1
|
Nair SR, Nihad M, Shenoy P S, Gupta S, Bose B. Unveiling the effects of micro and nano plastics in embryonic development. Toxicol Rep 2025; 14:101954. [PMID: 40104046 PMCID: PMC11914762 DOI: 10.1016/j.toxrep.2025.101954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 03/20/2025] Open
Abstract
The improper disposal and degradation of plastics causes the formation and spread of micro and nano-sized plastic particles in the ecosystem. The widespread presence of these micro and nanoplastics leads to their accumulation in the biotic and abiotic components of the environment, thereby affecting the cellular and metabolic functions of organisms. Despite being classified as xenobiotic agents, information about their sources and exposure related to reproductive health is limited. Micro and nano plastic exposure during early developmental stages can cause abnormal embryonic development. It can trigger neurotoxicity and inflammatory responses as well in the developing embryo. In embryonic development, a comprehensive study of their role in pluripotency, gastrulation, and multi-differentiation potential is scarce. Due to ethical concerns associated with the direct use of human embryos, pluripotent cells and its 3D in vitro models (with cell lines) are an alternative source for effective research. Thus, the 3D Embryoid body (EB) model provides a platform for conducting embryotoxicity and multi-differentiation potential research. Pluripotent stem cells such as embryonic and induced pluripotent stem cells derived embryoid bodies (EBs) serve as a robust 3D in vitro model that mimics characteristics similar to that of human embryos. Thus, the 3D EB model provides a platform for conducting embryotoxicity and multi-differentiation potential research. Accordingly, this review discusses the significance of 3D in vitro models in conducting effective embryotoxicity research. Further, we also evaluated the possible sources/routes of microplastic generation and analyzed their surface chemistry and cytotoxic effects reported till date.
Collapse
Affiliation(s)
- Sanjay R Nair
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka 575018, India
| | - Muhammad Nihad
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka 575018, India
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka 575018, India
| | - Sebanti Gupta
- Division of Data Analytics, Bioinformatics and Structural Biology, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka 575018, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka 575018, India
| |
Collapse
|
2
|
Rizwan A, Rehman U, Gupta G, Alsayari A, Wahab S, Kesharwani P. Polyglutamic acid in cancer nanomedicine: Advances in multifunctional delivery platforms. Int J Pharm 2025; 676:125623. [PMID: 40254191 DOI: 10.1016/j.ijpharm.2025.125623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Polyglutamic acid (PGA)-coated nanoparticles have emerged as a significant advancement in cancer nanomedicine due to their biocompatibility, biodegradability, and versatility. PGA enhances the stability and bioavailability of therapeutic agents, enabling controlled and sustained drug release with reduced systemic toxicity. Stimuli-responsive modifications to PGA allow for precise drug delivery tailored to the tumor microenvironment, improving specificity and therapeutic outcomes. PGA's potential extends to gene delivery, where it facilitates safe and efficient transfection, addressing critical challenges such as multidrug resistance. Additionally, PGA-coated nanoparticles play a pivotal role in theranostic, integrating diagnostic and therapeutic capabilities within a single platform for real-time monitoring and treatment optimization. These nanoparticles have demonstrated enhanced efficacy in chemotherapy, immunotherapy, and combination regimens, tackling persistent issues like poor tumor penetration and non-specific drug distribution. Advancements in stimuli-responsive designs, ligand functionalization, and payload customization highlight the adaptability of PGA-based platforms for precision oncology. However, challenges such as scalability, stability under physiological conditions, and regulatory compliance remain key obstacles to clinical translation. This review explores the design, development, and applications of PGA-coated nanoparticles, emphasizing their potential to transform cancer treatment through safer, more effective, and personalized therapeutic approaches.
Collapse
Affiliation(s)
- Asfi Rizwan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Urushi Rehman
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Garima Gupta
- Graphic Era Hill University, Dehradun 248002, India; School of Allied Medical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia; Health and Medical Research Centre, King Khalid University, Abha 61421, Saudi Arabia
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| |
Collapse
|
3
|
Maiocchi S, Burnham EE, Cartaya A, Lisi V, Buechler N, Pollard R, Babaki D, Bergmeier W, Pinkerton NM, Bahnson EM. Development of DNase-1 Loaded Polymeric Nanoparticles Synthesized by Inverse Flash Nanoprecipitation for Neutrophil-Mediated Drug Delivery to In Vitro Thrombi. Adv Healthc Mater 2025:e2404584. [PMID: 40341904 DOI: 10.1002/adhm.202404584] [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: 11/17/2024] [Revised: 03/31/2025] [Indexed: 05/11/2025]
Abstract
Activated neutrophils release Neutrophil Extracellular Traps (NETs), comprising decondensed chromatin, peroxidases, and serine proteases, which aid in host defense but are also implicated in thrombosis and resistance to thrombolysis. Recombinant DNase 1, which degrades NETs, may aid in thrombus dissolution synergistically with fibrinolytics. However, its short half-life and susceptibility to plasma proteases limit its therapeutic applicability. To address these limitations, DNase1 is encapsulated into polymeric nanoparticles (DNPs) using inverse Flash Nanoprecipitation (iFNP), a scalable nanoparticle synthesis technique. Previously only used with model proteins, the study demonstrates for the first time the feasibility of extending iFNP to the encapsulation of therapeutic proteins. Conditions that promote DNase1 solubility, preserve activity, and demonstrate release resulting in ex vivo NET degradation are detailed. Furthermore, the use of neutrophils, the source of NETs, as carriers for DNPs to enhance targeted delivery is investigated. These findings confirm that DNP-loaded neutrophils maintain key functionalities, including viability and oxidative burst, and associate with in vitro blood clots to deliver nanoparticles, and DNase1 protein. This study not only extends the feasibility of applying iFNP to encapsulate therapeutic proteins into polymeric nanoparticles, a promising alternative to lipid nanoparticles, but also contributes to the emerging literature on neutrophils as delivery vectors for nanocarriers.
Collapse
Affiliation(s)
- Sophie Maiocchi
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Erica E Burnham
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
| | - Ana Cartaya
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
- Light Microscopy Core Facility, Duke University, Durham, NC, 27710, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Veronica Lisi
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Nancy Buechler
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Rachel Pollard
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
| | - Danial Babaki
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Nathalie M Pinkerton
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
| | - Edward M Bahnson
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
4
|
Liu H, Li S, Yu X, Xu Q, Tang C, Yin C. Modulating the Protein Corona on Nanoparticles by Finely Tuning Cross-Linkers Improves Macrophage Targeting in Oral Small Interfering RNA Delivery. ACS NANO 2025; 19:16469-16487. [PMID: 40275505 DOI: 10.1021/acsnano.4c18033] [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: 04/26/2025]
Abstract
The protein corona (PC) plays an important role in regulating the in vivo fate of nanoparticles (NPs). Modulating the surface chemical properties of NPs to control PC formation provides an alternative impetus for the oral delivery of small interfering RNA (siRNA). Herein, using tripolyphosphate (TPP), hyaluronic acid, and poly-γ-glutamic acid as cross-linkers, three types of mannose-modified trimethyl chitosan-cysteine (MTC)-based NPs with distinct surface chemistries were prepared to encapsulate siRNA via ionic gelation. The MTC-based NPs that were cross-linked exclusively with TPP (MTC/TPP/siRNA NPs) exhibited greater thiol group accessibility on their surfaces, resulting in a stronger affinity for apolipoprotein (APO) B48 during translocation across intestinal epithelia. Moreover, intracellular transport of MTC/TPP/siRNA NPs via the endoplasmic reticulum and Golgi apparatus further increased adsorption of APOB48, a key component of chylomicrons, which follow a similar transport pathway. Benefiting from the elevated APOB48 levels within the PC, the orally delivered MTC/TPP/siRNA NPs showed higher uptake by hepatic macrophages and better therapeutic efficacy for acute liver injury. Our results elucidate the role of NP surface chemical characteristics and translocation mechanisms across intestinal epithelia in forming oral PC, providing valuable insights for designing NPs that achieve effective oral gene delivery by customizing PC formation in vivo.
Collapse
Affiliation(s)
- Hengqing Liu
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Shengqi Li
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xin Yu
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Qian Xu
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Cui Tang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
5
|
Xing Y, Zhou L, Chen Y, Zhang Q, Wu X, Gan Z, Wu K, Jiang D, Wei S, Chen H. A dual-peptides and specific promoter-modified nano gene delivery system for myocardial hypertrophy treatment. Int J Biol Macromol 2025; 311:143759. [PMID: 40319986 DOI: 10.1016/j.ijbiomac.2025.143759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/15/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Cardiac hypertrophy represents the heart's adaptive response to physiological or pathological stimuli, functioning to alleviate ventricular wall stress and preserve cardiac function and efficiency. However, pathological hypertrophy usually progresses to heart failure. Gene therapy, in contrast to conventional chemotherapeutic drugs, has the ability to impact cardiac hypertrophy directly, while the lack of adequate vectors limits its application. Gemini surfactants (GS) have been proven to be effective for transfection in vivo in earlier research. To diminish the natural liver targeting of GS nanoparticles, a dual-targeted myocardial biomimetic GS nanocomplex gene delivery system with functional peptides TAT and PCM modified and synergized with cardiac-specific promoter chicken cardiac troponin T promoter (cTnT) is designed in this study. Bioluminescence imaging reveals the utility of targeting hypertrophy myocardium, resulting in low localization in the liver upon systemic administration. Biochemical indicators, echocardiography, gross morphology and histology all indicate that GS-nanocomplexes attenuate ISO-induced cardiac hypertrophy. RNA sequencing results reflect different uptake pathways for different GS nanocomplexes, and the investigation of cellular uptake under various endocytosis inhibitors demonstrate that clathrin-mediated endocytosis (CME) serves as the primary endocytic pathway for GS-pDNA uptake and caveolin-mediated endocytosis (CVME) serves as the primary endocytic pathway for GS-pDNA-TP-RBCM uptake. The endocytic pathway for nanocomplexes is confirmed by CAV-1 silencing. In summary, this research presents a dual myocardium-targeted biomimetic GS nanocomplex for gene delivery for cardiomyocytes. The in vivo and in vitro targeting ability, good biocompatibility and helpful therapeutic efficacy for cardiac hypertrophy are verified, and the uptake mechanism and intracellular transport pathway of GS nanocomplex are revealed. This innovative approach provides a promising therapeutic strategy for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Yangchen Xing
- College of Pharmacy, Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 400016, PR China
| | - Lu Zhou
- College of Pharmacy, Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 400016, PR China
| | - Yuxin Chen
- College of Pharmacy, Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 400016, PR China
| | - Qianyu Zhang
- College of Pharmacy, Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 400016, PR China
| | - Xianwei Wu
- College of Pharmacy, Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 400016, PR China
| | - Zongjie Gan
- College of Pharmacy, Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 400016, PR China
| | - Kexin Wu
- College of Pharmacy, Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 400016, PR China; College of Pharmaceutical Sciences, Zhejiang University, Yuhangtang Road, Xihu District, Hangzhou 310012, PR China
| | - Dongjun Jiang
- College of Pharmacy, Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 400016, PR China
| | - Shiqi Wei
- College of Pharmacy, Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 400016, PR China
| | - Huali Chen
- College of Pharmacy, Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 400016, PR China.
| |
Collapse
|
6
|
Li S, Zhong J, Ma Y, Yue C, Lv W, Ye G, Tian X, Li X, Huang Y, Du L. Influences of chain length and conformation of guanidinylated linear synthetic polypeptides on nuclear delivery of siRNA with potential application in transcriptional gene silencing. Int J Biol Macromol 2025; 308:142743. [PMID: 40180092 DOI: 10.1016/j.ijbiomac.2025.142743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 03/16/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Transcriptional gene silencing (TGS) mediated by siRNA holds promise for long-term silencing efficacy, determined by effective nuclear delivery of siRNA. However, non-viral vectors for this purpose are limited. In this work, we synthesized guanidinylated linear synthetic polypeptides (GLSPs) to explore how chain length and conformation impact siRNA delivery, especially nuclear entry. Results show that helical conformations, particularly right-handed ones, enhance siRNA loading and silencing efficiency compared to unordered structures. Increasing chain length also improves these aspects. The endocytic pathways of carrier/siRNA nanocomplexes (NCs) are mainly determined by conformation, regardless of length. Notably, some NCs derived from right-handed helices can enter cells via direct membrane penetration, like bioactivity of cell penetrating peptides (CPPs). When the peptide chain of GLSPs is long enough, all vectors can rapidly deliver siRNA to the nucleus, similar to bioactivity of nuclear localization signal peptides (NLSPs). Interestingly, helicity of the vectors aids endosomal escape of NCs. Moreover, delivering siRNA to the nucleus via GLSPs induces TGS associated with DNA promoter methylation or histone deacetylation. This study clarifies the structure-activity relationship of GLSPs in siRNA delivery, providing new insights for designing non-viral carriers for TGS.
Collapse
Affiliation(s)
- Suifei Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Junyang Zhong
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yunxiao Ma
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China
| | - Chengfeng Yue
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Wenxia Lv
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Guodong Ye
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiumei Tian
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China
| | - Xin Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Yugang Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Lingran Du
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
7
|
Li Z, Zheng G, Fang C, Mei J, Liang H, Yang L. Comparation of brain-targeting chitosan/sodium tripolyphosphate and ovalbumin/sodium carboxymethylcellulose nanoparticles on dihydromyricetin delivery and cognitive impairment in obesity-related Alzheimer's disease. Int J Biol Macromol 2025; 306:141517. [PMID: 40020826 DOI: 10.1016/j.ijbiomac.2025.141517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/02/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The brain-gut axis plays an important role in regulating cognitive ability in obesity-related Alzheimer's disease (AD). In this study, we aimed to investigate the correlation between the barrier penetration ability of the DMY nanodelivery system in vivo and the regulation of the gut-brain axis to alleviate cognitive impairment. Brain-targeted peptide (TGN: TGNYKALHPHNG) and DMY loaded chitosan (CS)/sodium tripolyphosphate (TPP) nanoparticles (TGN-DMY-CS/TPP-NPs) and ovalbumin (OVA)/sodium carboxymethylcellulose (CMC) nanoparticles (TGN-DMY-OVA/CMC-NPs) were prepared. TGN-DMY-CS/TPP-NPs demonstrated superior mucus penetration and BBB targeting ability compared to TGN-DMY-OVA/CMC-NPs, while the latter showed notable intestinal accumulation. TGN-DMY-CS/TPP-NPs treatment significantly increased the relative abundance of Alistipes and Rikenellaceae_RC9_gut_group, and TGN-DMY-OVA/CMC-NPs treatment obviously enhanced the relative abundance of Lactobacillus. Furthermore, both nanoparticles alleviated lipid metabolism disorder, oxidative stress, and inflammation in the liver, reduced oxidative stress and neuroinflammation in the brain, inhibited neuronal apoptosis, and enhanced mitochondrial biogenesis and synaptic plasticity in obesity-related AD mice. Despite different mucus penetration and biodistribution, their similar efficacy in improving obesity-related AD is attributed to the gut-brain bidirectional connection.
Collapse
Affiliation(s)
- Zhiwei Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China; Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Guodong Zheng
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Chaoping Fang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Jingtao Mei
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Hanji Liang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Licong Yang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China.
| |
Collapse
|
8
|
Laguerre K, Caillot M, Casas-Solvas JM, Vargas-Berenguel A, Urbinati G, Gref R. Gemcitabine-monophosphate encapsulation in "stealth" MOFs effectively inhibits tumor growth in vivo. Int J Pharm 2025; 675:125537. [PMID: 40174810 DOI: 10.1016/j.ijpharm.2025.125537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Gemcitabine is a commonly used chemotherapeutic agent that faces limitations due to rapid clearance, limited stability, and significant adverse effects. Recent efforts in its nanoencapsulation within Metal Organic Frameworks (MOFs) aim to overcome these barriers, offering a promising platform due to MOFs' high drug-loading capacity, green synthesis methods, and in vivo biocompatibility. This study further explores the use of dextran-alendronate-PEG (DAP) coating to enhance MOF colloidal stability while maintaining substantial drug loading in the MOFs. Here, we demonstrate that DAP is essential for preventing the degradation of MOFs and enhancing their colloidal stability, enabling them to evade the mononuclear phagocytic system. The results also confirm that a high gemcitabine drug loading is preserved in DAP-coated MOFs. Furthermore, encapsulation improves the efficacy of the drug, as evidenced by a threefold decrease in the IC50 in prostate cancer cells compared to the free drug. Additionally, the use of the chorioallantoic membrane (CAM) assay, an alternative in vivo preclinical model, confirms that only gemcitabine-loaded MOFs effectively inhibit tumor growth. This work provides valuable insights into a biocompatible drug delivery system that improves the stability and therapeutic outcomes of gemcitabine in biological environments.
Collapse
Affiliation(s)
- Killian Laguerre
- Université Paris-Saclay, UMR CNRS 8214, Institut des Sciences Moléculaires d'Orsay 91405 Orsay, France; Université Paris-Saclay, UMR CNRS 9018, Institut Gustave Roussy, 94805 Villejuif, France
| | - Mélody Caillot
- Université Rouen Normandie, Inserm U1245, Centre Henri Becquerel, 76000 Rouen, France
| | - Juan Manuel Casas-Solvas
- Department of Chemistry and Physics, University of Almería, Ctra. de Sacramento s/n, 04120 Almería, Spain
| | - Antonio Vargas-Berenguel
- Department of Chemistry and Physics, University of Almería, Ctra. de Sacramento s/n, 04120 Almería, Spain
| | - Giorgia Urbinati
- Université Paris-Saclay, UMR CNRS 9018, Institut Gustave Roussy, 94805 Villejuif, France; Université Paris Cité, CNRS, INSERM, UTCBS (https://utcbs.u-paris.fr/), 4 avenue de l'Observatoire, F-75006 Paris, France.
| | - Ruxandra Gref
- Université Paris-Saclay, UMR CNRS 8214, Institut des Sciences Moléculaires d'Orsay 91405 Orsay, France.
| |
Collapse
|
9
|
Soh WWM, Zhu J, Zhang Z, Mazlan MDM, Chin EWM, Cheah CH, Goh ELK, Li J. Supramolecular Polycations with a Linear-Star Architecture Containing Hydrophobic Poly[( R, S)-3-hydroxybutyrate]: Formation of DNA Micelleplexes Coated with Apolipoprotein E3 for Blood-Brain Barrier Penetrating Gene Delivery. Biomacromolecules 2025; 26:2157-2170. [PMID: 40052737 DOI: 10.1021/acs.biomac.4c01412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
A novel blood-brain barrier (BBB)-penetrating supramolecular gene delivery system was developed utilizing a host-guest block-building strategy to systematically screen and optimize various block compositions. Linear poly(ethylene glycol) (PEG) was coupled with hydrophobic poly[(R,S)-β-hydroxybutyrate] (PHB) blocks of varying lengths with an adamantyl (Ad) end, giving the PEG-PHB-Ad guest polymers, which were complexed with the cationic 4-arm star-shaped β-cyclodextrin-poly(2-dimethylaminoethyl methacrylate) (βCD-pDMAEMA) host polymer, resulting in the formation of linear-star pseudoblock PEG-PHB-Ad/βCD-pDMAEMA copolymers. These amphiphilic supramolecular copolymers were thoroughly characterized and assessed for the formation of DNA micelleplex nanoparticles as a gene delivery system. Through a rational selection process, an optimal host-guest configuration was identified, considering critical factors such as cytotoxicity, gene transfection efficiency, serum stability, cellular uptake, and hemolytic activity. The optimized host-guest copolymer was subsequently coated with the targeting protein apolipoprotein E3 (ApoE3), endowing it with BBB-penetrating capabilities, which was validated through an in vitro BBB transwell model.
Collapse
Affiliation(s)
- Wilson Wee Mia Soh
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jingling Zhu
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
- NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Zhongxing Zhang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
- NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Muhammad Danial Mohd Mazlan
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Eunice W M Chin
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Chee Hoe Cheah
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Eyleen L K Goh
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Jun Li
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
- NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| |
Collapse
|
10
|
Dar AI, Jain V, Rani A, Pulukuri AJ, Gonzalez JC, Dhull A, Sharma R, Sharma A. Silibinin-Conjugated Galactose Dendrimers for Targeted Treatment of Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20980-21000. [PMID: 40146860 DOI: 10.1021/acsami.5c04744] [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: 03/29/2025]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most prevalent and lethal forms of liver cancer, contributing significantly to global cancer-related mortality. Conventional treatments, including surgical resection, liver transplantation, and systemic therapies such as multikinase inhibitors and immune checkpoint inhibitors, often face limitations such as systemic toxicity and drug resistance, emphasizing the urgent need for more effective therapeutic strategies. In this study, we developed a galactose-functionalized dendrimer (Gal24) conjugated with a natural flavonoid, silibinin, (Gal24-Sil), for HCC therapy. In our previous study, we developed a Gal24 dendrimer to target hepatocytes in vivo. Here, we further demonstrated that the conjugation of silibinin to Gal24 dendrimer platform significantly enhanced its solubility and efficacy. In vitro studies demonstrated that Gal24-Sil conjugates significantly improved the anticancer efficacy of silibinin in HepG2 and Hep3B liver cancer cells. The conjugate induced an inflammatory response and reactive oxygen species (ROS) generation, triggering cellular apoptosis and necrosis. Furthermore, Gal24-Sil effectively reduced cell proliferation by promoting mitochondrial membrane potential (MMP) depolarization and inducing DNA damage. Our findings demonstrate the potential of Gal24-Sil as a promising nanoplatform for HCC therapy, offering enhanced therapeutic efficacy over free Silibinin. This study highlights the broader applicability of the Gal24 dendrimer platform for addressing various liver diseases.
Collapse
Affiliation(s)
- Aqib Iqbal Dar
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Vikrantvir Jain
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Anu Rani
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Anunay James Pulukuri
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Joan Castaneda Gonzalez
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Anubhav Dhull
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Rishi Sharma
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Anjali Sharma
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| |
Collapse
|
11
|
Sun S, Liu P, Hu J, Zhang L, Li M, Lu X, Zhao Y, Zhi T, Huang C. Auto-focus scanning surface plasmon resonance microscopy. OPTICS EXPRESS 2025; 33:16551-16561. [PMID: 40219538 DOI: 10.1364/oe.557410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/22/2025] [Indexed: 04/14/2025]
Abstract
Wide-field inspection, nano detection, and real-time observation are essential for investigating biomolecular interaction processes. Surface plasmon resonance microscopy (SPRM) is a label-free, real-time, and nano-imaging method that is widely employed for the dynamic detection of nanoscale biomolecules. The field of view (FOV) of SPRM is limited by the usage of high NA objectives, and a scanning SPRM is required to obtain a large FOV. However, during the scanning, the focus drift introduced by the mechanical vibrations blurs the imaging quality of SPRM, making the detection deviate from the true status. To this end, this paper presents the development of autofocus scanning SPRM (AFS-SPRM) that is capable of performing automated real-time focus drift correction during auto-scanning, thereby enabling high-quality SPRM imaging with large FOV. Only 80 ms is taken to process each defocusing event, and the ability to maintain focus has been improved by 30 times by comparison with SPRM. The AFS-SPRM was successfully employed to distinguish nanoparticles of different sizes and to observe the changes of macrophages in a culture medium containing nanoparticles. This investigation illustrates the superior imaging capabilities of AFS-SPRM and demonstrates its potential for observing interactions between biomolecules at the nanoscale.
Collapse
|
12
|
Leite JMDS, Oliveira ACDJ, Dourado D, Santana LMD, Medeiros TS, Nadvorny D, Silva MLR, Rolim-Neto PJ, Moreira DRM, Formiga FR, Soares MFDLR, Soares-Sobrinho JL. Rifampicin-loaded phthalated cashew gum nano-embedded microparticles intended for pulmonary administration. Int J Biol Macromol 2025; 303:140693. [PMID: 39914544 DOI: 10.1016/j.ijbiomac.2025.140693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 03/19/2025]
Abstract
Tuberculosis is a serious infectious disease commonly treated with rifampicin (RIF), which has low water solubility and high permeability. Polymeric nanoparticles (PNs) are used for controlled drug delivery to improve drug efficacy. However, PNs can be easily expelled via pulmonary administration. Nano-embedded microparticles (NEMs) are designed to bypass pulmonary barriers. Cashew gum, a versatile heteropolysaccharide, was modified into phthalated cashew gum (PCG), which targets alveolar macrophages, to increase hydrophobicity and improve drug encapsulation efficiency. In this study, the PCG was successfully obtained. Polymeric nanoparticle (PN)-PCG-RIF was fabricated, and its performance characteristics were investigated. PN-PCG-RIF exhibits mucoadhesive properties. An in vitro release study showed the release of 66.57 % of RIF after 6 h. An in vitro cytotoxicity study in A549 cells showed that PN-PCG-RIF is cytocompatible. The cellular uptake study demonstrated efficient cellular internalization in J774 macrophages, which was attributed to the PCG composition binding to the galactose-type lectin C receptor (MGL-2/CD301b). NEM-RIF was optimized by the Box Behnken designer with a particle size of 240.80 nm, PdI of 0.185, and redispersion index of 1.63. Scanning electron microscopy revealed NEMs-RIF in the form of spherical agglomerates. Collectively, RIF-NEMs were successfully developed from PN-PCG-RIF, having potential for the treatment of tuberculosis.
Collapse
Affiliation(s)
- Joandra Maísa da Silva Leite
- Quality Control Center for Medicines and Related Products, Federal University of Pernambuco, Recife, PE, Brazil; Program of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| | | | - Douglas Dourado
- Aggeu Magalhães Institute, Oswaldo Cruz Foundation (FIOCRUZ), 50670-420, Recife, PE, Brazil
| | - Lucas Marinho de Santana
- Quality Control Center for Medicines and Related Products, Federal University of Pernambuco, Recife, PE, Brazil
| | - Thayse Silva Medeiros
- Aggeu Magalhães Institute, Oswaldo Cruz Foundation (FIOCRUZ), 50670-420, Recife, PE, Brazil
| | - Daniela Nadvorny
- Quality Control Center for Medicines and Related Products, Federal University of Pernambuco, Recife, PE, Brazil
| | | | - Pedro José Rolim-Neto
- Program of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| | | | - Fábio Rocha Formiga
- Program of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil; Aggeu Magalhães Institute, Oswaldo Cruz Foundation (FIOCRUZ), 50670-420, Recife, PE, Brazil; Faculty of Medical Sciences, University of Pernambuco, 50100-130, Recife, PE, Brazil
| | - Mônica Felts de La Roca Soares
- Quality Control Center for Medicines and Related Products, Federal University of Pernambuco, Recife, PE, Brazil; Program of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| | - José Lamartine Soares-Sobrinho
- Quality Control Center for Medicines and Related Products, Federal University of Pernambuco, Recife, PE, Brazil; Program of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil.
| |
Collapse
|
13
|
Li J, Zhan S, Yang W, Zhang H, Ma X, Chen F, Li A, Tong P, Jiang F, Cao Z, Delahunty I, Wang J, Wu Y, Liu Z, Li Z, Teng Y, Xu L, Xie J. Radiation-induced ferroptosis via liposomal delivery of 7-Dehydrocholesterol. J Nanobiotechnology 2025; 23:249. [PMID: 40133959 PMCID: PMC11938788 DOI: 10.1186/s12951-025-03303-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/08/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Ferroptosis is an emerging cell death mechanism characterized by uncontrolled lipid peroxidation. However, selectively inducing ferroptosis in cancer cells remains a challenge. METHODS We explore an approach that enables ferroptosis induction through external radiation. The key component of this technology is 7-dehydrocholesterol (7DHC), a natural biosynthetic precursor of cholesterol. To facilitate delivery, we demonstrate that 7DHC, like cholesterol, can be incorporated into the lipid layer of liposomes. To enhance targeting, we also introduced NTSmut, a ligand for the neurotensin receptor 1 (NTSR1), which is overexpressed in multiple malignancies, into liposomes. RESULTS Under radiation, 7DHC reacts with radiation-induced reactive oxygen species (ROS), initiating a radical chain reaction with polyunsaturated fatty acids (PUFAs) in cell membranes. This process results in direct lipid peroxidation and subsequent ferroptotic cell death. In vivo studies demonstrate that NTSmut-conjugated, 7DHC-loaded liposomes (N-7DHC-lipos) effectively accumulate in tumors and significantly enhance the efficacy of radiation therapy. CONCLUSION While conventional radiosensitizers primarily target DNA and its repair mechanisms, our study introduces a strategy to enhance radiotherapy by specifically activating ferroptosis within the irradiated area, thereby minimizing systemic toxicity. Such a strategy of controlled activation of ferroptosis offers a favorable therapeutic index and potentially opens avenues for clinical application.
Collapse
Grants
- R01CA247769, R01CA257851 NCI NIH HHS
- R01CA247769, R01CA257851 NCI NIH HHS
- R01CA247769, R01CA257851 NCI NIH HHS
- R01CA247769, R01CA257851 NCI NIH HHS
- R01CA247769, R01CA257851 NCI NIH HHS
- R01CA247769, R01CA257851 NCI NIH HHS
- R01CA247769, R01CA257851 NCI NIH HHS
- R01CA247769, R01CA257851 NCI NIH HHS
- R01CA247769, R01CA257851 NCI NIH HHS
- R01CA247769, R01CA257851 NCI NIH HHS
- R01CA247769, R01CA257851 NCI NIH HHS
- R01CA247769, R01CA257851 NCI NIH HHS
- P30CA138292 Winship Cancer Institute
- P30CA138292 Winship Cancer Institute
- R01HD092659 NIEHS NIH HHS
- R01HD092659 NIEHS NIH HHS
- R01HD092659 NIEHS NIH HHS
- R01HD092659 NIEHS NIH HHS
- R01DE028351 Congressionally Directed Medical Research Programs
- R01DE028351 Congressionally Directed Medical Research Programs
- National Cancer Institute
- National Institute of Environmental Health Sciences
Collapse
Affiliation(s)
- Jianwen Li
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Shuyue Zhan
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Wei Yang
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - He Zhang
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xinrui Ma
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Fanghui Chen
- Department of Hematology and Medical Oncology & Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Amy Li
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, 98195, USA
| | - Pakteema Tong
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, 98195, USA
| | - Fangchao Jiang
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Zhengwei Cao
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Ian Delahunty
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Jiayi Wang
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Yufei Wu
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Zhi Liu
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Zibo Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology & Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Libin Xu
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, 98195, USA
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
14
|
Li Y, Sun H, Cao D, Guo Y, Wu D, Yang M, Wang H, Shao X, Li Y, Liang Y. Overcoming Biological Barriers in Cancer Therapy: Cell Membrane-Based Nanocarrier Strategies for Precision Delivery. Int J Nanomedicine 2025; 20:3113-3145. [PMID: 40098719 PMCID: PMC11913051 DOI: 10.2147/ijn.s497510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Given the unique capabilities of natural cell membranes, such as prolonged blood circulation and homotypic targeting, extensive research has been devoted to developing cell membrane-inspired nanocarriers for cancer therapy, while most focused on overcoming one or a few biological barriers. In fact, the journey of nanosystems from systemic circulation to tumor cells involves intricate processes, encompassing blood circulation, tissue accumulation, cancer cell targeting, endocytosis, endosomal escape, intracellular trafficking to target sites, and therapeutic action, all of which pose limitations to their clinical translation. This underscores the necessity of meticulously considering these biological barriers in the design of cell membrane-mimetic nanocarriers. In this review, we delineate the functions and applications of diverse types of cell membranes in nanocarrier systems. We elaborate on the biological hurdles encountered at each stage of the biomimetic nanoparticle's odyssey to the target, and comprehensively discuss the obstacles imposed by the tumor microenvironment for precise delivery. Subsequently, we systematically review contemporary cell membrane-based strategies aimed at overcoming these multi-level biological barriers, encompassing hybrid cell membrane (HCM) camouflage, tumor microenvironment remodeling, endosomal/lysosomal escape, multidrug resistance (MDR) reversal, optimization of nanoparticle physicochemical properties, and so on. Finally, we outline potential strategies to accelerate the development of cell membrane-inspired precision nanocarriers and discuss the challenges that must be addressed to enhance their clinical applicability. This review serves as a guide for refining the study of cell membrane-mimetic nanosystems in surmounting in vivo delivery barriers, thereby significantly contributing to advancing the development and application of cell membrane-based nanoparticles in cancer delivery.
Collapse
Affiliation(s)
- Yuping Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People's Republic of China
| | - Hongfang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Dianchao Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Yang Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Dongyang Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Menghao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Hongming Wang
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People's Republic of China
| | - Xiaowei Shao
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People's Republic of China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Yan Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| |
Collapse
|
15
|
Nie W, Zhang X, Kang P, Lan J, Li Z, Gu D, Zhou R, Ding Y, Zhang T. Exploring the influence of polysaccharide on gastrointestinal stability, drug release and formation mechanism of nanoparticles in Zhimu and Huangbai herb pair decoction. Sci Rep 2025; 15:8293. [PMID: 40064967 PMCID: PMC11894175 DOI: 10.1038/s41598-024-82130-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/03/2024] [Indexed: 03/14/2025] Open
Abstract
Natural polysaccharides with multiple biological activities from traditional Chinese medicine (TCM) can be self-assembled. Natural nanoparticles have been found to exist in TCM decoctions. However, the influence of polysaccharides in TCM decoctions on the formation mechanism, drug release and gastrointestinal stability of nanoparticles remains unclear. Therefore, using Zhimu and Huangbai herb pair decoction (ZBD) as an example, this study aimed to reveal the effects of polysaccharides on the formation, gastrointestinal stability and in vitro release of nanoparticles (N-ZBD) in ZBD. First, N-ZBD was successfully isolated by high-speed centrifugation with an average particle size of 225.9 nm, PDI of 0.53 and zeta potential of -13.00 mV, and N-ZBD is mainly composed of small active compounds and polysaccharides. Interestingly, gastrointestinal stability results showed that N-ZBD had good stability while and the nanoparticles after polysaccharide removal (RPN-ZBD) were degraded within 4 h, indicating that polysaccharide can protect the stability of nanoparticles in the gastrointestinal environment. Moreover, in vitro drug release results showed that compared with free drugs and RPN-ZBD, N-ZBD has obvious slow-release behavior in simulated gastric fluid and simulated intestinal fluid, suggesting that polysaccharides contribute to the slow release of N-ZBD. The critical micelle concentration (CMC) of polysaccharides was 0.63 mg/mL, which could self-assemble with small active compounds to form N-ZBD. In conclusion, polysaccharides of ZBD were essential for N-ZBD formation, and could protect the stability of N-ZBD in the gastrointestinal environment and contribute to its slow drug release.
Collapse
Affiliation(s)
- Wenlong Nie
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xinyu Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ping Kang
- Human Resources Office, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Donghao Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Rui Zhou
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
16
|
Cicek N, Cobandede Z, Adiguzel S, Yilmaz H, Culha M. Synergistic anti-cancer effects of piezoelectric hexagonal boron nitride nanocarriers for controlled doxorubicin release. Nanomedicine (Lond) 2025; 20:455-466. [PMID: 39887263 PMCID: PMC11875468 DOI: 10.1080/17435889.2025.2459055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
AIMS This study aims to develop a piezoelectric drug delivery system based on hexagonal boron nitride nanosheets (hBNs). MATERIALS AND METHODS hBNs were synthesized using the chemical vapor deposition (CVD) method and characterized through imaging and spectroscopic techniques. Their piezoelectric properties were evaluated to confirm their functionality. Subsequently, the potential of hBNs as nanocarriers was assessed through in vitro experiments with doxorubicin (Dox) as a model drug. RESULTS The piezoelectric hBNs were successfully synthesized and exhibited efficient loading and controlled release of Dox. In vitro experiments conducted on PC3 (human prostate cancer) and PNT1A (normal adult prostate epithelial) cell lines demonstrated that ultrasound (US)-induced Dox-loaded hBNs (hBN-Dox) significantly inhibited the proliferation of prostate cancer cells, achieving efficacy at a much lower Dox concentration compared to conventional methods. The system enhanced reactive oxygen species (ROS) generation, impaired cancer cell colony formation, and induced both early and late apoptosis. CONCLUSIONS These findings highlight the potential of piezoelectric hBNs as nanocarriers for efficient drug delivery, leveraging the synergistic effect of piezoelectricity-induced drug release and the degradation products of hBNs in biological media. Their ability to enhance drug efficacy while reducing the required dose holds promise for advanced cancer therapies.
Collapse
Affiliation(s)
- Nilay Cicek
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla Istanbul, Turkey
| | - Zehra Cobandede
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla Istanbul, Turkey
| | - Sevin Adiguzel
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla Istanbul, Turkey
| | - Hulya Yilmaz
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla Istanbul, Turkey
| | - Mustafa Culha
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla Istanbul, Turkey
- Chemistry and Biochemistry Department, College of Science and Mathematics, Augusta University, Augusta, GA, USA
| |
Collapse
|
17
|
Jha K, Jaishwal P, Yadav TP, Singh SP. Self-assembling of coiled-coil peptides into virus-like particles: Basic principles, properties, design, and applications with special focus on vaccine design and delivery. Biophys Chem 2025; 318:107375. [PMID: 39674128 DOI: 10.1016/j.bpc.2024.107375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Self-assembling peptide nanoparticles (SAPN) based delivery systems, including virus-like particles (VLP), have shown great potential for becoming prominent in next-generation vaccine and drug development. The VLP can mimic properties of natural viral capsid in terms of size (20-200 nm), geometry (i.e., icosahedral structures), and the ability to generate a robust immune response (with multivalent epitopes) through activation of innate and/or adaptive immune signals. In this regard, coiled-coil (CC) domains are suitable building blocks for designing VLP because of their programmable interaction specificity, affinity, and well-established sequence-to-structure relationships. Generally, two CC domains with different oligomeric states (trimer and pentamer) are fused to form a monomeric protein through a short, flexible spacer sequence. By using combinations of symmetry axes (2-, 3- and 5- folds) that are unique to the geometry of the desired protein cage, it is possible, in principle, to assemble well-defined protein cages like VLP. In this review, we have discussed the crystallographic rules and the basic principles involved in the design of CC-based VLP. It also explored the functions of numerous noncovalent interactions in generating stable VLP structures, which play a crucial role in improving the properties of vaccine immunogenicity, drug delivery, and 3D cell culturing.
Collapse
Affiliation(s)
- Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Thakur Prasad Yadav
- Department of Physics, Faculty of Science, University of Allahabad, Prayagraj 211002, India.
| | | |
Collapse
|
18
|
Bresinsky M, Goepferich A. Control of biomedical nanoparticle distribution and drug release in vivo by complex particle design strategies. Eur J Pharm Biopharm 2025; 208:114634. [PMID: 39826847 DOI: 10.1016/j.ejpb.2025.114634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
The utilization of targeted nanoparticles as a selective drug delivery system is a powerful tool to increase the amount of active substance reaching the target site. This can increase therapeutic efficacy while reducing adverse drug effects. However, nanoparticles face several challenges: upon injection, the immediate adhesion of plasma proteins may mask targeting ligands, thereby diminishing the target cell selectivity. In addition, opsonization can lead to premature clearance and the widespread presence of receptors or enzymes limits the accuracy of target cell recognition. Nanoparticles may also suffer from endosomal entrapment, and controlled drug release can be hindered by premature burst release or insufficient particle retention at the target site. Various strategies have been developed to address these adverse events, such as the implementation of switchable particle properties, regulating the composition of the formed protein corona, or using click-chemistry based targeting approaches. This has resulted in increasingly complex particle designs, raising the question of whether this development actually improves the therapeutic efficacy in vivo. This review provides an overview of the challenges in targeted drug delivery and explores potential solutions described in the literature. Subsequently, appropriate strategies for the development of nanoparticular drug delivery concepts are discussed.
Collapse
Affiliation(s)
- Melanie Bresinsky
- Department of Pharmaceutical Technology, University of Regensburg 93053 Regensburg, Bavaria, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg 93053 Regensburg, Bavaria, Germany.
| |
Collapse
|
19
|
Itagaki M, Kamei N. Clathrin functions in intracellular sorting during receptor-mediated endocytosis. FEBS Lett 2025. [PMID: 39973445 DOI: 10.1002/1873-3468.70016] [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: 11/10/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/21/2025]
Abstract
Clathrin is a key protein involved in receptor-mediated endocytosis (RME), which is also known as clathrin-mediated endocytosis (CME). Ligand/receptor binding facilitates clathrin clustering and cell surface invagination. We recently found that the typical CME ligand transferrin (Tf) can be internalized into cells without the clathrin heavy chain (CLTC). In this study, we clarified the unknown roles of clathrin during intracellular trafficking. Microscopic observation revealed that Tf accumulates in the deeper compartments of CLTC-knockdown HeLa cells. A colocalization analysis with endosome/lysosome markers showed that endocytosed Tf moves to early/late endosomes and then to lysosomes in CLTC-knockdown cells. This suggests that clathrin contributes to the recycling and exocytosis of endocytosed molecules.
Collapse
Affiliation(s)
- Mai Itagaki
- Laboratory of Drug Delivery Systems, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Japan
| | - Noriyasu Kamei
- Laboratory of Drug Delivery Systems, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Japan
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Japan
| |
Collapse
|
20
|
Goswami R, Nagaraj H, Cicek YA, Nasim N, Mirza SS, Hassan MA, Mhaske R, Saravanan DM, Noonan C, Pham E, Mager J, Rotello VM. Polymer-siRNA nanovectors for treating lung inflammation. J Control Release 2025; 378:1092-1102. [PMID: 39730067 PMCID: PMC11830555 DOI: 10.1016/j.jconrel.2024.12.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/07/2024] [Accepted: 12/21/2024] [Indexed: 12/29/2024]
Abstract
Uncontrolled inflammation is the driver of numerous lung diseases. Current treatments, including corticosteroids and bronchodilators, can be effective. However, they often come with notable side effects. siRNA is a promising therapeutic modality for immune regulation. However, effective delivery of siRNA is challenged by issues related to cellular uptake and localization within tissues. This study investigates a series of guanidinium-functionalized polymers (Cn-Guan) designed to explore the effects of amphiphilicity on siRNA complexation and efficiency in vitro and in vivo. Nine polymers with varying side chain lengths (C3, C5, C7) and molecular weights (17 kDa, 30 kDa, 65 kDa) were synthesized, forming polyplexes with siRNA. Characterization revealed that C7-Guan/si_scr polymers exhibited the smallest polyplex sizes and the tightest complexation with siRNA. In vitro studies showed that 65 kDa polymers had the highest gene knockdown efficiency, with C3 and C5-Guan/si_TNF-α achieving ∼70 % knockdown, while C7-Guan/si_TNF-α achieved ∼30 %. In vivo, C7-Guan/Cy5-siRNA demonstrated the highest lung accumulation, and all polymers showed ∼70 % TNF-α knockdown with a low siRNA dosage (0.14 mg/kg) in a murine lung inflammation model. C7-Guan polymers, despite lower in vitro efficiency, were quite effective in vivo, potentially due to enhanced serum stability. These findings demonstrate that Cn-Guan/siRNA polyplexes are effective and safe for attenuating pulmonary inflammation and provide important insights for the development of future siRNA delivery vectors for lung disease treatment.
Collapse
Affiliation(s)
- Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Yagiz Anil Cicek
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Nourina Nasim
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Sarah S Mirza
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, MA 01003, USA
| | - Muhammad Aamir Hassan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Rukmini Mhaske
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Deepthika M Saravanan
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Cedar Noonan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Edward Pham
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Jesse Mager
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, MA 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA.
| |
Collapse
|
21
|
Zhang W, Liu H, Zhu B, Li W, Han X, Fu J, Luo R, Wang H, Wang J. Advances in Cytosolic Delivery of Proteins: Approaches, Challenges, and Emerging Technologies. Chem Biodivers 2025:e202401713. [PMID: 39921680 DOI: 10.1002/cbdv.202401713] [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: 07/14/2024] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 02/10/2025]
Abstract
Although therapeutic proteins have achieved recognized clinical success, they are inherently membrane impermeable, which limits them to acting only on extracellular or membrane-associated targets. Developing an efficient protein delivery method will provide a unique opportunity for intracellular target-related therapeutic proteins. In this review article, we summarize the different pathways by which cells take up proteins. These pathways fall into two main categories: One in which proteins are transported directly across the cell membrane and the other through endocytosis. At the same time, important features to ensure successful delivery through these pathways are highlighted. We then provide a comprehensive overview of the latest developments in the transduction of covalent protein modifications, such as coupling cell-penetrating motifs and supercharging, as well as the use of nanocarriers to mediate protein transport, such as liposomes, polymers, and inorganic nanoparticles. Finally, we emphasize the existing challenges of cytoplasmic protein delivery and provide an outlook for future progress.
Collapse
Affiliation(s)
- Wenyan Zhang
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Huiling Liu
- Gansu Provincial Hospital, Lanzhou, Gansu, China
| | | | - Wen Li
- Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xue Han
- Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Jiaojiao Fu
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Renjie Luo
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Haiyan Wang
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jinxia Wang
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
22
|
Kuriyama M, Hirose H, Kawaguchi Y, Michibata J, Maekawa M, Futaki S. KCNN4 as a genomic determinant of cytosolic delivery by the attenuated cationic lytic peptide L17E. Mol Ther 2025; 33:595-614. [PMID: 39748507 PMCID: PMC11852704 DOI: 10.1016/j.ymthe.2024.12.050] [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: 07/27/2024] [Revised: 11/19/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025] Open
Abstract
The development of a cytosolic delivery strategy for biopharmaceuticals is one of the central issues in drug development. Knowledge of the mechanisms underlying these processes may also pave the way for the discovery of novel delivery systems. L17E is an attenuated cationic amphiphilic lytic (ACAL) peptide developed by our research group that shows promise for cytosolic antibody delivery. In this study, given the high efficacy of L17E in cytosolic delivery, we investigated the mechanism of action of L17E in detail. L17E was found to achieve cytosolic delivery predominantly by transient disruption of the plasma membrane without the need for endocytosis. Importantly, the cell-line selectivity studies of L17E revealed a strong correlation between the efficiency of L17E-mediated delivery and the expression level of KCNN4, the gene encoding the calcium-activated potassium channel KCa3.1. Genetic and pharmacological regulation of KCNN4 expression and KCa3.1 activity, respectively, correlate closely with the efficiency of L17E-mediated cytosolic delivery, suggesting the importance of membrane-potential regulation by extracellular Ca2+ influx. Therefore, the activity of the L17E is relevant to the calcium-activated potassium channel.
Collapse
Affiliation(s)
- Masashi Kuriyama
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Hisaaki Hirose
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.
| | - Yoshimasa Kawaguchi
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Junya Michibata
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Masashi Maekawa
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo 105-8512, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.
| |
Collapse
|
23
|
Shiomi K, Hayashi K, Ishii H, Kamei T, Shimanouchi T, Nakamura H, Ichikawa S. Phase-separated cationic giant unilamellar vesicles as templates for the polymerization of tetraethyl orthosilicate (TEOS). BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184403. [PMID: 39626823 DOI: 10.1016/j.bbamem.2024.184403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024]
Abstract
Unlike homogeneous liposomes, phase-separated liposomes have the potential to be attractive soft materials because they exhibit different properties for each phase. In this study, phase separation was observed when liposomes were prepared using 1,2-dioleoyloxy-3-trimethylammonium propane chloride (DOTAP), 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), and cholesterol. The pH of the DOTAP-rich phase was evaluated using a coumarin derivative, and measurements showed that DOTAP molecules accumulated hydroxyl ions (OH-) in the DOTAP-rich phase. Such accumulation of OH- was not observed when homogeneous DSPC liposomes were used. The difference in local concentration of OH- in each phase was applied to prepare hollow silica particles with large pores. The OH- promotes the polymerization of tetraethyl orthosilicate (TEOS). Therefore, TEOS polymerized preferentially in the DOTAP-rich phase, whereas a silica membrane barely formed in the DSPC-rich phase.
Collapse
Affiliation(s)
- Kohei Shiomi
- Master's Program in Agro-Bioresources Science and Technology, Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan; Department of Materials Science and Chemical Engineering, Faculty of Advanced Engineering, National Institute of Technology, Nara College, 22 Yata-cho, Yamatokoriyama, Nara 639-1080, Japan
| | - Keita Hayashi
- Department of Chemical Engineering, National Institute of Technology, Nara College, 22 Yata-cho, Yamatokoriyama, Nara 639-1080, Japan.
| | - Haruyuki Ishii
- Department of Sustainable Environmental Engineering, Graduate School of Sciences and Technology for Innovation, Yamaguchi University, 2-16-1, Tokiwadai, Ube 755-8611, Japan
| | - Toshiyuki Kamei
- Department of Chemical Engineering, National Institute of Technology, Nara College, 22 Yata-cho, Yamatokoriyama, Nara 639-1080, Japan
| | - Toshinori Shimanouchi
- Division of Environmental Science, Graduate School of Environmental and Life Science, Okayama University, Kita-ku, Okayama 700-8530, Japan
| | - Hidemi Nakamura
- Department of Chemical Engineering, National Institute of Technology, Nara College, 22 Yata-cho, Yamatokoriyama, Nara 639-1080, Japan
| | - Sosaku Ichikawa
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| |
Collapse
|
24
|
Wu F, An X, Li S, Qiu C, Zhu Y, Ye Z, Song S, Wang Y, Shen D, Di X, Yao Y, Zhu W, Jiang X, Shi X, Chen R, Kou L. Enhancing chemoimmunotherapy for colorectal cancer with paclitaxel and alantolactone via CD44-Targeted nanoparticles: A STAT3 signaling pathway modulation approach. Asian J Pharm Sci 2025; 20:100993. [PMID: 39917727 PMCID: PMC11795048 DOI: 10.1016/j.ajps.2024.100993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/28/2024] [Accepted: 10/22/2024] [Indexed: 02/09/2025] Open
Abstract
Chemoimmunotherapy has the potential to enhance chemotherapy and modulate the immunosuppressive tumor microenvironment by activating immunogenic cell death (ICD), making it a promising strategy for clinical application. Alantolactone (A) was found to augment the anticancer efficacy of paclitaxel (P) at a molar ratio of 1:0.5 (P:A) through induction of more potent ICD via modulation of STAT3 signaling pathways. Nano drug delivery systems can synergistically combine natural drugs with conventional chemotherapeutic agents, thereby enhancing multi-drug chemoimmunotherapy. To improve tumor targeting ability and bioavailability of hydrophobic drugs, an amphiphilic prodrug conjugate (HA-PTX) was chemically modified with paclitaxel (PTX) and hyaluronic acid (HA) as a backbone. Based on this concept, CD44-targeted nanodrugs (A@HAP NPs) were developed for co-delivery of A and P in colorectal cancer treatment, aiming to achieve synergistic toxicity-based chemo-immunotherapy. The uniform size and high drug loading capacity of A@HAP NPs facilitated their accumulation within tumors through enhanced permeability and retention effect as well as HA-mediated targeting, providing a solid foundation for subsequent synergistic therapy and immunoregulation. In vitro and in vivo studies demonstrated that A@HAP NPs exhibited potent cytotoxicity against tumor cells while also remodeling the immune-suppressive tumor microenvironment by promoting antigen presentation and inducing dendritic cell maturation, thus offering a novel approach for colorectal cancer chemoimmunotherapy.
Collapse
Affiliation(s)
- Fugen Wu
- Department of Pediatrics, Wenling Hospital of Wenzhou Medical University, Wenling 317500, China
| | - Xingsi An
- Department of Pediatrics, Wenling Hospital of Wenzhou Medical University, Wenling 317500, China
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shize Li
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Chenyu Qiu
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yixuan Zhu
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhanzheng Ye
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shengnan Song
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Yunzhi Wang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Dingchao Shen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xinyu Di
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yinsha Yao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Wanling Zhu
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Xinyu Jiang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Ruijie Chen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Longfa Kou
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
25
|
Phan QT, Rabanel J, Mekhjian D, Saber J, Garcia Ac A, Zhang H, Gibson VP, Zaouter C, Hardy P, Patten SA, Boffito D, Banquy X. Core-Shell Bottlebrush Polymers: Unmatched Delivery of Small Active Compounds Deep Into Tissues. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408616. [PMID: 39679753 PMCID: PMC11798360 DOI: 10.1002/smll.202408616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/09/2024] [Indexed: 12/17/2024]
Abstract
The chemical structure of a delivery nanovehicle plays a pivotal role in determining the efficiency of drug delivery within the body. Leveraging the unique architecture of bottlebrush (BB) polymers-characterized by variations in backbone length, grafting density, and self-assembly morphology-offers a novel approach to understanding the influence of structural properties on biological behavior. In this study, developed a drug delivery system based on core-shell BB polymers synthesized using a "grafting-from" strategy. Comprehensive characterization techniques, including nuclear magnetic resonance (NMR), gel permeation chromatography (GPC), and atomic force microscopy (AFM), employed to confirm the polymers' structure. The BB polymers evaluated as carriers for molecules with differing hydrophobicity profiles, namely Rhodamine B and Paclitaxel. These nanocarriers systematically assessed for drug loading efficiency and penetration capabilities, compared to conventional polymeric micelles (PM) formed from linear amphiphilic polymers. BB-based nanocarriers exhibited superior cellular uptake in both 2D and 3D cell culture models when compared to PM. Furthermore, analysis of drug distribution and particle penetration highlighted the profound influence of polymer morphology on biological interactions. These findings underscore the potential of unimolecular carriers with precisely defined structures as promising drug delivery platforms for a wide range of biomedical applications.
Collapse
Affiliation(s)
- Quoc Thang Phan
- Faculty of PharmacyUniversité de Montréal2940 Chemin de PolytechniqueMontréalQuébecH3T 1J4Canada
| | - Jean‐Michel Rabanel
- Faculty of PharmacyUniversité de Montréal2940 Chemin de PolytechniqueMontréalQuébecH3T 1J4Canada
- School of Pharmaceutical SciencesFaculty of MedicineUniversity of OttawaRoger Guindon Hall, 451 Smyth RdOttawaOntarioK1H 8M5Canada
| | - Dikran Mekhjian
- Faculty of PharmacyUniversité de Montréal2940 Chemin de PolytechniqueMontréalQuébecH3T 1J4Canada
| | - Justine Saber
- Faculty of PharmacyUniversité de Montréal2940 Chemin de PolytechniqueMontréalQuébecH3T 1J4Canada
| | - Araceli Garcia Ac
- Faculty of PharmacyUniversité de Montréal2940 Chemin de PolytechniqueMontréalQuébecH3T 1J4Canada
| | - Hu Zhang
- Faculty of PharmacyUniversité de Montréal2940 Chemin de PolytechniqueMontréalQuébecH3T 1J4Canada
| | - Victor Passos Gibson
- Department of Pharmacology and PhysiologyUniversité de MontréalMontréalQuébecH3T 1J4Canada
| | - Charlotte Zaouter
- INRS Centre Armand‐Frappier Santé Biotechnologie531, boul. des PrairiesQuébecCanadaH7V 1B7
| | - Pierre Hardy
- Department of Pharmacology and PhysiologyUniversité de MontréalMontréalQuébecH3T 1J4Canada
| | | | - Daria Boffito
- Department of Chemical EngineeringPolytechnique Montréal2500 Chemin de PolytechniqueMontréalQuébecH3C 3A7Canada
| | - Xavier Banquy
- Faculty of PharmacyUniversité de Montréal2940 Chemin de PolytechniqueMontréalQuébecH3T 1J4Canada
- Biomedical Engineering InstituteUniversité de Montréal2940 Chemin de PolytechniqueMontréalQuébecH3T 1J4Canada
- Chemistry DepartmentFaculty of Arts and SciencesUniversité de Montréal2940 Chemin de PolytechniqueMontréalQuébecH3T 1J4Canada
| |
Collapse
|
26
|
Sriram A, Ithape H, Singh PK. Deep-insights: Nanoengineered gel-based localized drug delivery for arthritis management. Asian J Pharm Sci 2025; 20:101012. [PMID: 39995751 PMCID: PMC11848107 DOI: 10.1016/j.ajps.2024.101012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 05/15/2024] [Accepted: 07/03/2024] [Indexed: 02/26/2025] Open
Abstract
Arthritis is an inflammatory joint disorder that progressively impairs function and diminishes quality of life. Conventional therapies often prove ineffective, as oral administration lacks specificity, resulting in off-target side effects like hepatotoxicity and GIT-related issues. Intravenous administration causes systemic side effects. The characteristic joint-localized symptoms such as pain, stiffness, and inflammation make the localized drug delivery suitable for managing arthritis. Topical/transdermal/intra-articular routes have become viable options for drug delivery in treating arthritis. However, challenges with those localized drug delivery routes include skin barrier and cartilage impermeability. Additionally, conventional intra-articular drug delivery also leads to rapid clearance of drugs from the synovial joint tissue. To circumvent these limitations, researchers have developed nanocarriers that enhance drug permeability through skin and cartilage, influencing localized action. Gel-based nanoengineered therapy employs a gel matrix to incorporate the drug-encapsulated nanocarriers. This approach combines the benefits of gels and nanocarriers to enhance therapeutic effects and improve patient compliance. This review emphasizes deep insights into drug delivery using diverse gel-based novel nanocarriers, exploring their various applications embedded in hyaluronic acid (biopolymer)-based gels, carbopol-based gels, and others. Furthermore, this review discusses the influence of nanocarrier pharmacokinetics on the localization and therapeutic manipulation of macrophages mediated by nanocarriers. The ELVIS (extravasation through leaky vasculature and inflammatory cell-mediated sequestration) effect associated with arthritis is advantageous in drug delivery. Simply put, the ELVIS effect refers to the extravasation of nanocarriers through leaky vasculatures, which finally results in the accumulation of nanocarriers in the joint cavity.
Collapse
Affiliation(s)
| | | | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Telangana 500037, India
| |
Collapse
|
27
|
Lee Y, Kang S, Thuy LT, Son M, Park JY, Ahn SB, Kang M, Oh J, Choi JS, Lee M. Exosome-membrane and polymer-based hybrid-complex for systemic delivery of plasmid DNA into brains for the treatment of glioblastoma. Asian J Pharm Sci 2025; 20:101006. [PMID: 39931357 PMCID: PMC11808510 DOI: 10.1016/j.ajps.2024.101006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/02/2024] [Accepted: 10/12/2024] [Indexed: 02/13/2025] Open
Abstract
Herpes simplex virus thymidine kinase (HSVtk) gene therapy is a promising strategy for glioblastoma therapy. However, delivery of plasmid DNA (pDNA) encoding HSVtk into the brain by systemic administration is a challenge since pDNA can hardly penetrate the blood-brain barrier. In this study, an exosome-membrane (EM) and polymer-based hybrid complex was developed for systemic delivery of pDNA into the brain. Histidine/arginine-linked polyamidoamine (PHR) was used as a carrier. PHR binds to pDNA by electrostatic interaction. The pDNA/PHR complex was mixed with EM and subjected to extrusion to produce pDNA/PHR-EM hybrid complex. For glioblastoma targeting, T7 peptide was attached to the pDNA/PHR-EM complex. Both pDNA/PHR-EM and T7-decorated pDNA/PHR-EM (pDNA/PHR-EM-T7) had a surface charge of -5 mV and a size of 280 nm. Transfection assays indicated that pDNA/PHR-EM-T7 enhanced the transfection to C6 cells compared with pDNA/PHR-EM. Intravenous administration of pHSVtk/PHR-EM-T7 showed that pHSVtk/PHR-EM and pHSVtk/PHR-EM-T7 delivered pHSVtk more efficiently than pHSVtk/lipofectamine and pHSVtk/PHR into glioblastoma in vivo. pHSVtk/PHR-EM-T7 had higher delivery efficiency than pHSVtk/PHR-EM. As a result, the HSVtk expression and apoptosis levels in the tumors of the pHSVtk/PHR-EM-T7 group were higher than those of the other control groups. Therefore, the pDNA/PHR-EM-T7 hybrid complex is a useful carrier for systemic delivery of pHSVtk to glioblastoma.
Collapse
Affiliation(s)
- Youngki Lee
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Subin Kang
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Le Thi Thuy
- Department of Biochemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Mincheol Son
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Jae Young Park
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Sung Bin Ahn
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Minji Kang
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Jihun Oh
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Joon Sig Choi
- Department of Biochemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| |
Collapse
|
28
|
Ruzzante B, Fruzzetti F, Cattaneo M, Lauria Pinter G, Marcuzzo S, Candiani G, Bono N. Harnessing osmotic shock for enhanced intracellular delivery of (nano)cargos. Int J Pharm 2025; 669:125008. [PMID: 39638270 DOI: 10.1016/j.ijpharm.2024.125008] [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: 08/01/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Efficient intracellular delivery of exogenous (nano)materials is critical for both research and therapeutic applications. The physicochemical properties of the cargo play a crucial role in determining internalization efficacy. Consequently, significant research efforts are focused on developing innovative and effective methodologies to optimize (nano)material delivery. In this study, we utilized osmotic shock to enhance (nano)cargos internalization. We examined the effects of hypotonic/hypertonic shock on both primary and cell lines, assessing parameters such as cell viability, cell volume, membrane tension changes, and particle uptake. Our results indicate that short-lived osmotic shock does not harm cells. Hypotonic shock induced temporary shape changes lasting up to 5 min, followed by a 15-minute recovery period. Importantly, hypotonic shock increased the uptake of 100-nm and 500-nm particles by ∼ 3- and ∼ 5-fold, respectively, compared to isotonic conditions. In contrast, the hypertonic shock did not impact cell behavior or particle uptake. Notably, the internalization mechanisms triggered by osmotic shock operate independently of active endocytic pathways, making hypotonic stimulation particularly beneficial for hard-to-treat cells. When primary fibroblasts derived from amyotrophic lateral sclerosis (ALS)-patients were exposed to hypotonic shock in the presence of the therapeutic cargo icerguastat, there was an increased expression of miR-106b-5p compared to isotonic conditions. In conclusion, osmotic shock presents a promising strategy for improving drug delivery within cells and, potentially, in tissues such as muscles or skin, where localized drug administration is preferred.
Collapse
Affiliation(s)
- Beatrice Ruzzante
- genT_LΛB, Dept. of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Flaminia Fruzzetti
- genT_LΛB, Dept. of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Marco Cattaneo
- ALS Centre, 3rd Neurology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy; Ph.D. Program in Pharmacological Biomolecular Sciences, Experimental and Clinical, University of Milan, Milan, Italy
| | - Giuseppe Lauria Pinter
- ALS Centre, 3rd Neurology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Stefania Marcuzzo
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy; Brain-targeted Nanotechnologies (BraiNs) Lab, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Gabriele Candiani
- genT_LΛB, Dept. of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy; Brain-targeted Nanotechnologies (BraiNs) Lab, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Nina Bono
- genT_LΛB, Dept. of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy.
| |
Collapse
|
29
|
Zhang S, Peng S. Copper-Based biomaterials for anti-tumor therapy: Recent advances and perspectives. Acta Biomater 2025; 193:107-127. [PMID: 39800096 DOI: 10.1016/j.actbio.2025.01.014] [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: 08/26/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Copper, an essential trace element, is integral to numerous metabolic pathways across biological systems. In recent years, copper-based biomaterials have garnered significant interest due to their superior biocompatibility and multifaceted functionalities, particularly in the treatment of malignancies such as sarcomas and cancers. On the one hand, these copper-based materials serve as efficient carriers for a range of therapeutic agents, including chemotherapeutic drugs, small molecule inhibitors, and antibodies, allowing them for precise delivery and controlled release triggered by specific modifications and stimuli. On the other hand, they can induce cell death through mechanisms such as ferroptosis, cuproptosis, apoptosis, and pyroptosis, or inhibit the proliferation and invasion of cancer cells via their outstanding properties. Furthermore, advanced design approaches enable these materials to support tumor imaging and immune activation. Despite this progress, the full scope of their functional capabilities remains to be fully elucidated. This review provides an overview of the anti-tumor functions, underlying mechanisms, and design strategies of copper-based biomaterials, along with their advantages and limitations. The aim is to provide insights into the design, study, and development of novel multifunctional biomaterials, with the ultimate goal of accelerating the clinical application of copper-based nanomaterials in cancer therapy. STATEMENT OF SIGNIFICANCE: This study explores the groundbreaking potential of copper-based biomaterials in cancer therapy, uniquely combining biocompatibility with diverse therapeutic mechanisms such as targeted drug delivery and inhibition of cancer cells through specific cell death pathways. By enhancing tumor imaging and immune activation, copper-based nanomaterials have opened new avenues for cancer treatment. This review examines these multifunctional biomaterials, highlighting their advantages and current limitations while addressing gaps in existing research. The findings aim to accelerate clinical applications of these materials in the field of oncology, providing valuable insights for the design of next-generation copper-based therapies. Therefore, this work is highly relevant to researchers and practitioners focused on innovative cancer treatments.
Collapse
Affiliation(s)
- Shufang Zhang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education of Xiangya Hospital and School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Shuping Peng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education of Xiangya Hospital and School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
30
|
Ataollahi F, Amirheidari B, Amirheidari Z, Ataollahi M. Clinical and mechanistic insights into biomedical application of Se-enriched probiotics and biogenic selenium nanoparticles. Biotechnol Lett 2025; 47:18. [PMID: 39826010 DOI: 10.1007/s10529-024-03559-z] [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: 06/14/2024] [Revised: 11/13/2024] [Accepted: 12/16/2024] [Indexed: 01/20/2025]
Abstract
Selenium is an essential element with various industrial and medical applications, hence the current considerable attention towards the genesis and utilization of SeNPs. SeNPs and other nanoparticles could be achieved via physical and chemical methods, but these methods would not only require expensive equipment and specific reagents but are also not always environment friendly. Biogenesis of SeNPs could therefore be considered as a less troublesome alternative, which opens an excellent window to the selenium and nanoparticles' world. bSeNPs have proved to exert higher bioavailability, lower toxicity, and broader utility as compared to their non-bio counterparts. Many researchers have reported promising features of bSeNP such as anti-oxidant and anti-inflammatory, in vitro and in vivo. Considering this, bSeNPs have been tried as effective agents for health disorders, especially as constituents of probiotics. This article briefly reviews selenium, selenium nanoparticles, Se-enriched probiotics, and bSeNPs' usage in an array of health disorders. Obviously, there are very many articles on bSeNPs, but we wanted to summarize studies on prominent bSeNPs features published in the twenty-first century. This review is hoped to give an outlook to researchers for their future investigations, ultimately serving better care of health disorders.
Collapse
Affiliation(s)
- Farshid Ataollahi
- Extremophile and Productive Microorganisms Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Bagher Amirheidari
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Medical University Campus, Haft-Bagh Highway, Kerman, 76169-13555, Iran.
| | - Zohreh Amirheidari
- Extremophile and Productive Microorganisms Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahshid Ataollahi
- Extremophile and Productive Microorganisms Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
31
|
Soni S, Kori SK, Nema P, Iyer AK, Soni V, Kashaw SK. Cell-penetrating Peptides as Keys to Endosomal Escape and Intracellular Trafficking in Nanomedicine Delivery. Curr Med Chem 2025; 32:1288-1312. [PMID: 38362688 DOI: 10.2174/0109298673278936240107121907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 02/17/2024]
Abstract
This review article discusses the challenges of delivering cargoes to the cytoplasm, for example, proteins, peptides, and nucleic acids, and the mechanisms involved in endosomal escape. Endocytosis, endosomal maturation, and exocytosis pose significant barriers to effective cytoplasmic delivery. The article explores various endosomal escape mechanisms, such as the proton sponge effect, osmotic lysis, membrane fusion, pore formation, membrane destabilization/ disruption, and vesicle budding and collapse. Additionally, it discusses the role of lysosomes, glycocalyx, and molecular crowding in the cytoplasmic delivery process. Despite the recent advances in nonviral delivery systems, there is still a need to improve cytoplasmic delivery. Strategies such as fusogenic peptides, endosomolytic polymers, and cell-penetrating peptides have shown promise in improving endosomal escape and cytoplasmic delivery. More research is needed to refine these strategies and make them safer and more effective. In conclusion, the article highlights the challenges associated with cytoplasmic delivery and the importance of understanding the mechanisms involved in endosomal escape. A better understanding of these processes could result in the creation of greater effectiveness and safe delivery systems for various cargoes, including proteins, peptides, and nucleic acids.
Collapse
Affiliation(s)
- Sakshi Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Shivam K Kori
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Priyanshu Nema
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Arun K Iyer
- Use-inspired Biomaterials and Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan, USA
- Molecular Imaging Program, Karmanos Cancer Institute, Detroit, Michigan, USA
| | - Vandana Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Sushil K Kashaw
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| |
Collapse
|
32
|
Sanati M, Figueroa-Espada CG, Han EL, Mitchell MJ, Yavari SA. Bioengineered Nanomaterials for siRNA Therapy of Chemoresistant Cancers. ACS NANO 2024; 18:34425-34463. [PMID: 39666006 DOI: 10.1021/acsnano.4c11259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Chemoresistance remains a long-standing challenge after cancer treatment. Over the last two decades, RNA interference (RNAi) has emerged as a gene therapy modality to sensitize cancer cells to chemotherapy. However, the use of RNAi, specifically small-interfering RNA (siRNA), is hindered by biological barriers that limit its intracellular delivery. Nanoparticles can overcome these barriers by protecting siRNA in physiological environments and facilitating its delivery to cancer cells. In this review, we discuss the development of nanomaterials for siRNA delivery in cancer therapy, current challenges, and future perspectives for their implementation to overcome cancer chemoresistance.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand 97178, Iran
| | - Christian G Figueroa-Espada
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Emily L Han
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
| |
Collapse
|
33
|
Geng L, Matsumoto M, Yao F, Umino M, Kamiya M, Mukai H, Kawakami S. Microfluidic post-insertion of polyethylene glycol lipids and KK or RGD high functionality and quality lipids in milk-derived extracellular vesicles. Eur J Pharm Sci 2024; 203:106929. [PMID: 39389168 DOI: 10.1016/j.ejps.2024.106929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024]
Abstract
To achieve the desired delivery effect, extracellular vesicles (EVs) must bypass rapid clearance from circulation and exhibit affinity for target cells; however, it is difficult to simultaneously incorporate two materials into EVs. Post-insertion is a general modification method that can be performed by simply mixing different solutions. Previously, we have developed a microfluidic post-insertion method that supported fast and upscaled modification of EVs using KK-modified high-functionality and -quality (HFQ) lipids. Here, we used microfluidic post-insertion to achieve simultaneous incorporation of polyethylene glycol (PEG) lipids and KK or RGD-modified HFQ lipids into milk-derived EVs to avoid uptake from the reticuloendothelial system and increase the uptake into target cells. PEG lipid and HFQ lipids were formulated to produce micelles and subsequently mixed with EV solution using a microfluidic device. Compared to bulk mixing, microfluidic post-insertion showed higher cellular association. Altered cellular association capacities and endocytic pathways indicated simultaneous incorporation. The cellular association of modified EVs can be adjusted by altering the ratio of (EK)4-KK in micelles with slight changes in physicochemical properties. Furthermore, microfluidic post-insertion is also suitable for (SG)5-RGD, which is insoluble in phosphate-buffered saline (PBS). Our results may be valuable for the development and manufacture of functional EVs as drug delivery systems for clinical applications.
Collapse
Affiliation(s)
- Longjian Geng
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki 852-8588, Japan.
| | - Makoto Matsumoto
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki 852-8588, Japan.
| | - Feijie Yao
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki 852-8588, Japan.
| | - Mizuki Umino
- School of Pharmaceutical Sciences, Nagasaki University, 1-14 Bunkyo, Nagasaki-shi, Nagasaki 852-8521, Japan.
| | - Mariko Kamiya
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki 852-8588, Japan.
| | - Hidefumi Mukai
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki 852-8588, Japan.
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki 852-8588, Japan.
| |
Collapse
|
34
|
Wang Y, Sun C, Liu Z, Zhang S, Gao K, Yi F, Zhou W, Liu H. Nanoengineered Endocytic Biomaterials for Stem Cell Therapy. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202410714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Indexed: 01/05/2025]
Abstract
AbstractStem cells, ideal for the tissue repair and regeneration, possess extraordinary capabilities of multidirectional differentiation and self‐renewal. However, the limited spontaneous differentiation potential makes it challenging to harness them for tissue repair without external intervention. Although conventional approaches using biomolecules, small organic molecules, and ions have shown specific and effective functions, they face challenges such as in vivo diffusion and degradation, poor internalization, and side effects on adjacent cells. Nanoengineered biomaterials offer a solution by solidifying and nanosizing these soluble regulating molecules and ions, facilitating their uptake by stem cells. Once inside lysosomes, these nanoparticles release their contents in a controlled “molecule or ion storm,” efficiently altering the intracellular biological and chemical microenvironment to tune the differentiation of stem cells. This newly emerged approach for regulating stem cell fate has attracted much attention in recent years. This method has shown promising results and is poised to enhance clinical stem cell therapy. This review provides an overview of the design principles for nanoengineered biomaterials, discusses the categories and characteristics of nanoparticles, summarizes the application of nanoparticles in tissue repair and regeneration, and discusses the direction of nanoparticle‐enhanced stem cell therapy and prospects for its clinical application in regenerative medicine.
Collapse
Affiliation(s)
- Yingxue Wang
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Chunhui Sun
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Zhaoying Liu
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Shengmin Zhang
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Ke Gao
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Fan Yi
- School of Basic Medical Sciences Shandong University Jinan 250012 P. R. China
| | - Wenjuan Zhou
- School of Basic Medical Sciences Shandong University Jinan 250012 P. R. China
| | - Hong Liu
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
- State Key Laboratory of Crystal Materials Shandong University Jinan 250100 P. R. China
| |
Collapse
|
35
|
Ma Y, Li S, Lin X, Chen Y. A perspective of lipid nanoparticles for RNA delivery. EXPLORATION (BEIJING, CHINA) 2024; 4:20230147. [PMID: 39713203 PMCID: PMC11655307 DOI: 10.1002/exp.20230147] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/07/2024] [Indexed: 12/24/2024]
Abstract
Over the last two decades, lipid nanoparticles (LNPs) have evolved as an effective biocompatible and biodegradable RNA delivery platform in the fields of nanomedicine, biotechnology, and drug delivery. They are novel bionanomaterials that can be used to encapsulate a wide range of biomolecules, such as mRNA, as demonstrated by the current successes of COVID-19 mRNA vaccines. Therefore, it is important to provide a perspective on LNPs for RNA delivery, which further offers useful guidance for researchers who want to work in the RNA-based LNP field. This perspective first summarizes the approaches for the preparation of LNPs, followed by the introduction of the key characterization parameters. Then, the in vitro cell experiments to study LNP performance, including cell selection, cell viability, cellular association/uptake, endosomal escape, and their efficacy, were summarized. Finally, the in vivo animal experiments in the aspects of animal selection, administration, dosing and safety, and their therapeutic efficacy were discussed. The authors hope this perspective can offer valuable guidance to researchers who enter the field of RNA-based LNPs and help them understand the crucial parameters that RNA-based LNPs demand.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Shiyao Li
- School of ScienceRMIT UniversityBundooraVictoriaAustralia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical EngineeringThe University of MelbourneParkvilleVictoriaAustralia
| | - Xin Lin
- Department of Cell BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Yupeng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
36
|
Nasr SS, Paul P, Loretz B, Lehr CM. Realizing time-staggered expression of nucleic acid-encoded proteins by co-delivery of messenger RNA and plasmid DNA on a single nanocarrier. Drug Deliv Transl Res 2024; 14:3339-3353. [PMID: 39009932 DOI: 10.1007/s13346-024-01668-w] [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: 07/04/2024] [Indexed: 07/17/2024]
Abstract
Co-delivery of different protein-encoding polynucleotide species with varying expression kinetics of their therapeutic product will become a prominent requirement in the realm of combined nucleic acid(NA)-based therapies in the upcoming years. The current study explores the capacity for time-staggered expression of encoded proteins by simultaneous delivery of plasmid DNA (pDNA) in the core and mRNA on the shell of the same nanocarrier. The core is based on a Gelatin Type A-pDNA coacervate, thermally stabilized to form an irreversible nanogel stable enough for the deposition of cationic coats namely, protamine sulfate or LNP-related lipid mixtures. Only the protamine-coated nanocarriers remained colloidally stable following mRNA loading and could successfully co-transfect murine dendritic cell line DC2.4 with fluorescent reporter mRNA(mCherry) and pDNA (pAmCyan1). Further investigation of the protamine-coated nanosystem only, the transfection efficiency (percentage of transfected cells) and level of protein expression (mean fluorescence intensity, MFI) of mRNA and pDNA, simultaneously delivered by the same nanocarrier, were compared and kinetically assessed over 48 h in DC2.4 using flow cytometry. The onset of transfection for both nucleotides was initially delayed, with levels < 5% at 6 h. Thereafter, mRNA transfection reached 90% after 24 h and continued to slightly increase until 48 h. In contrast, pDNA transfection was clearly slower, reaching approximately 40% after 24 h, but continuing to increase to reach 94% at 48 h. The time course of protein expression (represented by MFI) for both NAs essentially followed that of transfection. Model-independent as well as model-dependent kinetic parameters applied to the data further confirmed such time-staggered expression of the two NA's where mRNA's rate of transfection and protein expression initially exceeded those of pDNA in the first 24 h of the experiment whereas the opposite was true during the second 24 h of the experiment where pDNA displayed the higher response rates. We expect that innovative nanocarriers capable of time-staggered co-delivery of different nucleotides could open new perspectives for multi-dosing, pulsatile or sustained expression of nucleic acid-based therapeutics in protein replacement, vaccination, and CRISPR-mediated gene editing scenarios.
Collapse
Affiliation(s)
- Sarah S Nasr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany.
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany.
- Fischell Department of Bioengineering, University of Maryland, College Park, USA.
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| | - Pascal Paul
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany.
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany.
| |
Collapse
|
37
|
Salmani-Zarchi H, Mousavi-Sagharchi SMA, Sepahdoost N, Ranjbar-Jamalabadi M, Gross JD, Jooya H, Samadi A. Antimicrobial Feature of Nanoparticles in the Antibiotic Resistance Era: From Mechanism to Application. Adv Biomed Res 2024; 13:113. [PMID: 39717242 PMCID: PMC11665187 DOI: 10.4103/abr.abr_92_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 12/25/2024] Open
Abstract
The growth of nanoscale sciences enables us to define and design new methods and materials for a better life. Health and disease prevention are the main issues in the human lifespan. Some nanoparticles (NPs) have antimicrobial properties that make them useful in many applications. In recent years, NPs have been used as antibiotics to overcome drug resistance or as drug carriers with antimicrobial features. They can also serve as antimicrobial coatings for implants in different body areas. The antimicrobial feature of NPs is based on different mechanisms. For example, the oxidative functions of NPs can inhibit nucleic acid replication and destroy the microbial cell membrane as well as interfere with their cellular functions and biochemical cycles. On the other hand, NPs can disrupt the pathogens' lifecycle by interrupting vital points of their life, such as virus uncoating and entry into human cells. Many types of NPs have been tested by different scientists for these purposes. Silver, gold, copper, and titanium have shown the most ability to inhibit and remove pathogens inside and outside the body. In this review, the authors endeavor to comprehensively describe the antimicrobial features of NPs and their applications for different biomedical goals.
Collapse
Affiliation(s)
- Hamed Salmani-Zarchi
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Nafise Sepahdoost
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahdieh Ranjbar-Jamalabadi
- Department of Polymer Engineering, Faculty of Chemical Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Jeffrey D. Gross
- ReCELLebrate Regenerative Medicine Clinic, Henderson, Nevada, USA
| | - Hossein Jooya
- Biochemistry Group, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ali Samadi
- Department of Basic Sciences, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| |
Collapse
|
38
|
Miao L, Zhu Y, Chang H, Zhang X. Nanotheranostics in Breast Cancer Bone Metastasis: Advanced Research Progress and Future Perspectives. Pharmaceutics 2024; 16:1491. [PMID: 39771471 PMCID: PMC11676679 DOI: 10.3390/pharmaceutics16121491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/27/2024] [Accepted: 11/14/2024] [Indexed: 01/11/2025] Open
Abstract
Breast cancer is the leading cause of cancer-related morbidity and mortality among women worldwide, with bone being the most common site of all metastatic breast cancer. Bone metastases are often associated with pain and skeletal-related events (SREs), indicating poor prognosis and poor quality of life. Most current therapies for breast cancer bone metastasis primarily serve palliative purposes, focusing on pain management, mitigating the risk of bone-related complications, and inhibiting tumor progression. The emergence of nanodelivery systems offers novel insights and potential solutions for the diagnosis and treatment of breast cancer-related bone metastasis. This article reviews the recent advancements and innovative applications of nanodrug delivery systems in the context of breast cancer bone metastasis and explores future directions in nanotheranostics.
Collapse
Affiliation(s)
- Lin Miao
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
- Graduate School, China Medical University, Shenyang 110122, China
| | - Yidan Zhu
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Hong Chang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
| | - Xinfeng Zhang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
- Graduate School, China Medical University, Shenyang 110122, China
| |
Collapse
|
39
|
Patel S, Salaman SD, Kapoor DU, Yadav R, Sharma S. Latest developments in biomaterial interfaces and drug delivery: challenges, innovations, and future outlook. Z NATURFORSCH C 2024:znc-2024-0208. [PMID: 39566511 DOI: 10.1515/znc-2024-0208] [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: 09/25/2024] [Accepted: 11/03/2024] [Indexed: 11/22/2024]
Abstract
An ideal drug carrier system should demonstrate optimal payload and release characteristics, thereby ensuring prolonged therapeutic index while minimizing adverse effects. The field of drug delivery has undergone significant advancements, particularly within the last two decades, owing to the revolutionary impact of biomaterials. The use of biomaterials presents significant due to their biocompatibility and biodegradability, which must be addressed in order to achieve effective drug delivery. The properties of the biomaterial and its interface are primarily influenced by their physicochemical attributes, physiological barriers, cellular trafficking, and immunomodulatory effects. By attuning these barriers, regulating the physicochemical properties, and masking the immune system's response, the bio interface can be effectively modulated, leading to the development of innovative supramolecular structures with enhanced effectiveness. With a comprehensive understanding of these technologies, there is a growing demand for repurposing existing drugs for new therapeutic indications within this space. This review aims to provide a substantial body of evidence showcasing the productiveness of biomaterials and their interface in drug delivery, as well as methods for mitigating and modulating barriers and physicochemical properties along with an examination of future prospects in this field.
Collapse
Affiliation(s)
- Saraswati Patel
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, 602105, Tamil Nadu, India
| | - Samsi D Salaman
- Apollo College of Pharmacy, Mevaloorkuppam, Kanchipuram, 602105, Tamil Nadu, India
| | - Devesh U Kapoor
- Dr. Dayaram Patel Pharmacy College, Sardar Baug, Station Road, 394601 Bardoli, Gujarat, India
| | - Richa Yadav
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O., Rajasthan, 304022, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O., Rajasthan, 304022, India
| |
Collapse
|
40
|
Du H, Ma Y, Wang X, Zhang J, Zhu L, Guan G, Pan S, Zhang Y, Wang J, Liu Z. Therapeutic Suppression of Atherosclerotic Burden and Vulnerability via Dll4 Inhibition in Plaque Macrophages Using Dual-Targeted Liposomes. ACS APPLIED BIO MATERIALS 2024; 7:7219-7232. [PMID: 39392531 DOI: 10.1021/acsabm.4c00923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Atherosclerosis, characterized by chronic inflammation within the arterial wall, remains a pivotal concern in cardiovascular health. We developed a dual-targeted liposomal system encapsulating Dll4-targeting siRNA, designed to selectively bind to pro-inflammatory M1 macrophages through surface conjugation with anti-F4/80 and anti-CD68 antibodies. The Dll4-targeting siRNA is then delivered to the macrophages, where it silences Dll4 expression, inhibiting Notch signaling and reducing plaque vulnerability. Emphasizing accuracy in targeting, the system demonstrates effective suppression of Dll4, a key modulator of atherosclerotic progression, and vulnerability via VSMCs phenotypic conversion and senescence. By employing liposomes for siRNA delivery, we observed enhanced stability and specificity of the siRNA. Alongside the therapeutic efficacy, our study also evaluated the safety profile and pharmacokinetics of the dual-targeted liposomal system, revealing favorable outcomes with minimal off-target effects and optimal biodistribution. The integration of RNA interference techniques with advanced nanotechnological methodologies signifies the importance of targeted delivery in this therapeutic approach. Preliminary findings suggest a potential attenuation in plaque development and vulnerability, indicating the therapeutic promise of this approach. This research emphasizes the potential of nanocarrier-mediated precision targeting combined with a reassuring safety and pharmacokinetic profile for advancing atherosclerosis therapeutic strategies.
Collapse
Affiliation(s)
- Haixia Du
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Department 403, PLA Rocket Force University of Engineering, Xi'an 710025, China
| | - Yanpeng Ma
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710003, China
- Traditional Chinese Medicine Inheritance and Innovation Platform, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Northwestern Polytechnical University, Affiliated Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Xiqiang Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710003, China
- Traditional Chinese Medicine Inheritance and Innovation Platform, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Northwestern Polytechnical University, Affiliated Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Junbo Zhang
- Department of Peripheral Vascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Ling Zhu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710003, China
- Traditional Chinese Medicine Inheritance and Innovation Platform, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Northwestern Polytechnical University, Affiliated Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Gongchang Guan
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710003, China
- Traditional Chinese Medicine Inheritance and Innovation Platform, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Northwestern Polytechnical University, Affiliated Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Shuo Pan
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710003, China
- Traditional Chinese Medicine Inheritance and Innovation Platform, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Northwestern Polytechnical University, Affiliated Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Yong Zhang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710003, China
- Traditional Chinese Medicine Inheritance and Innovation Platform, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Northwestern Polytechnical University, Affiliated Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Junkui Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710003, China
- Traditional Chinese Medicine Inheritance and Innovation Platform, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Northwestern Polytechnical University, Affiliated Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710003, China
- Traditional Chinese Medicine Inheritance and Innovation Platform, Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Northwestern Polytechnical University, Affiliated Shaanxi Provincial People's Hospital, Xi'an 710068, China
- Shaanxi Cardiovascular and Cerebrovascular Disease Precision Medicine "Belt and Road" Joint Laboratory, Xi'an 710068, China
| |
Collapse
|
41
|
Wang HD, Lv CL, Feng L, Guo JX, Zhao SY, Jiang P. The role of autophagy in brain health and disease: Insights into exosome and autophagy interactions. Heliyon 2024; 10:e38959. [PMID: 39524893 PMCID: PMC11546156 DOI: 10.1016/j.heliyon.2024.e38959] [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: 09/21/2023] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Effective management of cellular components is essential for maintaining brain health, and studies have identified several crucial biological processes in the brain. Among these, autophagy and the role of exosomes in cellular communication are critical for brain health and disease. The interaction between autophagy and exosomes in the nervous system, as well as their contributions to brain damage, have garnered significant attention. This review summarizes that exosomes and their cargoes have been implicated in the autophagy process in the pathophysiology of nervous system diseases. Furthermore, the onset and progression of neurological disorders may be affected by autophagy regulation of the secretion and release of exosomes. These findings may provide new insights into the potential mechanism by which autophagy mediates different exosome secretion and release, as well as the valuable biomedical applications of exosomes in the prevention and treatment of various brain diseases by targeting autophagy.
Collapse
Affiliation(s)
- Hai-Dong Wang
- Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/Nanjing Medical University Kangda College First Affiliated Hospital/The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Chao-Liang Lv
- Department of Spine Surgery, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Lei Feng
- Department of Neurosurgery, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Jin-Xiu Guo
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Shi-Yuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| |
Collapse
|
42
|
Tong S, Niu J, Wang Z, Jiao Y, Fu Y, Li D, Pan X, Sheng N, Yan L, Min P, Chen D, Cui S, Liu Y, Lin S. The Evolution of Microfluidic-Based Drug-Loading Techniques for Cells and Their Derivatives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403422. [PMID: 39152940 DOI: 10.1002/smll.202403422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/03/2024] [Indexed: 08/19/2024]
Abstract
Conventional drug delivery techniques face challenges related to targeting and adverse reactions. Recent years have witnessed significant advancements in nanoparticle-based drug carriers. Nevertheless, concerns persist regarding their safety and insufficient metabolism. Employing cells and their derivatives, such as cell membranes and extracellular vesicles (EVs), as drug carriers effectively addresses the challenges associated with nanoparticle carriers. However, an essential hurdle remains in efficiently loading drugs into these carriers. With the advancement of microfluidic technology and its advantages in precise manipulation at the micro- and nanoscales, as well as minimal sample loss, it has found extensive application in the loading of drugs using cells and their derivatives, thereby fostering the development of drug-loading techniques. This paper outlines the characteristics and benefits of utilizing cells and their derivatives as drug carriers and provides an overview of current drug-loading techniques, particularly those rooted in microfluidic technology. The significant potential for microfluidic technology in targeted disease therapy through drug delivery systems employing cells and their derivatives, is foreseen.
Collapse
Affiliation(s)
- Siyu Tong
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jiaqi Niu
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhitao Wang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yingao Jiao
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yanfei Fu
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Dongxia Li
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinni Pan
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Nengquan Sheng
- Department of General Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Li Yan
- Department of Geriatric Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Peiru Min
- Shanghai 9th People's Hospital, Shanghai Jiao Tong University College of Medicine, Shanghai, 200240, China
| | - Di Chen
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shengsheng Cui
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yanlei Liu
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shujing Lin
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
43
|
Jhaveri JR, Khare P, Paul Pinky P, Kamte YS, Chandwani MN, Milosevic J, Abraham N, Sun M, Stolz DB, Dave KM, Zheng SY, O'Donnell L, Manickam DS. Low pinocytic brain endothelial cells primarily utilize membrane fusion to internalize extracellular vesicles. Eur J Pharm Biopharm 2024; 204:114500. [PMID: 39303949 DOI: 10.1016/j.ejpb.2024.114500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Extracellular vesicles (EVs) are an emerging class of drug carriers and are primarily reported to be internalized into recipient cells via a combination of endocytic routes such as clathrin-mediated, caveolae-mediated and macropinocytosis pathways. In this work, (1) we investigated potential effects of homotypic vs. heterotypic interactions by studying the cellular uptake of homologous EVs (EV donor cells and recipient cells of the same type) vs. heterologous EVs (EV donor cells and recipient cells of different types) and (2) determined the route of EV internalization into low pinocytic/hard-to-deliver cell models such as brain endothelial cells (BECs). Homotypic interactions led to a greater extent of uptake into the recipient BECs compared to heterotypic interactions. However, we did not see a complete reduction in EV uptake into recipient BECs when endocytic pathways were blocked using pharmacological inhibitors and our findings from a R18-based fusion assay suggest that EVs primarily use membrane fusion to enter low-pinocytic recipient BECs instead of relying on endocytosis. Lipophilic PKH67 dye-labeled EVs but not intravesicular esterase-activated calcein ester-labeled EVs severely reduced particle uptake into BECs while phagocytic macrophages internalized EVs labeled with both dyes to comparable extents. Our results also highlight the importance of carefully choosing labeling dye chemistry to study EV uptake, especially in the case of low pinocytic cells such as BECs.
Collapse
Affiliation(s)
- Jhanvi R Jhaveri
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Purva Khare
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Paromita Paul Pinky
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Yashika S Kamte
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Manisha N Chandwani
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Jadranka Milosevic
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States; Captis Diagnostics Inc., Pittsburgh, PA, United States
| | - Nevil Abraham
- Unified Flow Core, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ming Sun
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Donna B Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Kandarp M Dave
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Si-Yang Zheng
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Lauren O'Donnell
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Devika S Manickam
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States.
| |
Collapse
|
44
|
Kesharwani P, Halwai K, Jha SK, Al Mughram MH, Almujri SS, Almalki WH, Sahebkar A. Folate-engineered chitosan nanoparticles: next-generation anticancer nanocarriers. Mol Cancer 2024; 23:244. [PMID: 39482651 PMCID: PMC11526716 DOI: 10.1186/s12943-024-02163-z] [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: 05/03/2024] [Accepted: 10/19/2024] [Indexed: 11/03/2024] Open
Abstract
Chitosan nanoparticles (NPs) are well-recognized as promising vehicles for delivering anticancer drugs due to their distinctive characteristics. They have the potential to enclose hydrophobic anticancer molecules, thereby enhancing their solubilities, permeabilities, and bioavailabilities; without the use of surfactant, i.e., through surfactant-free solubilization. This allows for higher drug concentrations at the tumor sites, prevents excessive toxicity imparted by surfactants, and could circumvent drug resistance. Moreover, biomedical engineers and formulation scientists can also fabricate chitosan NPs to slowly release anticancer agents. This keeps the drugs at the tumor site longer, makes therapy more effective, and lowers the frequency of dosing. Notably, some types of cancer cells (fallopian tube, epithelial tumors of the ovary, and primary peritoneum; lung, kidney, ependymal brain, uterus, breast, colon, and malignant pleural mesothelioma) have overexpression of folate receptors (FRs) on their outer surface, which lets folate-drug conjugate-incorporated NPs to target and kill them more effectively. Strikingly, there is evidence suggesting that the excessively produced FR&αgr (isoforms of the FR) stays consistent throughout treatment in ovarian and endometrial cancer, indicating resistance to conventional treatment; and in this regard, folate-anchored chitosan NPs can overcome it and improve the therapeutic outcomes. Interestingly, overly expressed FRs are present only in certain tumor types, which makes them a promising biomarker for predicting the effectiveness of FR-targeted therapy. On the other hand, the folate-modified chitosan NPs can also enhance the oral absorption of medicines, especially anticancer drugs, and pave the way for effective and long-term low-dose oral metronomic scheduling of poorly soluble and permeable drugs. In this review, we talked briefly about the techniques used to create, characterize, and tailor chitosan-based NPs; and delved deeper into the potential applications of folate-engineered chitosan NPs in treating various cancer types.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Kratika Halwai
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Saurav Kumar Jha
- Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Uttar Pradesh, Kanpur, 208016, India
| | - Mohammed H Al Mughram
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Postal Code 61421, Abha, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Postal Code 61421, Abha, Saudi Arabia
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
45
|
Carmona-Ribeiro AM, Pérez-Betancourt Y. Emerging Cationic Nanovaccines. Pharmaceutics 2024; 16:1362. [PMID: 39598488 PMCID: PMC11597065 DOI: 10.3390/pharmaceutics16111362] [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: 09/26/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Cationic vaccines of nanometric sizes can directly perform the delivery of antigen(s) and immunomodulator(s) to dendritic cells in the lymph nodes. The positively charged nanovaccines are taken up by antigen-presenting cells (APCs) of the lymphatic system often originating the cellular immunological defense required to fight intracellular microbial infections and the proliferation of cancers. Cationic molecules imparting the positive charges to nanovaccines exhibit a dose-dependent toxicity which needs to be systematically addressed. Against the coronavirus, mRNA cationic nanovaccines evolved rapidly. Nowadays cationic nanovaccines have been formulated against several infections with the advantage of cationic compounds granting protection of nucleic acids in vivo against biodegradation by nucleases. Up to the threshold concentration of cationic molecules for nanovaccine delivery, cationic nanovaccines perform well eliciting the desired Th 1 improved immune response in the absence of cytotoxicity. A second strategy in the literature involves dilution of cationic components in biocompatible polymeric matrixes. Polymeric nanoparticles incorporating cationic molecules at reduced concentrations for the cationic component often result in an absence of toxic effects. The progress in vaccinology against cancer involves in situ designs for cationic nanovaccines. The lysis of transformed cancer cells releases several tumoral antigens, which in the presence of cationic nanoadjuvants can be systemically presented for the prevention of metastatic cancer. In addition, these local cationic nanovaccines allow immunotherapeutic tumor treatment.
Collapse
Affiliation(s)
- Ana Maria Carmona-Ribeiro
- Biocolloids Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Yunys Pérez-Betancourt
- Department of Microbiology, University of Chicago, Cummings Life Science Center 920 E 58th St., Chicago, IL 60637, USA;
| |
Collapse
|
46
|
Meshram PR, Ranpise NS. Lymphatic targeting of cilnidipine by designing and developing a nanostructured lipid carrier drug delivery system. Drug Dev Ind Pharm 2024:1-17. [PMID: 39395164 DOI: 10.1080/03639045.2024.2415638] [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: 08/15/2024] [Revised: 09/19/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
OBJECTIVE The objective of current research is to design, develop, and optimize a cilnidipine (CLN) nanostructured lipid carrier (NLC)-based drug delivery system for the effective treatment of hypertension (HT). SIGNIFICANCE Oral administration of CLN-loaded NLC (CLN NLC) containing glyceryl monostearate (GMS) as a solid and isopropyl myristate (IPM) as a liquid lipid may show remarkable lymphatic uptake through payer patches. METHODS The emulsification probe sonication technique was used followed by optimization using 32 factorial designs. RESULTS The optimized batch showed a mean particle size of 115.4 ± 0.22 nm with encapsulation efficiency of 98.32 ± 0.23%, polydispersity index (PDI) of 0.342 ± 0.03, and zeta potential (ZP, ζ) was -60.5 ± 0.24 which indicate excellent physical stability. In vitro studies showed a controlled release of CLN NLCs. Pharmacokinetics studies determined the Cmax of NLCs (373.47 ± 15.1) indicates 2.3-fold enhancement compared with plain drug (160.64 ± 7.63). Pharmacodynamic studies indicated that CLN NLCs were maintaining systolic blood pressure in a controlled manner without any signs of side effects. CONCLUSION CLN NLCs significantly improved lymphatic delivery and proved to be effective in the treatment and management of HT. It has been proved that CLN NLCs are found to be better than any traditional CLN dosage form due to enhancement in solubility, absorption, bioavailability, intestinal permeability, avoidance of first-pass metabolism, P-glycoprotein efflux and reduction in dose-related side effects, achievement of controlled and sustained release action.
Collapse
|
47
|
Yamin MZ, Liu JY, Sayes CM. Comparative Assessment of Cellular Responses to Microscale Silica Morphologies in Human Gastrointestinal Cells: Insights for Occupational Health. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:1376. [PMID: 39457349 PMCID: PMC11508045 DOI: 10.3390/ijerph21101376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024]
Abstract
Silicon dioxide (SiO2), commonly known as silica, is a naturally occurring mineral extracted from the Earth's crust. It is widely used in commercial products such as food, medicine, and dental ceramics. There are few studies on the health effects of pyrogenic and colloidal silica after ingestion. No research has compared the impact of microscale morphologies on mitochondrial activity in colon cells after acute exposure. The results show that crystalline and amorphous silica had a concentration-independent effect on cells, with an initial increase in mitochondrial activity followed by a decrease. Vitreous silica did not affect cells. Diatomaceous earth and pyrogenic silica had a concentration-dependent response, causing a reduction in mitochondrial activity as concentration increased. Diatomaceous earth triggered the highest cellular response, with mitochondrial activity ranging from 78.84% ± 12.34 at the highest concentration (1000 ppm) to 62.54% ± 17.43 at the lowest concentration (0.01 ppm) and an average H2O2 concentration of 1.48 ± 0.15 RLUs. This research advances our understanding of silica's impact on human gastrointestinal cells, highlighting the need for ongoing exploration. These findings can improve risk mitigation strategies in silica-exposed environments.
Collapse
Affiliation(s)
| | | | - Christie M. Sayes
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, TX 76798-7266, USA; (M.Z.Y.); (J.Y.L.)
| |
Collapse
|
48
|
Tarab-Ravski D, Stotsky-Oterin L, Elisha A, Kundoor GR, Ramishetti S, Hazan-Halevy I, Haas H, Peer D. The future of genetic medicines delivered via targeted lipid nanoparticles to leukocytes. J Control Release 2024; 376:286-302. [PMID: 39401676 DOI: 10.1016/j.jconrel.2024.10.014] [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: 07/05/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Genetic medicines hold vast therapeutic potential, offering the ability to silence or induce gene expression, knock out genes, and even edit DNA fragments. Applying these therapeutic modalities to leukocytes offers a promising path for treating various conditions yet overcoming the obstacles of specific and efficient delivery to leukocytes remains a major bottleneck in their clinical translation. Lipid nanoparticles (LNPs) have emerged as the leading delivery system for nucleic acids due to their remarkable versatility and ability to improve their in vivo stability, pharmacokinetics, and therapeutic benefits. Equipping LNPs with targeting moieties can promote their specific cellular uptake and internalization to leukocytes, making targeted LNPs (tLNPs) an inseparable part of developing leukocyte-targeted gene therapy. However, despite the significant advancements in research, genetic medicines for leukocytes using targeted delivery approaches have not been translated into the clinic yet. Herein, we discuss the important aspects of designing tLNPs and highlight the considerations for choosing an appropriate bioconjugation strategy and targeting moiety. Furthermore, we provide our insights on limiting challenges and identify key areas for further research to advance these exciting therapies for patient care.
Collapse
Affiliation(s)
- Dana Tarab-Ravski
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Aviad Elisha
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Govinda Reddy Kundoor
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | | | - Inbal Hazan-Halevy
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Heinrich Haas
- NeoVac Ltd. 127 Olympic Ave., OX14 4SA, Milton Park, Oxfordshire, UK; Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg-University, Mainz, Germany
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
49
|
Qin Y, Ling X, Li Y, Wang J, Wang J, Rong Z, Cheng Y, Tao Z, Zhang H, Wei H, Yu CY. Histidine phosphatase-ferroptosis crosstalk modulation for efficient hepatocellular carcinoma treatment. J Nanobiotechnology 2024; 22:622. [PMID: 39402673 PMCID: PMC11476632 DOI: 10.1186/s12951-024-02918-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Altering the mechanisms of tumor cell death and overcoming the limitations of traditional chemotherapy is pivotal to contemporary tumor treatment. Inducing ferroptosis, while circumventing safety concerns associated with ferrous vectors, through nonferrous ferroptosis is a promising but underexplored frontier in cancer therapy. Histidine phosphatase (LHPP) has emerged as a novel therapeutic target in treating hepatocellular carcinoma (HCC), but the precise mechanism of LHPP against HCC remains unclear. Herein, we explore the effects of upregulating LHPP expression on ferroptosis and tumor immunogenicity induction by simply delivering a miRNA-363-5p inhibitor (miR-363-5pi) via a previously optimized gemcitabine-oleic acid (GOA) prodrug. Efficient miRNA encapsulation was achieved through hydrogen bonding at an optimized GOA/miRNA molar feed ratio of 250:1, affording spherical nanoparticles with a uniform hydrodynamic size of 147.1 nm and a negative potential of -21.5 mV. The mechanism of this LHPP-ferroptosis crosstalk is disclosed to be an inhibited phosphorylation of the PI3K/Akt pathway, leading to a remarkable tumor inhibition rate of 88.2% in nude mice bearing Bel-7402 tumor xenografts via a combination of LHPP-triggered nonferrous ferroptosis and GOA-induced chemotherapy. The biocompatibility of GOA/miR-363-5pi is strongly supported by their non-hematologic toxicity and insignificant organ damage. In addition, the tumor immunogenic activation potential of GOA/miR-363-5pi was finally explored. Overall, this study is the first work that elucidates the precise mechanism of LHPP for treating HCC via ferroptosis induction and achieves the transformation of chemotherapy and gene therapy into ferroptosis activation with tumor cell immunogenicity, which lays a new therapeutic foundation for the clinical treatment of HCC.
Collapse
Affiliation(s)
- Yang Qin
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xiaoli Ling
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yunxian Li
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jieqiong Wang
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jiaqi Wang
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zhuoyi Rong
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yao Cheng
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zhenghao Tao
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Haitao Zhang
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Hua Wei
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Cui-Yun Yu
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China.
- Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, 410006, China.
| |
Collapse
|
50
|
Martín‐Morales C, Caspani S, Desco M, Tavares de Sousa C, Gómez‐Gaviro MV. Controlled Drug Release Systems for Cerebrovascular Diseases. ADVANCED THERAPEUTICS 2024. [DOI: 10.1002/adtp.202400239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Indexed: 01/06/2025]
Abstract
AbstractThis review offers a comprehensive exploration of optimized drug delivery systems tailored for controlled release and their crucial role in addressing cerebrovascular diseases. Through an in‐depth analysis, various controlled release methods, including nanoparticles, liposomes, hydrogels, and other emerging technologies are examined. Highlighting the importance of precise drug targeting, it is delved into the underlying mechanisms of these delivery systems and their potential to improve therapeutic outcomes while minimizing adverse effects. Additionally, the specific applications of these optimized drug delivery systems in treating cerebrovascular disorders such as ischemic stroke, cerebral aneurysms, and intracranial hemorrhage are discussed. By shedding light on the advancements in drug delivery techniques and their implications in cerebrovascular medicine, this review offers valuable insights into the future of therapeutic interventions in neurology.
Collapse
Affiliation(s)
- Celia Martín‐Morales
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM) Doctor Esquerdo 46 Madrid 28007 Spain
| | - Sofia Caspani
- IFIMUP – Institute of Physics for Advanced Materials Departamento de Física e Astronomia, Nanotechnology and Photonics of University of Porto Faculdade de Ciências Universidade do Porto, Rua do Campo Alegre s/n Porto 4169‐007 Portugal
| | - Manuel Desco
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM) Doctor Esquerdo 46 Madrid 28007 Spain
- Departamento de Bioingeniería Universidad Carlos III de Madrid Leganés 28911 Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM) Madrid 28029 Spain
- Centro de Investigaciones Cardiovasculares (CNIC) Melchor Fernández Almagro Madrid 28029 Spain
| | - Célia Tavares de Sousa
- Departamento de Física Aplicada and IAdChem Facultad de Ciencias Universidad Autonoma de Madrid (UAM) Campus de Cantoblanco, C/ Francisco Tomas y Valiente, 7 Madrid 28049 Spain
| | - María Victoria Gómez‐Gaviro
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM) Doctor Esquerdo 46 Madrid 28007 Spain
- Departamento de Bioingeniería Universidad Carlos III de Madrid Leganés 28911 Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM) Madrid 28029 Spain
| |
Collapse
|