1
|
Kim YM, Kim H, Park SC, Lee M, Jang MK. Targeted drug delivery of cancer cell-derived extracellular vesicles decorated with a VEGFR-binding peptide. Colloids Surf B Biointerfaces 2025; 252:114661. [PMID: 40203507 DOI: 10.1016/j.colsurfb.2025.114661] [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: 01/15/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
Chemotherapy is commonly used to manage cancer lesions, but its use is limited or has a high risk of failure due to the side effects of the drugs. The use of a drug delivery system can minimize the side effects of drugs and maximize their anticancer effects. This study investigated the potential of tumor cell-derived small extracellular vesicles (sEV) as drug delivery vehicles for doxorubicin (Dox). In addition, the decoration of vascular endothelial growth factor receptor (VEGFR)-targeting peptide on the sEV provided an enhance specific cancer cell-targeting effect in vitro or homing capacity in vivo. The extrusion method was effective in loading Dox and displaying targeting peptide. The effective Dox release was resulted under acidic condition, an endosome pH. The growth of tumor masses on MCF-7 xenografted mice were significantly inhibited by this drug delivery system. We believe that our results will provide useful information for the development of chemotherapeutic drug delivery systems.
Collapse
Affiliation(s)
- Young-Min Kim
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea
| | - Hyeonseok Kim
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea
| | - Seong-Cheol Park
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea
| | - Mina Lee
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea.
| | - Mi-Kyeong Jang
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea.
| |
Collapse
|
2
|
Yang Y, Zhang C, Li H, He Q, Xie J, Liu H, Cui F, Lei Z, Qin X, Liu Y, Xu M, Huang S, Zhang X. A review of molecular interplay between inflammation and cancer: The role of lncRNAs in pathogenesis and therapeutic potential. Int J Biol Macromol 2025; 309:142824. [PMID: 40187457 DOI: 10.1016/j.ijbiomac.2025.142824] [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: 01/17/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
The inflammatory microenvironment (IME) has been demonstrated to facilitate the initiation and progression of tumors throughout the inflammatory process. Simultaneously, cancer can initiate or intensify the inflammatory response, thereby promoting tumor progression. This review examines the dual role of long non-coding RNAs (lncRNAs) in the interplay between inflammation and cancer. LncRNA modulate inflammation-induced cancer by influencing the activation of signaling pathways (NF-κB, Wnt/β-catenin, mTOR, etc), microRNA (miRNA) sponging, protein interactions, interactions with immune cells, and encoding short peptides. In contrast, lncRNAs also impact cancer-induced inflammatory processes by regulating cytokine expression, mediating tumor-derived extracellular vesicles (EVs), modulating intracellular reactive oxygen species (ROS) levels, and facilitating metabolic reprogramming. Furthermore, the therapeutic potential of lncRNA and the challenges of clinical translation were explicitly discussed as well. Overall, this review aims to provide a comprehensive and systematic resource for future researchers investigating the impact of lncRNAs on inflammation and cancer.
Collapse
Affiliation(s)
- Yan Yang
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China; School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Chuxi Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Huacui Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China; Tangshan Institute of Southwest Jiaotong University, Tangshan, China
| | - Qin He
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Jiang Xie
- Department of Pediatrics, The Third People's Hospital of Chengdu, Chengdu, China
| | - Hongmei Liu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Fenfang Cui
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Ziqin Lei
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Xiaoyan Qin
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Ying Liu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Min Xu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China.
| | - Shuai Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China.
| | - Xu Zhang
- Department of Pharmacy, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu University of TCM, Chengdu, China.
| |
Collapse
|
3
|
Delavari B, Bigdeli B, Khazeni S, Varamini P. Nanodiamond-Protein hybrid Nanoparticles: LHRH receptor targeted and co-delivery of doxorubicin and dasatinib for triple negative breast cancer therapy. Int J Pharm 2025; 675:125544. [PMID: 40187703 DOI: 10.1016/j.ijpharm.2025.125544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/28/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025]
Abstract
Triple Negative Breast Cancer (TNBC) is an aggressive type of breast cancer that is difficult to treat with conventional therapies. This study aimed to develop a novel therapeutic approach using a multifunctional protein-nanodiamond nanocomposite to co-deliver doxorubicin (DOX) and dasatinib (DAS) to cancer cells via luteinising hormone-releasing hormone receptors. Nanodiamonds help retain DOX in targeted cells, while α-lactalbumin efficiently encapsulates DAS, reducing side effects. We successfully formulated the nanocomposite with over 80 % drug loading efficiency for both drugs. The imine Schiff-base bond in the nanocomposite hydrolyzes in the acidic pH tumor environment, triggering approximately 65 % drug release after 72 h, compared to less than 20 % in neutral pH. In vitro studies showed enhanced uptake of DOX and DAS in TNBC cell lines, potentially overcoming drug resistance. The combined delivery showed enhanced synergistic cytotoxic effects in drug-resistant TNBC cell models. For example, in the MDA-MB-231 cell line, the IC50 of DOX dropped to 45.63 ng/ml, while in MDA-MB-468, DAS decreased to 35.85 ng/ml with nanoparticle therapy. In vivo experiments utilizing a TNBC mouse model demonstrated the therapeutic effectiveness of the nanocomposite, leading to a 55 % reduction in tumor growth and enhanced survival rates. All mice given the nanocomposite survived after 44 days, but most treated with the DOX/DAS mixture died by day 28. This research showcases multifunctional nanocomposites as targeted drug delivery systems for TNBC, improving drug uptake and cytotoxicity. This strategy presents a promising method for treating drug-resistant breast cancer, with potential clinical applications and synergy with other therapies.
Collapse
Affiliation(s)
- Behdad Delavari
- School of Biomedical Science, Faculty of Medicine and Health, University of New South Wales, 2052 NSW, Australia; School of Pharmacy, Faculty of Medicine and Health, University of Sydney, 2006 NSW, Australia.
| | - Bahareh Bigdeli
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, 2006 NSW, Australia.
| | - Sepideh Khazeni
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, 2006 NSW, Australia.
| | - Pegah Varamini
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, 2006 NSW, Australia; Nano Institute, University of Sydney, 2006 NSW, Australia.
| |
Collapse
|
4
|
Fang K, Yuan S, Zhang X, Zhang J, Sun SL, Li X. Regulation of immunogenic cell death and potential applications in cancer therapy. Front Immunol 2025; 16:1571212. [PMID: 40207233 PMCID: PMC11979251 DOI: 10.3389/fimmu.2025.1571212] [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: 02/05/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
Immunogenic cell death (ICD), a type of regulatory cell death, plays an important role in activating the adaptive immune response. Activation of the tumor-specific immune response is accompanied by the cell surface exposure of calreticulin and heat-shock proteins, the secretion of adenosine triphosphate, and the release of high mobility group box-1. In this review, we summarize and classify the latest types of ICD inducers and their molecular mechanisms, and discuss the effects and potential applications of inducing ICD by chemotherapy drugs, targeted drugs, and oncolytic viruses in clinical research. We also explore the potential role of epigenetic modifiers in the induction of ICD, and clarify the synergistic anti-tumor effects of nano-pulse stimulation, radiosensitizers for radiotherapy, photosensitizers for photodynamic therapy, photothermal therapy, and other physical stimulation, combined with radiotherapy and chemotherapy induced-ICD, in multimodal immunotherapy. In addition, we elucidate the molecular mechanism of ICD in detail, including the calcium imbalance, mitochondrial stress, and the interactions in the tumor microenvironment. Ultimately, this review aims to offer deeper insight into the factors and mechanisms of ICD induction and provide a theoretical basis for the future development of ICD-based immunotherapy.
Collapse
Affiliation(s)
- Kun Fang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Shuai Yuan
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Xue Zhang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Jingdong Zhang
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
- Department of Medical Oncology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
| | - Shu-lan Sun
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Xiaoxi Li
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| |
Collapse
|
5
|
Wang Y, Qi P, Shi S, Pang C, Wang W, Fang D. Targeted therapy for glioblastoma utilizing hyaluronic acid-engineered liposomes for adriamycin delivery. NANOTECHNOLOGY 2025; 36:125102. [PMID: 39842037 DOI: 10.1088/1361-6528/adacef] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 01/22/2025] [Indexed: 01/24/2025]
Abstract
Glioblastoma (GBM) is a malignant tumor with highly heterogeneous and invasive characteristics leading to a poor prognosis. The CD44 molecule, which is highly expressed in GBM, has emerged as a highly sought-after biological marker. Therapeutic strategies targeting the cell membrane protein CD44 have emerged, demonstrating novel therapeutic potential. In this study, we constructed a nanodrug system (HA-Liposome@Dox) based on hyaluronic acid-engineered liposomes delivering adriamycin to target GBM. The system efficiently encapsulated Dox inside the liposomes through a hydrophilic-hydrophobic interaction mechanism, and the resulting HA-Liposome@Dox exhibited excellent loading efficacy, attributed to its uniform particle size distribution and negatively charged surface. Further evaluation revealed that HA-Liposome@Dox possessed excellent stability and safety and could promote the effective uptake of drug particles by CD44-overexpressing tumor cells, thus exerting a more potent cell-killing effect. Notably, in the treatment of GBM, HA-Liposome@Dox demonstrated significantly greater tumor growth inhibition compared to free Dox and prolonged the survival of tumor-bearing mice. Taken together, the present study not only verified the feasibility of HA-Liposome@Dox as an effective therapeutic tool against GBM and other CD44-positively expressing tumors, but also opened a promising new avenue for the clinical treatment of this type of refractory malignancies.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| | - Peiyan Qi
- Guangzhou International Travel Healthcare Center, Guangzhou Customs District P.R.China, Guangzhou, People's Republic of China
| | - Shenbao Shi
- Department of Neurosurgery, Zhujiang Hospital Southern Medical University, Guangzhou, People's Republic of China
| | - Cong Pang
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| | - Weijie Wang
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| | - Dazhao Fang
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| |
Collapse
|
6
|
Bisht A, Avinash D, Sahu KK, Patel P, Das Gupta G, Kurmi BD. A comprehensive review on doxorubicin: mechanisms, toxicity, clinical trials, combination therapies and nanoformulations in breast cancer. Drug Deliv Transl Res 2025; 15:102-133. [PMID: 38884850 DOI: 10.1007/s13346-024-01648-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
Doxorubicin is a key treatment for breast cancer, but its effectiveness often comes with significant side effects. Its actions include DNA intercalation, topoisomerase II inhibition, and reactive oxygen species generation, leading to DNA damage and cell death. However, it can also cause heart problems and low blood cell counts. Current trials aim to improve doxorubicin therapy by adjusting doses, using different administration methods, and combining it with targeted treatments or immunotherapy. Nanoformulations show promise in enhancing doxorubicin's effectiveness by improving drug delivery, reducing side effects, and overcoming drug resistance. This review summarizes recent progress and difficulties in using doxorubicin for breast cancer, highlighting its mechanisms, side effects, ongoing trials, and the potential impact of nanoformulations. Understanding these different aspects is crucial in optimizing doxorubicin's use and improving outcomes for breast cancer patients. This review examines the toxicity of doxorubicin, a drug used in breast cancer treatment, and discusses strategies to mitigate adverse effects, such as cardioprotective agents and liposomal formulations. It also discusses clinical trials evaluating doxorubicin-based regimens, the evolving landscape of combination therapies, and the potential of nanoformulations to optimize delivery and reduce systemic toxicity. The review also discusses the potential of liposomes, nanoparticles, and polymeric micelles to enhance drug accumulation within tumor tissues while sparing healthy organs.
Collapse
Affiliation(s)
- Anjali Bisht
- Department of Pharmaceutical Quality Assurance, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Dubey Avinash
- Department of Pharmaceutical Quality Assurance, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, 17 km Stone, NH-2, Chaumuhan, Mathura, 281406, UP, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
7
|
Pal M, Bera A, Masarkar N, Upadhyay A, Mukherjee S, Roy M. Targeted Chemo-Phototherapy in Red Light with Novel Doxorubicin and Iron(III) Complex-Functionalized Gold Nanoconjugate (Dox-Fe@FA-AuNPs). Chem Asian J 2024; 19:e202400616. [PMID: 38923831 DOI: 10.1002/asia.202400616] [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: 05/30/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
The anticancer efficacy of doxorubicin, an anthracycline-based and FDA-approved chemotherapeutic drug, is significantly hindered by acquired chemoresistance and severe side effects despite its potent anticancer properties. To overcome these challenges, we developed an innovative therapeutic formulation that integrates targeted chemotherapy and phototherapy within a single platform using gold nanoparticles (AuNPs). This novel nanoconjugate, designated as Dox-Fe@FA-AuNPs, is co-functionalized with folic acid, doxorubicin, and an iron(III)-phenolate/carboxylate complex, enabling cancer-specific drug activation. Here, we report the synthesis, characterization, and comprehensive physico-chemical and biological evaluations of Dox-Fe@FA-AuNPs. The nanoconjugate exhibited excellent solubility, stability, and enhanced cellular uptake in folate receptor-positive cancer cells. The nanoconjugate was potently cytotoxic against HeLa and MDA-MB-231 cancer cells (HeLa: 105.5±16.52 μg mL-1; MDA-MB-231: 112.0±12.31 μg mL-1; MDA-MB-231 (3D): 156.31±19.35 μg mL-1) while less cytotoxic to the folate(-) cancer cells (MCF-7, A549 and HepG2). The cytotoxicity was attributed to the pH-dependent release of doxorubicin, which preferentially occurs in the acidic tumor microenvironment. Additionally, under red light irradiation, the nanoconjugate generated ROS, inducing caspase-3/7-dependent apoptosis with a photo-index (PI) >50, and inhibited cancer cell migration. Our findings underscore the potential of Dox-Fe@FA-AuNPs as a highly effective and sustainable platform for targeted chemo-phototherapy.
Collapse
Affiliation(s)
- Maynak Pal
- Department of Chemistry, National Institute of Technology Manipur, Langol, 795004, Imphal West, Manipur
| | - Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore, Bangalore, 560012, Karnataka
| | - Neha Masarkar
- Department of Biochemistry, AIIMS Bhopal, Saket Nagar, Bhopal, Madhya Pradesh
| | - Aarti Upadhyay
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore, Bangalore, 560012, Karnataka
| | - Sukhes Mukherjee
- Department of Biochemistry, AIIMS Bhopal, Saket Nagar, Bhopal, Madhya Pradesh
| | - Mithun Roy
- Department of Chemistry, National Institute of Technology Manipur, Langol, 795004, Imphal West, Manipur
- Department of Chemistry, National Institute of Technology Agartala, Jirania, 799046, Tripura West
| |
Collapse
|
8
|
Serras A, Faustino C, Pinheiro L. Functionalized Polymeric Micelles for Targeted Cancer Therapy: Steps from Conceptualization to Clinical Trials. Pharmaceutics 2024; 16:1047. [PMID: 39204392 PMCID: PMC11359152 DOI: 10.3390/pharmaceutics16081047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer is still ranked among the top three causes of death in the 30- to 69-year-old age group in most countries and carries considerable societal and macroeconomic costs that differ depending on the cancer type, geography, and patient gender. Despite advances in several pharmacological approaches, the lack of stability and specificity, dose-related toxicity, and limited bioavailability of chemotherapy (standard therapy) pose major obstacles in cancer treatment, with multidrug resistance being a driving factor in chemotherapy failure. The past three decades have been the stage for intense research activity on the topic of nanomedicine, which has resulted in many nanotherapeutics with reduced toxicity, increased bioavailability, and improved pharmacokinetics and therapeutic efficacy employing smart drug delivery systems (SDDSs). Polymeric micelles (PMs) have become an auspicious DDS for medicinal compounds, being used to encapsulate hydrophobic drugs that also exhibit substantial toxicity. Through preclinical animal testing, PMs improved pharmacokinetic profiles and increased efficacy, resulting in a higher safety profile for therapeutic drugs. This review focuses on PMs that are already in clinical trials, traveling the pathways from preclinical to clinical studies until introduction to the market.
Collapse
Affiliation(s)
| | - Célia Faustino
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa (ULisboa), Avenida Professor Gama PintoGama Pinto, 1649-003 Lisboa, Portugal; (A.S.); (L.P.)
| | | |
Collapse
|
9
|
Zanganeh S, Firoozpour L, Salavatipour MS, Sardari S, Cohan RA, Mohajel N. Critical Aggregation Concentration Can be a Predictor of Doxorubicin Delivery Performance of Self-Assembling Amphiphilic Peptides with Different Hydrophobic Tails. J Pharm Sci 2024; 113:2188-2197. [PMID: 38417791 DOI: 10.1016/j.xphs.2024.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/17/2024] [Accepted: 02/17/2024] [Indexed: 03/01/2024]
Abstract
Amphiphilic peptides hold great potential as drug delivery systems. A popular peptide design approach has been to place amino acids in the peptide sequence based on their known properties. On the other hand, the directed discovery approach aims to screen a sequence space for a desired property. However, screening amphiphilic peptides for desirable drug delivery properties is not possible without a quantity that is predictive of these properties. We studied the predictive power of critical aggregation concentration (CAC) values on the drug delivery performance of a series of amphiphilic peptides with different hydrophobic tails and close CAC values. The CAC values were predicted by our previously developed model and doxorubicin was used as a model hydrophobic drug. All peptides showed close drug loading, entrapment efficiency, and release profile. They also formed similar spherical particles by assembling in reverse β-sheet arrangements regardless of drug presence. Moreover, the assembled particles were able to accumulate doxorubicin inside ordinary as well as drug-resistant breast cancer cells and enhance its toxicity up to 39 and 17 folds, respectively. It can be concluded that similar drug delivery properties displayed by the peptides can be attributed to their similar hydrophilic-lipophilic balance as reflected in their close CAC values.
Collapse
Affiliation(s)
- Saeed Zanganeh
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran; Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, and Drug Design & Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | | | - Soroush Sardari
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran 1316943551, Iran.
| | - Nasir Mohajel
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran 1316943551, Iran.
| |
Collapse
|
10
|
Gaubert A, Castagnet T, Marsh J, Barthélémy P. Fluorinated GlycoNucleoLipid-based hydrogels as new spatiotemporal stimulable DDS. Drug Deliv Transl Res 2024; 14:2079-2084. [PMID: 38388815 DOI: 10.1007/s13346-024-01550-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
Achieving a controlled release of several active pharmaceutical ingredients (APIs) remains a challenge for improving their therapeutic effects and reduced their side effects. In the current work, stimulable Drug Delivery Systems (DDS) based on supramolecular hydrogels were designed by combining two APIs featuring anticancer activities, namely the doxorubicin and phenazine 14. In vitro studies revealed promising physicochemical properties for all the investigated API loaded gels. Fluorinated GlycoNucleoLipid (GNF) based supramolecular gels remain stable in the presence of either doxorubicin (Doxo) or phenazine 14 (Phe) as anticancer drugs. Noteworthy, the stiffness of the GNF-based supramolecular gels was enhanced in the presence of both APIs while maintaining their thixotropic properties. We demonstrated that the storage modulus (G') of the GNF gels was increased from 1.3 kPa to 9.3 kPa upon loading of both APIs within the same gels. With a low mechanical stimulation (within the LVR), a passive diffusion out of gels was observed for Dox whereas Phe remained trapped in the GNF gels over several hours. Also, in this work we showed that mechanical stress triggered the release of both Phe and Doxo at different rates.
Collapse
Affiliation(s)
- Alexandra Gaubert
- University of Bordeaux, ARNA laboratory, INSERM U1212, UMR CNRS 5320, F-33076, Bordeaux, France.
| | - Thibault Castagnet
- University of Bordeaux, ARNA laboratory, INSERM U1212, UMR CNRS 5320, F-33076, Bordeaux, France
| | - Jevon Marsh
- University of Bordeaux, ARNA laboratory, INSERM U1212, UMR CNRS 5320, F-33076, Bordeaux, France
| | - Philippe Barthélémy
- University of Bordeaux, ARNA laboratory, INSERM U1212, UMR CNRS 5320, F-33076, Bordeaux, France.
| |
Collapse
|
11
|
Jia N, Wang Q, Li W, Chen D, Hu H. Membrane Fusion Liposomes Deliver Antifibrotic and Chemotherapeutic Drugs Sequentially to Enhance Tumor Treatment Efficacy by Reshaping Tumor Microenvironment. Adv Healthc Mater 2024; 13:e2400219. [PMID: 38657266 DOI: 10.1002/adhm.202400219] [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: 01/19/2024] [Revised: 04/14/2024] [Indexed: 04/26/2024]
Abstract
The intricate tumor microenvironment in triple-negative breast cancer (TNBC) hampers chemotherapy and immunotherapy efficacy due to dense extracellular matrix (ECM) by tumor-associated fibroblasts (TAFs). Nanoparticle-based therapies, especially "all-in-one" nanoparticles, have shown great potential in combined drug delivery strategies to reshape the tumor microenvironment and enhance therapeutic efficiency. However, these "all-in-one" nanoparticles suffer from limitations in targeting different target cells, uncontrollable dosing ratio, and disregarding the impact of delivery schedules. This study prepared cell membrane fusion liposomes (TAFsomes and CCMsomes) to load FDA-approved antifibrotic drug pirfenidone (PFD/TAFsomes) and antitumor drug doxorubicin (DOX/CCMsomes). These liposomes can specifically target TAFs cells and tumor cells, and combined administration can effectively inhibit TAFs activity, reshape the tumor microenvironment (TME), and significantly enhance the tumor chemotherapy efficacy. Combined drug delivery defeats "all-in-one" liposomes (DOX/PFD/Liposomes, DOX/PFD/TAFsomes, and DOX/PFD/CCMsomes) by flexibly adjusting the drug delivery ratio. Moreover, an asynchronous delivery strategy that optimizes the administration schedule not only further improves the therapeutic effect, but also amplifies the effectiveness of α-PD-L1 immunotherapy by modulating the tumor immune microenvironment. This delivery strategy provides a personalized treatment approach with clinical translation potential, providing new ideas for enhancing the therapeutic effect against solid tumors such as TNBC.
Collapse
Affiliation(s)
- Nan Jia
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Qi Wang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Wenpan Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Haiyang Hu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| |
Collapse
|
12
|
Vitale R, Marzocco S, Popolo A. Role of Oxidative Stress and Inflammation in Doxorubicin-Induced Cardiotoxicity: A Brief Account. Int J Mol Sci 2024; 25:7477. [PMID: 39000584 PMCID: PMC11242665 DOI: 10.3390/ijms25137477] [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: 05/21/2024] [Revised: 07/01/2024] [Accepted: 07/06/2024] [Indexed: 07/16/2024] Open
Abstract
Cardiotoxicity is the main side effect of several chemotherapeutic drugs. Doxorubicin (Doxo) is one of the most used anthracyclines in the treatment of many tumors, but the development of acute and chronic cardiotoxicity limits its clinical usefulness. Different studies focused only on the effects of long-term Doxo administration, but recent data show that cardiomyocyte damage is an early event induced by Doxo after a single administration that can be followed by progressive functional decline, leading to overt heart failure. The knowledge of molecular mechanisms involved in the early stage of Doxo-induced cardiotoxicity is of paramount importance to treating and/or preventing it. This review aims to illustrate several mechanisms thought to underlie Doxo-induced cardiotoxicity, such as oxidative and nitrosative stress, inflammation, and mitochondrial dysfunction. Moreover, here we report data from both in vitro and in vivo studies indicating new therapeutic strategies to prevent Doxo-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | - Ada Popolo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (R.V.); (S.M.)
| |
Collapse
|
13
|
Hu K, Zhang D, Ma W, Gu Y, Zhao J, Mu X. Polydopamine-Based Nanoparticles for Synergistic Chemotherapy of Prostate Cancer. Int J Nanomedicine 2024; 19:6717-6730. [PMID: 38979530 PMCID: PMC11230127 DOI: 10.2147/ijn.s468946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024] Open
Abstract
Introduction Immune regulatory small molecule JQ1 can block its downstream effector PD-L1 pathway and effectively reverse the PD-L1 upregulation induced by doxorubicin (DOX). So the synergistic administration of chemotherapeutic drug DOX and JQ1 is expected to increase the sensitivity of tumors to immune checkpoint therapy and jointly enhance the body's own immunity, thus effectively killing tumor cells. Therefore, a drug delivery system loaded with DOX and JQ1 was devised in this study. Methods Polydopamine nanoparticles (PDA NPs) were synthesized through spontaneous polymerization. Under appropriate pH conditions, DOX and JQ1 were loaded onto the surface of PDA NPs, and the release of DOX and JQ1 were measured using UV-Vis or high performance liquid chromatography (HPLC). The mechanism of fabricated nanocomplex in vitro was investigated by cell uptake experiment, cell viability assays, apoptosis assays, and Western blot analysis. Finally, the tumor-bearing mouse model was used to evaluate the tumor-inhibiting efficacy and the biosafety in vivo. Results JQ1 and DOX were successfully loaded onto PDA NPs. PDA-DOX/JQ1 NPs inhibited the growth of prostate cancer cells, reduced the expression of apoptosis related proteins and induced apoptosis in vitro. The in vivo biodistribution indicated that PDA-DOX/JQ1 NPs could accumulated at the tumor sites through the EPR effect. In tumor-bearing mice, JQ1 delivered with PDA-DOX/JQ1 NPs reduced PD-L1 expression at tumor sites, generating significant tumor suppression. Furthermore, PDA-DOX/JQ1 NPs could reduce the side effects, and produce good synergistic treatment effect in vivo. Conclusion We have successfully prepared a multifunctional platform for synergistic prostate cancer therapy.
Collapse
Affiliation(s)
- Kebang Hu
- Department of Urology, Lequn Branch, The First Hospital of Jilin University, Changchun, 130031, People’s Republic of China
| | - Dongqi Zhang
- Department of Urology, Lequn Branch, The First Hospital of Jilin University, Changchun, 130031, People’s Republic of China
| | - Weiran Ma
- College of Pharmacy, Jilin University, Changchun, 130021, People’s Republic of China
| | - Yanzhi Gu
- College of Pharmacy, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jiang Zhao
- Department of Urology, Xi’an First Hospital, Xi’an, 710002, People’s Republic of China
| | - Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| |
Collapse
|
14
|
Szota M, Szwedowicz U, Rembialkowska N, Janicka-Klos A, Doveiko D, Chen Y, Kulbacka J, Jachimska B. Dendrimer Platforms for Targeted Doxorubicin Delivery-Physicochemical Properties in Context of Biological Responses. Int J Mol Sci 2024; 25:7201. [PMID: 39000306 PMCID: PMC11241532 DOI: 10.3390/ijms25137201] [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: 04/30/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
The unique structure of G4.0 PAMAM dendrimers allows a drug to be enclosed in internal spaces or immobilized on the surface. In the conducted research, the conditions for the formation of the active G4.0 PAMAM complex with doxorubicin hydrochloride (DOX) were optimized. The physicochemical properties of the system were monitored using dynamic light scattering (DLS), circular dichroism (CD), and fluorescence spectroscopy. The Quartz Crystal Microbalance with Dissipation Monitoring (QCM-D) method was chosen to determine the preferential conditions for the complex formation. The highest binding efficiency of the drug to the cationic dendrimer was observed under basic conditions when the DOX molecule was deprotonated. The decrease in the zeta potential of the complex confirms that DOX immobilizes through electrostatic interaction with the carrier's surface amine groups. The binding constants were determined from the fluorescence quenching of the DOX molecule in the presence of G4.0 PAMAM. The two-fold way of binding doxorubicin in the structure of dendrimers was visible in the Isothermal calorimetry (ITC) isotherm. Fluorescence spectra and release curves identified the reversible binding of DOX to the nanocarrier. Among the selected cancer cells, the most promising anticancer activity of the G4.0-DOX complex was observed in A375 malignant melanoma cells. Moreover, the preferred intracellular location of the complexes concerning the free drug was found, which is essential from a therapeutic point of view.
Collapse
Affiliation(s)
- Magdalena Szota
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, 30-239 Cracow, Poland
| | - Urszula Szwedowicz
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Nina Rembialkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Anna Janicka-Klos
- Department of Basic Chemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Daniel Doveiko
- Department of Physics, University of Strathclyde, Glasgow G4 0NG, UK
| | - Yu Chen
- Department of Physics, University of Strathclyde, Glasgow G4 0NG, UK
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Barbara Jachimska
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, 30-239 Cracow, Poland
| |
Collapse
|
15
|
Ito C, Taguchi K, Yamada T, Hanaya K, Enoki Y, Sugai T, Komatsu T, Matsumoto K. Dual delivery of carbon monoxide and doxorubicin using haemoglobin-albumin cluster: proof of concept for well-tolerated cancer therapy. J Mater Chem B 2024; 12:5600-5608. [PMID: 38738920 DOI: 10.1039/d4tb00123k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
A serious concern of doxorubicin (DOX) therapy is that it causes severe adverse effects, particularly cardiotoxicity. Carbon monoxide (CO) possesses powerful cytoprotective effects against drug-induced organ injury and is expected to ameliorate DOX-induced cardiotoxicity. In this study, a dual carrier of DOX and CO (CO-HemoAct-DOX) was fabricated based on a haemoglobin-albumin cluster (HemoAct), which is a protein cluster with a haemoglobin core structure wrapped by serum albumin. CO-HemoAct-DOX was synthesised by binding CO to a haemoglobin core and covalently conjugating (6-maleimidocaproyl)hydrazone derivative of DOX to an albumin shell. The average DOX/cluster ratio was about 2.6. In the in vitro cytotoxicity assay against cancer cells, the anti-tumour activity of CO-HemoAct-DOX was 10-fold lower than that of DOX in a 2D-cultured model, whereas CO-HemoAct-DOX suppressed the growth of tumour spheroids to the same extent as DOX in the 3D-cultured model. In colon-26 tumour-bearing mice, CO-HemoAct-DOX achieved DOX delivery to the tumour site and alleviated tumour growth more effectively than DOX. Furthermore, CO-HemoAct attenuated DOX-induced cardiomyocyte atrophy in H9c2 cells and elevated the levels of cardiac biomarkers in mice exposed to DOX. These results suggest that the dual delivery of CO and DOX using HemoAct is a promising strategy as an anti-tumour agent to realise well-tolerated cancer therapy with minimal cardiotoxicity.
Collapse
Affiliation(s)
- Chihiro Ito
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan.
| | - Kazuaki Taguchi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan.
| | - Taiga Yamada
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Kengo Hanaya
- Division of Organic and Biocatalytic Chemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Yuki Enoki
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan.
| | - Takeshi Sugai
- Division of Organic and Biocatalytic Chemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Teruyuki Komatsu
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Kazuaki Matsumoto
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan.
| |
Collapse
|
16
|
Sevieri M, Andreata F, Mainini F, Signati L, Piccotti F, Truffi M, Bonizzi A, Sitia L, Pigliacelli C, Morasso C, Tagliaferri B, Corsi F, Mazzucchelli S. Impact of doxorubicin-loaded ferritin nanocages (FerOX) vs. free doxorubicin on T lymphocytes: a translational clinical study on breast cancer patients undergoing neoadjuvant chemotherapy. J Nanobiotechnology 2024; 22:184. [PMID: 38622644 PMCID: PMC11020177 DOI: 10.1186/s12951-024-02441-4] [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: 11/06/2023] [Accepted: 03/25/2024] [Indexed: 04/17/2024] Open
Abstract
Despite the advent of numerous targeted therapies in clinical practice, anthracyclines, including doxorubicin (DOX), continue to play a pivotal role in breast cancer (BC) treatment. DOX directly disrupts DNA replication, demonstrating remarkable efficacy against BC cells. However, its non-specificity toward cancer cells leads to significant side effects, limiting its clinical utility. Interestingly, DOX can also enhance the antitumor immune response by promoting immunogenic cell death in BC cells, thereby facilitating the presentation of tumor antigens to the adaptive immune system. However, the generation of an adaptive immune response involves highly proliferative processes, which may be adversely affected by DOX-induced cytotoxicity. Therefore, understanding the impact of DOX on dividing T cells becomes crucial, to deepen our understanding and potentially devise strategies to shield anti-tumor immunity from DOX-induced toxicity. Our investigation focused on studying DOX uptake and its effects on human lymphocytes. We collected lymphocytes from healthy donors and BC patients undergoing neoadjuvant chemotherapy (NAC). Notably, patient-derived peripheral blood mononuclear cells (PBMC) promptly internalized DOX when incubated in vitro or isolated immediately after NAC. These DOX-treated PBMCs exhibited significant proliferative impairment compared to untreated cells or those isolated before treatment initiation. Intriguingly, among diverse lymphocyte sub-populations, CD8 + T cells exhibited the highest uptake of DOX. To address this concern, we explored a novel DOX formulation encapsulated in ferritin nanocages (FerOX). FerOX specifically targets tumors and effectively eradicates BC both in vitro and in vivo. Remarkably, only T cells treated with FerOX exhibited reduced DOX internalization, potentially minimizing cytotoxic effects on adaptive immunity.Our findings underscore the importance of optimizing DOX delivery to enhance its antitumor efficacy while minimizing adverse effects, highlighting the pivotal role played by FerOX in mitigating DOX-induced toxicity towards T-cells, thereby positioning it as a promising DOX formulation. This study contributes valuable insights to modern cancer therapy and immunomodulation.
Collapse
Affiliation(s)
- Marta Sevieri
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy
| | - Francesco Andreata
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Mainini
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy
| | - Lorena Signati
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| | | | - Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| | - Arianna Bonizzi
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| | - Leopoldo Sitia
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy
| | - Claudia Pigliacelli
- Laboratory of Supramolecular and Bio-Nanomaterials (SBNLab), Department of Chemistry, Materials, and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, 20131, Italy
| | - Carlo Morasso
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| | | | - Fabio Corsi
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy.
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy.
| | - Serena Mazzucchelli
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy.
| |
Collapse
|
17
|
Zhu M, Chen Y, Cheng L, Li X, Shen Y, Guo G, Xu X, Li H, Yang H, Liu C, He K. Calsyntenin-1 Promotes Doxorubicin-induced Dilated Cardiomyopathy in Rats. Cardiovasc Drugs Ther 2024; 38:237-252. [PMID: 36350487 PMCID: PMC10959838 DOI: 10.1007/s10557-022-07389-x] [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] [Accepted: 09/30/2022] [Indexed: 11/10/2022]
Abstract
PURPOSE Doxorubicin is an important cancer chemotherapeutic agent with severe cardiotoxic effects that eventually lead to dilated cardiomyopathy (DCM). Calsyntenin-1(CLSTN1) plays a critical role in the nervous system, but its relevance in cardiovascular diseases is unknown. We investigated the significance of CLSTN1 in doxorubicin-induced DCM. METHODS CLSTN1 expression in doxorubicin-induced DCM rats and H9c2 cells was determined using western blotting. To further explore the functions of CLSTN1, a cardiac-specific CLSTN1 overexpression rat model was constructed. The rats were subjected to analysis using echocardiographic, hemodynamic, and electrocardiographic parameters. Potential downstream molecules in CLSTN1 overexpression heart tissue were investigated using proteomics and western blotting. Finally, a knockdown of CLSTN1 was constructed to investigate the rescue function on doxorubicin-induced cell toxicity. RESULTS CLSTN1 protein expression increased drastically in doxorubicin-induced DCM rats and H9c2 cells. Under doxorubicin treatment, CLSTN1 protein-specific overexpression in the heart muscle promoted cardiac chamber enlargement and heart failure, while the knockdown of CLSTN1 reduced doxorubicin-induced cardiomyocyte toxicity in vitro. At the mechanistic level, overexpression of CLSTN1 downregulated SERCA2 expression and increased the phosphorylation levels of PI3K-Akt and CaMK2. CONCLUSION Our findings demonstrated that CLSTN1 promotes the pathogenesis of doxorubicin-induced DCM. CLSTN1 could be a therapeutic target to prevent the development of doxorubicin-induced DCM.
Collapse
Affiliation(s)
- Mingxiang Zhu
- Medical School of Chinese PLA, Beijing, 100853, China
- Medical Big Data Research Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yibing Chen
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Liting Cheng
- Medical School of Chinese PLA, Beijing, 100853, China
- Medical Big Data Research Center, Chinese PLA General Hospital, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xin Li
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanying Shen
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Ge Guo
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiang Xu
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Hanlu Li
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Hao Yang
- Department of Radiation Oncology, Inner Mongolia Cancer Hospital and Affiliated People's Hospital of Inner Mongolia Medical University, Huhhot, China
| | - Chunlei Liu
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China.
| | - Kunlun He
- Medical Big Data Research Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
18
|
Liu Q, Xiang Y, Yu Q, Lv Q, Xiang Z. A TME-activated nano-catalyst for triple synergistic therapy of colorectal cancer. Sci Rep 2024; 14:3328. [PMID: 38336997 PMCID: PMC10858196 DOI: 10.1038/s41598-024-53334-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Colorectal cancer cells are highly heterogeneous and exhibit various drug resistances, making personalized treatment necessary. This typically involves a combination of different treatment modalities such as surgery, radiation, and chemotherapy to increase patient survival. Inspired by this, synergistic therapy, which combines multiple modalities into a single nanotherapeutic drug, shows promise in treating cancer. In this study, a nano-catalyst based on calcium peroxide (CaO2) and the chemotherapeutic drug doxorubicin hydrochloride (DOX) co-loaded into HPB nanoparticles (HPB@CaO2/DOX-PAA) was developed to achieve synergistic cancer treatment through chemodynamic/chemo/photothermal (CDT/CT/PTT) mechanisms. After being endocytosed by cancer cells, the nano-catalyst decomposes, releasing cargo. During near-infrared light irradiation, HPB induces a photothermal effect, DOX exhibits significant RNA and DNA destruction capabilities, meanwhile CaO2 produces a large amount of H2O2 in the moderately acidic TME, which combines with Fe2+ ions derived from HPB to form the highly toxic •OH in a Fenton-like reaction, enhancing the chemodynamic treatment. Assays conducted ex vivo and in vivo have exhibited the efficacy of this triple synergistic therapeutic regimen, indicating its potential clinical application.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University Chongqing, Chongqing, China
- Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University Chongqing, Chongqing, China
- Department of Hepatobiliary Surgery, Suining First People's Hospital, Suining, China
| | - Yurong Xiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University Chongqing, Chongqing, China
- Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University Chongqing, Chongqing, China
| | - Qiang Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University Chongqing, Chongqing, China
- Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University Chongqing, Chongqing, China
| | - Quan Lv
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University Chongqing, Chongqing, China
- Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University Chongqing, Chongqing, China
| | - Zheng Xiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University Chongqing, Chongqing, China.
- Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University Chongqing, Chongqing, China.
| |
Collapse
|
19
|
Abstract
Anthracycline-induced cardiotoxicity (AIC) is a serious and common side effect of anthracycline therapy. Identification of genes and genetic variants associated with AIC risk has clinical potential as a cardiotoxicity predictive tool and to allow the development of personalized therapies. In this review, we provide an overview of the function of known AIC genes identified by association studies and categorize them based on their mechanistic implication in AIC. We also discuss the importance of functional validation of AIC-associated variants in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to advance the implementation of genetic predictive biomarkers. Finally, we review how patient-specific hiPSC-CMs can be used to identify novel patient-relevant functional targets and for the discovery of cardioprotectant drugs to prevent AIC. Implementation of functional validation and use of hiPSC-CMs for drug discovery will identify the next generation of highly effective and personalized cardioprotectants and accelerate the inclusion of approved AIC biomarkers into clinical practice.
Collapse
Affiliation(s)
- Romina B Cejas
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA;
| | - Kateryna Petrykey
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Paul W Burridge
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA;
| |
Collapse
|
20
|
Dai Q, Pan Y, Zhu X, Chen M, Xie L, Zhu Y, Wan G. Network Pharmacology along with Molecular Docking to Explore the Mechanism of Danshen Injection against Anthracycline-induced Cardiotoxicity and Transcriptome Validation. Curr Pharm Des 2024; 30:952-967. [PMID: 38482629 DOI: 10.2174/0113816128289845240305070522] [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: 10/28/2023] [Accepted: 02/20/2024] [Indexed: 06/21/2024]
Abstract
INTRODUCTION Although anthracyclines have demonstrated efficacy in cancer therapy, their utilization is constrained by cardiotoxicity. In contrast, Danshen injection (DSI), derived from Salvia miltiorrhiza, has a longstanding tradition of being employed to ameliorate cardiovascular ailments, including anthracycline- induced cardiotoxicity (AIC). Nonetheless, there is a notable dearth of comprehensive systematic investigation into the molecular mechanisms underlying DSI's effects on AIC. Consequently, this study was undertaken to explore the underlying mechanism by which DSI acted against AIC. METHODS Employing network pharmacology approach, the current investigation undertook a comprehensive analysis of the impact of DSI on AIC, which was further validated by transcriptome sequencing with in vitro AIC model. Additionally, molecular docking was conducted to evaluate the binding of active ingredients to core targets. A total of 3,404 AIC-related targets and 12 active ingredients in DSI, including chrysophanol, luteolin, tanshinone IIA, isoimperatorin, among others, were collected by differentially expressed analysis and database search, respectively. RESULTS The network pharmacology and enrichment analysis suggested 102 potential targets and 29 signaling pathways associated with the protective effect of DSI on AIC. Three core targets (CA12, NOS3, and POLH) and calcium signaling pathways were further validated by transcriptomic analysis of the in-vitro model. The high affinity of the active ingredients binding to corresponding targets was confirmed by molecular docking. CONCLUSION The present study suggested that DSI might exert a cardioprotective effect on AIC via the inhibition of CA12, NOS3, and POLH, as well as the modulation of calcium signaling. Further experiments are warranted to verify the findings.
Collapse
Affiliation(s)
- Quankai Dai
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yijun Pan
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Xiwen Zhu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Mengyao Chen
- Department of Oncology, Renmin Hospital, Institute of Medicine and Nursing, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Lin Xie
- Department of Oncology, Renmin Hospital, Institute of Medicine and Nursing, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yu Zhu
- Department of Research and Teaching, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Guoxing Wan
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| |
Collapse
|
21
|
Imran H, Tang Y, Wang S, Yan X, Liu C, Guo L, Wang E, Xu C. Optimized DOX Drug Deliveries via Chitosan-Mediated Nanoparticles and Stimuli Responses in Cancer Chemotherapy: A Review. Molecules 2023; 29:31. [PMID: 38202616 PMCID: PMC10780101 DOI: 10.3390/molecules29010031] [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: 10/15/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Chitosan nanoparticles (NPs) serve as useful multidrug delivery carriers in cancer chemotherapy. Chitosan has considerable potential in drug delivery systems (DDSs) for targeting tumor cells. Doxorubicin (DOX) has limited application due to its resistance and lack of specificity. Chitosan NPs have been used for DOX delivery because of their biocompatibility, biodegradability, drug encapsulation efficiency, and target specificity. In this review, various types of chitosan derivatives are discussed in DDSs to enhance the effectiveness of cancer treatments. Modified chitosan-DOX NP drug deliveries with other compounds also increase the penetration and efficiency of DOX against tumor cells. We also highlight the endogenous stimuli (pH, redox, enzyme) and exogenous stimuli (light, magnetic, ultrasound), and their positive effect on DOX drug delivery via chitosan NPs. Our study sheds light on the importance of chitosan NPs for DOX drug delivery in cancer treatment and may inspire the development of more effective approaches for cancer chemotherapy.
Collapse
Affiliation(s)
- HafizMuhammad Imran
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Yixin Tang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Siyuan Wang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Xiuzhang Yan
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Chang Liu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Lei Guo
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Erlei Wang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Caina Xu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| |
Collapse
|
22
|
Wei JB, Zeng XC, Ji KR, Zhang LY, Chen XM. Identification of Key Genes and Related Drugs of Adrenocortical Carcinoma by Integrated Bioinformatics Analysis. Horm Metab Res 2023. [PMID: 38109896 DOI: 10.1055/a-2209-0771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Adrenocortical carcinoma (ACC) is a malignant carcinoma with an extremely poor prognosis, and its pathogenesis remains to be understood to date, necessitating further investigation. This study aims to discover biomarkers and potential therapeutic agents for ACC through bioinformatics, enhancing clinical diagnosis and treatment strategies. Differentially expressed genes (DEGs) between ACC and normal adrenal cortex were screened out from the GSE19750 and GSE90713 datasets available in the GEO database. An online Venn diagram tool was utilized to identify the common DEGs between the two datasets. The identified DEGs were subjected to functional assessment, pathway enrichment, and identification of hub genes by performing the protein-protein interaction (PPI), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. The differences in the expressions of hub genes between ACC and normal adrenal cortex were validated at the GEPIA2 website, and the association of these genes with the overall patient survival was also assessed. Finally, on the QuartataWeb website, drugs related to the identified hub genes were determined. A total of 114 DEGs, 10 hub genes, and 69 known drugs that could interact with these genes were identified. The GO and KEGG analyses revealed a close association of the identified DEGs with cellular signal transduction. The 10 hub genes identified were overexpressed in ACC, in addition to being significantly associated with adverse prognosis in ACC. Three genes and the associated known drugs were identified as potential targets for ACC treatment.
Collapse
Affiliation(s)
- Jian-Bin Wei
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Xiao-Chun Zeng
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Kui-Rong Ji
- Department of Endocrinology, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Ling-Yi Zhang
- Department of Endocrinology, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Xiao-Min Chen
- Department of Endocrinology, Zhongshan Hospital Xiamen University, Xiamen, China
| |
Collapse
|
23
|
Rodrigues Esperandim T, Barcelos Ribeiro A, Silva Squarisi I, Teixeira Marcos de Souza L, Olimpio de Souza T, Oliveira Acésio N, Ferreira Conceição Santos M, Kenupp Bastos J, Ricardo Ambrósio S, Crispim Tavares D. Toxicological and chemoprevention studies of Brazilian brown propolis from Araucaria sp. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2023; 86:791-802. [PMID: 37592437 DOI: 10.1080/15287394.2023.2243976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Brazilian brown propolis (BBP) is a natural product derived predominantly from the south region of Brazil, where Araucaria forests are dominant. Despite its potential as a source of bioactive compounds with leishmanicidal, anti-inflammatory, nociceptive, and antimicrobial properties, BBP has not been comprehensively studied compared to green propolis. Therefore, this study aimed to determine the safety and chemopreventive potential of BBP. The cytotoxicity attributed to BBP was assessed using two different assays, while the Salmonella/microsome assay was employed to evaluate mutagenicity. The acute toxicity attributed to BBP was determined using a zebrafish model, while the chemopreventive potential was investigated utilizing Chinese hamster lung (V79) cell lines. Data demonstrated that BBP exerted cytotoxic effects at concentrations greater than or equal to 10 µg/ml and did not exhibit mutagenicity in Salmonella typhimurium strains TA98 and TA100. However, at the highest concentration tested (4000 µg/plate), BBP induced a significant increase in revertant colonies in S. typhimurium TA102 strain. The LC50 equivalent to 8.83 mg/L was obtained in the acute toxicity evaluation in zebrafish. BBP also showed antigenotoxic effect by significantly reducing chromosomal damage induced by the mutagen doxorubicin in V79 cell cultures at a concentration of 2.5 μg/ml. Compared to Brazilian green and red propolis, BBP exhibited greater toxicity. On the other hand, at lower concentrations, BBP displayed chemopreventive potential, which may be associated with the antioxidant capacity of the extract. These findings contribute to a better understanding of the biological properties and potential applications of BBP in treating various diseases.
Collapse
Affiliation(s)
| | - Arthur Barcelos Ribeiro
- University of Franca, Av. Dr. Armando Salles Oliveira, 201, 14.404-600 Franca, São Paulo, Brazil
| | - Iara Silva Squarisi
- University of Franca, Av. Dr. Armando Salles Oliveira, 201, 14.404-600 Franca, São Paulo, Brazil
| | | | - Thiago Olimpio de Souza
- University of Franca, Av. Dr. Armando Salles Oliveira, 201, 14.404-600 Franca, São Paulo, Brazil
| | - Nathália Oliveira Acésio
- University of Franca, Av. Dr. Armando Salles Oliveira, 201, 14.404-600 Franca, São Paulo, Brazil
| | | | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sérgio Ricardo Ambrósio
- University of Franca, Av. Dr. Armando Salles Oliveira, 201, 14.404-600 Franca, São Paulo, Brazil
| | - Denise Crispim Tavares
- University of Franca, Av. Dr. Armando Salles Oliveira, 201, 14.404-600 Franca, São Paulo, Brazil
| |
Collapse
|
24
|
Preeti, Sambhakar S, Malik R, Bhatia S, Al Harrasi A, Rani C, Saharan R, Kumar S, Geeta, Sehrawat R. Nanoemulsion: An Emerging Novel Technology for Improving the Bioavailability of Drugs. SCIENTIFICA 2023; 2023:6640103. [PMID: 37928749 PMCID: PMC10625491 DOI: 10.1155/2023/6640103] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/02/2023] [Accepted: 10/13/2023] [Indexed: 11/07/2023]
Abstract
The pharmaceutical sector has made considerable strides recently, emphasizing improving drug delivery methods to increase the bioavailability of various drugs. When used as a medication delivery method, nanoemulsions have multiple benefits. Their small droplet size, which is generally between 20 and 200 nanometers, creates a significant interfacial area for drug dissolution, improving the solubility and bioavailability of drugs that are weakly water-soluble. Additionally, nanoemulsions are a flexible platform for drug administration across various therapeutic areas since they can encapsulate hydrophilic and hydrophobic medicines. Nanoemulsion can be formulated in multiple dosage forms, for example, gels, creams, foams, aerosols, and sprays by using low-cost standard operative processes and also be taken orally, topically, topically, intravenously, intrapulmonary, intranasally, and intraocularly. The article explores nanoemulsion formulation and production methods, emphasizing the role of surfactants and cosurfactants in creating stable formulations. In order to customize nanoemulsions to particular medication delivery requirements, the choice of components and production techniques is crucial in assuring the stability and efficacy of the finished product. Nanoemulsions are a cutting-edge technology with a lot of potential for improving medication bioavailability in a variety of therapeutic contexts. They are a useful tool in the creation of innovative pharmaceutical formulations due to their capacity to enhance drug solubility, stability, and delivery. Nanoemulsions are positioned to play a crucial role in boosting medication delivery and enhancing patient outcomes as this field of study continues to advance.
Collapse
Affiliation(s)
- Preeti
- Banasthali Vidyapith, Vanasthali Road, Aliyabad, Rajasthan 304022, India
- Gurugram Global College of Pharmacy, Haily Mandi Rd, Farukh Nagar, Haryana 122506, India
| | - Sharda Sambhakar
- Banasthali Vidyapith, Vanasthali Road, Aliyabad, Rajasthan 304022, India
| | - Rohit Malik
- Gurugram Global College of Pharmacy, Haily Mandi Rd, Farukh Nagar, Haryana 122506, India
| | - Saurabh Bhatia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Oman
| | - Ahmed Al Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Oman
| | - Chanchal Rani
- Gurugram Global College of Pharmacy, Haily Mandi Rd, Farukh Nagar, Haryana 122506, India
| | - Renu Saharan
- Banasthali Vidyapith, Vanasthali Road, Aliyabad, Rajasthan 304022, India
- Maharishi Markandeswar Deemed to be University, Mullana, Ambala, Haryana 133203, India
| | - Suresh Kumar
- Ganpati Institute of Pharmacy, Yamunanagar, Haryana 135102, India
| | - Geeta
- Banasthali Vidyapith, Vanasthali Road, Aliyabad, Rajasthan 304022, India
| | - Renu Sehrawat
- School of Medical & Allied Sciences, K R Mangalam University, Gurugram, Haryana 122103, India
| |
Collapse
|
25
|
Mattioli R, Ilari A, Colotti B, Mosca L, Fazi F, Colotti G. Doxorubicin and other anthracyclines in cancers: Activity, chemoresistance and its overcoming. Mol Aspects Med 2023; 93:101205. [PMID: 37515939 DOI: 10.1016/j.mam.2023.101205] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/31/2023]
Abstract
Anthracyclines have been important and effective treatments against a number of cancers since their discovery. However, their use in therapy has been complicated by severe side effects and toxicity that occur during or after treatment, including cardiotoxicity. The mode of action of anthracyclines is complex, with several mechanisms proposed. It is possible that their high toxicity is due to the large set of processes involved in anthracycline action. The development of resistance is a major barrier to successful treatment when using anthracyclines. This resistance is based on a series of mechanisms that have been studied and addressed in recent years. This work provides an overview of the anthracyclines used in cancer therapy. It discusses their mechanisms of activity, toxicity, and chemoresistance, as well as the approaches used to improve their activity, decrease their toxicity, and overcome resistance.
Collapse
Affiliation(s)
- Roberto Mattioli
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy
| | - Beatrice Colotti
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Luciana Mosca
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy.
| |
Collapse
|
26
|
Yin L, Zhang K, Sun W, Zhang Y, Wang Y, Qin J. Carboxymethylcellulose based self-healing hydrogel with coupled DOX as Camptothecin loading carrier for synergetic colon cancer treatment. Int J Biol Macromol 2023; 249:126012. [PMID: 37517758 DOI: 10.1016/j.ijbiomac.2023.126012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/13/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023]
Abstract
The self-healing hydrogels have important applications in biomedication as drug release carrier. In this research, the Doxorubicin (DOX) was coupled onto oxidized carboxymethylcellulose (CMC) (CMC-Ald) to fabricate self-healing hydrogel with intrinsic antitumor property and loaded with Camptothecin (CPT) for synergetic antitumor treatment. The DOX coupled CMC-Ald (CMC-AD) was reacted with poly(aspartic hydrazide) (PAH) to fabricate injectable self-healing hydrogel. The coupled DOX avoided the burst release of the drug and the 100 % CPT loaded hydrogel could take the advantages of both drugs to enhance the synergetic antitumor therapeutic effect. The in vitro and in vivo results revealed the CPT loaded CMC-AD/PAH hydrogel showed enhanced antitumor property and reduced biotoxicity of the drugs. These properties demonstrate that the CMC-AD/PAH hydrogel has great application prospects in biomedication.
Collapse
Affiliation(s)
- Liping Yin
- College of Chemistry and Materials Science, Hebei University, Baoding City, Hebei Province 071002, China
| | - Kaiyue Zhang
- College of Chemistry and Materials Science, Hebei University, Baoding City, Hebei Province 071002, China
| | - Weichen Sun
- College of Chemistry and Materials Science, Hebei University, Baoding City, Hebei Province 071002, China
| | - Yu Zhang
- College of Chemistry and Materials Science, Hebei University, Baoding City, Hebei Province 071002, China
| | - Yong Wang
- Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-Autoimmune Diseases in Hebei Province, Hebei University, Baoding City, Hebei Province 071002, China
| | - Jianglei Qin
- College of Chemistry and Materials Science, Hebei University, Baoding City, Hebei Province 071002, China; Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-Autoimmune Diseases in Hebei Province, Hebei University, Baoding City, Hebei Province 071002, China.
| |
Collapse
|
27
|
Xu Y, Zhou A, Chen W, Ning X. Scaffold-Free Multicellular 3D Tissue Constructs Utilizing Bio-orthogonal Click Strategy. NANO LETTERS 2023; 23:8770-8778. [PMID: 37694972 DOI: 10.1021/acs.nanolett.3c02889] [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: 09/12/2023]
Abstract
Multicellular 3D tissue constructs (MTCs) are important in biomedical research due to their capacity to accurately mimic the structure and variation found in real tissues. This study presents a novel bio-orthogonal engineering strategy (BIEN), a transformative scaffold-free approach, to create advanced MTCs. BIEN harnesses the cellular biosynthetic machinery to incorporate bio-orthogonal azide reporters into cell surface glycoconjugates, followed by a click reaction with multiarm PEG, resulting in rapid assembly of MTCs. The implementation of this cutting-edge strategy culminates in the formation of uniform, heterogeneous spheroids, characterized by a high degree of intercellular junction and pluripotency. Remarkably, MTCs simulate tumor features, ensure cell heterogeneity, and significantly improve the subcutaneous xenograft model after transplantation, thereby bolstering both in vitro and in vivo research models. In conclusion, the utilization of the bio-orthogonal engineering strategy as a scaffold-free method to generate superior MTCs holds promising potential for driving advancements in cancer research.
Collapse
Affiliation(s)
- Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, People's Republic of China
| | - Anwei Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, School of Physics, Nanjing University, Nanjing 210093, People's Republic of China
| | - Weiwei Chen
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, People's Republic of China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, People's Republic of China
| |
Collapse
|
28
|
Cheng Y, Qu Z, Jiang Q, Xu T, Zheng H, Ye P, He M, Tong Y, Ma Y, Bao A. Functional Materials for Subcellular Targeting Strategies in Cancer Therapy: Progress and Prospects. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2305095. [PMID: 37665594 DOI: 10.1002/adma.202305095] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/26/2023] [Indexed: 09/05/2023]
Abstract
Neoadjuvant and adjuvant therapies have made significant progress in cancer treatment. However, tumor adjuvant therapy still faces challenges due to the intrinsic heterogeneity of cancer, genomic instability, and the formation of an immunosuppressive tumor microenvironment. Functional materials possess unique biological properties such as long circulation times, tumor-specific targeting, and immunomodulation. The combination of functional materials with natural substances and nanotechnology has led to the development of smart biomaterials with multiple functions, high biocompatibilities, and negligible immunogenicities, which can be used for precise cancer treatment. Recently, subcellular structure-targeting functional materials have received particular attention in various biomedical applications including the diagnosis, sensing, and imaging of tumors and drug delivery. Subcellular organelle-targeting materials can precisely accumulate therapeutic agents in organelles, considerably reduce the threshold dosages of therapeutic agents, and minimize drug-related side effects. This review provides a systematic and comprehensive overview of the research progress in subcellular organelle-targeted cancer therapy based on functional nanomaterials. Moreover, it explains the challenges and prospects of subcellular organelle-targeting functional materials in precision oncology. The review will serve as an excellent cutting-edge guide for researchers in the field of subcellular organelle-targeted cancer therapy.
Collapse
Affiliation(s)
- Yanxiang Cheng
- Department of Gynecology, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Zhen Qu
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Qian Jiang
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Tingting Xu
- Department of Clinical Laboratory, Wuhan Blood Center (WHBC), No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Hongyun Zheng
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Peng Ye
- Department of Pharmacy, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Mingdi He
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Yongqing Tong
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Yan Ma
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Anyu Bao
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| |
Collapse
|
29
|
Zhong X, Wei G, Liu B, Wang C, Wang J, Lu Y, Cui W, Guo H. Polyhedral Oligomeric Silsesquioxane-Based Nanoparticles for Efficient Chemotherapy of Glioblastoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207248. [PMID: 36725316 DOI: 10.1002/smll.202207248] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/10/2023] [Indexed: 05/04/2023]
Abstract
Glioblastoma (GBM) is the most common lethal brain tumor with dismal treatment outcomes and poor response to chemotherapy. As the regulatory center of cytogenetics and metabolism, most tumor chemotherapeutic molecules exert therapeutic effects in the nucleus. Nanodrugs showing the nuclear aggregation effect are expected to eliminate and fundamentally suppress tumor cells. In this study, a nanodrug delivery system based on polyhedral oligomeric silsesquioxane (POSS) is introduced to deliver drugs into the nuclei of GBM cells, effectively enhancing the therapeutic efficacy of chemotherapy. The nanoparticles are modified with folic acid and iRGD peptides molecules to improve their tumor cell targeting and uptake via receptor-mediated endocytosis. Nuclear aggregation allows for the direct delivery of chemotherapeutic drug temozolomide (TMZ) to the tumor cell nuclei, resulting in more significant DNA damage and inhibition of tumor cell proliferation. Herein, TMZ-loaded POSS nanoparticles can significantly improve the survival of GBM-bearing mice. Therefore, the modified POSS nanoparticles may serve as a promising drug-loaded delivery platform to improve chemotherapy outcomes in GBM patients.
Collapse
Affiliation(s)
- Xiangyang Zhong
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Gang Wei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Boyang Liu
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Chenyang Wang
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Juan Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Yong Lu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Hongbo Guo
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
30
|
Yang Z, Li H, Zhang W, Zhang M, He J, Yu Z, Sun X, Ni P. CD163 Monoclonal Antibody Modified Polymer Prodrug Nanoparticles for Targeting Tumor-Associated Macrophages (TAMs) to Enhance Anti-Tumor Effects. Pharmaceutics 2023; 15:pharmaceutics15041241. [PMID: 37111726 PMCID: PMC10144748 DOI: 10.3390/pharmaceutics15041241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/31/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Tumor-associated macrophages (TAMs)-based immunotherapy is a promising strategy. Since TAMs are mainly composed of M2-type macrophages, they have a promoting effect on tumor growth, invasion, and metastasis. M2-type macrophages contain a specific receptor CD163 on their surface, providing a prerequisite for active targeting to TAMs. In this study, we prepared CD163 monoclonal antibody modified doxorubicin-polymer prodrug nanoparticles (abbreviated as mAb-CD163-PDNPs) with pH responsiveness and targeted delivery. First, DOX was bonded with the aldehyde group of a copolymer by Schiff base reaction to form an amphiphilic polymer prodrug, which could self-assemble into nanoparticles in the aqueous solution. Then, mAb-CD163-PDNPs were generated through a "Click" reaction between the azide group on the surface of the prodrug nanoparticles and dibenzocyclocytyl-coupled CD163 monoclonal antibody (mAb-CD163-DBCO). The structure and assembly morphology of the prodrug and nanoparticles were characterized by 1H NMR, MALDI-TOF MS, FT-IR UV-vis spectroscopy, and dynamic light scattering (DLS). In vitro drug release behavior, cytotoxicity, and cell uptake were also investigated. The results show that the prodrug nanoparticles have regular morphology and stable structure, especially mAb-CD163-PDNPs, which can actively target TAMs at tumor sites, respond to the acidic environment in tumor cells, and release drugs. While depleting TAMs, mAb-CD163-PDNPs can actively enrich drugs at the tumor site and have a strong inhibitory effect on TAMs and tumor cells. The result of the in vivo test also shows a good therapeutic effect, with a tumor inhibition rate of 81%. This strategy of delivering anticancer drugs in TAMs provides a new way to develop targeted drugs for immunotherapy of malignant tumors.
Collapse
Affiliation(s)
- Zun Yang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Haijiao Li
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Wenrui Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Mingzu Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Jinlin He
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Zepeng Yu
- Center for Medical Ultrasound, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215001, China
| | - Xingwei Sun
- Intervention Department, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Peihong Ni
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| |
Collapse
|
31
|
Maneechote C, Chattipakorn SC, Chattipakorn N. Recent Advances in Mitochondrial Fission/Fusion-Targeted Therapy in Doxorubicin-Induced Cardiotoxicity. Pharmaceutics 2023; 15:pharmaceutics15041182. [PMID: 37111670 PMCID: PMC10143663 DOI: 10.3390/pharmaceutics15041182] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/09/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Doxorubicin (DOX) has been recognized as one of the most effective chemotherapies and extensively used in the clinical settings of human cancer. However, DOX-mediated cardiotoxicity is known to compromise the clinical effectiveness of chemotherapy, resulting in cardiomyopathy and heart failure. Recently, accumulation of dysfunctional mitochondria via alteration of the mitochondrial fission/fusion dynamic processes has been identified as a potential mechanism underlying DOX cardiotoxicity. DOX-induced excessive fission in conjunction with impaired fusion could severely promote mitochondrial fragmentation and cardiomyocyte death, while modulation of mitochondrial dynamic proteins using either fission inhibitors (e.g., Mdivi-1) or fusion promoters (e.g., M1) can provide cardioprotection against DOX-induced cardiotoxicity. In this review, we focus particularly on the roles of mitochondrial dynamic pathways and the current advanced therapies in mitochondrial dynamics-targeted anti-cardiotoxicity of DOX. This review summarizes all the novel insights into the development of anti-cardiotoxic effects of DOX via the targeting of mitochondrial dynamic pathways, thereby encouraging and guiding future clinical investigations to focus on the potential application of mitochondrial dynamic modulators in the setting of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
32
|
Pimenta BV, Madrid RRM, Mathews PD, Riske KA, Loh W, Angelov B, Angelova A, Mertins O. Interaction of polyelectrolyte-shell cubosomes with serum albumin for triggering drug release in gastrointestinal cancer. J Mater Chem B 2023; 11:2490-2503. [PMID: 36852541 DOI: 10.1039/d2tb02670h] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Nano-structured and functionalized materials for encapsulation, transport, targeting and controlled release of drugs are of high interest to overcome low bioavailability in oral administration. We develop lipid-based cubosomes, which are surface-functionalized with biocompatible chitosan-N-arginine and alginate, displaying internal liquid crystalline structures. Polyelectrolyte-shell (PS) cubosomes have pH-responsive characteristics profitable for oral delivery. The obtained PScubosomes can strongly interact with serum albumin, a protein which is released in the stomach under gastric cancer conditions. An effective thermodynamic PScubosome-protein interaction was characterized at pH 2.0 and 7.4 by isothermal titration calorimetry at 37 °C. A high increment of the albumin conformation transition temperature was evidenced by differential scanning calorimetry upon incubation with PScubosomes. The performed structural studies by synchrotron small-angle X-ray scattering (SAXS) revealed essential alterations in the internal liquid crystalline topology of the nanocarriers including an Im3m to Pn3m transition and a reduction of the cubic lattice parameters. The PScubosome nanoparticle interaction with serum albumin, leading to inner structural changes in a range of temperatures, promoted the release of water from the cubosomal nanochannels. Altogether, the results revealed effective interactions of the PScubosomes with albumin under simulated gastrointestinal pH conditions and suggested promising nanocarrier characteristics for triggered oral drug release.
Collapse
Affiliation(s)
- Barbara V Pimenta
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Rafael R M Madrid
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Patrick D Mathews
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Karin A Riske
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Watson Loh
- Institute of Chemistry, State University of Campinas (UNICAMP), 13083-970 Campinas, Brazil
| | - Borislav Angelov
- Institute of Physics, ELI Beamlines, Academy of Sciences of the Czech Republic, CZ-18221 Prague, Czech Republic
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France.
| | - Omar Mertins
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil. .,Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France.
| |
Collapse
|
33
|
Micellar Form of a Ferrocene-Containing Camphor Sulfonamide with Improved Aqueous Solubility and Tumor Curing Potential. Pharmaceutics 2023; 15:pharmaceutics15030791. [PMID: 36986651 PMCID: PMC10054005 DOI: 10.3390/pharmaceutics15030791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The discovery of new anticancer drugs with а higher, more specific activity and diminished side effects than the conventional chemotherapeutic agents is a tremendous challenge to contemporary medical research and development. To achieve a pronounced efficacy, the design of antitumor agents can combine various biologically active subunits in one molecule, which can affect different regulatory pathways in cancer cells. We recently demonstrated that a newly synthesized organometallic compound, a ferrocene-containing camphor sulfonamide (DK164), possesses promising antiproliferative activity against breast and lung cancer cells. However, it still encounters the problem of solubility in biological fluids. In this work, we describe a novel micellar form of DK164 with significantly improved solubility in aqueous medium. DK164 was embedded in biodegradable micelles based on a poly(ethylene oxide)-b-poly(α-cinnamyl-ε-caprolactone-co-ε-caprolactone)-b-poly(ethylene oxide) triblock copolymer (PEO113-b-P(CyCL3-co-CL46)-b-PEO113), and the physicochemical parameters (size, size distribution, zeta potential, encapsulation efficiency) and biological activity of the obtained system were studied. We used cytotoxicity assays and flow cytometry to determine the type of cell death, as well as immunocytochemistry to assess the influence of the encapsulated drug on the dynamics of cellular key proteins (p53 and NFkB) and the process of autophagy. According to our results, the micellar form of the organometallic ferrocene derivate (DK164-NP) exhibited several advantages compared to the free substance, such as higher metabolic stability, better cellular uptake, improved bioavailability, and long-term activity, maintaining nearly the same biological activity and anticancer properties of the drug.
Collapse
|
34
|
Stavroulaki D, Kyroglou I, Skourtis D, Athanasiou V, Thimi P, Sofianopoulou S, Kazaryan D, Fragouli PG, Labrianidou A, Dimas K, Patias G, Haddleton DM, Iatrou H. Influence of the Topology of Poly(L-Cysteine) on the Self-Assembly, Encapsulation and Release Profile of Doxorubicin on Dual-Responsive Hybrid Polypeptides. Pharmaceutics 2023; 15:790. [PMID: 36986652 PMCID: PMC10055909 DOI: 10.3390/pharmaceutics15030790] [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: 01/20/2023] [Revised: 02/16/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Τhe synthesis of a series of novel hybrid block copolypeptides based on poly(ethylene oxide) (PEO), poly(l-histidine) (PHis) and poly(l-cysteine) (PCys) is presented. The synthesis of the terpolymers was achieved through a ring-opening polymerization (ROP) of the corresponding protected N-carboxy anhydrides of Nim-Trityl-l-histidine and S-tert-butyl-l-cysteine, using an end-amine-functionalized poly(ethylene oxide) (mPEO-NH2) as macroinitiator, followed by the deprotection of the polypeptidic blocks. The topology of PCys was either the middle block, the end block or was randomly distributed along the PHis chain. These amphiphilic hybrid copolypeptides assemble in aqueous media to form micellar structures, comprised of an outer hydrophilic corona of PEO chains, and a pH- and redox-responsive hydrophobic layer based on PHis and PCys. Due to the presence of the thiol groups of PCys, a crosslinking process was achieved further stabilizing the nanoparticles (NPs) formed. Dynamic light scattering (DLS), static light scattering (SLS) and transmission electron microscopy (TEM) were utilized to obtain the structure of the NPs. Moreover, the pH and redox responsiveness in the presence of the reductive tripeptide of glutathione (GSH) was investigated at the empty as well as the loaded NPs. The ability of the synthesized polymers to mimic natural proteins was examined by Circular Dichroism (CD), while the study of zeta potential revealed the "stealth" properties of NPs. The anticancer drug doxorubicin (DOX) was efficiently encapsulated in the hydrophobic core of the nanostructures and released under pH and redox conditions that simulate the healthy and cancer tissue environment. It was found that the topology of PCys significantly altered the structure as well as the release profile of the NPs. Finally, in vitro cytotoxicity assay of the DOX-loaded NPs against three different breast cancer cell lines showed that the nanocarriers exhibited similar or slightly better activity as compared to the free drug, rendering these novel NPs very promising materials for drug delivery applications.
Collapse
Affiliation(s)
- Dimitra Stavroulaki
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Iro Kyroglou
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Dimitrios Skourtis
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Varvara Athanasiou
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Pandora Thimi
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Sosanna Sofianopoulou
- Hellenic Police Headquarters, Forensic Science Division, Chemical and Physical Examinations Department, GR-10442 Athens, Greece
| | - Diana Kazaryan
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Panagiota G. Fragouli
- DIDPE, Dyeing, Finishing, Dyestuffs and Advanced Polymers Laboratory, University of West Attica, 250 Thevon Street, GR-12241 Athens, Greece
| | - Andromahi Labrianidou
- Laboratory of Pharmacology, Faculty of Medicine, University of Thessaly, Viopolis, GR-41500 Larissa, Greece
| | - Konstantinos Dimas
- Laboratory of Pharmacology, Faculty of Medicine, University of Thessaly, Viopolis, GR-41500 Larissa, Greece
| | - Georgios Patias
- Department of Chemistry, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK
| | - David M. Haddleton
- Department of Chemistry, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK
| | - Hermis Iatrou
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| |
Collapse
|
35
|
Khabibullin VR, Chetyrkina MR, Obydennyy SI, Maksimov SV, Stepanov GV, Shtykov SN. Study on Doxorubicin Loading on Differently Functionalized Iron Oxide Nanoparticles: Implications for Controlled Drug-Delivery Application. Int J Mol Sci 2023; 24:4480. [PMID: 36901910 PMCID: PMC10002596 DOI: 10.3390/ijms24054480] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Nanoplatforms applied for the loading of anticancer drugs is a cutting-edge approach for drug delivery to tumors and reduction of toxic effects on healthy cells. In this study, we describe the synthesis and compare the sorption properties of four types of potential doxorubicin-carriers, in which iron oxide nanoparticles (IONs) are functionalized with cationic (polyethylenimine, PEI), anionic (polystyrenesulfonate, PSS), and nonionic (dextran) polymers, as well as with porous carbon. The IONs are thoroughly characterized by X-ray diffraction, IR spectroscopy, high resolution TEM (HRTEM), SEM, magnetic susceptibility, and the zeta-potential measurements in the pH range of 3-10. The degree of doxorubicin loading at pH 7.4, as well as the degree of desorption at pH 5.0, distinctive to cancerous tumor environment, are measured. Particles modified with PEI were shown to exhibit the highest loading capacity, while the greatest release at pH 5 (up to 30%) occurs from the surface of magnetite decorated with PSS. Such a slow release of the drug would imply a prolonged tumor-inhibiting action on the affected tissue or organ. Assessment of the toxicity (using Neuro2A cell line) for PEI- and PSS-modified IONs showed no negative effect. In conclusion, the preliminary evaluation of the effects of IONs coated with PSS and PEI on the rate of blood clotting was carried out. The results obtained can be taken into account when developing new drug delivery platforms.
Collapse
Affiliation(s)
- Vladislav R. Khabibullin
- Chemistry Department, Lomonosov Moscow State University, Lenin Hills, 119991 Moscow, Russia
- State Scientific Center of the Russian Federation, Joint Stock Company “State Order of the Red Banner of Labor Research Institute of Chemistry and Technology of Organoelement Compounds”, 105118 Moscow, Russia
| | | | - Sergei I. Obydennyy
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117198 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, 119334 Moscow, Russia
| | - Sergey V. Maksimov
- Chemistry Department, Lomonosov Moscow State University, Lenin Hills, 119991 Moscow, Russia
| | - Gennady V. Stepanov
- State Scientific Center of the Russian Federation, Joint Stock Company “State Order of the Red Banner of Labor Research Institute of Chemistry and Technology of Organoelement Compounds”, 105118 Moscow, Russia
| | - Sergei N. Shtykov
- Department of Analytical Chemistry and Chemical Ecology, Institute of Chemistry, Saratov State University, 410012 Saratov, Russia
| |
Collapse
|
36
|
Bognanni N, Viale M, La Piana L, Strano S, Gangemi R, Lombardo C, Cambria MT, Vecchio G. Hyaluronan-Cyclodextrin Conjugates as Doxorubicin Delivery Systems. Pharmaceutics 2023; 15:374. [PMID: 36839696 PMCID: PMC9963997 DOI: 10.3390/pharmaceutics15020374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
In the last years, nanoparticles based on cyclodextrins have been widely investigated for the delivery of anticancer drugs. In this work, we synthesized nanoparticles with a hyaluronic acid backbone functionalized with cyclodextrins under green conditions. We functionalized hyaluronic acid with two different molecular weights (about 11 kDa and 45 kDa) to compare their behavior as doxorubicin delivery systems. We found that the new hyaluronan-cyclodextrin conjugates increased the water solubility of doxorubicin. Moreover, we tested the antiproliferative activity of doxorubicin in the presence of the new cyclodextrin polymers in SK-N-SH and SK-N-SH-PMA (over-expressing CD44 receptor) cancer cells. We found that hyaluronan-cyclodextrin conjugates improved the uptake and antiproliferative activity of doxorubicin in the SK-N-SH-PMA compared to the SK-N-SH cell line at the ratio 8/1 doxorubicin/polymer. Notably, the system based on hyaluronan (45 kDa) was more effective as a drug carrier and significantly reduced the IC50 value of doxorubicin by about 56%. We also found that hyaluronic acid polymers determined an improved antiproliferative activity of doxorubicin (IC50 values are on average reduced by about 70% of free DOXO) in both cell lines at the ratio 16/1 doxorubicin/polymer.
Collapse
Affiliation(s)
- Noemi Bognanni
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Maurizio Viale
- UOC Bioterapie, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Luana La Piana
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Simone Strano
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Rosaria Gangemi
- UOC Bioterapie, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Cinzia Lombardo
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biochimica Medica, Università degli Studi di Catania, Via S. Sofia 97, 95125 Catania, Italy
| | - Maria Teresa Cambria
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biochimica Medica, Università degli Studi di Catania, Via S. Sofia 97, 95125 Catania, Italy
| | - Graziella Vecchio
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
37
|
Zhai J, Gu X, Liu Y, Hu Y, Jiang Y, Zhang Z. Chemotherapeutic and targeted drugs-induced immunogenic cell death in cancer models and antitumor therapy: An update review. Front Pharmacol 2023; 14:1152934. [PMID: 37153795 PMCID: PMC10160433 DOI: 10.3389/fphar.2023.1152934] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/04/2023] [Indexed: 05/10/2023] Open
Abstract
As traditional strategies for cancer treatment, some chemotherapy agents, such as doxorubicin, oxaliplatin, cyclophosphamide, bortezomib, and paclitaxel exert their anti-tumor effects by inducing immunogenic cell death (ICD) of tumor cells. ICD induces anti-tumor immunity through release of, or exposure to, damage-related molecular patterns (DAMPs), including high mobility group box 1 (HMGB1), calreticulin, adenosine triphosphate, and heat shock proteins. This leads to activation of tumor-specific immune responses, which can act in combination with the direct killing functions of chemotherapy drugs on cancer cells to further improve their curative effects. In this review, we highlight the molecular mechanisms underlying ICD, including those of several chemotherapeutic drugs in inducing DAMPs exposed during ICD to activate the immune system, as well as discussing the prospects for application and potential role of ICD in cancer immunotherapy, with the aim of providing valuable inspiration for future development of chemoimmunotherapy.
Collapse
|
38
|
Girigoswami A, Adhikesavan H, Mudenkattil S, Devi S, Girigoswami K. Role of Cerium Oxide Nanoparticles and Doxorubicin in Improving Cancer Management: A Mini Review. Curr Pharm Des 2023; 29:2640-2654. [PMID: 37957864 DOI: 10.2174/0113816128270290231029161741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 11/15/2023]
Abstract
Cancer is one of the significant issues with public health and the second leading cause of death worldwide. The three most lethal cancers in the general population are stomach, lung, and liver cancers, in which lung and breast cancers cause the majority of cancer-associated deaths among men and women, respectively. CeO2 nanoparticles have a cytoprotectant effect in normal cells and a cytotoxic effect in cancer cells that enables them to induce the reactive oxygen species (ROS) production within cancer cells, which in turn develops reactive nitrogen species (RNS) that interfere with intracellular activities, and this property makes them an excellent anticancer agent. Because of its biofilm suppression, free radical scavenging ability, redox activity, and other unique properties, attention has been bestowed on cerium oxide nanoparticles as a potential alternative to solve many biomedical issues in the future. This review mainly focuses on the combinatorial effect of cerium dioxide nanoparticles and Doxorubicin in cancer management.
Collapse
Affiliation(s)
- Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, 603103, Tamilnadu, India
| | - Harini Adhikesavan
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, 603103, Tamilnadu, India
| | - Shurfa Mudenkattil
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, 603103, Tamilnadu, India
| | - Sobita Devi
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, 603103, Tamilnadu, India
| | - Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, 603103, Tamilnadu, India
| |
Collapse
|
39
|
De Jong WH, Geertsma RE, Borchard G. Regulatory safety evaluation of nanomedical products: key issues to refine. Drug Deliv Transl Res 2022; 12:2042-2047. [PMID: 35908133 PMCID: PMC9358921 DOI: 10.1007/s13346-022-01208-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2022] [Indexed: 12/17/2022]
Abstract
Nanotechnologies enable great opportunities for the development and use of innovative (nano)medicines. As is common for scientific and technical developments, recognized safety evaluation methods for regulatory purposes are lagging behind. The specific properties responsible for the desired functioning also hamper the safety evaluation of such products. Pharmacokinetics determination of the active pharmaceutical ingredient as well as the nanomaterial component is crucial. Due to their particulate nature, nanomedicines, similar to all nanomaterials, are primarily removed from the circulation by phagocytizing cells that are part of the immune system. Therefore, the immune system can be potentially a specific target for adverse effects of nanomedicines, and thus needs special attention during the safety evaluation. This DDTR special issue on the results of the REFINE project on a regulatory science framework for nanomedical products presents a highly valuable body of knowledge needed to address regulatory challenges and gaps in currently available testing methods for the safety evaluation of nanomedicines.
Collapse
Affiliation(s)
- Wim H De Jong
- Formerly (retired) National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Robert E Geertsma
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | |
Collapse
|