Discovery, Johnson & Johnson Innovative Medicine, 301 Binney St, Cambridge, MA, 02141, USA
| | - Dajun Sun
- Pharmaceutical Product Development & Supply, Johnson & Johnson Innovative Medicine, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Giustino Di Pretoro
- Drug Product Development & Delivery, Johnson & Johnson Innovative Medicine, 347 Phoenixville Pike, Malvern, PA, 19355, USA
| | | |
Collapse
|
18
|
Dasaro SR, Singh A, Vlachos P, Ristroph KD. Mechanistic insights into how mixing factors govern polyelectrolyte-surfactant complexation in RNA lipid nanoparticle formulation. J Colloid Interface Sci 2025; 678:98-107. [PMID: 39182390 DOI: 10.1016/j.jcis.2024.08.150] [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: 04/22/2024] [Revised: 08/04/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
HYPOTHESIS Lipid nanoparticle self-assembly is a complex process that relies on ion pairing between nucleic acids and hydrophobic cationic lipid counterions for encapsulation. The chemical factors influencing this process, such as formulation composition, have been the focus of recent research. However, the physical factors, particularly the mixing protocol, which directly modulates these chemical factors, have yet to be mechanistically examined using a reproducible mixing platform comparable to the industry standard. We here utilize Flash NanoPrecipitation (FNP), a scalable rapid mixing platform, to isolate and systematically investigate how mixing factors influence this complexation step, first by using a model polyelectrolyte-surfactant system and then generalizing to a typical RNA lipid nanoparticle formulation. EXPERIMENTS Aqueous polystyrene sulfonate (PSS) and cetrimonium bromide (CTAB) solutions are rapidly homogenized using reproducible FNP mixing and controlled flow rates at different stoichiometric ratios and total solids concentrations to form polyelectrolyte-surfactant complexes (PESCs). Then, key mixing factors such as total flow rate, inlet stream relative volumetric flow rate, and magnitude of flow fluctuation are studied using both this PESC system and an RNA lipid nanoparticle formulation. FINDINGS Fluctuations in flow as low as ± 5 % of the total flow rate are found to severely compromise PESC formation. This result is replicated in the RNA lipid nanoparticle system, which exhibited significant differences in size (132.7 nm vs. 75.6 nm) and RNA encapsulation efficiency (34.0 % vs. 82.8 %) under fluctuating vs. steady flow. We explain these results in light of the chemical variables isolated and studied; slow or nonuniform mixing generates localized concentration gradients that disrupt the balance between the hydrophobic and electrostatic forces that drive complex formation. These experiments contribute to our understanding of the complexation stage of lipid nanoparticle formation and provide practical insights into the importance of developing controlled mixing protocols in industry.
Collapse
Affiliation(s)
- Sophia R Dasaro
- Department of Agricultural and Biological Engineering, Purdue University, 225 S. University St., West Lafayette, IN 47907, USA
| | - Abhishek Singh
- Department of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA
| | - Pavlos Vlachos
- Department of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA
| | - Kurt D Ristroph
- Department of Agricultural and Biological Engineering, Purdue University, 225 S. University St., West Lafayette, IN 47907, USA.
| |
Collapse
|
19
|
Bapat P, Taylor LS. Impact of HPMCAS Grade on the Release of Weakly Basic Drugs from Amorphous Solid Dispersions. Mol Pharm 2025; 22:397-407. [PMID: 39704640 DOI: 10.1021/acs.molpharmaceut.4c00986] [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] [Indexed: 12/21/2024]
Abstract
Oppositely charged species can form electrostatic interactions in aqueous solution, and these may lead to reduced solubility of the interacting components. Herein, insoluble complex formation between the lipophilic weakly basic drugs, cinnarizine or loratadine, and the enteric polymer, hydroxypropyl methylcellulose acetate succinate (HPMCAS), was studied and used to better understand drug and polymer release from their corresponding amorphous solid dispersions (ASDs). Surface area normalized release experiments were performed at various pH conditions for three different grades of HPMCAS, LF, MF and HF, as well as their ASDs. Both polymer and drug release rates were measured for the ASDs. Complexation tendency was evaluated by measuring the extent of polymer loss from the aqueous phase in the presence of the drug. Results showed that release from ASDs with HPMCAS-LF was less impacted by the presence of a cationic form of the drug than ASDs prepared with the HF grade. Furthermore, an increase in pH, leading to a reduction in the extent of ionized drug also led to an improvement in release rate. These observations provide a baseline to understand the role of drug-polymer electrostatic interactions on release from ASDs formulated with HPMCAS. Future studies should focus on adding complexity to media conditions by employing simulated intestinal fluids with solubilizing components.
Collapse
Affiliation(s)
- Pradnya Bapat
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
20
|
Schlosser CS, Rozek W, Mellor RD, Manka SW, Morris CJ, Brocchini S, Williams GR. A lipid-based delivery platform for thermo-responsive delivery of teriparatide. Int J Pharm 2024; 667:124853. [PMID: 39437847 DOI: 10.1016/j.ijpharm.2024.124853] [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: 09/19/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Teriparatide (and analogue peptides) are the only FDA approved anabolic treatments for osteoporosis. Current therapies are administered as a daily subcutaneous injection, which limits patient adherence and clinical efficacy. To achieve the desired anabolic effect, a controlled delivery system must ensure a pulsatile release profile over a prolonged period. Thermo-responsive formulations (e.g. liposomes) can undergo a temperature-related phase-transition which can allow active control of drug release. Herein, thermo-responsive liposomes were developed to permit control over teriparatide release rate through modulation of temperature. Entrapment of hydrophilic molecules, including peptides, within liposomes remains challenging due to the large volume of hydration. In this work, hydrophobic ion pairing was employed for the first time to enhance peptide entrapment within liposomes. The method resulted in a hydrophobic complex that achieved high teriparatide entrapment (>75 %) in sub-200 nm monodispersed liposomes. Hydrophobic ion pairing outperformed other entrapment approaches. Several liposomal formulations with transition temperatures between 38 and 50 °C were obtained by modulation of the phospholipid composition. In vitro assays demonstrated temperature-dependent release kinetics with faster rates of release observed at/above the transition temperature. The maintenance of biological activity of released teriparatide was demonstrated in a cell-based assay utilising the PTH1 receptor. Overall, this provides the first proof-of-concept of the suitability of thermo-responsive systems for pulsatile delivery of teriparatide and similar peptides.
Collapse
Affiliation(s)
- Corinna S Schlosser
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Wojciech Rozek
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Ryan D Mellor
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Szymon W Manka
- MRC Prion Unit at UCL, Institute of Prion Diseases, University College London, 33 Cleveland Street, London W1W 7FF, UK
| | - Christopher J Morris
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Steve Brocchini
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
21
|
Schlosser CS, Morris CJ, Brocchini S, Williams GR. Hydrophobic ion pairing as a novel approach to co-axial electrospraying of peptide-PLGA particles. Int J Pharm 2024; 667:124885. [PMID: 39491655 DOI: 10.1016/j.ijpharm.2024.124885] [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: 10/02/2024] [Revised: 10/26/2024] [Accepted: 10/26/2024] [Indexed: 11/05/2024]
Abstract
Electrospraying is a processing technique that has gained much interest to prepare polymeric particles. The technique operates at ambient temperature, thereby avoiding heat induced degradation of labile therapeutics (e.g. peptides and proteins). Exposure to organic solvents can be minimised by co-axial electrospraying through separation of core (aqueous) and shell (organic) solvents. However, aqueous solutions are often difficult to electrospray due to high surface tension. Immiscibility between the core-shell solvents creates a further process challenge. Herein, we describe for the first time the use of hydrophobic ion pairing (HIP) to encapsulate a polypeptide into polymeric particles prepared by co-axial electrospraying. Peptide ion pairs were prepared to incorporate a model peptide - teriparatide - into an organic solvent, permitting facile electrospraying while also protecting the peptide from denaturation. Teriparatide loaded PLGA particles were generated by electrospraying from aqueous or ethanolic peptide solutions (core). A PLGA solution in chloroform (with and without co-solvents) was employed as the shell solution. The aqueous core solution led to a teriparatide encapsulation efficiency of 79.2 ± 19.8 %, which was not significantly different from the ethanolic core (57.1 ± 14.5 %). When aqueous solutions were used the process lacked reproducibility, resulting in low process yields (61.3 ± 4.0 %). In contrast, when an organic core was used a dry powder bed was achieved with a yield of 102.2 ± 8.8 %. The peptide's integrity and biological functionality were retained after electrospraying as ion pairs, as evidenced in a cell-based PTH1 receptor binding assay.
Collapse
Affiliation(s)
- Corinna S Schlosser
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Christopher J Morris
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Steve Brocchini
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Gareth R Williams
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
22
|
Tsolaki E, Healy AM, Ferguson S. Development of polymer-encapsulated microparticles of a lipophilic API-IL and its lipid based formulations for enhanced solubilisation. Int J Pharm 2024; 667:124878. [PMID: 39491654 DOI: 10.1016/j.ijpharm.2024.124878] [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: 08/13/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Active Pharmaceutical Ingredient-Ionic liquids (API-ILs) have the potential to improve the bioavailability of BCS Class IV Drugs. However, the problematic physical handling properties of room temperature API-ILs have impaired clinical and commercial exploitation to date. Lipid-based formulations (LBFs) are used to improve the absorption of drugs with limited bioavailability. Nonetheless, LBFs face limitations such as low drug loading capacity and sub-par physical stability. A platform for transforming API-ILs into solid forms at high loadings via spray encapsulation with polymers has been developed and previously demonstrated for hydrophilic API-ILs. The current work demonstrates that this platform technology can be applied to a lipophilic API-IL of the BCS Class IV API, chlorpromazine, and to multi-component solutions comprising API-IL and a LBF. Furthermore, solidification of a type IIIB, liquid LBF was achieved via spray encapsulation with cellulose- and methacrylate- based polymers for the first time. The spray-encapsulated formulations had excellent physical handling properties, and successfully eluted the API-IL in aqueous media. The chlorpromazine release profiles from the API-IL, the API-IL containing LBF, and the solidified formulations, were evaluated in vitro using phosphate buffer (pH 6.8) and fasted state simulated intestinal fluid (FaSSIF). Spray-encapsulated formulations exhibited improved release profiles compared to the liquid formulations. Overall, these findings indicate that phase-separated, polymeric, solid formulations of liquid API forms represent a promising platform technology for developing oral solid dosage forms of poorly bioavailable drugs.
Collapse
Affiliation(s)
- Evangelia Tsolaki
- School of Chemical and Bioprocess Engineering, University College Dublin, Dublin 4, Ireland; SSPC, The SFI Research Centre for Pharmaceuticals, School of Chemical and Bioprocess Engineering, University College Dublin, Dublin 4, Ireland; EPSRC-SFI Centre for Doctoral Training in Transformative Pharmaceutical Technologies, Ireland.
| | - Anne Marie Healy
- EPSRC-SFI Centre for Doctoral Training in Transformative Pharmaceutical Technologies, Ireland; SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, Dublin 2, Ireland.
| | - Steven Ferguson
- School of Chemical and Bioprocess Engineering, University College Dublin, Dublin 4, Ireland; SSPC, The SFI Research Centre for Pharmaceuticals, School of Chemical and Bioprocess Engineering, University College Dublin, Dublin 4, Ireland; EPSRC-SFI Centre for Doctoral Training in Transformative Pharmaceutical Technologies, Ireland; National Institute for Bioprocessing Research and Training, 24 Foster's Ave, Belfield, Blackrock, Co. Dublin A94 X099, Ireland.
| |
Collapse
|
23
|
Lindner S, Ricci F, Holm R, Sun D, Washburn N, Michalowski CB, Di Pretoro G, Bernkop-Schnürch A. Hydrophobic ion pairing: lipophilicity improvement of anionic macromolecules by divalent cation mediated complex formation. Drug Deliv Transl Res 2024:10.1007/s13346-024-01760-1. [PMID: 39690372 DOI: 10.1007/s13346-024-01760-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2024] [Indexed: 12/19/2024]
Abstract
The aim of this study was to develop an alternative strategy to sufficiently increase the lipophilicity of anionic model macromolecules (MM) without the use of cationic counterions. Enoxaparin (ENO), insulin (INS) and poly-L-glutamic acid (PLG) were ion paired with anionic surfactants (sodium decanoate (DEC), sodium dodecyl sulfate (SDS), sodium stearate (SS) and sodium octadecyl sulfate (SOS)), mediated by divalent cations such as magnesium, calcium and zinc. Complexes were evaluated regarding their precipitation efficiency and logDn-butanol/water. SEDDS were developed, loaded with the complexes and characterized for their size and stability. Finally, payloads and logDSEDDS/release medium were determined. Divalent cation mediated ENO, INS and PLG complexes were successfully formed as underlined by high precipitation efficiencies above 90% in case of Zn2+-mediated complexes. Most pronounced increase in logDn-butanol/water was achieved for ENO-Zn2+-SOS (1.85), INS-Zn2+-SOS (0.80) and PLG-Zn2+-SS (0.48) providing suitable solubilities in commonly used SEDDS components. Developed SEDDS displayed droplet sizes below 200 nm without major changes after loading with MM complexes. Payloads up to 18.72 mg/ml could be achieved in developed SEDDS for ENO-Zn2+-SOS, and 2.44 mg/ml and 6.93 mg/ml for INS-Zn2+-SOS and PLG-Zn2+-SS, respectively. In general, highest lipophilicity enhancement and thus solubility in SEDDS was obtained with Zn2+-mediated complexes among the investigated cations and particularly with the highly negatively charged polysaccharide ENO. The formation of complexes between anionic MM and anionic surfactants mediated by divalent cations, substituting normally used cationic counterions exhibiting higher toxicity, offers a promising alternative to enhance their lipophilicity for oral drug delivery.
Collapse
Affiliation(s)
- Sera Lindner
- Thiomatrix Forschungs- Und Beratungs GmbH, Trientlgasse 65, 6020, Innsbruck, Austria
| | - Fabrizio Ricci
- Thiomatrix Forschungs- Und Beratungs GmbH, Trientlgasse 65, 6020, Innsbruck, Austria
| | - René Holm
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
| | - Dajun Sun
- Pharmaceutical Product Development & Supply, Johnson & Johnson Innovative Medicine, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Nathaniel Washburn
- Immunology Discovery, Johnson & Johnson Innovative Medicine, 301 Binney St, Cambridge, MA, 02141, USA
| | - Cecilia Bohns Michalowski
- Pharmaceutical Product Development & Supply, Johnson & Johnson Innovative Medicine, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Giustino Di Pretoro
- Drug Product Development & Delivery, Johnson & Johnson Innovative Medicine, 347 Phoenixville Pike, Malvern, PA, 19355, USA
| | | |
Collapse
|
24
|
Hussaini IM, Sulaiman AN, Abubakar SC, Abdulazeez TM, Abdullahi MM, Sulaiman MA, Madika A, Bishir M, Muhammad A. Unveiling the arsenal against antibiotic resistance: Antibacterial peptides as broad-spectrum weapons targeting multidrug-resistant bacteria. THE MICROBE 2024; 5:100169. [DOI: 10.1016/j.microb.2024.100169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
25
|
Sandmeier M, Ricci F, To D, Lindner S, Stengel D, Schifferle M, Koz S, Bernkop-Schnürch A. Design of self-emulsifying oral delivery systems for semaglutide: reverse micelles versus hydrophobic ion pairs. Drug Deliv Transl Res 2024:10.1007/s13346-024-01729-0. [PMID: 39427069 DOI: 10.1007/s13346-024-01729-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
It was the aim of this study to evaluate the potential of reverse micelles (RM) and hydrophobic ion pairs (HIP) for incorporation of semaglutide into self-emulsifying oral drug delivery systems. Reverse micelles loaded with semaglutide were formed with a cationic (ethyl lauroyl arginate, ELA) and an anionic surfactant (docusate, DOC), whereas HIP were formed between semaglutide and ELA. Maximum solubility of the peptide and the rate of dissolution was evaluated in various lipophilic phases (glycerol monocaprylocaprate:caprylic acid 1:4 (m/m), glycerol monolinoleate:caprylic acid 1:4 (m/m) and glycerol monocaprylocaprate:glycerol monolinoleate 1:4 (m/m)). Self-emulsifying drug delivery systems (SEDDS) loaded with RM and HIP were characterized regarding size distribution, zeta potential, cytocompatibility and Caco-2 permeability. Droplet sizes between 50 and 300 nm with polydispersity index (PDI) around 0.3 and zeta potentials between - 45 mV (RMDOC) and 36 mV (RMELA) were obtained. RM provided an almost 2-fold higher lipophilicity of semaglutide than HIP resulting in a 4.2-fold higher payload of SEDDS compared to HIP. SEDDS containing RM or HIP showed high cytocompatibilities with a cell survival above 75% for concentrations up to 0.1% on Caco-2 cells and acceptable hemolytic activity. Permeation studies across Caco-2 monolayer revealed an at least 2-fold increase in permeability of semaglutide for the developed formulations.
Collapse
Affiliation(s)
- Matthias Sandmeier
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria
- Thiomatrix Forschungs- und Beratungs GmbH, Trientlgasse 65, Innsbruck, 6020, Austria
| | - Fabrizio Ricci
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria
- Thiomatrix Forschungs- und Beratungs GmbH, Trientlgasse 65, Innsbruck, 6020, Austria
| | - Dennis To
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria
| | - Sera Lindner
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria
- Thiomatrix Forschungs- und Beratungs GmbH, Trientlgasse 65, Innsbruck, 6020, Austria
| | - Daniel Stengel
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria
| | - Michaela Schifferle
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria
| | - Saadet Koz
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria.
| |
Collapse
|
26
|
Henriksen NL, Serrano-Chávez E, Fuglsang-Madsen A, Jensen LK, Gottlieb H, Bue M, Andresen TL, Henriksen JR, Hansen AE. Gentamicin and clindamycin antibiotic-eluting depot technology eradicates S. aureus in an implant-associated osteomyelitis pig model without systemic antibiotics. Antimicrob Agents Chemother 2024; 68:e0069124. [PMID: 39287404 PMCID: PMC11459913 DOI: 10.1128/aac.00691-24] [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: 05/09/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
The therapeutic challenges of orthopedic device-related infections and emerging antimicrobial resistance have attracted attention to drug delivery technologies. This study evaluates the preclinical efficacy of local single- and dual-antibiotic therapy against implant-associated osteomyelitis (IAO) using a drug-eluting depot technology, CarboCell, that provides sustained release of high-dose antibiotics and allows for strategic in situ placement in relation to infectious lesions. Clindamycin and gentamicin were formulated in CarboCell compositions. One-stage-revision of tibial Staphylococcus aureus IAO was conducted in 19 pigs. Pigs were treated locally with CarboCell containing either gentamicin alone for 1 week or a co-formulation of gentamicin and clindamycin for 1 or 3 weeks. Bone, soft tissue, and antibiotic depots were collected for microbiology, histology, and HPLC analyses. Supporting in vivo release studies of CarboCell formulations were performed on mice. Both single- and dual-antibiotic CarboCell formulations were developed and capable of eradicating the infectious bacteria in bone and preventing colonization of implants inserted at revision. Eradication in soft tissue was observed in all pigs after 3 weeks and in 6/9 pigs after 1 week of treatment. Neutrophil counts in bone tissue were below the infection cut-off in all pigs receiving the dual-antibiotic therapies, but above in all pigs receiving the single-antibiotic therapy. Histological signs of active bone reorganization and healing were observed at 3 weeks. In conclusion, all CarboCell formulations demonstrated strong therapeutic activity against IAO, eradicating S. aureus in bone tissue and preventing colonization of implants even without the addition of systemic antibiotic therapy.
Collapse
Affiliation(s)
- Nicole L. Henriksen
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | | | - Louise K. Jensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Hans Gottlieb
- Department of Orthopedic Surgery, Herlev Hospital, Herlev, Denmark
| | - Mats Bue
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Orthopaedic Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas L. Andresen
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Jonas R. Henriksen
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Anders E. Hansen
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
27
|
Sun Y, Lu T, Pan J, He H, Xu M, Chen Y, Chen Y, Fang P, Ye X, Li S, Hu H, Yu S. Dual tobramycin and docosahexaenoic acid loaded nanoemulsions combating Pseudomonas aeruginosa-induced pulmonary infection. Colloids Surf B Biointerfaces 2024; 242:114088. [PMID: 39003845 DOI: 10.1016/j.colsurfb.2024.114088] [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: 04/12/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) typically forms biofilms in vivo, which exhibit high resistance and complicate eradication efforts. Additionally, persistent inflammation and excessive oxidative stress can lead to severe lung dysfunction, facilitating bacterial colonization and infection. Herein, we prepared oil-in-water (O/W) nanoemulsions (TD-αT NEs) by using PEG5k-block-PCL5k and α-tocopherol to encapsulate tobramycin (TOB). To enhance TOB's drug load, a hydrophobic ion pair (TDIP) composed of TOB and docosahexaenoic acid (DHA) was pre-prepared. TD-αT NEs was not only easily prepared and aerosolized, but stable in both physics and chemistry. The negatively charged TD-αT NEs facilitated penetration through mucus, reaching infection sites. Subsequently, TD-αT NEs permeated biofilms due to their small size and released drugs via lipase-triggered carrier dissociation, aiding in eradicating internal bacteria within biofilms (with a 16-fold reduction in CFU vs. free TOB group). TD-αT NEs simultaneously exerted superior anti-inflammatory effects, reducing levels of pro-inflammatory cytokines (NO, IL-6, IL-8, and TNF-α) while increasing the level of anti-inflammatory cytokine (IL-10). It was achieved through the upregulation of PPAR-γ and downregulation of NF-κB signaling, thus mitigating the lung damage. In addition, TD-αT NEs demonstrated strong antioxidant activity, alleviating the oxidative stress induced by P. aeruginosa. Notably, when administered via inhalation, TD-αT NEs significantly reduced the lung bacterial burden, lung inflammation, and oxidative stress in vivo compared to TOB solution. TD-αT NEs could prove beneficial in treating chronic pulmonary infections induced by P. aeruginosa through a comprehensive strategy, specifically enhancing biofilm eradication, reducing inflammation, and alleviating oxidative stress.
Collapse
Affiliation(s)
- Yingying Sun
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Tianli Lu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Jieyi Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Haonan He
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Mao Xu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Yujun Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Yan Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Pengchao Fang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Xiaoxing Ye
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Shuxuan Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Haiyan Hu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China; State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China.
| | - Shihui Yu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China; State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China.
| |
Collapse
|
28
|
Tan X, Zhou Y, Qin Y, Wu L, Yang R, Bao X, Jiang R, Sun X, Ying X, Ben Z, Dai Q, Zhang Z, Zeng K, Han M. Self-Healing Hydrogel Resulting from the Noncovalent Interaction between Ropivacaine and Low-Molecular-Weight Gelator Sodium Deoxycholate Achieves Stable and Endurable Local Analgesia in Vivo. ACS APPLIED MATERIALS & INTERFACES 2024; 16:45969-45988. [PMID: 39171973 DOI: 10.1021/acsami.4c07883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Regional analgesia based on the local anesthetic ropivacaine plays a crucial role in postoperative pain management and recovery; however, the short duration of analgesia limits its clinical potential. Various drug delivery systems such as microparticles and lipid carriers have been used to prolong the analgesic effect, yet most of them are prone to abrupt release from the site of administration or have poor analgesic effects of less than 48 h, which fail to meet the needs of postoperative analgesia. In this study, a low-molecular-weight gelator sodium deoxycholate-based hydrogel loaded with ropivacaine (DC-ROP gel) was designed for long-acting analgesia. The noncovalent interaction between ropivacaine and sodium deoxycholate helps to improve the stability and sustained release performance of the gel. This internal drug-binding hydrogel also avoids experiencing the burst release effect commonly seen in polymer hydrogels previously reported for the slow release of local anesthetics. DC-ROP gel exhibited the dual advantages of self-healing after compression and long-term controlled release. In mice with inflammatory pain, DC-ROP gel achieved peripheral nerve block for more than 1 week after a single injection. Histological and blood biochemical analyses confirmed that the DC-ROP gel did not produce systemic toxicity, and cytotoxicity experiments demonstrated that the DC-ROP gel resulted in low irritation. These results suggest that DC-ROP gel provides a promising strategy for local anesthetics in long-term postoperative pain management, broadening the potential of bile salt-based low-molecular-weight hydrogels for drug delivery.
Collapse
Affiliation(s)
- Xin Tan
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi Zhou
- National Narcotic Laboratory Zhejiang Regional Center, Hangzhou 310000, China
| | - Yaxin Qin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Linjie Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ruizhi Yang
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Xiaoyan Bao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ruolin Jiang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoyan Sun
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xufang Ying
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhiqing Ben
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qi Dai
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Afliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhicheng Zhang
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Afliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Kai Zeng
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Min Han
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Afliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, PR China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
29
|
Zhang J, Fang H, Dai Y, Li Y, Li L, Zuo S, Liu T, Sun Y, Shi X, He Z, Sun J, Sun B. Cholesterol sulfate-mediated ion-pairing facilitates the self-nanoassembly of hydrophilic cationic mitoxantrone. J Colloid Interface Sci 2024; 669:731-739. [PMID: 38735255 DOI: 10.1016/j.jcis.2024.05.029] [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: 01/20/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/14/2024]
Abstract
HYPOTHESIS Hydrophilic cationic drugs such as mitoxantrone hydrochloride (MTO) pose a significant delivery challenge to the development of nanodrug systems. Herein, we report the use of a hydrophobic ion-pairing strategy to enhance the nano-assembly of MTO. EXPERIMENTS We employed biocompatible sodium cholesteryl sulfate (SCS) as a modification module to form stable ion pairs with MTO, which balanced the intermolecular forces and facilitated nano-assembly. PEGylated MTO-SCS nanoassemblies (pMS NAs) were prepared via nanoprecipitation. We systematically evaluated the effect of the ratio of the drug module (MTO) to the modification module (SCS) on the nanoassemblies. FINDINGS The increased lipophilicity of MTO-SCS ion pair could significantly improve the encapsulation efficiency (∼97 %) and cellular uptake efficiency of MTO. The pMS NAs showed prolonged blood circulation, maintained the same level of tumor antiproliferative activity, and exhibited reduced toxicity compared with the free MTO solution. It is noteworthy that the stability, cellular uptake, cytotoxicity, and in vivo pharmacokinetic behavior of the pMS NAs increased in proportion to the molar ratio of SCS to MTO. This study presents a self-assembly strategy mediated by ion pairing to overcome the challenges commonly associated with the poor assembly ability of hydrophilic cationic drugs.
Collapse
Affiliation(s)
- Jingxuan Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hongkai Fang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuebin Dai
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yaqiao Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lingxiao Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shiyi Zuo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tian Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yixin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Bingjun Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
30
|
Chary PS, Shaikh S, Rajana N, Bhavana V, Mehra NK. Unlocking nature's arsenal: Nanotechnology for targeted delivery of venom toxins in cancer therapy. BIOMATERIALS ADVANCES 2024; 162:213903. [PMID: 38824828 DOI: 10.1016/j.bioadv.2024.213903] [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: 01/18/2024] [Revised: 04/24/2024] [Accepted: 05/19/2024] [Indexed: 06/04/2024]
Abstract
AIM The aim of the present review is to shed light on the nanotechnological approaches adopted to overcome the shortcomings associated with the delivery of venom peptides which possess inherent anti-cancer properties. BACKGROUND Venom peptides although have been reported to demonstrate anti-cancer effects, they suffer from several disadvantages such as in vivo instability, off-target adverse effects, limited drug loading and low bioavailability. This review presents a comprehensive compilation of different classes of nanocarriers while underscoring their advantages, disadvantages and potential to carry such peptide molecules for in vivo delivery. It also discusses various nanotechnological aspects such as methods of fabrication, analytical tools to assess these nanoparticulate formulations, modulation of nanocarrier polymer properties to enhance loading capacity, stability and improve their suitability to carry toxic peptide drugs. CONCLUSION Nanotechnological approaches bear great potential in delivering venom peptide-based molecules as anticancer agents by enhancing their bioavailability, stability, efficacy as well as offering a spatiotemporal delivery approach. However, the challenges associated with toxicity and biocompatibility of nanocarriers must be duly addressed. PERSPECTIVES The everlasting quest for new breakthroughs for safer delivery of venom peptides in human subjects is fuelled by unmet clinical needs in the current landscape of chemotherapy. In addition, exhaustive efforts are required in obtaining and purifying the venom peptides followed by designing and optimizing scale up technologies.
Collapse
Affiliation(s)
- Padakanti Sandeep Chary
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Samia Shaikh
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Naveen Rajana
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Valamla Bhavana
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India.
| |
Collapse
|
31
|
Polidori I, To D, Kali G, Bernkop-Schnürch A. Histidine-based ionizable cationic surfactants: novel biodegradable agents for hydrophilic macromolecular drug delivery. Drug Deliv Transl Res 2024; 14:2370-2385. [PMID: 38289467 PMCID: PMC11291603 DOI: 10.1007/s13346-023-01511-8] [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] [Accepted: 12/26/2023] [Indexed: 08/03/2024]
Abstract
The aim of this study was to design surfactants based on histidine (His) for hydrophobic ion-pairing and evaluate their safety and efficacy. Lauryl, palmitoyl and oleyl alcohol, as well as 2-hexyl-1-decanol were converted into surfactants with histidine as head-group via esterification. The synthesized His-surfactants were characterized regarding pKa, critical micellar concentration (CMC), biodegradability, toxicity on Caco-2 cells, and ability to provide endosomal escape. Furthermore, the suitability of these agents to be employed as counterions in hydrophobic ion pairing was evaluated. Chemical structures were confirmed by 1H-NMR, FT-IR, and MS. The synthesized surfactants showed pKa values ranging from 4.9 to 6.0 and CMC values in the range of 0.3 to 7.0 mM. Their biodegradability was proven by enzymatic cleavage within 24 h. Below the CMC, His-surfactants did not show cytotoxic effects on Caco-2 cells (cell viability > 80%). All His-surfactants showed the ability to provide endosomal escape in a pH-dependent manner in the range of 5.2 to 6.8. Complexes formed between His-surfactants and heparin or plasmid DNA (pDNA) via hydrophobic ion pairing showed at least 100-fold higher lipophilicity than the correspondent model drugs. According to these results, His-surfactants might be a promising safe tool for delivering hydrophilic macromolecular drugs and nucleic acids.
Collapse
Affiliation(s)
- Ilaria Polidori
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria
| | - Dennis To
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria
| | - Gergely Kali
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, 6020, Austria.
| |
Collapse
|
32
|
Ingabire D, Qin C, Meng T, Raynold AAM, Sudarjat H, Townsend EA, Pangeni R, Poudel S, Arriaga M, Zhao L, Chow WN, Banks M, Xu Q. Nor-LAAM loaded PLGA microparticles for treating opioid use disorder. J Control Release 2024; 373:93-104. [PMID: 38968971 PMCID: PMC11384420 DOI: 10.1016/j.jconrel.2024.06.071] [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: 04/08/2024] [Revised: 06/08/2024] [Accepted: 06/30/2024] [Indexed: 07/07/2024]
Abstract
The treatment landscape for opioid use disorder (OUD) faces challenges stemming from the limited efficacy of existing medications, poor adherence to prescribed regimens, and a heightened risk of fatal overdose post-treatment cessation. Therefore, there is a pressing need for innovative therapeutic strategies that enhance the effectiveness of interventions and the overall well-being of individuals with OUD. This study explored the therapeutic potential of nor-Levo-α-acetylmethadol (nor-LAAM) to treat OUD. We developed sustained release nor-LAAM-loaded poly (lactic-co-glycolic acid) (PLGA) microparticles (MP) using a hydrophobic ion pairing (HIP) approach. The nor-LAAM-MP prepared using HIP with pamoic acid had high drug loading and exhibited minimal initial burst release and sustained release. The nor-LAAM-MP was further optimized for desirable particle size, drug loading, and release kinetics. The lead nor-LAAM-MP (F4) had a relatively high drug loading (11 wt%) and an average diameter (19 μm) and maintained a sustained drug release for 4 weeks. A single subcutaneous injection of nor-LAAM-MP (F4) provided detectable nor-LAAM levels in rabbit plasma for at least 15 days. We further evaluated the therapeutic efficacy of nor-LAAM-MP (F4) in a well-established fentanyl-addiction rat model, and revealed a marked reduction in fentanyl choice and withdrawal symptoms in fentanyl-dependent rats. These findings provide insights into further developing long-acting nor-LAAM-MP for treating OUD. It has the potential to offer a new effective medication to the existing sparse armamentarium of products available to treat OUD.
Collapse
Affiliation(s)
- Diane Ingabire
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA; Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Chaolong Qin
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Tuo Meng
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | - Hadi Sudarjat
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - E Andrew Townsend
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Rudra Pangeni
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Sagun Poudel
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Michelle Arriaga
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Long Zhao
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Woon N Chow
- Department of Ophthalmology, Virginia Commonwealth University, Richmond, VA 23298, USA; Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Matthew Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | - Qingguo Xu
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, USA; Department of Ophthalmology, Virginia Commonwealth University, Richmond, VA 23298, USA; Center for Pharmaceutical Engineering, and Institute for Structural Biology, Drug Discovery & Development (ISB3D), Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
33
|
Tan X, Ke P, Chen Z, Zhou Y, Wu L, Bao X, Qin Y, Jiang R, Han M. Construction of injectable micron-sized polymorphic vesicles for prolonged local anesthesia with weekly sustained release of ropivacaine. Int J Pharm 2024; 661:124378. [PMID: 38925241 DOI: 10.1016/j.ijpharm.2024.124378] [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: 02/26/2024] [Revised: 05/22/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024]
Abstract
Currently, to overcome the short half-life of the local anesthetic ropivacaine, drug delivery systems such as nanoparticles and liposomes have been used to prolong the analgesic effect, but they are prone to abrupt release from the site of administration or have poor slow-release effects, which increases the risk of cardiotoxicity. In this study, injectable lipid suspensions based on ropivacaine-docusate sodium hydrophobic ion pairing (HIP) were designed to significantly prolong the duration of analgesia. The resulting ion-paired lipid suspension (HIP/LIPO) had a micrometer scale and a high zeta potential, which facilitates stable in situ retention. The strong interaction between docusate sodium and ropivacaine was verified using thermal and spectroscopic analyses, and the formation of micron-sized polymorphic vesicles was attributed to the mutual stabilizing interactions between ropivacaine-docusate sodium HIP, docusate sodium and lecithin. The HIP/LIPO delivery system could maintain drug release for more than 5 days in vitro and achieve high analgesic efficacy for more than 10 days in vivo, reducing the side effects associated with high drug doses. The stable HIP/LIPO delivery system is a promising strategy that offers a clinically beneficial alternative for postoperative pain management and other diseases.
Collapse
Affiliation(s)
- Xin Tan
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peng Ke
- Department of Anesthesiology, Fujian Provincial Hospital, Fujian Shengli Clinical Medical College, Fujian Medical University, Fuzhou 350108, China
| | - Ziying Chen
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi Zhou
- National Narcotic Laboratory Zhejiang Regional Center, Hangzhou 310000, China
| | - Linjie Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoyan Bao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yaxin Qin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ruolin Jiang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Min Han
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Afliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China; Jinhua Institute of Zhejiang University, Jinhua 321299, Zhejiang, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
34
|
Singh R, Sharma S, Kautu A, Joshi KB. Self-assembling short peptide amphiphiles as versatile delivery agents: a new frontier in antibacterial research. Chem Commun (Camb) 2024; 60:7687-7696. [PMID: 38958435 DOI: 10.1039/d4cc01762e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Self-assembling short peptide amphiphiles, crafted through a minimalistic approach, spontaneously generate well-ordered nanostructures, facilitating the creation of precise nanostructured biomaterials for diverse biomedical applications. The seamless integration of bioactive metal ions and nanoparticles endows them with the potential to serve as pioneering materials in combating bacterial infections. Nanomanipulation of these molecules' binary structures enables effective penetration of membranes, forming structured nanoarchitectures with antibacterial properties. Through a comprehensive exploration, we attempt to reveal the innovative potential of short peptide amphiphiles, particularly in conjugation with metal cations and nanoparticles, offering insights for future research trajectories.
Collapse
Affiliation(s)
- Ramesh Singh
- Department of Chemistry, School of Chemical Science and Technology, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, 470003, Madhya Pradesh, India.
| | - Shruti Sharma
- Department of Chemistry, School of Chemical Science and Technology, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, 470003, Madhya Pradesh, India.
| | - Aanand Kautu
- Department of Chemistry, School of Chemical Science and Technology, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, 470003, Madhya Pradesh, India.
| | - Khashti Ballabh Joshi
- Department of Chemistry, School of Chemical Science and Technology, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, 470003, Madhya Pradesh, India.
| |
Collapse
|
35
|
Deshmukh SH, Nachaki EO, Kuroda DG. Uncovering the binding nature of thiocyanate in contact ion pairs with lithium ions. J Chem Phys 2024; 161:034507. [PMID: 39017430 DOI: 10.1063/5.0216491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/28/2024] [Indexed: 07/18/2024] Open
Abstract
Ion pair formation is a fundamental molecular process that occurs in a wide variety of systems, including electrolytes, biological systems, and materials. In solution, the thiocyanate (SCN-) anion interacts with cations to form contact ion pairs (CIPs). Due to its ambidentate nature, thiocyanate can bind through either its sulfur or nitrogen atoms, depending on the solvent. This study focuses on the binding nature of thiocyanate with lithium ions as a function of the solvents using FTIR, 2D infrared spectroscopy (2DIR) spectroscopies, and theoretical calculations. The study reveals that the SCN- binding mode (S or N end) in CIPs can be identified through 2DIR spectroscopy but not by linear IR spectroscopy. Linear IR spectroscopy shows that the CN stretch frequencies are too close to one another to separate N- and S-bound CIPs. Moreover, the IR spectrum shows that the S-C stretch presents different frequencies for the salt in different solvents, but it is related to the anion speciation rather than to its binding mode. A similar trend is observed for the anion bend. 2DIR spectra show different dynamics for N-bound and S-bound thiocyanate. In particular, the frequency-frequency correlation function (FFCF) dynamics extracted from the 2DIR spectra have a single picosecond exponential decay for N-bound thiocyanate and a biexponential decay for S-bound thiocyanate, consistent with the binding mode of the anion. Finally, it is also observed that the binding mode also affects the line shape parameters, probably due to the different molecular mechanisms of the FFCF for N- and S-bound CIPs.
Collapse
Affiliation(s)
- Samadhan H Deshmukh
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, USA
| | - Ernest O Nachaki
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, USA
| | - Daniel G Kuroda
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, USA
| |
Collapse
|
36
|
Zhang H, Li Y, Fu Y, Jiao H, Wang X, Wang Q, Zhou M, Yong YC, Liu J. A structure-functionality insight into the bioactivity of microbial polysaccharides toward biomedical applications: A review. Carbohydr Polym 2024; 335:122078. [PMID: 38616098 DOI: 10.1016/j.carbpol.2024.122078] [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: 11/28/2023] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 04/16/2024]
Abstract
Microbial polysaccharides (MPs) are biopolymers secreted by microorganisms such as bacteria and fungi during their metabolic processes. Compared to polysaccharides derived from plants and animals, MPs have advantages such as wide sources, high production efficiency, and less susceptibility to natural environmental influences. The most attractive feature of MPs lies in their diverse biological activities, such as antioxidative, anti-tumor, antibacterial, and immunomodulatory activities, which have demonstrated immense potential for applications in functional foods, cosmetics, and biomedicine. These bioactivities are precisely regulated by their sophisticated molecular structure. However, the mechanisms underlying this precise regulation are not yet fully understood and continue to evolve. This article presents a comprehensive review of the most representative species of MPs, including their fermentation and purification processes and their biomedical applications in recent years. In particular, this work presents an in-depth analysis into the structure-activity relationships of MPs across multiple molecular levels. Additionally, this review discusses the challenges and prospects of investigating the structure-activity relationships, providing valuable insights into the broad and high-value utilization of MPs.
Collapse
Affiliation(s)
- Hongxing Zhang
- Biofuels Institute, School of Environment and Safety Engineering, c/o School of Emergency Management, Jiangsu University, Zhenjiang 212013, China
| | - Yan Li
- Biofuels Institute, School of Environment and Safety Engineering, c/o School of Emergency Management, Jiangsu University, Zhenjiang 212013, China
| | - Yinyi Fu
- Biofuels Institute, School of Environment and Safety Engineering, c/o School of Emergency Management, Jiangsu University, Zhenjiang 212013, China; Jiangsu Collaborative Innovation Center of Technology and Material of Water Treatment, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Haixin Jiao
- Biofuels Institute, School of Environment and Safety Engineering, c/o School of Emergency Management, Jiangsu University, Zhenjiang 212013, China
| | - Xiangyu Wang
- Biofuels Institute, School of Environment and Safety Engineering, c/o School of Emergency Management, Jiangsu University, Zhenjiang 212013, China
| | - Qianqian Wang
- Biofuels Institute, School of Environment and Safety Engineering, c/o School of Emergency Management, Jiangsu University, Zhenjiang 212013, China
| | - Mengbo Zhou
- Biofuels Institute, School of Environment and Safety Engineering, c/o School of Emergency Management, Jiangsu University, Zhenjiang 212013, China
| | - Yang-Chun Yong
- Biofuels Institute, School of Environment and Safety Engineering, c/o School of Emergency Management, Jiangsu University, Zhenjiang 212013, China; Jiangsu Collaborative Innovation Center of Technology and Material of Water Treatment, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jun Liu
- Biofuels Institute, School of Environment and Safety Engineering, c/o School of Emergency Management, Jiangsu University, Zhenjiang 212013, China; Jiangsu Collaborative Innovation Center of Technology and Material of Water Treatment, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
37
|
Han Y, Spicer J, Huang Y, Bunt C, Liu M, Wen J. Advancements in oral insulin: A century of research and the emergence of targeted nanoparticle strategies. EUR J LIPID SCI TECH 2024; 126. [DOI: 10.1002/ejlt.202300271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Indexed: 01/03/2025]
Abstract
AbstractWith the growing prevalence of diabetes, there is an urgent demand for a user‐friendly treatment option that minimizes side effects related to the use of subcutaneous injections. Scientists have dedicated over a century to developing an oral dosage form of insulin that can be administrated orally. The oral route of administration is the most desirable route for regularly dosed drugs in terms of safety and patient compliance. However, oral delivery of insulin remains a formidable challenge due to its intrinsically limited ability to cross the intestinal epithelium membrane and susceptibility to enzymatic degradation. This article reviews oral insulin research over the past decade, with a particular focus on surface modifications of nanoparticles (NPs). Various strategies involving controlling surface charges, utilizing protective proteins, and targeting specific receptors with ligands have been explored. Notably, surface modifications of the NPs for targeting specific intestinal receptors have shown promise in enhancing insulin oral absorption and bioavailability. Advanced technologies such as oral microneedles and gene therapy have also been developed, but their safety requires further assessment. Despite encouraging preclinical results across numerous strategies, the current clinical evidence is less optimistic. In summary, the present findings highlight the substantial journey that still lies ahead before achieving successful oral delivery of insulin.Practical Applications: This review provides a summary of recent progress in oral insulin delivery, particularly highlighting surface‐modified functional nanoparticles serving as an effective drug delivery system, which offers valuable information to the researchers. Due to the limited effectiveness of oral protein drugs caused by biological barriers, innovative technologies and drug delivery systems have been developed to overcome these obstacles and achieve therapeutic goals. This review concluded that surface modifications to nanoparticles can improve insulin stability and permeability, thereby enhancing oral bioavailability. It could assist researchers in developing more effective and patient‐friendly oral drug delivery systems.
Collapse
Affiliation(s)
- Yue Han
- School of Pharmacy Faculty of Medical and Health Sciences The University of Auckland Auckland New Zealand
| | - Julie Spicer
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland Auckland New Zealand
| | - Yuan Huang
- Key Laboratory of Drug Targeting and Drug Delivery, West China School of Pharmacy, Sichuan University Chengdu China
| | - Craig Bunt
- The Department of Food Science University of Otago Dunedin New Zealand
| | - Mengyang Liu
- School of Pharmacy Faculty of Medical and Health Sciences The University of Auckland Auckland New Zealand
| | - Jingyuan Wen
- School of Pharmacy Faculty of Medical and Health Sciences The University of Auckland Auckland New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland Auckland New Zealand
| |
Collapse
|
38
|
Haddadzadegan S, To D, Matteo Jörgensen A, Wibel R, Laffleur F, Bernkop-Schnürch A. Comparative Analysis of PEG-Free and PEG-Based Self-Emulsifying Drug Delivery Systems for Enhanced Oral Bioavailability of Therapeutic (Poly) Peptides. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307618. [PMID: 38308358 DOI: 10.1002/smll.202307618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/13/2024] [Indexed: 02/04/2024]
Abstract
This study aims to compare the potential of Polyethylene glycol (PEG-free and PEG-based self-emulsifying drug delivery systems (SEDDS) for the oral administration of insulin glargine (IG). Hydrophobic ion pairs (HIPs) of IG are formed using various counterions. HIPs are assessed for log P octanol/water and dissociation behavior. They are incorporated into SEDDS based on polyglycerol (PG) and zwitterionic surfactant (ZW) using response surface methodology and compared to conventional PEG-SEDDS in size, stability, and log D SEDDS/release medium. Oral IG bioavailability in PG/ZW-SEDDS and PEG-SEDDS is evaluated in rats. Among the various counterions studied, IG-BIS (bis(isotridecyl)sulfosuccinate) HIPs demonstrated the highest log P and an improved dissociation profile. PG/ZW-SEDDS and PEG-SEDDS have similar ≈40 nm sizes and are stable over 24 h. Both formulations have log D > 4 in water and >2 in 50 mM phosphate buffer pH 6.8. PG/ZW-SEDDS yielded an oral bioavailability of 2.13 ± 0.66% for IG, while the employment of PEG-SEDDS resulted in an oral bioavailability of 1.15 ± 0.35%. This study highlights the prospective utilization of PEG-free SEDDS involving the concurrent application of PG and ZW surfactants, an alternative to conventional PEG surfactants, for improved oral therapeutic (poly) peptide delivery.
Collapse
Affiliation(s)
- Soheil Haddadzadegan
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Dennis To
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Arne Matteo Jörgensen
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Richard Wibel
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Flavia Laffleur
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
39
|
Casalini T, Treacher K, Grant I, Marucci M. In Vitro Release from Polymeric Core/Shell Nanoparticles through the Lens of Multiscale Modeling. Mol Pharm 2024; 21:2684-2698. [PMID: 38687999 DOI: 10.1021/acs.molpharmaceut.3c00806] [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
The large number of studies involving nanoparticles for cancer therapy is due to their peculiar features: they protect loaded active molecules while extending circulation time and can extravasate from the blood flow to the tumor to deliver drugs directly in the target area. Mathematical modeling can provide a preliminary in silico exploration of design space to optimize an experimental activity that often relies on a trial-and-error approach. However, because of the characteristic size of these vectors (10-1000 nm), numerous phenomena of interest occur at different time and length scales, making a single modeling technique insufficient to fully characterize the system of interest. In this work we employed a multiscale modeling approach, which bridges the phenomena of interest across different scales, to study the in vitro release from polymeric core/shell nanoparticles for cancer therapy loaded with an active compound assembled as a hydrophobic ion pair. The "computational microscope" provided by molecular dynamics simulations was used to track drug molecules through the release process at an atomic scale. The outcomes suggested that the drug is mainly partitioned in the polymer and released as hydrophobic ion pair rather than a free molecule, and that the hydrophobic ion pair is preferentially partitioned in Tween 20 micelles in the release media. A model at macroscale, aimed at describing the release rate and elucidating the release mechanism, was developed according to the results from molecular simulations and validated against experimental data. The outcomes provided insights that are challenging to be obtained experimentally and which supported the development and validation of a release model at macroscale. Overall, the adopted multiscale approach corroborated the experimental findings and provided significant insights into the mechanisms of release.
Collapse
Affiliation(s)
- Tommaso Casalini
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg 11115 - 98499, Sweden
| | - Kevin Treacher
- New Modalities and Parenterals Development, Pharmaceutical Technology & Development, Operations & IT, AstraZeneca, Macclesfield, Cheshire SK10 2NA, United Kingdom
| | - Iain Grant
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, Cheshire SK10 2NA, United Kingdom
| | - Mariagrazia Marucci
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg 11115 - 98499, Sweden
| |
Collapse
|
40
|
Sirvi A, Jadhav K, Sangamwar AT. Enabling superior drug loading in lipid-based formulations with lipophilic salts for a brick dust molecule: Exploration of lipophilic counterions and in vitro-in vivo evaluation. Int J Pharm 2024; 656:124108. [PMID: 38604540 DOI: 10.1016/j.ijpharm.2024.124108] [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: 02/22/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Lipid-based formulations (LbFs) are an extensively used approach for oral delivery of poorly soluble drug compounds in the form of lipid suspension and lipid solution. However, the high target dose and inadequate lipid solubility limit the potential of brick dust molecules to be formulated as LbFs. Thus, the complexation of such molecules with a lipophilic counterion can be a plausible approach to improve the solubility in lipid-based solutions via reducing drug crystallinity and polar surface area. The study aimed to enhance drug loading in lipid solution for Nilotinib (Nil) through complexation or salt formation with different lipophilic counterions. We synthesized different lipophilic salts/ complexes via metathesis reactions and confirmed their formation by 1H NMR and FTIR. Docusate-based lipophilic salt showed improved solubility in medium-chain triglycerides (∼7 to 7.5-fold) and long-chain triglycerides (∼30 to 35-fold) based lipids compared to unformulated crystalline Nil. The increased lipid solubility could be attributed to the reduction in drug crystallinity which was further confirmed by the PXRD and DSC. Prototype LbFs were prepared to evaluate drug loading and their physicochemical characteristics. The findings suggested that structural features of counterion including chain length and lipophilicity affect the drug loading in LbF. In addition, physical stability testing of formulations was performed, inferring that aliphatic sulfate-based LbFs were stable with no sign of drug precipitation or salt disproportionation. An in vitro lipolysis-permeation study revealed that the primary driver of absorptive flux is the solubilization of the drug and reduced amount of lipid. Further, the in vivo characterization was conducted to measure the influence of increased drug load on oral bioavailability. Overall, the results revealed enhanced absorption of lipophilic salt-based LbF over unformulated crystalline Nil and conventional LbF (drug load equivalent to equilibrium solubility) which supports the idea that lipophilic salt-based LbF enhances drug loading, and supersaturation-mediated drug solubilization, unlocking the full potential of LbF.
Collapse
Affiliation(s)
- Arvind Sirvi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S Nagar, Punjab, India
| | - Karan Jadhav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S Nagar, Punjab, India
| | - Abhay T Sangamwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S Nagar, Punjab, India.
| |
Collapse
|
41
|
Pangua C, Espuelas S, Martínez-Ohárriz MC, Vizmanos JL, Irache JM. Mucus-penetrating and permeation enhancer albumin-based nanoparticles for oral delivery of macromolecules: Application to bevacizumab. Drug Deliv Transl Res 2024; 14:1189-1205. [PMID: 37880504 PMCID: PMC10984897 DOI: 10.1007/s13346-023-01454-0] [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] [Accepted: 10/16/2023] [Indexed: 10/27/2023]
Abstract
The oral administration of therapeutic proteins copes with important challenges (mainly degradation and poor absorption) making their potential therapeutic application extremely difficult. The aim of this study was to design and evaluate the potential of the combination between mucus-permeating nanoparticles and permeation enhancers as a carrier for the oral delivery of the monoclonal antibody bevacizumab, used as a model of therapeutic protein. For this purpose, bevacizumab was encapsulated in PEG-coated albumin nanoparticles as a hydrophobic ion-pairing complex with either sodium deoxycholate (DS) or sodium docusate (DOCU). In both cases, complex formation efficiencies close to 90% were found. The incorporation of either DS or DOCU in PEG-coated nanoparticles significantly increased their mean size, particularly when DOCU was used. Moreover, the diffusion in mucus of DOCU-loaded nanoparticles was significantly reduced, compared with DS ones. In a C. elegans model, DS or DOCU (free or nanoencapsulated) disrupted the intestinal epithelial integrity, but the overall survival of the worms was not affected. In rats, the relative oral bioavailability of bevacizumab incorporated in PEG-coated nanoparticles as a complex with DS (B-DS-NP-P) was 3.7%, a 1000-fold increase compared to free bevacizumab encapsulated in nanoparticles (B-NP-P). This important effect of DS may be explained not only by its capability to transiently disrupt tight junctions but also to their ability to increase the fluidity of membranes and to inhibit cytosolic and brush border enzymes. In summary, the current strategy may be useful to allow the therapeutic use of orally administered proteins, including monoclonal antibodies.
Collapse
Affiliation(s)
- Cristina Pangua
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, 31008, Pamplona, Spain
| | - Socorro Espuelas
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, 31008, Pamplona, Spain
| | | | - José Luis Vizmanos
- Department of Biochemistry & Genetics, School of Sciences, University of Navarra, 31008, Pamplona, Spain
| | - Juan M Irache
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, 31008, Pamplona, Spain.
- Institute for Health Research (IdiSNA), 31008, Pamplona, Spain.
| |
Collapse
|
42
|
Li Z, Kovshova T, Malinovskaya J, Valikhov M, Melnikov P, Osipova N, Maksimenko O, Dhakal N, Chernysheva A, Chekhonin V, Gelperina S, Wacker MG. Modeling the Drug delivery Lifecycle of PLG Nanoparticles Using Intravital Microscopy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306726. [PMID: 38152951 DOI: 10.1002/smll.202306726] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/29/2023] [Indexed: 12/29/2023]
Abstract
Polylactide-co-glycolide (PLG) nanoparticles hold immense promise for cancer therapy due to their enhanced efficacy and biodegradable matrix structure. Understanding their interactions with blood cells and subsequent biodistribution kinetics is crucial for optimizing their therapeutic potential. In this study, three doxorubicin-loaded PLG nanoparticle systems are synthesized and characterized, analyzing their size, zeta potential, morphology, and in vitro release behavior. Employing intravital microscopy in 4T1-tumor-bearing mice, real-time blood and tumor distribution kinetics are investigated. A mechanistic pharmacokinetic model is used to analyze biodistribution kinetics. Additionally, flow cytometry is utilized to identify cells involved in nanoparticle hitchhiking. Following intravenous injection, PLG nanoparticles exhibit an initial burst release (<1 min) and rapidly adsorb to blood cells (<5 min), hindering extravasation. Agglomeration leads to the clearance of one carrier species within 3 min. In stable dispersions, drug release rather than extravasation remains the dominant pathway for drug elimination from circulation. This comprehensive investigation provides valuable insights into the interplay between competing kinetics that influence the lifecycle of PLG nanoparticles post-injection. The findings advance the understanding of nanoparticle behavior and lay the foundation for improved cancer therapy strategies using nanoparticle-based drug delivery systems.
Collapse
Affiliation(s)
- Zhuoxuan Li
- Department of Pharmacy, National University of Singapore, 4 Science Drive 2, Singapore, 117544, Singapore
| | - Tatyana Kovshova
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow, 125047, Russia
| | - Julia Malinovskaya
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow, 125047, Russia
| | - Marat Valikhov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, Moscow, 119034, Russia
| | - Pavel Melnikov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, Moscow, 119034, Russia
| | - Nadezhda Osipova
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow, 125047, Russia
| | - Olga Maksimenko
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow, 125047, Russia
| | - Namrata Dhakal
- Department of Pharmacy, National University of Singapore, 4 Science Drive 2, Singapore, 117544, Singapore
| | - Anastasia Chernysheva
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, Moscow, 119034, Russia
| | - Vladimir Chekhonin
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, Moscow, 119034, Russia
| | - Svetlana Gelperina
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow, 125047, Russia
| | - Matthias G Wacker
- Department of Pharmacy, National University of Singapore, 4 Science Drive 2, Singapore, 117544, Singapore
| |
Collapse
|
43
|
Larsen NW, Kostrikov S, Hansen MB, Hjørringgaard CU, Larsen NB, Andresen TL, Kristensen K. Interactions of oral permeation enhancers with lipid membranes in simulated intestinal environments. Int J Pharm 2024; 654:123957. [PMID: 38430950 DOI: 10.1016/j.ijpharm.2024.123957] [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: 11/19/2023] [Revised: 02/17/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
The oral bioavailability of therapeutic peptides is generally low. To increase peptide transport across the gastrointestinal barrier, permeation enhancers are often used. Despite their widespread use, mechanistic knowledge of permeation enhancers is limited. To address this, we here investigate the interactions of six commonly used permeation enhancers with lipid membranes in simulated intestinal environments. Specifically, we study the interactions of the permeation enhancers sodium caprate, dodecyl maltoside, sodium cholate, sodium dodecyl sulfate, melittin, and penetratin with epithelial cell-like model membranes. To mimic the molecular composition of the real intestinal environment, the experiments are performed with two peptide drugs, salmon calcitonin and desB30 insulin, in fasted-state simulated intestinal fluid. Besides providing a comparison of the membrane interactions of the studied permeation enhancers, our results demonstrate that peptide drugs as well as intestinal-fluid components may substantially change the membrane activity of permeation enhancers. This highlights the importance of testing permeation enhancement in realistic physiological environments and carefully choosing a permeation enhancer for each individual peptide drug.
Collapse
Affiliation(s)
- Nanna Wichmann Larsen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Serhii Kostrikov
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Morten Borre Hansen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Claudia Ulrich Hjørringgaard
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Niels Bent Larsen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Thomas Lars Andresen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Kasper Kristensen
- DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
44
|
Yamashita S, Azuma K, Tanaka Y, Kimura S, Kiriyama A. Variations in the inner core affect the pharmacokinetics of indomethacin-encapsulated polymeric micelles. Int J Pharm 2024; 654:123933. [PMID: 38403090 DOI: 10.1016/j.ijpharm.2024.123933] [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: 11/09/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
Hydrophobic ion pairing (HIP) is a drug encapsulation technology that uses electrostatic interactions between a drug and an additive. However, although polymeric micelles can encapsulate hydrophobic drugs in the core, the encapsulated drug often leaks. Therefore, we designed polymeric micelles with HIP functionalized in a hydrophobic inner core using three diblock copolymers comprising polypeptides with different ratios of polar and hydrophobic amino acids and polyethylene glycol (PEG) to encapsulate indomethacin (IND). The three IND-encapsulated HIP micelles showed different area under the curve (AUC) values as an index of blood retention after intravenous injection in mice. Despite having the same PEG shell, IND-PEG-poly(H/F)n showed a 1.56-fold higher AUC than IND-PEG-poly(D/F)n. PEG interface morphologies were evaluated to determine the differences in pharmacokinetic parameters caused by changes in inner core HIP patterns. The micellarized diblock copolymer was desorbed from IND-PEG-poly(D/F)n due to electrostatic repulsion between IND and the diblock copolymer comprising aspartic acid. Our results suggest that changes in the HIP patterns of the micelle inner core affected the PEG interface morphologies, such as PEG density and diblock copolymer desorption from micelles. These phenomena might lead to changes in the interaction of plasma proteins and drug dispositions.
Collapse
Affiliation(s)
- Shugo Yamashita
- Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan.
| | - Karen Azuma
- Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| | - Yuka Tanaka
- Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| | - Shunsuke Kimura
- Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| | - Akiko Kiriyama
- Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| |
Collapse
|
45
|
Venga Mendes T, Figueiredo EC. Magnetic Particle Spray Mass Spectrometry. Anal Chem 2024; 96:5519-5526. [PMID: 38551631 DOI: 10.1021/acs.analchem.3c05680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
In this work, the concept of magnetic particle spray mass spectrometry (MPS-MS) is reported for the first time. Magnetic sorbent particles are used to extract the analytes from a liquid sample. The particles are magnetically attracted to the tip of a magnetic probe that is positioned at the entrance of the mass spectrometer. A solvent is dispensed on the particles, and a high voltage promotes the formation of the Taylor cone around the particles agglomerate. Analytes are desorbed by the solvent, ionized, and analyzed by mass spectrometry. MPS-MS is totally in consonance with the green chemistry principle. A minimal consumption of sample (100 μL), solvent (34 μL), and magnetic sorbent (500 μg) is needed per analysis for an excellent performance of MPS-MS in terms of sensitivity and selectivity. The determination of amitriptyline, citalopram, clomipramine, chlorpromazine, doxepin, haloperidol, nortriptyline, and venlafaxine in human plasma samples using magnetic restricted-access carbon nanotubes was carried out as a proof of principle. Limits of quantification of 10 μg L-1 and correlation coefficients higher than 0.98 were obtained for all of the analytes. Limits of detection ranged from 0.43 to 2.82 μg L-1. Precision (as relative standard deviation) and accuracies (as relative error) ranged from 3.6 to 23.6%, as well as -12.8 to 18.7%, respectively. MPS-MS opens a new line of developments in the association of sample preparation with ambient ionization. New sorbents, device configurations, and physical and chemical conditions can also be analyzed for the analysis of many other analytes in different samples.
Collapse
Affiliation(s)
- Tássia Venga Mendes
- Laboratory of Toxicant and Drug Analyses, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, 37130-000 Alfenas, MG Brazil
| | - Eduardo Costa Figueiredo
- Laboratory of Toxicant and Drug Analyses, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, 37130-000 Alfenas, MG Brazil
| |
Collapse
|
46
|
Dianzani C, Bozza A, Bordano V, Cangemi L, Ferraris C, Foglietta F, Monge C, Gallicchio M, Pizzimenti S, Marini E, Muntoni E, Valsania MC, Battaglia L. Cell Membrane Fragment-Wrapped Parenteral Nanoemulsions: A New Drug Delivery Tool to Target Gliomas. Cells 2024; 13:641. [PMID: 38607080 PMCID: PMC11011487 DOI: 10.3390/cells13070641] [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: 11/27/2023] [Revised: 02/28/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
Poor prognosis in high-grade gliomas is mainly due to fatal relapse after surgical resection in the absence of efficient chemotherapy, which is severely hampered by the blood-brain barrier. However, the leaky blood-brain-tumour barrier forms upon tumour growth and vascularization, allowing targeted nanocarrier-mediated drug delivery. The homotypic targeting ability of cell-membrane fragments obtained from cancer cells means that these fragments can be exploited to this aim. In this experimental work, injectable nanoemulsions, which have a long history of safe clinic usage, have been wrapped in glioma-cell membrane fragments via co-extrusion to give targeted, homogeneously sized, sterile formulations. These systems were then loaded with three different chemotherapeutics, in the form of hydrophobic ion pairs that can be released into the target site thanks to interactions with physiological components. The numerous assays performed in two-dimensional (2D) and three-dimensional (3D) cell models demonstrate that the proposed approach is a versatile drug-delivery platform with chemo-tactic properties towards glioma cells, with adhesive interactions between the target cell and the cell membrane fragments most likely being responsible for the effect. This approach's promising translational perspectives towards personalized nanomedicine mean that further in vivo studies are foreseen for the future.
Collapse
Affiliation(s)
- Chiara Dianzani
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10124 Turin, Italy; (C.D.); (A.B.); (V.B.); (L.C.); (C.F.); (F.F.); (C.M.); (M.G.); (E.M.); (E.M.)
| | - Annalisa Bozza
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10124 Turin, Italy; (C.D.); (A.B.); (V.B.); (L.C.); (C.F.); (F.F.); (C.M.); (M.G.); (E.M.); (E.M.)
| | - Valentina Bordano
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10124 Turin, Italy; (C.D.); (A.B.); (V.B.); (L.C.); (C.F.); (F.F.); (C.M.); (M.G.); (E.M.); (E.M.)
| | - Luigi Cangemi
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10124 Turin, Italy; (C.D.); (A.B.); (V.B.); (L.C.); (C.F.); (F.F.); (C.M.); (M.G.); (E.M.); (E.M.)
| | - Chiara Ferraris
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10124 Turin, Italy; (C.D.); (A.B.); (V.B.); (L.C.); (C.F.); (F.F.); (C.M.); (M.G.); (E.M.); (E.M.)
| | - Federica Foglietta
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10124 Turin, Italy; (C.D.); (A.B.); (V.B.); (L.C.); (C.F.); (F.F.); (C.M.); (M.G.); (E.M.); (E.M.)
| | - Chiara Monge
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10124 Turin, Italy; (C.D.); (A.B.); (V.B.); (L.C.); (C.F.); (F.F.); (C.M.); (M.G.); (E.M.); (E.M.)
| | - Margherita Gallicchio
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10124 Turin, Italy; (C.D.); (A.B.); (V.B.); (L.C.); (C.F.); (F.F.); (C.M.); (M.G.); (E.M.); (E.M.)
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10124 Turin, Italy;
| | - Elisabetta Marini
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10124 Turin, Italy; (C.D.); (A.B.); (V.B.); (L.C.); (C.F.); (F.F.); (C.M.); (M.G.); (E.M.); (E.M.)
| | - Elisabetta Muntoni
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10124 Turin, Italy; (C.D.); (A.B.); (V.B.); (L.C.); (C.F.); (F.F.); (C.M.); (M.G.); (E.M.); (E.M.)
| | - Maria Carmen Valsania
- Department of Chemistry, University of Turin, Via Quarello 15/a, 10135 Turin, Italy;
- Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Turin, 10124 Turin, Italy
| | - Luigi Battaglia
- Department of Drug Science and Technology, University of Turin, via Pietro Giuria 9, 10124 Turin, Italy; (C.D.); (A.B.); (V.B.); (L.C.); (C.F.); (F.F.); (C.M.); (M.G.); (E.M.); (E.M.)
- Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Turin, 10124 Turin, Italy
| |
Collapse
|
47
|
Jia Z, Wei X, Chen N, Xu X, Zhao G, Fu X, Wang H, Goldring MB, Goldring SR, Wang D. Thermoresponsive Polymeric Hydromorphone Prodrug Provides Sustained Local Analgesia without Apparent Adverse Effects. Mol Pharm 2024; 21:1838-1847. [PMID: 38413029 PMCID: PMC11210938 DOI: 10.1021/acs.molpharmaceut.3c01133] [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] [Indexed: 02/29/2024]
Abstract
The extensive use of opioids for chronic pain management has contributed significantly to the current opioid epidemic. While many alternative nonopioid analgesics are available, opioids remain the most potent analgesics for moderate to severe pain management. In addition to the implementation of multimodal analgesia, there is a pressing need for the development of more effective and safer opioids. In this study, we developed a thermoresponsive N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer-based hydromorphone (HMP) prodrug (ProGel-HMP, HMP content = 16.2 wt %, in base form). The aqueous solution of ProGel-HMP was free-flowing at 4 °C but became a hydrogel when the temperature was raised to ≥37 °C, allowing sustained local retention when administered in vivo. When tested in the destabilization of the medial meniscus (DMM) mouse model of osteoarthritis (OA), ProGel-HMP was retained after intra-articular injection in the OA knee joint for at least 2 weeks postinjection, with low extra-articular distribution. ProGel-HMP was not detected in the central nervous system (CNS). A single dose of ProGel-HMP produced rapid and sustained joint pain resolution for greater than 14 days when compared to saline and dose-equivalent HMP controls, likely mediated through peripheral μ-opioid receptors in the knee joint. Systemic analgesia effect was absent in the DMM mice treated with ProGel-HMP, as evident in the lack of difference in tail flick response between the ProGel-HMP-treated mice and the controls (i.e., Healthy, Saline, and Sham). Repeated dosing of ProGel-HMP did not induce tolerance. Collectively, these data support the further development of ProGel-HMP as a potent, safe, long-acting and nonaddictive analgesic for better clinical pain management.
Collapse
Affiliation(s)
- Zhenshan Jia
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6125, USA
| | - Xin Wei
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6125, USA
| | - Ningrong Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6125, USA
| | - Xiaoke Xu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6125, USA
| | - Gang Zhao
- Ensign Pharmaceutical, Omaha, NE 68106, USA
| | - Xin Fu
- Ensign Pharmaceutical, Omaha, NE 68106, USA
| | - Hanjun Wang
- Department of Anesthesiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-4455, USA
| | | | - Steven R. Goldring
- Ensign Pharmaceutical, Omaha, NE 68106, USA
- Hospital for Special Surgery, New York, NY, 10021, USA
| | - Dong Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6125, USA
- Ensign Pharmaceutical, Omaha, NE 68106, USA
- Department of Orthopaedic and Rehabilitation, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5640, USA
| |
Collapse
|
48
|
Bapat P, Paul S, Tseng YC, Taylor LS. Interplay of Drug-Polymer Interactions and Release Performance for HPMCAS-Based Amorphous Solid Dispersions. Mol Pharm 2024; 21:1466-1478. [PMID: 38346390 DOI: 10.1021/acs.molpharmaceut.3c01106] [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: 03/05/2024]
Abstract
The interplay between drug and polymer chemistry and its impact on drug release from an amorphous solid dispersion (ASD) is a relatively underexplored area. Herein, the release rates of several drugs of diverse chemistry from hydroxypropyl methylcellulose acetate succinate (HPMCAS)-based ASDs were explored using surface area normalized dissolution. The tendency of the drug to form an insoluble complex with HPMCAS was determined through coprecipitation experiments. The role of pH and the extent of drug ionization were probed to evaluate the role of electrostatic interactions in complex formation. Relationships between the extent of complexation and the drug release rate from an ASD were observed, whereby the drugs could be divided into two groups. Drugs with a low extent of insoluble complex formation with HPMCAS tended to be neutral or anionic and showed reasonable release at pH 6.8 even at higher drug loadings. Cationic drugs formed insoluble complexes with HPMCAS and showed poor release when formulated as an ASD. Thus, and somewhat counterintuitively, a weakly basic drug showed a reduced release rate from an ASD at a bulk solution pH where it was ionized, relative to when unionized. The opposite trend was observed in the absence of polymer for the neat amorphous drug. In conclusion, electrostatic interactions between HPMCAS and lipophilic cationic drugs led to insoluble complex formation, which in turn resulted in ASDs with poor release performance.
Collapse
Affiliation(s)
- Pradnya Bapat
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Shubhajit Paul
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Yin-Chao Tseng
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
49
|
Pinto SFT, Santos HA, Sarmento BFCC. New insights into nanomedicines for oral delivery of glucagon-like peptide-1 analogs. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1952. [PMID: 38500351 DOI: 10.1002/wnan.1952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/23/2024] [Accepted: 02/21/2024] [Indexed: 03/20/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder that arises when the body cannot respond fully to insulin, leading to impaired glucose tolerance. Currently, the treatment embraces non-pharmacological actions (e.g., diet and exercise) co-associated with the administration of antidiabetic drugs. Metformin is the first-line treatment for T2DM; nevertheless, alternative therapeutic strategies involving glucagon-like peptide-1 (GLP-1) analogs have been explored for managing the disease. GLP-1 analogs trigger insulin secretion and suppress glucagon release in a glucose-dependent manner thereby, reducing the risk of hyperglycemia. Additionally, GLP-1 analogs have an extended plasma half-life compared to the endogenous peptide due to their high resistance to degradation by dipeptidyl peptidase-4. However, GLP-1 analogs are mainly administered via subcutaneous route, which can be inconvenient for the patients. Even considering an oral delivery approach, GLP-1 analogs are exposed to the harsh conditions of the gastrointestinal tract (GIT) and the intestinal barriers (mucus and epithelium). Hereupon, there is an unmet need to develop non-invasive oral transmucosal drug delivery strategies, such as the incorporation of GLP-1 analogs into nanoplatforms, to overcome the GIT barriers. Nanotechnology has the potential to shield antidiabetic peptides against the acidic pH and enzymatic activity of the stomach. In addition, the nanoparticles can be coated and/or surface-conjugated with mucodiffusive polymers and target intestinal ligands to improve their transport through the intestinal mucus and epithelium. This review focuses on the main hurdles associated with the oral administration of GLP-1 and GLP-1 analogs, and the nanosystems developed to improve the oral bioavailability of the antidiabetic peptides. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Soraia Filipa Tavares Pinto
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Hélder Almeida Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bruno Filipe Carmelino Cardoso Sarmento
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Instituto Universitário de Ciências da Saúde (IUCS-CESPU), Gandra, Portugal
| |
Collapse
|
50
|
Longre S, Rana D, Rangra S, Jindal AB, Salave S, Vitore J, Benival D. Quality-by-Design Based Development of Doxycycline Hyclate-Loaded Polymeric Microspheres for Prolonged Drug Release. AAPS PharmSciTech 2024; 25:49. [PMID: 38424393 DOI: 10.1208/s12249-024-02760-7] [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: 10/07/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
This study explores a novel approach to address the challenges of delivering highly water-soluble drug molecules by employing hydrophobic ion-pairing (HIP) complexes within poly (lactic-co-glycolic acid) (PLGA) microspheres. The HIP complex, formed between doxycycline hyclate (DH) and docusate sodium (DS), renders the drug hydrophobic. The development of the microspheres was done using the QbD approach, namely, Box-Behnken Design (BBD). A comprehensive characterization of the HIP complex confirmed the successful conversion of DH. DH and the HIP complex were effectively loaded into PLGA microspheres using the oil-in-water (O/W) emulsion solvent evaporation method. Results demonstrated significant improvements in percentage entrapment efficiency (% EE) and drug loading (% DL) for DH within the HIP complex-loaded PLGA microspheres compared to DH-loaded microspheres alone. Additionally, the initial burst release of DH reduced to 3% within the initial 15 min, followed by sustained drug release over 8 days. The modified HIP complex strategy offers a promising platform for improving the delivery of highly water-soluble small molecules. It provides high % EE, % DL, minimal initial burst release, and sustained release, thus having the potential to enhance patient compliance and drug delivery efficiency.
Collapse
Affiliation(s)
- Suraj Longre
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, India
| | - Dhwani Rana
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, India
| | - Shagun Rangra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, India
| | - Anil B Jindal
- Department of Pharmacy, Birla Institute of Technology and Science Pilani (BITS PILANI), Pilani Campus, Rajasthan, 333031, India
| | - Sagar Salave
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, India
| | - Jyotsna Vitore
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, India
| | - Derajram Benival
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, India.
| |
Collapse
|