1
|
Guo Y, Patel HJ, Patel AS, Squillante E, Patel K. Albendazole nanosuspension coated granules for the rapid localized release and treatment of colorectal cancer. Colloids Surf B Biointerfaces 2024; 245:114320. [PMID: 39423765 DOI: 10.1016/j.colsurfb.2024.114320] [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: 06/17/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
Albendazole (ABZ), an anthelmintic drug, has been repurposed to treat various types of cancers. However, poor solubility of ABZ, resulting in low bioavailability, limits its application. Nanosuspension is a versatile method for enhancing the dissolution of hydrophobic molecules, but a successful drying has been the biggest challenge in the field. The objective of this research is to formulate and optimize ABZ nanosuspension (NS) coated granules for rapid delivery of ABZ for the treatment of colorectal cancer. ABZ NS was prepared by dual centrifugation method using Kollidon® VA64 and sodium lauryl sulphate (SLS) as stabilizers. The processing method was optimized to obtain a stable nanosuspension with particle size < 300 nm. The optimized ABZ NS was coated on microcrystalline cellulose (MCC) to form the nano-coated granules (NCG) and filled in EUDRACAP® for colon targeted delivery. The ABZ NS and NCG achieved ∼ 60 % and ∼55 % drug release, respectively, in presence of bile salt at colonic pH. Half-maximal inhibitory concentration (IC50) of ABZ NS was found to be 1.18 ± 0.081 µM and 3.59 ± 0.080 µM in two colorectal cancer cell lines: HCT 116 and HT-29, respectively. In addition, In vitro 3D tumor assay revealed that ABZ NS has superior tumor growth inhibition activity compared to the control and pure ABZ. The preparation of ABZ NCG in EUDRACAP® could be a promising approach to achieve colon targeted delivery and to repurpose ABZ for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Yi Guo
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | - Henis J Patel
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | - Akanksha S Patel
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | - Emilio Squillante
- College of Pharmacy and Health Sciences, St. John's University, NY, USA.
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John's University, NY, USA.
| |
Collapse
|
2
|
Yadav H, Maiti S. Poly(allylamine)-adorned heptylcarboxymethyl galactomannan nanocarriers of canagliflozin for controlling type-2 diabetes: Optimization by Box-Behnken design and in vivo performance. Int J Biol Macromol 2024; 277:134253. [PMID: 39084426 DOI: 10.1016/j.ijbiomac.2024.134253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/10/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
In the past three decades, the prevalence of type-2 diabetes has arisen dramatically in countries of all income levels. A novel, most effective nanotechnology-based strategy may reduce the prevalence of diabetes. Recently, the shell-crosslinked polysaccharide-based micellar nanocarriers (MNCs) have shown great promise in terms of stability, controlled drug release, and improved in vivo performance. In this study, heptyl carboxymethyl guar gum was synthesized and characterized by ATR-FTIR, 1HNMR spectroscopy, surface charge, critical micelle concentration (23.9 μg/mL), and cytotoxicity analysis. Box-Behnken design was used to optimize the diameter, zeta potential, drug entrapment efficiency (DEE), and drug release characteristics of poly (allylamine)-crosslinked MNCs containing canagliflozin. The optimized MNCs revealed spherical morphology under TEM and had 149.3 nm diameter (PDI 21.2 %), +53.8 mV zeta potential, and 84 % DEE. The MNCs released about 63 % of the drug in 12 h under varying pH of the simulated gastrointestinal fluid. DSC and x-ray analyses suggested amorphous dispersion of drugs in the MNCs. CAM assay demonstrated the biocompatibility of the MNCs. The MNCs showed hemolysis of <1 %, 85 % mucin adsorption, and stability over three months. The MNCs demonstrated excellent anti-diabetic efficacy in streptozotocin-nicotinamide-induced diabetic rats, continuously lowering blood glucose levels up to 12 h.
Collapse
Affiliation(s)
- Harsh Yadav
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh 484887, India
| | - Sabyasachi Maiti
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh 484887, India.
| |
Collapse
|
3
|
Littrell CA, Takacs GP, Sankara CS, Sherman A, Rubach KA, Garcia JS, Bell CA, Lnu T, Harrison JK, Zhang F. Systemically targeting monocytic myloid-derrived suppressor cells using dendrimers and their cell-level biodistribution kinetics. J Control Release 2024; 374:181-193. [PMID: 39103055 DOI: 10.1016/j.jconrel.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
The focus of nanoparticles in vivo trafficking has been mostly on their tissue-level biodistribution and clearance. Recent progress in the nanomedicine field suggests that the targeting of nanoparticles to immune cells can be used to modulate the immune response and enhance therapeutic delivery to the diseased tissue. In the presence of tumor lesions, monocytic-myeloid-derived suppressor cells (M-MDSCs) expand significantly in the bone marrow, egress into peripheral blood, and traffic to the solid tumor, where they help maintain an immuno-suppressive tumor microenvironment. In this study, we investigated the interaction between PAMAM dendrimers and M-MDSCs in two murine models of glioblastoma, by examining the cell-level biodistribution kinetics of the systemically injected dendrimers. We found that M-MDSCs in the tumor and lymphoid organs can efficiently endocytose hydroxyl dendrimers. Interestingly, the trafficking of M-MDSCs from the bone marrow to the tumor contributed to the deposition of hydroxyl dendrimers in the tumor. M-MDSCs showed different capacities of endocytosing dendrimers of different functionalities in vivo. This differential uptake was mediated by the unique serum proteins associated with each dendrimer surface functionality. The results of this study set up the framework for developing dendrimer-based immunotherapy to target M-MDSCs for cancer treatment.
Collapse
Affiliation(s)
- Chad A Littrell
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Gregory P Takacs
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Chenikkayala Siva Sankara
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Alexandra Sherman
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Kai A Rubach
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Julia S Garcia
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Coral A Bell
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Tejashwini Lnu
- Department of Chemical Engineering, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Jeffrey K Harrison
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Fan Zhang
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States; Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States; Department of Chemical Engineering, University of Florida College of Pharmacy, Gainesville, FL, United States.
| |
Collapse
|
4
|
Kapoor DU, Garg R, Maheshwari R, Gaur M, Sharma D, Prajapati BG. Advancing psoriasis drug delivery through topical liposomes. Z NATURFORSCH C 2024; 0:znc-2024-0118. [PMID: 39037729 DOI: 10.1515/znc-2024-0118] [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: 05/14/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
Psoriasis, recognized as a chronic inflammatory skin disorder, disrupts immune system functionality. Global estimates by the World Psoriasis Day consortium indicate its impact on approximately 130 million people, constituting 4 to 5 percent of the worldwide population. Conventional drug delivery systems, mainly designed to alleviate psoriasis symptoms, fall short in achieving targeted action and optimal bioavailability due to inherent challenges such as the drug's brief half-life, instability, and a deficiency in ensuring both safety and efficacy. Liposomes, employed in drug delivery systems, emerge as highly promising carriers for augmenting the therapeutic efficacy of topically applied drugs. These small unilamellar vesicles demonstrate enhanced penetration capabilities, facilitating drug delivery through the stratum corneum layer of skin. This comprehensive review article illuminates diverse facets of liposomes as a promising drug delivery system to treat psoriasis. Addressing various aspects such as formulation strategies, encapsulation techniques, and targeted delivery, the review underscores the potential of liposomes in enhancing the efficacy and specificity of psoriasis treatments.
Collapse
Affiliation(s)
- Devesh U Kapoor
- Dr. Dayaram Patel Pharmacy College, Bardoli 394601, Gujarat, India
| | - Rahul Garg
- Asian College of Pharmacy, Rajasthan University of Health Sciences, Udaipur, Rajasthan 313001, India
| | - Rahul Maheshwari
- School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-University, Green Industrial Park, TSIIC, 509301, Jadcherla, Hyderabad, India
| | - Mansi Gaur
- Rajasthan Pharmacy College, Rajasthan University of Health Sciences, Jaipur 302026, India
| | - Deepak Sharma
- Institute of Pharmacy, Assam Don Bosco University, Tapesia, Assam 782402, India
| | - Bhupendra G Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva 384012, India
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| |
Collapse
|
5
|
Bahrami K, Lee E, Morse B, Lanier OL, Peppas NA. Design of nanoparticle-based systems for the systemic delivery of chemotherapeutics: Alternative potential routes via sublingual and buccal administration for systemic drug delivery. Drug Deliv Transl Res 2024; 14:1173-1188. [PMID: 38151650 DOI: 10.1007/s13346-023-01493-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 12/29/2023]
Abstract
Conventional therapeutic approaches for cancer generally involve chemo- and radiation therapies that often exhibit low efficacy and induce toxic side effects. Recent years have seen significant advancements in the use of protein biologics as a promising alternative treatment option. Nanotechnology-based systems have shown great potential in providing more specific and targeted cancer treatments, thus improving upon many of the limitations associated with current treatments. The unique properties of biomaterial carriers at the nanoscale have been proven to enhance both the performance of the incorporated therapeutic agent and tumor targeting; however, many of these systems are delivered intravenously, which can cause hazardous side effects. Buccal and sublingual delivery systems offer an alternative route for more efficient delivery of nanotechnologies and drug absorption into systemic circulation. This review concentrates on emerging buccal and sublingual nanoparticle delivery systems for chemo- and protein therapeutics, their development, efficacy, and potential areas of improvement in the field. Several factors contribute to the development of effective buccal or sublingual nanoparticle delivery systems, including targeting efficiency of the nanoparticulate carriers, drug release, and carrier biocompatibility. Furthermore, the potential utilization of buccal and sublingual multilayer films combined with nanoparticle chemotherapeutic systems is outlined as a future avenue for in vitro and in vivo research.
Collapse
Affiliation(s)
- Kiana Bahrami
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
- Institute of Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA
- Department of Chemical and Biomolecular Engineering, New York University, Brooklyn, NY, USA
| | - Elaine Lee
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
- Institute of Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA
- School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Brinkley Morse
- Institute of Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
- Department of Neurology, Dell Medical School, University of Texas, Austin, USA
| | - Olivia L Lanier
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
- Institute of Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA.
- Institute of Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA.
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA.
- Department of Surgery and Perioperative Care, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
- Department of Pediatrics, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
6
|
Wang L, Quine S, Frickenstein AN, Lee M, Yang W, Sheth VM, Bourlon MD, He Y, Lyu S, Garcia-Contreras L, Zhao YD, Wilhelm S. Exploring and Analyzing the Systemic Delivery Barriers for Nanoparticles. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2308446. [PMID: 38828467 PMCID: PMC11142462 DOI: 10.1002/adfm.202308446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Indexed: 06/05/2024]
Abstract
Most nanomedicines require efficient in vivo delivery to elicit diagnostic and therapeutic effects. However, en route to their intended tissues, systemically administered nanoparticles often encounter delivery barriers. To describe these barriers, we propose the term "nanoparticle blood removal pathways" (NBRP), which summarizes the interactions between nanoparticles and the body's various cell-dependent and cell-independent blood clearance mechanisms. We reviewed nanoparticle design and biological modulation strategies to mitigate nanoparticle-NBRP interactions. As these interactions affect nanoparticle delivery, we studied the preclinical literature from 2011-2021 and analyzed nanoparticle blood circulation and organ biodistribution data. Our findings revealed that nanoparticle surface chemistry affected the in vivo behavior more than other nanoparticle design parameters. Combinatory biological-PEG surface modification improved the blood area under the curve by ~418%, with a decrease in liver accumulation of up to 47%. A greater understanding of nanoparticle-NBRP interactions and associated delivery trends will provide new nanoparticle design and biological modulation strategies for safer, more effective, and more efficient nanomedicines.
Collapse
Affiliation(s)
- Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Skyler Quine
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Alex N. Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Michael Lee
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Wen Yang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Vinit M. Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Margaret D. Bourlon
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yuxin He
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Shanxin Lyu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Lucila Garcia-Contreras
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yan D. Zhao
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73012, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), Norman, Oklahoma, 73019, USA
| |
Collapse
|
7
|
Mehta P, Shende P. Evasion of opsonization of macromolecules using novel surface-modification and biological-camouflage-mediated techniques for next-generation drug delivery. Cell Biochem Funct 2023; 41:1031-1043. [PMID: 37933222 DOI: 10.1002/cbf.3880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/14/2023] [Accepted: 10/21/2023] [Indexed: 11/08/2023]
Abstract
Opsonization plays a pivotal role in hindering controlled drug release from nanoformulations due to macrophage-mediated nanoparticle destruction. While first and second-generation delivery systems, such as lipoplexes (50-150 nm) and quantum dots, hold immense potential in revolutionizing disease treatment through spatiotemporal controlled drug delivery, their therapeutic efficacy is restricted by the selective labeling of nanoparticles for uptake by reticuloendothelial system and mononuclear phagocyte system via various molecular forces, such as electrostatic, hydrophobic, and van der Waals bonds. This review article presents novel insights into surface-modification techniques utilizing macromolecule-mediated approaches, including PEGylation, di-block copolymerization, and multi-block polymerization. These techniques induce stealth properties by generating steric forces to repel micromolecular-opsonins, such as fibrinogen, thereby mitigating opsonization effects. Moreover, advanced biological methods, like cellular hitchhiking and dysopsonic protein adsorption, are highlighted for their potential to induce biological camouflage by adsorbing onto the nanoparticulate surface, leading to immune escape. These significant findings pave the way for the development of long-circulating next-generation nanoplatforms capable of delivering superior therapy to patients. Future integration of artificial intelligence-based algorithms, integrated with nanoparticle properties such as shape, size, and surface chemistry, can aid in elucidating nanoparticulate-surface morphology and predicting interactions with the immune system, providing valuable insights into the probable path of opsonization.
Collapse
Affiliation(s)
- Parth Mehta
- Department of Pharmaceutics, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be-University, Mumbai, India
| | - Pravin Shende
- Department of Pharmaceutics, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be-University, Mumbai, India
| |
Collapse
|
8
|
Miao G, He Y, Lai K, Zhao Y, He P, Tan G, Wang X. Accelerated blood clearance of PEGylated nanoparticles induced by PEG-based pharmaceutical excipients. J Control Release 2023; 363:12-26. [PMID: 37717659 DOI: 10.1016/j.jconrel.2023.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023]
Abstract
PEGylated nanomedicines have been extensively developed and applied to cancer therapy. However, the antitumor efficacy of these nanoparticles is hampered by the accelerated blood clearance (ABC) effect caused by anti-PEG antibodies in vivo. There is still limited understanding about the cause of pre-existing anti-PEG antibodies in the human body. Herein, we discovered that PEG-based pharmaceutical excipients, commonly used in clinical and daily settings, could induce anti-PEG antibodies in vivo and lead to considerable potential clinical impacts on pharmacokinetics and pharmacodynamics of PEGylated nanoparticles. Specifically, we investigated the ability of poloxamer 188 (F68) and poloxamer 407 (F127), the two most frequently used PEG-based pharmaceutical excipients, to elicit the production of anti-PEG antibodies and influence the pharmacokinetics of PEGylated nanoparticles, with PEGylated liposome nanoparticles (L-NPs) as a model. Anti-PEG IgG and IgM levels were significantly boosted 3.8- and 32.2-fold, respectively, after pre-injection with F68, leading to rapid clearance of subsequently injected L-NPs from circulation due to the capture by neutrophils and monocytes. However, pre-injection of F127 did not induce the production of anti-PEG IgG, although there was a 7.7-fold increase in IgM level, which resulted in minimal effect on circulation time of L-NPs. Furthermore, the potential clinical impacts of F68 and F127 were further inspected for PEGylated liposomal doxorubicin (PLD). It was found that administering F68 prior to treatment led to over a one-third decrease in the antitumor effectiveness of PLD, while F127 had a negligible impact. Our study elucidates the mechanism by which PEG-based pharmaceutical excipients influence the effectiveness of PEGylated nanomedicines. It also highlights the significance of considering the potential for an ABC effect induced by PEG-based pharmaceutical excipients in patients.
Collapse
Affiliation(s)
- Guifeng Miao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Yuejian He
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Keren Lai
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Yan Zhao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Peiyi He
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Guozhu Tan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Xiaorui Wang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China.
| |
Collapse
|
9
|
Kataria S, Qi J, Lin CW, Li Z, Dane EL, Iyer AM, Sacane J, Irvine DJ, Belcher AM. Noninvasive In Vivo Imaging of T-Cells during Cancer Immunotherapy Using Rare-Earth Nanoparticles. ACS NANO 2023; 17:17908-17919. [PMID: 37676036 DOI: 10.1021/acsnano.3c03882] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Only a minority of patients respond positively to cancer immunotherapy, and addressing this variability is an active area of immunotherapy research. Infiltration of tumors by immune cells is one of the most significant prognostic indicators of response and disease-free survival. However, the ability to noninvasively sample the tumor microenvironment for immune cells remains limited. Imaging in the near-infrared-II region using rare-earth nanocrystals is emerging as a powerful imaging tool for high-resolution deep-tissue imaging. In this paper, we demonstrate that these nanoparticles can be used for noninvasive in vivo imaging of tumor-infiltrating T-cells in a highly aggressive melanoma tumor model. We present nanoparticle synthesis and surface modification strategies for the generation of small, ultrabright, and biocompatible rare-earth nanocrystals necessary for deep tissue imaging of rare cell types. The ability to noninvasively monitor the immune contexture of a tumor during immunotherapy could lead to early identification of nonresponding patients in real time, leading to earlier interventions and better outcomes.
Collapse
Affiliation(s)
- Swati Kataria
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Jifa Qi
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Ching-Wei Lin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Zhongming Li
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Eric L Dane
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Archana Mahadevan Iyer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Jay Sacane
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02139, United States
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, United States
| | - Angela M Belcher
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
10
|
Amirahmadi M, Hosseinkhani S, Hosseini M, Yaghmei P, Heydari A. Fe 3O 4@SiO 2@NiAl-LDH microspheres implication in separation, kinetic and structural properties of phenylalanine dehydrogenase. Heliyon 2023; 9:e19429. [PMID: 37809670 PMCID: PMC10558515 DOI: 10.1016/j.heliyon.2023.e19429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/12/2023] [Accepted: 08/22/2023] [Indexed: 10/10/2023] Open
Abstract
Fe3O4@SiO2@NiAl-LDH three-components microsphere contains a Fe3O4@SiO2 magnetic core and a layered double hydroxide with nickel cation provide the binding ability to (His)-tagged-protein and exhibits high performance in protein separation and purification. The morphology and chemistry of the synthesized Fe3O4@SiO2@NiAl-LDH microspheres were characterized by energy-dispersive X-ray spectroscopy (EDX), scanning electron microscopy (SEM), X-ray diffraction (XRD), Fourier transform infrared (FTIR), vibrating sample magnetometer (VSM), Dynamic light scattering (DLS). Purified enzyme was assesed with SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis and intrinsic fluorescence spectroscopy. In this study, the separation of phenylalanine dehydrogenase (PheDH) by Fe3O4@SiO2@NiAl -LDH was performed and the effect of microsphere was investigated on the kinetic and structural properties of PheDH. After purification, kinetic parameters such as Km, Vmax, Kcat, kcat/Km, optimum temperature, thermal stability, and and activation energy were evaluated and compared according to the mentioned methods. The interaction between the enzyme and the microsphere displayed a high performance in protein binding capacity. The results also revealed that the kinetic parameters of the enzyme changed in a dose-dependent manner in the presence of a microsphere. Moreover, the results of intrinsic fluorescence and Circular Dichroism (CD) confirmed the structural changes of the protein in the interaction with the microsphere.
Collapse
Affiliation(s)
- Mozhgan Amirahmadi
- Department of Biochemistry, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-175, Iran
| | - Morteza Hosseini
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran 1417614418, Iran
| | - Paricher Yaghmei
- Department of Biochemistry, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Akbar Heydari
- Chemistry Department, Tarbiat Modares University, P.O. Box 14155-4838, Tehran, Iran
| |
Collapse
|
11
|
Shafaei N, Khorshidi S, Karkhaneh A. The immune-stealth polymeric coating on drug delivery nanocarriers: In vitro engineering and in vivo fate. J Biomater Appl 2023:8853282231185352. [PMID: 37480331 DOI: 10.1177/08853282231185352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Although essential nanosystems such as nanoparticles and nanocarriers are desirable options for transporting various drug molecules into the biological environment, they rapidly remove from the circulatory system due to their interaction with multiple in vivo barriers, especially the immune barrier, which will result in their short-term effects. In order to improve their effectiveness and durability in the circulatory system, the polymer coatings can use to cover the surface of nanoparticles and nanocarriers to conceal them from the immune system. Due to their different properties (like charge, elasticity, and hydrophilicity/hydrophobicity), these coatings can improve drug delivery nanosystem durability and therapeutic applications. The mentioned coatings have different types and are divided into various categories, such as synthetic polymers, polysaccharides, and zwitterionic polymers. Each of these polymers has unique properties based on its category, origin, and chemical structure that make them suitable for producing stealth drug delivery nanocarriers. In this review article, we have tried to explain the importance of these diverse polymer coatings in determining the fate of drug nanocarriers and then introduced the different types of these coatings and, finally, described various methods that directly and indirectly analyze the nanocoatings to determine the stability of nanoparticles in the body.
Collapse
Affiliation(s)
- Nadia Shafaei
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Sajedeh Khorshidi
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Akbar Karkhaneh
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
12
|
Fudalej M, Kwaśniewska D, Nurzyński P, Badowska-Kozakiewicz A, Mękal D, Czerw A, Sygit K, Deptała A. New Treatment Options in Metastatic Pancreatic Cancer. Cancers (Basel) 2023; 15:cancers15082327. [PMID: 37190255 DOI: 10.3390/cancers15082327] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/12/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023] Open
Abstract
Pancreatic cancer (PC) is the seventh leading cause of cancer death across the world. Poor prognosis of PC is associated with several factors, such as diagnosis at an advanced stage, early distant metastases, and remarkable resistance to most conventional treatment options. The pathogenesis of PC seems to be significantly more complicated than originally assumed, and findings in other solid tumours cannot be extrapolated to this malignancy. To develop effective treatment schemes prolonging patient survival, a multidirectional approach encompassing different aspects of the cancer is needed. Particular directions have been established; however, further studies bringing them all together and connecting the strengths of each therapy are needed. This review summarises the current literature and provides an overview of new or emerging therapeutic strategies for the more effective management of metastatic PC.
Collapse
Affiliation(s)
- Marta Fudalej
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Daria Kwaśniewska
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Paweł Nurzyński
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | | | - Dominika Mękal
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Aleksandra Czerw
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland
- Department of Economic and System Analyses, National Institute of Public Health NIH-National Research Institute, 00-791 Warsaw, Poland
| | - Katarzyna Sygit
- Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland
| | - Andrzej Deptała
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| |
Collapse
|
13
|
Zhang X, Misra SK, Moitra P, Zhang X, Jeong SJ, Stitham J, Rodriguez-Velez A, Park A, Yeh YS, Gillanders WE, Fan D, Diwan A, Cho J, Epelman S, Lodhi IJ, Pan D, Razani B. Use of acidic nanoparticles to rescue macrophage lysosomal dysfunction in atherosclerosis. Autophagy 2023; 19:886-903. [PMID: 35982578 PMCID: PMC9980706 DOI: 10.1080/15548627.2022.2108252] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/19/2022] Open
Abstract
Dysfunction in the macrophage lysosomal system including reduced acidity and diminished degradative capacity is a hallmark of atherosclerosis, leading to blunted clearance of excess cellular debris and lipids in plaques and contributing to lesion progression. Devising strategies to rescue this macrophage lysosomal dysfunction is a novel therapeutic measure. Nanoparticles have emerged as an effective platform to both target specific tissues and serve as drug delivery vehicles. In most cases, administered nanoparticles are taken up non-selectively by the mononuclear phagocyte system including monocytes/macrophages leading to the undesirable degradation of cargo in lysosomes. We took advantage of this default route to target macrophage lysosomes to rectify their acidity in disease states such as atherosclerosis. Herein, we develop and test two commonly used acidic nanoparticles, poly-lactide-co-glycolic acid (PLGA) and polylactic acid (PLA), both in vitro and in vivo. Our results in cultured macrophages indicate that the PLGA-based nanoparticles are the most effective at trafficking to and enhancing acidification of lysosomes. PLGA nanoparticles also provide functional benefits including enhanced lysosomal degradation, promotion of macroautophagy/autophagy and protein aggregate removal, and reduced apoptosis and inflammasome activation. We demonstrate the utility of this system in vivo, showing nanoparticle accumulation in, and lysosomal acidification of, macrophages in atherosclerotic plaques. Long-term administration of PLGA nanoparticles results in significant reductions in surrogates of plaque complexity with reduced apoptosis, necrotic core formation, and cytotoxic protein aggregates and increased fibrous cap formation. Taken together, our data support the use of acidic nanoparticles to rescue macrophage lysosomal dysfunction in the treatment of atherosclerosis.Abbreviations: BCA: brachiocephalic arteries; FACS: fluorescence activated cell sorting; FITC: fluorescein-5-isothiocyanatel; IL1B: interleukin 1 beta; LAMP: lysosomal associated membrane protein; LIPA/LAL: lipase A, lysosomal acid type; LSDs: lysosomal storage disorders; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MFI: mean fluorescence intensity; MPS: mononuclear phagocyte system; PEGHDE: polyethylene glycol hexadecyl ether; PLA: polylactic acid; PLGA: poly-lactide-co-glycolic acid; SQSTM1/p62: sequestosome 1.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | - Santosh Kumar Misra
- Department of Bioengineering, University of Illinois at Urbana Champaign, IL, USA
| | - Parikshit Moitra
- Departments of Diagnostic Radiology and Nuclear Medicine and Pediatrics, Baltimore, Maryland, USA
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania16802, USA
| | - Xiuli Zhang
- Department of Surgery, Washington University, St. Louis, MO, USA
| | - Se-Jin Jeong
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | - Jeremiah Stitham
- Cardiovascular Division, Washington University, St. Louis, MO, USA
- Division of Endocrinology, Metabolism, and Lipid Research, St. Louis, MO, USA
| | | | - Arick Park
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | - Yu-Sheng Yeh
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | | | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Abhinav Diwan
- Cardiovascular Division, Washington University, St. Louis, MO, USA
- John Cochran Division, VA Medical Center, St. Louis, MO, USA
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Slava Epelman
- Peter Munk Cardiac Center, Toronto General Hospital Research Institute, University Health Network, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Irfan J. Lodhi
- Division of Endocrinology, Metabolism, and Lipid Research, St. Louis, MO, USA
| | - Dipanjan Pan
- Department of Bioengineering, University of Illinois at Urbana Champaign, IL, USA
- Departments of Diagnostic Radiology and Nuclear Medicine and Pediatrics, Baltimore, Maryland, USA
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania16802, USA
| | - Babak Razani
- Cardiovascular Division, Washington University, St. Louis, MO, USA
- John Cochran Division, VA Medical Center, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
14
|
Elmowafy M, Shalaby K, Elkomy MH, Alsaidan OA, Gomaa HAM, Abdelgawad MA, Mostafa EM. Polymeric Nanoparticles for Delivery of Natural Bioactive Agents: Recent Advances and Challenges. Polymers (Basel) 2023; 15:1123. [PMID: 36904364 PMCID: PMC10007077 DOI: 10.3390/polym15051123] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
In the last few decades, several natural bioactive agents have been widely utilized in the treatment and prevention of many diseases owing to their unique and versatile therapeutic effects, including antioxidant, anti-inflammatory, anticancer, and neuroprotective action. However, their poor aqueous solubility, poor bioavailability, low GIT stability, extensive metabolism as well as short duration of action are the most shortfalls hampering their biomedical/pharmaceutical applications. Different drug delivery platforms have developed in this regard, and a captivating tool of this has been the fabrication of nanocarriers. In particular, polymeric nanoparticles were reported to offer proficient delivery of various natural bioactive agents with good entrapment potential and stability, an efficiently controlled release, improved bioavailability, and fascinating therapeutic efficacy. In addition, surface decoration and polymer functionalization have opened the door to improving the characteristics of polymeric nanoparticles and alleviating the reported toxicity. Herein, a review of the state of knowledge on polymeric nanoparticles loaded with natural bioactive agents is presented. The review focuses on frequently used polymeric materials and their corresponding methods of fabrication, the needs of such systems for natural bioactive agents, polymeric nanoparticles loaded with natural bioactive agents in the literature, and the potential role of polymer functionalization, hybrid systems, and stimuli-responsive systems in overcoming most of the system drawbacks. This exploration may offer a thorough idea of viewing the polymeric nanoparticles as a potential candidate for the delivery of natural bioactive agents as well as the challenges and the combating tools used to overcome any hurdles.
Collapse
Affiliation(s)
- Mohammed Elmowafy
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Khaled Shalaby
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Mohammed H. Elkomy
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Hesham A. M. Gomaa
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Ehab M. Mostafa
- Department of Pharmacognosy, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| |
Collapse
|
15
|
Bi F, Yu D, Wei Z, Wei H, Ren H, Wang Y, Ren D, Hua Z, Huang B, Yang G. Core-Shell Polymeric Nanostructures with Intracellular ATP-Fueled dsRNA Delivery toward Genetic Control of Insect Pests. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:2762-2772. [PMID: 36745409 DOI: 10.1021/acs.jafc.2c05737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Transgenic RNA interference (RNAi) represents a burgeoning and promising alternative avenue to manage plant diseases and insect pests in plants. Nonviral nanostructured dsRNA carriers have been demonstrated to possess great potential to facilitate the application of RNAi. However, it remains a critical challenge to achieve the targeted and effective release of dsRNA into the pest cells, limiting the efficiency of the biological control of pests and diseases in practical applications. In this study, we designed and constructed a new type of core-shell polymeric nanostructure (CSPN) with controllable structure, eco-friendliness, and good biocompatibility, on which dsRNA can be efficiently loaded. Once loaded into CSPNs, the dsRNA can be effectively prevented from nonsense degradation by enzymes before entering cells, and it shows targeted and image-guided release triggered by intracellular ATP, which significantly increases the efficiency of gene transfection. Significantly, the in vivo study of the typical lepidoptera silkworm after oral feeding demonstrates the potential of dsCHT10 in CSPNs for a much better knockdown efficiency than that of naked dsCHT10. This innovation enables the nanotechnology developed for the disease microenvironment-triggered release of therapeutic genes for application in sustainable crop protection.
Collapse
Affiliation(s)
- Feihu Bi
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Deshui Yu
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Zengming Wei
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Hanchen Wei
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Hui Ren
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Yulong Wang
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Dalong Ren
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Zan Hua
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Bo Huang
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Guang Yang
- Biomass Molecular Engineering Center and Department of Materials Science and Engineering, Anhui Agricultural University, Hefei 230036, Anhui, China
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei 230036, Anhui, China
| |
Collapse
|
16
|
Colby AH, Kirsch J, Patwa AN, Liu R, Hollister B, McCulloch W, Burdette JE, Pearce CJ, Oberliels NH, Colson YL, Liu K, Grinstaff MW. Radiolabeled Biodistribution of Expansile Nanoparticles: Intraperitoneal Administration Results in Tumor Specific Accumulation. ACS NANO 2023; 17:2212-2221. [PMID: 36701244 PMCID: PMC9933882 DOI: 10.1021/acsnano.2c08451] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/23/2023] [Indexed: 05/18/2023]
Abstract
Nanoparticle biodistribution in vivo is an essential component to the success of nanoparticle-based drug delivery systems. Previous studies with fluorescently labeled expansile nanoparticles, or "eNPs", demonstrated a high specificity of eNPs to tumors that is achieved through a materials-based targeting strategy. However, fluorescent labeling techniques are primarily qualitative in nature and the gold-standard for quantitative evaluation of biodistribution is through radiolabeling. In this manuscript, we synthesize 14C-labeled eNPs to quantitatively evaluate the biodistribution of these particles in a murine model of intraperitoneal mesothelioma via liquid scintillation counting. The results demonstrate a strong specificity of eNPs for tumors that lasts one to 2 weeks postinjection with an overall delivery efficiency to the tumor tissue of 30% of the injected dose which is congruent with prior reports of preclinical efficacy of the technology. Importantly, the route of administration is essential to the eNP's material-based targeting strategy with intraperitoneal administration leading to tumoral accumulation while, in contrast, intravenous administration leads to rapid clearance via the reticuloendothelial system and low tumoral accumulation. A comparison against nanoparticle delivery systems published over the past decade shows that the 30% tumoral delivery efficiency of the eNP is significantly higher than the 0.7% median delivery efficiency of other systems with sufficient quantitative data to define this metric. These results lay a foundation for targeting intraperitoneal tumors and encourage efforts to explore alternative, nonintravenous routes, of delivery to accelerate the translation of nanoparticle therapies to the clinic.
Collapse
Affiliation(s)
- Aaron H. Colby
- Boston
University, Boston, Massachusetts 02215, United States
- Ionic
Pharmaceuticals, LLC, Watertown, Massachusetts 02472, United States
| | - Jack Kirsch
- Boston
University, Boston, Massachusetts 02215, United States
| | - Amit N. Patwa
- Boston
University, Boston, Massachusetts 02215, United States
- Navrachana
University, Vadodara 391410, India
| | - Rong Liu
- Massachusetts
General Hospital, Boston, Massachusetts 02114, United States
| | - Beth Hollister
- HighRock
Consulting, Oxford, North Carolina 27565, United States
| | - William McCulloch
- Alba BioPharm
Advisors, Inc., Raleigh, North Carolina 27614, United States
| | - Joanna E. Burdette
- University
of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Cedric J. Pearce
- Mycosynthetix,
Inc., Hillsborough, North Carolina 27278, United States
| | - Nicholas H. Oberliels
- Ionic
Pharmaceuticals, LLC, Watertown, Massachusetts 02472, United States
- University
of North Carolina at Greensboro, Greensboro, North Carolina 27412, United States
| | - Yolonda L. Colson
- Massachusetts
General Hospital, Boston, Massachusetts 02114, United States
| | - Kebin Liu
- Augusta
University, Augusta, Georgia 30912, United States
| | - Mark W. Grinstaff
- Boston
University, Boston, Massachusetts 02215, United States
- Ionic
Pharmaceuticals, LLC, Watertown, Massachusetts 02472, United States
| |
Collapse
|
17
|
Suvarna V, Sawant N, Desai N. A Review on Recent Advances in Mannose-Functionalized Targeted Nanocarrier Delivery Systems in Cancer and Infective Therapeutics. Crit Rev Ther Drug Carrier Syst 2023; 40:43-82. [PMID: 36734913 DOI: 10.1615/critrevtherdrugcarriersyst.2022041853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Unmodified nanocarriers used in the chemotherapy of cancers and various infectious diseases exhibit prolonged blood circulation time, prevent enzymatic degradation and increase chemical stability of encapsulated therapeutics. However, off-target effect and lack of specificity associated with unmodified nanoparticles (NPs) limit their applications in the health care system. Mannose (Man) receptors with significant overexpression on antigen-presenting cells and macrophages are among the most admired targets for cancer and anti-infective therapeutics. Therefore, development of Man functionalized nanocarriers targeting Man receptors, for target specific drug delivery in the chemotherapy have been extensively studied. Present review expounds diverse Man-conjugated NPs with their potential for targeted drug delivery, improved biodistribution profiles and localization. Additionally, the review gives detailed account of the interactions of mannosylated NPs with various biological systems and their characterization not discussed in earlier published reports is discussed.
Collapse
Affiliation(s)
- Vasanti Suvarna
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai 400056, Maharashtra, India
| | - Niserga Sawant
- C.U. Shah College of Pharmacy, SNDT Women's University, Santacruz (W), Mumbai 400049, Maharashtra, India
| | - Namita Desai
- Department of Pharmaceutics, C. U. Shah College of Pharmacy, SNDT Women's University, Santacruz (W), Mumbai - 400049, Maharashtra, India
| |
Collapse
|
18
|
Kandasamy G, Maity D. Current Advancements in Self-assembling Nanocarriers-Based siRNA Delivery for Cancer Therapy. Colloids Surf B Biointerfaces 2022; 221:113002. [PMID: 36370645 DOI: 10.1016/j.colsurfb.2022.113002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/01/2022] [Accepted: 10/30/2022] [Indexed: 11/07/2022]
Abstract
Different therapeutic practices for treating cancers have significantly evolved to compensate and/or overcome the failures in conventional methodologies. The demonstrated potentiality in completely inhibiting the tumors and in preventing cancer relapse has made nucleic acids therapy (NAT)/gene therapy as an attractive practice. This has been made possible because NAT-based cancer treatments are highly focused on the fundamental mechanisms - i.e., silencing the expression of oncogenic genes responsible for producing abnormal proteins (via messenger RNAs (mRNAs)). However, the future clinical translation of NAT is majorly dependent upon the effective delivery of the exogenous nucleic acids (especially RNAs - e.g., short interfering RNAs (siRNAs) - herein called biological drugs). Moreover, nano-based vehicles (i.e., nanocarriers) are involved in delivering them to prevent degradation and undesired bioaccumulation while enhancing the stability of siRNAs. Herein, we have initially discussed about three major types of self-assembling nanocarriers (liposomes, polymeric nanoparticles and exosomes). Later, we have majorly reviewed recent developments in non-targeted/targeted nanocarriers for delivery of biological drugs (individual/dual) to silence the most important genes/mRNAs accountable for inducing protein abnormality. These proteins include polo-like kinase 1 (PLK1), survivin, vascular endothelial growth factor (VEGF), B-cell lymphoma/leukaemia-2 (Bcl-2) and multi-drug resistance (MDR). Besides, the consequent therapeutic effects on cancer growth, invasion and/or metastasis have also been discussed. Finally, we have comprehensively reviewed the improvements achieved in the cutting-edge cancer therapeutics while delivering siRNAs in combination with clinically approved chemotherapeutic drugs.
Collapse
|
19
|
Recent approaches to mRNA vaccine delivery by lipid-based vectors prepared by continuous-flow microfluidic devices. Future Med Chem 2022; 14:1561-1581. [DOI: 10.4155/fmc-2022-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Advancements in nanotechnology have resulted in the introduction of several nonviral delivery vectors for the nontoxic, efficient delivery of encapsulated mRNA-based vaccines. Lipid- and polymer-based nanoparticles (NP) have proven to be the most potent delivery systems, providing increased delivery efficiency and protection of mRNA molecules from degradation. Here, the authors provide an overview of the recent studies carried out using lipid NPs and their functionalized forms, polymeric and lipid-polymer hybrid nanocarriers utilized mainly for the encapsulation of mRNAs for gene and immune therapeutic applications. A microfluidic system as a prevalent methodology for the preparation of NPs with continuous flow enables NP size tuning, rapid mixing and production reproducibility. Continuous-flow microfluidic devices for lipid and polymeric encapsulated RNA NP production are specifically reviewed.
Collapse
|
20
|
Wu JLY, Stordy BP, Nguyen LNM, Deutschman CP, Chan WCW. A proposed mathematical description of in vivo nanoparticle delivery. Adv Drug Deliv Rev 2022; 189:114520. [PMID: 36041671 DOI: 10.1016/j.addr.2022.114520] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/10/2022] [Accepted: 08/23/2022] [Indexed: 02/06/2023]
Abstract
Nanoparticles are promising vehicles for the precise delivery of molecular therapies to diseased sites. Nanoparticles interact with a series of tissues and cells before they reach their target, which causes less than 1% of administered nanoparticles to be delivered to these target sites. Researchers have been studying the nano-bio interactions that mediate nanoparticle delivery to develop guidelines for designing nanoparticles with enhanced delivery properties. In this review article, we describe these nano-bio interactions with a series of mathematical equations that quantitatively define the nanoparticle delivery process. We employ a compartment model framework to describe delivery where nanoparticles are either (1) at the site of administration, (2) in the vicinity of target cells, (3) internalized by the target cells, or (4) sequestered away in off-target sites or eliminated from the body. This framework explains how different biological processes govern nanoparticle transport between these compartments, and the role of intercompartmental transport rates in determining the final nanoparticle delivery efficiency. Our framework provides guiding principles to engineer nanoparticles for improved targeted delivery.
Collapse
Affiliation(s)
- Jamie L Y Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Benjamin P Stordy
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Luan N M Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Christopher P Deutschman
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Warren C W Chan
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON M5S 3E5, Canada; Department of Materials Science & Engineering, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada.
| |
Collapse
|
21
|
Szabó R, Rácz CP, Dulf FV. Bioavailability Improvement Strategies for Icariin and Its Derivates: A Review. Int J Mol Sci 2022; 23:ijms23147519. [PMID: 35886867 PMCID: PMC9318307 DOI: 10.3390/ijms23147519] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
In recent years, there has been considerable interest in icariin (ICA) and its derivates, icariside II (ICS) and icaritin (ICT), due to their wide range of potential applications in preventing cancer, cardiovascular disease, osteoporosis, delaying the effects of Alzheimer’s disease, treating erectile dysfunction, etc. However, their poor water solubility and membrane permeability, resulting in low bioavailability, dampens their potential beneficial effects. In this regard, several strategies have been developed, such as pharmaceutical technologies, structural transformations, and absorption enhancers. All these strategies manage to improve the bioavailability of the above-mentioned flavonoids, thus increasing their concentration in the desired places. This paper focuses on gathering the latest knowledge on strategies to improve bioavailability for enhancing the efficacy of icariin, icariside II, and icaritin. We conclude that there is an opportunity for many further improvements in this field. To the best of our knowledge, no such review articles scoping the bioavailability improvement of icariin and its derivates have been published to date. Therefore, this paper can be a good starting point for all those who want to deepen their understanding of the field.
Collapse
Affiliation(s)
- Róbert Szabó
- Department of Environmental and Plant Protection, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Calea Mănăştur 3-5, 400372 Cluj-Napoca, Romania;
| | - Csaba Pál Rácz
- Faculty of Chemistry and Chemical Engineering, Babeș-Bolyai University of Cluj-Napoca, Arany János 11, 400028 Cluj-Napoca, Romania;
| | - Francisc Vasile Dulf
- Department of Environmental and Plant Protection, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Calea Mănăştur 3-5, 400372 Cluj-Napoca, Romania;
- Correspondence:
| |
Collapse
|
22
|
Honmane SM, Charde MS, Salunkhe SS, Choudhari PB, Nangare SN. Polydopamine surface-modified nanocarriers for improved anticancer activity: Current progress and future prospects. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
23
|
Small tumour microparticle enhances drug delivery efficiency and therapeutic antitumour efficacy. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00125-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Targeted delivery of chemotherapeutic drugs to tumour cells is a major challenge for cancer chemotherapy. Recent studies show that tumour cell-derived microparticles can be used as vectors to package chemotherapeutic drugs, and selectively deliver drugs to tumour cells. Nevertheless, since the particle size range of microparticles is relatively wide, the sizes may exhibit different pharmacokinetic characteristics in the body, which will have a great impact on the application of drug-loaded microparticles. Here in this report, we compare the characteristics, distribution in vivo and antitumour efficacy of small microparticles (SMPs, ≤ 200 nm) and large microparticles (LMPs, > 200 nm) which loaded with methotrexate, in order to screen out more suitable carrier sizes.
Results
In vivo and in vitro studies have proved that the drug-loaded vesicles of SMPs (mainly 100–200 nm) are more reasonable, and the drug content and maintenance in tumour tissues. The time is significantly higher than that of LMPs (mainly 400–500 nm). At the same time, we found that SMPs can be better taken up and processed by DC cells to promote the proliferation of specific T cells. SMPs show obvious advantages in both drug delivery and immune activation, which is verified by the comparison of the efficacy of SMPs and LMPs in the treatment of solid tumours in mice.
Conclusions
The present data demonstrate that the SMPs had a higher cumulative concentration in tumour tissue, and the tumour suppressive effect was also significantly better than that of LMPs. It provides important process parameters for the drug-loaded vesicle delivery system. Future works will aim to expand production scale and improve the separation and purification process of the microparticles. Although the research and application of drug-loaded vesicles derived from tumour cells is still in its infancy, it has broad prospects for tumour therapy.
Collapse
|
24
|
Gairola A, Benjamin A, Weatherston JD, Cirillo JD, Wu HJ. Recent Developments in Drug Delivery for Treatment of Tuberculosis by Targeting Macrophages. ADVANCED THERAPEUTICS 2022; 5:2100193. [PMID: 36203881 PMCID: PMC9531895 DOI: 10.1002/adtp.202100193] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Indexed: 11/10/2022]
Abstract
Tuberculosis (TB) is among the greatest public health and safety concerns in the 21st century, Mycobacterium tuberculosis, which causes TB, infects alveolar macrophages and uses these cells as one of its primary sites of replication. The current TB treatment regimen, which consist of chemotherapy involving a combination of 3-4 antimicrobials for a duration of 6-12 months, is marked with significant side effects, toxicity, and poor compliance. Targeted drug delivery offers a strategy that could overcome many of the problems of current TB treatment by specifically targeting infected macrophages. Recent advances in nanotechnology and material science have opened an avenue to explore drug carriers that actively and passively target macrophages. This approach can increase the drug penetration into macrophages by using ligands on the nanocarrier that interact with specific receptors for macrophages. This review encompasses the recent development of drug carriers specifically targeting macrophages actively and passively. Future directions and challenges associated with development of effective TB treatment is also discussed.
Collapse
Affiliation(s)
- Anirudh Gairola
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Aaron Benjamin
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Joshua D Weatherston
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Jeffrey D Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Hung-Jen Wu
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
25
|
Nsairat H, Khater D, Sayed U, Odeh F, Al Bawab A, Alshaer W. Liposomes: structure, composition, types, and clinical applications. Heliyon 2022; 8:e09394. [PMID: 35600452 PMCID: PMC9118483 DOI: 10.1016/j.heliyon.2022.e09394] [Citation(s) in RCA: 284] [Impact Index Per Article: 142.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/19/2022] [Accepted: 05/06/2022] [Indexed: 12/18/2022] Open
Abstract
Liposomes are now considered the most commonly used nanocarriers for various potentially active hydrophobic and hydrophilic molecules due to their high biocompatibility, biodegradability, and low immunogenicity. Liposomes also proved to enhance drug solubility and controlled distribution, as well as their capacity for surface modifications for targeted, prolonged, and sustained release. Based on the composition, liposomes can be considered to have evolved from conventional, long-circulating, targeted, and immune-liposomes to stimuli-responsive and actively targeted liposomes. Many liposomal-based drug delivery systems are currently clinically approved to treat several diseases, such as cancer, fungal and viral infections; more liposomes have reached advanced phases in clinical trials. This review describes liposomes structure, composition, preparation methods, and clinical applications.
Collapse
Affiliation(s)
- Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Dima Khater
- Department of Chemistry, Faculty of Arts and Science, Applied Science Private University, Amman, Jordan
| | - Usama Sayed
- Department of Biology, The University of Jordan, Amman, 11942, Jordan
| | - Fadwa Odeh
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan
| | - Abeer Al Bawab
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.,Hamdi Mango Center for Scientific Research, The University of Jordan, Amman, 11942, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
26
|
Malhotra S, Dumoga S, Singh N. Red blood cells membrane-derived nanoparticles: Applications and key challenges in their clinical translation. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1776. [PMID: 35106966 DOI: 10.1002/wnan.1776] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/16/2021] [Accepted: 10/12/2021] [Indexed: 12/20/2022]
Abstract
Cellular membrane-derived nanoparticles, particularly of red blood cells (RBCs), represent an emerging class of drug delivery systems. The lack of nucleus and organelles in these cells makes them easy to process and empty out intracellular contents. The empty vesicle membranes can then be either used as a coating on nanoparticles or can be reassembled into a nanovesicle. Engineered RBCs membrane has unique ability to retain its lipid bilayer architecture with host's proteins during top-down approach, thus allowing it to form stable nanoformulations mimicking RBCs stealth properties. In addition, its core-shell structure allows loading of different drug molecules, and its surface chemistry can be manipulated by facile conjugation with ligands on the shell. The remarkable ability of RBCs membrane to fuse with membranes of other cells enables the formation of hybrid nanovesicles. In this review, we highlight the biomedical applications of such vesicles and discuss the potential challenges related to its clinical translation. Although nano-RBCs retain much of the host's proteins, which may give an edge over synthetic nanoparticles in terms of lower immunogenicity, its production at industrial level is more challenging. This review gives the critical analysis of barriers involved in the translation of RBCs-derived nanoparticles from preclinical to clinical level. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Lipid-Based Structures Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Sahil Malhotra
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Shweta Dumoga
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India.,Biomedical Engineering unit, All India Institute of Medical Sciences New Delhi, New Delhi, India
| |
Collapse
|
27
|
Re-directing nanomedicines to the spleen: A potential technology for peripheral immunomodulation. J Control Release 2022; 350:60-79. [DOI: 10.1016/j.jconrel.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 11/23/2022]
|
28
|
Kesharwani P, Chadar R, Sheikh A, Rizg WY, Safhi AY. CD44-Targeted Nanocarrier for Cancer Therapy. Front Pharmacol 2022; 12:800481. [PMID: 35431911 PMCID: PMC9008230 DOI: 10.3389/fphar.2021.800481] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023] Open
Abstract
Cluster of differentiation 44 (CD44) is a cell surface glycoprotein overexpressed in varieties of solid tumors including pancreatic, breast, ovary, brain, and lung cancers. It is a multi-structural glycoprotein of the cell surface which is majorly involved in cell proliferation, cell-to-cell interaction, cellular migration, inflammation, and generation of immune responses. Numerous studies focus on the development of nanocarriers for active targeting of the CD44 receptor to improve efficacy of targeting chemotherapy and achieve precise chemotherapy by defining the release, uptake, and accumulation of therapeutic agents. The CD44 receptor has a selective binding affinity towards hyaluronic and chondroitin sulfate (CS). Taking this into consideration, this review focused on the role of CD44 in cancer and its therapy using several nanocarriers such as polymeric/non-polymeric nanoparticles, dendrimer, micelles, carbon nanotubes, nanogels, nanoemulsions etc., for targeted delivery of several chemotherapeutic molecules and nucleic acid. This review also illuminates the role of hyaluronic acid (HA) in cancer therapy, interaction of HA with CD44, and various approaches to target CD44-overexpressed neoplastic cells.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- *Correspondence: Prashant Kesharwani,
| | - Rahul Chadar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Waleed Y. Rizg
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Awaji Y Safhi
- Department of Pharmaceutics, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
29
|
Liposomal-Based Formulations: A Path from Basic Research to Temozolomide Delivery Inside Glioblastoma Tissue. Pharmaceutics 2022; 14:pharmaceutics14020308. [PMID: 35214041 PMCID: PMC8875825 DOI: 10.3390/pharmaceutics14020308] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma (GBM) is a lethal brain cancer with a very difficult therapeutic approach and ultimately frustrating results. Currently, therapeutic success is mainly limited by the high degree of genetic and phenotypic heterogeneity, the blood brain barrier (BBB), as well as increased drug resistance. Temozolomide (TMZ), a monofunctional alkylating agent, is the first line chemotherapeutic drug for GBM treatment. Yet, the therapeutic efficacy of TMZ suffers from its inability to cross the BBB and very short half-life (~2 h), which requires high doses of this drug for a proper therapeutic effect. Encapsulation in a (nano)carrier is a promising strategy to effectively improve the therapeutic effect of TMZ against GBM. Although research on liposomes as carriers for therapeutic agents is still at an early stage, their integration in GBM treatment has a great potential to advance understanding and treating this disease. In this review, we provide a critical discussion on the preparation methods and physico-chemical properties of liposomes, with a particular emphasis on TMZ-liposomal formulations targeting GBM developed within the last decade. Furthermore, an overview on liposome-based formulations applied to translational oncology and clinical trials formulations in GBM treatment is provided. We emphasize that despite many years of intense research, more careful investigations are still needed to solve the main issues related to the manufacture of reproducible liposomal TMZ formulations for guaranteed translation to the market.
Collapse
|
30
|
Markova E, Taneska L, Kostovska M, Shalabalija D, Mihailova L, Glavas Dodov M, Makreski P, Geskovski N, Petrushevska M, N Taravari A, Simonoska Crcarevska M. Design and evaluation of nanostructured lipid carriers loaded with Salvia officinalis extract for Alzheimer's disease treatment. J Biomed Mater Res B Appl Biomater 2022; 110:1368-1390. [PMID: 35019231 DOI: 10.1002/jbm.b.35006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/24/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022]
Abstract
Considering the potential of Salvia officinalis in prevention and treatment of Alzheimer's disease (AD), as well as the ability of nanostructured lipid carriers (NLC) to successfully deliver drug molecules across blood-brain barrier (BBB), the objective of this study was design, development, optimization and characterization of freeze-dried salvia officinalis extract (FSE) loaded NLC intended for intranasal administration. NLC were prepared by solvent evaporation method and the optimization was carried out using central composite design (CCD) of experiments. Further, the optimized formulation (NLCo) was coated either with chitosan (NLCc) or poloxamer (NLCp). Surface characterization of the particles demonstrated a spherical shape with smooth exterior. Particle size of optimal formulations after 0.45 μm pore size filtration ranged from 127 ± 0.68 nm to 140 ± 0.74 nm. The zeta potential was -25.6 ± 0.404 mV; 22.4 ± 1.106 mV and - 6.74 ± 0.609 mV for NLCo, NLCc, and NLCp, respectively. Differential scanning calorimetry (DSC) confirmed the formation of NLC whereas Fourier-transform infrared spectroscopy confirmed the FSE encapsulation into particles. All formulations showcased relatively high drug loading (>86.74 mcg FSE/mg solid lipid) and were characterized by prolonged and controlled release that followed Peppas-Sahlin in vitro release kinetic model. Protein adsorption studies revealed the lowest adsorption of the proteins onto NLCp (43.53 ± 0.07%) and highest protein adsorption onto NLCc (55.97 ± 0.75%) surface. The modified ORAC assay demonstrated higher antioxidative activity for NLCo (95.31 ± 1.86%) and NLCc (97.76 ± 4.00%) as compared to FSE (90.30 ± 1.53%). Results obtained from cell cultures tests pointed to the potential of prepared NLCs for FSE brain targeting and controlled release.
Collapse
Affiliation(s)
- Elena Markova
- Institute of Pharmaceutical Technology, Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Skopje, North Macedonia
| | - Lea Taneska
- Institute of Pharmaceutical Technology, Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Skopje, North Macedonia
| | - Monika Kostovska
- Institute of Pharmaceutical Technology, Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Skopje, North Macedonia
| | - Dushko Shalabalija
- Institute of Pharmaceutical Technology, Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Skopje, North Macedonia
| | - Ljubica Mihailova
- Institute of Pharmaceutical Technology, Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Skopje, North Macedonia
| | - Marija Glavas Dodov
- Institute of Pharmaceutical Technology, Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Skopje, North Macedonia
| | - Petre Makreski
- Institute of Chemistry, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University in Skopje, Skopje, North Macedonia
| | - Nikola Geskovski
- Institute of Pharmaceutical Technology, Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Skopje, North Macedonia
| | - Marija Petrushevska
- Institute of Pharmacology, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, Skopje, North Macedonia
| | - Arben N Taravari
- University Clinic for Neurology, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, Skopje, North Macedonia
| | - Maja Simonoska Crcarevska
- Institute of Pharmaceutical Technology, Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Skopje, North Macedonia
| |
Collapse
|
31
|
Suffian IFBM, Al-Jamal KT. Bioengineering of virus-like particles as dynamic nanocarriers for in vivo delivery and targeting to solid tumours. Adv Drug Deliv Rev 2022; 180:114030. [PMID: 34736988 DOI: 10.1016/j.addr.2021.114030] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 09/16/2021] [Accepted: 10/27/2021] [Indexed: 12/12/2022]
Abstract
Virus-like particles (VLPs) are known as self-assembled, non-replicative and non-infectious protein particles, which imitate the formation and structure of original wild type viruses, however, lack the viral genome and/or their fragments. The capacity of VLPs to encompass small molecules like nucleic acids and others has made them as novel vessels of nanocarriers for drug delivery applications. In addition, VLPs surface have the capacity to achieve variation of the surface display via several modification strategies including genetic modification, chemical modification, and non-covalent modification. Among the VLPs nanocarriers, Hepatitis B virus core (HBc) particles have been the most encouraging candidate. HBc particles are hollow nanoparticles in the range of 30-34 nm in diameter and 7 nm thick envelopes, consisting of 180 or 240 copies of identical polypeptide monomer. They also employ a distinctive position among the VLPs carriers due to the high-level synthesis, which serves as a strong protective capsid shell and efficient self-assembly properties. This review highlights on the bioengineering of HBc particles as dynamic nanocarriers for in vivo delivery and specific targeting to solid tumours.
Collapse
Affiliation(s)
- Izzat F B M Suffian
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia (Kuantan Campus), Jalan Sultan Ahmad Shah, Bandar Indera Mahkota, 25200 Kuantan, Pahang, Malaysia.
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
32
|
Habibzadeh Mashatooki M, Ghalami-Choobar B. Improved drug delivery and competitive adsorption of paclitaxel and mitomycin C anticancer drugs on the Boron-nitride nanoparticles: A molecular dynamics insight. Phys Chem Chem Phys 2022; 24:6639-6654. [DOI: 10.1039/d1cp04006e] [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 competitive aggregated adsorption and molecular interactions between paclitaxel (PX) and mitomycin C (MMC) molecules on the surface of boron nitride nanosheet (BNNS) was investigated using molecular dynamics method. BNNS...
Collapse
|
33
|
Raza F, Zafar H, Khan MW, Ullah A, Khan AU, Baseer A, Fareed R, Sohail M. Recent advances in the targeted delivery of paclitaxel nanomedicine for cancer therapy. MATERIALS ADVANCES 2022; 3:2268-2290. [DOI: 10.1039/d1ma00961c] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cancer cases have reached an all-time high in the current era.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | | | - Aftab Ullah
- Department of Pharmacy, Shantou University Medical College, Shantou, 515041, P. R. China
| | | | - Abdul Baseer
- Department of Pharmacy, Abasyn University, Peshawar, Pakistan
| | - Rameesha Fareed
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Islamabad, Pakistan
| | - Muhammad Sohail
- School of Pharmacy, Yantai University, Shandong, 264005, China
| |
Collapse
|
34
|
Duan R, Qi M. Amphiphilic triblock copolymer as the gas chromatographic stationary phase with high-resolution performance towards a wide range of isomers and the components of lemon essential oil. J Chromatogr A 2021; 1658:462611. [PMID: 34666270 DOI: 10.1016/j.chroma.2021.462611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 02/02/2023]
Abstract
This work presents the investigation of using the amphiphilic triblock copolymer composed of poly(ethylene oxide)(PEO)-poly(propylene oxide) (PPO)-poly(ethylene oxide) (PEO) (denoted as EPE) as the stationary phase for gas chromatographic (GC) analyses. The EPE capillary column exhibited moderate polarity and column efficiency of 4348 plates/m determined by naphthalene at 120 °C (k = 11.52). Different from the PEG and polysiloxane homopolymers, it showed high-resolution performance towards a wide range of aliphatic and aromatic isomers in terms of polarity and acid-base properties. Particularly, the EPE column displayed distinct advantages for separating the critical isomers of alkanes, anilines and phenols and the components of the lemon essential oil over the commercial PEG and polysiloxane columns. In addition, the EPE column exhibited excellent separation repeatability and reproducibility with the relative standard deviation (RSD) values in the range of 0.03% - 0.08% for run-to-run, 0.14% - 0.61% for day-to-day and 3.1% - 4.0% for column-to-column, respectively. Moreover, the EPE column was investigated in terms of thermal stability, the minimum allowable operating temperature (MiAOT) and sample loadability. Its application to GC-MS analysis of the essential oil demonstrated its feasibility for practical analyses. This work demonstrates the promising future of triblock copolymers as a new class of selective stationary phases for GC analyses, which is barely reported up to date. The findings of this work is of important value for fundamental researches and practical applications.
Collapse
Affiliation(s)
- Ruijuan Duan
- Key Laboratory of Cluster Science, Ministry of Education of China, Beijing Key Laboratory of Photoelectronic/Electrophotonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Meiling Qi
- Key Laboratory of Cluster Science, Ministry of Education of China, Beijing Key Laboratory of Photoelectronic/Electrophotonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
35
|
Marsili L, Dal Bo M, Berti F, Toffoli G. Thermoresponsive Chitosan-Grafted-Poly( N-vinylcaprolactam) Microgels via Ionotropic Gelation for Oncological Applications. Pharmaceutics 2021; 13:1654. [PMID: 34683947 PMCID: PMC8539247 DOI: 10.3390/pharmaceutics13101654] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 12/19/2022] Open
Abstract
Microgels can be considered soft, porous and deformable particles with an internal gel structure swollen by a solvent and an average size between 100 and 1000 nm. Due to their biocompatibility, colloidal stability, their unique dynamicity and the permeability of their architecture, they are emerging as important candidates for drug delivery systems, sensing and biocatalysis. In clinical applications, the research on responsive microgels is aimed at the development of "smart" delivery systems that undergo a critical change in conformation and size in reaction to a change in environmental conditions (temperature, magnetic fields, pH, concentration gradient). Recent achievements in biodegradable polymer fabrication have resulted in new appealing strategies, including the combination of synthetic and natural-origin polymers with inorganic nanoparticles, as well as the possibility of controlling drug release remotely. In this review, we provide a literature review on the use of dual and multi-responsive chitosan-grafted-poly-(N-vinylcaprolactam) (CP) microgels in drug delivery and oncological applications.
Collapse
Affiliation(s)
- Lorenzo Marsili
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, 34127 Trieste, Italy;
- Experimental and Clinical Pharmacology Unit, CRO National Cancer Institute IRCCS, Via Franco Gallini 2, 33081 Aviano, Italy; (M.D.B.); (G.T.)
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, CRO National Cancer Institute IRCCS, Via Franco Gallini 2, 33081 Aviano, Italy; (M.D.B.); (G.T.)
| | - Federico Berti
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, 34127 Trieste, Italy;
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, CRO National Cancer Institute IRCCS, Via Franco Gallini 2, 33081 Aviano, Italy; (M.D.B.); (G.T.)
| |
Collapse
|
36
|
Sa-nguanmoo N, Namdee K, Khongkow M, Ruktanonchai U, Zhao Y, Liang XJ. Review: Development of SARS-CoV-2 immuno-enhanced COVID-19 vaccines with nano-platform. NANO RESEARCH 2021; 15:2196-2225. [PMID: 34659650 PMCID: PMC8501370 DOI: 10.1007/s12274-021-3832-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 05/04/2023]
Abstract
Vaccination is the most effective way to prevent coronavirus disease 2019 (COVID-19). Vaccine development approaches consist of viral vector vaccines, DNA vaccine, RNA vaccine, live attenuated virus, and recombinant proteins, which elicit a specific immune response. The use of nanoparticles displaying antigen is one of the alternative approaches to conventional vaccines. This is due to the fact that nano-based vaccines are stable, able to target, form images, and offer an opportunity to enhance the immune responses. The diameters of ultrafine nanoparticles are in the range of 1-100 nm. The application of nanotechnology on vaccine design provides precise fabrication of nanomaterials with desirable properties and ability to eliminate undesirable features. To be successful, nanomaterials must be uptaken into the cell, especially into the target and able to modulate cellular functions at the subcellular levels. The advantages of nano-based vaccines are the ability to protect a cargo such as RNA, DNA, protein, or synthesis substance and have enhanced stability in a broad range of pH, ambient temperatures, and humidity for long-term storage. Moreover, nano-based vaccines can be engineered to overcome biological barriers such as nonspecific distribution in order to elicit functions in antigen presenting cells. In this review, we will summarize on the developing COVID-19 vaccine strategies and how the nanotechnology can enhance antigen presentation and strong immunogenicity using advanced technology in nanocarrier to deliver antigens. The discussion about their safe, effective, and affordable vaccines to immunize against COVID-19 will be highlighted.
Collapse
Affiliation(s)
- Nawamin Sa-nguanmoo
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Katawut Namdee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - Mattaka Khongkow
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - Uracha Ruktanonchai
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - YongXiang Zhao
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumour Theranostics and Therapy, Guangxi Medical University, Nanning, 530021 China
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
37
|
Gharaibeh L, Alshaer W, Wehaibi S, Al Buqain R, Alqudah DA, Al-Kadash A, Al-Azzawi H, Awidi A, Bustanji Y. Fabrication of aptamer-guided siRNA loaded lipopolyplexes for gene silencing of notch 1 in MDA-mb-231 triple negative breast cancer cell line. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102733] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
38
|
Mohsen AM. Cationic Polymeric Nanoparticles for Improved Ocular Delivery and Antimycotic Activity of Terconazole. J Pharm Sci 2021; 111:458-468. [PMID: 34547306 DOI: 10.1016/j.xphs.2021.09.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/11/2021] [Accepted: 09/11/2021] [Indexed: 12/14/2022]
Abstract
Terconazole (TCZ) is a broad-spectrum antifungal triazole that is particularly active against Candida species, but its poor water solubility hinders its ocular absorption and restricts its application. This study aims to fabricate TCZ-loaded cationic polymeric nanoparticles to enhance the ocular delivery and antimycotic activity of terconazole. TCZ-loaded nanoparticles were developed by nanoprecipitation method employing Eudragit RLPO®. They were characterized by entrapment efficiency (EE%), particle size (PS), zeta potential (ZP), morphology, Fourier transform infrared spectroscopy (FT-IR), and X-ray powder diffraction (XRPD). In-vitro antimycotic activity was evaluated by measuring zone of inhibition (ZI), minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC). The developed nanoparticles were spherical with moderate to high EE% (44.03-71.14%), a nanometric PS (49.41-78.72 nm), and a positively charged ZP (≥ +21.47). In-vitro release studies revealed sustained release of drug up to 24 h. FT-IR of TCZ-loaded nanoparticles revealed distinctive peaks for Eudragit RLPO® and Poloxamer-188, with disappearance of the TCZ characteristic peaks. XRPD revealed the amorphous state of TCZ within the polymer matrix. Mucoadhesive studies proved the mucoadhesive property of the developed TCZ nanoparticles. In-vitro antimycotic studies, assessed by ZI, MIC and MFC, revealed enhanced antimycotic activity of TCZ-loaded nanoparticles against Candida albicans, relative to plain TCZ. No irritation or abnormal changes to the rabbits' eyes for plain and medicated polymeric nanoparticles were found by the in-vivo Draize test. These findings reveal that the cationic polymeric nanoparticles can be regarded as a potential drug delivery system for enhancing the ocular antimycotic activity of TCZ.
Collapse
Affiliation(s)
- Amira Mohamed Mohsen
- Pharmaceutical Technology Department, National Research Centre, El-Buhouth St., Dokki, Cairo 12622, Egypt.
| |
Collapse
|
39
|
Large DE, Abdelmessih RG, Fink EA, Auguste DT. Liposome composition in drug delivery design, synthesis, characterization, and clinical application. Adv Drug Deliv Rev 2021; 176:113851. [PMID: 34224787 DOI: 10.1016/j.addr.2021.113851] [Citation(s) in RCA: 334] [Impact Index Per Article: 111.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
Liposomal drug delivery represents a highly adaptable therapeutic platform for treating a wide range of diseases. Natural and synthetic lipids, as well as surfactants, are commonly utilized in the synthesis of liposomal drug delivery vehicles. The molecular diversity in the composition of liposomes enables drug delivery with unique physiological functions, such as pH response, prolonged blood circulation, and reduced systemic toxicity. Herein, we discuss the impact of composition on liposome synthesis, function, and clinical utility.
Collapse
|
40
|
Recent Advances and Challenges in Nanodelivery Systems for Antimicrobial Peptides (AMPs). Antibiotics (Basel) 2021; 10:antibiotics10080990. [PMID: 34439040 PMCID: PMC8388958 DOI: 10.3390/antibiotics10080990] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/10/2021] [Accepted: 08/14/2021] [Indexed: 02/07/2023] Open
Abstract
Antimicrobial peptides (AMPs) can be used as alternative therapeutic agents to traditional antibiotics. These peptides have abundant natural template sources and can be isolated from animals, plants, and microorganisms. They are amphiphilic and mostly net positively charged, and they have a broad-spectrum inhibitory effect on bacteria, fungi, and viruses. AMPs possess significant rapid killing effects and do not interact with specific receptors on bacterial surfaces. As a result, drug resistance is rarely observed with treatments. AMPs, however, have some operational problems, such as a susceptibility to enzymatic (protease) degradation, toxicity in vivo, and unclear pharmacokinetics. However, nanodelivery systems loaded with AMPs provide a safe mechanism of packaging such peptides before they exert their antimicrobial actions, facilitate targeted delivery to the sites of infection, and control the release rate of peptides and reduce their toxic side effects. However, nanodelivery systems using AMPs are at an early stage of development and are still in the laboratory phase of development. There are also some challenges in incorporating AMPs into nanodelivery systems. Herein, an insight into the nanotechnology challenges in delivering AMPs, current advances, and remaining technological challenges are discussed in depth.
Collapse
|
41
|
Gupta N, Yadav V, Patel R. A brief review of the essential role of nanovehicles for improving the therapeutic efficacy of pharmacological agents against tumours. Curr Drug Deliv 2021; 19:301-316. [PMID: 34391379 DOI: 10.2174/1567201818666210813144105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/05/2021] [Accepted: 06/16/2021] [Indexed: 11/22/2022]
Abstract
Cancer is the leading cause of death globally. There are several differences between cancer cells and normal cells. From all the therapies, chemotherapy is the most prominent therapy to treat cancer. However, the conventional drug delivery that is used to deliver poorly aqueous soluble chemotherapeutic agents has several obstacles such as whole-body distribution, rapid excretion, degradation before reaching the infected site, side effects, etc. Nanoformulation of these aqueous insoluble agents is the emerging delivery system for targeted and increasing solubility. Among all the three methods (physical, chemical and biological) chemical and biological methods are mostly used for the synthesis of nanovehicles (NVs) of different sizes, shapes and dimensions. A passive targeting delivery system in which NVs supports the pharmacological agents (drugs/genes) is a good way for resolving the obstacles with a conventional delivery system. It enhances the therapeutic efficacy of pharmacological agents (drugs/genes). These NVs have several specific characters like small size, large surface area to volume ratio, surface functionalization, etc. However, this delivery is not able to deliver site-specific delivery of drugs. An active targeting delivery system in which pharmacological agents are loaded on NVs to attack directly on cancer cells and tissues is a superior way for delivering the pharmacological agents compared to a passive targeting delivery system. Various targeting ligands have been investigated and applied for targeting the delivery of drugs such as sugar, vitamin, antibodies, protein, peptides, etc. These targeted ligand supports to guide the NVs accumulated directly on the cancer cells with a higher level of cellular internalization compared to passive targeting and conventional delivery system.
Collapse
Affiliation(s)
- Nitin Gupta
- School of Nano Sciences, Central University of Gujarat, Gandhinagar- 382030, Gujarat, India
| | - Virendra Yadav
- Department of Microbiology, School of Life Sciences, Jaipur National University, Jaipur- 341503, Rajasthan, India
| | - Rakesh Patel
- Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Mehsana- 384012, Gujarat, India
| |
Collapse
|
42
|
Muljajew I, Huschke S, Ramoji A, Cseresnyés Z, Hoeppener S, Nischang I, Foo W, Popp J, Figge MT, Weber C, Bauer M, Schubert US, Press AT. Stealth Effect of Short Polyoxazolines in Graft Copolymers: Minor Changes of Backbone End Group Determine Liver Cell-Type Specificity. ACS NANO 2021; 15:12298-12313. [PMID: 34270899 DOI: 10.1021/acsnano.1c04213] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Dye-loaded micelles of 10 nm diameter formed from amphiphilic graft copolymers composed of a hydrophobic poly(methyl methacrylate) backbone and hydrophilic poly(2-ethyl-2-oxazoline) side chains with a degree of polymerization of 15 were investigated concerning their cellular interaction and uptake in vitro as well as their interaction with local and circulating cells of the reticuloendothelial system in the liver by intravital microscopy. Despite the high molar mass of the individual macromolecules (Mn ≈ 20 kg mol-1), backbone end group modification by attachment of a hydrophilic anionic fluorescent probe strongly affected the in vivo performance. To understand these effects, the end group was additionally modified by the attachment of four methacrylic acid repeating units. Although various micelles appeared similar in dynamic light scattering and cryo-transmission electron microscopy, changes in the micelles were evident from principal component analysis of the Raman spectra. Whereas an efficient stealth effect was found for micelles formed from polymers with anionically charged or thiol end groups, a hydrophobic end group altered the micelles' structure sufficiently to adapt cell-type specificity and stealth properties in the liver.
Collapse
Affiliation(s)
- Irina Muljajew
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Sophie Huschke
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Anuradha Ramoji
- Institute for Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Helmholtzweg 4, 07745 Jena, Germany
- Leibniz Institute of Photonic Technology (IPHT) Jena, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Zoltán Cseresnyés
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute Jena, Adolf-Reichwein-Strasse 23, 07745 Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Ivo Nischang
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Wanling Foo
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Jürgen Popp
- Institute for Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Helmholtzweg 4, 07745 Jena, Germany
- Leibniz Institute of Photonic Technology (IPHT) Jena, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Marc Thilo Figge
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute Jena, Adolf-Reichwein-Strasse 23, 07745 Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Neugasse 24, 07743 Jena, Germany
| | - Christine Weber
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Ulrich S Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Adrian T Press
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
- Medical Faculty, Friedrich Schiller University Jena, Bachstraße 18, 07743 Jena, Germany
| |
Collapse
|
43
|
|
44
|
Rehman TU, Bratlie KM. Improving selective targeting to cancer-associated fibroblasts by modifying liposomes with arginine based materials. J Drug Target 2021; 30:94-107. [PMID: 34116612 DOI: 10.1080/1061186x.2021.1941059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A library of arginine-like surface modifiers was tested to improve the targetability of DOPE:DOPC liposomes towards myofibroblasts in a tumour microenvironment. Liposomes were characterised using zeta potential and dynamic light scattering. Cell viability remained unchanged for all liposomes. Liposomes were encapsulated using doxorubicin (DOX) with an encapsulation efficiency >94%. The toxicity of DOX-loaded liposomes was calculated via half-maximal inhibitory concentration (IC50) for fibroblasts and myofibroblasts. These liposomes resulted in significantly lower IC50-values for myofibroblasts compared to fibroblasts, making them more toxic towards the myofibroblasts. Furthermore, a significant increase in cell internalisation was observed for myofibroblasts compared to fibroblasts, using fluorescein-loaded liposomes. Most importantly, a novel regression model was constructed to predict the IC50-values for different modifications using their physicochemical properties. Fourteen modifications (A-N) were used to train and validate this model; subsequently, this regression model predicted IC50-values for three new modifications (O, P and Q) for both fibroblasts and myofibroblasts. Predicted and measured IC50-values showed no significant difference for fibroblasts. For myofibroblasts, modification O showed no significant difference. This study demonstrates that the tested surface modifications can improve targeting to myofibroblasts in the presence of fibroblasts and hence are suitable drug delivery vehicles for myofibroblasts in a tumour microenvironment.
Collapse
Affiliation(s)
- Tanzeel Ur Rehman
- Department of Materials Science & Engineering, Iowa State University, Ames, IA, USA
| | - Kaitlin M Bratlie
- Department of Materials Science & Engineering, Iowa State University, Ames, IA, USA.,Department of Chemical & Biological Engineering, Iowa State University, Ames, IA, USA
| |
Collapse
|
45
|
Zhao Q, Zhang S, Wu F, Li D, Zhang X, Chen W, Xing B. Rationales Design von Nanogelen zur Überwindung biologischer Barrieren auf verschiedenen Verabreichungswegen. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.201911048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Qing Zhao
- Key Laboratory of Pollution Ecology and Environmental Engineering Institute of Applied Ecology Chinese Academy of Sciences Shenyang 110016 China
| | - Siyu Zhang
- Key Laboratory of Pollution Ecology and Environmental Engineering Institute of Applied Ecology Chinese Academy of Sciences Shenyang 110016 China
| | - Fengchang Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment Chinese Research Academy of Environmental Sciences Beijing 100012 China
| | - Dengyu Li
- Key Laboratory of Pollution Ecology and Environmental Engineering Institute of Applied Ecology Chinese Academy of Sciences Shenyang 110016 China
| | - Xuejiao Zhang
- Key Laboratory of Pollution Ecology and Environmental Engineering Institute of Applied Ecology Chinese Academy of Sciences Shenyang 110016 China
| | - Wei Chen
- Department of Pharmaceutical Engineering School of Engineering China Pharmaceutical University Nanjing 211198 China
| | - Baoshan Xing
- Stockbridge School of Agriculture University of Massachusetts Amherst MA 01003 USA
| |
Collapse
|
46
|
Sun Y, Chen D, Zhao Y, Zhou K, Zhang B, Wang H, Xie S. Exploitation of nanocrystal suspension as an effective oral formulation for oxfendazole. Drug Deliv Transl Res 2021; 12:1219-1229. [PMID: 34148210 DOI: 10.1007/s13346-021-01012-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
An oxfendazole (OFZ) nanocrystal suspension was prepared by acid-base neutralization and crystallization combined with ultrasonic dispersion to overcome the challenge of its poor oral bioavailability. The nanosuspensions were screened and optimized by single-factor experiments and an orthogonal design using size and appearance as indices. The morphology (differential scanning calorimetry (DSC), X-ray powder diffraction (XRPD)) properties and pharmacokinetics of the best formulation were further developed. The results showed that the best cosolvent and stabilizer were malic acid and hydrogenated castor oil polyoxyethylene ether (HEL-40), respectively. Scanning electron microscopy demonstrated that the oxfendazole nanocrystals are irregular sheets with relative uniformity. The prepared nanocrystals have an average particle diameter of 431 ± 18 nm, a polydispersity index (PDI) of 0.376 ± 0.128, a zeta potential of 2.30 ± 0.44 mV, and a sedimentation coefficient of 0.993. The equilibrium solubility of nanocrystals in different solvents was significantly improved by 2.02-109.99-fold compared to OFZ crude. In 0.5% SDS-PBS (pH 2) and 0.5% SDS-PBS (pH 8) solution, oxfendazole nanocrystals were completely released within 5 min, while the OFZ crude only released 60.26% and 28.31%, respectively. The pharmacokinetics showed that the Cmax, Tmax, and AUC0-∞ of OFZ nanosuspension and OFZ granules in rats after oral dosage at 50 mg/kg were 4.23 and 13.63 μg/mL, 2.04 and 1.67 h, and 111.36 and 295.80 μg*h/mL, respectively. The relative bioavailability of the oxfendazole nanosuspension was 265.61% compared to the OFZ granules. These results showed that the nanosuspension might be a promising oral formulation for the hardly soluble OFZ.
Collapse
Affiliation(s)
- Yuzhu Sun
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, 430070, Hubei, China
| | - Dongmei Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, 430070, Hubei, China.,MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Ying Zhao
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, 430070, Hubei, China
| | - Kaixiang Zhou
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, 430070, Hubei, China
| | - Bao Zhang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, 430070, Hubei, China
| | - Haiting Wang
- Qilu Animal Health Products Co. Ltd. No, 243 Gongye North Road, Shandong, 250100, Jinan, People's Republic of China
| | - Shuyu Xie
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, 430070, Hubei, China.
| |
Collapse
|
47
|
Martin JD, Miyazaki T, Cabral H. Remodeling tumor microenvironment with nanomedicines. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1730. [PMID: 34124849 DOI: 10.1002/wnan.1730] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/17/2022]
Abstract
The tumor microenvironment (TME) has been recognized as a major contributor to cancer malignancy and therapeutic resistance. Thus, strategies directed to re-engineer the TME are emerging as promising approaches for improving the efficacy of antitumor therapies by enhancing tumor perfusion and drug delivery, as well as alleviating the immunosuppressive TME. In this regard, nanomedicine has shown great potential for developing effective treatments capable of re-modeling the TME by controlling drug action in a spatiotemporal manner and allowing long-lasting modulatory effects on the TME. Herein, we review recent progress on TME re-engineering by using nanomedicine, particularly focusing on formulations controlling TME characteristics through targeted interaction with cellular components of the TME. Importantly, the TME should be re-engineering to a quiescent phenotype rather than be destroyed. Finally, immediate challenges and future perspectives of TME-re-engineering nanomedicines are discussed, anticipating further innovation in this growing field. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
| | - Takuya Miyazaki
- Kanagawa Institute of Industrial Science and Technology, Ebina, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
48
|
Hertig JB, Shah VP, Flühmann B, Mühlebach S, Stemer G, Surugue J, Moss R, Di Francesco T. Tackling the challenges of nanomedicines: are we ready? Am J Health Syst Pharm 2021; 78:1047-1056. [PMID: 33599767 PMCID: PMC7929390 DOI: 10.1093/ajhp/zxab048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE This review provides an overview of the proceedings of the symposium "Tackling the Challenges of Nanomedicines: Are We Ready?" organized by the International Pharmaceutical Federation (FIP) Hospital Pharmacy Section and Non-Biological Complex Drugs (NBCDs) Working Group at the 2019 FIP World Congress of Pharmacy and Pharmaceutical Sciences. Debate centered on reasons underlying the current complex regulatory landscape for nanomedicines and their follow-on products (referred to as nanosimilars) and the pivotal role of hospital pharmacists in selecting, handling, and guiding usage of nanomedicines and nanosimilars. SUMMARY The evaluation and use of nanomedicines are recognized among scientific, pharmaceutical, and regulatory bodies as complex. Interchangeability and substitutability of nanomedicines and nanosimilars are confounded by a lack of pharmaceutical and pharmacological equivalence, reflecting the inherent complex nature of these drug products and manufacturing processes. Consequences include implications for clinical safety and efficacy and, ultimately, comparability. Local regulatory approvals of some nanomedicines have occurred, but there is no standard to ensure streamlined evaluation and use of consistent measures of therapeutic equivalence of reference products and their nanosimilars. Hospital pharmacists are expected to be experts in the selection, handling, and substitution of nanomedicines and familiarize themselves with the limitations of current methods of assessing pharmaceutical and clinical equivalence of nanosimilars in order to ensure informed formulary decision-making and eventual patient benefit. CONCLUSION Supportive guidance for pharmacists focusing on the substitutability and/or interchangeability of nanomedicines and their nanosimilars is needed. Current FIP guidance for pharmacists on therapeutic interchange and substitution should be extended to include nanomedicines and nanosimilars.
Collapse
Affiliation(s)
- John B Hertig
- Department of Pharmacy Practice, Butler University College of Pharmacy and Health Sciences, Indianapolis, IN, USA
| | | | | | - Stefan Mühlebach
- Division of Clinical Pharmacy & Epidemiology and Hospital Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Gunar Stemer
- Pharmacy Department, Vienna General Hospital–Medical University Campus, Vienna, Austria
| | - Jacqueline Surugue
- Hospital Pharmacy Department, Georges Renon General Hospital, Niort, France
| | - Rob Moss
- Hospital Pharmacy Section, International Pharmaceutical Federation, The Hague, Netherlands
| | | |
Collapse
|
49
|
Systemic metastasis-targeted nanotherapeutic reinforces tumor surgical resection and chemotherapy. Nat Commun 2021; 12:3187. [PMID: 34045459 PMCID: PMC8160269 DOI: 10.1038/s41467-021-23466-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 04/21/2021] [Indexed: 01/26/2023] Open
Abstract
Failure of conventional clinical therapies such as tumor resection and chemotherapy are mainly due to the ineffective control of tumor metastasis. Metastasis consists of three steps: (i) tumor cells extravasate from the primary sites into the circulation system via epithelial-mesenchymal transition (EMT), (ii) the circulating tumor cells (CTCs) form “micro-thrombi” with platelets to evade the immune surveillance in circulation, and (iii) the CTCs colonize in the pre-metastatic niche. Here, we design a systemic metastasis-targeted nanotherapeutic (H@CaPP) composed of an anti-inflammatory agent, piceatannol, and an anti-thrombotic agent, low molecular weight heparin, to hinder the multiple steps of tumor metastasis. H@CaPP is found efficiently impeded EMT, inhibited the formation of “micro-thrombi”, and prevented the development of pre-metastatic niche. When combined with surgical resection or chemotherapy, H@CaPP efficiently inhibits tumor metastasis and prolonged overall survival of tumor-bearing mice. Collectively, we provide a simple and effective systemic metastasis-targeted nanotherapeutic for combating tumor metastasis. Failure of conventional clinical therapies such as tumor resection and chemotherapy are mainly due to the ineffective control of tumor metastasis. Here, the authors show that a systemic metastasis-targeted nanotherapeutic may offer a powerful adjunct therapy for suppressing tumor metastasis.
Collapse
|
50
|
Insight into drug encapsulation in polymeric nanoparticles using microfluidic nanoprecipitation. Chem Eng Sci 2021. [DOI: 10.1016/j.ces.2021.116468] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|