1
|
Iorio AL, Lenci E, Marzano C, Bucaletti E, Tirinnanzi B, Casati G, Giunti L, Dallari C, Credi C, Sardi I, Trabocchi A. Oxime Linked Doxorubicin Glycoconjugates Improve the Specific Targeting of Glioblastoma in High-Grade Glioma Therapy. ACS Med Chem Lett 2024; 15:1953-1960. [PMID: 39563793 PMCID: PMC11571026 DOI: 10.1021/acsmedchemlett.4c00398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 11/21/2024] Open
Abstract
The treatment of glioblastoma (GBM) represents an urgent challenge for public health due to the inability to effectively deliver anticancer agents, such as doxorubicin (DOX), through the blood-brain barrier (BBB). Herein we report the synthesis of two novel DOX glycoconjugates using an oxime linkage that maintained the intercalation capability of the planar anthracycline ring of DOX, as demonstrated by UV-vis and fluorescence experiments in the presence of DNA. The biological effect of DOX glycoconjugates was evaluated in GBM cell lines, showing an enhanced cytotoxic and pro-apoptotic effect of 7 as compared to 4 and to conventional DOX. These data were confirmed in an in vitro coculture BBB model in which DOX glycoconjugate 7 showed high capability to cross a cellular monolayer and exert its cytotoxic effect on GBM cells. The results show that conjugation with glucose may represent a helpful tool to increase chemotherapy effectiveness in poor-responding GBM patients.
Collapse
Affiliation(s)
- Anna Lisa Iorio
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, 50139 Florence, Italy
| | - Elena Lenci
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 13, 50019 Sesto Fiorentino, Florence, Italy
| | - Chiara Marzano
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Elisabetta Bucaletti
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 13, 50019 Sesto Fiorentino, Florence, Italy
| | - Bianca Tirinnanzi
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Giacomo Casati
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Laura Giunti
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Caterina Dallari
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, 50019 Sesto Fiorentino, Florence, Italy
- National Institute of Optics National Research Council, 50019 Sesto Fiorentino, Florence, Italy
| | - Caterina Credi
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, 50019 Sesto Fiorentino, Florence, Italy
- National Institute of Optics National Research Council, 50019 Sesto Fiorentino, Florence, Italy
| | - Iacopo Sardi
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Andrea Trabocchi
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 13, 50019 Sesto Fiorentino, Florence, Italy
| |
Collapse
|
2
|
Fang X, Zeng J, Li Y, Yu H, Wu Z, Qi X. Hydroxychloroquine loaded hollow apoferritin nanocages for cancer drug repurposing and autophagy inhibition. Eur J Pharm Biopharm 2024; 203:114473. [PMID: 39186959 DOI: 10.1016/j.ejpb.2024.114473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/31/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
Hydroxychloroquine sulfate (HCQ) is currently being repurposed for cancer treatment. The antitumor mechanism of HCQ is inhibition of cellular autophagy, but its therapeutic potential is severely limited by poor solubility, lack of tumor targeting and lower cellular uptake. Therefore, utilization of human H-chain apoferritin (HFn) composed only of heavy subunits is an attractive approach for tumor targeting drug delivery. This study focused on pH-triggered encapsulation of HCQ within the inner cavity of HFn to form HFn@HCQ nanoparticles for tumor-targeted drug delivery. Characterization using a range of techniques has been used to confirm the successful establishment of HFn@HCQ. HFn@HCQ exhibited pH-responsive release behavior, with almost no drug release at pH 7.4, but 80% release at pH 5.0. Owing to its intrinsic binding to transferrin receptor 1 (TfR1), HFn@HCQ was significantly internalized through TfR1-mediated endocytosis, with a 4.4-fold difference of internalization amount across cell lines. Additionally, HFn@HCQ enhanced the antitumor effect against four different cancer cell lines when compared against HCQ alone, especially in TfR1 high-expressing cells, where the inhibitory effect was 3-fold higher than free HCQ. The autophagy inhibition of HFn@HCQ has been demonstrated, which is a major pathway to induce cancer cell death. According to current findings, HFn based drug delivery is a promising strategy to target and kill TfR1 overexpressing tumor cells.
Collapse
Affiliation(s)
- Xinning Fang
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jia Zeng
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yitong Li
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Han Yu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenghong Wu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; Hangzhou Innovative Institute of Pharmaceutics, China Pharmaceutical University, Hangzhou 310018, China.
| |
Collapse
|
3
|
Singh AK, Ruiz D, Rasheed MSU, Avery TD, Turner DJL, Abell AD, Grace PM. Systemic and targeted activation of Nrf2 reverses doxorubicin-induced cognitive impairments and sensorimotor deficits in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598291. [PMID: 38915544 PMCID: PMC11195070 DOI: 10.1101/2024.06.10.598291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
While cancer survivorship has increased due to advances in treatments, chemotherapy often carries long-lived neurotoxic side effects which reduce quality of life. Commonly affected domains include memory, executive function, attention, processing speed and sensorimotor function, colloquially known as chemotherapy-induced cognitive impairment (CICI) or "chemobrain". Oxidative stress and neuroimmune signaling in the brain have been mechanistically linked to the deleterious effects of chemotherapy on cognition and sensorimotor function. With this in mind, we tested if activation of the master regulator of antioxidant response nuclear factor E2-related factor 2 (Nrf2) alleviates cognitive and sensorimotor impairments induced by doxorubicin. The FDA-approved systemic Nrf2 activator, diroximel fumarate (DRF) was used, along with our recently developed prodrug 1c which has the advantage of specifically releasing monomethyl fumarate at sites of oxidative stress. DRF and 1c both reversed doxorubicin-induced deficits in executive function, spatial and working memory, as well as decrements in fine motor coordination and grip strength, across both male and female mice. Both treatments reversed doxorubicin-induced loss of synaptic proteins and microglia phenotypic transition in the hippocampus. Doxorubicin-induced myelin damage in the corpus callosum was reversed by both Nrf2 activators. These results demonstrate the therapeutic potential of Nrf2 activators to reverse doxorubicin-induced cognitive impairments, motor incoordination, and associated structural and phenotypic changes in the brain. The localized release of monomethyl fumarate by 1c has the potential to diminish unwanted effects of fumarates while retaining efficacy.
Collapse
Affiliation(s)
- Anand Kumar Singh
- Laboratories of Neuroimmunology, Department of Symptom Research, and the MD Anderson Pain Research Consortium, University of Texas MD Anderson Cancer Center, Houston, USA
| | - David Ruiz
- Laboratories of Neuroimmunology, Department of Symptom Research, and the MD Anderson Pain Research Consortium, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Mohd Sami Ur Rasheed
- Laboratories of Neuroimmunology, Department of Symptom Research, and the MD Anderson Pain Research Consortium, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Thomas D Avery
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Institute for Photonics and Advanced Sensing (IPAS), Department of Chemistry, The University of Adelaide, Adelaide, Australia
| | - Dion J L Turner
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Institute for Photonics and Advanced Sensing (IPAS), Department of Chemistry, The University of Adelaide, Adelaide, Australia
| | - Andrew D Abell
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Institute for Photonics and Advanced Sensing (IPAS), Department of Chemistry, The University of Adelaide, Adelaide, Australia
| | - Peter M Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, and the MD Anderson Pain Research Consortium, University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
4
|
Cassioli ML, Fay M, Turyanska L, Bradshaw TD, Thomas NR, Pordea A. Encapsulation of copper phenanthroline within horse spleen apoferritin: characterisation, cytotoxic activity and ability to retain temozolomide. RSC Adv 2024; 14:14008-14016. [PMID: 38686295 PMCID: PMC11056943 DOI: 10.1039/d3ra07430g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/13/2024] [Indexed: 05/02/2024] Open
Abstract
Protein capsules are promising drug delivery vehicles for cancer research therapies. Apoferritin (AFt) is a self-assembling 12 nm diameter hollow nanocage with many desirable features for drug delivery, however, control of drug retention inside the protein cage remains challenging. Here we report the encapsulation of copper(ii)-1,10-phenanthroline (Cu(phen)) within the horse spleen AFt (HSAFt) nanocage, by diffusion of the metal through the pores between the protein subunits. Transmission electron microscopy revealed the formation of organised copper adducts inside HSAFt, without affecting protein integrity. These structures proved stable during storage (>4 months at -20 °C). Exposure to physiologically relevant conditions (37 °C) showed some selectivity in cargo release after 24 h at pH 5.5, relevant to the internalisation of AFt within the endosome (60% release), compared to pH 7.4, relevant to the bloodstream (40% release). Co-encapsulation of temozolomide, a prodrug used to treat glioblastoma multiforme, and Cu(phen) enabled entrapment of an average of 339 TMZ molecules per cage. In vitro results from MTT and clonogenic assays identified cytotoxic activity of the Cu(phen), HSAFt-Cu(phen) and HSAFt-Cu(phen)-TMZ adducts against colorectal cancer cells (HCT-116) and glioblastoma cells (U373V, U373M). However, the presence of the metal also contributed to more potent activity toward healthy MRC5 fibroblasts, a result that requires further investigation to assess the clinical viability of this system.
Collapse
Affiliation(s)
| | - Michael Fay
- Nanoscale and Microscale Research Centre, University of Nottingham NG7 2RD UK
| | | | - Tracey D Bradshaw
- Biodiscovery Institute, School of Pharmacy, University of Nottingham NG7 2RD UK
| | - Neil R Thomas
- Biodiscovery Institute, School of Chemistry, University of Nottingham NG7 2RD UK
| | - Anca Pordea
- Faculty of Engineering, University of Nottingham NG7 2RD UK
| |
Collapse
|
5
|
Oraiopoulou ME, Tzamali E, Psycharakis SE, Tzedakis G, Makatounakis T, Manolitsi K, Drakos E, Vakis AF, Zacharakis G, Papamatheakis J, Sakkalis V. The Temozolomide-Doxorubicin paradox in Glioblastoma in vitro-in silico preclinical drug-screening. Sci Rep 2024; 14:3759. [PMID: 38355655 PMCID: PMC10866941 DOI: 10.1038/s41598-024-53684-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
Adjuvant Temozolomide is considered the front-line Glioblastoma chemotherapeutic treatment; yet not all patients respond. Latest trends in clinical trials usually refer to Doxorubicin; yet it can lead to severe side-effects if administered in high doses. While Glioblastoma prognosis remains poor, little is known about the combination of the two chemotherapeutics. Patient-derived spheroids were generated and treated with a range of Temozolomide/Doxorubicin concentrations either as monotherapy or in combination. Optical microscopy was used to monitor the growth pattern and cell death. Based on the monotherapy experiments, we developed a probabilistic mathematical framework in order to describe the drug-induced effect at the single-cell level and simulate drug doses in combination assuming probabilistic independence. Doxorubicin was found to be effective in doses even four orders of magnitude less than Temozolomide in monotherapy. The combination therapy doses tested in vitro were able to lead to irreversible growth inhibition at doses where monotherapy resulted in relapse. In our simulations, we assumed both drugs are anti-mitotic; Temozolomide has a growth-arrest effect, while Doxorubicin is able to cumulatively cause necrosis. Interestingly, under no mechanistic synergy assumption, the in silico predictions underestimate the in vitro results. In silico models allow the exploration of a variety of potential underlying hypotheses. The simulated-biological discrepancy at certain doses indicates a supra-additive response when both drugs are combined. Our results suggest a Temozolomide-Doxorubicin dual chemotherapeutic scheme to both disable proliferation and increase cytotoxicity against Glioblastoma.
Collapse
Affiliation(s)
- Mariam-Eleni Oraiopoulou
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
- Cancer Research UK - Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Eleftheria Tzamali
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Stylianos E Psycharakis
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
- School of Medicine, University of Crete, Heraklion, Greece
| | - Georgios Tzedakis
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Takis Makatounakis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
| | - Katina Manolitsi
- University General Hospital of Heraklion (PAGNI), Heraklion, Greece
| | - Elias Drakos
- School of Medicine, University of Crete, Heraklion, Greece
- University General Hospital of Heraklion (PAGNI), Heraklion, Greece
| | - Antonis F Vakis
- School of Medicine, University of Crete, Heraklion, Greece
- University General Hospital of Heraklion (PAGNI), Heraklion, Greece
| | - Giannis Zacharakis
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
| | - Joseph Papamatheakis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Vangelis Sakkalis
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece.
| |
Collapse
|
6
|
Xu H, Zhang Y, Li L, Ren Y, Qian F, Wang L, Ma H, Quan A, Liu H, Yu R. The nanoprodrug of polytemozolomide combines with MGMT siRNA to enhance the effect of temozolomide in glioma. Drug Deliv 2023; 30:1-13. [PMID: 36579448 PMCID: PMC9809344 DOI: 10.1080/10717544.2022.2152911] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Temozolomide (TMZ) is a conventional chemotherapeutic drug for glioma, however, its clinical application and efficacy is severely restricted by its drug resistance properties. O6-methylguanine-DNA methyltransferase (MGMT) is a DNA repair enzyme, which can repair the DNA damage caused by TMZ. A large number of clinical data show that reducing the expression of MGMT can enhance the chemotherapeutic efficacy of TMZ. Therefore, in order to improve the resistance of glioma to TMZ, an angiopep-2 (A2) modified nanoprodrug of polytemozolomide (P(TMZ)n) that combines with MGMT siRNA (siMGMT) targeting MGMT was developed (A2/T/D/siMGMT). It not only increased the amount of TMZ within tumor lesion site, but also reduced MGMT expression in glioma. The in vitro experiments indicated that the A2/T/D/siMGMT effectively enhanced the cellular uptake of TMZ and siMGMT, and resulted in a significant cell apoptosis and cytotoxicity in the glioma cells. The in vivo experiments showed that glioma growth was inhibited and the survival time of animals were prolonged remarkably after A2/T/D/siMGMT was injected via tail vein. The results showed that the therapeutic effect of A2/T/D/siMGMT in the treatment of glioma was significantly improved.
Collapse
Affiliation(s)
- Haoyue Xu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yongkang Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Linfeng Li
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yanhong Ren
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Feng Qian
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Lansheng Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Hongwei Ma
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Ankang Quan
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Hongmei Liu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China,Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China,Hongmei Liu Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China,CONTACT Yu Rutong;
| |
Collapse
|
7
|
Zhao F, Wang X, Zhu W, Zhao D, Ye C, Guo Y, Dou Y. Low-dose pleiotropic radiosensitive nanoformulations for three-pronged radiochemotherapy of hypoxic brain glioblastoma under BOLD/DWI monitoring. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00159-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abstract
Background
Hypoxia-mediated radioresistance is the main obstacle to the successful treatment of glioblastoma (GBM). Enhancing hypoxic radiosensitivity and alleviating tumor hypoxia are both effective means to improve therapeutic efficacy, and the combination of the two is highly desirable and meaningful.
Results
Herein, we construct a low-dose pleiotropic radiosensitive nanoformulation consisting of a high-Z atomic nanocrystal core and mesoporous silica shell, surface-modified with angiopep-2 (ANG) peptide and loaded with nitric oxide (NO) donor and hypoxia-activated prodrug (AQ4N). Benefiting from ANG-mediated transcytosis, this nanoformulation can efficiently cross the BBB and accumulate preferentially in the brain. Low-dose radiation triggers this nanoformulation to exert a three-pronged synergistic therapeutic effect through high-Z-atom-dependent dose deposition enhancement, NO-mediated hypoxia relief, and AQ4N-induced hypoxia-selective killing, thereby significantly inhibiting GBM in situ growth while prolonging survival and maintaining stable body weight in the glioma-bearing mice. Meanwhile, the proposed in vivo 9.4 T BOLD/DWI can realize real-time dynamic assessment of local oxygen supply and radiosensitivity to monitor the therapeutic response of GBM.
Conclusions
This work provides a promising alternative for hypoxia-specific GBM-targeted comprehensive therapy, noninvasive monitoring, and precise prognosis.
Graphical Abstract
Collapse
|
8
|
Sandal P, Kumari L, Patel P, Singh A, Singh D, Gupta GD, Kurmi BD. Doxorubicin Conjugates: An Efficient Approach for Enhanced Therapeutic Efficacy with Reduced Side Effects. Assay Drug Dev Technol 2023; 21:137-156. [PMID: 37083490 DOI: 10.1089/adt.2022.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Abstract
Continuous drug delivery modification is the scientific approach and is a basic need for the efficient therapeutic efficacy of active drug molecules. Polymer-drug conjugates have long been a hallmark of the drug delivery sector, with various conjugates on the market or in clinical trials. Improved drug solubilization, extended blood circulation, decreased immunogenicity, controlled release behavior, and increased safety are the advantages of conjugating drugs to the polymeric carrier like polyethylene glycol (PEG). Polymer therapies have evolved over the last decade, resulting in polymer-drug conjugates with diverse topologies and chemical properties. Traditional nondegradable polymeric carriers like PEG and hydroxy propyl methacrylate have been clinically employed to fabricate polymer-drug conjugates. Still, functionalized polymer-drug conjugates are increasingly being used to increase localized drug delivery and ease of removal. Researchers have developed multifunctional carriers that can "see and treat" patients using medicinal and diagnostic chemicals. This review focused on the various conjugation approaches for attaching the doxorubicin to different polymers to achieve enhanced therapeutic efficacy, that is, increased bioavailability and reduced adverse effects.
Collapse
Affiliation(s)
- Pallavi Sandal
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | - Lakshmi Kumari
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Punjab, India
| | - Amrinder Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | - Dilpreet Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | | | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| |
Collapse
|
9
|
Jatyan R, Singh P, Sahel DK, Karthik YG, Mittal A, Chitkara D. Polymeric and small molecule-conjugates of temozolomide as improved therapeutic agents for glioblastoma multiforme. J Control Release 2022; 350:494-513. [PMID: 35985493 DOI: 10.1016/j.jconrel.2022.08.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/31/2022] [Accepted: 08/12/2022] [Indexed: 11/15/2022]
Abstract
Temozolomide (TMZ), an imidazotetrazine, is a second-generation DNA alkylating agent used as a first-line treatment of glioblastoma multiforme (GBM). It was approved by FDA in 2005 and declared a blockbuster drug in 2008. Although TMZ has shown 100% oral bioavailability and crosses the blood-brain barrier effectively, however it suffers from limitations such as a short half-life (∼1.8 h), rapid metabolism, and lesser accumulation in the brain (∼10-20%). Additionally, development of chemoresistance has been associated with its use. Since it is a potential chemotherapeutic agent with an unmet medical need, advanced delivery strategies have been explored to overcome the associated limitations of TMZ. Nanocarriers including liposomes, solid lipid nanoparticles (SLNs), nanostructure lipid carriers (NLCs), and polymeric nanoparticles have demonstrated their ability to improve its circulation time, stability, tissue-specific accumulation, sustained release, and cellular uptake. Because of the appreciable water solubility of TMZ (∼5 mg/mL), the physical loading of TMZ in these nanocarriers is always challenging. Alternatively, the conjugation approach, wherein TMZ has been conjugated to polymers or small molecules, has been explored with improved outcomes in vitro and in vivo. This review emphasized the practical evidence of the conjugation strategy to improve the therapeutic potential of TMZ in the treatment of glioblastoma multiforme.
Collapse
Affiliation(s)
- Reena Jatyan
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Prabhjeet Singh
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Deepak Kumar Sahel
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Y G Karthik
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India.
| |
Collapse
|
10
|
Ye Z, Gao L, Cai J, Wang Y, Li Y, Tong S, Yan T, Sun Q, Qi Y, Xu Y, Jiang H, Zhang S, Zhao L, Zhang S, Chen Q. Esterase-responsive and size-optimized prodrug nanoparticles for effective intracranial drug delivery and glioblastoma treatment. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 44:102581. [PMID: 35811067 DOI: 10.1016/j.nano.2022.102581] [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: 02/28/2022] [Revised: 06/03/2022] [Accepted: 06/23/2022] [Indexed: 06/15/2023]
Abstract
Glioblastoma multiforme (GBM) is the intracranial malignancy with the highest rates of morbidity and mortality. Chemotherapy is often ineffective against GBM due to the presence of the blood-brain barrier (BBB); however, the application of nanotechnology is expected to overcome this limitation. Poly(lactic-co-glycolic acid) (PLGA) is a degradable and nontoxic functional polymer with good biocompatibility that is widely used in the pharmaceutical industry. Previous studies have shown that the ability of PLGA nanoparticles (NPs) to penetrate the BBB is largely determined by their size; however, determination of the optimal PLGA NP size requires further research. Here, we report a tandutinib-based prodrug (proTan), which responds to the GBM microenvironment, that was combined with NPs to overcome the BBB. AMD3100-PLGA NPs loaded with proTan inhibited tumor growth and effectively prolonged the survival of tumor-bearing mice.
Collapse
Affiliation(s)
- Zhang Ye
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Lun Gao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jiayang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yixuan Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yong Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Shiao Tong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Tengfeng Yan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Qian Sun
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yangzhi Qi
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yang Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Hongxiang Jiang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Si Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Linyao Zhao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Shenqi Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
11
|
Aragon-Sanabria V, Aditya A, Zhang L, Chen F, Yoo B, Cao T, Madajewski B, Lee R, Turker MZ, Ma K, Monette S, Chen P, Wu J, Ruan S, Overholtzer M, Zanzonico P, Rudin CM, Brennan C, Wiesner U, Zhang L. Ultrasmall Nanoparticle Delivery of Doxorubicin Improves Therapeutic Index for High-Grade Glioma. Clin Cancer Res 2022; 28:2938-2952. [PMID: 35499557 DOI: 10.1158/1078-0432.ccr-21-4053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/11/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: Despite dramatic growth in the number of small molecule drugs developed to treat solid tumors, durable therapeutic options to control primary central nervous system malignancies are relatively scarce. Chemotherapeutic agents which appear biologically potent in model systems have often been found to be marginally effective at best when given systemically in clinical trials. This work presents for the first time an ultrasmall (< 8 nm) multimodal core-shell silica nanoparticle, Cornell prime dots (or C' dots), for the efficacious treatment of high-grade gliomas. Experimental Design: This work presents first-in-kind renally-clearable ultrasmall (< 8 nm) multimodal Cornell prime dots (or C' dots) with surface-conjugated doxorubicin via pH-sensitive linkers for the efficacious treatment in two different clinically relevant high-grade glioma models. Results: Optimal drug-per-particle ratios of as-developed nanoparticle-drug conjugates were established and used to obtain favorable pharmacokinetic profiles. The in vivo efficacy results showed significantly improved biological, therapeutic, and toxicological properties over the native drug after intravenous administration in platelet-derived growth factor-driven genetically engineered mouse model, and an epidermal growth factor expressing patient-derived xenograft (EGFR PDX) model. Conclusions: Ultrasmall C' dot-drug conjugates showed great translational potential over doxorubicin for improving the therapeutic outcome of patients with high-grade gliomas, even without a cancer-targeting moiety.
Collapse
Affiliation(s)
| | - Anusha Aditya
- Memorial Sloan Kettering Cancer Center, New York, United States
| | - Li Zhang
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Feng Chen
- Memorial Sloan Kettering Cancer Center, United States
| | | | - Tianye Cao
- Memorial Sloan Kettering Cancer Center, New York, United States
| | - Brian Madajewski
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | | | - Kai Ma
- Cornell University, Ithaca, NY, United States
| | - Sebastien Monette
- Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, New York, United States
| | - Peiming Chen
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jing Wu
- Hunter College, United States
| | - Shutian Ruan
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Pat Zanzonico
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Charles M. Rudin
- Memorial Sloan Kettering Cancer Center, New York, New York, United States
| | - Cameron Brennan
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Li Zhang
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
12
|
Zhou YS, Wang W, Chen N, Wang LC, Huang JB. Research progress of anti-glioma chemotherapeutic drugs (Review). Oncol Rep 2022; 47:101. [PMID: 35362540 PMCID: PMC8990335 DOI: 10.3892/or.2022.8312] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Glioma is the most common primary intracranial malignancy in the central nervous system. At present, the most important treatment option is surgical resection of the tumor combined with radiotherapy and chemotherapy. The principle of operation is to remove the tumor to the maximal extent on the basis of preserving brain function. However, prominent invasive and infiltrative proliferation of glioma tumor cells into the surrounding normal tissues frequently reduces the efficacy of treatment. This in turn worsens the prognosis, because the tumor cannot be completely removed, which can readily relapse. Chemotherapeutic agents when applied individually have demonstrated limited efficacy for the treatment of glioma. However, multiple different chemotherapeutic agents can be used in combination with other treatment modalities to improve the efficacy while circumventing systemic toxicity and drug resistance. Therefore, it is pivotal to unravel the inhibitory mechanism mediated by the different chemotherapeutic drugs on glioma cells in preclinical studies. The aim of the present review is to provide a summary for understanding the effects of different chemotherapeutic drugs in glioma, in addition to providing a reference for the preclinical research into novel chemotherapeutic agents for future clinical application.
Collapse
Affiliation(s)
- Yi-Shu Zhou
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Wei Wang
- Department of Radiology and Research Institute for Translation Medicine on Molecular Function and Artificial Intelligence Imaging, The First People's Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Na Chen
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Li-Cui Wang
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Jin-Bai Huang
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
13
|
Liu XY, Yang JB, Wu CY, Tang Q, Lu ZL, Lin L. [12]aneN3-Conjugated AIEgens with Two-Photon Imaging Property for Synergistic Gene/Photodynamic Therapy in Vitro and in Vivo. J Mater Chem B 2022; 10:945-957. [DOI: 10.1039/d1tb02352g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Six amphiphiles (TTC-L-M-1/2/3/4/5/6), each consisting of hydrophilic macrocyclic polyamine triazole-[12]aneN3 (M) and hydrophobic photosensitizer tetraphenylethenethiophene modified cyanoacrylate (TTC) moiety linked with alkyl chains (L), have been designed and synthesized for...
Collapse
|
14
|
Du K, Xia Q, Sun J, Feng F. Visible Light and Glutathione Dually Responsive Delivery of a Polymer-Conjugated Temozolomide Intermediate for Glioblastoma Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:55851-55861. [PMID: 34788006 DOI: 10.1021/acsami.1c16962] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Temozolomide (TMZ) is a prodrug of 5-(3-methyltriazene-1-yl)imidazole-4-carboxamide (MTIC, short-lived) and used as a first-line therapy drug for glioblastoma multiforme (GBM). However, little progress has been made in regulating the kinetics of TMZ to MTIC degradation to improve the therapeutic effect, particularly in the case of TMZ-resistant GBM. In this work, we introduced a strategy to cage MTIC by N-acylation of the triazene moiety to boost the MTIC stability, designed a diblock copolymer-based MTIC prodrug installed with a disulfide linkage, and achieved self-assembled polymer micelles without the concern of MTIC leakage under physiological conditions. Polymer micelles could be induced to disassemble by stimuli factors such as glutathione (GSH) and visible light irradiation through thiol/sulfide exchange and homolytic sulfide scission mechanisms, which contributed to MTIC release in GSH-dependent and GSH-independent pathways. The in vitro results demonstrated that microenvironment-responsive polymeric micelles benefited the suppression of both TMZ-sensitive and TMZ-resistant GBM cells. The chemistry of polymer-MTIC prodrug provided a new option for TMZ-based glioma treatment.
Collapse
Affiliation(s)
- Ke Du
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qiuyu Xia
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jian Sun
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Fude Feng
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
15
|
Wang X, Hu A, Du K, Feng F. Biomimetic Polymer-Templated Copper Nanoparticles Stabilize a Temozolomide Intermediate for Chemotherapy against Glioblastoma Multiforme. ACS APPLIED BIO MATERIALS 2021; 4:8004-8012. [DOI: 10.1021/acsabm.1c00915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xia Wang
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Andi Hu
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ke Du
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Fude Feng
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
16
|
Zhang Y, Cui H, Zhang R, Zhang H, Huang W. Nanoparticulation of Prodrug into Medicines for Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101454. [PMID: 34323373 PMCID: PMC8456229 DOI: 10.1002/advs.202101454] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/16/2021] [Indexed: 05/28/2023]
Abstract
This article provides a broad spectrum about the nanoprodrug fabrication advances co-driven by prodrug and nanotechnology development to potentiate cancer treatment. The nanoprodrug inherits the features of both prodrug concept and nanomedicine know-how, attempts to solve underexploited challenge in cancer treatment cooperatively. Prodrugs can release bioactive drugs on-demand at specific sites to reduce systemic toxicity, this is done by using the special properties of the tumor microenvironment, such as pH value, glutathione concentration, and specific overexpressed enzymes; or by using exogenous stimulation, such as light, heat, and ultrasound. The nanotechnology, manipulating the matter within nanoscale, has high relevance to certain biological conditions, and has been widely utilized in cancer therapy. Together, the marriage of prodrug strategy which shield the side effects of parent drug and nanotechnology with pinpoint delivery capability has conceived highly camouflaged Trojan horse to maneuver cancerous threats.
Collapse
Affiliation(s)
- Yuezhou Zhang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
- Ningbo Institute of Northwestern Polytechnical University, 218 Qingyi Road, Ningbo, 315103, China
| | - Huaguang Cui
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
- Ningbo Institute of Northwestern Polytechnical University, 218 Qingyi Road, Ningbo, 315103, China
| | - Ruiqi Zhang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
- Ningbo Institute of Northwestern Polytechnical University, 218 Qingyi Road, Ningbo, 315103, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, FI-00520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-00520, Finland
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
- Ningbo Institute of Northwestern Polytechnical University, 218 Qingyi Road, Ningbo, 315103, China
| |
Collapse
|
17
|
Du J, Zhang A, Li J, Liu X, Wu S, Wang B, Wang Y, Jia H. Doxorubicin-Induced Cognitive Impairment: The Mechanistic Insights. Front Oncol 2021; 11:673340. [PMID: 34055643 PMCID: PMC8158153 DOI: 10.3389/fonc.2021.673340] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
Chemotherapy can significantly prolong the survival of patients with breast cancer; Nevertheless, the majority of patients receiving chemotherapy such as doxorubicin may have cognitive deficits that manifest as impairments in learning, reasoning, attention, and memory. The phenomenon of chemotherapy-induced cognitive decline is termed as chemotherapy-related cognitive impairment (CRCI) or chemo-brain. Doxorubicin (DOX), a commonly used drug in adjuvant chemotherapy for patients with breast cancer, has been reported to induce chemo-brain through a variety of mechanisms including DNA damage, oxidative stress, inflammation, dysregulation of apoptosis and autophagy, changes in neurotransmitter levels, mitochondrial dysfunction, glial cell interactions, neurogenesis inhibition, and epigenetic factors. These mechanisms do not operate independently but are inter-related, coordinately contributing to the development of chemo-brain. Here we review the relationships of these mechanisms and pathways in attempt to provide mechanistic insights into the doxorubicin-induced cognitive impairment.
Collapse
Affiliation(s)
- Jiajia Du
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Aoxue Zhang
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Xin Liu
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuai Wu
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Bin Wang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Hongyan Jia
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
18
|
Veroniaina H, Pan X, Wu Z, Qi X. Apoferritin: a potential nanocarrier for cancer imaging and drug delivery. Expert Rev Anticancer Ther 2021; 21:901-913. [PMID: 33844625 DOI: 10.1080/14737140.2021.1910027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Introduction: As a protein-based biomaterial for potential cancer targeting delivery, apoferritin has recently attracted interest.Areas covered: In this review, we discuss the development of this cage-like protein as an endogenous nanocarrier that can hold molecules in its cavity. We present the specific characterizations and formulations of apoferritin nanocarriers, and outline the recent progress of the protein as an appropriate tumor-delivery vehicle in different therapeutic strategies to treat solid tumors. Finally, we propose how the application for cancer drug repurposing delivery within apoferritin could expand cancer treatment in the future.Expert opinion: Being a ubiquitous iron storage protein that exists in many living organisms, apoferritin is promising as a cancer tumor-targeting nanocarrier. By exploiting its versatility, apoferritin could be used for cancer repurposed drug delivery and could reduce the high cost of new drug discovery development and shorten the formulation process.
Collapse
Affiliation(s)
| | - Xiuhua Pan
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhenghong Wu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
19
|
Small interfering RNA (siRNA) to target genes and molecular pathways in glioblastoma therapy: Current status with an emphasis on delivery systems. Life Sci 2021; 275:119368. [PMID: 33741417 DOI: 10.1016/j.lfs.2021.119368] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 02/08/2023]
Abstract
Glioblastoma multiforme (GBM) is one of the worst brain tumors arising from glial cells, causing many deaths annually. Surgery, chemotherapy, radiotherapy and immunotherapy are used for GBM treatment. However, GBM is still an incurable disease, and new approaches are required for its successful treatment. Because mutations and amplifications occurring in several genes are responsible for the progression and aggressive behavior of GBM cells, genetic approaches are of great importance in its treatment. Small interfering RNA (siRNA) is a new emerging tool to silence the genes responsible for disease progression, particularly cancer. SiRNA can be used for GBM treatment by down-regulating genes such as VEGF, STAT3, ELTD1 or EGFR. Furthermore, the use of siRNA can promote the chemosensitivity of GBM cells. However, the efficiency of siRNA in GBM is limited via its degradation by enzymes, and its off-targeting effects. SiRNA-loaded carriers, especially nanovehicles that are ligand-functionalized by CXCR4 or angiopep-2, can be used for the protection and targeted delivery of siRNA. Nanostructures can provide a platform for co-delivery of siRNA plus anti-tumor drugs as another benefit. The prepared nanovehicles should be stable and biocompatible in order to be tested in human studies.
Collapse
|
20
|
Verebová V, Beneš J, Staničová J. Biophysical Characterization and Anticancer Activities of Photosensitive Phytoanthraquinones Represented by Hypericin and Its Model Compounds. Molecules 2020; 25:E5666. [PMID: 33271809 PMCID: PMC7731333 DOI: 10.3390/molecules25235666] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/17/2022] Open
Abstract
Photosensitive compounds found in herbs have been reported in recent years as having a variety of interesting medicinal and biological activities. In this review, we focus on photosensitizers such as hypericin and its model compounds emodin, quinizarin, and danthron, which have antiviral, antifungal, antineoplastic, and antitumor effects. They can be utilized as potential agents in photodynamic therapy, especially in photodynamic therapy (PDT) for cancer. We aimed to give a comprehensive summary of the physical and chemical properties of these interesting molecules, emphasizing their mechanism of action in relation to their different interactions with biomacromolecules, specifically with DNA.
Collapse
Affiliation(s)
- Valéria Verebová
- Department of Chemistry, Biochemistry and Biophysics, University of Veterinary Medicine & Pharmacy, Komenského 73, 041 81 Košice, Slovakia;
| | - Jiří Beneš
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Kateřinská 1, 121 08 Prague, Czech Republic;
| | - Jana Staničová
- Department of Chemistry, Biochemistry and Biophysics, University of Veterinary Medicine & Pharmacy, Komenského 73, 041 81 Košice, Slovakia;
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Kateřinská 1, 121 08 Prague, Czech Republic;
| |
Collapse
|
21
|
Liu J, Zhang Y, Chen T, Chen H, He H, Jin T, Wang J, Ke Y. Environmentally Self-Adaptative Nanocarriers Suppress Glioma Proliferation and Stemness via Codelivery of shCD163 and Doxorubicin. ACS APPLIED MATERIALS & INTERFACES 2020; 12:52354-52369. [PMID: 33196179 DOI: 10.1021/acsami.0c14288] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Gliomas-devastating intracranial tumors with a dismal outcome-are in dire need of innovative treatment. Although nanodrugs have been utilized as a target therapy for certain types of solid tumors, their therapeutic effects in gliomas are limited due to the complications of the systemic circulation, blood-brain barrier (BBB), and specific glioma environment. Thus, we aimed to establish a nanoliposome adaptable to different environments by codelivery of shCD163 and doxorubicin (DOX) to treat gliomas. In this study, we first synthesized pH-sensitive DSPE-cRGD-Hz-PEG2000 to form an environmentally self-adaptative nanoliposome (cRGD-DDD Lip) via a thin film method. We used in vitro BBB models, in vitro cell uptake experiments, and in vivo biodistribution assays to confirm the long circulation time and low cell uptake of the cRGD-DDD Lip as a result of the poly(ethylene glycol) (PEG) shell of cRGD-DDD Lip in the neutral pH systemic circulation. Moreover, the cRGD-DDD Lip bypassed the BBB and attached to the intracranial glioma following the removal of the PEG shell and the exposure of cRGD to the weakly acidic tumor microenvironment. We further assembled the shCD163/DOX@cRGD-DDD Lip through cRGD-DDD Lip loading of shCD163 and DOX. In vitro, cell proliferation and self-renewal of glioma cells were inhibited by the shCD163/DOX@cRGD-DDD Lip due to the toxicity of DOX and the suppression of shCD163 via the CD163 pathway. In vivo, the shCD163/DOX@cRGD-DDD Lip disturbed the progression of in situ gliomas by inhibiting the growth and stemness of glioma cells and prevented the recurrence of gliomas after resection. In conclusion, the cRGD-DDD Lip may be a promising nanodrug-loading platform to cope with different environments and the shCD163/DOX@cRGD-DDD Lip may potentially be a novel nanodrug for glioma therapy.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/pharmacology
- Antibiotics, Antineoplastic/therapeutic use
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/metabolism
- Brain Neoplasms/drug therapy
- Brain Neoplasms/mortality
- Brain Neoplasms/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Doxorubicin/chemistry
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Glioma/drug therapy
- Glioma/mortality
- Glioma/pathology
- Humans
- Liposomes/chemistry
- Mice
- Mice, Nude
- Nanoparticles/chemistry
- Nanoparticles/metabolism
- Oligopeptides/chemistry
- Polyethylene Glycols/chemistry
- RNA Interference
- RNA, Small Interfering/chemistry
- RNA, Small Interfering/metabolism
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Survival Rate
- Tissue Distribution
Collapse
Affiliation(s)
- Jie Liu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yuxuan Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Taoliang Chen
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Huajian Chen
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Haoqi He
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Tao Jin
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jihui Wang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yiquan Ke
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
22
|
Horescu C, Elena Cioc C, Tuta C, Sevastre AS, Tache DE, Alexandru O, Artene SA, Danoiu S, Dricu A, Stefana Oana P. The effect of temozolomide in combination with doxorubicin in glioblastoma cells in vitro. J Immunoassay Immunochem 2020; 41:1033-1043. [PMID: 33021886 DOI: 10.1080/15321819.2020.1819309] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Prior to 2000, the DNA alkylating agents nitrosoureas were used as standard treatment of glioblastoma. Current treatments for glioblastoma patients consist of surgery followed by radiation in combination with temozolomide. Despite therapeutic advances, the prognosis for glioblastoma patients remains grim, with a five-year overall survival below 15%. In this study, our team analyzed the interaction between temozolomide and doxorubicin in a glioblastoma cell line, in vitro. MATERIALS AND METHOD The cell line, established from a patient who underwent surgery at the "Bagdasar Arseni Emergency Hospital", was exposed to 10 µM and 100 µM of temozolomide and 10 nM and 100 nM of doxorubicin, respectively, over a period of 7, 10 and 14 days, in monotherapy and in combination. RESULTS The results showed that both temozolomide (66.5% cytotoxicity for the 10 µM dose at 14 days) de and doxorubicin (66.8% cytotoxicity for the 10 nM dose after 14 days) were very effective in killing cancer cells in monotherapy, but failed to produce a synergistic effect when used in combination. CONCLUSION While the results may be discouraging, they present an interesting prospect into how certain drug interactions can impact treatment response.
Collapse
Affiliation(s)
- Cristina Horescu
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Catalina Elena Cioc
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Cristian Tuta
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Ani-Simona Sevastre
- Faculty of Pharmacy, Pharmaceutical Technology Department, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Daniela Elise Tache
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Oana Alexandru
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | | | - Suzana Danoiu
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Anica Dricu
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Purcaru Stefana Oana
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| |
Collapse
|