1
|
Păduraru L, Panainte AD, Peptu CA, Apostu M, Vieriu M, Bibire T, Sava A, Bibire N. Smart Drug Delivery Systems Based on Cyclodextrins and Chitosan for Cancer Therapy. Pharmaceuticals (Basel) 2025; 18:564. [PMID: 40283999 PMCID: PMC12030441 DOI: 10.3390/ph18040564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Despite improvements in therapeutic approaches like immunotherapy and gene therapy, cancer still remains a serious threat to world health due to its high incidence and mortality rates. Limitations of conventional therapy include suboptimal targeting, multidrug resistance, and systemic toxicity. A major challenge in current oncology therapies is the development of new delivery methods for antineoplastic drugs that act directly on target. One approach involves the complexation of antitumor drugs with cyclodextrins (CDs) and chitosan (CS) as an attempt to counteract their primary limitations: low water solubility and bioavailability, diminished in vitro and in vivo stability, and high dose-dependent toxicity. All those drawbacks may potentially exclude some therapeutic candidates from clinical trials, thus their integration into smart delivery systems or drug-targeting technologies must be implemented. We intended to overview new drug delivery systems based on chitosan or cyclodextrins with regard to the current diagnosis and cancer management. This narrative review encompasses full-length articles published in English between 2019 and 2025 (including online ahead of print versions) in PubMed-indexed journals, focusing on recent research on the encapsulation of diverse antitumor drugs within those nanosystems that exhibit responsiveness to various stimuli such as pH, redox potential, and folate receptor levels, thereby enhancing the release of bioactive compounds at tumor sites. The majority of the cited references focus on the most notable research, studies of novel applications, and scientific advancements in the field of nanostructures and functional materials employed in oncological therapies over the last six years. Certainly, there are additional stimuli with research potential that can facilitate the drug's release upon activation, such as reactive oxygen species (ROS), various enzymes, ATP level, or hypoxia; however, our review exclusively addresses the aforementioned stimuli presented in a comprehensive manner.
Collapse
Affiliation(s)
- Larisa Păduraru
- Department of Analytical Chemistry, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy from Iasi, 16th Universitatii Street, 700116 Iasi, Romania; (L.P.); (M.A.); (M.V.); (A.S.); (N.B.)
| | - Alina-Diana Panainte
- Department of Analytical Chemistry, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy from Iasi, 16th Universitatii Street, 700116 Iasi, Romania; (L.P.); (M.A.); (M.V.); (A.S.); (N.B.)
| | - Cătălina-Anișoara Peptu
- Department of Natural and Synthetic Polymers, Faculty of Chemical Engineering and Environmental Protection, “Gheorghe Asachi” Technical University of Iasi, 71st Prof. Dr. Docent Dimitrie Mangeron Street, 700050 Iasi, Romania
| | - Mihai Apostu
- Department of Analytical Chemistry, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy from Iasi, 16th Universitatii Street, 700116 Iasi, Romania; (L.P.); (M.A.); (M.V.); (A.S.); (N.B.)
| | - Mădălina Vieriu
- Department of Analytical Chemistry, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy from Iasi, 16th Universitatii Street, 700116 Iasi, Romania; (L.P.); (M.A.); (M.V.); (A.S.); (N.B.)
| | - Tudor Bibire
- “St. Spiridon” County Clinical Emergency Hospital, 1st Independentei Blvd., 700111 Iasi, Romania;
| | - Alexandru Sava
- Department of Analytical Chemistry, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy from Iasi, 16th Universitatii Street, 700116 Iasi, Romania; (L.P.); (M.A.); (M.V.); (A.S.); (N.B.)
| | - Nela Bibire
- Department of Analytical Chemistry, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy from Iasi, 16th Universitatii Street, 700116 Iasi, Romania; (L.P.); (M.A.); (M.V.); (A.S.); (N.B.)
| |
Collapse
|
2
|
Chang CY, Huang SH, Chen CY, Jian CB, Chang CC, Chang YY, Jung M, Lee HM, Cheng B. Monocyte-adhesive peptidyl liposomes for harnessing monocyte homing to tumor tissues. J Control Release 2025; 382:113672. [PMID: 40185332 DOI: 10.1016/j.jconrel.2025.113672] [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: 03/22/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
In current drug delivery strategies, the efficiency of most carriers still largely depends on their ability to passively infiltrate target tissues. To overcome this limitation, we developed monocyte-adhesive peptidyl liposomes, termed monocyte-mediated drug carriers (MMDCs). These carriers are designed to exploit the innate chemotactic properties of monocytes, which actively home to diseased tissues. MMDCs were shown to effectively hitchhike on circulating monocytes (THP-1 cells) under physiological flow conditions. Their targeting specificity was further demonstrated in a 3D microfluidic culture system consisting of human breast cancer spheroids (MDA-MB-231) embedded in a collagen matrix, overlaid with a human endothelial cell-derived barrier. MMDCs underwent trans-endothelial migration via monocyte hitchhiking and selectively recognized collagen matrices containing MDA-MB-231 cells, but not those embedded with non-cancerous cells. In vitro assays revealed that doxorubicin encapsulated in MMDCs was released into the extracellular environment following phagocytosis of the carriers by THP-1-derived macrophages. In a xenograft mouse model, MMDCs exhibited high tumor-targeting efficiency. By harnessing the homing capability of monocytes, MMDCs significantly improved drug biodistribution at the disease site, thereby enhancing the therapeutic efficacy of the encapsulated agents.
Collapse
Affiliation(s)
- Chia-Yu Chang
- Graduate Institute of Biomedical Engineering, National Chung-Hsing University, Taichung, Taiwan
| | - Shih-Hsun Huang
- Graduate Institute of Biomedical Engineering, National Chung-Hsing University, Taichung, Taiwan
| | | | - Cheng-Bang Jian
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan; Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan; Nano Science and Technology Program, Taiwan International Graduate Program, Academia Sinica and National Taiwan University, Taiwan
| | - Ching-Chung Chang
- Doctoral Program in Tissue Engineering and Regenerative Medicine, National Chung-Hsing University, Taichung, Taiwan
| | | | - Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Hsien-Ming Lee
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan.
| | - Bill Cheng
- Graduate Institute of Biomedical Engineering, National Chung-Hsing University, Taichung, Taiwan.
| |
Collapse
|
3
|
Menachem R, Nudelman I, Vorontsova A, Livneh I, Sela M, Benguigui M, Manobla B, Shammai Y, Deo A, Buxbaum C, Bessler R, Raviv Z, Shklover J, Sznitman J, Ciechanover A, Schroeder A, Shaked Y. Bone Marrow-Targeted Liposomes Loaded with Bortezomib Overcome Multiple Myeloma Resistance. ACS NANO 2025; 19:11684-11701. [PMID: 40117329 PMCID: PMC11966756 DOI: 10.1021/acsnano.4c10597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/23/2025]
Abstract
Multiple myeloma (MM) poses a significant therapeutic challenge due to its persistent progression and low survival rate. Although the proteasome inhibitor bortezomib has revolutionized MM treatment, MM aggressiveness and drug resistance remain critical concerns. To tackle this problem, we developed AMD3100-targeted Bortezomib Liposomes (ATBL) designed for the targeted delivery of bortezomib to MM cells. Uptake of ATBL into MM cells was dependent on CXCR4 and was enhanced compared to nontargeted liposomes, both in vitro and in vivo. Treating MM-bearing mice with ATBL achieved superior therapeutic efficacy compared to treatment with free bortezomib or nontargeted bortezomib-loaded liposomes. Notably, the therapeutic activity of ATBL was limited in mice inoculated with CXCR4-knockdown MM cells, highlighting CXCR4 as a potential biomarker for ATBL response. Importantly, ATBL was effective against an aggressive and bortezomib-resistant MM clone both in vitro and in vivo. Toxicity and biodistribution profiles demonstrated the safety and bone marrow-targeting ability of ATBL. Collectively, this study highlights ATBL as a promising next-generation proteasome inhibitor-based therapy that incorporates bone marrow-targeting ability and sensitizing elements to overcome drug resistance in MM.
Collapse
Affiliation(s)
- Rotem Menachem
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Igor Nudelman
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Avital Vorontsova
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Ido Livneh
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Mor Sela
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Madeleine Benguigui
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Bar Manobla
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Yael Shammai
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Abhilash Deo
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Chen Buxbaum
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Ron Bessler
- Faculty
of Biomedical Engineering, Technion−Israel
Institute of Technology, Haifa 3200001, Israel
| | - Ziv Raviv
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Jeny Shklover
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Josué Sznitman
- Faculty
of Biomedical Engineering, Technion−Israel
Institute of Technology, Haifa 3200001, Israel
| | - Aaron Ciechanover
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Avi Schroeder
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Yuval Shaked
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| |
Collapse
|
4
|
Xu M, Duan M, Chen M, Mahal A, Yang L, Meng C, Zhang Z, Ren J, Obaidullah AJ, Li S, Wang C. Study on the activity of targeted delivery of DOX against melanoma by exosome-like nanovesicles of Rhodiola rosea. Biochim Biophys Acta Gen Subj 2025; 1869:130776. [PMID: 39970993 DOI: 10.1016/j.bbagen.2025.130776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/21/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Melanoma is the main cause of death from skin cancer. The current treatment methods have prominent toxic side effects. In order to more effectively inhibit melanoma and reduce the toxic side effects during treatment, this paper constructs an engineering system using DSPE-PEG2000-pYEEIE(pYEEIE) molecules to modify exosome-like nanovesicles vesicles of Rhodiola rosea (RELNs) and load Doxorubicin (DOX). As a drug system, the aim is to achieve better targeting activity of the system towards melanoma cell A375. The results showed that the morphology and particle size of the prepared RELNs met the defined criteria for evaluating extracellular vesicles. The pYEEIE-RELNs-DOX drug delivery system has a better inhibitory effect on cell proliferation compared to DOX and RELNs-DOX. At the same time, the pYEEIE-RELN-DOX drug delivery system also showed better targeting towards tumor cells. In summary, this study proposes for the first time RELNs as a new generation of drug delivery carriers and uses them for drug delivery and inhibition of melanoma cell toxicity. This lays the foundation for subsequent animal and clinical experiments, and provides new ideas for the treatment of skin cancer caused by melanoma.
Collapse
Affiliation(s)
- Moxun Xu
- College of Pharmacy, Jiamusi University, Jiamusi 154007, PR China
| | - Meitao Duan
- School of Pharmacy, Xiamen Medical College, Xiamen 361023, PR China
| | - Ming Chen
- School of Pharmacy, Xiamen Medical College, Xiamen 361023, PR China
| | - Ahmed Mahal
- Department of Medical Biochemical Analysis, College of Health Technology, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Lin Yang
- College of Pharmacy, Jiamusi University, Jiamusi 154007, PR China
| | - Chen Meng
- College of Pharmacy, Jiamusi University, Jiamusi 154007, PR China
| | - Zhiqiang Zhang
- School of Pharmacy, Xiamen Medical College, Xiamen 361023, PR China
| | - Jungang Ren
- School of Pharmacy, Xiamen Medical College, Xiamen 361023, PR China
| | - Ahmad J Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P. O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Shuxian Li
- College of Pharmacy, Jiamusi University, Jiamusi 154007, PR China.
| | - Chen Wang
- School of Pharmacy, Xiamen Medical College, Xiamen 361023, PR China.
| |
Collapse
|
5
|
Mostafa MAH, Khojah HMJ. Nanoparticle-based delivery systems for phytochemicals in cancer therapy: molecular mechanisms, clinical evidence, and emerging trends. Drug Dev Ind Pharm 2025:1-17. [PMID: 40116905 DOI: 10.1080/03639045.2025.2483425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/16/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
OBJECTIVE This review examines recent advancements in nanoparticle-based delivery systems for phytochemicals, focusing on their role in overcoming multidrug resistance, improving therapeutic efficacy, and facilitating clinical translation. SIGNIFICANCE This review highlights recent advances in nanoparticle-enabled phytochemical delivery to enhance bioavailability, improve therapeutic outcomes, and enable targeted applications. By comparing various nanoparticle systems, formulation methods, and efficacy data, it identifies gaps in current research and guides the development of more effective, next-generation phytochemical-loaded nanocarriers. METHODS A systematic review of literature published between 2000 and 2024 was conducted using PubMed, Scopus, and Web of Science. Articles focusing on nanoparticle-based phytochemical delivery in cancer therapy were included. KEY FINDINGS Compounds such as curcumin, resveratrol, quercetin, and epigallocatechin gallate demonstrate enhanced anti-cancer efficacy when encapsulated in nanoparticles, leading to improved bioavailability, increased tumor cell targeting, and reduced toxicity. Clinical trials indicate tumor regression and fewer adverse effects. Emerging approaches-such as nanogels, hybrid nanoparticles, and combination therapies with immune checkpoint inhibitors-further refine treatment efficacy. CONCLUSIONS Nanoparticle-based delivery systems significantly improve the therapeutic potential of phytochemicals, making them promising candidates for safer, more effective cancer treatments. However, challenges related to regulatory guidelines, scalability, and long-term safety must be addressed to fully realize their clinical potential.
Collapse
Affiliation(s)
- Mahmoud A H Mostafa
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University (Assiut Branch), Assiut, Egypt
| | - Hani M J Khojah
- Department of Pharmacy Practice, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| |
Collapse
|
6
|
Hosseinikhah SM, Farhoudi L, Mirzavi F, Vahdat-Lasemi F, Arabi L, Gheybi F, Sazgarnia A, Alavizadeh SH, Jaafari MR. Ultrasound-assisted efficient targeting of doxorubicin to the tumor microenvironment by lyso-thermosensitive liposomes of varying phase transition temperatures. Eur J Pharm Sci 2025; 206:107024. [PMID: 39863136 DOI: 10.1016/j.ejps.2025.107024] [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: 10/08/2024] [Revised: 01/04/2025] [Accepted: 01/23/2025] [Indexed: 01/27/2025]
Abstract
Premature drug release is the primary hindrance to the effective function of the lyso-thermosensitive liposomes (LTSLs) of doxorubicin (Dox), known as ThermoDox® for the treatment of cancer. Herein, we have optimized LTSLs by using a combination of phospholipids (PLs) with high transition temperatures (Tm) to improve the therapeutic outcome in an assisted ultrasound approach. For this, several Dox LTSLs were prepared using the remote loading method at varying molar ratios (0 to 90 %) of DPPC (Tm 41 °C) and HSPC (Tm 54.5 °C), as well as a constant molar ratio of MSPC (10 %), DSPE-mPEG2000 (4 %). The treatment efficacy was explored by using ultrasound as external hyperthermia (HT) (40-42℃) in mice bearing C26 murine colon carcinoma. All the formulations had an average diameter of around 110 nm, PDI ≤ 0.15, zeta potential of around -12 mV, and Dox encapsulation of >90 %. The cytotoxicity results indicated a higher IC50 value of Dox-LTSLs compared to the ThermoDox® (F0: DPPC:MSPC:DSPE-mPEG2000, 90:10:4), attributed to the faster Dox release in F0 formulation devoid of HSPC. Among various formulations, F25 (DPPC: MSPC: DSPE-mPEG2000: HSPC, 65:10:4:25) showed the highest cellular uptake at 42℃ and significantly improved the antitumor and survival efficacy in mice bearing C26 colon carcinoma in combination with ultrasonic HT compared to F0. Collectively, results demonstrated that optimizing the rigidity of the liposomal bilayers through the combinatorial selection of PLs of different transition temperatures could improve the plasma stability of the liposome, and hence ameliorate the outcome of therapy in assistance with an effective HT approach.
Collapse
Affiliation(s)
- Seyedeh Maryam Hosseinikhah
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Farhoudi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Fatemeh Vahdat-Lasemi
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Gheybi
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ameneh Sazgarnia
- Department of Medical Physics, Research Center of Medical Physics, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Zhang Z, Tang Y, Luo D, Qiu J, Chen L. Advances in nanotechnology for targeting cancer-associated fibroblasts: A review of multi-strategy drug delivery and preclinical insights. APL Bioeng 2025; 9:011502. [PMID: 40094065 PMCID: PMC11910205 DOI: 10.1063/5.0244706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) play a crucial role in the tumor microenvironment by promoting tumor growth, immune evasion, and metastasis. Recently, drug delivery systems targeting CAFs have emerged as a promising long-term and effective approach to cancer treatment. Advances in nanotechnology, in particular, have led to the development of nanomedicine delivery systems designed specifically to target CAFs, offering new possibilities for precise and personalized cancer therapies. This article reviews recent progress in drug delivery using nanocarriers that target CAFs. Additionally, we explore the potential of combining multiple therapies, such as chemotherapy and immunotherapy, with nanocarriers to enhance efficacy and overcome drug resistance. Although many preclinical studies show promise, the clinical application of nanomedicine still faces considerable challenges, especially in terms of drug penetration and large-scale production. Therefore, this review aims to provide a fresh perspective on CAF-targeted drug delivery systems and highlight potential future research directions and clinical applications.
Collapse
|
8
|
Li Z, Ran Q, Qu C, Hu S, Cui S, Zhou Y, Shen B, Yang B. Sigma-1 receptor activation attenuates DOX-induced cardiotoxicity by alleviating endoplasmic reticulum stress and mitochondrial calcium overload via PERK and IP3R-VDAC1-MCU signaling pathways. Biol Direct 2025; 20:23. [PMID: 40001213 PMCID: PMC11853590 DOI: 10.1186/s13062-025-00617-y] [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: 10/30/2024] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Doxorubicin (DOX) is an anthracycline with potent antitumor properties and rare yet serious cardiotoxic side effects that limit its clinical application. The sigma-1 receptor is a stress-triggered chaperone often dysregulated in diseases and has known cardioprotective effects. Although its anti-oxidative stress and anti-apoptotic effects have been demonstrated, its effectiveness in DOX-induced cardiotoxicity has never been explored. This study investigated the potential role of the activated sigma-1 receptor in a DOX-induced murine cardiotoxicity model to elucidate the receptor's mechanism of action. METHODS We established the model in C57BL/6 mice by daily intraperitoneal injections of fluvoxamine (Flv) for 4 consecutive weeks to activate the receptor and by weekly intraperitoneal injections of DOX at 5 mg/kg for 3 weeks. We performed in vitro experiments using cardiomyocytes of neonatal Sprague-Dawley rats to verify the protective effect of the sigma-1 receptor. RESULTS We found that sigma-1 expression in the heart decreased in the DOX-treated mice, and activating the receptor with Flv improved cardiac function. Moreover, Flv pretreatment inhibited cardiomyocyte apoptosis and endoplasmic reticulum stress and increased the expression of the Bcl2 apoptosis regulator (Bcl2), effectively alleviating the pathophysiological manifestations in mice. In addition, activating the receptor exerted cardioprotective effects by modulating endoplasmic reticulum stress through the PRKR-like endoplasmic reticulum kinase (PERK) signaling pathway. It also reduced mitochondrial and endoplasmic reticulum contact and alleviated mitochondrial calcium overload through the IP3R-VDAC1-MCU signaling pathway. CONCLUSION In conclusion, our study emphasizes the therapeutic potential of activating sigma-1 receptors against DOX-induced cardiotoxicity, suggesting sigma-1 receptors as potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Zixuan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Qian Ran
- Department of Cardiology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Shan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Shengyu Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - You Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Bo Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| |
Collapse
|
9
|
Jiang T, Jia T, Yin Y, Li T, Song X, Feng W, Wang S, Ding L, Chen Y, Zhang Q. Cuproptosis-Inducing Functional Nanocomposites for Enhanced and Synergistic Cancer Radiotherapy. ACS NANO 2025; 19:5429-5446. [PMID: 39895200 DOI: 10.1021/acsnano.4c13753] [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: 02/04/2025]
Abstract
Radiotherapy is crucial in local cancer management and needs advancements. Tumor cells elevate intracellular copper levels to promote growth and resist radiation; thus, targeted copper delivery to mitochondria could enhance radiotherapy by inducing cuproptosis in tumor cells. In this study, we engineered a multifunctional nanoliposome complex, termed Lipo-Ele@CuO2, which encapsulates both copper peroxide (CuO2) and the copper chelator elesclomol, which can delivery Cu ions to the mitochondria. The Lipo-Ele@CuO2 complex induces mitochondria-mediated cuproptosis in tumor cells and synergistically enhances the efficacy of radiotherapy. CuO2 acts as a copper donor and exhibits inherent sensitivity to acidic environments. Additionally, it depletes intracellular glutathione, thereby sensitizing cells to cuproptosis. Leveraging its pH-responsive properties in the acidic tumor microenvironment, the Lipo-Ele@CuO2 facilitate the controlled release of elesclomol, efficiently delivering copper ions to mitochondria at tumor sites. The combined in vitro and in vivo studies demonstrate that Lipo-Ele@CuO2-based therapy significantly improves antitumor efficacy and exhibits excellent safety profiles, effectively inducing cuproptosis in tumor cells and boosting the effectiveness of radiotherapy. Furthermore, metabolomic and transcriptomic analyses reveal that this combination therapy precipitates significant alterations in tumor energy metabolism, notably repressing genes related to iron-sulfur cluster assembly and glycolysis, thereby confirming the induction of cuproptosis. This therapeutic strategy provides a viable approach for addressing clinical radiotherapy resistance and demonstrates significant translational potential.
Collapse
Affiliation(s)
- Tiaoyan Jiang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, P. R. China
| | - Tianying Jia
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, P. R. China
| | - Yipengchen Yin
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, P. R. China
| | - Tianyu Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Sheng Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
| | - Li Ding
- Department of Medical Ultrasound, National Clinical Research Center of Interventional Medicine, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Tongji University, Shanghai 200072, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang 325088, P. R. China
| | - Qin Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, P. R. China
| |
Collapse
|
10
|
Buchalska B, Kamińska K, Kowara M, Sobiborowicz-Sadowska A, Cudnoch-Jędrzejewska A. Doxorubicin or Epirubicin Versus Liposomal Doxorubicin Therapy-Differences in Cardiotoxicity. Cardiovasc Toxicol 2025; 25:248-268. [PMID: 39810066 DOI: 10.1007/s12012-024-09952-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025]
Abstract
Doxorubicin (DOX) is an important drug used in the treatment of many malignancies. Unfortunately DOX causes various side effects, with cardiotoxicity being the most characteristic. Risk factors for DOX induced cardiotoxicity (DIC) include cumulative dose of DOX, preexisting cardiovascular diseases, dyslipidemia, diabetes, smoking, along with the use of other cardiotoxic agents. Development of DIC is associated with many pathological phenomena - increased oxidative stress, as well as upregulation of ferroptosis, apoptosis, necrosis, and autophagy. In DIC expression of many microRNAs is also deregulated. In order to avoid cardiotoxicity and still use DOX effectively DOX derivatives such as epirubicin were synthesized. Nowadays a new liposomal form of DOX (L-DOX) appeared as an alternative to conventional treatment with greatly reduced cardiotoxicity. L-DOX can be divided into two groups of substances - pegylated (PLD) with increased solubility and stability, and non-pegylated (NLPD). Many metaanalyses, clinical along with preclinical studies have shown L-DOX treatment is associated with a smaller decrease of left ventricular ejection fraction (LVEF) and other heart functions, but efficacy of this treatment is comparable to the use of convenctional DOX.
Collapse
Affiliation(s)
- Barbara Buchalska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Katarzyna Kamińska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland.
| | - Michał Kowara
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Aleksandra Sobiborowicz-Sadowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| |
Collapse
|
11
|
Iftode L, Cadinoiu AN, Raţă DM, Atanase LI, Vochiţa G, Rădulescu L, Popa M, Gherghel D. Double Peptide-Functionalized Carboxymethyl Chitosan-Coated Liposomes Loaded with Dexamethasone as a Potential Strategy for Active Targeting Drug Delivery. Int J Mol Sci 2025; 26:922. [PMID: 39940692 PMCID: PMC11816442 DOI: 10.3390/ijms26030922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/10/2025] [Accepted: 01/19/2025] [Indexed: 02/16/2025] Open
Abstract
Liposomes are intensively used as nanocarriers for biology, biochemistry, medicine, and in the cosmetics industry and their non-toxic and biocompatible nature makes these vesicles attractive systems for biomedical applications. Moreover, the conjugation of specific ligands to liposomes increases their cellular uptake and therapeutic efficiency. Considering these aspects, the aim of the present study was to obtain new formulations of cationic liposomes coated with dual-peptide functionalized carboxymethyl chitosan (CMCS) for the treatment of inner ear diseases. In order to achieve efficient active targeting and ensuring a high efficacy of the treatment, CMCS was functionalized with Tet1 peptide, to target specific ear cells, and TAT peptide, to ensure cellular penetration. Furthermore, dexamethasone phosphate was loaded as a model drug for the treatment of ear inflammation. The infrared spectroscopy confirmed the functionalization of CMCS with the two specific peptides. The mean diameter of the uncovered liposomes varied between 167 and 198 nm whereas the CMCS-coated liposomes ranged from 179 to 202 nm. TEM analysis showed the spherical shape and unilamellar structure of liposomes. The release efficiency of dexamethasone phosphate after 24 h from the uncoated liposomes was between 37 and 40% and it appeared that the coated liposomes modulated this release. The obtained results demonstrated that the liposomes are hemocompatible since, for a tested concentration of 100 µg/mL, the liposome suspension had a lysis of erythrocytes lower than 2.5% after 180 min of incubation. In addition, the peptide-functionalized CMCS-coated liposomes induced a non-significant effect on the viability of normal V79-4 cells after 48 h, at the highest doses. Values of 71.31% were recorded (CLCP-1), 77.28% (CLCP-2) and 74.36% (CLCP-3), correlated with cytotoxic effects of 28.69%, 22.72%, and 25.64%.
Collapse
Affiliation(s)
- Loredana Iftode
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.I.); (L.R.)
- “Cristofor Simionescu” Faculty of Chemical Engineering and Environmental Protection, “Gheorghe Asachi” Technical University, 700050 Iasi, Romania
| | - Anca Niculina Cadinoiu
- Department of Biomaterials, Faculty of Medical Dentistry, “Apollonia” University of Iasi, 700511 Iasi, Romania; (D.M.R.); (L.I.A.)
| | - Delia Mihaela Raţă
- Department of Biomaterials, Faculty of Medical Dentistry, “Apollonia” University of Iasi, 700511 Iasi, Romania; (D.M.R.); (L.I.A.)
| | - Leonard Ionuț Atanase
- Department of Biomaterials, Faculty of Medical Dentistry, “Apollonia” University of Iasi, 700511 Iasi, Romania; (D.M.R.); (L.I.A.)
- Academy of Romanian Scientists, 050044 Bucharest, Romania
| | - Gabriela Vochiţa
- Institute of Biological Research Iasi, Branch of NIRDBS—National Institute of Research and Development of Biological Sciences Bucharest, 700107 Iasi, Romania; (G.V.); (D.G.)
| | - Luminița Rădulescu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.I.); (L.R.)
| | - Marcel Popa
- “Cristofor Simionescu” Faculty of Chemical Engineering and Environmental Protection, “Gheorghe Asachi” Technical University, 700050 Iasi, Romania
- Department of Biomaterials, Faculty of Medical Dentistry, “Apollonia” University of Iasi, 700511 Iasi, Romania; (D.M.R.); (L.I.A.)
- Academy of Romanian Scientists, 050044 Bucharest, Romania
| | - Daniela Gherghel
- Institute of Biological Research Iasi, Branch of NIRDBS—National Institute of Research and Development of Biological Sciences Bucharest, 700107 Iasi, Romania; (G.V.); (D.G.)
| |
Collapse
|
12
|
Mancoo A, Silva M, Lopes C, Loureiro M, Pinto V, Ramalho JFCB, Carvalho P, Gouveia CAJ, Rocha S, Bordeira SMP, Sampaio PM, Turpin A, Gersen H, Mumtaz M. Toward Resolving Heterogeneous Mixtures of Nanocarriers in Drug Delivery Systems through Light Scattering and Machine Learning. ACS NANO 2025; 19:2388-2404. [PMID: 39772474 DOI: 10.1021/acsnano.4c12963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Nanocarriers (NCs) have emerged as a revolutionary approach in targeted drug delivery, promising to enhance drug efficacy and reduce toxicity through precise targeting and controlled release mechanisms. Despite their potential, the clinical adoption of NCs is hindered by challenges in their physicochemical characterization, essential for ensuring drug safety, efficacy, and quality control. Traditional characterization methods, such as dynamic light scattering and nanoparticle tracking analysis, offer limited insights, primarily focusing on particle size and concentration, while techniques like high-performance liquid chromatography and mass spectrometry are hampered by extensive sample preparation, high costs, and potential sample degradation. Addressing these limitations, this work presents a cost-effective methodology leveraging light scattering and optical forces, combined with machine learning algorithms, to characterize polydisperse nanoparticle mixtures, including lipid-based NCs. We prove that our approach provides quantification of the relative concentration of complex nanoparticle suspensions by detecting changes in refractive index and polydispersity without extensive sample preparation or destruction, offering a high-throughput solution for NC characterization in drug delivery systems. Experimental validation demonstrates the method's efficacy in characterizing commercially available synthetic nanoparticles and Doxoves, a liposomal formulation of Doxorubicin used in cancer treatment, marking a significant advancement toward reliable, noninvasive characterization techniques that can accelerate the clinical translation of nanocarrier-based therapeutics.
Collapse
Affiliation(s)
- Allan Mancoo
- iLoF-Intelligent Lab on Fiber, Rua de Godim 389, 4300 Porto, Portugal
| | - Mariana Silva
- iLoF-Intelligent Lab on Fiber, Rua de Godim 389, 4300 Porto, Portugal
| | - Claudia Lopes
- iLoF-Intelligent Lab on Fiber, Rua de Godim 389, 4300 Porto, Portugal
| | - Maria Loureiro
- iLoF-Intelligent Lab on Fiber, Rua de Godim 389, 4300 Porto, Portugal
| | - Vanessa Pinto
- iLoF-Intelligent Lab on Fiber, Rua de Godim 389, 4300 Porto, Portugal
| | | | - Patricia Carvalho
- iLoF-Intelligent Lab on Fiber, Rua de Godim 389, 4300 Porto, Portugal
| | | | - Sara Rocha
- iLoF-Intelligent Lab on Fiber, Rua de Godim 389, 4300 Porto, Portugal
| | | | - Paula M Sampaio
- iLoF-Intelligent Lab on Fiber, Rua de Godim 389, 4300 Porto, Portugal
- Departamento de Bioquímica, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Alex Turpin
- iLoF-Intelligent Lab on Fiber, Rua de Godim 389, 4300 Porto, Portugal
- School of Physics and Astronomy, University of Glasgow, G12 8QQ Glasgow, U.K
| | - Henkjan Gersen
- iLoF-Intelligent Lab on Fiber, Rua de Godim 389, 4300 Porto, Portugal
- H.H. Wills Physics Laboratory, University of Bristol, BS8 1TL Bristol, U.K
| | - Mehak Mumtaz
- iLoF-Intelligent Lab on Fiber, Rua de Godim 389, 4300 Porto, Portugal
| |
Collapse
|
13
|
Basingab FS, Alshahrani OA, Alansari IH, Almarghalani NA, Alshelali NH, Alsaiary AH, Alharbi N, Zaher KA. From Pioneering Discoveries to Innovative Therapies: A Journey Through the History and Advancements of Nanoparticles in Breast Cancer Treatment. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:27-51. [PMID: 39867813 PMCID: PMC11761866 DOI: 10.2147/bctt.s501448] [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: 10/17/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025]
Abstract
Nanoparticle technology has revolutionized breast cancer treatment by offering innovative solutions addressing the gaps in traditional treatment methods. This paper aimed to comprehensively explore the historical journey and advancements of nanoparticles in breast cancer treatment, highlighting their transformative impact on modern medicine. The discussion traces the evolution of nanoparticle-based therapies from their early conceptualization to their current applications and future potential. We initially explored the historical context of breast cancer treatment, highlighting the limitations of conventional therapies, such as surgery, radiation, and chemotherapy. The advent of nanotechnology has introduced a new era characterized by the development of various nanoparticles, including liposomes, dendrimers, and gold nanoparticles, designed to target cancer cells with remarkable precision. We further described the mechanisms of action for nanoparticles, including passive and active targeting, and reviewed significant breakthroughs and clinical trials that have validated their efficacy. Current applications of nanoparticles in breast cancer treatment have been examined, showcasing clinically approved therapies and comparing their effectiveness with traditional methods. This article also discusses the latest advancements in nanoparticle research, including drug delivery systems and combination therapy innovations, while addressing the current technical, biological, and regulatory challenges. The technical challenges include efficient and targeted delivery to tumor sites without affecting healthy tissue; biological, such as potential toxicity, immune system activation, or resistance mechanisms; economic, involving high production and scaling costs; and regulatory, requiring rigorous testing for safety, efficacy, and long-term effects to meet stringent approval standards. Finally, we have explored emerging trends, the potential for personalized medicine, and the ethical and social implications of this transformative technology. In conclusion, through comprehensive analysis and case studies, this paper underscores the profound impact of nanoparticles on breast cancer treatment and their future potential.
Collapse
Affiliation(s)
- Fatemah S Basingab
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Omniah A Alshahrani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Ibtehal H Alansari
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Nada A Almarghalani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Nada H Alshelali
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Abeer Hamad Alsaiary
- Biology Department, College of Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Najwa Alharbi
- Department of Biology Science, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Kawther A Zaher
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| |
Collapse
|
14
|
Dragomir RD, Mercioni MA, Negru Ș, Popovici D, Săftescu S, Blidari AR, Sas I. Comparison of Hepatic Function and Chemotherapy-Induced Side Effects Between Pegylated Liposomal Doxorubicin (PLD), Topotecan (TOPO), and Gemcitabine in Platinum-Resistant Ovarian Cancer (PROC). J Pers Med 2025; 15:39. [PMID: 39852231 PMCID: PMC11766750 DOI: 10.3390/jpm15010039] [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: 11/02/2024] [Revised: 12/13/2024] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: Platinum-resistant ovarian cancer (PROC) is a major therapeutic challenge, as it responds poorly to standard platinum-based treatment, has limited treatment options, and offers a generally unfavorable prognosis. Chemotherapeutic agents like pegylated liposomal doxorubicin (PLD), topotecan (TOPO), and gemcitabine (GEM) are used for this setting, but with varying efficacy and toxicity profiles, leading to an increasing need to understand the optimal balance between treatment effectiveness and tolerability for improving patient outcomes. This study evaluates the efficacy and side effects of PLD, TOPO, and GEM, focusing on progression-free survival (PFS), overall survival (OS), and safety profiles. Methods: We conducted a retrospective observational study that included 856 PROC patients treated with PLD (n = 383), TOPO (n = 352), or GEM (n = 121) at the OncoHelp Oncology Center from January 2018 to December 2023. Inclusion criteria encompass diagnosis, prior platinum therapy, and Eastern Cooperative Oncology Group (ECOG) status (0-2). Treatment protocols followed standard dosing, with adjustments for toxicity. Primary endpoints included PFS and OS, with safety assessed by incidence of grade 3 and 4 toxicities per CTCAE v5.0. Kaplan-Meier analysis and Cox regression were used to compare survival, and statistical significance was set at p < 0.05. Results: TOPO showed higher toxicity than PLD and GEM, including liver damage, hematological and non-hematological side effects, while PLD induced more skin toxicity. In terms of survival, minor differences were seen between the three chemotherapeutic agents, with a slight advantage for PLD for better disease control. Conclusions: Given the comparable results in OS across the regimens, treatment decisions should be based on other factors such as patient tolerance and quality of life.
Collapse
Affiliation(s)
- Radu-Dumitru Dragomir
- Department of Obstetrics and Gynecology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (R.-D.D.); (I.S.)
| | - Marina Adriana Mercioni
- Faculty of Electronics, Telecommunications and Information Technologies, Politehnica University, 300223 Timișoara, Romania
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Șerban Negru
- Department of Oncology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (Ș.N.); (D.P.); (S.S.)
| | - Dorel Popovici
- Department of Oncology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (Ș.N.); (D.P.); (S.S.)
| | - Sorin Săftescu
- Department of Oncology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (Ș.N.); (D.P.); (S.S.)
| | - Andiana Roxana Blidari
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Ioan Sas
- Department of Obstetrics and Gynecology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (R.-D.D.); (I.S.)
| |
Collapse
|
15
|
Hillmann J, Maass N, Bauerschlag DO, Flörkemeier I. Promising new drugs and therapeutic approaches for treatment of ovarian cancer-targeting the hallmarks of cancer. BMC Med 2025; 23:10. [PMID: 39762846 PMCID: PMC11706140 DOI: 10.1186/s12916-024-03826-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Ovarian cancer remains the most lethal gynecological malignancy. Despite the approval of promising targeted therapy such as bevacizumab and PARP inhibitors, 5-year survival has not improved significantly. Thus, there is an urgent need for new therapeutics. New advancements in therapeutic strategies target the pivotal hallmarks of cancer. This review is giving an updated overview of innovative and upcoming therapies for the treatment of ovarian cancer that focuses specific on the hallmarks of cancer. The hallmarks of cancer constitute a broad concept to reenact complexity of malignancies and furthermore identify possible targets for new treatment strategies. For this purpose, we analyzed approvals and current clinical phase III studies (registered at ClinicalTrials.gov (National Library of Medicine, National Institutes of Health; U.S. Department of Health and Human Services, 2024)) for new drugs on the basis of their mechanisms of action and identified new target approaches. A broad spectrum of new promising drugs is currently under investigation in clinical phase III studies targeting mainly the hallmarks "self-sufficiency in growth signals," "genomic instability," and "angiogenesis." The benefit of immune checkpoint inhibitors in ovarian cancer has been demonstrated for the first time. Besides, targeting the tumor microenvironment is of growing interest. Replicative immortality, energy metabolism, tumor promoting inflammation, and the microbiome of ovarian cancer are still barely targeted by drugs. Nevertheless, precision medicine, which focuses on specific disease characteristics, is becoming increasingly important in cancer treatment.
Collapse
Affiliation(s)
- Julia Hillmann
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Nicolai Maass
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Dirk O Bauerschlag
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany.
- Department of Gynaecology, Jena University Hospital, Jena, Germany.
| | - Inken Flörkemeier
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany.
| |
Collapse
|
16
|
Tayal R, Mannan A, Singh S, Dhiman S, Singh TG. Unveiling the Complexities: Exploring Mechanisms of Anthracyclineinduced Cardiotoxicity. Curr Cardiol Rev 2025; 21:42-77. [PMID: 39484769 DOI: 10.2174/011573403x322928241021100631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 11/03/2024] Open
Abstract
The coexistence of cancer and heart disease, both prominent causes of illness and death, is further exacerbated by the detrimental impact of chemotherapy. Anthracycline-induced cardiotoxicity is an unfortunate side effect of highly effective therapy in treating different types of cancer; it presents a significant challenge for both clinicians and patients due to the considerable risk of cardiotoxicity. Despite significant progress in understanding these mechanisms, challenges persist in identifying effective preventive and therapeutic strategies, rendering it a subject of continued research even after three decades of intensive global investigation. The molecular targets and signaling pathways explored provide insights for developing targeted therapies, emphasizing the need for continued research to bridge the gap between preclinical understanding and clinical applications. This review provides a comprehensive exploration of the intricate mechanisms underlying anthracycline-induced cardiotoxicity, elucidating the interplay of various signaling pathways leading to adverse cellular events, including cardiotoxicity and death. It highlights the extensive involvement of pathways associated with oxidative stress, inflammation, apoptosis, and cellular stress responses, offering insights into potential and unexplored targets for therapeutic intervention in mitigating anthracycline-induced cardiac complications. A comprehensive understanding of the interplay between anthracyclines and these complexes signaling pathways is crucial for developing strategies to prevent or mitigate the associated cardiotoxicity. Further research is needed to outline the specific contributions of these pathways and identify potential therapeutic targets to improve the safety and efficacy of anthracycline-based cancer treatment. Ultimately, advancements in understanding anthracycline-induced cardiotoxicity mechanisms will facilitate the development of more efficacious preventive and treatment approaches, thereby improving outcomes for cancer patients undergoing anthracycline-based chemotherapy.
Collapse
Affiliation(s)
- Rohit Tayal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | |
Collapse
|
17
|
Jiao Y, Yang L, Wang R, Song G, Fu J, Wang J, Gao N, Wang H. Drug Delivery Across the Blood-Brain Barrier: A New Strategy for the Treatment of Neurological Diseases. Pharmaceutics 2024; 16:1611. [PMID: 39771589 PMCID: PMC11677317 DOI: 10.3390/pharmaceutics16121611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
The blood-brain barrier (BBB) serves as a highly selective barrier between the blood and the central nervous system (CNS), and its main function is to protect the brain from foreign substances. This physiological property plays a crucial role in maintaining CNS homeostasis, but at the same time greatly limits the delivery of drug molecules to the CNS, thus posing a major challenge for the treatment of neurological diseases. Given that the high incidence and low cure rate of neurological diseases have become a global public health problem, the development of effective BBB penetration technologies is important for enhancing the efficiency of CNS drug delivery, reducing systemic toxicity, and improving the therapeutic outcomes of neurological diseases. This review describes the physiological and pathological properties of the BBB, as well as the current challenges of trans-BBB drug delivery, detailing the structural basis of the BBB and its role in CNS protection. Secondly, this paper reviews the drug delivery strategies for the BBB in recent years, including physical, biological and chemical approaches, as well as nanoparticle-based delivery technologies, and provides a comprehensive assessment of the effectiveness, advantages and limitations of these delivery strategies. It is hoped that the review in this paper will provide valuable references and inspiration for future researchers in therapeutic studies of neurological diseases.
Collapse
Affiliation(s)
- Yimai Jiao
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Luosen Yang
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300401, China;
| | - Rujuan Wang
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Guoqiang Song
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Jingxuan Fu
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300401, China;
| | - Jinping Wang
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Na Gao
- Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China
| | - Hui Wang
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| |
Collapse
|
18
|
Muhammad FA, Altalbawy FMA, Mandaliya V, Saraswat SK, Rekha MM, Aulakh D, Chahar M, Mahdi MS, Jaber MA, Alhadrawi M. Targeting breast tumor extracellular matrix and stroma utilizing nanoparticles. Clin Transl Oncol 2024:10.1007/s12094-024-03793-x. [PMID: 39692807 DOI: 10.1007/s12094-024-03793-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024]
Abstract
Breast cancer is a complicated malignancy and is known as the most common cancer in women. Considerable experiments have been devoted to explore the basic impacts of the tumor stroma, particularly the extracellular matrix (ECM) and stromal components, on tumor growth and resistance to treatment. ECM is made up of an intricate system of proteins, glycosaminoglycans, and proteoglycans, and maintains structural support and controls key signaling pathways involved in breast tumors. ECM can block different drugs such as chemotherapy and immunotherapy drugs from entering the tumor stroma. Furthermore, the stromal elements, such as cancer-associated fibroblasts (CAFs), immune cells, and blood vessels, have crucial impacts on tumor development and therapeutic resistance. Recently, promising outcomes have been achieved in using nanotechnology for delivering drugs to tumor stroma and crossing ECM in breast malignancies. Nanoparticles have various benefits for targeting the breast tumor stroma, such as improved permeability and retention, extended circulation time, and the ability to actively target the area. This review covers the latest developments in nanoparticle therapies that focus on breast tumor ECM and stroma. We will explore different approaches using nanoparticles to target the delivery of anticancer drugs like chemotherapy, small molecule drugs, various antitumor products, and other specific synthetic therapeutic agents to the breast tumor stroma. Furthermore, we will investigate the utilization of nanoparticles in altering the stromal elements, such as reprogramming CAFs and immune cells, and also remodeling ECM.
Collapse
Affiliation(s)
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, 12613, Egypt.
| | - Viralkumar Mandaliya
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | | | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Damanjeet Aulakh
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology Chitkara University, Rajpura, Punjab, 140401, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering and Technology, NIMS University Rajasthan, Jaipur, India
| | | | | | - Merwa Alhadrawi
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
19
|
Díaz-Tejeiro C, López de Sá A, Poyatos-Racionero E, Ballestín P, Bartolomé J, Calvo E, Moreno V, Moris F, Pérez-Segura P, Gyorffy B, Pandiella A, Ocaña A. Understanding the Preclinical Efficacy of Antibody-Drug Conjugates. Int J Mol Sci 2024; 25:12875. [PMID: 39684584 DOI: 10.3390/ijms252312875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Antibody-drug conjugates (ADCs) represent a therapeutic modality that guides chemotherapies to tumoral cells by using antibodies against tumor-associated antigens (TAAs). The antibody and the chemotherapy or payload are attached by a chemical structure called the linker. The strategy for the development of this type of drug was based on several rational pillars, including the use of a very potent payload and the use of specific antibodies acting only on antigens expressed on tumoral cells. In this article, by using data from all approved ADCs that have received regulatory approval, we analyze the potential contribution of each ADC component to preclinical activity. We suggest that payload potency and the drug-to-antibody ratio (DAR) have a less relevant role in relation to efficacy than previously considered. Additionally, we have observed that some ADCs have been developed against antigens also present in non-transformed tissues, which could suggest that TAA specificity is not a mandatory requirement. Finally, we have identified that ADCs with payloads harboring more favorable physicochemical characteristics showed better potential activity. In this article, we also review other aspects that should be taken into consideration for ADC design, including linker structure, stability, conjugation type, pharmacokinetics, receptor internalization, and recycling. Based on currently available data, our study summaries different concepts that should be considered in the design of novel ADCs in the future.
Collapse
Affiliation(s)
- Cristina Díaz-Tejeiro
- Experimental Therapeutics Unit, Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Alfonso López de Sá
- Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | | | - Pablo Ballestín
- Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Jorge Bartolomé
- Experimental Therapeutics Unit, Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
- Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Emiliano Calvo
- START Madrid Centro Integral Oncológico Clara Campal, 28050 Madrid, Spain
| | - Víctor Moreno
- START Madrid-FJD, Hospital Fundación Jiménez Díaz, 28040 Madrid, Spain
| | | | - Pedro Pérez-Segura
- Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Balazs Gyorffy
- Department of Biophysics, Medical School, University of Pecs, H-7624 Pecs, Hungary
- Department of Bioinformatics, Semmelweis University, H-1094 Budapest, Hungary
- Research Centre for Natural Sciences, Cancer Biomarker Research Group, Institute of Enzymology, Magyar Tudósok Krt. 2, H-1117 Budapest, Hungary
| | - Atanasio Pandiella
- Cancer Research Center, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), 28029 Madrid, Spain
| | - Alberto Ocaña
- Experimental Therapeutics Unit, Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
- Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
- START Madrid-FJD, Hospital Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
20
|
Mashweu AR, Azov VA. Nanotechnology in Drug Delivery: Anatomy and Molecular Insight into the Self-Assembly of Peptide-Based Hydrogels. Molecules 2024; 29:5654. [PMID: 39683812 PMCID: PMC11643151 DOI: 10.3390/molecules29235654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
The bioavailability, release, and stability of pharmaceuticals under physicochemical conditions is the major cause of drug candidates failing during their clinical trials. Therefore, extensive efforts have been invested in the development of novel drug delivery systems that are able to transport drugs to a desired site and improve bioavailability. Hydrogels, and peptide hydrogels in particular, have been extensively investigated due to their excellent biocompatibility and biodegradability properties. However, peptide hydrogels often have weak mechanical strength, which limits their therapeutic efficacy. Therefore, a number of methods for improving their rheological properties have been established. This review will cover the broad area of drug delivery, focusing on the recent developments in this research field. We will discuss the variety of different types of nanocarrier drug delivery systems and then, more specifically, the significance and perspectives of peptide-based hydrogels. In particular, the interplay of intermolecular forces that govern the self-assembly of peptide hydrogels, progress made in understanding the distinct morphologies of hydrogels, and applications of non-canonical amino acids in hydrogel design will be discussed in more detail.
Collapse
Affiliation(s)
- Adelaide R. Mashweu
- Department of Chemistry, University of the Free State, P.O. Box 339, Bloemfontein 9300, South Africa
| | - Vladimir A. Azov
- Department of Chemistry, University of the Free State, P.O. Box 339, Bloemfontein 9300, South Africa
| |
Collapse
|
21
|
Sukumar VK, Tai YK, Chan CW, Iversen JN, Wu KY, Fong CHH, Lim JSJ, Franco-Obregón A. Brief Magnetic Field Exposure Stimulates Doxorubicin Uptake into Breast Cancer Cells in Association with TRPC1 Expression: A Precision Oncology Methodology to Enhance Chemotherapeutic Outcome. Cancers (Basel) 2024; 16:3860. [PMID: 39594815 PMCID: PMC11592624 DOI: 10.3390/cancers16223860] [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: 10/21/2024] [Revised: 11/04/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Doxorubicin (DOX) is commonly used as a chemotherapeutic agent for the treatment of breast cancer. Nonetheless, its systemic delivery via intravenous injection and toxicity towards healthy tissues commonly result in a broad range of detrimental side effects. Breast cancer severity was previously shown to be correlated with TRPC1 channel expression that conferred upon it enhanced vulnerability to pulsed electromagnetic field (PEMF) therapy. PEMF therapy was also previously shown to enhance breast cancer cell vulnerability to DOX in vitro and in vivo that correlated with TRPC1 expression and mitochondrial respiratory rates. Methods: DOX uptake was assessed by measuring its innate autofluorescence within murine 4T1 or human MCF7 breast cancer cells following magnetic exposure. Cellular vulnerability to doxorubicin uptake was assessed by monitoring mitochondrial activity and cellular DNA content. Results: Here, we demonstrate that 10 min of PEMF exposure could augment DOX uptake into 4T1 and MCF7 breast cancer cells. DOX uptake could be increased by TRPC1 overexpression, whereas inhibiting the activity of TRPC1 channels with SKF-96356 or genetic knockdown, precluded DOX uptake. PEMF exposure enhances DOX-mediated killing of breast cancer cells, reducing the IC50 value of DOX by half, whereas muscle cells, representative of collateral tissues, were less sensitive to PEMF-enhanced DOX-mediated cytotoxicity. Vesicular loading of DOX correlated with TRPC1 expression. Conclusions: This study presents a novel TRPC1-mediated mechanism through which PEMF therapy may enhance DOX cytotoxicity in breast cancer cells, paving the way for the development of localized non-invasive PEMF platforms to improve cancer outcomes with lower systemic levels of DOX.
Collapse
Affiliation(s)
- Viresh Krishnan Sukumar
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Yee Kit Tai
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Ching Wan Chan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Jan Nikolas Iversen
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Kwan Yu Wu
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Charlene Hui Hua Fong
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Joline Si Jing Lim
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- Experimental Therapeutics Programme, Cancer Science Institute, Singapore 117599, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore 119228, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore 119074, Singapore
| | - Alfredo Franco-Obregón
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| |
Collapse
|
22
|
Liu Y, Zhang D, Kong M, Wang Y, Mei H, Shan C, Meng J, Zou Y, Wang J. Synaptic vesicle protein 2-targeted doxorubicin-loaded liposome for effective neuroblastoma therapy. Biomed Pharmacother 2024; 180:117548. [PMID: 39413621 DOI: 10.1016/j.biopha.2024.117548] [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: 07/08/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024] Open
Abstract
Neuroblastoma, a pediatric cancer originating from neural crest tissues of the sympathetic nervous system, poses significant treatment challenges due to its molecular diversity and restricted druggable targets. While chemotherapy is a common treatment, its drawbacks, including poor targeting of cancer cells and nonspecific cytotoxicity, highlight the urgent need for innovative and effective therapeutic strategies. Herein, we developed a novel drug by coupling the receptor binding domain of botulinum neurotoxin type A (Hc) fused with monomeric streptavidin (mSA) to biotin coated doxorubicin (Dox)-loaded liposome, via interaction between mSA and biotin. The resultant Hc-coated liposome (Hc-Lipo@Dox) actively targeted the recycling synaptic vesicle 2 protein (SV2) abundantly expressed on the surface of neuroblastoma cells. Our results revealed that Hc-Lipo@Dox more effectively entered the neuroblastoma SH-SY5Y cells, inducing apoptosis compared to non-targeted liposome and free Dox. Moreover, Hc-Lipo@Dox rapidly enriched Dox in the subcutaneously implanted neuroblastoma tumor in nude mice, resulting potent anti-neuroblastoma effect compared to non-targeted liposomes or free Dox. Importantly, Hc-Lipo@Dox significantly improved the survival rate of treated mice, while also exhibiting a favorable safety profile with no discernible impact on mobility or observable side effects. These findings highlight the potential of SV2-targeted Dox liposome as a promising and well-tolerated chemotherapy approach for neuroblastoma treatment. Moreover, the technology established here has broader applications for various cancer therapies by substituting the Hc moiety with other tumor-specific targeting moieties.
Collapse
Affiliation(s)
- Yang Liu
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Dongya Zhang
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Miaomiao Kong
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Yibin Wang
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Huiyuan Mei
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Chunxu Shan
- School of Biotechnology, Dublin City University, Collins Avenue, Dublin, Ireland.
| | - Jianghui Meng
- School of Biotechnology, Dublin City University, Collins Avenue, Dublin, Ireland.
| | - Yan Zou
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Jiafu Wang
- School of Biotechnology, Dublin City University, Collins Avenue, Dublin, Ireland.
| |
Collapse
|
23
|
Alsunbul M, El-Masry TA, El Zahaby EI, Gaballa MMS, El-Nagar MMF. Potential Protective Effect of Orlistat: A Formulation of Nanocrystals Targeting Inflammation, Oxidative Stress, and Apoptosis in an Experimental Model of Doxorubicin-Induced Cardiotoxicity. Pharmaceutics 2024; 16:1356. [PMID: 39598480 PMCID: PMC11597902 DOI: 10.3390/pharmaceutics16111356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Doxorubicin (DOX) is a widely used chemotherapeutic agent; nevertheless, cardiotoxicity limits its effectiveness. Orlistat (Orli) is an irreversible lipase enzyme inhibitor with poor solubility and bioavailability. Furthermore, Orli has a favorable impact on the decrease in cardiometabolic risk variables. Thus, this study aimed to investigate the novel use of Orlistat Nanocrystals (Orli-Nanocrystals) to mitigate DOX-induced cardiotoxicity and to identify probable pathways behind the cardioprotective effects. Methods: The pharmacokinetic parameters-area under % dose/g heart time curve (AUC0→4h), Drug targeting index (DTI), and relative targeting efficiency (RTE)-were calculated. Furthermore, experimental design mice were categorized into six groups: a (1) Normal control group, (2) Orli-Free group, (3) Orli-Nanocrystals group, (4) DOX group, (5) Orli-Free-DOX group, and (6) Orli-Nanocrystals-DOX group. All treatments were intraperitoneally injected once daily for 14 days with a single dose of DOX (15 mg/kg) on the 12th day for 4, 5, and 6 groups. Results: The pharmacokinetic parameters (Cmax, AUC) following oral administration of Orli-Nanocrystals presented a significant difference (higher values) in comparison to Orli due to the enhanced extent of the absorption of nanocrystals and, subsequently, their distribution to the heart. The study results indicated that DOX caused significant cardiotoxicity, as revealed by a remarkable rise in cardiac function biomarkers like LDH and CK-MB, which involve enzyme activities. Additionally, cardiac MDA content also increased; however, glutathione peroxidase, catalase, and superoxide dismutase activities were decreased. In the same context, DOX was found to have a remarkable downregulation in Nrf2, HO-1, Sirt-1, and Bcl2, while the upregulation of NF-κB, TNF-α, and BAX gene and protein expression occurred. Pretreatment with Orli-Nanocrystals displayed the most notable recovery of the altered immunohistochemical, histological, and biochemical characteristics as compared to the Orli-Free group. Conclusions: This work is the first investigation into the potential use of antioxidant, anti-inflammatory, and anti-apoptotic characteristics of Orli-Nanocrystals to protect against DOX-induced cardiotoxicity in vivo.
Collapse
Affiliation(s)
- Maha Alsunbul
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia;
| | - Thanaa A. El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt;
| | - Enas I. El Zahaby
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 35712, Egypt;
| | - Mohamed M. S. Gaballa
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt;
| | - Maysa M. F. El-Nagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt;
| |
Collapse
|
24
|
Alzahrani RR, Alkhulaifi MM, Al Jeraisy M, Albekairy AM, Ali R, Alrfaei BM, Ehaideb SN, Al-Asmari AI, Qahtani SA, Halwani A, Yassin AEB, Halwani MA. Enhancing Gentamicin Antibacterial Activity by Co-Encapsulation with Thymoquinone in Liposomal Formulation. Pharmaceutics 2024; 16:1330. [PMID: 39458659 PMCID: PMC11510464 DOI: 10.3390/pharmaceutics16101330] [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: 08/04/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND AND PURPOSE Gentamicin (GEN) is a broad-spectrum antibiotic that cannot be prescribed freely because of its toxicity. Thymoquinone (THQ), a phytochemical, has antibacterial, antioxidant, and toxicity-reducing properties. However, its hydrophobicity and light sensitivity make it challenging to utilize. This incited the idea of co-encapsulating GEN and THQ in liposomes (Lipo-GEN-THQ). METHOD Lipo-GEN-THQ were characterized using the zeta-potential, dynamic light scattering, Fourier transform infrared spectroscopy, and transmission electron microscope (TEM). The liposomes' stability was evaluated under different storage and biological conditions. Lipo-GEN-THQ's efficacy was investigated by the minimum inhibitory/bactericidal concentrations (MICs-MBCs), time-kill curves, and antibiofilm and antiadhesion assays. Bacterial interactions with the empty and GEN-THQ-loaded liposomes were evaluated using TEM. RESULTS The Lipo-GEN-THQ were spherical, monodispersed, and negatively charged. The Lipo-GEN-THQ were relatively stable and released GEN sustainably over 24 h. The liposomes exhibited significantly higher antibacterial activity than free GEN, as evidenced by the four-fold lower MIC and biofilm eradication in resistant E. coli strain (EC-219). TEM images display how the empty liposomes fused closely to the tested bacteria and how the loaded liposomes caused ultrastructure damage and intracellular component release. An antiadhesion assay showed that the Lipo-GEN-THQ and free GEN (0.125 mg/L) similarly inhibited Escherichia coli (EC-157) adhesion to the A549 cells (68% vs. 64%). CONCLUSIONS The Lipo-THQ-GEN enhanced GEN by combining it with THQ within the liposomes, reducing the effective dose. The reduction in the GEN dose after adding THQ may indirectly reduce the toxicity and aid in developing an enhanced and safer form of GEN.
Collapse
Affiliation(s)
- Raghad R. Alzahrani
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (R.R.A.); (M.M.A.)
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Manal M. Alkhulaifi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (R.R.A.); (M.M.A.)
| | - Majed Al Jeraisy
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia; (M.A.J.); (A.H.)
| | - Abdulkareem M. Albekairy
- Department of Pharmacy Practice, College of Pharmacy, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia;
- Pharmaceutical Care Department, King Abdulaziz Medical City, National Guard Health Affairs, Riyadh 11481, Saudi Arabia
| | - Rizwan Ali
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia;
| | - Bahauddeen M. Alrfaei
- Stem Cells and Regenerative Medicine, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, National Guard Health Affairs, Riyadh 11481, Saudi Arabia;
| | - Salleh N. Ehaideb
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard—Health Affairs, Riyadh 11481, Saudi Arabia;
| | - Ahmed I. Al-Asmari
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Sultan Al Qahtani
- Department of Basic Medical Sciences, College of Medicine, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia;
| | - Abdulaziz Halwani
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia; (M.A.J.); (A.H.)
- College of Dentistry, King Saud bin Abdul Aziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Alaa Eldeen B. Yassin
- College of Pharmacy, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Majed A. Halwani
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| |
Collapse
|
25
|
Li W, Xu K, Lan M, Gao J, Dou L, Yang Y, Wang Q, Yan M, Li S, Ma Q, Tian W, Chen B, Cui J, Zhang X, Cai J, Wang H, Sun L, Li J, Huang X, Shen T. The dual functions of the pentacyclic triterpenoid madecassic acid in ameliorating doxorubicin-induced cardiotoxicity and enhancing the antitumor efficacy of doxorubicin. Int J Biol Sci 2024; 20:5396-5414. [PMID: 39494326 PMCID: PMC11528453 DOI: 10.7150/ijbs.97418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
Doxorubicin (DOX) is an anthracycline that has excellent anticancer effects during tumor chemotherapy, but it can cause cardiotoxic effects and its clinical use has been limited. Therefore, finding new drugs or methods to prevent or reverse the cardiac damage caused by DOX therapy in cancer patients is essential. Previous studies have identified potential cardioprotective effects of Centella asiatica (C. asiatica), and madecassic acid (MA) is a pentacyclic triterpenoid derived from C. asiatica. However, the pharmacological effects of MA on the heart and tumors during tumor chemotherapy are not fully understood. The aim of this study was to investigate the pharmacological function and molecular mechanisms of MA in the heart and tumor during chemotherapy. In a DOX-induced acute heart failure mouse model and a cardiomyocyte injury model, MA reduced cardiomyocyte oxidative stress and the inflammatory response, improved mitochondrial function, and attenuated autophagic flux blockade and apoptosis. Interestingly, MA significantly increased the expression and activity of SIRT1. When SIRT1 was knocked down, the protective effect of MA on cardiomyocytes was significantly inhibited, suggesting that MA may exert cardioprotective effects through the SIRT1 pathway. Interestingly, in contrast to its cardioprotective effect, MA could synergize with DOX and significantly contribute to the anticancer chemotherapeutic effect of DOX by inhibiting proliferation, migration and invasion; promoting apoptosis; and suppressing tumor progression by inhibiting the expression of the DDX5 pathway in tumor cells. Here, we identified the pharmacological functions of the pentacyclic triterpenoid MA in ameliorating DOX-induced cardiotoxicity and enhancing the antitumor efficacy of DOX.
Collapse
Affiliation(s)
- Wenlin Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Kun Xu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Ming Lan
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Junpeng Gao
- Biomedical Pioneering lnnovation Center, School of Life Sciences, Peking University, Beijing 100871, China
| | - Lin Dou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Yao Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Que Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Mingjing Yan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
- Peking University Fifth School of Clinical Medicine, Beijing, 100730, China
| | - Sainan Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Qinan Ma
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Weimeng Tian
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Beidong Chen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Ju Cui
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Xiyue Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Jianping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Hua Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Liang Sun
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Xiuqing Huang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Tao Shen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Peking University Fifth School of Clinical Medicine, Beijing, 100730, China
| |
Collapse
|
26
|
Mehryab F, Ebrahimi M, Baharvand H, Haeri A, Shekari F. Extracellular vesicle-based formulation of doxorubicin: drug loading optimization, characterization, and cytotoxicity evaluation in tumor spheroids. Pharm Dev Technol 2024; 29:727-737. [PMID: 39072404 DOI: 10.1080/10837450.2024.2384448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/26/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Doxorubicin (DOX) is a chemotherapeutic with considerable efficacy, but its application is limited due to cardiotoxicity. Nanoparticles can improve DOX efficacy and prevent its adverse effects. Herein, DOX-loaded extracellular vesicles (DOX-EVs) were prepared using different loading methods including incubation, electroporation, and sonication in different hydration buffers to permeabilize nanostructures or desalinize DOX for improved entrapment. Different protein:drug (µg:µg) ratios of 1:10, 1:5, and 1:2, and incubation parameters were also investigated. The optimal formulation was characterized by western blotting, electron microscopy, Zetasizer, infrared spectroscopy, and release study. The cellular uptake and efficacy were investigated in MCF-7 spheroids via MTS assay, spheroid formation assay (SFA), confocal microscopy, and flow cytometry. The percentage of entrapment efficiency (EE) of formulations was improved from 1.0 ± 0.1 to 22.0 ± 1.4 using a protein:drug ratio of 1:2 and sonication in Tween 80 (0.1%w/v) containing buffer. Characterization studies verified the vesicles' identity, spherical morphology, and controlled drug release properties. Cellular studies revealed the accumulation and cytotoxicity of DOX-EVs in the spheroids, and SFA and confocal microscopy confirmed the efficacy and cellular localization. Flow cytometry results revealed a comparable and amplified efficacy for DOX-EV formulations with different cell origins. Overall, the EV formulation of DOX can be applied as a promising alternative with potential advantages.
Collapse
Affiliation(s)
- Fatemeh Mehryab
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
27
|
Xu N, Wu J, Wang W, Sun S, Sun M, Bian Y, Zhang H, Liu S, Yu G. Anti-tumor therapy of glycyrrhetinic acid targeted liposome co-delivery of doxorubicin and berberine for hepatocellular carcinoma. Drug Deliv Transl Res 2024; 14:2386-2402. [PMID: 38236508 DOI: 10.1007/s13346-023-01512-7] [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] [Accepted: 12/26/2023] [Indexed: 01/19/2024]
Abstract
During the development of hepatocellular carcinoma (HCC), hepatic stellate cells undergo activation and transform into cancer-associated fibroblasts (CAFs) due to the influence of tumor cells. The interaction between CAFs and tumor cells can compromise the effectiveness of chemotherapy drugs and promote tumor proliferation, invasion, and metastasis. This study explores the potential of glycyrrhetinic acid (GA)-modified liposomes (lip-GA) as a strategy for co-delivery of berberine (Ber) and doxorubicin (Dox) to treat HCC. The characterizations of liposomes, including particle size, zeta potential, polydispersity index, stability and in vitro drug release, were investigated. The study evaluated the anti-proliferation and anti-migration effects of Dox&Ber@lip-GA on the Huh-7 + LX-2 cell model were through MTT and wound-healing assays. Additionally, the in vivo drug distribution and anti-tumor efficacy were investigated using the H22 + NIH-3T3-bearing mouse model. The results indicated that Dox&Ber@lip-GA exhibited a nanoscale particle size, accumulated specifically in the tumor region, and was efficiently taken up by tumor cells. Compared to other groups, Dox&Ber@lip-GA demonstrated higher cytotoxicity and lower migration rates. Additionally, it significantly reduced the deposition of extracellular matrix (ECM) and inhibited tumor angiogenesis, thereby suppressing tumor growth. In conclusion, Dox&Ber@lip-GA exhibited superior anti-tumor effects both in vitro and in vivo, highlighting its potential as an effective therapeutic strategy for combating HCC.
Collapse
Affiliation(s)
- Na Xu
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
- Department of Oncology, The First Affiliated Hospital of Weifang Medical College: Weifang People's Hospital, Weifang, China
| | - Jingliang Wu
- School of Nursing, Weifang University of Science and Technology, Weifang, China.
| | - Weihao Wang
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
| | - Shujie Sun
- School of Nursing, Weifang University of Science and Technology, Weifang, China
| | - Mengmeng Sun
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
- Department of Oncology, The First Affiliated Hospital of Weifang Medical College: Weifang People's Hospital, Weifang, China
| | - Yandong Bian
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
| | - Huien Zhang
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
| | - Shuzhen Liu
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
- Department of Oncology, The First Affiliated Hospital of Weifang Medical College: Weifang People's Hospital, Weifang, China
| | - Guohua Yu
- School of Clinical Medicine, Weifang Medicine University, Weifang, China.
- Department of Oncology, The First Affiliated Hospital of Weifang Medical College: Weifang People's Hospital, Weifang, China.
| |
Collapse
|
28
|
Bourang S, Noruzpour M, Jahanbakhsh Godekahriz S, Ebrahimi HAC, Amani A, Asghari Zakaria R, Yaghoubi H. Application of nanoparticles in breast cancer treatment: a systematic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6459-6505. [PMID: 38700795 DOI: 10.1007/s00210-024-03082-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/02/2024] [Indexed: 09/25/2024]
Abstract
It is estimated that cancer is the second leading cause of death worldwide. The primary or secondary cause of cancer-related mortality for women is breast cancer. The main treatment method for different types of cancer is chemotherapy with drugs. Because of less water solubility of chemotherapy drugs or their inability to pass through membranes, their body absorbs them inadequately, which lowers the treatment's effectiveness. Drug specificity and pharmacokinetics can be changed by nanotechnology using nanoparticles. Instead, targeted drug delivery allows medications to be delivered to the targeted sites. In this review, we focused on nanoparticles as carriers in targeted drug delivery, their characteristics, structure, and the previous studies related to breast cancer. It was shown that nanoparticles could reduce the negative effects of chemotherapy drugs while increasing their effectiveness. Lipid-based nanocarriers demonstrated notable results in this instance, and some products that are undergoing various stages of clinical trials are among the examples. Nanoparticles based on metal or polymers demonstrated a comparable level of efficacy. With the number of cancer cases rising globally, many researchers are now looking into novel treatment approaches, particularly the use of nanotechnology and nanoparticles in the treatment of cancer. In order to help clinicians, this article aimed to gather more information about various areas of nanoparticle application in breast cancer therapy, such as modifying their synthesis and physicochemical characterization. It also sought to gain a deeper understanding of the mechanisms underlying the interactions between nanoparticles and biologically normal or infected tissues.
Collapse
Affiliation(s)
- Shima Bourang
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mehran Noruzpour
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Sodabeh Jahanbakhsh Godekahriz
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hossein Ali Ca Ebrahimi
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Amin Amani
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Rasool Asghari Zakaria
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hashem Yaghoubi
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| |
Collapse
|
29
|
Díaz-Tejeiro C, Arenas-Moreira M, Sanvicente A, Paniagua-Herranz L, Clemente-Casares P, Bravo I, Alonso-Moreno C, Nieto-Jiménez C, Ocaña A. Antitumoral activity of a CDK12 inhibitor in colorectal cancer through a liposomal formulation. Biomed Pharmacother 2024; 178:117165. [PMID: 39059354 DOI: 10.1016/j.biopha.2024.117165] [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: 03/27/2024] [Revised: 07/04/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. Recent experiments suggest that CDK12 can be a good therapeutic target in CRC, and therefore, novel inhibitors targeting this protein are currently in preclinical development. Lipid-based formulations of chemical entities have demonstrated the ability to enhance activity while improving the safety profile. In the present work, we explore the antitumor activity of a new CDK12 inhibitor (CDK12-IN-E9, CDK12i) and its lipid-based formulation (LP-CDK12i) in CRC models, to increase efficacy. SW620, SW480 and HCT116 CRC cell lines were used to evaluate the inhibitor and the liposomal formulation using MTT proliferation assay, 3D invasion cultures, flow cytometry, Western blotting and immunofluorescence experiments. Free-cholesterol liposomal formulations of CDK12i (LP-CDK12i) were obtained by solvent injection method and fully characterized by size, shape, polydispersity, encapsulation efficiency, and release profile and stability assessments. LP-CDK12i induced a higher antiproliferative effect compared with CDK12i as a free agent. The IC50 value was lower across all cell lines tested, leading to a reduction in cell proliferation and the formation of 3D structures. Evaluation of apoptosis revealed an increase in cell death, while biochemical studies demonstrated modifications of apoptosis and DNA damage components. In conclusion, we confirm the role of targeting CDK12 for the treatment of CRC and describe, for the first time, a liposomal formulation of a CDK12i with higher antiproliferative activity compared with the free compound.
Collapse
Affiliation(s)
- Cristina Díaz-Tejeiro
- Experimental Therapeutics in Cancer Unit, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdISSC), Madrid, Spain; Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - María Arenas-Moreira
- Universidad de Castilla-La Mancha, Unidad nanoDrug, Facultad de Farmacia-Centro de Innovación en Química Avanzada (ORFEO-CINQA), Albacete 02008, Spain
| | - Adrián Sanvicente
- Experimental Therapeutics in Cancer Unit, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdISSC), Madrid, Spain; Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Lucía Paniagua-Herranz
- Experimental Therapeutics in Cancer Unit, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Pilar Clemente-Casares
- Laboratorio de Virología Molecular, Centro Regional de Investigaciones Biomédicas, Facultad de Farmacia de Albacete, Universidad de Castilla-La Mancha, Albacete 02008, Spain; Grupo de Medicina Molecular, Laboratorio de Virología Molecular, Instituto de Biomedicina (IB)), Facultad de Farmacia de Albacete, Universidad de Castilla-La Mancha, Albacete 02008, Spain
| | - Ivan Bravo
- Universidad de Castilla-La Mancha, Unidad nanoDrug, Facultad de Farmacia-Centro de Innovación en Química Avanzada (ORFEO-CINQA), Albacete 02008, Spain
| | - Carlos Alonso-Moreno
- Universidad de Castilla-La Mancha, Unidad nanoDrug, Facultad de Farmacia-Centro de Innovación en Química Avanzada (ORFEO-CINQA), Albacete 02008, Spain
| | - Cristina Nieto-Jiménez
- Experimental Therapeutics in Cancer Unit, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - Alberto Ocaña
- Experimental Therapeutics in Cancer Unit, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdISSC), Madrid, Spain; Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), and CIBERONC, Madrid, Spain; START Madrid-Fundación Jiménez Díaz (FJD) Early Phase Program, Fundación Jiménez Díaz Hospital, Madrid, Spain.
| |
Collapse
|
30
|
Aloss K, Leroy Viana PH, Bokhari SMZ, Giunashvili N, Schvarcz CA, Bócsi D, Koós Z, Benyó Z, Hamar P. Ivermectin Synergizes with Modulated Electro-hyperthermia and Improves Its Anticancer Effects in a Triple-Negative Breast Cancer Mouse Model. ACS Pharmacol Transl Sci 2024; 7:2496-2506. [PMID: 39144564 PMCID: PMC11320741 DOI: 10.1021/acsptsci.4c00314] [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: 05/27/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 08/16/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype, with limited treatment options. Modulated electro-hyperthermia (mEHT) is a novel adjuvant cancer therapy that induces selective cancer damage. However, mEHT upregulates heat shock protein beta 1 (HSPB1), a cancer-promoting stress chaperone molecule. Thus, we investigated whether ivermectin (IVM), an anthelmintic drug, may synergize with mEHT and enhance its anticancer effects by inhibiting HSPB1 phosphorylation. Isogenic 4T1 TNBC cells were inoculated into BALB/c mice and treated with mEHT, IVM, or a combination of both. IVM synergistically improved the tumor growth inhibition achieved by mEHT. Moreover, IVM downregulated mEHT-induced HSPB1 phosphorylation. Thus, the strongest cancer tissue damage was observed in the mEHT + IVM-treated tumors, coupled with the strongest apoptosis induction and proliferation inhibition. In addition, there was no significant body weight loss in mice treated with mEHT and IVM, indicating that this combination was well-tolerated. In conclusion, mEHT combined with IVM is a new, effective, and safe option for the treatment of TNBC.
Collapse
Affiliation(s)
- Kenan Aloss
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26., Budapest 1085, Hungary
- Department
of Pharmacology and Pharmacotherapy, Semmelweis
University, Budapest 1089, Hungary
| | | | | | - Nino Giunashvili
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26., Budapest 1085, Hungary
| | - Csaba András Schvarcz
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26., Budapest 1085, Hungary
- HUN-REN-SU
Cerebrovascular and Neurocognitive Diseases Research Group, Tűzoltó utca 37-47., Budapest 1094, Hungary
| | - Dániel Bócsi
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26., Budapest 1085, Hungary
| | - Zoltán Koós
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26., Budapest 1085, Hungary
| | - Zoltán Benyó
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26., Budapest 1085, Hungary
- HUN-REN-SU
Cerebrovascular and Neurocognitive Diseases Research Group, Tűzoltó utca 37-47., Budapest 1094, Hungary
| | - Péter Hamar
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26., Budapest 1085, Hungary
| |
Collapse
|
31
|
Visan AI, Negut I. Development and Applications of PLGA Hydrogels for Sustained Delivery of Therapeutic Agents. Gels 2024; 10:497. [PMID: 39195026 DOI: 10.3390/gels10080497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Poly(lactic-co-glycolic acid) (PLGA) hydrogels are highly utilized in biomedical research due to their biocompatibility, biodegradability, and other versatile properties. This review comprehensively explores their synthesis, properties, sustained release mechanisms, and applications in drug delivery. The introduction underscores the significance of PLGA hydrogels in addressing challenges like short half-lives and systemic toxicity in conventional drug formulations. Synthesis methods, including emulsion solvent evaporation, solvent casting, electrospinning, thermal gelation, and photopolymerization, are described in detail and their role in tailoring hydrogel properties for specific applications is highlighted. Sustained release mechanisms-such as diffusion-controlled, degradation-controlled, swelling-controlled, and combined systems-are analyzed alongside key kinetic models (zero-order, first-order, Higuchi, and Peppas models) for designing controlled drug delivery systems. Applications of PLGA hydrogels in drug delivery are discussed, highlighting their effectiveness in localized and sustained chemotherapy for cancer, as well as in the delivery of antibiotics and antimicrobials to combat infections. Challenges and future prospects in PLGA hydrogel research are discussed, with a focus on improving drug loading efficiency, improving release control mechanisms, and promoting clinical translation. In summary, PLGA hydrogels provide a promising platform for the sustained delivery of therapeutic agents and meet diverse biomedical requirements. Future advancements in materials science and biomedical engineering are anticipated to further optimize their efficacy and applicability in clinical settings. This review consolidates the current understanding and outlines future research directions for PLGA hydrogels, emphasizing their potential to revolutionize therapeutic delivery and improve patient outcomes.
Collapse
Affiliation(s)
- Anita Ioana Visan
- National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Romania
| | - Irina Negut
- National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Romania
| |
Collapse
|
32
|
Stavilă C, Minuti AE, Herea DD, Lăbuşcă L, Gherca D, Lupu N, Chiriac H. Synergistic Effect of Chemotherapy and Magnetomechanical Actuation of Fe-Cr-Nb-B Magnetic Particles on Cancer Cells. ACS OMEGA 2024; 9:30518-30533. [PMID: 39035922 PMCID: PMC11256100 DOI: 10.1021/acsomega.4c02189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/23/2024]
Abstract
The present study is aimed at developing an innovative method for efficient cancer cell destruction by exploiting the magnetomechanical actuation (MMA) of Fe-Cr-Nb-B magnetic particles (MPs), which are loaded with clinically approved chemotherapeutic drugs. To achieve this objective, Fe68.2Cr11.5Nb0.3B20 magnetic nanoparticles were produced by mechanically grinding amorphous ribbon precursors with the same composition. These nanoparticles display high anisotropy, a parallelepipedic shape with an amorphous structure, and a ferromagnetic behavior. MPs were loaded with the antitumoral drugs mitoxantrone (MTX) or doxorubicin (DOX). In our study, we used adipose-derived mesenchymal stem cells and human osteosarcoma cells to test drug-loaded MPs for their biocompatibility, cytotoxicity, and cellular internalization. Further tests involved exposing cells to magnetomechanical actuation and simultaneous MPs-targeted chemotherapy followed by cell viability/death assays, such as MTT and LDH, and live/dead cell staining. Results demonstrate that cancer cell death was induced by the synergistic action of chemotherapeutic drugs and magnetomechanical actuation. The nanoparticle vehicles helped overcome drug resistance, decreasing the high dose of drugs used in conventional therapies as well as the time intervals needed for MMA to affect cancer cell viability. The proposed approach highlights the possibility of using a new, targeted, and effective cancer treatment with very few side effects.
Collapse
Affiliation(s)
- Cristina Stavilă
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
- Faculty
of Physics, “Alexandru Ioan Cuza”
University, Iasi 700506, Romania
| | - Anca Emanuela Minuti
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| | - Dumitru Daniel Herea
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| | - Luminiţa Lăbuşcă
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| | - Daniel Gherca
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| | - Nicoleta Lupu
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| | - Horia Chiriac
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| |
Collapse
|
33
|
Babanyinah GK, Bhadran A, Polara H, Wang H, Shah T, Biewer MC, Stefan MC. Maleimide functionalized polycaprolactone micelles for glutathione quenching and doxorubicin delivery. Chem Sci 2024; 15:9987-10001. [PMID: 38966382 PMCID: PMC11220601 DOI: 10.1039/d4sc01625d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/16/2024] [Indexed: 07/06/2024] Open
Abstract
High glutathione production is known to be one of the defense mechanisms by which many cancer cells survive elevated oxidative stress. By explicitly targeting glutathione in these cancer cells and diminishing its levels, oxidative stress can be intensified, ultimately triggering apoptosis or programmed cell death. Herein, we developed a novel approach by creating maleimide-functionalized polycaprolactone polymers, specifically using 2,3-diiodomaleimide functionality to reduce the level of glutathione in cancer cells. Polycaprolactone was chosen to conjugate the 2,3-diiodomaleimide functionality due to its biodegradable and biocompatible properties. The amphiphilic block copolymer was synthesized using PEG as a macroinitiator to make corresponding polymeric micelles. The resulting 2,3-diiodomaleimide-conjugated polycaprolactone micelles effectively quenched glutathione, even at low concentrations (0.01 mg mL-1). Furthermore, we loaded these micelles with the anticancer drug doxorubicin (DOX), which exhibited pH-dependent drug release. We obtained a loading capacity (LC) of 3.5% for the micelles, one of the highest LC reported among functional PCL-based micelles. Moreover, the enhanced LC doesn't affect their release profile. Cytotoxicity experiments demonstrated that empty and DOX-loaded micelles inhibited cancer cell growth, with the DOX-loaded micelles displaying the highest cytotoxicity. The ability of the polymer to quench intracellular GSH was also confirmed. This approach of attaching maleimide to polycaprolactone polymers shows promise in depleting elevated glutathione levels in cancer cells, potentially improving cancer treatment efficacy.
Collapse
Affiliation(s)
- Godwin K Babanyinah
- Department of Chemistry and Biochemistry, University of Texas at Dallas Richardson TX USA
| | - Abhi Bhadran
- Department of Chemistry and Biochemistry, University of Texas at Dallas Richardson TX USA
| | - Himanshu Polara
- Department of Chemistry and Biochemistry, University of Texas at Dallas Richardson TX USA
| | - Hanghang Wang
- Department of Chemistry and Biochemistry, University of Texas at Dallas Richardson TX USA
| | - Tejas Shah
- Department of Chemistry and Biochemistry, University of Texas at Dallas Richardson TX USA
| | - Michael C Biewer
- Department of Chemistry and Biochemistry, University of Texas at Dallas Richardson TX USA
| | - Mihaela C Stefan
- Department of Chemistry and Biochemistry, University of Texas at Dallas Richardson TX USA
| |
Collapse
|
34
|
Aloss K, Hamar P. Augmentation of the EPR effect by mild hyperthermia to improve nanoparticle delivery to the tumor. Biochim Biophys Acta Rev Cancer 2024; 1879:189109. [PMID: 38750699 DOI: 10.1016/j.bbcan.2024.189109] [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: 02/12/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
The clinical translation of the nanoparticle (NP)-based anticancer therapies is still unsatisfactory due to the heterogeneity of the enhanced permeability and retention (EPR) effect. Despite the promising preclinical outcome of the pharmacological EPR enhancers, their systemic toxicity can limit their clinical application. Hyperthermia (HT) presents an efficient tool to augment the EPR by improving tumor blood flow (TBF) and vascular permeability, lowering interstitial fluid pressure (IFP), and disrupting the structure of the extracellular matrix (ECM). Furthermore, the HT-triggered intravascular release approach can overcome the EPR effect. In contrast to pharmacological approaches, HT is safe and can be focused to cancer tissues. Moreover, HT conveys direct anti-cancer effects, which improve the efficacy of the anti-cancer agents encapsulated in NPs. However, the clinical application of HT is challenging due to the heterogeneous distribution of temperature within the tumor, the length of the treatment and the complexity of monitoring.
Collapse
Affiliation(s)
- Kenan Aloss
- Institute of Translational Medicine - Semmelweis University - 1094, Tűzoltó utca, 37-49, Budapest, Hungary
| | - Péter Hamar
- Institute of Translational Medicine - Semmelweis University - 1094, Tűzoltó utca, 37-49, Budapest, Hungary.
| |
Collapse
|
35
|
Zheng H, An G, Yang X, Huang L, Wang N, Zhu Y. Iron-Based Metal-Organic Frameworks as Multiple Cascade Synergistic Therapeutic Effect Nano-Drug Delivery Systems for Effective Tumor Elimination. Pharmaceuticals (Basel) 2024; 17:812. [PMID: 38931479 PMCID: PMC11206809 DOI: 10.3390/ph17060812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Efforts have been made to improve the therapeutic efficiency of tumor treatments, and metal-organic frameworks (MOFs) have shown excellent potential in tumor therapy. Monotherapy for the treatment of tumors has limited effects due to the limitation of response conditions and inevitable multidrug resistance, which seriously affect the clinical therapeutic effect. In this study, we chose to construct a multiple cascade synergistic tumor drug delivery system MIL-101(Fe)-DOX-TCPP-MnO2@PDA-Ag (MDTM@P-Ag) using MOFs as drug carriers. Under near-infrared (NIR) laser irradiation, 5,10,15,20-tetrakis(4-carboxyphenyl)porphyrin (TCPP) and Ag NPs loaded on MDTM@P-Ag can be activated to generate cytotoxic reactive oxygen species (ROS) and achieve photothermal conversion, thus effectively inducing the apoptosis of tumor cells and achieving a combined photodynamic/photothermal therapy. Once released at the tumor site, manganese dioxide (MnO2) can catalyze the decomposition of hydrogen peroxide (H2O2) in the acidic microenvironment of the tumor to generate oxygen (O2) and alleviate the hypoxic environment of the tumor. Fe3+/Mn2+ will mediate a Fenton/Fenton-like reaction to generate cytotoxic hydroxyl radicals (·OH), while depleting the high concentration of glutathione (GSH) in the tumor, thus enhancing the chemodynamic therapeutic effect. The successful preparation of the tumor drug delivery system and its good synergistic chemodynamic/photodynamic/photothermal therapeutic effect in tumor treatment can be demonstrated by the experimental results of material characterization, performance testing and in vitro experiments.
Collapse
Affiliation(s)
- Heming Zheng
- State Key Laboratory of Featured Metal Materials and Life-Cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; (H.Z.); (G.A.); (Y.Z.)
| | - Guanghui An
- State Key Laboratory of Featured Metal Materials and Life-Cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; (H.Z.); (G.A.); (Y.Z.)
| | - Xiaohui Yang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Lei Huang
- School of Stomatology, Minzhu Clinic of Stomatology Hospital Affiliated to Guangxi Medical University, Nanning 530007, China;
| | - Nannan Wang
- State Key Laboratory of Featured Metal Materials and Life-Cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; (H.Z.); (G.A.); (Y.Z.)
| | - Yanqiu Zhu
- State Key Laboratory of Featured Metal Materials and Life-Cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; (H.Z.); (G.A.); (Y.Z.)
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter EX4 4QF, UK
| |
Collapse
|
36
|
Zhang J, Ye Q, Yang X, Li T, Huang S, Zhou P, Feng Y, Liu H, Xie K. Dual immunotherapy alternating with anti-PD-1 antibody plus liposomal doxorubicin show good efficacy in prostate epithelioid hemangioendothelioma: a case report. Front Immunol 2024; 15:1384111. [PMID: 38947327 PMCID: PMC11211375 DOI: 10.3389/fimmu.2024.1384111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Epithelioid hemangioendothelioma is a rare vascular malignancy, and currently, there is no standard treatment regimen for this disease and existing treatment options have limited efficacy. In this case report, we present a patient with lung and lymph node metastases from prostate epithelioid hemangioendothelioma who achieved a significant partial response. This was accomplished through alternating nivolumab therapy with ipilimumab and liposomal doxorubicin, resulting in a progression-free-survival more than 6 months to date. The treatment was well-tolerated throughout. Our report suggests that dual immunotherapy alternating with anti-PD-1antibody plus doxorubicin may be a potential treatment modality for epithelioid hemangioendothelioma. However, larger sample studies are necessary to ascertain the effectiveness of this treatment strategy and it is essential to continue monitoring this patient to sustain progression-free survival and overall survival.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Oncology, Chengdu BOE Hospital, Chengdu, Sichuan, China
| | - Qin Ye
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xudan Yang
- Department of Pathology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Tenglong Li
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shan Huang
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ping Zhou
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yumei Feng
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Hao Liu
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ke Xie
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
37
|
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
|
38
|
Paschoal Barbosa LM, Gomes ER, de Barros ALB, Cassali GD, de Carvalho AT, Silva JDO, Pádua AL, Oliveira MC. Hybrid Nanosystem Formed by DOX-Loaded Liposomes and Extracellular Vesicles from MDA-MB-231 Is Effective against Breast Cancer Cells with Different Molecular Profiles. Pharmaceutics 2024; 16:739. [PMID: 38931861 PMCID: PMC11206866 DOI: 10.3390/pharmaceutics16060739] [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: 05/10/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Drug delivery selectivity is a challenge for cancer treatment. A hybrid pegylated pH-sensitive liposome-extracellular vesicle isolated from human breast cancer cell MDA-MB-231 was developed to investigate its in vitro activity against breast cancer cells of different molecular profiles to overcome this inconvenience. The hybrid nanosystem was produced by film hydration, and doxorubicin (DOX) was encapsulated in this system using the ammonium sulfate gradient method. The characterization of this hybrid nanosystem revealed a mean diameter of 140.20 ± 2.70 nm, a polydispersity index of 0.102 ± 0.033, an encapsulation efficiency of doxorubicin of 88.9% ± 2.4, and a great storage stability for 90 days at 4 °C. The fusion of extracellular vesicles with liposomes was confirmed by nanoflow cytometry using PE-conjugated human anti-CD63. This hybrid nanosystem demonstrated cytotoxicity against human breast cancer cell lines with different molecular subtypes, enhanced anti-migration properties, and exhibited similar cellular uptake to the free DOX treatment. Preliminary acute toxicity assessments using Balb/C female mice indicated a median lethal dose of 15-17.5 mg/kg, with no evidence of splenic, liver, heart, bone marrow, and renal damage at a dose of 15 mg/kg. These findings suggest the hybrid formulation as a versatile nanocarrier for the treatment of various breast cancer subtypes.
Collapse
Affiliation(s)
- Luiza Marques Paschoal Barbosa
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (E.R.G.)
| | - Eliza Rocha Gomes
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (E.R.G.)
- Institute of Regenerative Medicine and Biotherapies (IRMB), Hôpital Saint-Eloi, 34295 Montpellier, France
| | - André Luis Branco de Barros
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (E.R.G.)
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Geovanni Dantas Cassali
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Andréa Teixeira de Carvalho
- Instituto René Rachou, Fiocruz Minas, Av. Augusto de Lima, 1715, Barro Preto, Belo Horizonte 30190-002, MG, Brazil
| | - Juliana de Oliveira Silva
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (E.R.G.)
| | - Ana Luiza Pádua
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (E.R.G.)
| | - Mônica Cristina Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (E.R.G.)
| |
Collapse
|
39
|
Zhao C, Liu H, Huang S, Guo Y, Xu L. Metal-Organic Framework-Capped Gold Nanorod Hybrids for Combinatorial Cancer Therapy. Molecules 2024; 29:2384. [PMID: 38792244 PMCID: PMC11124105 DOI: 10.3390/molecules29102384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Recently, nanomaterials have attracted extensive attention in cancer-targeting therapy and as drug delivery vehicles owing to their unique surface and size properties. Multifunctional combinations of nanomaterials have become a research hotspot as researchers aim to provide a full understanding of their nanomaterial characteristics. In this study, metal-organic framework-capped gold nanorod hybrids were synthesized. Our research explored their ability to kill tumor cells by locally increasing the temperature via photothermal conclusion. The specific peroxidase-like activity endows the hybrids with the ability to disrupt the oxidative balance in vitro. Simultaneously, chemotherapeutic drugs are administered and delivered by loading and transportation for effective combinatorial cancer treatment, thereby enhancing the curative effect and reducing the unpredictable toxicity and side effects of large doses of chemotherapeutic drugs. These studies can improve combinatorial cancer therapy and enhance cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Yi Guo
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (C.Z.); (H.L.); (S.H.)
| | - Li Xu
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (C.Z.); (H.L.); (S.H.)
| |
Collapse
|
40
|
Gabriel EM, Bahr D, Rachamala HK, Madamsetty VS, Shreeder B, Bagaria S, Escobedo AL, Reid JM, Mukhopadhyay D. Liposomal Phenylephrine Nanoparticles Enhance the Antitumor Activity of Intratumoral Chemotherapy in a Preclinical Model of Melanoma. ACS Biomater Sci Eng 2024; 10:3412-3424. [PMID: 38613483 PMCID: PMC11301277 DOI: 10.1021/acsbiomaterials.4c00078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2024]
Abstract
Intratumoral injection of anticancer agents has limited efficacy and is not routinely used for most cancers. In this study, we aimed to improve the efficacy of intratumoral chemotherapy using a novel approach comprising peri-tumoral injection of sustained-release liposomal nanoparticles containing phenylephrine, which is a potent vasoconstrictor. Using a preclinical model of melanoma, we have previously shown that systemically administered (intravenous) phenylephrine could transiently shunt blood flow to the tumor at the time of drug delivery, which in turn improved antitumor responses. This approach was called dynamic control of tumor-associated vessels. Herein, we used liposomal phenylephrine nanoparticles as a "local" dynamic control strategy for the B16 melanoma. Local dynamic control was shown to increase the retention and exposure time of tumors to intratumorally injected chemotherapy (melphalan). C57BL/6 mice bearing B16 tumors were treated with intratumoral melphalan and peri-tumoral injection of sustained-release liposomal phenylephrine nanoparticles (i.e., the local dynamic control protocol). These mice had statistically significantly improved antitumor responses compared to melphalan alone (p = 0.0011), whereby 58.3% obtained long-term complete clinical response. Our novel approach of local dynamic control demonstrated significantly enhanced antitumor efficacy and is the subject of future clinical trials being designed by our group.
Collapse
Affiliation(s)
- Emmanuel M. Gabriel
- Department of Surgery, Division of Surgical Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Deborah Bahr
- Department of Molecular Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | | | - Barath Shreeder
- Department of Immunology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Sanjay Bagaria
- Department of Surgery, Division of Surgical Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Joel M. Reid
- Department of Pharmacology, Mayo Clinic, Rochester, MN, 55902, USA
| | | |
Collapse
|
41
|
Pusta A, Tertis M, Ardusadan C, Mirel S, Cristea C. Electrochemical Sensing Device for Carboplatin Monitoring in Proof-of-Concept Drug Delivery Nanosystems. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:793. [PMID: 38727386 PMCID: PMC11085464 DOI: 10.3390/nano14090793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024]
Abstract
(1) Background: Carboplatin (CBP) is a chemotherapeutic drug widely used in the treatment of a variety of cancers. Despite its efficiency, CBP is associated with side effects that greatly limit its clinical use. To mitigate these effects, CBP can be encapsulated in targeted delivery systems, such as liposomes. Ensuring the adequate loading and release of CBP from these carriers requires strict control in pharmaceutical formulation development, demanding modern, rapid, and robust analytical methods. The aim of this study was the development of a sensor for the fast and accurate quantification of CBP and its application on proof-of-concept CBP-loaded nanosomes. (2) Methods: Screen-printed electrodes were obtained in-lab and the electrochemical behavior of CBP was tested on the obtained electrodes. (3) Results: The in-lab screen-printed electrodes demonstrated superior properties compared to commercial ones. The novel sensors demonstrated accurate detection of CBP on a dynamic range from 5 to 500 μg/mL (13.5-1350 μM). The method was successfully applied on CBP loaded and released from nanosomes, with strong correlations with a spectrophotometric method used as control. (4) Conclusions: This study demonstrates the viability of electrochemical techniques as alternative options during the initial phases of pharmaceutical formulation development.
Collapse
Affiliation(s)
- Alexandra Pusta
- Department of Analytical Chemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (A.P.); (C.A.); (C.C.)
- Department of Medical Devices, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Mihaela Tertis
- Department of Analytical Chemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (A.P.); (C.A.); (C.C.)
| | - Catalina Ardusadan
- Department of Analytical Chemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (A.P.); (C.A.); (C.C.)
| | - Simona Mirel
- Department of Medical Devices, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Cecilia Cristea
- Department of Analytical Chemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (A.P.); (C.A.); (C.C.)
| |
Collapse
|
42
|
Kimura T, Okada K, Morohashi Y, Kato Y, Mori M, Kato H, Matsumoto T, Shimoyama S. Quantification of Unencapsulated Drug in Target Tissues Demonstrates Pharmacological Properties and Therapeutic Effects of Liposomal Topotecan (FF-10850). Pharm Res 2024; 41:795-806. [PMID: 38536615 PMCID: PMC11024016 DOI: 10.1007/s11095-023-03652-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/27/2023] [Indexed: 04/18/2024]
Abstract
PURPOSE Quantifying unencapsulated drug concentrations in tissues is crucial for understanding the mechanisms underlying the efficacy and safety of liposomal drugs; however, the methodology for this has not been fully established. Herein, we aimed to investigate the enhanced therapeutic potential of a pegylated liposomal formulation of topotecan (FF-10850) by analyzing the concentrations of the unencapsulated drug in target tissues, to guide the improvement of its dosing regimen. METHODS We developed a method for measuring unencapsulated topotecan concentrations in tumor and bone marrow interstitial fluid (BM-ISF) and applied this method to pharmacokinetic assessments. The ratios of the area under the concentration-time curves (AUCs) between tumor and BM-ISF were calculated for total and unencapsulated topotecan. DNA damage and antitumor effects of FF-10850 or non-liposomal topotecan (TPT) were evaluated in an ES-2 mice xenograft model. RESULTS FF-10850 exhibited a much larger AUC ratio between tumor and BM-ISF for unencapsulated topotecan (2.96), but not for total topotecan (0.752), than TPT (0.833). FF-10850 promoted milder DNA damage in the bone marrow than TPT; however, FF-10850 and TPT elicited comparable DNA damage in the tumor. These findings highlight the greater tumor exposure to unencapsulated topotecan and lower bone marrow exposure to FF-10850 than TPT. The dosing regimen was successfully improved based on the kinetics of unencapsulated topotecan and DNA damage. CONCLUSIONS Tissue pharmacokinetics of unencapsulated topotecan elucidated the favorable pharmacological properties of FF-10850. Evaluation of tissue exposure to an unencapsulated drug with appropriate pharmacodynamic markers can be valuable in optimizing liposomal drugs and dosing regimens.
Collapse
Affiliation(s)
- Toshifumi Kimura
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan.
| | - Ken Okada
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Yasushi Morohashi
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Mikinaga Mori
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Hiroshi Kato
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Takeshi Matsumoto
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Susumu Shimoyama
- FUJIFILM Pharmaceuticals U.S.A., Inc, One Broadway, Cambridge, MA, 02142, USA
| |
Collapse
|
43
|
Qin S, Wen Z, Huang H, Wu W. Use of novel taurine-chitosan mediated liposomes for enhancing the oral absorption of doxorubicin via the TAUT transporter. Carbohydr Polym 2024; 329:121780. [PMID: 38286550 DOI: 10.1016/j.carbpol.2024.121780] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024]
Abstract
Our research aimed to enhance the oral bioavailability of doxorubicin hydrochloride (DOX·HCl) while minimizing the potential for myocardial toxicity. To achieve this goal, we developed a new method that utilizes a coating material to encapsulate the drug in liposomes, which can specifically target intestinal taurine transporter proteins. This coating material, TAU-CS, was created by combining taurine with chitosan. We characterized TAU-CS using various methods, including 1H NMR, FT-IR, and scanning electron microscopy (SEM). The resulting liposomes exhibited a regular spherical morphology, with a particle size of 195.7 nm, an encapsulation efficiency of 91.23 %, and a zeta potential of +11.65 mV. Under simulated gastrointestinal conditions, TAU-CS/LIP@DOX·HCl exhibited good stability and slow release. Pharmacokinetic studies revealed that, compared with DOX·HCl, TAU-CS/LIP@DOX·HCl had a relative bioavailability of 342 %. Intracellular uptake, immunofluorescence imaging, and permeation assays confirmed that the taurine transporter protein mediates the intestinal uptake of these liposomes. Our study suggested that liposomes coated with TAU-CS could serve as an effective oral delivery system and that targeting the taurine transporter protein shows promise in enhancing drug absorption.
Collapse
Affiliation(s)
- Shuiling Qin
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Zhiwei Wen
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Huajie Huang
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Wei Wu
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China.
| |
Collapse
|
44
|
Ding L, Agrawal P, Singh SK, Chhonker YS, Sun J, Murry DJ. Polymer-Based Drug Delivery Systems for Cancer Therapeutics. Polymers (Basel) 2024; 16:843. [PMID: 38543448 PMCID: PMC10974363 DOI: 10.3390/polym16060843] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 11/12/2024] Open
Abstract
Chemotherapy together with surgery and/or radiotherapy are the most common therapeutic methods for treating cancer. However, the off-target effects of chemotherapy are known to produce side effects and dose-limiting toxicities. Novel delivery platforms based on natural and synthetic polymers with enhanced pharmacokinetic and therapeutic potential for the treatment of cancer have grown tremendously over the past 10 years. Polymers can facilitate selective targeting, enhance and prolong circulation, improve delivery, and provide the controlled release of cargos through various mechanisms, including physical adsorption, chemical conjugation, and/or internal loading. Notably, polymers that are biodegradable, biocompatible, and physicochemically stable are considered to be ideal delivery carriers. This biomimetic and bio-inspired system offers a bright future for effective drug delivery with the potential to overcome the obstacles encountered. This review focuses on the barriers that impact the success of chemotherapy drug delivery as well as the recent developments based on natural and synthetic polymers as platforms for improving drug delivery for treating cancer.
Collapse
Affiliation(s)
- Ling Ding
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
| | - Prachi Agrawal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (P.A.); (J.S.)
| | - Sandeep K. Singh
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
| | - Yashpal S. Chhonker
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
| | - Jingjing Sun
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (P.A.); (J.S.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Daryl J. Murry
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
45
|
Püsküllüoğlu M, Michalak I. The therapeutic potential of natural metabolites in targeting endocrine-independent HER-2-negative breast cancer. Front Pharmacol 2024; 15:1349242. [PMID: 38500769 PMCID: PMC10944949 DOI: 10.3389/fphar.2024.1349242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/16/2024] [Indexed: 03/20/2024] Open
Abstract
Breast cancer (BC) is a heterogenous disease, with prognosis and treatment options depending on Estrogen, Progesterone receptor, and Human Epidermal Growth Factor Receptor-2 (HER-2) status. HER-2 negative, endocrine-independent BC presents a significant clinical challenge with limited treatment options. To date, promising strategies like immune checkpoint inhibitors have not yielded breakthroughs in patient prognosis. Despite being considered archaic, agents derived from natural sources, mainly plants, remain backbone of current treatment. In this context, we critically analyze novel naturally-derived drug candidates, elucidate their intricate mechanisms of action, and evaluate their pre-clinical in vitro and in vivo activity in endocrine-independent HER-2 negative BC. Since pre-clinical research success often does not directly correlate with drug approval, we focus on ongoing clinical trials to uncover current trends. Finally, we demonstrate the potential of combining cutting-edge technologies, such as antibody-drug conjugates or nanomedicine, with naturally-derived agents, offering new opportunities that utilize both traditional cytotoxic agents and new metabolites.
Collapse
Affiliation(s)
- Mirosława Püsküllüoğlu
- Department of Clinical Oncology, Maria Skłodowska-Curie National Research Institute of Oncology, Kraków, Poland
| | - Izabela Michalak
- Wrocław University of Science and Technology, Faculty of Chemistry, Department of Advanced Material Technologies, Wrocław, Poland
| |
Collapse
|
46
|
Su H, Jia J, Mao Y, Zhu R, Li Z. A real-world analysis of FDA Adverse Event Reporting System (FAERS) events for liposomal and conventional doxorubicins. Sci Rep 2024; 14:5095. [PMID: 38429374 PMCID: PMC10907704 DOI: 10.1038/s41598-024-55185-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 02/21/2024] [Indexed: 03/03/2024] Open
Abstract
The clinical application of conventional doxorubicin (CDOX) was constrained by its side effects. Liposomal doxorubicin was developed to mitigate these limitations, showing improved toxicity profiles. However, the adverse events associated with liposomal doxorubicin and CDOX have not yet been comprehensively evaluated in clinical settings. The FAERS data from January 2004 to December 2022 were collected to analyze the adverse events of liposomal doxorubicin and CDOX. Disproportionate analysis and Bayesian analysis were employed to quantify this association. Our analysis incorporated 68,803 adverse event reports related to Doxil/Caelyx, Myocet and CDOX. The relative odds ratios (RORs, 95%CI) for febrile neutropenia associated with CDOX, Doxil/Caelyx, and Myocet were 42.45 (41.44; 43.48), 17.53 (16.02; 19.20), and 34.68 (26.63; 45.15) respectively. For cardiotoxicity, they were 38.87(36.41;41.49), 17.96 (14.10; 22.86), and 37.36 (19.34; 72.17). For Palmar-Plantar Erythrodysesthesia (PPE), the RORs were 6.16 (5.69; 6.68), 36.13 (32.60; 40.06), and 19.69 (11.59; 33.44). Regarding onset time, significant differences adverse events including neutropenia, PPE, pneumonia and malignant neoplasm progression. This study indicates that clinical monitoring for symptoms of cardiotoxicity of CDOX and Myocet, and PPE and interstitial lung disease of Doxil should be performed. Additionally, the onset time of febrile neutropenia, malignant neoplasm progression, and pneumonia associated with Doxil and Myocet merits particular attention. Continuous surveillance, risk evaluations, and additional comparative studies between liposomal doxorubicin and CDOX were recommended.
Collapse
Affiliation(s)
- Huiling Su
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing Jia
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuxiang Mao
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Riran Zhu
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhengjun Li
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
47
|
El-Tanani M, Nsairat H, Aljabali AA, Matalka II, Alkilany AM, Tambuwala MM. Dual-loaded liposomal carriers to combat chemotherapeutic resistance in breast cancer. Expert Opin Drug Deliv 2024; 21:309-324. [PMID: 38284386 DOI: 10.1080/17425247.2024.2311812] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/25/2024] [Indexed: 01/30/2024]
Abstract
INTRODUCTION The resistance to chemotherapy is a significant hurdle in breast cancer treatment, prompting the exploration of innovative strategies. This review discusses the potential of dual-loaded liposomal carriers to combat chemoresistance and improve outcomes for breast cancer patients. AREAS COVERED This review discusses breast cancer chemotherapy resistance and dual-loaded liposomal carriers. Drug efflux pumps, DNA repair pathways, and signaling alterations are discussed as chemoresistance mechanisms. Liposomes can encapsulate several medicines and cargo kinds, according to the review. It examines how these carriers improve medication delivery, cancer cell targeting, and tumor microenvironment regulation. Also examined are dual-loaded liposomal carrier improvement challenges and techniques. EXPERT OPINION The use of dual-loaded liposomal carriers represents a promising and innovative strategy in the battle against chemotherapy resistance in breast cancer. This article has explored the various mechanisms of chemoresistance in breast cancer, emphasizing the potential of dual-loaded liposomal carriers to overcome these challenges. These carriers offer versatility, enabling the encapsulation and precise targeting of multiple drugs with different modes of action, a crucial advantage when dealing with the complexity of breast cancer treatment.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Pharmacy, Yarmouk University, Irbid, Jordan
| | - Ismail I Matalka
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Department of Pathology and Microbiology, Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | | | | |
Collapse
|
48
|
Nagar N, Naidu G, Mishra A, Poluri KM. Protein-Based Nanocarriers and Nanotherapeutics for Infection and Inflammation. J Pharmacol Exp Ther 2024; 388:91-109. [PMID: 37699711 DOI: 10.1124/jpet.123.001673] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 08/04/2023] [Accepted: 08/21/2023] [Indexed: 09/14/2023] Open
Abstract
Infectious and inflammatory diseases are one of the leading causes of death globally. The status quo has become more prominent with the onset of the coronavirus disease 2019 (COVID-19) pandemic. To combat these potential crises, proteins have been proven as highly efficacious drugs, drug targets, and biomarkers. On the other hand, advancements in nanotechnology have aided efficient and sustained drug delivery due to their nano-dimension-acquired advantages. Combining both strategies together, the protein nanoplatforms are equipped with the advantageous intrinsic properties of proteins as well as nanoformulations, eloquently changing the field of nanomedicine. Proteins can act as carriers, therapeutics, diagnostics, and theranostics in their nanoform as fusion proteins or as composites with other organic/inorganic materials. Protein-based nanoplatforms have been extensively explored to target the major infectious and inflammatory diseases of clinical concern. The current review comprehensively deliberated proteins as nanocarriers for drugs and nanotherapeutics for inflammatory and infectious agents, with special emphasis on cancer and viral diseases. A plethora of proteins from diverse organisms have aided in the synthesis of protein-based nanoformulations. The current study specifically presented the proteins of human and pathogenic origin to dwell upon the field of protein nanotechnology, emphasizing their pharmacological advantages. Further, the successful clinical translation and current bottlenecks of the protein-based nanoformulations associated with the infection-inflammation paradigm have also been discussed comprehensively. SIGNIFICANCE STATEMENT: This review discusses the plethora of promising protein-based nanocarriers and nanotherapeutics explored for infectious and inflammatory ailments, with particular emphasis on protein nanoparticles of human and pathogenic origin with reference to the advantages, ADME (absorption, distribution, metabolism, and excretion parameters), and current bottlenecks in development of protein-based nanotherapeutic interventions.
Collapse
Affiliation(s)
- Nupur Nagar
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| | - Goutami Naidu
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| | - Amit Mishra
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| |
Collapse
|
49
|
Lukácsi S, Munkácsy G, Győrffy B. Harnessing Hyperthermia: Molecular, Cellular, and Immunological Insights for Enhanced Anticancer Therapies. Integr Cancer Ther 2024; 23:15347354241242094. [PMID: 38818970 PMCID: PMC11143831 DOI: 10.1177/15347354241242094] [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: 12/28/2023] [Revised: 02/25/2024] [Accepted: 03/11/2024] [Indexed: 06/01/2024] Open
Abstract
Hyperthermia, the raising of tumor temperature (≥39°C), holds great promise as an adjuvant treatment for cancer therapy. This review focuses on 2 key aspects of hyperthermia: its molecular and cellular effects and its impact on the immune system. Hyperthermia has profound effects on critical biological processes. Increased temperatures inhibit DNA repair enzymes, making cancer cells more sensitive to chemotherapy and radiation. Elevated temperatures also induce cell cycle arrest and trigger apoptotic pathways. Furthermore, hyperthermia modifies the expression of heat shock proteins, which play vital roles in cancer therapy, including enhancing immune responses. Hyperthermic treatments also have a significant impact on the body's immune response against tumors, potentially improving the efficacy of immune checkpoint inhibitors. Mild systemic hyperthermia (39°C-41°C) mimics fever, activating immune cells and raising metabolic rates. Intense heat above 50°C can release tumor antigens, enhancing immune reactions. Using photothermal nanoparticles for targeted heating and drug delivery can also modulate the immune response. Hyperthermia emerges as a cost-effective and well-tolerated adjuvant therapy when integrated with immunotherapy. This comprehensive review serves as a valuable resource for the selection of patient-specific treatments and the guidance of future experimental studies.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
| | - Gyöngyi Munkácsy
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
| | - Balázs Győrffy
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
- University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| |
Collapse
|
50
|
Liu X, Liu G, Mao Y, Luo J, Cao Y, Tan W, Li W, Yu H, Jia X, Li H. Engineering extracellular vesicles mimetics for targeted chemotherapy of drug-resistant ovary cancer. Nanomedicine (Lond) 2024; 19:25-41. [PMID: 38059464 DOI: 10.2217/nnm-2023-0289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
Abstract
Aim: To develop nanocarriers for targeting the delivery of chemotherapeutics to overcome multidrug-resistant ovarian cancer. Materials & methods: Doxorubicin-loaded nanovesicles were obtained through serial extrusion, followed by loading of P-glycoprotein siRNA and folic acid. The targeting ability and anticancer efficacy of the nanovesicles were evaluated. Results: The doxorubicin-loaded nanovesicles showed a high production yield. The presence of P-glycoprotein siRNA and folic acid resulted in reversed drug resistance and tumor targeting. This nanoplatform tremendously inhibited the viability of multidrug-resistant ovarian cancer cells, which was able to target tumor tissue and suppress tumor growth without adverse effects. Conclusion: These bioengineered nanovesicles could serve as novel extracellular vesicles mimetics for chemotherapeutics delivery to overcome multidrug resistance.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity & Child Health Care Hospital, Nanjing, 210001, China
| | - Guangquan Liu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity & Child Health Care Hospital, Nanjing, 210001, China
| | - Yinghua Mao
- Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China
| | - Jie Luo
- Department of Healthcare, General Hospital of Eastern Theater Command, Nanjing, 210002, China
| | - Yongping Cao
- Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China
| | - Weilong Tan
- Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China
| | - Wenhao Li
- Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China
| | - Huanhuan Yu
- Department of Clinical Pharmacy, General Hospital of Eastern Theater Command, Nanjing, 210002, China
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity & Child Health Care Hospital, Nanjing, 210001, China
| | - Hong Li
- Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China
| |
Collapse
|