1
|
Gulyaev IA, Sokol MB, Mollaeva MR, Klimenko MA, Yabbarov NG, Chirkina MV, Nikolskaya ED. Polymeric Drug Delivery Systems in Biomedicine. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S233-S262. [PMID: 40164161 DOI: 10.1134/s0006297924603976] [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: 09/24/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 04/02/2025]
Abstract
Our review examines the key aspects of using polymeric carriers in biomedicine. The section "Polymers for Biomedicine" provides an overview of different types of polymers, their structural features and properties that determine their use as drug delivery vehicles. The section "Polymeric Carriers" characterizes the role of polymeric delivery systems in modern medicine. The main forms of polymeric carriers are described, as well as their key advantages for drug delivery. The section "Preclinical and Clinical Trials of Polymeric Drug Carriers" reviews the examples of clinical and preclinical studies of polymeric forms used for antitumor therapy, therapy for bacterial and infectious diseases. The final section "Targeted Drug Delivery Systems" is devoted to the discussion of approaches, as well as ligands that provide targeted drug delivery using polymeric carriers. We have paid special attention to modern approaches for increasing the efficacy of antibacterial therapy using vector molecules.
Collapse
Affiliation(s)
- Ivan A Gulyaev
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Maria B Sokol
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Mariia R Mollaeva
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Maksim A Klimenko
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Nikita G Yabbarov
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Margarita V Chirkina
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Elena D Nikolskaya
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia.
| |
Collapse
|
2
|
Cheng X, Pan R, Tang J, Yu K, Zhang H, Zhao X. Preliminary Study on Pharmacokinetics and Antitumor Pharmacodynamics of Folic Acid Modified Crebanine Polyethyleneglycol-Polylactic Acid Hydroxyacetic Acid Copolymer Nanoparticles. Int J Nanomedicine 2024; 19:10513-10536. [PMID: 39435040 PMCID: PMC11492913 DOI: 10.2147/ijn.s477027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024] Open
Abstract
Purpose Liver cancer is associated significantly with morbidity and mortality. The combination of low-intensity ultrasound with nanomedicine delivery systems holds promise as an alternative for the treatment for liver cancer. This study focuses on the utilization of folic acid (FA) modified nanoparticles, which are loaded with fluorescent dye DiR and liquid fluorocarbon (PFP). These nanoparticles have the potential to enhance liver cancer targeting under ultrasound stimulation and future applications in vivo. Methods The pharmacokinetics and tissue distribution of folic acid-modified Crebanine polyethylene glycol-polylactic acid copolymer nanoparticles (FA-Cre@PEG-PLGA NPs) were investigated. The pharmacokinetic parameters, liver targeting, and in vivo distribution were assessed. Additionally, the inhibitory impacts of FA-Cre@PEG-PLGA NPs in combination with ultrasonic irradiation on the proliferation and acute toxicity of H22 cells of mouse hepatoma were investigated in vitro. The tumor targeting and anti-tumor efficacy of FA-Cre@PEG-PLGA NPs were assessed utilizing a small animal in vivo imaging system and an in situ hepatocellular carcinoma transplantation model, respectively. Results The pharmacokinetic studies and tissue distribution tests demonstrated that FA-Cre@PEG-PLGA NPs conspicuously prolonged the half-life and retention time of the drug in rats, and the liver targeting effect was pronounced. Additionally, the in vivo acute toxicity test indicated that FA-Cre@PEG-PLGA NPs had minimal adverse reactions and could fulfill the aim of attenuating the drug. The outcomes of the animal experiments further substantiated that FA-Cre@PEG-PLGA NPs had a longer retention time at the tumor site, a superior anti-tumor effect, and less damage to liver and kidney tissue. Conclusion The integration of FA-Cre@PEG-PLGA NPs with ultrasound irradiation demonstrated exceptional safety and potent anti-tumor efficacy in vivo, presenting a promising therapeutic strategy for the treatment of liver cancer through the combination of ultrasound technology with a nanomedicine delivery system.
Collapse
Affiliation(s)
- Xin Cheng
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
- The Key Laboratory of External Drug Delivery System and Preparation Technology in University of Yunnan Province, Kunming, 650500, People’s Republic of China
- Yunnan Key Laboratory of Dai and Yi Medicines, Kunming, 650500, People’s Republic of China
- Laboratory Animal Center, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
| | - Rui Pan
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
- Yunnan Key Laboratory of Dai and Yi Medicines, Kunming, 650500, People’s Republic of China
- Laboratory Animal Center, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Kunming, 650500, People’s Republic of China
| | - Junze Tang
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
- Laboratory Animal Center, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
| | - Kun Yu
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
- Laboratory Animal Center, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
| | - Hailiang Zhang
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
- Yunnan Key Laboratory of Dai and Yi Medicines, Kunming, 650500, People’s Republic of China
- Laboratory Animal Center, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Kunming, 650500, People’s Republic of China
| | - Xiaoyu Zhao
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
- Laboratory Animal Center, Yunnan University of Chinese Medicine, Kunming, 650500, People’s Republic of China
| |
Collapse
|
3
|
Saddam Hussain M, Khetan R, Albrecht H, Krasowska M, Blencowe A. Oligoelectrolyte-mediated, pH-triggered release of hydrophobic drugs from non-responsive micelles: Influence of oligo(2-vinyl pyridine)-loading on drug-loading, release and cytotoxicity. Int J Pharm 2024; 661:124368. [PMID: 38925236 DOI: 10.1016/j.ijpharm.2024.124368] [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: 04/29/2024] [Revised: 06/03/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
pH-responsive polymeric micelles have been extensively studied for nanomedicine and take advantage of pH differentials in tissues for the delivery of large doses of cytotoxic drugs at specific target sites. Despite significant advances in this area, there is a lack of versatile and adaptable strategies to render micelles pH-responsive that could be widely applied to different payloads and applications. To address this deficiency, we introduce the concept of oligoelectrolyte-mediated, pH-triggered release of hydrophobic drugs from non-responsive polymeric micelles as a highly effective approach with broad scope. Herein, we investigate the influence of the oligoelectrolyte, oligo(2-vinyl pyridine) (OVP), loading and polymer molecular weight on the pH-sensitivity, drug loading/release and cytotoxicity of poly(ethylene glycol-b-ε-caprolactone) (PEG-b-PCL) micelles using copolymers with either short or long hydrophobic blocks (PEG4PCL4 and PEG10PCL10, respectively). The micelles were characterized as a function of pH (7.4 to 3.5). Dynamic light scattering (DLS) revealed narrow particle size distributions (PSDs) for both the blank and OVP-loaded micelles at pH 7.4. While OVP encapsulation resulted in an increase in the hydrodynamic diameter (Dh) (cf. blank micelles), a decrease in the pH below 6.5 led to a decrease in the Dh consistent with the ionization and release of OVP and core collapse, which were further supported by proton nuclear magnetic resonance (1H NMR) spectroscopy and UV-visible (UV-vis) spectrophotometry. The change in zeta potential (ζ) with pH for the OVP-loaded PEG4PCL4 and PEG10PCL10 micelles was different, suggesting that the location/distribution of OVP in the micelles is influenced by the polymer molecular weight. In general, co-encapsulation of drugs (doxorubicin (DOX), gossypol (GP), paclitaxel (PX) or 7-ethyl-10-hydroxycamptothecin (SN38)) and OVP in the micelles proceeded efficiently with high encapsulation efficiency percentages (EE%). In vitro release studies revealed the rapid, pH-triggered release of drugs from OVP-loaded PEG10PCL10 micelles within hours, with higher OVP loadings providing faster and more complete release. In comparison, no triggered release was observed for the OVP-loaded PEG4PCL4 micelles, implying a strong molecular weight dependency. In metabolic assays the drug- and OVP-loaded PEG10PCL10 micelles were found to result in significant enhancement of the cytotoxicity compared to drug-loaded micelles (no OVP) or other controls. Importantly, micelles with low OVP loadings were found to be nearly as effective as those with high OVP loadings. These results provide key insights into the tunability of the oligoelectrolyte-mediated approach for the effective formulation of pH-responsive micelles and pH-triggered drug release.
Collapse
Affiliation(s)
- Md Saddam Hussain
- Applied Chemistry and Translational Biomaterials (ACTB) Group, Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, SA 5000, Australia; Department of Pharmacy, Faculty of Science, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Riya Khetan
- Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, SA, 5000, Australia
| | - Hugo Albrecht
- Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, SA, 5000, Australia
| | - Marta Krasowska
- Future Industries Institute, UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Anton Blencowe
- Applied Chemistry and Translational Biomaterials (ACTB) Group, Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
4
|
Assiri AA, Glover K, Mishra D, Waite D, Vora LK, Thakur RRS. Block copolymer micelles as ocular drug delivery systems. Drug Discov Today 2024; 29:104098. [PMID: 38997002 DOI: 10.1016/j.drudis.2024.104098] [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: 02/27/2024] [Revised: 06/07/2024] [Accepted: 07/06/2024] [Indexed: 07/14/2024]
Abstract
Block copolymer micelles, formed by the self-assembly of amphiphilic polymers, address formulation challenges, such as poor drug solubility and permeability. These micelles offer advantages including a smaller size, easier preparation, sterilization, and superior solubilization, compared with other nanocarriers. Preclinical studies have shown promising results, advancing them toward clinical trials. Their mucoadhesive properties enhance and prolong contact with the ocular surface, and their small size allows deeper penetration through tissues, such as the cornea. Additionally, copolymeric micelles improve the solubility and stability of hydrophobic drugs, sustain drug release, and allow for surface modifications to enhance biocompatibility. Despite these benefits, long-term stability remains a challenge. In this review, we highlight the preclinical performance, structural frameworks, preparation techniques, physicochemical properties, current developments, and prospects of block copolymer micelles as ocular drug delivery systems.
Collapse
Affiliation(s)
- Ahmad A Assiri
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast, UK; Department of Pharmacognosy, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Katie Glover
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast, UK
| | - Deepakkumar Mishra
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast, UK
| | - David Waite
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast, UK
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast, UK.
| | | |
Collapse
|
5
|
Fu WY, Chiu YL, Huang SC, Huang WY, Hsu FT, Lee HY, Wang TW, Keng PY. Boron Neutron Capture Therapy Enhanced by Boronate Ester Polymer Micelles: Synthesis, Stability, and Tumor Inhibition Studies. Biomacromolecules 2024; 25:4215-4232. [PMID: 38845149 PMCID: PMC11238341 DOI: 10.1021/acs.biomac.4c00298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 07/09/2024]
Abstract
Boron neutron capture therapy (BNCT) targets invasive, radioresistant cancers but requires a selective and high B-10 loading boron drug. This manuscript investigates boron-rich poly(ethylene glycol)-block-(poly(4-vinylphenyl boronate ester)) polymer micelles synthesized via atom transfer radical polymerization for their potential application in BNCT. Transmission electron microscopy (TEM) revealed spherical micelles with a uniform size of 43 ± 10 nm, ideal for drug delivery. Additionally, probe sonication proved effective in maintaining the micelles' size and morphology postlyophilization and reconstitution. In vitro studies with B16-F10 melanoma cells demonstrated a 38-fold increase in boron accumulation compared to the borophenylalanine drug for BNCT. In vivo studies in a B16-F10 tumor-bearing mouse model confirmed enhanced tumor selectivity and accumulation, with a tumor-to-blood (T/B) ratio of 2.5, surpassing BPA's T/B ratio of 1.8. As a result, mice treated with these micelles experienced a significant delay in tumor growth, highlighting their potential for BNCT and warranting further research.
Collapse
Affiliation(s)
- Wan Yun Fu
- Department of Material Science
and Engineering, National Tsing Hua University, Hsinchu City 300, Taiwan
| | - Yi-Lin Chiu
- Department of Material Science
and Engineering, National Tsing Hua University, Hsinchu City 300, Taiwan
| | - Shi-Chih Huang
- Department of Material Science
and Engineering, National Tsing Hua University, Hsinchu City 300, Taiwan
| | - Wei-Yuan Huang
- Department of Material Science
and Engineering, National Tsing Hua University, Hsinchu City 300, Taiwan
| | - Fang-Tzu Hsu
- Department of Material Science
and Engineering, National Tsing Hua University, Hsinchu City 300, Taiwan
| | - Han Yu Lee
- Department of Material Science
and Engineering, National Tsing Hua University, Hsinchu City 300, Taiwan
| | - Tzu-Wei Wang
- Department of Material Science
and Engineering, National Tsing Hua University, Hsinchu City 300, Taiwan
| | - Pei Yuin Keng
- Department of Material Science
and Engineering, National Tsing Hua University, Hsinchu City 300, Taiwan
| |
Collapse
|
6
|
Abstract
The recent emergence of nanomedicine has revolutionized the therapeutic landscape and necessitated the creation of more sophisticated drug delivery systems. Polymeric nanoparticles sit at the forefront of numerous promising drug delivery designs, due to their unmatched control over physiochemical properties such as size, shape, architecture, charge, and surface functionality. Furthermore, polymeric nanoparticles have the ability to navigate various biological barriers to precisely target specific sites within the body, encapsulate a diverse range of therapeutic cargo and efficiently release this cargo in response to internal and external stimuli. However, despite these remarkable advantages, the presence of polymeric nanoparticles in wider clinical application is minimal. This review will provide a comprehensive understanding of polymeric nanoparticles as drug delivery vehicles. The biological barriers affecting drug delivery will be outlined first, followed by a comprehensive description of the various nanoparticle designs and preparation methods, beginning with the polymers on which they are based. The review will meticulously explore the current performance of polymeric nanoparticles against a myriad of diseases including cancer, viral and bacterial infections, before finally evaluating the advantages and crucial challenges that will determine their wider clinical potential in the decades to come.
Collapse
Affiliation(s)
- Maximilian
A. Beach
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Umeka Nayanathara
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yanting Gao
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Changhe Zhang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yijun Xiong
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yufu Wang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Georgina K. Such
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
7
|
Shalmani AA, Ahmed Z, Sheybanifard M, Wang A, Weiler M, Buhl EM, Klinkenberg G, Schmid R, Hennink W, Kiessling F, Metselaar JM, Lammers T, Peña Q, Shi Y. Effect of Radical Polymerization Method on Pharmaceutical Properties of Π Electron-Stabilized HPMA-Based Polymeric Micelles. Biomacromolecules 2023; 24:4444-4453. [PMID: 36753733 DOI: 10.1021/acs.biomac.2c01261] [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: 02/10/2023]
Abstract
Polymeric micelles are among the most extensively used drug delivery systems. Key properties of micelles, such as size, size distribution, drug loading, and drug release kinetics, are crucial for proper therapeutic performance. Whether polymers from more controlled polymerization methods produce micelles with more favorable properties remains elusive. To address this question, we synthesized methoxy poly(ethylene glycol)-b-(N-(2-benzoyloxypropyl)methacrylamide) (mPEG-b-p(HPMAm-Bz)) block copolymers of three different comparable molecular weights (∼9, 13, and 20 kDa), via both conventional free radical (FR) and reversible addition-fragmentation chain transfer (RAFT) polymerization. The polymers were subsequently employed to prepare empty and paclitaxel-loaded micelles. While FR polymers had relatively high dispersities (Đ ∼ 1.5-1.7) compared to their RAFT counterparts (Đ ∼ 1.1-1.3), they formed micelles with similar pharmaceutical properties (e.g., size, size distribution, critical micelle concentration, cytotoxicity, and drug loading and retention). Our findings suggest that pharmaceutical properties of mPEG-b-p(HPMAm-Bz) micelles do not depend on the synthesis route of their constituent polymers.
Collapse
Affiliation(s)
- Armin Azadkhah Shalmani
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Zaheer Ahmed
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Maryam Sheybanifard
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Alec Wang
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Marek Weiler
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Institute of Pathology, RWTH University Hospital, 52074 Aachen, Germany
| | - Geir Klinkenberg
- Department of Biotechnology and Nanomedicine, SINTEF Industry, 7034, Trondheim, Norway
| | - Ruth Schmid
- Department of Biotechnology and Nanomedicine, SINTEF Industry, 7034, Trondheim, Norway
| | - Wim Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Josbert M Metselaar
- 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
| | - Yang Shi
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| |
Collapse
|
8
|
Hebels ER, Bindt F, Walther J, van Geijn M, Weterings J, Hu Q, Colombo C, Liskamp R, Rijcken C, Hennink WE, Vermonden T. Orthogonal Covalent Entrapment of Cargo into Biodegradable Polymeric Micelles via Native Chemical Ligation. Biomacromolecules 2023; 24:4385-4396. [PMID: 36044412 PMCID: PMC10565831 DOI: 10.1021/acs.biomac.2c00865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/18/2022] [Indexed: 11/29/2022]
Abstract
Polymeric micelles (PMs) are promising platforms for enhanced tissue targeting of entrapped therapeutic agents. Strategies to circumvent premature release of entrapped drugs include cross-linking of the micellar core as well as covalent attachment of the drug cargo. The chemistry employed to obtain cross-linked micelles needs to be mild to also allow entrapment of fragile molecules, such as certain peptides, proteins, oligonucleotides, and fluorescent dyes. Native chemical ligation (NCL) is a mild bio-orthogonal reaction between a N-terminal cysteine residue and a thioester that proceeds under physiological conditions. Here, we designed a trifunctional cross-linker containing two cysteine residues for the micelle core-cross-linking reaction and an azide residue for ring-strained alkyne conjugation of fluorescent dyes. We applied this approach to thermosensitive methoxypolyethylene glycol-b-N-(2-hydroxypropyl)methacrylamide-lactate (mPEG-b-HPMAmLacn) based block copolymers of a core-cross-linked polymeric micelle (CCPM) system by attaching thioester residues (using ethyl thioglycolate-succinic anhydride, ETSA) for NCL cross-linking with the trifunctional cross-linker under physiological conditions. By use of mild copper-free click chemistry, we coupled fluorescent dyes, Sulfo.Cy5 and BODIPY, to the core via the azide residue present on the cross-linker by triazole ring formation. In addition, we employed a recently developed cycloheptyne strain promoted click reagent (TMTHSI, CliCr) in comparison to the frequently employed cyclooctyne derivative (DBCO), both achieving successful dye entrapment. The size of the resulting CCPMs could be tuned between 50 and 100 nm by varying the molecular weight of the thermosensitive block and ETSA content. In vitro cell experiments showed successful internalization of the dye entrapped CCPMs, which did not affect cell viability up to a polymer concentration of 2 mg/mL in PC3 cells. These fluorescent dye entrapped CCPMs can be applied in diagnostic imaging and the chemistry developed in this study serves as a steppingstone toward covalently entrapped fragile drug compounds with tunable release in CCPMs.
Collapse
Affiliation(s)
- Erik R. Hebels
- Division
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Felix Bindt
- Division
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Johanna Walther
- Division
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands
| | | | | | - Qizhi Hu
- Cristal
Therapeutics, 6229 EV Maastricht, The Netherlands
| | | | - Rob Liskamp
- Cristal
Therapeutics, 6229 EV Maastricht, The Netherlands
| | | | - Wim E. Hennink
- Division
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Tina Vermonden
- Division
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
9
|
Zhang F, Pei G, Huang B, Xu J, Zhang L. Exploring release mechanisms by disrupting π-π stacking regions in stable micelles. J Mater Chem B 2023; 11:9246-9259. [PMID: 37721031 DOI: 10.1039/d3tb01388j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
π-π stacking strategies can enhance the stability performance of delivery platforms but are often restricted by incomplete drug release performance, even with the help of crosslinking strategies. Therefore, there has been considerable interest in enhancing the drug release performance by disrupting the π-π stacking region (structural rearrangements). Herein, we synthesized poly(3-(isobutyloxy)-2-oxopropyl benzoate)-b-poly(2-hydroxybutyl methacrylate)-co-poly((ethylene glycol)methylether methacrylate) [PBOOPMA-b-P(HBMA-co-PEGMA), PHB] and revealed the drug release mechanism of PHB-based micelles. The structural rearrangements derived from the crosslinking strategy were revealed to improve the early release performance by 43-55% using micellar dissolutions. Moreover, the esterase-responsive strategy was elucidated to induce reassembly with 77-79% size variation, intensifying the structural rearrangements, which was also synergistic with the crosslinking strategy. Based on the advantages of improving drug release performance, the esterase-responsive strategy was considered a promising candidate for enhancing late release performance. Meanwhile, it is believed that such responsive modulation (crosslinking, esterase-responsive) in the π-π stacking region will become highly promising for subsequent research. Finally, the biosafety of 95.81% at 400 mg L-1 and drug cytotoxicity of IC50 ≈ 2.5 mg L-1 of PHB-EDE@CPT were also validated, confirming the broad application prospects of PHB-based crosslinked micelles.
Collapse
Affiliation(s)
- Fusheng Zhang
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Gongcui Pei
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Baihao Huang
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Jianchang Xu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lijuan Zhang
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
10
|
Wei G, Zhang S, Yu S, Lu W. Intravital Microscopy Reveals Endothelial Transcytosis Contributing to Significant Tumor Accumulation of Albumin Nanoparticles. Pharmaceutics 2023; 15:519. [PMID: 36839841 PMCID: PMC9960641 DOI: 10.3390/pharmaceutics15020519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
The principle of enhanced permeability and retention (EPR) effect has been used to design anti-cancer nanomedicines over decades. However, it is being challenged due to the poor clinical outcome of nanoparticles and controversial physiological foundation. Herein, we use a near-infrared-II (1000-1700 nm, NIR-II) fluorescence probe BPBBT to investigate the pathway for the entry of human serum albumin-bound nanoparticles (BPBBT-HSA NPs) into tumor compared with BPBBT micelles with phospholipid-poly (ethylene glycol) of the similar particle size about 110 nm. The plasma elimination half-life of BPBBT micelles was 2.8-fold of that of BPBBT-HSA NPs. However, the area under the BPBBT concentration in tumor-time curve to 48 h post-injection (AUCtumor0→48h) of BPBBT-HSA NPs was 7.2-fold of that of BPBBT micelles. The intravital NIR-II fluorescence microscopy revealed that BPBBT-HSA NPs but not BPBBT micelles were transported from the tumor vasculature into tumor parenchyma with high efficiency, and endocytosed by the tumor cells within 3 h post-injection in vivo. This effect was blocked by cross-linking BPBBT-HSA NPs to denature HSA, resulting in the AUCtumor0→48h decreased to 22% of that of BPBBT-HSA NPs. Our results demonstrated that the active process of endothelial transcytosis is the dominant pathway for albumin-bound nanoparticles' entry into tumor.
Collapse
Affiliation(s)
| | | | | | - Wei Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy & Minhang Hospital, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
11
|
Rijcken CJF, De Lorenzi F, Biancacci I, Hanssen RGJM, Thewissen M, Hu Q, Atrafi F, Liskamp RMJ, Mathijssen RHJ, Miedema IHC, Menke-van der Houven van Oordt CW, van Dongen GAMS, Vugts DJ, Timmers M, Hennink WE, Lammers T. Design, development and clinical translation of CriPec®-based core-crosslinked polymeric micelles. Adv Drug Deliv Rev 2022; 191:114613. [PMID: 36343757 DOI: 10.1016/j.addr.2022.114613] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/18/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Nanomedicines are used to improve the efficacy and safety of pharmacotherapeutic interventions. Unraveling the biological behavior of nanomedicines, including their biodistribution and target site accumulation, is essential to establish design criteria that contribute to superior performance. CriPec® technology is based on amphiphilic methoxy-poly(ethylene glycol)-b-poly[N-(2-hydroxypropyl) methacrylamide lactate] (mPEG-b-pHPMAmLacn) block copolymers, which are designed to upon self-assembly covalently entrap active pharmaceutical ingredients (API) in core-crosslinked polymeric micelles (CCPM). Key features of CCPM are a prolonged circulation time, high concentrations at pathological sites, and low levels of accumulation in the majority of healthy tissues. Proprietary hydrolysable linkers allow for tunable and sustained release of entrapped API, including hydrophobic and hydrophilic small molecules, as well as peptides and oligonucleotides. Preclinical imaging experiments provided valuable information on their tumor and tissue accumulation and distribution, as well as on uptake by cancer, healthy and immune cells. The frontrunner formulation CPC634, which refers to 65 nm-sized CCPM entrapping the chemotherapeutic drug docetaxel, showed excellent pharmacokinetic properties, safety, tumor accumulation and antitumor efficacy in multiple animal models. In the clinic, CPC634 also demonstrated favorable pharmacokinetics, good tolerability, signs of efficacy, and enhanced localization in tumor tissue as compared to conventional docetaxel. PET imaging of radiolabeled CPC634 showed quantifiable accumulation in ∼50 % of tumors and metastases in advanced-stage cancer patients, and demonstrated potential for use in a theranostic setting even when applied at a companion diagnostic dose. Altogether, the preclinical and clinical results obtained to date demonstrate that mPEG-b-pHPMAmLacn CCPM based on CriPec® technology are a potent, tunable, broadly applicable and well-tolerable platform for targeted drug delivery and improved anticancer therapy.
Collapse
Affiliation(s)
| | - Federica De Lorenzi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Ilaria Biancacci
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | | | | | - Qizhi Hu
- Cristal Therapeutics, Maastricht, the Netherlands
| | - Florence Atrafi
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Iris H C Miedema
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| | - C Willemien Menke-van der Houven van Oordt
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| | - Guus A M S van Dongen
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Danielle J Vugts
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Matt Timmers
- Cristal Therapeutics, Maastricht, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht University, Utrecht, the Netherlands
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
12
|
Poellmann MJ, Javius-Jones K, Hopkins C, Lee JW, Hong S. Dendritic-Linear Copolymer and Dendron Lipid Nanoparticles for Drug and Gene Delivery. Bioconjug Chem 2022; 33:2008-2017. [PMID: 35512322 DOI: 10.1021/acs.bioconjchem.2c00128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Polymers constitute a diverse class of macromolecules that have demonstrated their unique advantages to be utilized for drug or gene delivery applications. In particular, polymers with a highly ordered, hyperbranched structure─"dendrons"─offer significant benefits to the design of such nanomedicines. The incorporation of dendrons into block copolymer micelles can endow various unique properties that are not typically observed from linear polymer counterparts. Specifically, the dendritic structure induces the conical shape of unimers that form micelles, thereby improving the thermodynamic stability and achieving a low critical micelle concentration (CMC). Furthermore, through a high density of highly ordered functional groups, dendrons can enhance gene complexation, drug loading, and stimuli-responsive behavior. In addition, outward-branching dendrons can support a high density of nonfouling polymers, such as poly(ethylene glycol), for serum stability and variable densities of multifunctional groups for multivalent cellular targeting and interactions. In this paper, we review the design considerations for dendron-lipid nanoparticles and dendron micelles formed from amphiphilic block copolymers intended for gene transfection and cancer drug delivery applications. These technologies are early in preclinical development and, as with other nanomedicines, face many obstacles on the way to clinical adoption. Nevertheless, the utility of dendron micelles for drug delivery remains relatively underexplored, and we believe there are significant and dramatic advancements to be made in tumor targeting with these platforms.
Collapse
Affiliation(s)
- Michael J Poellmann
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Kaila Javius-Jones
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Caroline Hopkins
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Jin Woong Lee
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States.,Wisconsin Center for NanoBioSystems, University of Wisconsin, Madison, Wisconsin 53705, United States.,Yonsei Frontier Lab and Department of Pharmacy, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
13
|
Timmers M, Weterings J, van Geijn M, Bell R, Lenting PE, Rijcken CJ, Vermonden T, Hennink WE, Liskamp RM. A New Class of Tunable Acid-Sensitive Linkers for Native Drug Release Based on the Trityl Protecting Group. Bioconjug Chem 2022; 33:1707-1715. [PMID: 35979909 PMCID: PMC9501768 DOI: 10.1021/acs.bioconjchem.2c00310] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/03/2022] [Indexed: 12/24/2022]
Abstract
Core-cross-linked polymeric micelles (CCPMs) are a promising nanoparticle platform due to favorable properties such as their long circulation and tumor disposition exploiting the enhanced permeability and retention (EPR) effect. Sustained release of covalently linked drugs from the hydrophobic core of the CCPM can be achieved by a biodegradable linker that connects the drug and the core. This study investigates the suitability of trityl-based linkers for the design of acid-triggered native active pharmaceutical ingredient (API) release from CCPMs. Trityl linker derivatives with different substituent patterns were synthesized and conjugated to model API compounds such as DMXAA-amine, doxorubicin, and gemcitabine, and their release kinetics were studied. Hereafter, API release from CCPMs based on mPEG-b-pHPMAmLac block copolymers was investigated. Variation of the trityl substitution pattern showed tunability of the API release rate from the trityl-based linker with t1/2 varying from <1.0 to 5.0 h at pH 5.0 and t1/2 from 6.5 to >24 h at pH 7.4, all at 37 °C. A clear difference in release kinetics was found between gemcitabine and doxorubicin, with gemcitabine showing no detectable release for 72 h at pH 5.0 and doxorubicin showing a t1/2 of less than 1 h. Based on these findings, we show that the reaction mechanism of trityl deprotection plays an important role in the API release kinetics. The first step in this mechanism, which is protonation of the trityl-bound amine, is pKa-dependent, which explains the difference in release rate. In conclusion, acid-sensitive and tunable trityl linkers are highly promising for the design of linker-API conjugates and for their use in CCPMs.
Collapse
Affiliation(s)
- Matt Timmers
- Cristal
Therapeutics, Maastricht 6229 EV, The Netherlands
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
| | | | | | - Roel Bell
- Symeres, Nijmegen 6546 BB, The
Netherlands
| | - Peter E. Lenting
- School
of Chemistry, University of Glasgow, Glasgow G12 8QQ, U.K.
| | | | - Tina Vermonden
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Wim E. Hennink
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Rob M.J. Liskamp
- Cristal
Therapeutics, Maastricht 6229 EV, The Netherlands
- School
of Chemistry, University of Glasgow, Glasgow G12 8QQ, U.K.
- Department
of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht 6229 ER, The Netherlands
| |
Collapse
|
14
|
Wang Y, Fens MH, van Kronenburg NCH, Shi Y, Lammers T, Heger M, van Nostrum CF, Hennink WE. Magnetic beads for the evaluation of drug release from biotinylated polymeric micelles in biological media. J Control Release 2022; 349:954-962. [PMID: 35931210 DOI: 10.1016/j.jconrel.2022.07.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/22/2022] [Accepted: 07/31/2022] [Indexed: 11/16/2022]
Abstract
To improve the reliability of in vitro release studies of drug delivery systems, we developed a novel in vitro method for the evaluation of drug release from polymeric micelles in complex biological media. Polymeric micelles based on poly(N-2-hydroxypropyl methacrylamide)-block-poly(N-2-benzoyloxypropyl methacrylamide) (p(HPMAm)-b-p(HPMAm-Bz)) of which 10% of the chains was functionalized with biotin at the p(HPMAm) terminus were prepared using a solvent extraction method. The size of the micelles when loaded with a hydrophobic agent, namely paclitaxel (a clinically used cytostatic drug) or curcumin (a compound with multiple pharmacological activities), was around 65 nm. The biotin decoration allowed the binding of the micelles to streptavidin-coated magnetic beads which occurred within 10 min and reached a binding efficiency of 90 ± 6%. Drug release in different media was studied after the magnetic separation of micelles bound to the streptavidin-coated beads, by determination of the released drug in the media as well as the retained drug in the micellar fraction bound to the beads. The in vitro release of paclitaxel and curcumin at 37 °C in PBS, PBS containing 2% v/v Tween 80, PBS containing 4.5% w/v bovine serum albumin, mouse plasma, and whole mouse blood was highly medium-dependent. In all media studied, paclitaxel showed superior micellar retention compared to curcumin. Importantly, the presence of serum proteins accelerated the release of both paclitaxel and curcumin. The results presented in this study show great potential for predicting drug release from nanomedicines in biological media which in turn is crucial for their further pharmaceutical development.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands
| | - Marcel H Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands
| | - Nicky C H van Kronenburg
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Yang Shi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbecktrasse 55, 52074 Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbecktrasse 55, 52074 Aachen, Germany
| | - Michal Heger
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands; Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing 314001, Zhejiang, PR China
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| |
Collapse
|
15
|
The in vivo fate of polymeric micelles. Adv Drug Deliv Rev 2022; 188:114463. [PMID: 35905947 DOI: 10.1016/j.addr.2022.114463] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/10/2022] [Accepted: 07/15/2022] [Indexed: 12/12/2022]
Abstract
This review aims to provide a systemic analysis of the in vivo, as well as subcellular, fate of polymeric micelles (PMs), starting from the entry of PMs into the body. Few PMs are able to cross the biological barriers intact and reach the circulation. In the blood, PMs demonstrate fairly good stability mainly owing to formation of protein corona despite controversial results reported by different groups. Although the exterior hydrophilic shells render PMs "long-circulating", the biodistribution of PMs into the mononuclear phagocyte systems (MPS) is dominant as compared with non-MPS organs and tissues. Evidence emerges to support that the copolymer poly(ethylene glycol)-poly(lactic acid) (PEG-PLA) is first broken down into pieces of PEG and PLA and then remnants to be eliminated from the body finally. At the cellular level, PMs tend to be internalized via endocytosis due to their particulate nature and disassembled and degraded within the cell. Recent findings on the effect of particle size, surface characteristics and shape are also reviewed. It is envisaged that unraveling the in vivo and subcellular fate sheds light on the performing mechanisms and gears up the clinical translation of PMs.
Collapse
|
16
|
Varela-Moreira A, van Leur H, Krijgsman D, Ecker V, Braun M, Buchner M, H.A.M. Fens M, Hennink WE, Schiffelers RM. Utilizing In Vitro Drug Release Assays to Predict In Vivo Retention of Micelles. Int J Pharm 2022; 618:121638. [DOI: 10.1016/j.ijpharm.2022.121638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
|
17
|
Sadat SMA, Wuest M, Paiva IM, Munira S, Sarrami N, Sanaee F, Yang X, Paladino M, Binkhathlan Z, Karimi-Busheri F, Martin GR, Jirik FR, Murray D, Gamper AM, Hall DG, Weinfeld M, Lavasanifar A. Nano-Delivery of a Novel Inhibitor of Polynucleotide Kinase/Phosphatase (PNKP) for Targeted Sensitization of Colorectal Cancer to Radiation-Induced DNA Damage. Front Oncol 2022; 11:772920. [PMID: 35004293 PMCID: PMC8733593 DOI: 10.3389/fonc.2021.772920] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/24/2021] [Indexed: 12/29/2022] Open
Abstract
Inhibition of the DNA repair enzyme polynucleotide kinase/phosphatase (PNKP) increases the sensitivity of cancer cells to DNA damage by ionizing radiation (IR). We have developed a novel inhibitor of PNKP, i.e., A83B4C63, as a potential radio-sensitizer for the treatment of solid tumors. Systemic delivery of A83B4C63, however, may sensitize both cancer and normal cells to DNA damaging therapeutics. Preferential delivery of A83B4C63 to solid tumors by nanoparticles (NP) was proposed to reduce potential side effects of this PNKP inhibitor to normal tissue, particularly when combined with DNA damaging therapies. Here, we investigated the radio-sensitizing activity of A83B4C63 encapsulated in NPs (NP/A83) based on methoxy poly(ethylene oxide)-b-poly(α-benzyl carboxylate-ε-caprolactone) (mPEO-b-PBCL) or solubilized with the aid of Cremophor EL: Ethanol (CE/A83) in human HCT116 colorectal cancer (CRC) models. Levels of γ-H2AX were measured and the biodistribution of CE/A83 and NP/A83 administered intravenously was determined in subcutaneous HCT116 CRC xenografts. The radio-sensitization effect of A83B4C63 was measured following fractionated tumor irradiation using an image-guided Small Animal Radiation Research Platform (SARRP), with 24 h pre-administration of CE/A83 and NP/A83 to Luc+/HCT116 bearing mice. Therapeutic effects were analyzed by monitoring tumor growth and functional imaging using Positron Emission Tomography (PET) and [18F]-fluoro-3’-deoxy-3’-L:-fluorothymidine ([18F]FLT) as a radiotracer for cell proliferation. The results showed an increased persistence of DNA damage in cells treated with a combination of CE/A83 or NP/A83 and IR compared to those only exposed to IR. Significantly higher tumor growth delay in mice treated with a combination of IR and NP/A83 than those treated with IR plus CE/A83 was observed. [18F]FLT PET displayed significant functional changes for tumor proliferation for the drug-loaded NP. This observation was attributed to the higher A83B4C63 levels in the tumors for NP/A83-treated mice compared to those treated with CE/A83. Overall, the results demonstrated a potential for A83B4C63-loaded NP as a novel radio-sensitizer for the treatment of CRC.
Collapse
Affiliation(s)
- Sams M A Sadat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Melinda Wuest
- Department of Oncology, Cross Cancer Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Igor M Paiva
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Sirazum Munira
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Nasim Sarrami
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Forughalsadat Sanaee
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Xiaoyan Yang
- Department of Oncology, Cross Cancer Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Marco Paladino
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Ziyad Binkhathlan
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Feridoun Karimi-Busheri
- Department of Oncology, Cross Cancer Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Gary R Martin
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Frank R Jirik
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - David Murray
- Department of Oncology, Cross Cancer Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Armin M Gamper
- Department of Oncology, Cross Cancer Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Dennis G Hall
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Michael Weinfeld
- Department of Oncology, Cross Cancer Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Department of Chemical and Material Engineering, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
18
|
Wang Y, Thies-Weesie DM, Bosman ED, van Steenbergen MJ, van den Dikkenberg J, Shi Y, Lammers T, van Nostrum CF, Hennink WE. Tuning the size of all-HPMA polymeric micelles fabricated by solvent extraction. J Control Release 2022; 343:338-346. [DOI: 10.1016/j.jconrel.2022.01.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 12/14/2022]
|
19
|
Zhang X, Wei Z, Liu K, Wang L, Yang W. A 3B-type miktoarm star polymer nanoassemblies prepared by reversible addition–fragmentation chain transfer (RAFT) dispersion polymerization. Polym Chem 2022. [DOI: 10.1039/d2py00935h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The investigation on a series of A3B-type miktoarm star polymer assemblies by RAFT PISA has revealed the role of A3B architecture in delaying morphological transitions, and the formation of larger vesicles as well as other interesting morphologies.
Collapse
Affiliation(s)
- Xinru Zhang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zhiqiang Wei
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Kai Liu
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Li Wang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Wantai Yang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Engineering Research Center for the Syntheses and Applications of Waterborne Polymers, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, Beijing 100029, China
| |
Collapse
|
20
|
Bochkarev LN, Parshina YP, Gracheva YV, Kovylina TA, Lermontova SA, Klapshina LG, Konev AN, Lopatin MA, Lukina MM, Komarova AD, Shcheslavskiy VI, Shirmanova MV. Red Light-Emitting Water-Soluble Luminescent Iridium-Containing Polynorbornenes: Synthesis, Characterization and Oxygen Sensing Properties in Biological Tissues In Vivo. Molecules 2021; 26:6349. [PMID: 34770757 PMCID: PMC8587708 DOI: 10.3390/molecules26216349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022] Open
Abstract
New water-soluble polynorbornenes P1-P4 containing oligoether, amino acid groups and luminophoric complexes of iridium(III) were synthesized by ring-opening metathesis polymerization. The polymeric products in organic solvents and in water demonstrate intense photoluminescence in the red spectral region. The polymers P1 and P3 with 1-phenylisoquinoline cyclometalating ligands in iridium fragments reveal 4-6 fold higher emission quantum yields in solutions than those of P2 and P4 that contain iridium complexes with 1-(thien-2-yl)isoquinoline cyclometalating ligands. The emission parameters of P1-P4 in degassed solutions essentially differ from those in the aerated solutions showing oxygen-dependent quenching of phosphorescence. Biological testing of P1 and P3 demonstrates that the polymers do not penetrate into live cultured cancer cells and normal skin fibroblasts and do not possess cytotoxicity within the concentrations and time ranges reasonable for biological studies. In vivo, the polymers display longer phosphorescence lifetimes in mouse tumors than in muscle, as measured using phosphorescence lifetime imaging (PLIM), which correlates with tumor hypoxia. Therefore, preliminary evaluation of the synthesized polymers shows their suitability for noninvasive in vivo assessments of oxygen levels in biological tissues.
Collapse
Affiliation(s)
- Leonid N. Bochkarev
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Yulia P. Parshina
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Yana V. Gracheva
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Tatyana A. Kovylina
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Svetlana A. Lermontova
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Larisa G. Klapshina
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Aleksey N. Konev
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Mikhail A. Lopatin
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Maria M. Lukina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.M.L.); (A.D.K.); (V.I.S.); (M.V.S.)
| | - Anastasia D. Komarova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.M.L.); (A.D.K.); (V.I.S.); (M.V.S.)
| | - Vladislav I. Shcheslavskiy
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.M.L.); (A.D.K.); (V.I.S.); (M.V.S.)
- Becker&Hickl GmbH, Nunsdorfer Ring 7-9, 12277 Berlin, Germany
| | - Marina V. Shirmanova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.M.L.); (A.D.K.); (V.I.S.); (M.V.S.)
| |
Collapse
|
21
|
Repp L, Unterberger CJ, Ye Z, Feltenberger JB, Swanson SM, Marker PC, Kwon GS. Oligo(Lactic Acid) 8-Docetaxel Prodrug-Loaded PEG- b-PLA Micelles for Prostate Cancer. NANOMATERIALS 2021; 11:nano11102745. [PMID: 34685195 PMCID: PMC8540550 DOI: 10.3390/nano11102745] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 01/14/2023]
Abstract
Docetaxel (DTX) is among the most frequently prescribed chemotherapy drugs and has recently been shown to extend survival in advanced prostate cancer patients. However, the poor water solubility of DTX prevents full exploitation of this potent anticancer drug. The current marketed formulation, Taxotere®, contains a toxic co-solvent that induces adverse reactions following intravenous injection. Nano-sized polymeric micelles have been proposed to create safer, water-soluble carriers for DTX, but many have failed to reach the clinic due to poor carrier stability in vivo. In this study, we aimed to improve micelle stability by synthesizing an ester prodrug of DTX, oligo(lactic acid)8-docetaxel (o(LA)8-DTX), for augmented compatibility with the core of poly(ethylene glycol)-b-poly(lactic acid) (PEG-b-PLA) micelles. Due to the enhancement of drug-carrier compatibility, we were able to load 50% (w/w) prodrug within the micelle, solubilize 20 mg/mL o(LA)8-DTX (~12 mg/mL DTX-equivalent) in aqueous media, and delay payload release. While the micelle core prohibited premature degradation, o(LA)8-DTX was rapidly converted to parent drug DTX through intramolecular backbiting (t1/2 = 6.3 h) or esterase-mediated degradation (t1/2 = 2.5 h) following release. Most importantly, o(LA)8-DTX micelles proved to be as efficacious but less toxic than Taxotere® in a preclinical mouse model of prostate cancer.
Collapse
Affiliation(s)
- Lauren Repp
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; (L.R.); (C.J.U.); (S.M.S.); (P.C.M.)
| | - Christopher J. Unterberger
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; (L.R.); (C.J.U.); (S.M.S.); (P.C.M.)
| | - Zhengqing Ye
- Medicinal Chemistry Center, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; (Z.Y.); (J.B.F.)
| | - John B. Feltenberger
- Medicinal Chemistry Center, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; (Z.Y.); (J.B.F.)
| | - Steven M. Swanson
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; (L.R.); (C.J.U.); (S.M.S.); (P.C.M.)
| | - Paul C. Marker
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; (L.R.); (C.J.U.); (S.M.S.); (P.C.M.)
| | - Glen S. Kwon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; (L.R.); (C.J.U.); (S.M.S.); (P.C.M.)
- Correspondence:
| |
Collapse
|
22
|
Hu X, Jazani AM, Oh JK. Recent advances in development of imine-based acid-degradable polymeric nanoassemblies for intracellular drug delivery. POLYMER 2021. [DOI: 10.1016/j.polymer.2021.124024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
Pharmaceutical Formulations with P-Glycoprotein Inhibitory Effect as Promising Approaches for Enhancing Oral Drug Absorption and Bioavailability. Pharmaceutics 2021; 13:pharmaceutics13071103. [PMID: 34371794 PMCID: PMC8309061 DOI: 10.3390/pharmaceutics13071103] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 02/08/2023] Open
Abstract
P-glycoprotein (P-gp) is crucial in the active transport of various substrates with diverse structures out of cells, resulting in poor intestinal permeation and limited bioavailability following oral administration. P-gp inhibitors, including small molecule drugs, natural constituents, and pharmaceutically inert excipients, have been exploited to overcome P-gp efflux and enhance the oral absorption and bioavailability of many P-gp substrates. The co-administration of small molecule P-gp inhibitors with P-gp substrates can result in drug–drug interactions and increased side effects due to the pharmacological activity of these molecules. On the other hand, pharmaceutically inert excipients, including polymers, surfactants, and lipid-based excipients, are safe, pharmaceutically acceptable, and are not absorbed from the gut. Notably, they can be incorporated in pharmaceutical formulations to enhance drug solubility, absorption, and bioavailability due to the formulation itself and the P-gp inhibitory effects of the excipients. Different formulations with inherent P-gp inhibitory activity have been developed. These include micelles, emulsions, liposomes, solid lipid nanoparticles, polymeric nanoparticles, microspheres, dendrimers, and solid dispersions. They can bypass P-gp by different mechanisms related to their properties. In this review, we briefly introduce P-gp and P-gp inhibitors, and we extensively summarize the current development of oral drug delivery systems that can bypass and inhibit P-gp to improve the oral absorption and bioavailability of P-gp substrates. Since many drugs are limited by P-gp-mediated efflux, this review is helpful for designing suitable formulations of P-gp substrates to enhance their oral absorption and bioavailability.
Collapse
|
24
|
Biswas S. Polymeric micelles as drug-delivery systems in cancer: challenges and opportunities. Nanomedicine (Lond) 2021; 16:1541-1544. [PMID: 34169749 DOI: 10.2217/nnm-2021-0081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tweetable abstract Micelles are nanocarriers for hydrophobic chemotherapeutic drugs. This editorial discusses the current status of preclinical micellar research and sheds light on the possibility of their clinical translation.
Collapse
Affiliation(s)
- Swati Biswas
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad, Telangana, 500078, India
| |
Collapse
|
25
|
Braatz D, Dimde M, Ma G, Zhong Y, Tully M, Grötzinger C, Zhang Y, Mavroskoufis A, Schirner M, Zhong Z, Ballauff M, Haag R. Toolbox of Biodegradable Dendritic (Poly glycerol sulfate)-SS-poly(ester) Micelles for Cancer Treatment: Stability, Drug Release, and Tumor Targeting. Biomacromolecules 2021; 22:2625-2640. [PMID: 34076415 DOI: 10.1021/acs.biomac.1c00333] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In this paper, we present well-defined dPGS-SS-PCL/PLGA/PLA micellar systems demonstrating excellent capabilities as a drug delivery platform in light of high stability and precise in vitro and in vivo drug release combined with active targetability to tumors. These six amphiphilic block copolymers were each targeted in two different molecular weights (8 or 16 kDa) and characterized using 1H NMR, gel permeation chromatography (GPC), and elemental analysis. The block copolymer micelles showed monodispersed size distributions of 81-187 nm, strong negative charges between -52 and -41 mV, and low critical micelle concentrations (CMCs) of up to 1.13-3.58 mg/L (134-527 nM). The serum stability was determined as 94% after 24 h. The drug-loading efficiency for Sunitinib ranges from 38 to 83% (8-17 wt %). The release was selectively triggered by glutathione (GSH) and lipase, reaching 85% after 5 days, while only 20% leaching was observed under physiological conditions. Both the in vitro and in vivo studies showed sustained release of Sunitinib over 1 week. CCK-8 assays on HeLa lines demonstrated the high cell compatibility (1 mg/mL, 94% cell viability, 48 h) and the high cancer cell toxicity of Sunitinib-loaded micelles (IC50 2.5 μg/mL). By in vivo fluorescence imaging studies on HT-29 tumor-bearing mice, the targetability of dPGS7.8-SS-PCL7.8 enabled substantial accumulation in tumor tissue compared to nonsulfated dPG3.9-SS-PCL7.8. As a proof of concept, Sunitinib-loaded dPGS-SS-poly(ester) micelles improved the antitumor efficacy of the chemotherapeutic. A tenfold lower dosage of loaded Sunitinib led to an even higher tumor growth inhibition compared to the free drug, as demonstrated in a HeLa human cervical tumor-bearing mice model. No toxicity for the organism was observed, confirming the good biocompatibility of the system.
Collapse
Affiliation(s)
- Daniel Braatz
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Mathias Dimde
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Guoxin Ma
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Yinan Zhong
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Michael Tully
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Carsten Grötzinger
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, 13353 Berlin, Germany
| | - Yuanyuan Zhang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Alexandros Mavroskoufis
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Michael Schirner
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Matthias Ballauff
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| |
Collapse
|
26
|
Multifunctional polymeric micellar nanomedicine in the diagnosis and treatment of cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112186. [PMID: 34082985 DOI: 10.1016/j.msec.2021.112186] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
Polymeric micelles are a prevalent topic of research for the past decade, especially concerning their fitting ability to deliver drug and diagnostic agents. This delivery system offers outstanding advantages, such as biocompatibility, high loading efficiency, water-solubility, and good stability in biological fluids, to name a few. The multifunctional polymeric micellar architect offers the added capability to adapt its surface to meet the looked-for clinical needs. This review cross-talks the recent reports, proof-of-concept studies, patents, and clinical trials that utilize polymeric micellar family architectures concerning cancer targeted delivery of anticancer drugs, gene therapeutics, and diagnostic agents. The manuscript also expounds on the underlying opportunities, allied challenges, and ways to resolve their bench-to-bedside translation for allied clinical applications.
Collapse
|
27
|
A synthetically lethal nanomedicine delivering novel inhibitors of polynucleotide kinase 3'-phosphatase (PNKP) for targeted therapy of PTEN-deficient colorectal cancer. J Control Release 2021; 334:335-352. [PMID: 33933518 DOI: 10.1016/j.jconrel.2021.04.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 11/21/2022]
Abstract
Phosphatase and TENsin homolog deleted on chromosome 10 (PTEN) is a major tumor-suppressor protein that is lost in up to 75% of aggressive colorectal cancers (CRC). The co-depletion of PTEN and a DNA repair protein, polynucleotide kinase 3'-phosphatase (PNKP), has been shown to lead to synthetic lethality in several cancer types including CRC. This finding inspired the development of novel PNKP inhibitors as potential new drugs against PTEN-deficient CRC. Here, we report on the in vitro and in vivo evaluation of a nano-encapsulated potent, but poorly water-soluble lead PNKP inhibitor, A83B4C63, as a new targeted therapeutic for PTEN-deficient CRC. Our data confirmed the binding of A83B4C63, as free or nanoparticle (NP) formulation, to intracellular PNKP using the cellular thermal shift assay (CETSA), in vitro and in vivo. Dose escalating toxicity studies in healthy CD-1 mice, based on measurement of animal weight changes and biochemical blood analysis, revealed the safety of both free and nano-encapsulated A83B4C63, at assessed doses of ≤50 mg/kg. Nano-carriers of A83B4C63 effectively inhibited the growth of HCT116/PTEN-/- xenografts in NIH-III nude mice following intravenous (IV) administration, but not that of wild-type HCT116/PTEN+/+ xenografts. This was in contrast to IV administration of A83B4C63 solubilized with the aid of Cremophor EL: Ethanol (CE), which led to similar tumor growth to that of formulation excipients (NP or CE without drug) or 5% dextrose. This observation was attributed to the higher levels of A83B4C63 delivered to tumor tissue by its NP formulation. Our data provide evidence for the success of NPs of A83B4C63, as novel synthetically lethal nano-therapeutics in the treatment of PTEN-deficient CRC. This research also highlights the potential of successful application of nanomedicine in the drug development process.
Collapse
|
28
|
Gjuroski I, Furrer J, Vermathen M. Probing the Interactions of Porphyrins with Macromolecules Using NMR Spectroscopy Techniques. Molecules 2021; 26:1942. [PMID: 33808335 PMCID: PMC8037866 DOI: 10.3390/molecules26071942] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 12/11/2022] Open
Abstract
Porphyrinic compounds are widespread in nature and play key roles in biological processes such as oxygen transport in blood, enzymatic redox reactions or photosynthesis. In addition, both naturally derived as well as synthetic porphyrinic compounds are extensively explored for biomedical and technical applications such as photodynamic therapy (PDT) or photovoltaic systems, respectively. Their unique electronic structures and photophysical properties make this class of compounds so interesting for the multiple functions encountered. It is therefore not surprising that optical methods are typically the prevalent analytical tool applied in characterization and processes involving porphyrinic compounds. However, a wealth of complementary information can be obtained from NMR spectroscopic techniques. Based on the advantage of providing structural and dynamic information with atomic resolution simultaneously, NMR spectroscopy is a powerful method for studying molecular interactions between porphyrinic compounds and macromolecules. Such interactions are of special interest in medical applications of porphyrinic photosensitizers that are mostly combined with macromolecular carrier systems. The macromolecular surrounding typically stabilizes the encapsulated drug and may also modify its physical properties. Moreover, the interaction with macromolecular physiological components needs to be explored to understand and control mechanisms of action and therapeutic efficacy. This review focuses on such non-covalent interactions of porphyrinic drugs with synthetic polymers as well as with biomolecules such as phospholipids or proteins. A brief introduction into various NMR spectroscopic techniques is given including chemical shift perturbation methods, NOE enhancement spectroscopy, relaxation time measurements and diffusion-ordered spectroscopy. How these NMR tools are used to address porphyrin-macromolecule interactions with respect to their function in biomedical applications is the central point of the current review.
Collapse
Affiliation(s)
| | | | - Martina Vermathen
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland; (I.G.); (J.F.)
| |
Collapse
|
29
|
Ghezzi M, Pescina S, Padula C, Santi P, Del Favero E, Cantù L, Nicoli S. Polymeric micelles in drug delivery: An insight of the techniques for their characterization and assessment in biorelevant conditions. J Control Release 2021; 332:312-336. [PMID: 33652113 DOI: 10.1016/j.jconrel.2021.02.031] [Citation(s) in RCA: 493] [Impact Index Per Article: 123.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/16/2022]
Abstract
Polymeric micelles, i.e. aggregation colloids formed in solution by self-assembling of amphiphilic polymers, represent an innovative tool to overcome several issues related to drug administration, from the low water-solubility to the poor drug permeability across biological barriers. With respect to other nanocarriers, polymeric micelles generally display smaller size, easier preparation and sterilization processes, and good solubilization properties, unfortunately associated with a lower stability in biological fluids and a more complicated characterization. Particularly challenging is the study of their interaction with the biological environment, essential to predict the real in vivo behavior after administration. In this review, after a general presentation on micelles features and properties, different characterization techniques are discussed, from the ones used for the determination of micelles basic characteristics (critical micellar concentration, size, surface charge, morphology) to the more complex approaches used to figure out micelles kinetic stability, drug release and behavior in the presence of biological substrates (fluids, cells and tissues). The techniques presented (such as dynamic light scattering, AFM, cryo-TEM, X-ray scattering, FRET, symmetrical flow field-flow fractionation (AF4) and density ultracentrifugation), each one with their own advantages and limitations, can be combined to achieve a deeper comprehension of polymeric micelles in vivo behavior. The set-up and validation of adequate methods for micelles description represent the essential starting point for their development and clinical success.
Collapse
Affiliation(s)
- M Ghezzi
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - S Pescina
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - C Padula
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - P Santi
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - E Del Favero
- Department of Medical Biotechnologies and Translational Medicine, LITA, University of Milan, Segrate, Italy
| | - L Cantù
- Department of Medical Biotechnologies and Translational Medicine, LITA, University of Milan, Segrate, Italy
| | - S Nicoli
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy.
| |
Collapse
|
30
|
Cook A, Decuzzi P. Harnessing Endogenous Stimuli for Responsive Materials in Theranostics. ACS NANO 2021; 15:2068-2098. [PMID: 33555171 PMCID: PMC7905878 DOI: 10.1021/acsnano.0c09115] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/02/2021] [Indexed: 05/04/2023]
Abstract
Materials that respond to endogenous stimuli are being leveraged to enhance spatiotemporal control in a range of biomedical applications from drug delivery to diagnostic tools. The design of materials that undergo morphological or chemical changes in response to specific biological cues or pathologies will be an important area of research for improving efficacies of existing therapies and imaging agents, while also being promising for developing personalized theranostic systems. Internal stimuli-responsive systems can be engineered across length scales from nanometers to macroscopic and can respond to endogenous signals such as enzymes, pH, glucose, ATP, hypoxia, redox signals, and nucleic acids by incorporating synthetic bio-inspired moieties or natural building blocks. This Review will summarize response mechanisms and fabrication strategies used in internal stimuli-responsive materials with a focus on drug delivery and imaging for a broad range of pathologies, including cancer, diabetes, vascular disorders, inflammation, and microbial infections. We will also discuss observed challenges, future research directions, and clinical translation aspects of these responsive materials.
Collapse
Affiliation(s)
- Alexander
B. Cook
- Laboratory of Nanotechnology
for Precision Medicine, Istituto Italiano
di Tecnologia, Via Morego
30, 16163 Genova, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology
for Precision Medicine, Istituto Italiano
di Tecnologia, Via Morego
30, 16163 Genova, Italy
| |
Collapse
|
31
|
Bagheri M, Fens MH, Kleijn TG, Capomaccio RB, Mehn D, Krawczyk PM, Scutigliani EM, Gurinov A, Baldus M, van Kronenburg NCH, Kok RJ, Heger M, van Nostrum CF, Hennink WE. In Vitro and In Vivo Studies on HPMA-Based Polymeric Micelles Loaded with Curcumin. Mol Pharm 2021; 18:1247-1263. [PMID: 33464911 PMCID: PMC7927141 DOI: 10.1021/acs.molpharmaceut.0c01114] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Curcumin-loaded polymeric micelles composed of poly(ethylene glycol)-b-poly(N-2-benzoyloxypropyl methacrylamide) (mPEG-b-p(HPMA-Bz)) were prepared to solubilize and improve the pharmacokinetics of curcumin. Curcumin-loaded micelles were prepared by a nanoprecipitation method using mPEG5kDa-b-p(HPMA-Bz) copolymers with varying molecular weight of the hydrophobic block (5.2, 10.0, and 17.1 kDa). At equal curcumin loading, micelles composed of mPEG5kDa-b-p(HPMA-Bz)17.1kDa showed better curcumin retention in both phosphate-buffered saline (PBS) and plasma at 37 °C than micelles based on block copolymers with smaller hydrophobic blocks. No change in micelle size was observed during 24 h incubation in plasma using asymmetrical flow field-flow fractionation (AF4), attesting to particle stability. However, 22-49% of the curcumin loading was released from the micelles during 24 h from formulations with the highest to the lowest molecular weight p(HPMA-Bz), respectively, in plasma. AF4 analysis further showed that the released curcumin was subsequently solubilized by albumin. In vitro analyses revealed that the curcumin-loaded mPEG5kDa-b-p(HPMA-Bz)17.1kDa micelles were internalized by different types of cancer cells, resulting in curcumin-induced cell death. Intravenously administered curcumin-loaded, Cy7-labeled mPEG5kDa-b-p(HPMA-Bz)17.1kDa micelles in mice at 50 mg curcumin/kg showed a long circulation half-life for the micelles (t1/2 = 42 h), in line with the AF4 results. In contrast, the circulation time of curcumin was considerably shorter than that of the micelles (t1/2α = 0.11, t1/2β = 2.5 h) but ∼5 times longer than has been reported for free curcumin (t1/2α = 0.02 h). The faster clearance of curcumin in vivo compared to in vitro studies can be attributed to the interaction of curcumin with blood cells. Despite the excellent solubilizing effect of these micelles, no cytostatic effect was achieved in neuroblastoma-bearing mice, possibly because of the low sensitivity of the Neuro2A cells to curcumin.
Collapse
Affiliation(s)
- Mahsa Bagheri
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Marcel H Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Tony G Kleijn
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands.,Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing 314001, P. R. China
| | - Robin B Capomaccio
- European Commission, Joint Research Centre (JRC), 21027 Ispra, VA, Italy
| | - Dora Mehn
- European Commission, Joint Research Centre (JRC), 21027 Ispra, VA, Italy
| | - Przemek M Krawczyk
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Enzo M Scutigliani
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Andrei Gurinov
- NMR Spectroscopy Group, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Marc Baldus
- NMR Spectroscopy Group, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Nicky C H van Kronenburg
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Robbert J Kok
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Michal Heger
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands.,Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing 314001, P. R. China
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
32
|
Enzymatic synthesis of PEGylated lactide-diester-diol copolyesters for highly efficient targeted anticancer drug delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 115:111125. [DOI: 10.1016/j.msec.2020.111125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/19/2020] [Accepted: 05/25/2020] [Indexed: 01/04/2023]
|
33
|
Hwang D, Ramsey JD, Kabanov AV. Polymeric micelles for the delivery of poorly soluble drugs: From nanoformulation to clinical approval. Adv Drug Deliv Rev 2020; 156:80-118. [PMID: 32980449 DOI: 10.1016/j.addr.2020.09.009] [Citation(s) in RCA: 329] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 01/04/2023]
Abstract
Over the last three decades, polymeric micelles have emerged as a highly promising drug delivery platform for therapeutic compounds. Particularly, poorly soluble small molecules with high potency and significant toxicity were encapsulated in polymeric micelles. Polymeric micelles have shown improved pharmacokinetic profiles in preclinical animal models and enhanced efficacy with a superior safety profile for therapeutic drugs. Several polymeric micelle formulations have reached the clinical stage and are either in clinical trials or are approved for human use. This furthers interest in this field and underscores the need for additional learning of how to best design and apply these micellar carriers to improve the clinical outcomes of many drugs. In this review, we provide detailed information on polymeric micelles for the solubilization of poorly soluble small molecules in topics such as the design of block copolymers, experimental and theoretical analysis of drug encapsulation in polymeric micelles, pharmacokinetics of drugs in polymeric micelles, regulatory approval pathways of nanomedicines, and current outcomes from micelle formulations in clinical trials. We aim to describe the latest information on advanced analytical approaches for elucidating molecular interactions within the core of polymeric micelles for effective solubilization as well as for analyzing nanomedicine's pharmacokinetic profiles. Taking into account the considerations described within, academic and industrial researchers can continue to elucidate novel interactions in polymeric micelles and capitalize on their potential as drug delivery vehicles to help improve therapeutic outcomes in systemic delivery.
Collapse
Affiliation(s)
- Duhyeong Hwang
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Jacob D Ramsey
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M. V. Lomonosov Moscow State University, Moscow 119992, Russia.
| |
Collapse
|
34
|
Zhang L, Zhang Y, Zhao G, Yang H, Wang X, Yu R, Liu H, Li S. Preparation of Poly(MTZ) n -(DMAEMA) m Micelles and Study on Their Antibacterial Property. ACS OMEGA 2020; 5:23053-23061. [PMID: 32954155 PMCID: PMC7495777 DOI: 10.1021/acsomega.0c02774] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/14/2020] [Indexed: 06/11/2023]
Abstract
Bacterial infections are the most common type of clinical infection. The abuse of clinical antibiotics has led to the frequent appearance of drug-resistant strains and even some super bacteria. In this study, we synthesized Poly(MTZ) n -(DMAEMA) m polymer micelles with cations on the surface. The synthesis of this novel polymer comes in two steps. First, Poly(MTZ) n was synthesized with metronidazole (MTZ) referred as the macromolecular chain transfer agent and v-501 as the initiator for initiating the polymerization of 4-cyanopentanoic acid dithiobenzoate. Then, novel polymer micelles were synthesized with Poly(MTZ) n referred as the macromolecular chain transfer agent and v-501 as the initiator for initiating the polymerization of the monomer 2-(dimethylamino) ethyl methacrylate, which could adsorb to the negatively charged bacterial surface via electrostatic interaction and enhance bactericidal activity. Scanning electron microscopy showed that the micelles could be accurately targeted to the surface of bacteria, and the zone of inhibition assay confirmed that the micelles could enhance the sensitivity of bacteria to drugs. Hence, Poly(MTZ) n -(DMAEMA) m polymer micelles will have potential use for the clinical treatment of anaerobic infections in the future.
Collapse
Affiliation(s)
- Long Zhang
- Insititute of Nervous
System Diseases, Xuzhou Medical University, Xuzhou 221002, P. R. China
| | - Yongkang Zhang
- Insititute of Nervous
System Diseases, Xuzhou Medical University, Xuzhou 221002, P. R. China
| | - Gang Zhao
- Insititute of Nervous
System Diseases, Xuzhou Medical University, Xuzhou 221002, P. R. China
| | - Han Yang
- Insititute of Nervous
System Diseases, Xuzhou Medical University, Xuzhou 221002, P. R. China
| | - Xiaoqian Wang
- Insititute of Nervous
System Diseases, Xuzhou Medical University, Xuzhou 221002, P. R. China
| | - Rutong Yu
- Insititute of Nervous
System Diseases, Xuzhou Medical University, Xuzhou 221002, P. R. China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, P. R. China
| | - Hongmei Liu
- Insititute of Nervous
System Diseases, Xuzhou Medical University, Xuzhou 221002, P. R. China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, P. R. China
| | - Shanshan Li
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Jiangsu Medical
Engineering Research Center of Gene Detection, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| |
Collapse
|
35
|
Wang Y, van Steenbergen MJ, Beztsinna N, Shi Y, Lammers T, van Nostrum CF, Hennink WE. Biotin-decorated all-HPMA polymeric micelles for paclitaxel delivery. J Control Release 2020; 328:970-984. [PMID: 32926885 DOI: 10.1016/j.jconrel.2020.09.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/21/2020] [Accepted: 09/07/2020] [Indexed: 12/28/2022]
Abstract
To avoid poly(ethylene glycol)-related issues of nanomedicines such as accelerated blood clearance, fully N-2-hydroxypropyl methacrylamide (HPMAm)-based polymeric micelles decorated with biotin for drug delivery were designed. To this end, a biotin-functionalized chain transfer agent (CTA), 4-cyano-4-[(dodecylsulfanylthiocarbonyl)-sulfanyl]pentanoic acid (biotin-CDTPA), was synthesized for reversible addition-fragmentation chain-transfer (RAFT) polymerization. Amphiphilic poly(N-2-hydroxypropyl methacrylamide)-block-poly(N-2-benzoyloxypropyl methacrylamide) (p(HPMAm)-b-p(HPMAm-Bz)) with molecular weights ranging from 8 to 24 kDa were synthesized using CDTPA or biotin-CDTPA as CTA and 2,2'-azobis(2-methylpropionitrile) as initiator. The copolymers self-assembled in aqueous media into micelles with sizes of 40-90 nm which positively correlated to the chain length of the hydrophobic block in the polymers, whereas the critical micelle concentrations decreased with increasing hydrophobic block length. The polymer with a molecular weight of 22.1 kDa was used to prepare paclitaxel-loaded micelles which had sizes between 61 and 70 nm, and a maximum loading capacity of around 10 wt%. A549 lung cancer cells overexpressing the biotin receptor, internalized the biotin-decorated micelles more efficiently than non-targeted micelles, while very low internalization of both types of micelles by HEK293 human embryonic kidney cells lacking the biotin receptor was observed. As a consequence, the paclitaxel-loaded micelles with biotin decoration exhibited stronger cytotoxicity in A549 cells than non-targeted micelles. Overall, a synthetic pathway to obtain actively targeted poly(ethylene glycol)-free micelles fully based on a poly(HPMAm) backbone was established. These polymeric micelles are promising systems for the delivery of hydrophobic anticancer drugs.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| | - Mies J van Steenbergen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| | - Nataliia Beztsinna
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| | - Yang Shi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbecktrasse 55, 52074 Aachen, Germany.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbecktrasse 55, 52074 Aachen, Germany.
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| |
Collapse
|
36
|
Docetaxel and its nanoformulations: how delivery strategies could impact the therapeutic outcome? Ther Deliv 2020; 11:755-759. [PMID: 32873188 DOI: 10.4155/tde-2020-0088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
37
|
Li S, Shan X, Wang Y, Chen Q, Sun J, He Z, Sun B, Luo C. Dimeric prodrug-based nanomedicines for cancer therapy. J Control Release 2020; 326:510-522. [PMID: 32721523 DOI: 10.1016/j.jconrel.2020.07.036] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 01/10/2023]
Abstract
With the rapid development of conjugation chemistry and biomedical nanotechnology, prodrug-based nanosystems (PNS) have emerged as promising drug delivery nanoplatforms. Dimeric prodrug, as an emerging branch of prodrug, has been widely investigated by covalently conjugating two same or different drug molecules. In recent years, great progress has been made in dimeric prodrug-based nanosystems (DPNS) for cancer therapy. Many advantages offered by DPNS have significantly facilitated the delivery efficiency of anticancer drugs, such as high drug loading capacity, favorable pharmacokinetics, tumor stimuli-sensitive drug release and facile combination theranostics. Given the rapid developments in this field, we here outline the latest updates of DPNS in cancer treatment, focusing on dimeric prodrug-encapsulated nanosystems, dimeric prodrug-nanoassemblies and tumor stimuli-responsive DPNS. Moreover, the design principle, advantages and challenges of DPNS for clinical cancer therapy are also highlighted.
Collapse
Affiliation(s)
- Shumeng Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xinzhu Shan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yuequan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Qin Chen
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, PR China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
38
|
Jeevan BGC, Szlenk CT, Gao J, Dong X, Wang Z, Natesan S. Molecular Dynamics Simulations Provide Insight into the Loading Efficiency of Proresolving Lipid Mediators Resolvin D1 and D2 in Cell Membrane-Derived Nanovesicles. Mol Pharm 2020; 17:2155-2164. [PMID: 32374613 PMCID: PMC7313724 DOI: 10.1021/acs.molpharmaceut.0c00299] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Resolvins D1 and D2 (RvDs) are structural isomers and metabolites of docosahexaenoic acid, an omega-3 fatty acid, enzymatically produced in our body in response to acute inflammation or microbial invasion. Resolvins have been shown to play an essential role in the resolution of inflammation, tissue repair, and return to homeostasis and thus are actively pursued as potential therapeutics in treating inflammatory disorders and infectious diseases. However, effective in vivo delivery of RvDs continues to be a challenging task. Recent studies demonstrated that RvD1 or RvD2 loaded in cell membrane-derived nanovesicles significantly increased therapeutic efficacy in treating murine peritonitis and ischemic stroke, respectively. The mechanistic details of how the subtle structural difference between RvD1 and RvD2 alters their molecular interactions with the membrane lipids of the nanovesicles and thus affects the loading efficiency remain unknown. Here, we report the encapsulation profiles of the neutral and ionized species of both RvD1 and RvD2 determined with the cell membrane-derived nanovesicles at pH values 5.4 and 7.4, respectively. Also, we performed microsecond time-scale all-atom molecular dynamics (MD) simulations in explicit water to elucidate the molecular interactions of both neutral and ionized species of RvD1 and RvD2 with the lipid bilayer using a model membrane system, containing 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) and cholesterol. We found that the differences in the position and chirality of hydroxyl groups in RvD1 and RvD2 affected their location, orientation, and conformations within the bilayer. Surprisingly, the deprotonation of their carboxyl group caused their orientation and conformation to change from a fully extended one that is oriented in parallel to the membrane plane to a J-shaped bent conformation that is oriented perpendicular to the bilayer plane. Our studies offer valuable insight into the molecular interactions of RvD1/D2 with the lipid bilayer in atomistic details and provide a mechanistic explanation for the observed differences in the encapsulation profiles of RvD1 and RvD2, which may facilitate the rational design of nanovesicle-based therapeutics for treating inflammatory diseases.
Collapse
Affiliation(s)
- B. GC Jeevan
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Christopher T. Szlenk
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Jin Gao
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Xinyue Dong
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Zhenjia Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Senthil Natesan
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
39
|
Liu Y, Fens MH, Lou B, van Kronenburg NC, Maas-Bakker RF, Kok RJ, Oliveira S, Hennink WE, van Nostrum CF. π-π-Stacked Poly(ε-caprolactone)- b-poly(ethylene glycol) Micelles Loaded with a Photosensitizer for Photodynamic Therapy. Pharmaceutics 2020; 12:pharmaceutics12040338. [PMID: 32283871 PMCID: PMC7238042 DOI: 10.3390/pharmaceutics12040338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
To improve the in vivo stability of poly(ε-caprolactone)-b-poly(ethylene glycol) (PCL-PEG)-based micelles and cargo retention by π-π stacking interactions, pendant aromatic rings were introduced by copolymerization of ε-caprolactone with benzyl 5-methyl-2-oxo-1,3-dioxane-5-carboxylate (TMC-Bz). It was shown that the incorporation of aromatic rings yielded smaller micelles (18–30 nm) with better colloidal stability in PBS than micelles without aromatic groups. The circulation time of i.v. injected micelles containing multiple pendant aromatic groups was longer (t½-α: ~0.7 h; t½-β: 2.9 h) than that of micelles with a single terminal aromatic group (t½ < 0.3 h). In addition, the in vitro partitioning of the encapsulated photosensitizer (meta-tetra(hydroxyphenyl)chlorin, mTHPC) between micelles and human plasma was favored towards micelles for those that contained the pendant aromatic groups. However, this was not sufficient to fully retain mTHPC in the micelles in vivo, as indicated by similar biodistribution patterns of micellar mTHPC compared to free mTHPC, and unequal biodistribution patterns of mTHPC and the host micelles. Our study points out that more detailed in vitro methods are necessary to more reliably predict in vivo outcomes. Furthermore, additional measures beyond π-π stacking are needed to stably incorporate mTHPC in micelles in order to benefit from the use of micelles as targeted delivery systems.
Collapse
Affiliation(s)
- Yanna Liu
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Marcel H.A.M. Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Bo Lou
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Nicky C.H. van Kronenburg
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Roel F.M. Maas-Bakker
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Robbert J. Kok
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Sabrina Oliveira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Wim E. Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
| | - Cornelus F. van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands; (Y.L.); (B.L.); (N.C.H.v.K.); (R.J.K.); (S.O.); (W.E.H.)
- Correspondence: ; Tel.: +31-620274607
| |
Collapse
|
40
|
Shi H, van Steenbergen MJ, Lou B, Liu Y, Hennink WE, Kok RJ. Folate decorated polymeric micelles for targeted delivery of the kinase inhibitor dactolisib to cancer cells. Int J Pharm 2020; 582:119305. [PMID: 32278056 DOI: 10.1016/j.ijpharm.2020.119305] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/11/2022]
Abstract
One of the main challenges in clinical translation of polymeric micelles is retention of the drug in the nanocarrier system upon its systemic administration. Core crosslinking and coupling of the drug to the micellar backbone are common strategies to overcome these issues. In the present study, polymeric micelles were prepared for tumor cell targeting of the kinase inhibitor dactolisib which inhibits both the mammalian Target of Rapamycin (mTOR) kinase and phosphatidylinositol-3-kinase (PI3K). We employed platinum(II)-based linker chemistry to couple dactolisib to the core of poly(ethylene glycol)-b-poly(acrylic acid) (PEG-b-PAA) polymeric micelles. The formed dactolisib-PEG-PAA unimers are amphiphilic and self-assemble in an aqueous milieu into core-shell polymeric micelles. Folate was conjugated onto the surface of the micelles to yield folate-decorated polymeric micelles which can target folate receptor over-expressing tumor cells. Fluorescently labeled polymeric micelles were prepared using a lissamine-platinum complex linked in a similar manner as dactolisib. Dactolisib polymeric micelles showed good colloidal stability in water and released the coupled drug in buffers containing chloride or glutathione. Folate decorated micelles were avidly internalized by folate-receptor-positive KB cells and displayed targeted cellular cytotoxicity at 50-75 nM IC50. In conclusion, we have prepared a novel type of folate-receptor targeted polymeric micelles in which platinum(II) linker chemistry modulates drug retention and sustained release of the coupled inhibitor dactolisib.
Collapse
Affiliation(s)
- Haili Shi
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands
| | - Mies J van Steenbergen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands
| | - Bo Lou
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands
| | - Yanna Liu
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands
| | - Robbert J Kok
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands.
| |
Collapse
|
41
|
Vinayak M, Maurya AK. Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development. Anticancer Agents Med Chem 2020; 19:1560-1576. [PMID: 31284873 DOI: 10.2174/1871520619666190705150214] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 12/27/2022]
Abstract
The spread of metastatic cancer cell is the main cause of death worldwide. Cellular and molecular basis of the action of phytochemicals in the modulation of metastatic cancer highlights the importance of fruits and vegetables. Quercetin is a natural bioflavonoid present in fruits, vegetables, seeds, berries, and tea. The cancer-preventive activity of quercetin is well documented due to its anti-inflammatory, anti-proliferative and anti-angiogenic activities. However, poor water solubility and delivery, chemical instability, short half-life, and low-bioavailability of quercetin limit its clinical application in cancer chemoprevention. A better understanding of the molecular mechanism of controlled and regulated drug delivery is essential for the development of novel and effective therapies. To overcome the limitations of accessibility by quercetin, it can be delivered as nanoconjugated quercetin. Nanoconjugated quercetin has attracted much attention due to its controlled drug release, long retention in tumor, enhanced anticancer potential, and promising clinical application. The pharmacological effect of quercetin conjugated nanoparticles typically depends on drug carriers used such as liposomes, silver nanoparticles, silica nanoparticles, PLGA (Poly lactic-co-glycolic acid), PLA (poly(D,L-lactic acid)) nanoparticles, polymeric micelles, chitosan nanoparticles, etc. In this review, we described various delivery systems of nanoconjugated quercetin like liposomes, silver nanoparticles, PLGA (Poly lactic-co-glycolic acid), and polymeric micelles including DOX conjugated micelles, metal conjugated micelles, nucleic acid conjugated micelles, and antibody-conjugated micelles on in vitro and in vivo tumor models; as well as validated their potential as promising onco-therapeutic agents in light of recent updates.
Collapse
Affiliation(s)
- Manjula Vinayak
- Biochemistry & Molecular Biology Laboratory, Centre for Advanced Study in Zoology, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Akhilendra K Maurya
- Biochemistry & Molecular Biology Laboratory, Centre for Advanced Study in Zoology, Institute of Science, Banaras Hindu University, Varanasi-221005, India.,Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| |
Collapse
|
42
|
Multiple analyte profiling (MAP) index as a powerful diagnostic and therapeutic monitoring tool. Methods 2020; 190:26-32. [PMID: 32243921 DOI: 10.1016/j.ymeth.2020.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/09/2020] [Accepted: 03/29/2020] [Indexed: 11/20/2022] Open
Abstract
A robust data mining algorithm is presented as a critical solution to the challenge of managing intensive data generated from the recently developed multiplexing techniques, which allow simultaneous detection of up to 500 biomarkers in a few microliters of a single sample. Furthermore, detailed methodology is provided for exploiting the new algorithm along with examples for description of the first application as a powerful diagnostic and therapeutic monitoring tool in the management of breast cancer, as a disease model.
Collapse
|
43
|
Liu Y, Scrivano L, Peterson JD, Fens MHAM, Hernández IB, Mesquita B, Toraño JS, Hennink WE, van Nostrum CF, Oliveira S. EGFR-Targeted Nanobody Functionalized Polymeric Micelles Loaded with mTHPC for Selective Photodynamic Therapy. Mol Pharm 2020; 17:1276-1292. [PMID: 32142290 PMCID: PMC7140040 DOI: 10.1021/acs.molpharmaceut.9b01280] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
meta-Tetra(hydroxyphenyl)chlorin
(mTHPC) is one
of the most potent second-generation photosensitizers, clinically
used for photodynamic therapy (PDT) of head and neck squamous cell
carcinomas. However, improvements are still required concerning its
present formulation (i.e., Foscan, a solution of mTHPC in ethanol/propylene
glycol (40:60 w/w)), as mTHPC has the tendency to aggregate in aqueous
media, e.g., biological fluids, and it has limited tumor specificity.
In the present study, polymeric micelles with three different diameters
(17, 24, and 45 nm) based on benzyl-poly(ε-caprolactone)-b-poly(ethylene glycol) (PCLn-PEG; n = 9, 15, or 23) were prepared with mTHPC
loadings ranging from 0.5 to 10 wt % using a film-hydration method
as advanced nanoformulations for this photosensitizer. To favor the
uptake of the micelles by cancer cells that overexpress the epidermal
growth factor receptor (EGFR), the micelles were decorated with an
EGFR-targeted nanobody (named EGa1) through maleimide-thiol chemistry.
The enhanced binding of the EGFR-targeted micelles at 4 °C to
EGFR-overexpressing A431 cells, compared to low-EGFR-expressing HeLa
cells, confirmed the specificity of the micelles. In addition, an
enhanced uptake of mTHPC-loaded micelles by A431 cells was observed
when these were decorated with the EGa1 nanobody, compared to nontargeted
micelles. Both binding and uptake of targeted micelles were blocked
by an excess of free EGa1 nanobody, demonstrating that these processes
occur through EGFR. In line with this, mTHPC loaded in EGa1-conjugated
PCL23-PEG (EGa1-P23) micelles demonstrated 4
times higher photocytotoxicity on A431 cells, compared to micelles
lacking the nanobody. Importantly, EGa1-P23 micelles also
showed selective PDT against A431 cells compared to the low-EGFR-expressing
HeLa cells. Finally, an in vivo pharmacokinetic study
shows that after intravenous injection, mTHPC incorporated in the
P23 micelles displayed prolonged blood circulation kinetics,
compared to free mTHPC, independently of the presence of EGa1. Thus,
these results make these micelles a promising nanomedicine formulation
for selective therapy.
Collapse
Affiliation(s)
- Yanna Liu
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Luca Scrivano
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Julia Denise Peterson
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Marcel H A M Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Irati Beltrán Hernández
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands.,Division of Cell Biology, Department of Biology, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Bárbara Mesquita
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Javier Sastre Toraño
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Sabrina Oliveira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands.,Division of Cell Biology, Department of Biology, Utrecht University, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
44
|
Yu C, Tan X, Xu Z, Zhu G, Teng W, Zhao Q, Liang Z, Wu Z, Xiong D. Smart drug carrier based on polyurethane material for enhanced and controlled DOX release triggered by redox stimulus. REACT FUNCT POLYM 2020. [DOI: 10.1016/j.reactfunctpolym.2020.104507] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Bisso S, Leroux JC. Nanopharmaceuticals: A focus on their clinical translatability. Int J Pharm 2020; 578:119098. [DOI: 10.1016/j.ijpharm.2020.119098] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 12/19/2022]
|
46
|
Yang X, Lian K, Tan Y, Zhu Y, Liu X, Zeng Y, Yu T, Meng T, Yuan H, Hu F. Selective uptake of chitosan polymeric micelles by circulating monocytes for enhanced tumor targeting. Carbohydr Polym 2020; 229:115435. [DOI: 10.1016/j.carbpol.2019.115435] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/26/2019] [Accepted: 10/03/2019] [Indexed: 01/08/2023]
|
47
|
Ghilu S, Li Q, Fontaine SD, Santi DV, Kurmasheva RT, Zheng S, Houghton PJ. Prospective use of the single-mouse experimental design for the evaluation of PLX038A. Cancer Chemother Pharmacol 2020; 85:251-263. [PMID: 31927611 PMCID: PMC7039322 DOI: 10.1007/s00280-019-04017-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Defining robust criteria for drug activity in preclinical studies allows for fewer animals per treatment group, and potentially allows for inclusion of additional cancer models that more accurately represent genetic diversity and, potentially, allows for tumor sensitivity biomarker identification. METHODS Using a single-mouse design, 32 pediatric xenograft tumor models representing diverse pediatric cancer types [Ewing sarcoma (9), brain (4), rhabdomyosarcoma (10), Wilms tumor (4), and non-CNS rhabdoid tumors (5)] were evaluated for response to a single administration of pegylated-SN38 (PLX038A), a controlled-release PEGylated formulation of SN-38. Endpoints measured were percent tumor regression, and event-free survival (EFS). The correlation between response to PLX038A was compared to that for ten models treated with irinotecan (2.5 mg/kg × 5 days × 2 cycles), using a traditional design (10 mice/group). Correlations between tumor sensitivity, genetic mutations and gene expression were sought. Models showing no disease at week 20 were categorized as 'extreme responders' to PLX038A, whereas those with EFS less than 5 weeks were categorized as 'resistant'. RESULTS The activity of PLX038A was evaluable in 31/32 models. PLX038A induced > 50% volume regressions in 25 models (78%). Initial tumor volume regression correlated only modestly with EFS (r2 = 0.238), but sensitivity to PLX038A was better correlated with response to irinotecan when one tumor hypersensitive to PLX038A was omitted (r2 = 0.6844). Mutations in 53BP1 were observed in three of six sensitive tumor models compared to none in resistant models (n = 6). CONCLUSIONS This study demonstrates the feasibility of using a single-mouse design for assessing the antitumor activity of an agent, while encompassing greater genetic diversity representative of childhood cancers. PLX038A was highly active in most xenograft models, and tumor sensitivity to PLX038A was correlated with sensitivity to irinotecan, validating the single-mouse design in identifying agents with the same mechanism of action. Biomarkers that correlated with model sensitivity included wild-type TP53, or mutant TP53 but with a mutation in 53BP1, thus a defect in DNA damage response. These results support the value of the single-mouse experimental design.
Collapse
Affiliation(s)
- Samson Ghilu
- Greehey Children's Cancer Research Institute, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Qilin Li
- Greehey Children's Cancer Research Institute, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Shaun D Fontaine
- ProLynx LLC, 455 Mission Bay Blvd, South San Francisco, CA, 94158, USA
| | - Daniel V Santi
- ProLynx LLC, 455 Mission Bay Blvd, South San Francisco, CA, 94158, USA
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Siyuan Zheng
- Greehey Children's Cancer Research Institute, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Peter J Houghton
- Greehey Children's Cancer Research Institute, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| |
Collapse
|
48
|
Malviya N, Sonkar C, Ganguly R, Bhattacherjee D, Bhabak KP, Mukhopadhyay S. Novel Approach to Generate a Self-Deliverable Ru(II)-Based Anticancer Agent in the Self-Reacting Confined Gel Space. ACS APPLIED MATERIALS & INTERFACES 2019; 11:47606-47618. [PMID: 31755256 DOI: 10.1021/acsami.9b17075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Finding the most effective method for cancer treatment is one of the thought-provoking tasks. Drug delivery by collapsing of metallogel to the cancer cell is an appealing way out. Cancer cells have an acidic environment due to excessive accumulation of lactic acid. In this work, the novel G5 gelator with a strategically free carboxylic acid arm has been designed and fabricated and characterized by several spectroscopic and microscopic techniques. These experiments suggest the formation of an ordered supramolecular gel with clover-leaf-like morphology. Mechanical properties from rheological measurements suggest the viscoelastic nature of the gel. Furthermore, we have obtained crystals of G5 from the pure dimethyl sulfoxide solution, whereas gelation gets induced by addition of water. This G5 gelator loses its gelation capability once the carboxylate is esterified by layering with methanol, which furnished the crystals of Me-G5' (G5' = G5-H). Further, the G5 gelator is used for the formation of ruthenium metallogel. Interestingly, we obtained the monomeric species [Ru(G5')(η6-p-cymene)Cl] [Ru(II)G5] only in confined gel space upon addition of a [Ru2(η6-p-cymene)2Cl4] dimer to G5. The Ru(II)G5 metallogel has an inherent anticancer property with an IC50 value of 10.53 μM for the A549 cancer cell line. Treatment of the Ru(II)G5 metallogel by lactic acid for mimicking the acidic environment of the malignant cell results in collapsing of the gel by releasing the ruthenium metal ion. This released ruthenium ion binds with the lactic acid derivative making the gelator G5 free and producing a new compound Ru(II)L, which has also shown the anticancer property. The molecular docking study revealed that the released G5 could interact with a monocarboxylate transporter to disrupt the lactate transport chain, which might induce apoptosis.
Collapse
Affiliation(s)
| | | | - Rakesh Ganguly
- Division of Chemistry and Biological Chemistry , Nanyang Technological University , 639798 Singapore
| | - Debojit Bhattacherjee
- Department of Chemistry , Indian Institute of Technology Guwahati , Guwahati 781039 , India
| | - Krishna Pada Bhabak
- Department of Chemistry , Indian Institute of Technology Guwahati , Guwahati 781039 , India
| | | |
Collapse
|
49
|
Gjuroski I, Girousi E, Meyer C, Hertig D, Stojkov D, Fux M, Schnidrig N, Bucher J, Pfister S, Sauser L, Simon HU, Vermathen P, Furrer J, Vermathen M. Evaluation of polyvinylpyrrolidone and block copolymer micelle encapsulation of serine chlorin e6 and chlorin e4 on their reactivity towards albumin and transferrin and their cell uptake. J Control Release 2019; 316:150-167. [PMID: 31689463 DOI: 10.1016/j.jconrel.2019.10.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/02/2019] [Accepted: 10/02/2019] [Indexed: 01/10/2023]
Abstract
Encapsulation of porphyrinic photosensitizers (PSs) into polymeric carriers plays an important role in enhancing their efficiency as drugs in photodynamic therapy (PDT). Porphyrin aggregation and low solubility as well as the preservation of the advantageous photophysical properties pose a challenge on the design of efficient PS-carrier systems. Block copolymer micelles (BCMs) and polyvinylpyrrolidone (PVP) are promising drug delivery vehicles for physical entrapment of PSs. BCMs exhibit enhanced dynamics as compared to the less flexible PVP network. In the current work the question is addressed how these different dynamics affect PS encapsulation, release from the carrier, reaction with serum proteins, and cellular uptake. The porphyrinic compounds serine-amide of chlorin e6 (SerCE) and chlorin e4 (CE4) were used as model PSs with different lipophilicity and aggregation properties. 1H NMR and fluorescence spectroscopy were applied to study their interactions with PVP and BCMs consisting of Kolliphor P188 (KP). Both chlorins were well encapsulated by the carriers and had improved photophysical properties. Compared to SerCE, the more lipophilic CE4 exhibited stronger hydrophobic interactions with the BCM core, stabilizing the system and preventing exchange with the surrounding medium as was shown by NMR NOESY and DOSY experiments. PVP and BCMs protected the encapsulated chlorins against interaction with human transferrin (Tf). However, SerCE and CE4 were released from BCMs in favor of binding to human serum albumin (HSA) while PVP prevented interaction with HSA. Fluorescence spectroscopic studies revealed that HSA binds to the surface of PVP forming a protein corona. PVP and BCMs reduced cellular uptake of the chlorins. However, encapsulation into BCMs resulted in more efficient cell internalization for CE4 than for SerCE. HSA significantly lowered both, free and carrier-mediated cell uptake for CE4 and SerCE. In conclusion, PVP appears as the more universal delivery system covering a broad range of host molecules with respect to polarity, whereas BCMs require a higher drug-carrier compatibility. Poorly soluble hydrophobic PSs benefit stronger from BCM-type carriers due to enhanced bioavailability through disaggregation and solubilization allowing for more efficient cell uptake. In addition, increased PS-carrier hydrophobic interactions have a stabilizing effect. For more hydrophilic PSs, the main advantage of polymeric carriers like PVP or poloxamer micelles lies in their protection during the transport through the bloodstream. HSA binding plays an important role for drug release and cell uptake in carrier-mediated delivery to the target tissue.
Collapse
Affiliation(s)
- Ilche Gjuroski
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland.
| | - Eleftheria Girousi
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Christoph Meyer
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Damian Hertig
- Department of BioMedical Research and Radiology, University of Bern and Inselspital, sitem-insel AG, Freiburgstrasse 3, CH-3010, Bern, Switzerland; Institute of Clinical Chemistry, University of Bern and Inselspital, CH-3010, Bern, Switzerland
| | - Darko Stojkov
- Institute of Pharmacology, University of Bern and Inselspital, CH-3010, Bern, Switzerland
| | - Michaela Fux
- Institute of Clinical Chemistry, University of Bern and Inselspital, CH-3010, Bern, Switzerland
| | - Nicolas Schnidrig
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Jan Bucher
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Sara Pfister
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Luca Sauser
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern and Inselspital, CH-3010, Bern, Switzerland
| | - Peter Vermathen
- Department of BioMedical Research and Radiology, University of Bern and Inselspital, sitem-insel AG, Freiburgstrasse 3, CH-3010, Bern, Switzerland
| | - Julien Furrer
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Martina Vermathen
- University of Bern, Department of Chemistry and Biochemistry, Freiestrasse 3, CH-3012, Bern, Switzerland.
| |
Collapse
|
50
|
Bresseleers J, Bagheri M, Storm G, Metselaar JM, Hennink WE, Meeuwissen SA, van Hest JCM. Scale-Up of the Manufacturing Process To Produce Docetaxel-Loaded mPEG- b-p(HPMA-Bz) Block Copolymer Micelles for Pharmaceutical Applications. Org Process Res Dev 2019; 23:2707-2715. [PMID: 32952390 PMCID: PMC7493301 DOI: 10.1021/acs.oprd.9b00387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Indexed: 12/22/2022]
Abstract
![]()
An
efficient, scalable, and good manufacturing practice (GMP) compatible
process was developed for the production of docetaxel-loaded poly(ethylene
glycol)-b-poly(N-2-benzoyloxypropyl
methacrylamide) (mPEG-b-p(HPMA-Bz)) micelles. First,
the synthesis of the mPEG-b-p(HPMA-Bz) block copolymer
was optimized through step-by-step investigation of the batch synthesis
procedures. This resulted in the production of 1 kg of mPEG-b-p(HPMA-Bz) block copolymer with a 5 kDa PEG block and
an overall molecular weight of 22.5 kDa. Second, the reproducibility
and scalability of micelle formation was investigated for both batch
and continuous flow setups by assessing critical process parameters.
This resulted in the development of a new and highly efficient continuous
flow process, which led to the production of 100 mL of unloaded micelles
with a size of 55 nm. Finally, the loading of the micelles with the
anticancer drug docetaxel was successfully fine-tuned to obtain precise
control on the loaded micelle characteristics. As a result, 100 mL
of docetaxel-loaded micelles (20 mg/mL polymer and 5 mg/mL docetaxel
in the feed) with a size of 55 nm, an encapsulation efficiency of
65%, a loading capacity of 14%, and stable for at least 2 months in
water at room temperature were produced with the newly developed continuous
flow process. In conclusion, this study paves the way for efficient
and robust large-scale production of docetaxel-loaded micelles with
high encapsulation efficiencies and stability, which is crucial for
their applicability as a clinically relevant drug delivery platform.
Collapse
Affiliation(s)
- Jaleesa Bresseleers
- ChemConnection BV - Ardena Oss, 5349 AB Oss, The Netherlands.,Department of Bio-Organic chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Mahsa Bagheri
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, 3508 TB Utrecht, The Netherlands.,Section - Targeted Therapeutics, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands
| | - Josbert M Metselaar
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging RWTH University Clinic, 52074 Aachen, Germany
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, 3508 TB Utrecht, The Netherlands
| | | | - Jan C M van Hest
- Department of Bio-Organic chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|