1
|
Monem AS, Ghannam MM, ElBaz TA, Fahmy HM. Synthesis, characterization, and cytotoxicity evaluation of magnetoliposomes in human breast cancer MCF-7 cells. Biochem Biophys Res Commun 2025; 772:152016. [PMID: 40412366 DOI: 10.1016/j.bbrc.2025.152016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/30/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
This study examines and evaluates the potential lethality of the highly mobile phospholipid bilayer of magnetoliposomes (MNLPS). Ferrous oxide nanoparticles (MNPs) measuring less than 20 nm in diameter were synthesized and enclosed within liposomes made of single and mixed phospholipids, yielding liposomes with diameters of approximately 200 nm. The physical attributes of the MNPs, including magnetic susceptibility, Zeta potential, size distribution, optical anisotropy spectrum, and nonlinear optical properties, were carefully measured. The liposomes were synthesized via the rotary evaporation method and contained a significant concentration of MNPs. The size distribution and Zeta potential of the magnetoliposomes (MNLPS) were examined using dynamic light scattering (DLS) and transmission electron microscopy (TEM). The gel-to-liquid crystalline transition characteristics of the ML were examined utilizing differential scanning calorimetry (DSC) and depolarized light scattering. The nonlinear optical spectra of the MNP and ML were analyzed below and above their primary transition temperature. The potential cytotoxicity of the ML against human breast cancer cells (MCF-7) was examined with and without a rotating magnetic field. This work successfully produced liposome-encapsulated magnetic nanoparticles using alternating magnetic field therapy for potential cancer cell eradication. The DLS, Zeta potential, and TEM analyses validated the suitable particle size distribution. The magnetic susceptibility measurements verified that the magnetic properties of the liposome-encapsulated magnetic nanoparticles fall within the permitted range. Consequently, magnetoliposomes subjected to a highly mobile alternating magnetic field are advocated as a therapeutic agent for cancer treatment, showing potential in vitro efficacy. The findings of our MTT assay show that MNLPS may show improved efficacy when subjected to magnetic field irradiation. Treatment of MCF-7 breast cancer cells with MNLPS, followed by magnetic field irradiation, significantly exhibited enhanced toxicity. The increased cytotoxicity of the combination therapy against cancer cells is due to membrane permeability, improved intracellular uptake of magnetoliposomes, and hyperthermia. Additional research is necessary to determine the optimal doses of MNLPS, intensity, duration of exposure, mobility, setup of the magnetic field, and type for magnetic field application.
Collapse
Affiliation(s)
- Ahmed Sultan Monem
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Toka Ashraf ElBaz
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt.
| | - Heba Mohamed Fahmy
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt.
| |
Collapse
|
2
|
Xu Y, Saiding Q, Zhou X, Wang R, Wang J, Cui W, Chen X. Promoting mitocytosis via gene-engineered aligned fibers for fascia regeneration. J Control Release 2025; 382:113725. [PMID: 40233829 DOI: 10.1016/j.jconrel.2025.113725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/17/2025]
Abstract
The abnormal accumulation of damaged mitochondria severely impedes tissue repair, and conventional therapeutic approaches, such as drug treatments, are often ineffective to remove damaged mitochondria. In this study, we developed gene-engineered aligned electrospun fibers by integrating microfluidic chip technology with a micro-sol oriented electrospinning technique. This study is the first to demonstrate the repair of damaged fascia by promoting mitocytosis through upregulating tetraspanin-9 (TSPAN9). The key gene for mitochondrial exocytosis, TSPAN9, was initially encapsulated into liposomes using microfluidic chip technology. Subsequently, core-shell structured aligned electrospun fibers were fabricated via oriented micro-sol electrospinning, where TSPAN9-loaded liposomes were protected by hyaluronic acid (HA) in the core layer, while aligned polylactic acid (PLA) fibers formed the outer shell layer. In vitro studies revealed that the aligned fibers closely mimicked the oriented structure of fascia tissue and significantly enhanced cell migration by providing directional physical cues. By sustained release of gene-loaded liposomes into cells, mitochondrial homeostasis was effectively restored, mitochondrial respiration was recovered, reactive oxygen species levels were reduced, and mitochondrial membrane potential was maintained. In vivo studies confirmed that these gene-engineered fibers effectively suppressed inflammatory responses and promoted fascia regeneration by facilitating the removal of damaged mitochondria through mitocytosis. In conclusion, gene-engineered fibers developed in this study, which enhance mitocytosis, offer a novel and promising therapeutic strategy for fascia tissue repair.
Collapse
Affiliation(s)
- Yiru Xu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xue Zhou
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Rui Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China
| | - Juan Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China..
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China..
| | - Xinliang Chen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China.
| |
Collapse
|
3
|
Li W, Wang R, Su Z, Li S, Zhao G, Wang Q, Cao H, Zhang L. Self-Iron-Enriched Bacterial Membrane Nanovesicles for Cascade and Multi-Modal Antitumor Therapy. ACS Biomater Sci Eng 2025. [PMID: 40387446 DOI: 10.1021/acsbiomaterials.5c00217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
The integration of microbiology and nanotechnology offers a novel strategy for cancer treatment. In this study, we innovatively propose the use of Pseudomonas aeruginosa bacterial membranes as nanocarriers. These membranes possess a simple and unique self-enriching property for iron, which, in addition to the inherent immune effects of the membrane itself, can facilitate tumor chemodynamic therapy through Fenton reactions. The system encapsulates the anticancer drug β-Lapachone, which can generate a large amount of hydrogen peroxide within cells, further serving as a substrate for the Fenton reaction, leading to a cascade reaction that achieves a synergistic effect of three therapeutic modalities in tumor treatment. Moreover, the aptamer AS1411 is used to enhance tumor targeting and optimize drug delivery within the tumor microenvironment. This investigation presents a multimodal antitumor strategy that demonstrates enhanced antitumor effects both in vitro and in vivo, providing a new paradigm for the antitumor application of bacterial membrane nanocarriers.
Collapse
Affiliation(s)
- Weizheng Li
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, China
| | - Ruiqi Wang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, China
| | - Zhenzhen Su
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, China
| | - Shang Li
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, China
| | - Guoping Zhao
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266000, China
| | - Qinghua Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250000, China
| | - Hongqian Cao
- Department of Health Inspection and Quarantine, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, China
| | - Lei Zhang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266000, China
| |
Collapse
|
4
|
Pisani S, Isaac GT, Dorati R, Genta I, Bertino G, Benazzo M, Conti B. Assessing liposomal nanocarriers for targeted drug delivery through electroporation. Int J Pharm 2025; 679:125708. [PMID: 40354904 DOI: 10.1016/j.ijpharm.2025.125708] [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: 03/10/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Electroporation (EP) is a technique that temporarily increases cell membrane permeability through high-voltage electrical pulses, facilitating the internalization of hydrophilic drugs. When used in clinics, reversible EP offers significant advantages in drug delivery with minimal systemic toxicity, making it a promising approach in cancer therapy (Electrochemotherapy). However, is still challenging to increase therapeutic efficacy, such as increasing the amount of drug internalized by cells after EP. To address these limitations, integrating nanocarriers-particularly liposomes-into EP-based drug delivery strategies has shown great promise. Due to their structural similarity to cell membranes, liposomes can undergo electroporation without causing irreversible cell damage, enabling localized and controlled drug release at targeted sites. This study preliminary evaluates the effectiveness of positively charged gentamicin sulfate loaded liposomes (GS-Lipo) in enhancing gentamicin sulfate uptake through electroporation. The focus is on liposome behavior under EP, drug release, and cellular internalization. The results reveal a strong interplay between liposomes and EP. While EP minimally affects liposome size (sizes lower than 250 nm before and after EP) and PDI, it significantly enhances intracellular uptake and drug release by creating transient pores in liposomal bilayer, facilitating gentamicin diffusion. In vitro uptake studies performed with fluorescent liposomes and GS-Lipo, confirmed superior performance when combined treatment (EP + GS-Lipo) is used. By optimizing electroporation parameters (160 V, 200 V, and 250 V), this study succeeds in maximizing intracellular drug concentration, with the long-term goal of improving therapeutic outcomes, particularly in cancer treatment.
Collapse
Affiliation(s)
- Silvia Pisani
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy.
| | | | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Giulia Bertino
- Department of Otorhinolaryngology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Marco Benazzo
- Department of Otorhinolaryngology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| |
Collapse
|
5
|
Huang Y, Li Q, He F, Yang T, Zhou Q, Zheng Y, Li Y. Cationic Azobenzene Tag to Enhance Liposomal Prodrug Retention and Tumor-Targeting Prodrug Activation for Improved Antitumor Efficacy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:26323-26337. [PMID: 40285708 DOI: 10.1021/acsami.5c01634] [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: 04/29/2025]
Abstract
In this study, we reported a cationic azobenzene (Azo) tag to increase the retention of camptothecin (CPT) prodrugs in liposomes driven by π-π stacking interaction between Azo. Compared with a cationic CPT prodrug without Azo, the liposome-encapsulating Azo-linked CPT prodrugs (AzoCPT-Lips) exhibited slower prodrug leakage in plasma and a longer blood circulation time in mice. The AzoCPT-Lips had a high encapsulation efficiency (95%), loading capacity (20%, by weight), and good storage stability. The AzoCPT was efficiently taken up by 4T1 tumor cells (100-fold higher than CPT) and readily converted into active CPT in the cytoplasm to exert 10-fold higher cytotoxicity than free CPT. More importantly, AzoCPT-Lips resulted in 5-20 times higher tumor distribution of active CPT than that of CPT solution or those in other tissues, which further led to more potent antitumor activity and lower toxicities in the 4T1 breast cancer xenograft. Such a cationic Azo tag represents an effective strategy for developing liposomal antitumor drugs with improved antitumor efficacy.
Collapse
Affiliation(s)
- Yuanyuan Huang
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Qiunan Li
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Fei He
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Tao Yang
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Qing Zhou
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu 610051, China
| | - Yaxin Zheng
- Key Laboratory of Structure-Specific Small Molecule Drugs at Chengdu Medical College of Sichuan Province, School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yang Li
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
6
|
Gong J, Zhang W, Balthasar JP. Camptothein-Based Anti-Cancer Therapies and Strategies to Improve Their Therapeutic Index. Cancers (Basel) 2025; 17:1032. [PMID: 40149365 PMCID: PMC11941615 DOI: 10.3390/cancers17061032] [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: 02/06/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Camptothecin and its derivatives (CPTs) are potent antineoplastic agents that exert their effects by inhibiting DNA topoisomerase I, leading to apoptosis during cell proliferation. Since their discovery in the 1960s, CPTs have faced challenges such as low water solubility, pH-dependent lactone ring instability, and severe off-target toxicities. Despite extensive research, only two CPTs, irinotecan and topotecan, have received health authority approval. Ongoing clinical trials continue to explore the use of CPTs in combination with targeted therapies and immunotherapies to expand their clinical use. Drug delivery systems, including liposomes and antibody-drug conjugates (ADCs), have significantly enhanced the therapeutic index of CPTs. Liposomal irinotecan (Onivyde®, Ipsen, Paris, France) and two ADCs delivering CPT payloads, trastuzumab deruxtecan (Enhertu®, Daiichi Sankyo, Tokyo, Japan) and sacituzumab govitecan (Trodelvy®, Gilead Sciences, Inc., Foster City, CA, USA), have demonstrated substantial efficacy and safety. There is promise that novel strategies such as inverse targeting and co-dosing with anti-idiotypic distribution enhancers may expand the utility of CPT ADCs. This review highlights CPT therapies in clinical use and discusses approaches to further enhance their therapeutic selectivity.
Collapse
Affiliation(s)
| | | | - Joseph P. Balthasar
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA (W.Z.)
| |
Collapse
|
7
|
Ren F, Gan Z, Zhang Q, He D, Chen B, Wu X, Zeng X, Wu K, Xing Y, Zhang Y, Chen H. Construction and evaluation of liposomal drug delivery system for an ALK/HDACs dual-targeted inhibitor with sustained release and enhanced antitumor effect. Drug Deliv Transl Res 2025; 15:939-954. [PMID: 39112826 DOI: 10.1007/s13346-024-01647-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2024] [Indexed: 02/01/2025]
Abstract
ALK/HDACs dual target inhibitor (PT-54) was a 2,4-pyrimidinediamine derivative synthesized based on the pharmacophore merged strategy that inhibits both anaplastic lymphoma kinase (ALK) and histone deacetylases (HDACs), which has demonstrated significant efficacy in treating multiple cancers. However, its poor solubility in water limited its clinical application. In this study, we prepared PT-54 liposomes (PT-54-LPs) by the membrane hydration method to overcome this defect. The encapsulation efficiency (EE) and particle size were used as evaluation indicators to explore the preparation conditions of PT-54-LPs. The morphology, particle size, EE, drug loading content (DLC), drug release properties, and stability of PT-54-LPs were further investigated. In vitro drug release studies showed that PT-54-LPs exhibited significant slow-release properties compared with free PT-54. PT-54-LPs also showed better tumor inhibitory effects than free PT-54 without significant adverse effects. These results suggested that PT-54-LPs displayed sustained drug release and significantly improved the tumor selectivity of PT-54. Thus, PT-54-LPs showed significant promise in enhancing anticancer efficiency.
Collapse
Affiliation(s)
- Fang Ren
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Medical School Road, Yuzhong District, Chongqing, 400042, PR China
| | - Zongjie Gan
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Medical School Road, Yuzhong District, Chongqing, 400042, PR China
| | - Qianyu Zhang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Medical School Road, Yuzhong District, Chongqing, 400042, PR China
| | - Dan He
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Medical School Road, Yuzhong District, Chongqing, 400042, PR China
| | - Baoyan Chen
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Medical School Road, Yuzhong District, Chongqing, 400042, PR China
| | - Xianwei Wu
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Medical School Road, Yuzhong District, Chongqing, 400042, PR China
| | - Xiaolin Zeng
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Medical School Road, Yuzhong District, Chongqing, 400042, PR China
| | - Kexin Wu
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Medical School Road, Yuzhong District, Chongqing, 400042, PR China
| | - Yangchen Xing
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Medical School Road, Yuzhong District, Chongqing, 400042, PR China
| | - Yan Zhang
- Yaopharma Co, Ltd, No. 100, Xingguang Ave, Chongqing, 401121, China
| | - Huali Chen
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Medical School Road, Yuzhong District, Chongqing, 400042, PR China.
| |
Collapse
|
8
|
Graham W, Torbett-Dougherty M, Islam A, Soleimani S, Bruce-Tagoe TA, Johnson JA. Magnetic Nanoparticles and Drug Delivery Systems for Anti-Cancer Applications: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:285. [PMID: 39997849 PMCID: PMC11858650 DOI: 10.3390/nano15040285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Cancer continues to be a prominent fatal health issue worldwide, driving the urgent need for more effective treatment strategies. The pressing demand has sparked significant interest in the development of advanced drug delivery systems for chemotherapeutics. The advent of nanotechnology offers a groundbreaking approach, presenting a promising pathway to revolutionize cancer treatment and improve patient outcomes. Nanomedicine-based drug delivery systems have demonstrated the capability of improving the pharmacokinetic properties and accumulation of chemotherapeutic agents in cancer sites while minimizing the adverse side effects. Despite these advantages, most NDDSs exhibit only limited improvement in cancer treatment during clinical trials. The recent development of magnetic nanoparticles (MNPs) for biomedical applications has revealed a potential opportunity to further enhance the performance of NDDSs. The magnetic properties of MNPs can be utilized to increase the targeting capabilities of NDDSs, improve the controlled release of chemotherapeutic agents, and weaken the chemoresistance of tumors with magnetic hyperthermia. In this review, we will explore recent advancements in research for NDDSs for oncology applications, how MNPs and their properties can augment the capabilities of NDDSs when complexed with them and emphasize the challenges and safety concerns of incorporating these systems into cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Jacqueline Ann Johnson
- Department of Mechanical, Aerospace, and Biomedical Engineering, University of Tennessee, Knoxville, TN 37996, USA; (W.G.); (M.T.-D.); (A.I.); (S.S.); (T.A.B.-T.)
| |
Collapse
|
9
|
Tang Y, Shen Q, Lin P, Chen Z, Fan D, Zhuo M, Gan Y, Su Y, Qian Q, Lin L, Xue E, Chen Z. aPD-L1-facilitated theranostic and tumor microenvironment remodeling of pancreatic cancer via docetaxel-loaded phase-transformation nanoparticles triggered by low-intensity pulsed ultrasound. J Nanobiotechnology 2025; 23:48. [PMID: 39871305 PMCID: PMC11773723 DOI: 10.1186/s12951-025-03105-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025] Open
Abstract
Early diagnosis of pancreatic ductal adenocarcinoma (PDAC) is challenging because of its depth, which often leads to misdiagnosis during ultrasound examinations. The unique PDAC tumor microenvironment (TME) is characterized by significant fibrous tissue growth, and high interstitial pressure hinders drug penetration into tumors. Additionally, hypoxia and immune suppression within the tumor contribute to poor responses to radiotherapy and chemotherapy, ultimately leading to an unfavorable prognosis. In this study, aPD-L1-modified docetaxel and perfluoropentane-loaded liquid‒vapor phase-transformation lipid nanoparticles (aPDL1-DTX/PFP@Lipid) were synthesized and had an average diameter of 61.63 nm with 84.3% antibody modification. We demonstrated that the nanoparticles (NPs) exhibited excellent PDAC-targeting capabilities both in vitro and in vivo. Upon exposure to low-intensity pulsed ultrasound (LIPUS) stimulation, the NPs underwent a phase transformation to form microbubbles with substantial molecular ultrasound diagnostic effects, and combined treatment resulted in a tumor growth inhibition rate of 88.91%. This treatment strategy also led to the infiltration of CD8+ T cells, the downregulation of Treg cells, the promotion of M1 macrophage polarization, the inhibition of fibrosis to reduce tumor stromal pressure, and the facilitation of perfluoropentane (PFP) gasification to release O2 and improve tumor hypoxia. In conclusion, aPD-L1-modified liquid‒vapor phase-transformation nanoparticles loaded with docetaxel (DTX) and PFP were successfully combined with ultrasound for the molecular diagnosis and targeted treatment of PDAC. aPDL1-DTX/PFP@Lipid could reshape the PDAC TME, offering a new approach for ultrasound-mediated diagnosis and treatment with promising clinical applications.
Collapse
Affiliation(s)
- Yi Tang
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China
| | - Qingling Shen
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China
| | - Peng Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Zhixin Chen
- Fujian College Association Instrumental Analysis Center, Fuzhou University, 2 Xueyuan Road, Fuzhou, China
| | - Denghui Fan
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, China
| | - Minling Zhuo
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Yajiao Gan
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Yixi Su
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Qingfu Qian
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Liwu Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Ensheng Xue
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Zhikui Chen
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China.
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China.
| |
Collapse
|
10
|
Ciftci F, Özarslan AC, Kantarci İC, Yelkenci A, Tavukcuoglu O, Ghorbanpour M. Advances in Drug Targeting, Drug Delivery, and Nanotechnology Applications: Therapeutic Significance in Cancer Treatment. Pharmaceutics 2025; 17:121. [PMID: 39861768 PMCID: PMC11769154 DOI: 10.3390/pharmaceutics17010121] [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: 12/03/2024] [Revised: 01/01/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
In the 21st century, thanks to advances in biotechnology and developing pharmaceutical technology, significant progress is being made in effective drug design. Drug targeting aims to ensure that the drug acts only in the pathological area; it is defined as the ability to accumulate selectively and quantitatively in the target tissue or organ, regardless of the chemical structure of the active drug substance and the method of administration. With drug targeting, conventional, biotechnological and gene-derived drugs target the body's organs, tissues, and cells that can be selectively transported to specific regions. These systems serve as drug carriers and regulate the timing of release. Despite having many advantageous features, these systems have limitations in thoroughly treating complex diseases such as cancer. Therefore, combining these systems with nanoparticle technologies is imperative to treat cancer at both local and systemic levels effectively. The nanocarrier-based drug delivery method involves encapsulating target-specific drug molecules into polymeric or vesicular systems. Various drug delivery systems (DDS) were investigated and discussed in this review article. The first part discussed active and passive delivery systems, hydrogels, thermoplastics, microdevices and transdermal-based drug delivery systems. The second part discussed drug carrier systems in nanobiotechnology (carbon nanotubes, nanoparticles, coated, pegylated, solid lipid nanoparticles and smart polymeric nanogels). In the third part, drug targeting advantages were discussed, and finally, market research of commercial drugs used in cancer nanotechnological approaches was included.
Collapse
Affiliation(s)
- Fatih Ciftci
- Department of Biomedical Engineering, Faculty of Engineering, Fatih Sultan Mehmet Vakıf University, Istanbul 34015, Turkey
- Department of Technology Transfer Office, Fatih Sultan Mehmet Vakıf University, Istanbul 34015, Turkey
| | - Ali Can Özarslan
- Department of Metallurgical and Materials Engineering, Istanbul University-Cerrahpasa, Istanbul 34320, Turkey;
| | - İmran Cagri Kantarci
- Department of Bioengineering, Faculty of Chemistry-Metallurgy, Yildiz Technical University, Istanbul 34210, Turkey;
| | - Aslihan Yelkenci
- Department of Pediatric Dentistry, Faculty of Dentistry, University of Health Sciences, Istanbul 34668, Turkey;
| | - Ozlem Tavukcuoglu
- Department of Biochemistry, Faculty of Hamidiye Pharmacy, University of Health Sciences, Istanbul 34668, Turkey;
| | - Mansour Ghorbanpour
- Department of Medicinal Plants, Faculty of Agriculture and Natural Resources, Arak University, Arak 38156-8-8349, Iran;
| |
Collapse
|
11
|
Chen X, Li F, Cui B, Yan Q, Qiu C, Zhu Z, Wen L, Chen W. Liposomes-mediated enhanced antitumor effect of docetaxel with BRD4-PROTAC as synergist for breast cancer chemotherapy/immunotherapy. Int J Pharm 2025; 668:124973. [PMID: 39566696 DOI: 10.1016/j.ijpharm.2024.124973] [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: 07/31/2024] [Revised: 11/07/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
It has been reported that proteolysis-targeting chimeras (PROTACs) can effectively degrade intracellular oncogenic proteins, providing an ideal strategy for cancer treatment. ARV825, a bromodomain-containing protein 4 (BRD4)-PROTAC, has demonstrated the capacity to enhance the antitumor effect of the classic chemotherapeutic agent docetaxel (DTX). However, there are three major challenges to the broader in vivo application of ARV825: poor solubility, poor permeability, and off-target effects. Additionally, the efficient co-delivery of ARV825 and DTX to tumor tissues for a synergistic therapeutic effect remains unresolved. In this study, liposomes were utilized as co-delivery vehicles for ARV825 and DTX to effectively address these issues. The well-established liposomes significantly improved the solubility of both ARV825 and DTX while maintaining a sustained release profile in blood-mimetic conditions. The co-loaded liposomes accumulated in tumor tissues via the enhanced permeability and retention (EPR) effect. After internalization, ARV825 effectively degraded intracellular BRD4 proteins and downregulated the expression of both Bcl-2 and PD-L1 proteins, thereby increasing tumor cell apoptosis and enhancing the tumor immune response. This, in turn, augmented the antitumor effect of DTX in vivo without undesired side effects. In conclusion, BRD4-PROTAC may serve as a promising synergistic agent alongside the conventional chemotherapeutic agent DTX, with liposomes functioning as effective co-delivery vehicles.
Collapse
Affiliation(s)
- Xixi Chen
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Fang Li
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China
| | - Binghui Cui
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Qingyi Yan
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Caisheng Qiu
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Zengyan Zhu
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China
| | - Lijuan Wen
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China.
| | - Weiliang Chen
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China.
| |
Collapse
|
12
|
Oopkaew L, Injongkol Y, Kungwan N, Rungrotmongkol T. Theoretical investigation of structure and electronic properties in Cisplatin-citrate complexes. J Comput Chem 2025; 46:e27511. [PMID: 39644131 DOI: 10.1002/jcc.27511] [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: 03/16/2024] [Revised: 08/22/2024] [Accepted: 09/16/2024] [Indexed: 12/09/2024]
Abstract
Cisplatin (CDDP) is an effective Platinum (Pt) based anticancer drug used in chemotherapy. However, its effectiveness is limited due to its instability in solvents, along with the side effects it causes due to DNA damage. Nanoparticles (NPs) were developed in vitro to address these issues by loading CDDP into various types of NPs, including metal, lipid, and biological NPs. Citrate was employed as a biocompatible compound in nanomedicine to reduce cytotoxicity and enhance stability. In our study, the physicochemical and electronic properties of CDDP and citrate have been investigated using density functional theory (DFT), with a comparison of their behavior in water and DMSO. Additionally, TD-DFT was applied to analyze the UV-Vis spectra results. Six complexes have been proposed to better understand the interaction between citrate and CDDP. The results demonstrated that the CDDP could form stable complexes with citrate in both water and DMSO, and the considered complexes exhibited UV-Vis spectra within the experiment range. The frontier orbitals, electron densities mapping, and electrostatic potential analysis revealed that complex 5, where citrate di-substituted on two chlorides, is the most likely and effective complex. In summary, our investigation sheds light on the potential of CDDP-citrate complexes to address the limitations of CDDP, offering insights into their stability and interaction in solvents and highlighting the promising efficacy of specific complex formations for future therapeutic applications.
Collapse
Affiliation(s)
- Lipika Oopkaew
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Yuwanda Injongkol
- Futuristic Science Research Center, School of Science, Walailak University, Nakhon Si Thammarat, Thailand
- Functional Materials and Nanotechnology Center of Excellence, Walailak University, Nakhon Si Thammarat, Thailand
| | - Nawee Kungwan
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Materials Science and Technology, Chiang Mai University, Chiang Mai, Thailand
| | - Thanyada Rungrotmongkol
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
13
|
Matalqah S, Lafi Z, Mhaidat Q, Asha N, Yousef Asha S. 'Applications of machine learning in liposomal formulation and development'. Pharm Dev Technol 2025; 30:126-136. [PMID: 39780760 DOI: 10.1080/10837450.2024.2448777] [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: 10/12/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025]
Abstract
Machine learning (ML) has emerged as a transformative tool in drug delivery, particularly in the design and optimization of liposomal formulations. This review focuses on the intersection of ML and liposomal technology, highlighting how advanced algorithms are accelerating formulation processes, predicting key parameters, and enabling personalized therapies. ML-driven approaches are restructuring formulation development by optimizing liposome size, stability, and encapsulation efficiency while refining drug release profiles. Additionally, the integration of ML enhances therapeutic outcomes by enabling precision-targeted delivery and minimizing side effects. This review presents current breakthroughs, challenges, and future opportunities in applying ML to liposomal systems, aiming to improve therapeutic efficacy and patient outcomes in various disease treatments.
Collapse
Affiliation(s)
- Sina Matalqah
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Zainab Lafi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | | | | | | |
Collapse
|
14
|
Yin C, Wei ZJ, Long K, Sun M, Zhang Z, Wang Y, Wang W, Yuan Z. pH-Responsive Persistent Luminescent Nanosystem with Sensitized NIR Imaging and Ratiometric Imaging Modes for Tumor Surgery Navigation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:69071-69085. [PMID: 39648513 DOI: 10.1021/acsami.4c17747] [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: 12/10/2024]
Abstract
Owing to autofluorescence-free feature, persistent luminescent (PersL) nanoparticles (PLNPs) become potential materials for tumor surgical navigation. However, it is still challenging to enhance PersL intensity, contrast ratio, and imaging stability so as to meet clinical demand and avoid missed detection of microlesions. Herein, integrating a tumor microenvironment (TME)-responsive strategy, sensitization enhancement, and internal-standard ratiometric method, a dual-mode PersL imaging strategy is proposed: After loading pH-responsive fluorescent molecule Rh-ADM on PLNPs ZnGa2O4:Cr3+,Mn2+ (ZGCM-Rh8), the fluorescence resonance energy transfer (FRET) pathways between Cr3+ and Rh-ADM, as well as Mn2+ and Rh-ADM, could sensitize the NIR PersL emitted by Cr3+ and quench the green PersL from Mn2+ at acidic TME, respectively. As a result, ZGCM-Rh8 is endowed with WLED (white light LED)-excited NIR imaging mode and UV-excited ratiometric imaging mode. Under WLED, ZGCM-Rh8 realizes 4.5-fold PersL enhancement and 97.9 as the maximum tumor contrast after precise control of Rh-ADM contents, helping with the preoperative diagnosis of deep lesions. Under UV, ZGCM-Rh8 conducts ratiometric PersL imaging steadily, and the "NIR/Vis" ratios at the tumor keep larger than 110, succeeding in detecting out a 1.5 mm small lesion and serving thorough surgical elimination of H22 ectopic intramuscular tumor in balb/c mice. To our knowledge, ZGCM-Rh8 is the first to realize pH-responsive PersL sensitization and apply ratiometric PersL imaging technology to surgical navigation.
Collapse
Affiliation(s)
- Chang Yin
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zi-Jin Wei
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Kai Long
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Mengjie Sun
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhouyu Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yifei Wang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Wei Wang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhi Yuan
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| |
Collapse
|
15
|
Feng L, Wei R, Wu J, Chen X, Wen Y, Chen J. Cyclodextrin Drugs in Liposomes: Preparation and Application of Anticancer Drug Carriers. AAPS PharmSciTech 2024; 26:3. [PMID: 39638889 DOI: 10.1208/s12249-024-02999-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Cyclodextrin complexes have been widely used in pharmaceutical applications, but disadvantages such as the rapid clearance of cyclodextrins from the blood stream after in vivo administration or their replacement by other molecules in the biological medium with higher luminal affinity for cyclodextrins limit the application of cyclodextrins as drug carriers. Liposome-encapsulated hydrophobic drugs have low and unstable drug loading rates. Drug-in-CD-in-liposome (DCL), which encapsulate cyclodextrin inclusion complexes into liposomes, combine the advantages of both delivery systems, can effectively avoid the leakage and rapid release of lipophilic drugs in the lipid bilayer, and help to maintain the integrity of liposomes. This paper focuses on the preparation method, characterization and application of DCL, with a view to providing methods and references for the research and application of DCL technology.
Collapse
Affiliation(s)
- Lanni Feng
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Shanghai WeiEr Lab, Shanghai, 201707, China
| | - Ruting Wei
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Shanghai WeiEr Lab, Shanghai, 201707, China
| | - Jiali Wu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Shanghai WeiEr Lab, Shanghai, 201707, China
| | | | - Yan Wen
- Department of Pharmacy, Changzheng Hospital, Naval Medical University, No.415, Fengyang Road, Shanghai, 200003, China
| | - Jianming Chen
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
- Shanghai WeiEr Lab, Shanghai, 201707, China.
| |
Collapse
|
16
|
Jourdain MA, Eyer J. Recent advances in liposomes and peptide-based therapeutics for glioblastoma treatment. J Control Release 2024; 376:732-752. [PMID: 39437968 DOI: 10.1016/j.jconrel.2024.10.037] [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: 02/07/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
In the context of glioblastoma treatment, the penetration of drugs is drastically limited by the blood-brain-barrier (BBB). Emerging therapies have focused on the field of therapeutic peptides for their excellent BBB targeting properties that promote a deep tumor penetration. Peptide-based strategies are also renowned for their abilities of driving cargo such as liposomal system allowing an active targeting of receptors overexpressed on GBM cells. This review provides a detailed description of the internalization mechanisms of specific GBM homing and penetrating peptides as well as the latest in vitro/in vivo studies of liposomes functionalized with them. The purpose of this review is to summarize a selection of promising pre-clinical results that demonstrate the advantages of this nanosystem, including an increase of tumor cell targeting, triggering drug accumulation and thus a strong antitumor effect. Aware of the early stage of these studies, many challenges need to be overcome to promote peptide-directed liposome at clinical level. In particular, the lack of suitable production, the difficulty to characterize the nanosystem and therapeutic competition leaded by antibodies.
Collapse
Affiliation(s)
- M-A Jourdain
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France.
| | - J Eyer
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| |
Collapse
|
17
|
Liu L, Yang M, Chen Z. Surface functionalized nanomaterial systems for targeted therapy of endocrine related tumors: a review of recent advancements. Drug Deliv 2024; 31:2390022. [PMID: 39138394 PMCID: PMC11328606 DOI: 10.1080/10717544.2024.2390022] [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: 03/19/2024] [Revised: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024] Open
Abstract
The application of multidisciplinary techniques in the management of endocrine-related cancers is crucial for harnessing the advantages of multiple disciplines and their coordinated efforts in eliminating tumors. Due to the malignant characteristics of cancer cells, they possess the capacity to develop resistance to traditional treatments such as chemotherapy and radiotherapy. Nevertheless, despite diligent endeavors to enhance the prediction of outcomes, the overall survival rate for individuals afflicted with endocrine-related malignancy remains quite miserable. Hence, it is imperative to investigate innovative therapy strategies. The latest advancements in therapeutic tactics have offered novel approaches for the therapy of various endocrine tumors. This paper examines the advancements in nano-drug delivery techniques and the utilization of nanomaterials for precise cancer cures through targeted therapy. This review provides a thorough analysis of the potential of combined drug delivery strategies in the treatment of thyroid cancer, adrenal gland tumors, and pancreatic cancer. The objective of this study is to gain a deeper understanding of current therapeutic approaches, stimulate the development of new drug DDS, and improve the effectiveness of treatment for patients with these diseases. The intracellular uptake of pharmaceuticals into cancer cells can be significantly improved through the implantation of synthetic or natural substances into nanoparticles, resulting in a substantial reduction in the development of endocrine malignancies.
Collapse
Affiliation(s)
- Limei Liu
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Miao Yang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ziyang Chen
- Department of Gastroenterology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
18
|
Han H, Santos HA. Nano- and Micro-Platforms in Therapeutic Proteins Delivery for Cancer Therapy: Materials and Strategies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409522. [PMID: 39263818 DOI: 10.1002/adma.202409522] [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: 07/03/2024] [Revised: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Proteins have emerged as promising therapeutics in oncology due to their great specificity. Many treatment strategies are developed based on protein biologics, such as immunotherapy, starvation therapy, and pro-apoptosis therapy, while some protein biologics have entered the clinics. However, clinical translation is severely impeded by instability, short circulation time, poor transmembrane transportation, and immunogenicity. Micro- and nano-particles-based drug delivery platforms are designed to solve those problems and enhance protein therapeutic efficacy. This review first summarizes the different types of therapeutic proteins in clinical and research stages, highlighting their administration limitations. Next, various types of micro- and nano-particles are described to demonstrate how they can overcome those limitations. The potential of micro- and nano-particles are then explored to enhance the therapeutic efficacy of proteins by combinational therapies. Finally, the challenges and future directions of protein biologics carriers are discussed for optimized protein delivery.
Collapse
Affiliation(s)
- Huijie Han
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
19
|
Jäck N, Hemming A, Hartmann L. Synthesis of Dual-Responsive Amphiphilic Glycomacromolecules: Controlled Release of Glycan Ligands via pH and UV Stimuli. Macromol Rapid Commun 2024; 45:e2400439. [PMID: 39037337 DOI: 10.1002/marc.202400439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 06/25/2024] [Indexed: 07/23/2024]
Abstract
This work presents a versatile strategy for the synthesis of dual stimuli-responsive amphiphilic glycomacromolecules with tailored release properties. Amphiphilic precision glycomacromolecules (APGs) derived from tailor-made building blocks using solid phase polymer synthesis form glycofunctionalized micelles, a versatile class of materials with applications in drug delivery, as antiinfection agents as well as simple cell mimetics. In this work, this concept is extended by integrating cleavable building blocks into APGs now allowing stimuli-responsive release of glycan ligands or destruction of the micelles. This study incorporates a newly designed acid-labile building block, 4-(4-(((((9H-fluoren-9-yl)methoxy)carbonyl)amino)methyl)-1,3-dioxolan-2-yl)benzoic acid (DBA), suitable also for other types of solid phase or amide chemistry, and an established UV-cleavable 2-nitrobenzyl linker (PL). The results demonstrate that both linkers can be cleaved independently and thus allow dual stimuli-responsive release from the APG micelles. By choosing the APG design e.g., placing the cleavable linkers between glycomacromolecular blocks presenting different types of carbohydrates, they can tune APG and micellar stability as well as the interaction and cluster formation with a carbohydrate-recognizing lectin. Such dual-responsive glycofunctionalized micelles have wide potential for use in drug delivery applications or for the development as anti-adhesion agents in antiviral and antibacterial treatments.
Collapse
Affiliation(s)
- Nicholas Jäck
- Institute of Macromolecular Chemistry, Albert-Ludwigs-Universität Freiburg, Stefan-Meier-Str. 31, 79104, Freiburg, Germany
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
- Cluster of Excellence livMatS @ FIT - Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Georges-Köhler-Allee 105, D-79110, Freiburg, Germany
| | - Arne Hemming
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Laura Hartmann
- Institute of Macromolecular Chemistry, Albert-Ludwigs-Universität Freiburg, Stefan-Meier-Str. 31, 79104, Freiburg, Germany
- Cluster of Excellence livMatS @ FIT - Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Georges-Köhler-Allee 105, D-79110, Freiburg, Germany
| |
Collapse
|
20
|
He Y, Feng Y, Qiu D, Lin M, Jin H, Hu Z, Huang X, Ma S, He Y, Lai M, Jin W, Liu J. Regulation of IFP in solid tumours through acoustic pressure to enhance infiltration of nanoparticles of various sizes. J Drug Target 2024; 32:964-976. [PMID: 38884143 DOI: 10.1080/1061186x.2024.2367579] [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: 02/16/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Numerous nanomedicines have been developed recently that can accumulate selectively in tumours due to the enhanced permeability and retention (EPR) effect. However, the high interstitial fluid pressure (IFP) in solid tumours limits the targeted delivery of nanomedicines. We were previously able to relieve intra-tumoural IFP by low-frequency non-focused ultrasound (LFNFU) through ultrasonic targeted microbubble destruction (UTMD), improving the targeted delivery of FITC-dextran. However, the accumulation of nanoparticles of different sizes and the optimal acoustic pressure were not evaluated. In this study, we synthesised Cy5.5-conjugated mesoporous silica nanoparticles (Cy5.5-MSNs) of different sizes using a one-pot method. The Cy5.5-MSNs exhibited excellent stability and biosafety regardless of size. MCF7 tumour-bearing mice were subjected to UTMD over a range of acoustic pressures (0.5, 0.8, 1.5 and 2.0 MPa), and injected intravenously with Cy5.5-MSNs. Blood perfusion, tumour IFP and intra-tumoural accumulation of Cy5.5-MSNs were analysed. Blood perfusion and IFP initially rose, and then declined, as acoustic pressure intensified. Furthermore, UTMD significantly enhanced the accumulation of differentially sized Cy5.5-MSNs in tumour tissues compared to that of the control group, and the increase was sevenfold higher at an acoustic pressure of 1.5 MPa. Taken together, UTMD enhanced the infiltration and accumulation of Cy5.5-MSNs of different sizes in solid tumours by reducing intra-tumour IFP.
Collapse
Affiliation(s)
- Yangcheng He
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Yuyi Feng
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Danxai Qiu
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - MinHua Lin
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Hai Jin
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Zhiwen Hu
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Xue Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Suihong Ma
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Yan He
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Meiqi Lai
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Wenhui Jin
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Jianhua Liu
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| |
Collapse
|
21
|
Hart M, Isuri RK, Ramos D, Osharovich SA, Rodriguez AE, Harmsen S, Dudek GC, Huck JL, Holt DE, Popov AV, Singhal S, Delikatny EJ. Non-Small Cell Lung Cancer Imaging Using a Phospholipase A2 Activatable Fluorophore. CHEMICAL & BIOMEDICAL IMAGING 2024; 2:490-500. [PMID: 39056064 PMCID: PMC11267604 DOI: 10.1021/cbmi.4c00026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 07/28/2024]
Abstract
Lung cancer, the most common cause of cancer-related death in the United States, requires advanced intraoperative detection methods to improve evaluation of surgical margins. In this study we employed DDAO-arachidonate (DDAO-A), a phospholipase A2 (PLA2) activatable fluorophore, designed for the specific optical identification of lung cancers in real-time during surgery. The in vitro fluorescence activation of DDAO-A by porcine sPLA2 was tested in various liposomal formulations, with 100 nm extruded EggPC showing the best overall characteristics. Extruded EggPC liposomes containing DDAO-A were tested for their stability under various storage conditions, demonstrating excellent stability for up to 4 weeks when stored at -20 °C or below. Cell studies using KLN 205 and LLC1 lung cancer cell lines showed DDAO-A activation was proportional to cell number. DDAO-A showed preferential activation by human recombinant cPLA2, an isoform highly specific to arachidonic acid-containing lipids, when compared to a control probe, DDAO palmitate (DDAO-P). In vivo studies with DBA/2 mice bearing KLN 205 lung tumors recapitulated these results, with preferential activation of DDAO-A relative to DDAO-P following intratumoral injection. Topical application of DDAO-A-containing liposomes to human (n = 10) and canine (n = 3) lung cancers ex vivo demonstrated the preferential activation of DDAO-A in tumor tissue relative to adjacent normal lung tissue, with fluorescent tumor-to-normal ratios (TNR) of up to 5.2:1. The combined results highlight DDAO-A as a promising candidate for clinical applications, showcasing its potential utility in intraoperative and back-table imaging and topical administration during lung cancer surgeries. By addressing the challenge of residual microscopic disease at resection margins and offering stability in liposomal formulations, DDAO-A emerges as a potentially valuable tool for advancing precision lung cancer surgery and improving curative resection rates.
Collapse
Affiliation(s)
- Michael
C. Hart
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ritesh K. Isuri
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department
of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104, United States
| | - Drew Ramos
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sofya A. Osharovich
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Andrea E. Rodriguez
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Stefan Harmsen
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Grace C. Dudek
- Department
of Biology, University of Pennsylvania, 102 Leidy Laboratories 433 S University
Ave, Philadelphia, Pennsylvania 19104, United States
| | - Jennifer L. Huck
- Department
of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - David E. Holt
- Department
of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Anatoliy V. Popov
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sunil Singhal
- Department
of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Edward J. Delikatny
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
22
|
Nguyen-Huu AM, Le NTT, Vo Do MH, Dong Yen PN, Nguyen-Dinh TD, Nguyen NH, Nguyen DH. Development and Characterization of Quercetin-Loaded Polymeric Liposomes with Gelatin-Poly(ethylene glycol)-Folic Acid Coating to Increase Their Long-Circulating and Anticancer Activity. ACS APPLIED BIO MATERIALS 2024; 7:4454-4470. [PMID: 38857443 DOI: 10.1021/acsabm.4c00334] [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: 06/12/2024]
Abstract
Liposomes as drug-delivery systems have been researched and applied in multiple scientific reports and introduced as patented products with interesting therapeutic properties. Despite various advantages, this drug carrier faces major difficulties in its innate stability, cancer cell specificity, and control over the release of hydrophobic drugs, particularly quercetin, a naturally derived drug that carries many desirable characteristics for anticancer treatment. To improve the effectiveness of liposomes to deliver quercetin by tackling and mitigating the mentioned hurdles, we developed a strategy to establish the ability to passively target cancerous cells, as well as to increase the bioavailability of loaded drugs by incorporating poly(ethylene glycol), gelatin, and folic acid moieties to modify the liposomal system's surface. This research developed a chemically synthesized gelatin, poly(ethylene glycol), and folic acid as a single polymer to coat drug-loaded liposome systems. Liposomes were coated with gelatin-poly(ethylene glycol)-folic acid by electrostatic interaction, characterized by their size, morphology, ζ potential, drug loading efficiency, infrared structures, differential scanning calorimetry spectra, and drug-releasing profiles, and then evaluated for their cytotoxicity to MCF-7 breast cancer cells, as well as cellular uptake, analyzed by confocal imaging to further elaborate on the in vitro behavior of the coated liposome. The results indicated an unusual change in size with increased coating materials, followed by increased colloidal stability, ζ potential, and improved cytotoxicity to cancer cells, as shown by the cellular viability test with MCF-7. Cellular uptake also confirmed these results, providing data for the effects of biopolymer coating, while confirming that folic acid can increase the uptake of liposome by cancer cells. In consideration of such results, the modified gelatin-poly(ethylene glycol)-folic acid-coated liposome can be a potential system in delivering the assigned anticancer compound. This modified biopolymer showed excellent properties as a coating material and should be considered for further practical applications in the future.
Collapse
Affiliation(s)
- Anh-Minh Nguyen-Huu
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
- Department of Chemical and Material Engineering, Vlab, New Jersey Institute of Technology, Newark, New Jersey 07001, United States
| | - Ngoc Thuy Trang Le
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City 100000, Vietnam
| | - Minh Hoang Vo Do
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
| | - Pham Nguyen Dong Yen
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
| | - Tien-Dung Nguyen-Dinh
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City 100000, Vietnam
| | - Ngoc Hoi Nguyen
- Institute of Chemical Technology, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City 100000, Vietnam
| | - Dai Hai Nguyen
- Institute of Chemical Technology, Vietnam Academy of Science and Technology, Ho Chi Minh City 70000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City 100000, Vietnam
| |
Collapse
|
23
|
Kalra J, Baker J, Sun X, Kyle A, Minchinton A, Bally MB. Accumulation of liposomes in metastatic tumor sites is not necessary for anti-cancer drug efficacy. J Transl Med 2024; 22:621. [PMID: 38961395 PMCID: PMC11223361 DOI: 10.1186/s12967-024-05428-9] [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: 02/27/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND The tumor microenvironment is profoundly heterogeneous particularly when comparing sites of metastases. Establishing the extent of this heterogeneity may provide guidance on how best to design lipid-based drug delivery systems to treat metastatic disease. Building on our previous research, the current study employs a murine model of metastatic cancer to explore the distribution of ~ 100 nm liposomes. METHODS Female NCr nude mice were inoculated with a fluorescently labeled, Her2/neu-positive, trastuzumab-resistant breast cancer cell line, JIMT-1mkate, either in the mammary fat pad to create an orthotopic tumor (OT), or via intracardiac injection (IC) to establish tumors throughout the body. Animals were dosed with fluorescent and radio-labeled liposomes. In vivo and ex vivo fluorescent imaging was used to track liposome distribution over a period of 48 h. Liposome distribution in orthotopic tumors was compared to sites of tumor growth that arose following IC injection. RESULTS A significant amount of inter-vessel heterogeneity for DiR distribution was observed, with most tumor blood vessels showing little to no presence of the DiR-labelled liposomes. Further, there was limited extravascular distribution of DiR liposomes in the perivascular regions around DiR-positive vessels. While all OT tumors contained at least some DiR-positive vessels, many metastases had very little or none. Despite the apparent limited distribution of liposomes within metastases, two liposomal drug formulations, Irinophore C and Doxil, showed similar efficacy for both the OT and IC JIMT-1mkate models. CONCLUSION These findings suggest that liposomal formulations achieve therapeutic benefits through mechanisms that extend beyond the enhanced permeability and retention effect.
Collapse
Affiliation(s)
- Jessica Kalra
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada.
| | - Jennifer Baker
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - XuXin Sun
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Alastair Kyle
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Andrew Minchinton
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marcel B Bally
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- NanoMedicine Innovation Network, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
Moon Y, Cho H, Kim K. Nano-Delivery of Immunogenic Cell Death Inducers and Immune Checkpoint Blockade Agents: Single-Nanostructure Strategies for Enhancing Immunotherapy. Pharmaceutics 2024; 16:795. [PMID: 38931916 PMCID: PMC11207855 DOI: 10.3390/pharmaceutics16060795] [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: 05/20/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Cancer immunotherapy has revolutionized oncology by harnessing the patient's immune system to target and eliminate cancer cells. However, immune checkpoint blockades (ICBs) face limitations such as low response rates, particularly in immunologically 'cold' tumors. Enhancing tumor immunogenicity through immunogenic cell death (ICD) inducers and advanced drug delivery systems represents a promising solution. This review discusses the development and application of various nanocarriers, including polymeric nanoparticles, liposomes, peptide-based nanoparticles, and inorganic nanoparticles, designed to deliver ICD inducers and ICBs effectively. These nanocarriers improve therapeutic outcomes by converting cold tumors into hot tumors, thus enhancing immune responses and reducing systemic toxicity. By focusing on single-nanoparticle systems that co-deliver both ICD inducers and ICBs, this review highlights their potential in achieving higher drug concentrations at tumor sites, improving pharmacokinetics and pharmacodynamics, and facilitating clinical translation. Future research should aim to optimize these nanocarrier systems for better in vivo performance and clinical applications, ultimately advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Yujeong Moon
- Department of Bioengineering, Korea University, Seoul 02841, Republic of Korea;
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hanhee Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea;
| | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea;
| |
Collapse
|
25
|
Zhao M, Zhu X, Li B, Yan C, Wu C, He L, Cao J, Lu F, Chen H, Li W. Potent cancer therapy by liposome microstructure tailoring with active-to-passive targeting and shell-to-core thermosensitive features. Mater Today Bio 2024; 26:101035. [PMID: 38586871 PMCID: PMC10995888 DOI: 10.1016/j.mtbio.2024.101035] [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: 11/29/2023] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024] Open
Abstract
Liposomes have been widely studied as drug carriers for clinical application, and the key issue is how to achieve effective delivery through targeting strategies. Even though certain cell-level targeting or EPR effect designs have been developed, reaching sufficient drug concentration in intracellular regions remains a challenge due to the singularity of functionality. Herein, benefiting from the unique features of tumor from tissue to cell, a dual-thermosensitive and dual-targeting liposome (DTSL) was creatively fabricated through fine microstructure tailoring, which holds intelligent both tissue-regulated active-to-passive binding and membrane-derived homologous-fusion (HF) properties. At the micro level, DTSL can actively capture tumor cells and accompany the enhanced HF effect stimulated by self-constriction, which achieves a synergistic promotion effect targeting tissues to cells. As a result, this first active-then passive targeting process makes drug delivery more accurate and effective, and after dynamic targeting into cells, the nucleus of DTSL undergoes further thermally responsive contraction, fully releasing internal drugs. In vivo experiments showed that liposomes with dual targeting and dual thermosensitive features almost completely inhibited tumor growth. Summarized, these results provide a reference for a rational design and microstructural tailoring of the liposomal co-delivery system of drugs, suggesting that active-to-passive dual-targeting DTSL can function as a new strategy for cancer treatment.
Collapse
Affiliation(s)
- Mengxin Zhao
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
| | - Xiaodong Zhu
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
| | - Bailing Li
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chenyang Yan
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
| | - Cong Wu
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
| | - Lei He
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jingyi Cao
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Fanglin Lu
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Han Chen
- Department of General Surgery, 905th Hospital of People's Liberation Army Navy, Naval Medical University, Shanghai, 200433, China
| | - Wei Li
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
26
|
Peddinti V, Rout B, Agnihotri TG, Gomte SS, Jain A. Functionalized liposomes: an enticing nanocarrier for management of glioma. J Liposome Res 2024; 34:349-367. [PMID: 37855432 DOI: 10.1080/08982104.2023.2270060] [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: 06/06/2023] [Accepted: 10/07/2023] [Indexed: 10/20/2023]
Abstract
Glioma is one of the most severe central nervous systems (CNS)-specific tumors, with rapidly growing malignant glial cells accounting for roughly half of all brain tumors and having a poor survival rate ranging from 12 to 15 months. Despite being the most often used technique for glioma therapy, conventional chemotherapy suffers from low permeability of the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) to anticancer drugs. When it comes to nanocarriers, liposomes are thought of as one of the most promising nanocarrier systems for glioma treatment. However, owing to BBB tight junctions, non-targeted liposomes, which passively accumulate in most cancer cells primarily via the increased permeability and retention effect (EPR), would not be suitable for glioma treatment. The surface modification of liposomes with various active targeting ligands has shown encouraging outcomes in the recent times by allowing various chemotherapy drugs to pass across the BBB and BBTB and enter glioma cells. This review article introduces by briefly outlining the landscape of glioma, its classification, and some of the pathogenic causes. Further, it discusses major barriers for delivering drugs to glioma such as the BBB, BBTB, and tumor microenvironment. It further discusses modified liposomes such as long-acting circulating liposomes, actively targeted liposomes, stimuli responsive liposomes. Finally, it highlighted the limitations of liposomes in the treatment of glioma and the various actively targeted liposomes undergoing clinical trials for the treatment of glioma.
Collapse
Affiliation(s)
- Vasu Peddinti
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Biswajit Rout
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| |
Collapse
|
27
|
Fidan Y, Muçaj S, Timur SS, Gürsoy RN. Recent advances in liposome-based targeted cancer therapy. J Liposome Res 2024; 34:316-334. [PMID: 37814217 DOI: 10.1080/08982104.2023.2268710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 09/29/2023] [Indexed: 10/11/2023]
Abstract
Nano-drug delivery systems have opened new pathways for tumor treatment by overcoming some of the limitations of conventional drugs, such as physiological degradation, short half-life, and rapid release. Liposomes are promising nanocarrier systems due to their biocompatibility, low toxicity, and high inclusivity, as well as their enhanced drug bioavailability. Various strategies for active targeting of liposomal formulations have been investigated to achieve the highest drug efficacy. This review aims to summarize current developments in novel liposomal formulations, particularly ligand-targeted liposomes (such as folate, transferrin, hyaluronic acid, antibodies, aptamer, and peptide, etc.) used for the therapy of various cancers and provide an insight on the challenges and future of liposomes for scientists and pharmaceutical companies.
Collapse
Affiliation(s)
- Yeliz Fidan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Stela Muçaj
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Selin Seda Timur
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - R Neslihan Gürsoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
28
|
Liu J, Han Y, Zhao M, Wang L, Hu H, Chen D. Unlocking the power of immunotherapy: Combinatorial delivery of plasmid IL-15 and gemcitabine to synergistically remodeling the tumor microenvironment. Int J Pharm 2024; 655:124027. [PMID: 38554742 DOI: 10.1016/j.ijpharm.2024.124027] [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: 11/30/2023] [Revised: 03/03/2024] [Accepted: 03/19/2024] [Indexed: 04/02/2024]
Abstract
Cancer immunotherapy has emerged as a promising clinical treatment strategy in recent years. Unfortunately, the satisfactory antitumor therapeutic efficacy of immunotherapy is limited by intricate immunosuppressive tumor microenvironment (ITM). To remodel the ITM and alleviate the immune evasion, we constructed FA-PEG-modified liposomes to deliver plasmid IL-15 (pIL-15) and gemcitabine (GEM) (FPCL@pIL-15 + FPGL), respectively. The FPCL@pIL-15 (150 nm) and FPGL (120 nm) exhibited symmetrically spherical structures as well as desirable penetration and accumulation on tumor tissue depending on folic acid (FA) specialized targeting function. The transfected expression of IL-15 efficiently fosters the proliferation and co-activation of Natural killer (NK) cells and CD8+T cells through binding to IL-15R. FPGL upregulated the expression of Natural killer group 2 member D ligands (NKG2DLs) and reinforced recognition by NK cells to alleviate the immune evasion, and simultaneously promoted activation of CD8+T cells through immunogenic cell death (ICD) effects. More importantly, the combinatorial administration achieved intended anti-tumor efficacy in the subcutaneous 4T1 tumor model. In essence, we demonstrated that combining FPCL@pIL-15 with FPGL synergistically stimulates and mobilizes the immune system to reverse the ITM and trigger an anti-tumor immune response, indicating a tremendous potential for application in immunotherapy.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Yanyan Han
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Ming Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Leyuan Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Haiyang Hu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China.
| | - Dawei Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China.
| |
Collapse
|
29
|
Ma W, Zhao Q, Zhu S, Wang X, Zhang C, Ma D, Li N, Yin Y. Construction of glutathione-responsive paclitaxel prodrug nanoparticles for image-guided targeted delivery and breast cancer therapy. RSC Adv 2024; 14:12796-12806. [PMID: 38645515 PMCID: PMC11027725 DOI: 10.1039/d4ra00610k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/29/2024] [Indexed: 04/23/2024] Open
Abstract
Paclitaxel (PTX) remains an essential drug in the treatment of breast cancer. To improve metabolic stability and real-time monitoring of drug location, we develop a visualized nano-prodrug. Novel hyaluronic acid (HA)-coated glutathione (GSH)-sensitive chitosan (CS)-based nano-prodrug (HA/TPE-CS-SS-PTX NPs) with aggregation-induced emission effects (AIE) were accomplished. The prodrug NPs (drug loading 29.32%, particle size 105 nm, regular sphericity) exhibit excellent fluorescence stability. The prodrug NPs could target tumor cells with high expression of CD44 and decompose in the presence of high concentrations of glutathione. In vitro evaluations revealed that the prodrug NPs have significant cytotoxicity on 4T1 cells, and due to their excellent AIE characteristics, their position in cells can be tracked. Moreover, the prodrug NPs also shown superior anti-tumor effects in vivo experimental. Overall, the HA/TPE-CS-SS-PTX NPs we constructed have excellent bio-imaging capabilities and can be served as a potential nanomedicine for PTX delivery against breast cancer.
Collapse
Affiliation(s)
- Weiwei Ma
- School of Pharmacy, Xinxiang Medical University Henan 453003 P. R. China
| | - Qiufeng Zhao
- School of Pharmacy, Xinxiang Medical University Henan 453003 P. R. China
| | - Shilong Zhu
- School of Pharmacy, Xinxiang Medical University Henan 453003 P. R. China
| | - Xinyue Wang
- School of Pharmacy, Xinxiang Medical University Henan 453003 P. R. China
| | - Chuangchuang Zhang
- School of Pharmacy, Xinxiang Medical University Henan 453003 P. R. China
| | - Daming Ma
- School of Pharmacy, Xinxiang Medical University Henan 453003 P. R. China
| | - Na Li
- School of Pharmacy, Xinxiang Medical University Henan 453003 P. R. China
| | - Yanyan Yin
- School of Pharmacy, Xinxiang Medical University Henan 453003 P. R. China
| |
Collapse
|
30
|
Huang J, Chen J. Pharmacokinetics and pharmacodynamic evaluation of hyaluronic acid-modified imatinib-loaded PEGylated liposomes in CD44-positive Gist882 tumor-bearing mice. J Liposome Res 2024; 34:97-112. [PMID: 37401372 DOI: 10.1080/08982104.2023.2228888] [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: 05/02/2023] [Accepted: 06/14/2023] [Indexed: 07/05/2023]
Abstract
To develop a PEGylated and CD44-targeted liposomes, enabled by surface coating with hyaluronic acid (HA) via amide bond to improve the efficacy of imatinib mesylate (IM), for tumor-targeted cytoplasmic drug delivery. HA was covalently grafted on DSPE-PEG2000-NH2 polymer. HA-modified or unmodified PEGylated liposomes were prepared with ethanol injection method, and the stability, drug release, and cytotoxicity of these liposomes were studied. Meanwhile, intracellular drug delivery efficiency, antitumor efficacy, and pharmacokinetics were also investigated. Ex vivo fluorescence biodistribution was also detected by small animal imaging. In addition, endocytosis mechanism was also explored HA-coated PEGylated liposomes (137.5 nm ± 10.24) had a negative zeta potential (-29.3 mV ± 5.44) and high drug loading (27.8%, w/w). The liposomes were stable with cumulative drug leakage (<60%) under physiological conditions. Blank liposomes were nontoxic to Gist882 cells, and IM-loaded liposomes had higher cytotoxicity to Gist882 cells. HA-modified PEGylated liposomes were internalized more effectively than non-HA coating via CD44-mediated endocytosis. Besides, the cellular uptake of HA-modified liposomes also partly depends on caveolin-medicated endocytosis and micropinocytosis. In rats, both liposomes produced a prolonged half-life of IM (HA/Lp/IM: 14.97h; Lp/IM: 11.15h) by 3- to 4.5-folds compared with the IM solution (3.61h). HA-decorated PEGylated liposomes encapsulated IM exhibited strong inhibitory effect on tumor growth in Gist882 cell-bearing nude mice and formation of 2D/3D tumor spheroids. The Ki67 immunohistochemistry result was consistent with the above results. IM-loaded PEGylated liposomes modified with HA exerted the excellent anti-tumor effect on tumor-bearing mice and more drugs accumulated into the tumor site.
Collapse
Affiliation(s)
- Ju Huang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, China
| | - Jian Chen
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| |
Collapse
|
31
|
Bandyopadhyay S, Zhao Z, East AK, Hernandez RT, Forzano JA, Shapiro BA, Yadav AK, Swartchick CB, Chan J. Activity-Based Nitric Oxide-Responsive Porphyrin for Site-Selective and Nascent Cancer Ablation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:9680-9689. [PMID: 38364813 DOI: 10.1021/acsami.3c15604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
Nitric oxide (NO) generated within the tumor microenvironment is an established driver of cancer progression and metastasis. Recent efforts have focused on leveraging this feature to target cancer through the development of diagnostic imaging agents and activatable chemotherapeutics. In this context, porphyrins represent an extraordinarily promising class of molecules, owing to their demonstrated use within both modalities. However, the remodeling of a standard porphyrin to afford a responsive chemical that can distinguish elevated NO from physiological levels has remained a significant research challenge. In this study, we employed a photoinduced electron transfer strategy to develop a panel of NO-activatable porphyrin photosensitizers (NOxPorfins) augmented with real-time fluorescence monitoring capabilities. The lead compound, NOxPorfin-1, features an o-phenylenediamine trigger that can effectively capture NO (via N2O3) to yield a triazole product that exhibits a 7.5-fold enhancement and a 70-fold turn-on response in the singlet oxygen quantum yield and fluorescence signal, respectively. Beyond demonstrating excellent in vitro responsiveness and selectivity toward NO, we showcase the potent photodynamic therapy (PDT) effect of NOxPorfin-1 in murine breast cancer and human non-small cellular lung cancer cells. Further, to highlight the in vivo efficacy, two key studies were executed. First, we utilized NOxPorfin-1 to ablate murine breast tumors in a site-selective manner without causing substantial collateral damage to healthy tissue. Second, we established a nascent human lung cancer model to demonstrate the unprecedented ability of NOxPorfin-1 to halt tumor growth and progression completely. The results of the latter study have tremendous implications for applying PDT to target metastatic lesions.
Collapse
Affiliation(s)
- Suritra Bandyopadhyay
- Department of Chemistry, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology and Cancer Center at Illinois, University of Illinois at Urbana─Champaign, 405 N. Mathews Avenue, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Zhenxiang Zhao
- Department of Chemistry, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology and Cancer Center at Illinois, University of Illinois at Urbana─Champaign, 405 N. Mathews Avenue, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Amanda K East
- Department of Chemistry, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology and Cancer Center at Illinois, University of Illinois at Urbana─Champaign, 405 N. Mathews Avenue, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Rodrigo Tapia Hernandez
- Department of Chemistry, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology and Cancer Center at Illinois, University of Illinois at Urbana─Champaign, 405 N. Mathews Avenue, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Joseph A Forzano
- Department of Chemistry, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology and Cancer Center at Illinois, University of Illinois at Urbana─Champaign, 405 N. Mathews Avenue, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Benjamin A Shapiro
- Department of Chemistry, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology and Cancer Center at Illinois, University of Illinois at Urbana─Champaign, 405 N. Mathews Avenue, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Anuj K Yadav
- Department of Chemistry, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology and Cancer Center at Illinois, University of Illinois at Urbana─Champaign, 405 N. Mathews Avenue, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Chelsea B Swartchick
- Department of Chemistry, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology and Cancer Center at Illinois, University of Illinois at Urbana─Champaign, 405 N. Mathews Avenue, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Jefferson Chan
- Department of Chemistry, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology and Cancer Center at Illinois, University of Illinois at Urbana─Champaign, 405 N. Mathews Avenue, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
32
|
Ren L, Liu S, Zhong J, Zhang L. Revolutionizing targeting precision: microfluidics-enabled smart microcapsules for tailored delivery and controlled release. LAB ON A CHIP 2024; 24:1367-1393. [PMID: 38314845 DOI: 10.1039/d3lc00835e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
As promising delivery systems, smart microcapsules have garnered significant attention owing to their targeted delivery loaded with diverse active materials. By precisely manipulating fluids on the micrometer scale, microfluidic has emerged as a powerful tool for tailoring delivery systems based on potential applications. The desirable characteristics of smart microcapsules are associated with encapsulation capacity, targeted delivery capability, and controlled release of encapsulants. In this review, we briefly describe the principles of droplet-based microfluidics for smart microcapsules. Subsequently, we summarize smart microcapsules as delivery systems for efficient encapsulation and focus on target delivery patterns, including passive targets, active targets, and microfluidics-assisted targets. Additionally, based on release mechanisms, we review controlled release modes adjusted by smart membranes and on/off gates. Finally, we discuss existing challenges and potential implications associated with smart microcapsules.
Collapse
Affiliation(s)
- Lingling Ren
- School of Petroleum Engineering, China University of Petroleum (East China), Qingdao, Shandong, China.
| | - Shuang Liu
- School of Petroleum Engineering, China University of Petroleum (East China), Qingdao, Shandong, China.
| | - Junjie Zhong
- School of Petroleum Engineering, China University of Petroleum (East China), Qingdao, Shandong, China.
| | - Liyuan Zhang
- School of Petroleum Engineering, China University of Petroleum (East China), Qingdao, Shandong, China.
| |
Collapse
|
33
|
Li J, Ye E, Huang J, Xu M, Zhang J, Zhang J, Cai H, Zhang J. Cysteine-modified PEGylated nanoparticles for targeted delivery of methylprednisolone to pancreatitis. Eur J Pharm Biopharm 2024; 195:114179. [PMID: 38199584 DOI: 10.1016/j.ejpb.2024.114179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/24/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024]
Abstract
The timely suppression of inflammatory mediator production and mitigation of their effects on pancreatic acinar cells are crucial for the successful management of acute pancreatitis. To achieve effective treatment, we present a novel approach utilizing cysteine modified PEG nanoparticles for both precise accumulation at the site of pancreatitis and specific targeting of acinar cells. Methylprednisolone, a nonsteroidal anti-inflammatory drug, was tailored to enhance its circulation time in the bloodstream, preferentially accumulate in the pancreas and enhance cell uptake efficiency by acinar cells through specifically targeting L-Type amino acid transporter 1. The nanosystem significantly downregulated pro-inflammatory cytokines in plasma, resulting in the effective suppression of inflammation in acinar cells within an acute pancreatitis rat model. The utilization of the dual targeted therapy strategy holds considerable potential for the clinical management of pancreatitis.
Collapse
Affiliation(s)
- Jianbo Li
- Henan Key Laboratory for Pharmacology of Liver Diseases, BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ertai Ye
- Henan Key Laboratory for Pharmacology of Liver Diseases, BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jingwen Huang
- Henan Key Laboratory for Pharmacology of Liver Diseases, BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Meng Xu
- Henan Key Laboratory for Pharmacology of Liver Diseases, BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; School of Basic Medical Science, Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Jieke Zhang
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Junrong Zhang
- Department of Pathogen Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Huijie Cai
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jinjie Zhang
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
34
|
Mohammad-Jafari K, Naghib SM, Mozafari MR. Cisplatin-based Liposomal Nanocarriers for Drug Delivery in Lung Cancer Therapy: Recent Progress and Future Outlooks. Curr Pharm Des 2024; 30:2850-2881. [PMID: 39051580 DOI: 10.2174/0113816128304923240704113319] [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: 02/13/2024] [Accepted: 05/23/2024] [Indexed: 07/27/2024]
Abstract
In order to improve the treatment of lung cancer, this paper looks at the development of cisplatinbased liposomal nanocarriers. It focuses on addressing the drawbacks of conventional cisplatin therapy, including systemic toxicity, inadequate tumor targeting, and drug resistance. Liposomes, or spherical lipid vesicles, offer a potentially effective way to encapsulate cisplatin, enhancing its transport and minimizing harmful effects on healthy tissues. The article discusses many liposomal cisplatin formulations, including pH-sensitive liposomes, sterically stabilized liposomes, and liposomes coupled with specific ligands like EGFR antibodies. These novel formulations show promise in reducing cisplatin resistance, optimizing pharmacokinetics, and boosting therapeutic results in the two in vitro and in vivo models. They also take advantage of the Enhanced Permeability and Retention (EPR) effect in the direction of improved tumor accumulation. The study highlights the need for more investigation to move these liposomal formulations from experimental to clinical settings, highlighting their potential to offer less harmful and more effective cancer therapy alternatives.
Collapse
Affiliation(s)
- Kave Mohammad-Jafari
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
35
|
Saraf S, Jain SK. pH-sensitive liposomes bearing a chemotherapeutic agent and a natural apoptosis modulator for effective intracellular delivery to the solid tumor. Drug Deliv Transl Res 2023; 13:2961-2981. [PMID: 37306925 DOI: 10.1007/s13346-023-01364-1] [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: 05/11/2023] [Indexed: 06/13/2023]
Abstract
The intracellular delivery of the drug to the solid tumor is a major challenge in the treatment of solid tumors. This project aims to increase cytosolic drug delivery using the endosomal escape of drugs. Topotecan (TPT) and capsaicin were used for the treatment of solid tumors. The pH-dependent conversion of active lactone form to inactive carboxylic form is a major problem of TPT that limits its therapeutic use. Liposomal encapsulation of TPT improved the stability of active lactone form and increased the therapeutic efficacy of TPT. Endosomal degradation of liposomes may reduce the content in the target cells. To solve these problems, pH-sensitive liposomes (pSLPs) were developed which improved the intracellular drug delivery by the endosomal escape of drugs. The liposomes (LPs) bearing the drug(s) were prepared using the cast film method and optimized for various formulation and process variables using the Design-Expert 7 software by employing the Box-Behnken design (BBD). The developed hyaluronic acid (HA)-conjugated pSLPs (HA-pSLPs) displayed a vesicle size of 166.5 ± 2.31 nm, zeta potential - 30.53 ± 0.91, and entrapment efficiency of 44.39 ± 1.78%, and 73.48 ± 2.15% for TPT and CAP, respectively. HA-pSLPs displayed better cytotoxicity in comparison to free drugs either single or in combination on the MCF-7 cell line. The apoptosis and cellular uptake of HA-pSLPs were increased ⁓ 4.45-fold and ⁓ 6.95-fold as compared to unconjugated pSLPs, respectively. The pharmacokinetic studies in Balb/c mice demonstrated that HA-pSLPs increased the half-life, MRT, and AUC in comparison to the free drug solution. The HA-pSLPs formulation has shown remarkable tumor regression as compared to PpSLPs, pSLPs, and free drug combinations. These results demonstrated that TPT- and CAP-loaded HA-pSLPs offer a potential platform for targeted drug delivery to solid tumors.
Collapse
Affiliation(s)
- Shivani Saraf
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, M.P, India, 470003
| | - Sanjay K Jain
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, M.P, India, 470003.
| |
Collapse
|
36
|
Cardoso RV, Pereira PR, Freitas CS, Mattos ÉBDA, Silva AVDF, Midlej VDV, Vericimo MA, Conte-Júnior CA, Paschoalin VMF. Tarin-Loaded Nanoliposomes Activate Apoptosis and Autophagy and Inhibit the Migration of Human Mammary Adenocarcinoma Cells. Int J Nanomedicine 2023; 18:6393-6408. [PMID: 37954458 PMCID: PMC10638905 DOI: 10.2147/ijn.s434626] [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: 09/09/2023] [Accepted: 10/28/2023] [Indexed: 11/14/2023] Open
Abstract
Background Tarin, a lectin purified from Colocasia esculenta, promotes in vitro and in vivo immunomodulatory effects allied to promising anticancer and antimetastatic effects against human adenocarcinoma mammary cells. This makes this 47 kDa-protein a natural candidate against human breast cancer, a leading cause of death among women. Tarin encapsulated in pegylated nanoliposomes displays increased effectiveness in controlling the proliferation of a mammary adenocarcinoma lineage comprising MDA-MB-231 cells. Methods The mechanisms enrolled in anticancer and antimetastatic responses were investigated by treating MDA-MB-231 cells with nano-encapsulated tarin at 72 μg/mL for up to 48h through flow cytometry and transmission electron microscopy (TEM). The safety of nano-encapsulated tarin towards healthy tissue was also assessed by the resazurin viability assay, and the effect of nanoencapsulated tarin on cell migration was evaluated by scratch assays. Results Ultrastructural analyses of MDA-MB-231 cells exposed to nanoencapsulated tarin revealed the accumulation of autophagosomes and damaged organelles, compatible with autophagy-dependent cell death. On the other hand, the flow cytometry investigation detected the increased occurrence of acidic vacuolar organelles, a late autophagosome trait, along with the enhanced presence of apoptotic cells, activated caspase-3/7, and cell cycle arrest at G0/G1. No deleterious effects were observed in healthy fibroblast cells following tarin nanoencapsulated exposition, in contrast to reduced viability in cells exposed to free tarin. The migration of MDA-MB-231 cells was inhibited by nano-encapsulated tarin, with delayed movement by 24 h compared to free tarin. Conclusion The nanoliposome formulation delivers tarin in a delayed and sustained manner, as evidenced by the belated and potent antitumoral and anti-migration effects on adenocarcinoma cells, with no toxicity to healthy cells. Although further investigations are required to fully understand antitumorigenic tarin mechanisms, the activation of both apoptotic and autophagic machineries along with the caspase-3/7 pathway, and cell cycle arrest may comprise a part of these mechanisms.
Collapse
Affiliation(s)
- Raiane Vieira Cardoso
- Departamento de Bioquímica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Cyntia Silva Freitas
- Departamento de Bioquímica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | |
Collapse
|
37
|
Petruk N, Sousa S, Croset M, Polari L, Zlatev H, Selander K, Mönkkönen J, Clézardin P, Määttä J. Liposome-encapsulated zoledronate increases inflammatory macrophage population in TNBC tumours. Eur J Pharm Sci 2023; 190:106571. [PMID: 37652236 DOI: 10.1016/j.ejps.2023.106571] [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: 03/08/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Tumour associated macrophages (TAMs) are important players in breast tumour progression and metastasis. Clinical and preclinical evidence suggests a role for zoledronate (ZOL) in breast cancer metastasis prevention. Further, zoledronate is able to induce inflammatory activation of monocytes and macrophages, which can be favourable in cancer treatments. The inherent bone tropism of zoledronate limits its availability in soft tissues and tumours. In this study we utilised an orthotopic murine breast cancer model to evaluate the possibility to use liposomes (EMP-LIP) to target zoledronate to tumours to modify TAM activation. METHODS Triple-negative breast cancer 4T1 cells were inoculated in the 4th mammary fat pad of female Balb/c mice. Animals were divided according to the treatment: vehicle, ZOL, EMP-LIP and liposome encapsulated zoledronate (ZOL-LIP). Treatment was done intravenously (with tumour resection) and intraperitoneally (without tumour resection). Tumour growth was followed by bioluminescence in vivo imaging (IVIS) and calliper measurements. Tumour-infiltrating macrophages were assessed by immunohistochemical and immunofluorescence staining. Protein and RNA expression levels of inflammatory transcription factors and cytokines were measured by Western Blotting and Taqman RT-qPCR. RESULTS Liposome encapsulated zoledronate (ZOL-LIP) treatment suppressed migration of 4T1 cell in vitro. Tumour growth and expression of the angiogenic marker CD34 were reduced upon both ZOL and ZOL-LIP treatment in vivo. Long-term ZOL-LIP treatment resulted in shift towards M1-type macrophage polarization, increased CD4 T cell infiltration and activation of NF-κB indicating changes in intratumoural inflammation, whereas ZOL treatment showed similar but non-significant trends. Moreover, ZOL-LIP had a lower bisphosphonate accumulation in bone compared to free ZOL. CONCLUSION Results show that the decreased bisphosphonate accumulation in bone promotes the systemic anti-tumour effect of ZOL-LIP by increasing inflammatory response in TNBC tumours via M1-type macrophage activation.
Collapse
Affiliation(s)
- Nataliia Petruk
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sofia Sousa
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | | | - Lauri Polari
- Institute of Biomedicine, University of Turku, Turku, Finland; Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Hristo Zlatev
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Katri Selander
- Department of Oncology and Radiation Therapy, Oulu University Hospital, Oulu, Finland
| | - Jukka Mönkkönen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | | | - Jorma Määttä
- Institute of Biomedicine, University of Turku, Turku, Finland; Turku Center for Disease Modeling, University of Turku, Turku, Finland.
| |
Collapse
|
38
|
Bernardi M, Signore G, Moscardini A, Pugliese LA, Pesce L, Beltram F, Cardarelli F. Fluorescence Lifetime Nanoscopy of Liposomal Irinotecan Onivyde: From Manufacturing to Intracellular Processing. ACS APPLIED BIO MATERIALS 2023; 6:4277-4289. [PMID: 37699572 PMCID: PMC10583229 DOI: 10.1021/acsabm.3c00478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/27/2023] [Indexed: 09/14/2023]
Abstract
Onivyde was approved by the Food and Drug Administration (FDA) in 2015 for the treatment of solid tumors, including metastatic pancreatic cancer. It is designed to encapsulate irinotecan at high concentration, increase its blood-circulation lifetime, and deliver it to cells where it is enzymatically converted into SN-38, a metabolite with 100- to 1000-fold higher anticancer activity. Despite a rewarding clinical path, little is known about the physical state of encapsulated irinotecan within Onivyde and how this synthetic identity changes throughout the process from manufacturing to intracellular processing. Herein, we exploit irinotecan intrinsic fluorescence and fluorescence lifetime imaging microscopy (FLIM) to selectively probe the supramolecular organization of the drug. FLIM analysis on the manufacturer's formulation reveals the presence of two coexisting physical states within Onivyde liposomes: (i) gelated/precipitated irinotecan and (ii) liposome-membrane-associated irinotecan, the presence of which is not inferable from the manufacturer's indications. FLIM in combination with high-performance liquid chromatography (HPLC) and a membrane-impermeable dynamic quencher of irinotecan reveals rapid (within minutes) and complete chemical dissolution of the gelated/precipitated phase upon Onivyde dilution in standard cell-culturing medium with extensive leakage of the prodrug from liposomes. Indeed, confocal imaging and cell-proliferation assays show that encapsulated and nonencapsulated irinotecan formulations are similar in terms of cell-uptake mechanism and cell-division inhibition. Finally, 2-channel FLIM analysis discriminates the signature of irinotecan from that of its red-shifted SN-38 metabolite, demonstrating the appearance of the latter as a result of Onivyde intracellular processing. The findings presented in this study offer fresh insights into the synthetic identity of Onivyde and its transformation from production to in vitro administration. Moreover, these results serve as another validation of the effectiveness of FLIM analysis in elucidating the supramolecular organization of encapsulated fluorescent drugs. This research underscores the importance of leveraging advanced imaging techniques to deepen our understanding of drug formulations and optimize their performance in delivery applications.
Collapse
Affiliation(s)
- Mario Bernardi
- Scuola
Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Giovanni Signore
- Biochemistry
Unit, Department of Biology, University
of Pisa, via San Zeno
51, 56123 Pisa, Italy
- Institute
of Clinical Physiology, National Research
Council, 56124 Pisa, Italy
| | - Aldo Moscardini
- Scuola
Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Licia Anna Pugliese
- Scuola
Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Luca Pesce
- Scuola
Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Fabio Beltram
- Scuola
Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
- NEST,
Istituto Nanoscienze-CNR, Piazza S. Silvestro, 12, I-56127 Pisa, Italy
| | - Francesco Cardarelli
- Scuola
Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
- NEST,
Istituto Nanoscienze-CNR, Piazza S. Silvestro, 12, I-56127 Pisa, Italy
| |
Collapse
|
39
|
Liu WS, Wu LL, Chen CM, Zheng H, Gao J, Lu ZM, Li M. Lipid-hybrid cell-derived biomimetic functional materials: A state-of-the-art multifunctional weapon against tumors. Mater Today Bio 2023; 22:100751. [PMID: 37636983 PMCID: PMC10448342 DOI: 10.1016/j.mtbio.2023.100751] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Tumors are among the leading causes of death worldwide. Cell-derived biomimetic functional materials have shown great promise in the treatment of tumors. These materials are derived from cell membranes, extracellular vesicles and bacterial outer membrane vesicles and may evade immune recognition, improve drug targeting and activate antitumor immunity. However, their use is limited owing to their low drug-loading capacity and complex preparation methods. Liposomes are artificial bionic membranes that have high drug-loading capacity and can be prepared and modified easily. Although they can overcome the disadvantages of cell-derived biomimetic functional materials, they lack natural active targeting ability. Lipids can be hybridized with cell membranes, extracellular vesicles or bacterial outer membrane vesicles to form lipid-hybrid cell-derived biomimetic functional materials. These materials negate the disadvantages of both liposomes and cell-derived components and represent a promising delivery platform in the treatment of tumors. This review focuses on the design strategies, applications and mechanisms of action of lipid-hybrid cell-derived biomimetic functional materials and summarizes the prospects of their further development and the challenges associated with it.
Collapse
Affiliation(s)
- Wen-Shang Liu
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| | - Li-Li Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Cui-Min Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Hao Zheng
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zheng-Mao Lu
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Meng Li
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| |
Collapse
|
40
|
Cho E, Mun SJ, Jeon M, Kim HK, Baek H, Ham YS, Gil WJ, Kim JW, Yang CS. Tumor-targeted liposomes with platycodin D2 promote apoptosis in colorectal cancer. Mater Today Bio 2023; 22:100745. [PMID: 37576871 PMCID: PMC10415802 DOI: 10.1016/j.mtbio.2023.100745] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/28/2023] [Accepted: 07/24/2023] [Indexed: 08/15/2023] Open
Abstract
Conventional chemotherapy for colorectal cancer (CRC), though efficacious, is discouraging due to its limited targeting capability, lack of selectivity, and chemotherapy-associated side effects. With the advent of nanomedicines, a liposomal delivery system making use of a combination of anticancer phytochemicals is fast gaining popularity as one of the most promising nanoplatforms for CRC treatment. Rising evidence supports phytochemicals such as platycosides for their anticancer potency. To this end, a combination therapy including tumor-targeted liposomes along with phytochemicals might have a greater therapeutic potential against cancer. In this study, we developed acidity-triggered rational membrane (ATRAM) along with conjugated platycodin D2 (PCD2) and liposomes (PCD2-Lipo-ATRAM) as a tumor-targeting therapy. The PCD2-Lipo-ATRAM treatment demonstrated a successful tumor-targeting ability in the CRC xenografts, in which PCD2 not only exerted a potent antitumor effect by inducing apoptotic cell death and but also functioned as a liposome membrane stabilizer. Moreover, PCD2-Lipo-ATRAM suppressed antiapoptotic BCL-2 family proteins, resulting in enhanced cytotoxicity toward CRC cells by inducing intrinsic caspase-9/-3 mediated apoptosis. Thus, our data has shown that tumor-targeting PCD2-based liposomal systems represent a promising strategy for CRC therapy, since they directly target the tumors, unlike other therapies that can miss the target.
Collapse
Affiliation(s)
- Euni Cho
- Department of Bionano Engineering Technology, Hanyang University, Seoul, 04673, South Korea
- Center for Bionano Intelligence Education and Research, Ansan, 15588, South Korea
| | - Seok-Jun Mun
- Department of Bionano Engineering Technology, Hanyang University, Seoul, 04673, South Korea
- Center for Bionano Intelligence Education and Research, Ansan, 15588, South Korea
| | - Minha Jeon
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Hyo Keun Kim
- Center for Bionano Intelligence Education and Research, Ansan, 15588, South Korea
- Department of Molecular and Life Science, Hanyang University, Ansan, 15588, South Korea
| | - Hwira Baek
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Yu Seong Ham
- Center for Bionano Intelligence Education and Research, Ansan, 15588, South Korea
- Department of Molecular and Life Science, Hanyang University, Ansan, 15588, South Korea
| | - Woo Jin Gil
- Center for Bionano Intelligence Education and Research, Ansan, 15588, South Korea
- Department of Molecular and Life Science, Hanyang University, Ansan, 15588, South Korea
| | - Jin Woong Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Chul-Su Yang
- Center for Bionano Intelligence Education and Research, Ansan, 15588, South Korea
- Department of Molecular and Life Science, Hanyang University, Ansan, 15588, South Korea
- Department of Medicinal and Life Science, Hanyang University, Ansan, 15588, South Korea
| |
Collapse
|
41
|
Lu X, Cheng H, Xu Q, Tan X. Encapsulation of STING Agonist cGAMP with Folic Acid-Conjugated Liposomes Significantly Enhances Antitumor Pharmacodynamic Effect. Cancer Biother Radiopharm 2023; 38:543-557. [PMID: 33719535 DOI: 10.1089/cbr.2020.4085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background: 2',3'-cGAMP (2',3'-cyclic AMP-GMP) has been reported as an agonist of the STING (stimulator of interferon genes) signaling pathway. However, cGAMP has poor membrane permeability and can be hydrolyzed by ectonucleotide pyrophosphatase/phosphodiesterase (ENPP1), limiting its ability to activate the STING-IRF3 pathway. This study aimed to investigate that the folate-targeted liposomal cGAMP could overcome the defects of free cGAMP to enhance the antitumor effect. Materials and Methods: cGAMP was encapsulated in PEGylated folic acid-targeted liposomes to construct a carrier-delivered formulation. The particle size and morphology were detected by dynamic light scattering and transmission electron microscopy. The sustained-release ability was measured by drug release and pharmacokinetics. Animal models were applied to evaluate the tumor inhibition efficiency in vivo. Flow cytometry, enzyme-linked immunosorbent assay, and real-time polymerase chain reaction were used to detect the expression of immune cells, secreted cytokines, and target genes. The activation of the STING-IRF3 pathway was evaluated by immunofluorescence. Results: Physical characters of liposomes revealed that the prepared liposomes were stable in neutral humoral environments and released more internal drugs in acidic tumor tissues. Systemic therapy with liposomes on Colorectal 26 tumor-bearing mice in vivo effectively inhibited tumor growth via stimulating the expression of CD8+ T cells and reversed the immunosuppressed tumor microenvironment (TME). Conclusions: The study suggests that the folic acid-targeted cGAMP-loaded liposomes deliver drugs to the TME to enhance the STING agonist activity, improving the efficiency of tumor therapy via the cGAMP-STING-IRF3 pathway.
Collapse
Affiliation(s)
- Xing Lu
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hao Cheng
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qiming Xu
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiangshi Tan
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Dubey AK, Mostafavi E. Biomaterials-mediated CRISPR/Cas9 delivery: recent challenges and opportunities in gene therapy. Front Chem 2023; 11:1259435. [PMID: 37841202 PMCID: PMC10568484 DOI: 10.3389/fchem.2023.1259435] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The use of biomaterials in delivering CRISPR/Cas9 for gene therapy in infectious diseases holds tremendous potential. This innovative approach combines the advantages of CRISPR/Cas9 with the protective properties of biomaterials, enabling accurate and efficient gene editing while enhancing safety. Biomaterials play a vital role in shielding CRISPR/Cas9 components, such as lipid nanoparticles or viral vectors, from immunological processes and degradation, extending their effectiveness. By utilizing the flexibility of biomaterials, tailored systems can be designed to address specific genetic diseases, paving the way for personalized therapeutics. Furthermore, this delivery method offers promising avenues in combating viral illnesses by precisely modifying pathogen genomes, and reducing their pathogenicity. Biomaterials facilitate site-specific gene modifications, ensuring effective delivery to infected cells while minimizing off-target effects. However, challenges remain, including optimizing delivery efficiency, reducing off-target effects, ensuring long-term safety, and establishing scalable production techniques. Thorough research, pre-clinical investigations, and rigorous safety evaluations are imperative for successful translation from the laboratory to clinical applications. In this review, we discussed how CRISPR/Cas9 delivery using biomaterials revolutionizes gene therapy and infectious disease treatment, offering precise and safe editing capabilities with the potential to significantly improve human health and quality of life.
Collapse
Affiliation(s)
- Ankit Kumar Dubey
- Global Research and Publishing Foundation, New Delhi, India
- Institute of Scholars, Bengaluru, Karnataka, India
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
43
|
Nasr M, Kira AY, Saber S, Essa EA, El-Gizawy SA. Lactosylated Chitosan Nanoparticles Potentiate the Anticancer Effects of Telmisartan In Vitro and in a N-Nitrosodiethylamine-Induced Mice Model of Hepatocellular Carcinoma. Mol Pharm 2023; 20:4758-4769. [PMID: 37585079 DOI: 10.1021/acs.molpharmaceut.3c00542] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
Hepatocellular carcinoma (HCC) is the leading cause of cancer-related mortality worldwide. Telmisartan (TLM), a BSC class II drug, has been reported to have antiproliferative activity in HCC. However, its therapeutic activity is limited by poor bioavailability and unpredictable distribution. This work aimed to enhance TLM's liver uptake for HCC management through passive and active targeting pathways utilizing chitosan nanoparticles decorated with lactose (LCH NPs) as a delivery system. In vitro cell cytotoxicity and cellular uptake studies indicated that TLM-LCH NPs significantly (p < 0.05) enhanced the antiproliferative activity and cellular uptake percentage of TLM. In vivo bioavailability and liver biodistribution studies indicated that TLM-LCH NPs significantly (p < 0.05) enhanced TLM concentrations in plasma and the liver. The relative liver uptake of TLM from TLM-LCH NPs was 2-fold higher than that of unmodified NPs and 5-fold higher than that of plain TLM suspension. In vivo studies of a N-nitrosodiethylamine-induced HCC model revealed that administration of TLM through LCH NPs improved liver histology and resulted in lower serum alpha-fetoprotein (AFP), matrix metalloproteinase 2 (MMP-2), vascular endothelial growth factor (VEGF) levels, and liver weight index compared to plain TLM and TLM-loaded unmodified NPs. These results reflected the high potentiality of LCH NPs as a liver-targeted delivery system for TLM in the treatment of HCC.
Collapse
Affiliation(s)
- Mohamed Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Helwan University, Cairo 11790, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| | - Ahmed Y Kira
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| | - Ebtessam A Essa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Sanaa A El-Gizawy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| |
Collapse
|
44
|
Pisani S, Di Martino D, Cerri S, Genta I, Dorati R, Bertino G, Benazzo M, Conti B. Investigation and Comparison of Active and Passive Encapsulation Methods for Loading Proteins into Liposomes. Int J Mol Sci 2023; 24:13542. [PMID: 37686348 PMCID: PMC10487800 DOI: 10.3390/ijms241713542] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/26/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
In this work, four different active encapsulation methods, microfluidic (MF), sonication (SC), freeze-thawing (FT), and electroporation (EP), were investigated to load a model protein (bovine serum albumin-BSA) into neutral liposomes made from 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC):cholesterol (Chol) and charged liposomes made from DSPC:Chol:Dioleoyl-3-trimethylammonium propane (DOTAP), DSPC:Chol:1,2-dioleoyl-sn-glycero-3-phospho-L-serine (DOPS), and DSPC:Chol:phosphatidylethanolamine (PE). The aim was to increase the protein encapsulation efficiency (EE%) by keeping the liposome size below 200 nm and the PDI value below 0.7, which warrants a nearly monodisperse preparation. Electroporation (100 V) yielded the best results in terms of EE%, with a dramatic increase in liposome size (>600 nm). The FT active-loading method, either applied to neutral or charged liposomes, allowed for obtaining suitable EE%, keeping the liposome size range below 200 nm with a suitable PDI index. Cationic liposomes (DSPC:Chol:DOTAP) loaded with the FT active method showed the best results in terms of EE% (7.2 ± 0.8%) and size (131.2 ± 11.4 nm, 0.140 PDI). In vitro release of BSA from AM neutral and charged liposomes resulted slower compared to PM liposomes and was affected by incubation temperature (37 °C, 4 °C). The empty charged liposomes tested for cell viability on Human Normal Dermal Fibroblast (HNDF) confirmed their cytocompatibility also at high concentrations (1010 particles/mL) and cellular uptake at 4 °C and 37 °C. It can be concluded that even if both microfluidic passive and active methods are more easily transferable to an industrial scale, the FT active-loading method turned out to be the best in terms of BSA encapsulation efficiencies, keeping liposome size below 200 nm.
Collapse
Affiliation(s)
- Silvia Pisani
- Department of Drug Sciences, University of Pavia, Viale T. Taramelli 12, 27100 Pavia, Italy; (S.P.); (I.G.); (R.D.)
| | - Deborah Di Martino
- Unit of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (D.D.M.); (S.C.)
| | - Silvia Cerri
- Unit of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (D.D.M.); (S.C.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, Viale T. Taramelli 12, 27100 Pavia, Italy; (S.P.); (I.G.); (R.D.)
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, Viale T. Taramelli 12, 27100 Pavia, Italy; (S.P.); (I.G.); (R.D.)
| | - Giulia Bertino
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (G.B.); (M.B.)
| | - Marco Benazzo
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (G.B.); (M.B.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, Viale T. Taramelli 12, 27100 Pavia, Italy; (S.P.); (I.G.); (R.D.)
| |
Collapse
|
45
|
Zhang Y, Fang X, Huang W, Li Q, Jiang H, Wang C, Liu H. Plasmon Resonance Energy Transfer Nanoruler for Pinpointing Molecular Distance and Interaction on the Living Cell Membrane. NANO LETTERS 2023; 23:7750-7757. [PMID: 37387534 DOI: 10.1021/acs.nanolett.3c01629] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Developing novel strategies to measure nanoscale distance and molecular interaction on a living cell membrane is of great significance but challenging. Here we develop a model of a linker-free plasmon resonance energy transfer, termed "PRET nanoruler", which is composed of a single-sized nanogold-antibody conjugates donor (G26@antiCD71) and a fluorophore-labeled XQ-2d aptamer receptor (XQ-2d-Cy3), that produces a separation distance (r) dependent energy transfer (ηPRET). Both the theoretical finite element simulation and experiments evidence the observable PRET between single G26NPs and XQ-2d-Cy3. Regardless of the size of ηPRET, we could confirm r is less than 5 nm, the separation of two binding sites is in the range of 13.0-18.0 nm. There is a competitive binding of Tf and XQ-2d-Cy3 on CD71 receptors. PRET nanoruler realizes the estimation of the nanoscale separation distance, and determines the molecular interaction and competitive binding. It is an alternative tool for observing nanoscale single molecular events in the future.
Collapse
Affiliation(s)
- Yu Zhang
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Xingru Fang
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Wenwen Huang
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Qi Li
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Hao Jiang
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Chao Wang
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei 230027, China
| | - Honglin Liu
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| |
Collapse
|
46
|
Kou P, Levy ES, Nguyen AD, Zhang D, Chen S, Cui Y, Zhang X, Broccatelli F, Pizzano J, Cantley J, Bortolon E, Rousseau E, Berlin M, Dragovich P, Sethuraman V. Development of Liposome Systems for Enhancing the PK Properties of Bivalent PROTACs. Pharmaceutics 2023; 15:2098. [PMID: 37631312 PMCID: PMC10458015 DOI: 10.3390/pharmaceutics15082098] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Proteolysis-Targeting Chimeras (PROTACs) are a promising new technology in drug development. They have rapidly evolved in recent years, with several of them in clinical trials. While most of these advances have been associated with monovalent protein degraders, bivalent PROTACs have also entered clinical trials, although progression to market has been limited. One of the reasons is the complex physicochemical properties of the heterobifunctional PROTACs. A promising strategy to improve pharmacokinetics of highly lipophilic compounds, such as PROTACs, is encapsulation in liposome systems. Here we describe liposome systems for intravenous administration to enhance the PK properties of two bivalent PROTAC molecules, by reducing clearance and increasing systemic coverage. We developed and characterized a PROTAC-in-cyclodextrin liposome system where the drug was retained in the liposome core. In PK studies at 1 mg/kg for GNE-01 the PROTAC-in-cyclodextrin liposome, compared to the solution formulation, showed a 80- and a 380-fold enhancement in AUC for mouse and rat studies, respectively. We further investigated the same PROTAC-in-cyclodextrin liposome system with the second PROTAC (GNE-02), where we monitored both lipid and drug concentrations in vivo. Similarly, in a mouse PK study of GEN-02, the PROTAC-in-cyclodextrin liposome system exhibited enhancement in plasma concentration of a 23× increase over the conventional solution formulation. Importantly, the lipid CL correlated with the drug CL. Additionally, we investigated a conventional liposome approach for GNE-02, where the PROTAC resides in the lipid bilayer. Here, a 5× increase in AUC was observed, compared to the conventional solution formulation, and the drug CL was faster than the lipid CL. These results indicate that the different liposome systems can be tailored to translate across multiple PROTAC systems to modulate and improve plasma concentrations. Optimization of the liposomes could further improve tumor concentration and improve the overall therapeutic index (TI). This delivery technology may be well suited to bring novel protein targeted PROTACs into clinics.
Collapse
Affiliation(s)
- Ponien Kou
- Small Molecules Pharmaceutics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (P.K.); (E.S.L.); (A.D.N.)
| | - Elizabeth S. Levy
- Small Molecules Pharmaceutics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (P.K.); (E.S.L.); (A.D.N.)
| | - An D. Nguyen
- Small Molecules Pharmaceutics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (P.K.); (E.S.L.); (A.D.N.)
| | - Donglu Zhang
- Drug Metabolism & Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (D.Z.); (S.C.); (Y.C.); (X.Z.); (F.B.)
| | - Shu Chen
- Drug Metabolism & Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (D.Z.); (S.C.); (Y.C.); (X.Z.); (F.B.)
| | - Yusi Cui
- Drug Metabolism & Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (D.Z.); (S.C.); (Y.C.); (X.Z.); (F.B.)
| | - Xing Zhang
- Drug Metabolism & Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (D.Z.); (S.C.); (Y.C.); (X.Z.); (F.B.)
| | - Fabio Broccatelli
- Drug Metabolism & Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (D.Z.); (S.C.); (Y.C.); (X.Z.); (F.B.)
| | - Jennifer Pizzano
- Arvinas LLC, 5 Science Park, New Haven, CT 06511, USA; (J.P.); (J.C.); (E.B.); (E.R.); (M.B.)
| | - Jennifer Cantley
- Arvinas LLC, 5 Science Park, New Haven, CT 06511, USA; (J.P.); (J.C.); (E.B.); (E.R.); (M.B.)
| | - Elizabeth Bortolon
- Arvinas LLC, 5 Science Park, New Haven, CT 06511, USA; (J.P.); (J.C.); (E.B.); (E.R.); (M.B.)
| | - Emma Rousseau
- Arvinas LLC, 5 Science Park, New Haven, CT 06511, USA; (J.P.); (J.C.); (E.B.); (E.R.); (M.B.)
| | - Michael Berlin
- Arvinas LLC, 5 Science Park, New Haven, CT 06511, USA; (J.P.); (J.C.); (E.B.); (E.R.); (M.B.)
| | - Peter Dragovich
- Medicinal Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA;
| | - Vijay Sethuraman
- Small Molecules Pharmaceutics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (P.K.); (E.S.L.); (A.D.N.)
| |
Collapse
|
47
|
Raju R, Abuwatfa WH, Pitt WG, Husseini GA. Liposomes for the Treatment of Brain Cancer-A Review. Pharmaceuticals (Basel) 2023; 16:1056. [PMID: 37630971 PMCID: PMC10458450 DOI: 10.3390/ph16081056] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/09/2023] [Accepted: 07/19/2023] [Indexed: 08/27/2023] Open
Abstract
Due to their biocompatibility, non-toxicity, and surface-conjugation capabilities, liposomes are effective nanocarriers that can encapsulate chemotherapeutic drugs and facilitate targeted delivery across the blood-brain barrier (BBB). Additionally, strategies have been explored to synthesize liposomes that respond to internal and/or external stimuli to release their payload controllably. Although research into liposomes for brain cancer treatment is still in its infancy, these systems have great potential to fundamentally change the drug delivery landscape. This review paper attempts to consolidate relevant literature regarding the delivery to the brain using nanocarriers, particularly liposomes. The paper first briefly explains conventional treatment modalities for cancer, followed by describing the blood-brain barrier and ways, challenges, and techniques involved in transporting drugs across the BBB. Various nanocarrier systems are introduced, with attention to liposomes, due to their ability to circumvent the challenges imposed by the BBB. Relevant studies involving liposomal systems researched to treat brain tumors are reviewed in vitro, in vivo, and clinical studies. Finally, the challenges associated with the use of liposomes to treat brain tumors and how they can be addressed are presented.
Collapse
Affiliation(s)
- Richu Raju
- Biomedical Engineering Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Waad H. Abuwatfa
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - William G. Pitt
- Department of Chemical Engineering, Brigham Young University, Provo, UT 84602, USA;
| | - Ghaleb A. Husseini
- Biomedical Engineering Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
48
|
Sezgin-Bayindir Z, Losada-Barreiro S, Fernández-Bravo S, Bravo-Díaz C. Innovative Delivery and Release Systems for Antioxidants and Other Active Substances in the Treatment of Cancer. Pharmaceuticals (Basel) 2023; 16:1038. [PMID: 37513948 PMCID: PMC10383431 DOI: 10.3390/ph16071038] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer is one of the major diseases leading to death worldwide, and the fight against the disease is still challenging. Cancer diseases are usually associated with increased oxidative stress and the accumulation of reactive oxygen and nitrogen species as a result of metabolic alterations or signaling aberrations. While numerous antioxidants exhibit potential therapeutic properties, their clinical efficiency against cancer is limited and even unproven. Conventional anticancer antioxidants and drugs have, among others, the great disadvantage of low bioavailability, poor targeting efficiency, and serious side effects, constraining their use in the fight against diseases. Here, we review the rationale for and recent advances in potential delivery systems that could eventually be employed in clinical research on antioxidant therapy in cancer. We also review some of the various strategies aimed at enhancing the solubility of poorly water-soluble active drugs, including engineered delivery systems such as lipid-based, polymeric, and inorganic formulations. The use of cyclodextrins, micro- and nanoemulsions, and thermosensitive smart liposomes as useful systems for the delivery and release of poorly aqueous-soluble drugs, improving their bioactivity and stability, is also addressed. We also provide some details on their formulation processes and their use in a variety of medical applications. Finally, we briefly cover a case study specifically focused on the use of delivery systems to minimize oral cancer and associated dental problems.
Collapse
Affiliation(s)
- Zerrin Sezgin-Bayindir
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Ankara 06560, Turkey
| | - Sonia Losada-Barreiro
- Departamento de Química-Física, Facultade de Química, Universidade de Vigo, 36200 Vigo, Spain
| | - Sofía Fernández-Bravo
- Odontology Department, Primary Health Care Unit, Galician Health Service (SERGAS), Camiño do Lodairo s/n, 15570 Narón, Spain
| | - Carlos Bravo-Díaz
- Departamento de Química-Física, Facultade de Química, Universidade de Vigo, 36200 Vigo, Spain
| |
Collapse
|
49
|
Tsuneishi T, Kojima K, Kubota F, Harashima H, Yamada Y, Sudo Y. Development of light-induced disruptive liposomes (LiDL) as a photoswitchable carrier for intracellular substance delivery. Chem Commun (Camb) 2023; 59:7591-7594. [PMID: 37254694 DOI: 10.1039/d3cc02056h] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Light-driven inward proton pump rhodopsin RmXeR was embedded in pH-sensitive liposomes. Substance release from the proteoliposomes was observed following light illumination both in vitro and in cells, indicating the successful production of light-induced disruptive liposomes (LiDL). Thus, LiDL is a photoswitchable carrier utilized for intracellular substance delivery.
Collapse
Affiliation(s)
- Taichi Tsuneishi
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan.
| | - Keiichi Kojima
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan.
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan
| | - Fumika Kubota
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Yuki Sudo
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan.
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan
| |
Collapse
|
50
|
Hariharan K, Mehta T, Shah J, Dave H, Sami A, Omri A. Localized Delivery of Erlotinib Using Liposomal Gel Formulations for the Treatment of Oral Squamous Cell Carcinoma. Int J Pharm 2023:123144. [PMID: 37330155 DOI: 10.1016/j.ijpharm.2023.123144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/15/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Oral cancer accounts for more than 350000 cases worldwide with 90% of them being oral squamous cell carcinomas (OSCC). The current treatment modalities of chemoradiation have poor outcomes along with harmful effects to neighbouring healthy tissues. The present study aimed to deliver Erlotinib (ERB), locally at the site of tumor arising in the oral cavity. ERB was encapsulated in liposomal formulations (ERB Lipo) and optimized using full factorial, 32 experimental design. The optimized batch was then coated with chitosan to obtain CS-ERB Lipo and were characterized further. Both liposomal ERB formulations had size less than 200nm and PDI less than 0.4. Zeta potential was upto -50mV for ERB Lipo and upto + 25mV for CS-ERB Lipo indicating stable formulation. Liposomal formulations were freeze dried and loaded into gel to study in-vitro release and chemotherapeutic evaluation. CS-ERB Lipo showed sustained release upto 36 h from gel as compared to control formulation. In-vitro cell viability studies showed potent anti-cancer activity on KB-cells. In-vivo studies showed better pharmacological efficacy in terms of tumor volume reduction for ERB LIPO gel (49.19%) and CS-ERB Lipo gel (55.27%) as compared to plain ERB Gel (38.88%) applied locally. Histology also revealed that formulation could alleviate dysplasia condition to hyperplasia. The locoregional therapy of ERB Lipo gel and CS-ERB Lipo gel thus show promising outcome in improving pre-malignant and early-stage oral cavity cancers.
Collapse
Affiliation(s)
- Kartik Hariharan
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Ahmedabad, India-382481
| | - Tejal Mehta
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Ahmedabad, India-382481
| | - Jigna Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, SG Highway, Ahmedabad, India-382481
| | - Heena Dave
- Institute of Science, Nirma University, SG Highway, Ahmedabad, India-382481
| | - Anam Sami
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Ahmedabad, India-382481
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury ON P3E 2C6, Ontario, Canada
| |
Collapse
|