1
|
Fattahi N, Aghaz F, Rezaei A, Ramazani A, Heydari A, Hosseininezhad S, Jung WK. pH-responsive magnetic CuFe 2O 4-PMAA nanogel conjugated with amino-modified lignin for controlled breast cancer drug delivery. Sci Rep 2024; 14:25987. [PMID: 39472530 PMCID: PMC11522326 DOI: 10.1038/s41598-024-77414-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
In this study, a novel magnetic and pH-responsive nanocarrier was developed, incorporating both natural and synthetic polymers, for delivering curcumin (CUR) to breast cancer cells. For this purpose, CuFe2O4@poly(methacrylic acid) (CuFe2O4@PMAA) nanogel was developed and conjugated with amino-modified lignin (Lignin-adipic acid dihydrazide conjugate, Lig-ADH) to achieve the CuFe2O4@PMAA@Lig-ADH nanocarrier. The morphology, structure, and physical properties of the synthesized nanomaterials were examined using a range of techniques, including transmission electron microscopy (TEM), field emission scanning electron microscopy (FESEM), Fourier transform infrared (FT-IR) spectroscopy, X-ray diffraction (XRD), energy dispersive X-ray (EDX), and vibrating sample magnetometer (VSM). The synthesized nanocarrier exhibited a spherical shape, with an average diameter of approximately 15 nm, and demonstrated good magnetic responsiveness. Moreover, the in vitro drug release was found to be pH-dependent, with an increased release rate in acidic conditions. To evaluate cytotoxicity, the survival of MCF-7 cells was measured using the MTT assay for 24 h. Notably, the synthesized CuFe2O4@PMAA@Lig-ADH@CUR and CUR exhibited significant cytotoxic effects, effectively eliminating MCF-7 cells with IC50 values of 39.80 µg/mL and 4.27 µg/mL, respectively. Also, the significant intracellular uptake of NPs was confirmed by FITC and DAPI staining after 4 h. This research highlighted the potential of CuFe2O4@PMAA@Lig-ADH@CUR as a highly effective nano-delivery system and demonstrated a straightforward method for utilizing renewable lignin.
Collapse
Affiliation(s)
- Nadia Fattahi
- The Organic Chemistry Research Laboratory (OCRL), Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan, 45371-38791, Iran
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
| | - Faranak Aghaz
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 67158-47141, Iran
| | - Aram Rezaei
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 67158-47141, Iran.
| | - Ali Ramazani
- The Organic Chemistry Research Laboratory (OCRL), Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan, 45371-38791, Iran.
| | - Abolfazl Heydari
- Polymer Institute of the Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, 845 41, Slovakia
| | - Seyedmohammad Hosseininezhad
- The Organic Chemistry Research Laboratory (OCRL), Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan, 45371-38791, Iran
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea.
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea.
- Major of Biomedical Engineering, Division of Smart Healthcare and New-Senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan, 48513, Korea.
| |
Collapse
|
2
|
Rothe R, Xu Y, Wodtke J, Brandt F, Meister S, Laube M, Lollini PL, Zhang Y, Pietzsch J, Hauser S. Programmable Release of Chemotherapeutics from Ferrocene-Based Injectable Hydrogels Slows Melanoma Growth. Adv Healthc Mater 2024; 13:e2400265. [PMID: 39007274 DOI: 10.1002/adhm.202400265] [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: 01/23/2024] [Revised: 07/02/2024] [Indexed: 07/16/2024]
Abstract
Hydrogel-based injectable drug delivery systems provide temporally and spatially controlled drug release with reduced adverse effects on healthy tissues. Therefore, they represent a promising therapeutic option for unresectable solid tumor entities. In this study, a peptide-starPEG/hyaluronic acid-based physical hydrogel is modified with ferrocene to provide a programmable drug release orchestrated by matrix-drug interaction and local reactive oxygen species (ROS). The injectable ROS-responsive hydrogel (hiROSponse) exhibits adequate biocompatibility and biodegradability, which are important for clinical applications. HiROSponse is loaded with the two cytostatic drugs (hiROSponsedox/ptx) doxorubicin (dox) and paclitaxel (ptx). Dox is a hydrophilic compound and its release is mainly controlled by Fickian diffusion, while the hydrophobic interactions between ptx and ferrocene can control its release and thus be regulated by the oxidation of ferrocene to the more hydrophilic state of ferrocenium. In a syngeneic malignant melanoma-bearing mouse model, hiROSponsedox/ptx slows tumor growth without causing adverse side effects and doubles the relative survival probability. Programmable release is further demonstrated in a tumor model with a low physiological ROS level, where dox release, low dose local irradiation, and the resulting ROS-triggered ptx release lead to tumor growth inhibition and increased survival.
Collapse
Affiliation(s)
- Rebecca Rothe
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Yong Xu
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Tatzberg 41, 01307, Dresden, Germany
| | - Johanna Wodtke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Florian Brandt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Sebastian Meister
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Markus Laube
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Pier-Luigi Lollini
- Alma Mater Studiorum, University of Bologna, Department of Medical and Surgical Sciences, Viale Filopanti 22, Bologna, 40126, Italy
| | - Yixin Zhang
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Tatzberg 41, 01307, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Sandra Hauser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| |
Collapse
|
3
|
Jayaswal N, Srivastava S, Kumar S, Belagodu Sridhar S, Khalid A, Najmi A, Zoghebi K, Alhazmi HA, Mohan S, Tambuwala MM. Precision arrows: Navigating breast cancer with nanotechnology siRNA. Int J Pharm 2024; 662:124403. [PMID: 38944167 DOI: 10.1016/j.ijpharm.2024.124403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Nanotechnology-based drug delivery systems, including siRNA, present an innovative approach to treating breast cancer, which disproportionately affects women. These systems enable personalized and targeted therapies, adept at managing drug resistance and minimizing off-target effects. This review delves into the current landscape of nanotechnology-derived siRNA transport systems for breast cancer treatment, discussing their mechanisms of action, preclinical and clinical research, therapeutic applications, challenges, and future prospects. Emphasis is placed on the importance of targeted delivery and precise gene silencing in improving therapeutic efficacy and patient outcomes. The review addresses specific hurdles such as specificity, biodistribution, immunological reactions, and regulatory approval, offering potential solutions and avenues for future research. SiRNA drug delivery systems hold promise in revolutionizing cancer care and improving patient outcomes, but realizing their full potential necessitates ongoing research, innovation, and collaboration. Understanding the intricacies of siRNA delivery mechanisms is pivotal for designing effective cancer treatments, overcoming challenges, and advancing siRNA-based therapies for various diseases, including cancer. The article provides a comprehensive review of the methods involved in siRNA transport for therapeutic applications, particularly in cancer treatment, elucidating the complex journey of siRNA molecules from extracellular space to intracellular targets. Key mechanisms such as endocytosis, receptor-mediated uptake, and membrane fusion are explored, alongside innovative delivery vehicles and technologies that enhance siRNA delivery efficiency. Moreover, the article discusses challenges and opportunities in the field, including issues related to specificity, biodistribution, immune response, and clinical translation. By comprehending the mechanisms of siRNA delivery, researchers can design and develop more effective siRNA-based therapies for various diseases, including cancer.
Collapse
Affiliation(s)
- Nandani Jayaswal
- Faculty of Pharmaceutical Sciences, Mahayogi Gorakhnath University, Gorakhpur, 273007, India
| | - Shriyansh Srivastava
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 273007, India; Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, India.
| | - Sachin Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 273007, India
| | | | - Asaad Khalid
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia.
| | - Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Khalid Zoghebi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Hassan A Alhazmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK; RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE.
| |
Collapse
|
4
|
Kong F, Lu Z, Xiong Y, Zhou L, Ye Q. A novel cancer-associated fibroblasts risk score model predict survival and immunotherapy in lung adenocarcinoma. Mol Genet Genomics 2024; 299:70. [PMID: 39017768 DOI: 10.1007/s00438-024-02156-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/09/2024] [Indexed: 07/18/2024]
Abstract
Lung adenocarcinoma (LUAD) is the leading cause of cancer-related death worldwide. Cancer-associated fibroblasts (CAFs) are a special type of fibroblasts, which play an important role in the development and immune escape of tumors. Weighted gene co-expression network analysis (WGCNA) was used to construct the co-expression module. In combination with univariate Cox regression and analysis of least absolute shrinkage operator (LASSO), characteristics associated with CAFs were developed for a prognostic model. The migration and proliferation of lung cancer cells were evaluated in vitro. Finally, the expression levels of proteins were analyzed by Western blot. LASSO Cox regression algorithm was then performed to select hub genes. Finally, a total of 2 Genes (COL5A2, COL6A2) were obtained. We then divided LUAD patients into high- and low-risk groups based on CAFs risk scores. Survival analysis, CAFs score correlation analysis and tumor mutation load analysis showed that COL5A2 and COL6A2 were high-risk genes for LUAD. Human Protein Atlas (HPA), western blot and PCR results showed that COL5A2 and COL6A2 were up-regulated in LUAD tissues. When COL5A2 and COL6A2 were knocked down, the proliferation, invasion and migration of lung cancer cells were significantly decreased. Finally, COL5A2 can affect LUAD progression through the Wnt/β-Catenin and TGF-β signaling pathways. Our CAFs risk score model offers a new approach for predicting the prognosis of LUAD patients. Furthermore, the identification of high-risk genes COL5A2 and COL6A2 and drug sensitivity analysis can provide valuable candidate clues for clinical treatment of LUAD.
Collapse
Affiliation(s)
- Fanhua Kong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, Hubei, 430071, China
| | - Zhongshan Lu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, Hubei, 430071, China
| | - Yan Xiong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, Hubei, 430071, China.
| | - Lihua Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, Hubei, 430071, China.
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, Hubei, 430071, China.
- The 3rd Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, Changsha, China.
| |
Collapse
|
5
|
Adams SC, Nambiar AK, Bressler EM, Raut CP, Colson YL, Wong WW, Grinstaff MW. Immunotherapies for locally aggressive cancers. Adv Drug Deliv Rev 2024; 210:115331. [PMID: 38729264 PMCID: PMC11228555 DOI: 10.1016/j.addr.2024.115331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/31/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
Improving surgical resection outcomes for locally aggressive tumors is key to inducing durable locoregional disease control and preventing progression to metastatic disease. Macroscopically complete resection of the tumor is the standard of care for many cancers, including breast, ovarian, lung, sarcoma, and mesothelioma. Advancements in cancer diagnostics are increasing the number of surgically eligible cases through early detection. Thus, a unique opportunity arises to improve patient outcomes with decreased recurrence rates via intraoperative delivery treatments using local drug delivery strategies after the tumor has been resected. Of the current systemic treatments (e.g., chemotherapy, targeted therapies, and immunotherapies), immunotherapies are the latest approach to offer significant benefits. Intraoperative strategies benefit from direct access to the tumor microenvironment which improves drug uptake to the tumor and simultaneously minimizes the risk of drug entering healthy tissues thereby resulting in fewer or less toxic adverse events. We review the current state of immunotherapy development and discuss the opportunities that intraoperative treatment provides. We conclude by summarizing progress in current research, identifying areas for exploration, and discussing future prospects in sustained remission.
Collapse
Affiliation(s)
- Sarah C Adams
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Arun K Nambiar
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Eric M Bressler
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Chandrajit P Raut
- Department of Surgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Yolonda L Colson
- Massachusetts General Hospital, Department of Surgery, Boston, MA 02114, USA.
| | - Wilson W Wong
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Chemistry, Boston University, Boston MA 02215, USA.
| |
Collapse
|
6
|
Alshehri AM, Wilson OC. Biomimetic Hydrogel Strategies for Cancer Therapy. Gels 2024; 10:437. [PMID: 39057460 PMCID: PMC11275631 DOI: 10.3390/gels10070437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Recent developments in biomimetic hydrogel research have expanded the scope of biomedical technologies that can be used to model, diagnose, and treat a wide range of medical conditions. Cancer presents one of the most intractable challenges in this arena due to the surreptitious mechanisms that it employs to evade detection and treatment. In order to address these challenges, biomimetic design principles can be adapted to beat cancer at its own game. Biomimetic design strategies are inspired by natural biological systems and offer promising opportunities for developing life-changing methods to model, detect, diagnose, treat, and cure various types of static and metastatic cancers. In particular, focusing on the cellular and subcellular phenomena that serve as fundamental drivers for the peculiar behavioral traits of cancer can provide rich insights into eradicating cancer in all of its manifestations. This review highlights promising developments in biomimetic nanocomposite hydrogels that contribute to cancer therapies via enhanced drug delivery strategies and modeling cancer mechanobiology phenomena in relation to metastasis and synergistic sensing systems. Creative efforts to amplify biomimetic design research to advance the development of more effective cancer therapies will be discussed in alignment with international collaborative goals to cure cancer.
Collapse
Affiliation(s)
- Awatef M. Alshehri
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
- Department of Nanomedicine, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdelaziz University for Health Sciences (KSAU-HS), Ministry of National Guard-Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia;
| | - Otto C. Wilson
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
| |
Collapse
|
7
|
Voznyuk AA, Makarets YA, Advakhova DY, Khafizov KA, Lugovoi ME, Zakharova VA, Senatov FS, Koudan EV. Biodegradable Local Chemotherapy Platform with Prolonged and Controlled Release of Doxorubicin for the Prevention of Local Tumor Recurrence. ACS APPLIED BIO MATERIALS 2024; 7:2472-2487. [PMID: 38480461 DOI: 10.1021/acsabm.4c00078] [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: 04/16/2024]
Abstract
Local recurrence after surgical and therapeutic treatment remains a significant clinical problem in oncology. Recurrence may be due to imperfections in existing therapies, particularly chemotherapy. To improve antitumor activity and prevent local cancer recurrence while keeping toxicity at acceptable levels, we have developed and demonstrated a biodegradable local chemotherapy platform that provides controlled and prolonged drug release. The platform consists of a polycaprolactone (PCL) substrate, which provides the structural integrity of the platform and the predominant unidirectional drug release, and a thin multilayer coating (∼200 nm) containing doxorubicin (DOX). The coating is an electrostatic complex obtained by the layer-by-layer (LbL) assembly and consists of natural polyelectrolytes [poly-γ-glutamic acid (γ-PGA) and chitosan (CS) or poly-l-lysine (PLL)]. To improve the release stability, an ionic conjugate of DOX and γ-PGA was prepared and incorporated into the multilayer coating. By varying the structure of the coating by adding empty (without DOX) bilayers, we were able to control the kinetics of drug release. The resulting platforms contained equal numbers of empty bilayers and DOX-loaded bilayers (15 + 15 or 30 + 30 bilayers) with a maximum loading of 566 ng/cm2. The platforms demonstrated prolonged and fairly uniform drug release for more than 5 months while retaining antitumor activity in vitro on ovarian cancer cells (SKOV-3). The empty platforms (without DOX) showed good cytocompatibility and no cytotoxicity to human fibroblasts and SKOV-3 cells. This study presents the development of a local chemotherapy platform consisting of a PCL-based substrate which provides structural stability and a biodegradable polyelectrolyte layered coating which combines layers containing a polyanion ionic complex with DOX with empty bilayers to ensure prolonged and controlled drug release. Our results may provide a basis for improving the efficacy of chemotherapy using drug delivery systems.
Collapse
Affiliation(s)
- Amina A Voznyuk
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Yulia A Makarets
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Darya Yu Advakhova
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Krestina A Khafizov
- Haute École de la Province de Namur, Henri Blès st. 192, Namur 5000, Belgium
| | - Maksim E Lugovoi
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Vasilina A Zakharova
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Fedor S Senatov
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Elizaveta V Koudan
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| |
Collapse
|
8
|
Li Z, Liu X, Xiao J, Jiang H, Ma L, Luo Y, Wang M, Zhu Y, Jiang H, Yao H, Ngai T, Guo Q. Ultrastable Iodinated Oil-Based Pickering Emulsion Enables Locoregional Sustained Codelivery of Hypoxia Inducible Factor-1 Inhibitor and Anticancer Drugs for Tumor Combination Chemotherapy. ACS Biomater Sci Eng 2024; 10:2270-2281. [PMID: 38536862 DOI: 10.1021/acsbiomaterials.3c01887] [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: 04/09/2024]
Abstract
Tumor hypoxia-associated drug resistance presents a major challenge for cancer chemotherapy. However, sustained delivery systems with a high loading capability of hypoxia-inducible factor-1 (HIF-1) inhibitors are still limited. Here, we developed an ultrastable iodinated oil-based Pickering emulsion (PE) to achieve locally sustained codelivery of a HIF-1 inhibitor of acriflavine and an anticancer drug of doxorubicin for tumor synergistic chemotherapy. The PE exhibited facile injectability for intratumoral administration, great radiopacity for in vivo examination, excellent physical stability (>1 mo), and long-term sustained release capability of both hydrophilic drugs (i.e., acriflavine and doxorubicin). We found that the codelivery of acriflavine and doxorubicin from the PE promoted the local accumulation and retention of both drugs using an acellular liver organ model and demonstrated significant inhibition of tumor growth in a 4T1 tumor-bearing mouse model, improving the chemotherapeutic efficacy through the synergistic effects of direct cytotoxicity with the functional suppression of HIF-1 pathways of tumor cells. Such an iodinated oil-based PE provides a great injectable sustained delivery platform of hydrophilic drugs for locoregional chemotherapy.
Collapse
Affiliation(s)
- Zhihua Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaoya Liu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jingyu Xiao
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hang Jiang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Le Ma
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yucheng Luo
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Meijuan Wang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuwei Zhu
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, PR China
| | - Hongliang Jiang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hanyang Yao
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - To Ngai
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, PR China
| | - Qiongyu Guo
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
9
|
Ebrahimnia M, Alavi S, Vaezi H, Karamat Iradmousa M, Haeri A. Exploring the vast potentials and probable limitations of novel and nanostructured implantable drug delivery systems for cancer treatment. EXCLI JOURNAL 2024; 23:143-179. [PMID: 38487087 PMCID: PMC10938236 DOI: 10.17179/excli2023-6747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/08/2024] [Indexed: 03/17/2024]
Abstract
Conventional cancer chemotherapy regimens, albeit successful to some extent, suffer from some significant drawbacks, such as high-dose requirements, limited bioavailability, low therapeutic indices, emergence of multiple drug resistance, off-target distribution, and adverse effects. The main goal of developing implantable drug delivery systems (IDDS) is to address these challenges and maintain anti-cancer drugs directly at the intended sites of therapeutic action while minimizing inevitable side effects. IDDS possess numerous advantages over conventional drug delivery, including controlled drug release patterns, one-time drug administration, as well as loading and stabilizing poorly water-soluble chemotherapy drugs. Here, we summarized conventional and novel (three-dimensional (3D) printing and microfluidic) preparation techniques of different IDDS, including nanofibers, films, hydrogels, wafers, sponges, and osmotic pumps. These systems could be designed with high biocompatibility and biodegradability features using a wide variety of natural and synthetic polymers. We also reviewed the published data on these systems in cancer therapy with a particular focus on their release behavior. Various release profiles could be attained in IDDS, which enable predictable, adjustable, and sustained drug releases. Furthermore, multi-step or stimuli-responsive drug release could be obtained in these systems. The studies mentioned in this article have proven the effectiveness of IDDS for treating different cancer types with high prevalence, including breast cancer, and aggressive cancer types, such as glioblastoma and liver cancer. Additionally, the challenges in applying IDDS for efficacious cancer therapy and their potential future developments are also discussed. Considering the high potential of IDDS for further advancements, such as programmable release and degradation features, further clinical trials are needed to ensure their efficiency. The overall goal of this review is to expand our understanding of the behavior of commonly investigated IDDS and to identify the barriers that should be addressed in the pursuit of more efficient therapies for cancer. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Maryam Ebrahimnia
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sonia Alavi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Hamed Vaezi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdieh Karamat Iradmousa
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Bakhrushina EO, Mikhel IB, Buraya LM, Moiseev ED, Zubareva IM, Belyatskaya AV, Evzikov GY, Bondarenko AP, Krasnyuk II, Krasnyuk II. Implantation of In Situ Gelling Systems for the Delivery of Chemotherapeutic Agents. Gels 2024; 10:44. [PMID: 38247767 PMCID: PMC10815592 DOI: 10.3390/gels10010044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024] Open
Abstract
Implantation is a modern method of administering chemotherapeutic agents, with a highly targeted effect and better patient tolerance due to the low frequency of administration. Implants are capable of controlled release, which makes them a viable alternative to infusional chemotherapy, allowing patients to enjoy a better quality of life without the need for prolonged hospitalization. Compared to subcutaneous implantation, intratumoral implantation has a number of significant advantages in terms of targeting and side effects, but this area of chemotherapy is still poorly understood in terms of clinical trials. At the same time, there are more known developments of drugs in the form of implants and injections for intratumoral administration. The disadvantages of classical intratumoral implants are the need for surgical intervention to install the system and the increased risk of tumor rupture noted by some specialists. The new generation of implants are in situ implants-systems formed in the tumor due to a phase transition (sol-gel transition) under the influence of various stimuli. Among this systems some are highly selective for a certain type of malignant neoplasm. Such systems are injected and have all the advantages of intratumoral injections, but due to the phase transition occurring in situ, they form depot forms that allow the long-term release of chemotherapeutic agents.
Collapse
Affiliation(s)
- Elena O. Bakhrushina
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Iosif B. Mikhel
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Liliya M. Buraya
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Egor D. Moiseev
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Irina M. Zubareva
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
- Department of Pharmacology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia
| | - Anastasia V. Belyatskaya
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Grigory Y. Evzikov
- Department of Nervous Diseases and Neurosurgery, N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia;
| | | | - Ivan I. Krasnyuk
- Department of Analytical, Physical and Colloidal Chemistry, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia;
| | - Ivan I. Krasnyuk
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| |
Collapse
|
11
|
Sadri E, Khoee S, Moayeri S, Haji Ali B, Pirhajati Mahabadi V, Shirvalilou S, Khoei S. Enhanced anti-tumor activity of transferrin/folate dual-targeting magnetic nanoparticles using chemo-thermo therapy on retinoblastoma cancer cells Y79. Sci Rep 2023; 13:22358. [PMID: 38102193 PMCID: PMC10724238 DOI: 10.1038/s41598-023-49171-5] [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: 07/19/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023] Open
Abstract
Malignant neoplasms are one of the main causes of death, especially in children, on a global scale, despite strenuous efforts made at advancing both diagnostic and therapeutic modalities. In this regard, a new nanocarrier Vincristine (VCR)-loaded Pluronic f127 polymer-coated magnetic nanoparticles conjugated with folic acid and transferrin (PMNP-VCR-FA-TF) were synthesized and characterized by various methods. The cytotoxicity of these nanoparticles was evaluated in vitro and ex vivo conditions. The in vitro anti-tumor effect of the nanoparticles was evaluated by colony formation assay (CFA) and reactive oxygen species (ROS) in Y79 cell line. The results showed that nanoparticles with two ligands conferred greater toxicity toward Y79 cancer cells than ARPE19 normal cells. Under an alternating magnetic field (AMF), these nanoparticles demonstrated a high specific absorption rate. The CFA and ROS results indicated that the AMF in combination with PMNP-VCR-FA-TF conferred the highest cytotoxicity toward Y79 cells compared with other groups (P < 0.05). PMNP-VCR-FA-TF could play an important role in converting externally applied radiofrequency energy into heat in cancer cells. The present study confirmed that dual targeting chemo-hyperthermia using PMNP-VCR-FA-TF was significantly more effective than hyperthermia or chemotherapy alone, providing a promising platform for precision drug delivery as an essential component in the chemotherapy of retinoblastoma.
Collapse
Affiliation(s)
- Elaheh Sadri
- Finetech in Medicine Research Center, Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Khoee
- Department of Polymer Chemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Samaneh Moayeri
- Department of Polymer Chemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Bahareh Haji Ali
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Vahid Pirhajati Mahabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sakine Shirvalilou
- Finetech in Medicine Research Center, Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran.
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Samideh Khoei
- Finetech in Medicine Research Center, Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran.
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Gunay G, Maier KN, Hamsici S, Carvalho F, Timog TA, Acar H. Peptide aggregation-induced immunogenic cell death in a breast cancer spheroid model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.31.565012. [PMID: 37961293 PMCID: PMC10635027 DOI: 10.1101/2023.10.31.565012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Utilizing multicellular aggregates (spheroids) for in vitro cancer research offers a physiologically relevant model that closely mirrors the intricate tumor microenvironment, capturing properties of solid tumors such as cell interactions and drug resistance. In this research, we investigated the Peptide-Aggregation Induced Immunogenic Response (PAIIR), an innovative method employing engineered peptides we designed specifically to induce immunogenic cell death (ICD). We contrasted PAIIR-induced ICD with standard ICD and non-ICD inducer chemotherapeutics within the context of three-dimensional breast cancer tumor spheroids. Our findings reveal that PAIIR outperforms traditional chemotherapeutics in its efficacy to stimulate ICD. This is marked by the release of key damage-associated molecular patterns (DAMPs), which bolster the phagocytic clearance of dying cancer cells by dendritic cells (DCs) and, in turn, activate powerful anti-tumor immune responses. Additionally, we observed that PAIIR results in elevated dendritic cell activation and increased antitumor cytokine presence. This study not only showcases the utility of tumor spheroids for efficient high-throughput screening but also emphasizes PAIIR's potential as a formidable immunotherapeutic strategy against breast cancer, setting the stage for deeper exploration and potential clinical implementation.
Collapse
|
13
|
Kim J, Song S, Gwak M, Cho H, Yun WS, Hwang N, Kim J, Lee JS, Kim DH, Kim H, Jeon SI, Kim TI, Kim K. Micro-syringe chip-guided intratumoral administration of lipid nanoparticles for targeted anticancer therapy. Biomater Res 2023; 27:102. [PMID: 37845762 PMCID: PMC10577945 DOI: 10.1186/s40824-023-00440-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Nano-sized drug delivery system has been widely studied as a potential technique to promote tumor-specific delivery of anticancer drugs due to its passive targeting property, but resulting in very restricted improvements in its systemic administration so far. There is a requirement for a different approach that dramatically increases the targeting efficiency of therapeutic agents at targeted tumor tissues. METHODS To improve the tumor-specific accumulation of anticancer drugs and minimize their undesirable toxicity to normal tissues, a tumor-implantable micro-syringe chip (MSC) with a drug reservoir is fabricated. As a clinically established delivery system, six liposome nanoparticles (LNPs) with different compositions and surface chemistry are prepared and their physicochemical properties and cellular uptake are examined in vitro. Subsequently, MSC-guided intratumoral administration is studied to identify the most appropriate for the higher tumor targeting efficacy with a uniform intratumoral distribution. For efficient cancer treatment, pro-apoptotic anticancer prodrugs (SMAC-P-FRRG-DOX) are encapsulated to the optimal LNPs (SMAC-P-FRRG-DOX encapsulating LNPs; ApoLNPs), then the ApoLNPs are loaded into the 1 μL-volume drug reservoir of MSC to be delivered intratumorally for 9 h. The tumor accumulation and therapeutic effect of ApoLNPs administered via MSC guidance are evaluated and compared to those of intravenous and intratumoral administration of ApoLNP in 4T1 tumor-bearing mice. RESULTS MSC is precisely fabricated to have a 0.5 × 4.5 mm needle and 1 μL-volume drug reservoir to achieve the uniform intratumoral distribution of LNPs in targeted tumor tissues. Six liposome nanoparticles with different compositions of 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine (PC), 1,2-dioleoyl-sn-glycero-3-phospho-L-serine (PS), 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethylene glycol)2000] (PEG2000-DSPE) are prepared with average sizes of 100-120 nm and loaded into the 1 μL-volume drug reservoir in MSC. Importantly negatively charged 10 mol% of PS-containing LNPs are very slowly infused into the tumor tissue through the micro-syringe of the MSC over 6 h. The intratumoral targeting efficiency of MSC guidance is 93.5%, effectively assisting the homogeneous diffusion of LNPs throughout the tumor tissue at 3.8- and 2.7-fold higher concentrations compared to the intravenous and intratumoral administrations of LNPs, respectively. Among the six LNP candidates 10 mol% of PS-containing LNPs are finally selected for preparing pro-apoptotic SMAC-P-FRRG-DOX anticancer prodrug-encapsulated LNPs (ApoLNPs) due to their moderate endocytosis rate high tumor accumulation and homogenous intratumoral distribution. The ApoLNPs show a high therapeutic effect specifically to cathepsin B-overexpressing cancer cells with 6.6 μM of IC50 value while its IC50 against normal cells is 230.7 μM. The MSC-guided administration of ApoLNPs efficiently inhibits tumor growth wherein the size of the tumor is 4.7- and 2.2-fold smaller than those treated with saline and intratumoral ApoLNP without MSC, respectively. Moreover, the ApoLNPs remarkably reduce the inhibitor of apoptosis proteins (IAPs) level in tumor tissues confirming their efficacy even in cancers with high drug resistance. CONCLUSION The MSC-guided administration of LNPs greatly enhances the therapeutic efficiency of anticancer drugs via the slow diffusion mechanism through micro-syringe to tumor tissues for 6 h, whereas they bypass most hurdles of systemic delivery including hepatic metabolism, rapid renal clearance, and interaction with blood components or other normal tissues, resulting in the minimum toxicity to normal tissues. The negatively charged ApoLNPs with cancer cell-specific pro-apoptotic prodrug (SMAC-P-FRRG-DOX) show the highest tumor-targeting efficacy when they are treated with the MSC guidance, compared to their intravenous or intratumoral administration in 4T1 tumor-bearing mice. The MSC-guided administration of anticancer drug-encapsulated LNPs is expected to be a potent platform system that facilitates overcoming the limitations of systemic drug administration with low delivery efficiency and serious side effects.
Collapse
Affiliation(s)
- Jeongrae Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Woman's University, Seoul, 03760, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Sunejeong Song
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Woman's University, Seoul, 03760, Republic of Korea
| | - Minjun Gwak
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Hanhee Cho
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Woman's University, Seoul, 03760, Republic of Korea
| | - Wan Su Yun
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Namcheol Hwang
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Jinseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Woman's University, Seoul, 03760, Republic of Korea
| | - Jun Seo Lee
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Hyuncheol Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Seong Ik Jeon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Woman's University, Seoul, 03760, Republic of Korea.
| | - Tae-Il Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
| | - Kwangmeyung Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Woman's University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
14
|
Jin X, Zhao J, Li H, Zheng M, Shao J, Chen Z. Research trends and hot spots in global nanotechnology applications in liver cancer: a bibliometric and visual analysis (2000-2022). Front Oncol 2023; 13:1192597. [PMID: 37664074 PMCID: PMC10472833 DOI: 10.3389/fonc.2023.1192597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/11/2023] [Indexed: 09/05/2023] Open
Abstract
Background Liver cancer (LC) is one of the most common malignancies. Currently, nanotechnology has made great progress in LC research, and many studies on LC nanotechnology have been published. This study aims to discuss the current status, hot spots, and research trends in this field through bibliometric analysis. Methods The Web of Science Core Collection (WoSCC) database was searched for papers related to hepatocellular carcinoma (HCC) included from January 2000 to November 2022, and its research hotspots and trends were visualized and analyzed with the help of VOSviewer. In addition, a search was conducted to find LC papers related to nanotechnology. Then we used the visual analysis software VOSviewer and CiteSpace to evaluate the contributions of countries/regions, authors, and journals related to the topic and analyze keywords to understand the research priorities and hot spots in the field as well as the development direction. Results There are 1908 papers in the highly cited literature on LC, and its research hotspots are pathogenesis, risk factors, and survival rate. The literature on the application of nanotechnology in LC had 921 papers. Among them, China (n=560, 60.8%) and the United States (n=170, 18.5%) were the countries with the highest number of published papers. Wang Yan (n=11) and Llovet JM (n=131) were the first authors and co-cited authors, respectively. The International Journal of Nanomedicine was the most prolific academic journal (n=41). In addition to "hepatocellular carcinoma" and "nanoparticles", the most frequent keyword was "drug delivery". In recent years, "metastasis" and "diagnosis" appeared in the keyword bursts. This indicates that the application of nanoparticles in the early diagnosis and drug delivery of LC (including liver metastasis) has a good prospect. Conclusion Nanotechnology has received more and more attention in the medical field in recent years. As nanoparticles are easily localized in organelles and cells, they can increase drug permeability in tumor tissues, improve drug delivery efficiency and reduce drug toxicity. Our research results were the first scientific evaluation of the application of nanotechnology in LC, providing scholars with research hotspots and development trends.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhanguo Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
15
|
Caputo TM, Cusano AM, Principe S, Cicatiello P, Celetti G, Aliberti A, Micco A, Ruvo M, Tagliamonte M, Ragone C, Minopoli M, Carriero MV, Buonaguro L, Cusano A. Sorafenib-Loaded PLGA Carriers for Enhanced Drug Delivery and Cellular Uptake in Liver Cancer Cells. Int J Nanomedicine 2023; 18:4121-4142. [PMID: 37525693 PMCID: PMC10387258 DOI: 10.2147/ijn.s415968] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/29/2023] [Indexed: 08/02/2023] Open
Abstract
Introduction Currently, conventional treatments of hepatocellular carcinoma (HCC) are not selective enough for tumor tissue and lead to multidrug resistance and drug toxicity. Although sorafenib (SOR) is the standard first-line systemic therapy approved for the clinical treatment of HCC, its poor aqueous solubility and rapid clearance result in low absorption efficiency and severely limit its use for local treatment. Methods Herein, we present the synthesis of biodegradable polymeric Poly (D, L-Lactide-co-glycolide) (PLGA) particles loaded with SOR (PS) by emulsion-solvent evaporation process. The particles are carefully characterized focusing on particle size, surface charge, morphology, drug loading content, encapsulation efficiency, in vitro stability, drug release behaviour and tested on HepG2 cells. Additionally, PLGA particles have been coupled on side emitting optical fibers (seOF) integrated in a microfluidic device for light-triggered local release. Results PS have a size of 248 nm, tunable surface charge and a uniform and spherical shape without aggregation. PS shows encapsulation efficiency of 89.7% and the highest drug loading (8.9%) between the SOR-loaded PLGA formulations. Treating HepG2 cells with PS containing SOR at 7.5 µM their viability is dampened to 40%, 30% and 17% after 48, 129 and 168 hours of incubation, respectively. Conclusion The high PS stability, their sustained release profile and the rapid cellular uptake corroborate the enhanced cytotoxicity effect on HepG2. With the prospect of developing biomedical tools to control the spatial and temporal release of drugs, we successfully demonstrated the potentiality of seOF for light-triggered local release of the carriers. Our prototypical system paves the way to new devices integrating microfluidics, optical fibers, and advanced carriers capable to deliver minimally invasive locoregional cancer treatments.
Collapse
Affiliation(s)
- Tania Mariastella Caputo
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
| | - Angela Maria Cusano
- CeRICTscrl Regional Center Information Communication Technology, Palazzo Ex Poste, Benevento, Italy
| | - Sofia Principe
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
| | - Paola Cicatiello
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
| | - Giorgia Celetti
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
| | - Anna Aliberti
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
| | - Alberto Micco
- CeRICTscrl Regional Center Information Communication Technology, Palazzo Ex Poste, Benevento, Italy
| | - Menotti Ruvo
- Institute of Biostructure and Bioimaging, National Research Council, Naples, Italy
| | - Maria Tagliamonte
- Innovative Immunological Models Unit, Istituto Nazionale Tumori - IRCCS - “Fond G. Pascale”, Naples, Italy
| | - Concetta Ragone
- Innovative Immunological Models Unit, Istituto Nazionale Tumori - IRCCS - “Fond G. Pascale”, Naples, Italy
| | - Michele Minopoli
- Neoplastic Progression Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Naples, Italy
| | - Maria Vincenza Carriero
- Neoplastic Progression Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Naples, Italy
| | - Luigi Buonaguro
- Innovative Immunological Models Unit, Istituto Nazionale Tumori - IRCCS - “Fond G. Pascale”, Naples, Italy
| | - Andrea Cusano
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
- CeRICTscrl Regional Center Information Communication Technology, Palazzo Ex Poste, Benevento, Italy
| |
Collapse
|
16
|
Li Y, Bi D, Hu Z, Yang Y, Liu Y, Leung WK. Hydrogel-Forming Microneedles with Applications in Oral Diseases Management. MATERIALS (BASEL, SWITZERLAND) 2023; 16:4805. [PMID: 37445119 DOI: 10.3390/ma16134805] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023]
Abstract
Controlled drug delivery in the oral cavity poses challenges such as bacterial contamination, saliva dilution, and inactivation by salivary enzymes upon ingestion. Microneedles offer a location-specific, minimally invasive, and retentive approach. Hydrogel-forming microneedles (HFMs) have emerged for dental diagnostics and therapeutics. HFMs penetrate the stratum corneum, undergo swelling upon contact, secure attachment, and enable sustained transdermal or transmucosal drug delivery. Commonly employed polymers such as polyvinyl alcohol (PVA) and polyvinyl pyrrolidone are crosslinked with tartaric acid or its derivatives while incorporating therapeutic agents. Microneedle patches provide suture-free and painless drug delivery to keratinized or non-keratinized mucosa, facilitating site-specific treatment and patient compliance. This review comprehensively discusses HFMs' applications in dentistry such as local anesthesia, oral ulcer management, periodontal treatment, etc., encompassing animal experiments, clinical trials, and their fundamental impact and limitations, for example, restricted drug carrying capacity and, until now, a low number of dental clinical trial reports. The review explores the advantages and future perspectives of HFMs for oral drug delivery.
Collapse
Affiliation(s)
- Yuqing Li
- Periodontology and Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Duohang Bi
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhekai Hu
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Yanqi Yang
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Yijing Liu
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wai Keung Leung
- Periodontology and Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
17
|
Mohammadi A, Eivazzadeh-Keihan R, Aliabadi HAM, Kashtiaray A, Cohan RA, Bani MS, Komijani S, Etminan A, salehpour N, Maleki A, Mahdavi M. Magnetic carboxymethyl cellulose-silk fibroin hydrogel: a ternary nanobiocomposite exhibiting excellent biological activity and in vitro hyperthermia of cancer therapy. J Biotechnol 2023; 367:71-80. [PMID: 37028560 DOI: 10.1016/j.jbiotec.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
In this work, a magnetic nanobiocomposite scaffold based on carboxymethylcellulose (CMC) hydrogel, silk fibroin (SF), and magnetite nanoparticles was fabricated. The structural properties of this new magnetic nanobiocomposite were characterized by various analyses such as FT-IR, XRD, EDX, FE-SEM, TGA and VSM. According to the particle size histogram, most of the particles were between 55-77nm and the value of saturation magnetization of this nanobiocomposite was reported 41.65emu.g- 1. Hemolysis and MTT tests showed that the designed magnetic nanobiocomposite was compatible with the blood. In addition, the viability percentage of HEK293T normal cells did not change significantly, and the proliferation rate of BT549 cancer cells decreased in its vicinity. EC50 values for HEK293T normal cells after 48h and 72h were 3958 and 2566, respectively. Also, these values for BT549 cancer cells after 48h and 72h were 0.4545 and 0.9967, respectively. The efficiency of fabricated magnetic nanobiocomposite was appraised in a magnetic fluid hyperthermia manner. The specific absorption rate (SAR) of 69W/g (for the 1mg/mL sample at 200kHz) was measured under the alternating magnetic field (AMF).
Collapse
|
18
|
Younis MR, He Y, Yao X, He G, Liu H, Huang P, Lin J. Acidity/carbon dioxide-sensitive triblock polymer-grafted photoactivated vesicles for programmed release of chemotherapeutic drugs against glioblastoma. Acta Biomater 2023; 157:442-450. [PMID: 36470393 DOI: 10.1016/j.actbio.2022.11.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022]
Abstract
Controllable release of chemotherapeutic drugs in tumor sites remains a big challenge for precision therapy. Herein, we developed acidity/carbon dioxide (H+/CO2)-sensitive poly (ethylene glycol)-b-poly (2-(diisopropylamino) ethyl methacrylate)-b-polystyrene triblock polymer (PEG-b-PDPA-b-PS) grafted photoactivated vesicles for programmed release of chemotherapeutic drugs against glioblastoma. In brief, gold nanoparticles (GNPs) were firstly tethered with the H+/CO2-sensitive PEG-b-PDPA-b-PS polymer. Next, the CO2 precursor (ammonium bicarbonate, NH4HCO3) and doxorubicin (DOX) were loaded during self-assembly process of PEG-b-PDPA-b-PS-tethered GNPs, thus obtaining the multifunctional gold vesicles (denoted as GVND). The programmed multi-stimuli responsive drug release by GVND was undergone in multiple steps as follows: 1) the vesicular architecture of GVND was first swelled in tumor acidic microenvironment, 2) the GVND were partially broken under near-infrared (NIR) laser irradiation, 3) the mild hyperthermia generated by GV triggered the thermal decomposition of encapsulated NH4HCO3, leading to the in situ generation of CO2, 4) the generated CO2 reacted with PDPA of PEG-b-PDPA-b-PS, changing the hydrophilicity and hydrophobicity of GVND, thus vastly breaking its vesicular architecture, finally resulting in a "bomb-like" release of DOX in tumor tissues. Such a multi-stimuli responsive programmed drug delivery and mild hyperthermia under NIR laser activation displayed strong antitumor efficacy and completely eradicated U87MG glioblastoma tumor. This work presented a promising strategy to realize precision drug delivery for chemotherapy against glioblastoma. STATEMENT OF SIGNIFICANCE.
Collapse
Affiliation(s)
- Muhammad Rizwan Younis
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Yaling He
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Xikuang Yao
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China; Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, Jiangsu 211816, China
| | - Gang He
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Hengke Liu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China.
| |
Collapse
|
19
|
Jeshvaghani PA, Pourmadadi M, Yazdian F, Rashedi H, Khoshmaram K, Nigjeh MN. Synthesis and characterization of a novel, pH-responsive sustained release nanocarrier using polyethylene glycol, graphene oxide, and natural silk fibroin protein by a green nano emulsification method to enhance cancer treatment. Int J Biol Macromol 2023; 226:1100-1115. [PMID: 36435465 DOI: 10.1016/j.ijbiomac.2022.11.226] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
In this study, for the first time, by employing a simple and efficient double nano-emulsification method and using sweet almond oil as the organic phase, polyethylene glycol (PEG)/graphene oxide (GO)/silk fibroin (SF) hydrogel-nanocomposite was synthesized. The aim of the research was to fabricate a biocompatible targeted pH-sensitive sustained release carrier, improve the drug loading capacity and enhance the anticancer effect of doxorubicin (DOX) drug. The obtained values for the entrapment (%EE) and loading efficacy (%LE) were 87.75 ± 0.7 % and 46 ± 1 %, respectively, and these high values were due to the use of GO with a large specific surface area and the electrostatic interaction between the drug and SF. The Fourier transform infrared spectroscopy (FTIR) and X-ray diffraction (XRD) analyses confirmed the presence of all the components in the nanocomposite and the suitable interaction between them. Based on the results of dynamic light scattering analysis (DLS) and zeta potential analysis, the mean size of the carrier particles and its surface charge were 293.7 nm and -102.9 mV, respectively. The high negative charge was caused by the presence of hydroxyl groups in GO and SF and it caused proper stability of the nanocomposite. The spherical core-shell structure with its homogeneous surface was also observed in the field emission scanning electron microscopy (FE-SEM) image. The cumulative release percentage of the nanocarrier reached 95.75 after 96 h and it is higher in the acidic environment at all times. The results of fitting the release data to the kinetic models suggested that the mechanism of release was dissolution-controlled anomalous at pH 7.4 and diffusion-controlled anomalous at pH 5.4. The results of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay and flow cytometry showed an increase in toxicity on MCF-7 cells and improved apoptotic cell death compared to the free drug. Consequently, the findings of this research introduced and confirmed PEG/GO/SF nanocomposite as an attractive novel drug delivery system for pH-sensitive and sustained delivery of chemotherapeutic agents in biomedicine.
Collapse
Affiliation(s)
| | - Mehrab Pourmadadi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Fatemeh Yazdian
- Department of Life Science Engineering, Faculty of New Science and Technologies, University of Tehran, Tehran, Iran.
| | - Hamid Rashedi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran.
| | - Keyvan Khoshmaram
- Department of Life Science Engineering, Faculty of New Science and Technologies, University of Tehran, Tehran, Iran
| | - Mona Navaei Nigjeh
- Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran; Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
20
|
Kumbhar P, Kolekar K, Khot C, Dabhole S, Salawi A, Sabei FY, Mohite A, Kole K, Mhatre S, Jha NK, Manjappa A, Singh SK, Dua K, Disouza J, Patravale V. Co-crystal nanoarchitectonics as an emerging strategy in attenuating cancer: Fundamentals and applications. J Control Release 2023; 353:1150-1170. [PMID: 36566843 DOI: 10.1016/j.jconrel.2022.12.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Cancer ranks as the second foremost cause of death in various corners of the globe. The clinical uses of assorted anticancer therapeutics have been limited owing to the poor physicochemical attributes, pharmacokinetic performance, and lethal toxicities. Various sorts of co-crystals or nano co-crystals or co-crystals-laden nanocarriers have presented great promise in targeting cancer via improved physicochemical attributes, pharmacokinetic performance, and reduced toxicities. These systems have also demonstrated the controlled cargo release and passive targeting via enhanced permeation and retention (EPR) effect. In addition, regional delivery of co-crystals via inhalation and transdermal route displayed remarkable potential in targeting lung and skin cancer effectively. However, more research is required on the use of co-crystals in cancer and their commercialization. The present review mainly emphasizes co-crystals as emerging avenues in the treatment of various cancers by modulating the physicochemical and pharmacokinetic attributes of approved anticancer therapeutics. The worth of co-crystals in cancer treatment, computational paths in the co-crystals screening, diverse experimental techniques of co-crystals fabrication, and sorts of co-crystals and their noteworthy applications in targeting cancer are also discussed. Besides, the game changer approaches like nano co-crystals and co-crystals-laden nanocarriers, and co-crystals in regional delivery in cancer are also explained with reported case studies. Furthermore, regulatory directives for pharmaceutical co-crystals and their scale-up, and challenges are also highlighted with concluding remarks and future initiatives. In essence, co-crystals and nano co-crystals emerge to be a promising strategy in overwhelming cancers through improving anticancer efficacy, safety, patient compliance, and reducing the cost.
Collapse
Affiliation(s)
- Popat Kumbhar
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur, Maharashtra 416113, India
| | - Kaustubh Kolekar
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur, Maharashtra 416113, India
| | - Chinmayee Khot
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur, Maharashtra 416113, India
| | - Swati Dabhole
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur, Maharashtra 416113, India
| | - Ahmad Salawi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Fahad Y Sabei
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Akshay Mohite
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur, Maharashtra 416113, India
| | - Kapil Kole
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur, Maharashtra 416113, India
| | - Susmit Mhatre
- Department of Pharmacy Sciences, School of Pharmacy and Health Professionals, Creighton University, Omaha, NE 68178, USA
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, 201310, Uttar Pradesh, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India
| | - Arehalli Manjappa
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur, Maharashtra 416113, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun 248007, India
| | - John Disouza
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur, Maharashtra 416113, India.
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, Maharashtra 400019, India.
| |
Collapse
|
21
|
Bressler EM, Chu NQ, Sabatelle RC, Mahvi DA, Korunes-Miller JT, Nagashima F, Ichinose F, Liu R, Grinstaff MW, Colson YL, Raut CP. Doxorubicin-Loaded Polymeric Meshes Prevent Local Recurrence after Sarcoma Resection While Avoiding Cardiotoxicity. Cancer Res 2022; 82:4474-4484. [PMID: 36169924 PMCID: PMC9948765 DOI: 10.1158/0008-5472.can-22-0734] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/04/2022] [Accepted: 09/23/2022] [Indexed: 01/27/2023]
Abstract
Surgery is the only potentially curative treatment for localized soft-tissue sarcomas. However, for sarcomas arising in the retroperitoneum, locoregional recurrence rates are 35% to 59% despite resection. Doxorubicin (DOX) is the standard first-line systemic chemotherapy for advanced soft-tissue sarcoma, yet its intravenous administration yields limited clinical efficacy and results in dose-limiting cardiotoxicity. We report the fabrication and optimization of a novel electrospun poly(caprolactone) (PCL) surgical mesh coated with layers of a hydrophobic polymer (poly(glycerol monostearate-co-caprolactone), PGC-C18), which delivers DOX directly to the operative bed following sarcoma resection. In xenograft models of liposarcoma and chondrosarcoma, DOX-loaded meshes (DoM) increased overall survival 4-fold compared with systemically administered DOX and prevented local recurrence in all but one animal. Importantly, mice implanted with DoMs exhibited preserved cardiac function, whereas mice receiving an equivalent dose systemically displayed a 23% decrease from baseline in both cardiac output and ejection fraction 20 days after administration. Collectively, this work demonstrates a feasible therapeutic approach to simultaneously prevent post-surgical tumor recurrence and minimize cardiotoxicity in soft-tissue sarcoma. SIGNIFICANCE A proof-of-principle study in animal models shows that a novel local drug delivery approach can prevent tumor recurrence as well as drug-related adverse events following surgical resection of soft-tissue sarcomas.
Collapse
Affiliation(s)
- Eric M. Bressler
- Department of Biomedical Engineering, Boston University, Boston, MA 02114
| | - Ngoc-Quynh Chu
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
| | | | - David A. Mahvi
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
| | | | - Fumiaki Nagashima
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
| | - Rong Liu
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
| | - Mark W. Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02114,Department of Chemistry, Boston University, Boston, MA 02114,Co-corresponding authors Mark W. Grinstaff, Room 519, 590 Commonwealth Ave, Boston MA, Boston, MA 02215, Tel: 718-358-3429, ; Yolonda L. Colson, Massachusetts General Hospital, 55 Fruit Street, Founders 7, Boston, MA 02114, Office: 617-726-5600, ; Chandrajit P. Raut, Brigham and Women's Hospital · , 75 Francis St, Boston, MA 02115, Tel: 617-632-5982,
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115,Co-corresponding authors Mark W. Grinstaff, Room 519, 590 Commonwealth Ave, Boston MA, Boston, MA 02215, Tel: 718-358-3429, ; Yolonda L. Colson, Massachusetts General Hospital, 55 Fruit Street, Founders 7, Boston, MA 02114, Office: 617-726-5600, ; Chandrajit P. Raut, Brigham and Women's Hospital · , 75 Francis St, Boston, MA 02115, Tel: 617-632-5982,
| | - Chandrajit P. Raut
- Department of Surgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115,Co-corresponding authors Mark W. Grinstaff, Room 519, 590 Commonwealth Ave, Boston MA, Boston, MA 02215, Tel: 718-358-3429, ; Yolonda L. Colson, Massachusetts General Hospital, 55 Fruit Street, Founders 7, Boston, MA 02114, Office: 617-726-5600, ; Chandrajit P. Raut, Brigham and Women's Hospital · , 75 Francis St, Boston, MA 02115, Tel: 617-632-5982,
| |
Collapse
|
22
|
Najibi AJ, Larkin K, Feng Z, Jeffreys N, Dacus MT, Rustagi Y, Hodi FS, Mooney DJ. Chemotherapy Dose Shapes the Expression of Immune-Interacting Markers on Cancer Cells. Cell Mol Bioeng 2022; 15:535-551. [PMID: 36531864 PMCID: PMC9751245 DOI: 10.1007/s12195-022-00742-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/21/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction Tumor and immune cells interact through a variety of cell-surface proteins that can either restrain or promote tumor progression. The impacts of cytotoxic chemotherapy dose and delivery route on this interaction profile remain incompletely understood, and could support the development of more effective combination therapies for cancer treatment. Methods and Results Here, we found that exposure to the anthracycline doxorubicin altered the expression of numerous immune-interacting markers (MHC-I, PD-L1, PD-L2, CD47, Fas, and calreticulin) on live melanoma, breast cancer, and leukemia cells in a dose-dependent manner in vitro. Notably, an intermediate dose best induced immunogenic cell death and the expression of immune-activating markers without maximizing expression of markers associated with immune suppression. Bone marrow-derived dendritic cells exposed to ovalbumin-expressing melanoma treated with intermediate doxorubicin dose became activated and best presented tumor antigen. In a murine melanoma model, both the doxorubicin dose and delivery location (systemic infusion versus local administration) affected the expression of these markers on live tumor cells. Particularly, local release of doxorubicin from a hydrogel increased calreticulin expression on tumor cells without inducing immune-suppressive markers, in a manner dependent on the loaded dose. Doxorubicin exposure also altered the expression of immune-interacting markers in patient-derived melanoma cells. Conclusions Together, these results illustrate how standard-of-care chemotherapy, when administered in various manners, can lead to distinct expression of immunogenic markers on cancer cells. These findings may inform development of chemo-immunotherapy combinations for cancer treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-022-00742-y.
Collapse
Affiliation(s)
- Alexander J. Najibi
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| | - Kerry Larkin
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| | - Zhaoqianqi Feng
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| | - Nicholas Jeffreys
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| | - Mason T. Dacus
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| | - Yashika Rustagi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215 USA
| | - F. Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215 USA
| | - David J. Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115 USA
| |
Collapse
|
23
|
Kong X, Feng M, Wu L, He Y, Mao H, Gu Z. Biodegradable gemcitabine-loaded microdevice with sustained local drug delivery and improved tumor recurrence inhibition abilities for postoperative pancreatic tumor treatment. Drug Deliv 2022; 29:1595-1607. [PMID: 35612309 PMCID: PMC9176693 DOI: 10.1080/10717544.2022.2075984] [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: 02/22/2022] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 11/05/2022] Open
Abstract
At present, the 10-year survival rate of patients with pancreatic cancer is still less than 4%, mainly due to the high cancer recurrence rate caused by incomplete surgery and lack of effective postoperative adjuvant treatment. Systemic chemotherapy remains the only choice for patients after surgery; however, it is accompanied by off-target effects and server systemic toxicity. Herein, we proposed a biodegradable microdevice for local sustained drug delivery and postoperative pancreatic cancer treatment as an alternative and safe option. Biodegradable poly(l-lactic-co-glycolic acid) (P(L)LGA) was developed as the matrix material, gemcitabine hydrochloride (GEM·HCl) was chosen as the therapeutic drug and polyethylene glycol (PEG) was employed as the drug release-controlled regulator. Through adjusting the amount and molecular weight of PEG, the controllable degradation of matrix and the sustained release of GEM·HCl were obtained, thus overcoming the unstable drug release properties of traditional microdevices. The drug release mechanism of microdevice and the regulating action of PEG were studied in detail. More importantly, in the treatment of the postoperative recurrence model of subcutaneous pancreatic tumor in mice, the microdevice showed effective inhibition of postoperative in situ recurrences of pancreatic tumors with excellent biosafety and minimum systemic toxicity. The microdevice developed in this study provides an option for postoperative adjuvant pancreatic treatment, and greatly broadens the application prospects of traditional chemotherapy drugs.
Collapse
Affiliation(s)
- Xiangming Kong
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
| | - Miao Feng
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
| | - Lihuang Wu
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
| | - Yiyan He
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
- NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing, PR China
- Suqian Advanced Materials Industry Technology Innovation Center of Nanjing Tech University, Nanjing, PR China
| | - Hongli Mao
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
- NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing, PR China
- Suqian Advanced Materials Industry Technology Innovation Center of Nanjing Tech University, Nanjing, PR China
| | - Zhongwei Gu
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
- NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing, PR China
- Suqian Advanced Materials Industry Technology Innovation Center of Nanjing Tech University, Nanjing, PR China
| |
Collapse
|
24
|
Paca AM, Singh M, Ajibade PA. Synthesis, characterization and in vitro anticancer studies of Ru(III) dithiocarbamate complexes. J COORD CHEM 2022. [DOI: 10.1080/00958972.2022.2145472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Athandwe M. Paca
- School of Chemistry and Physics, University of KwaZulu-Natal, Scottsville, South Africa
| | - Moganavelli Singh
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Peter A. Ajibade
- School of Chemistry and Physics, University of KwaZulu-Natal, Scottsville, South Africa
| |
Collapse
|
25
|
Drug-device systems based on biodegradable metals for bone applications: Potential, development and challenges. Biocybern Biomed Eng 2022. [DOI: 10.1016/j.bbe.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
26
|
Krukiewicz K, Kazek-Kęsik A, Brzychczy-Włoch M, Łos MJ, Ateba CN, Mehrbod P, Ghavami S, Shyntum DY. Recent Advances in the Control of Clinically Important Biofilms. Int J Mol Sci 2022; 23:9526. [PMID: 36076921 PMCID: PMC9455909 DOI: 10.3390/ijms23179526] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Biofilms are complex structures formed by bacteria, fungi, or even viruses on biotic and abiotic surfaces, and they can be found in almost any part of the human body. The prevalence of biofilm-associated diseases has increased in recent years, mainly because of the frequent use of indwelling medical devices that create opportunities for clinically important bacteria and fungi to form biofilms either on the device or on the neighboring tissues. As a result of their resistance to antibiotics and host immunity factors, biofilms have been associated with the development or persistence of several clinically important diseases. The inability to completely eradicate biofilms drastically increases the burden of disease on both the patient and the healthcare system. Therefore, it is crucial to develop innovative ways to tackle the growth and development of biofilms. This review focuses on dental- and implant-associated biofilm infections, their prevalence in humans, and potential therapeutic intervention strategies, including the recent advances in pharmacology and biomedical engineering. It lists current strategies used to control the formation of clinically important biofilms, including novel antibiotics and their carriers, antiseptics and disinfectants, small molecule anti-biofilm agents, surface treatment strategies, and nanostructure functionalization, as well as multifunctional coatings particularly suitable for providing antibacterial effects to the surface of implants, to treat either dental- or implant-related bacterial infections.
Collapse
Affiliation(s)
- Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
- Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Konarskiego 22B, 44-100 Gliwice, Poland
| | - Alicja Kazek-Kęsik
- Department of Inorganic Chemistry, Analytical Chemistry and Electrochemistry, Silesian University of Technology, 44-100 Gliwice, Poland
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8 Street, 44-100 Gliwice, Poland
| | - Monika Brzychczy-Włoch
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Krakow, Poland
| | - Marek J. Łos
- Department of Pathology, Pomeranian Medical University, 71-344 Szczecin, Poland
| | - Collins Njie Ateba
- Food Security and Safety Niche Area, North West University, Private Bag X2046, Mahikeng 2735, South Africa
| | - Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Saeid Ghavami
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland
- Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 3P5, Canada
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada
| | - Divine Yufetar Shyntum
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8 Street, 44-100 Gliwice, Poland
| |
Collapse
|
27
|
Mamalis D, Panagiotopoulou A, Couladouros EA, Tzeli D, Vidali VP. A DFT Study towards the Amide cis‐trans Isomerization Process of the Myc‐Max Inhibitor Mycro 3 and Its Photophysical Properties; Synthesis and NMR Studies of the trans‐Conformation. ChemistrySelect 2022. [DOI: 10.1002/slct.202201639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Dimitrios Mamalis
- Laboratory of Physical Chemistry Department of Chemistry National and Kapodistrian University of Athens Panepistimiopolis Athens 157 71 Greece
- Institute of Nanoscience and Nanotechnology National Center for Scientific Research “Demokritos” Ag. Paraskevi 153 41 Athens Greece
- Theoretical and Physical Chemistry Institute National Hellenic Research Foundation, 48 Vassileos Constantinou Ave. Athens 116 35 Greece
| | - Angeliki Panagiotopoulou
- Institute of Biosciences and Applications National Center for Scientific Research “Demokritos”, Ag. Paraskevi 153 41 Athens Greece
| | - Elias A. Couladouros
- Agricultural University of Athens Department of Food Science and Human Nutrition Iera Odos 75 Athens 118 55 Greece
| | - Demeter Tzeli
- Laboratory of Physical Chemistry Department of Chemistry National and Kapodistrian University of Athens Panepistimiopolis Athens 157 71 Greece
- Theoretical and Physical Chemistry Institute National Hellenic Research Foundation, 48 Vassileos Constantinou Ave. Athens 116 35 Greece
| | - Veroniki P. Vidali
- Institute of Nanoscience and Nanotechnology National Center for Scientific Research “Demokritos” Ag. Paraskevi 153 41 Athens Greece
| |
Collapse
|
28
|
Özcan Bülbül E, Üstündağ Okur N, Mısırlı D, Cevher E, Tsanaktsis V, Bingöl Özakpınar Ö, Siafaka PI. Applying quality by design approach for the determination of potent paclitaxel loaded poly(lactic acid) based implants for localized tumor drug delivery. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2067538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Ece Özcan Bülbül
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istinye University, Istanbul, Turkey
| | - Neslihan Üstündağ Okur
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Health Sciences, Istanbul, Turkey
| | - Duygu Mısırlı
- Department of Biochemistry, Faculty of Pharmacy, University of Health Sciences, Istanbul, Turkey
| | - Erdal Cevher
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, Istanbul, Turkey
| | - Vasilios Tsanaktsis
- Faculty of Sciences, School of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Panoraia I. Siafaka
- School of Health Studies, KES College, Nicosia, Cyprus
- Faculty of Pharmacy, European University Cyprus, Nicosia, Cyprus
| |
Collapse
|
29
|
|
30
|
Janardhanam LSL, Bandi SP, Venuganti VVK. Functionalized LbL Film for Localized Delivery of STAT3 siRNA and Oxaliplatin Combination to Treat Colon Cancer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:10030-10046. [PMID: 35170934 DOI: 10.1021/acsami.1c22166] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The aim of the study was to develop and evaluate the efficacy of a functionalized layer-by-layer (LbL) assembled film entrapped with oxaliplatin (OX) and signal transducer and activator of transcription 3 (STAT3) siRNA in the localized treatment of colon cancer. The LbL film was prepared by the sequential layering of chitosan (CS) and alginate to attain desired physical and mechanical properties. The film was functionalized by coating folic acid-conjugated CS on one side. On the other side, polycaprolactone was coated as a backing layer to provide directional drug release. OX was entrapped within the layers of the film, while STAT3 siRNA was complexed with CS to form nanoparticles before entrapment in the LbL film. The CS-siRNA nanoparticles were taken up by the colon carcinoma, Caco-2 cells within 3 h and provided concentration-dependent reduction in STAT3 protein expression. The functionalized LbL film (F-LbL film) selectively adhered to the colon cancer tissue in the mice model, whereas the nonfunctionalized film adhered to the normal colon tissue. The combination of OX and STAT3 siRNA provided significantly greater tumor regression, survival rate, and STAT3 protein suppression after localized delivery through oral administration compared with intravenous administration. Taken together, the F-LbL film can selectively bind to colon tumors for localized delivery of drugs to treat colon cancer.
Collapse
Affiliation(s)
- Leela Sai Lokesh Janardhanam
- Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Hyderabad 500078, Telangana State, India
| | - Sony Priyanka Bandi
- Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Hyderabad 500078, Telangana State, India
| | | |
Collapse
|
31
|
Karbakhshzadeh A, Derakhshande M, Farhami N, Hosseinian A, Ebrahimiasl S, Ebadi A. Study the Adsorption of Letrozole Drug on the Silicon Doped Graphdiyne Monolayer: a DFT Investigation. SILICON 2022. [PMCID: PMC8109220 DOI: 10.1007/s12633-021-01143-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
In the current study, by employing first-principles computations, the adsorption behavior of letrozole (LET) was investigated on the pristine graphdiyne nanosheet (GDY) as well as Si-doped graphdiyne (SiGDY). According to the adsorption energy, charge transfer value, and the change in the bang gap energy, the tendency of the pristine GDY towards LET is insignificant. However, the interaction of LET with SiGDY was strong and the adsorption energy was approximately − 19.20 kcal/mol. In addition, the associated electrical conductivity with SiGDY increased by approximately 23.53 % following the adsorption of LET. The results show that SiGDY can be employed as an electronic sensor to detect LET. Furthermore, LET is detected by SiGDY in the water phase based on the magnitude of solvation energy. Finally, a considerable charge-transfer between LET and SiGDY is a precondition for the adsorption of the LET molecule with proper binding energies, which delivers the Si atoms with a significant positive charge.
Collapse
Affiliation(s)
| | - Maryam Derakhshande
- Department of Chemistry, Faculty of Chemical Engineering, Islamic Azad University, Mahshahr Branch, Mahshahr, Iran
| | - Nabieh Farhami
- Department of Chemistry, Faculty of Chemical Engineering, Islamic Azad University, Mahshahr Branch, Mahshahr, Iran
| | - Akram Hosseinian
- School of Engineering Science, College of Engineering, University of Tehran, P. O. Box 11365-4563, Tehran, Iran
| | - Saeideh Ebrahimiasl
- Department of Chemistry, Ahar Branch, Islamic Azad University, Ahar, Iran
- Industrial Nanotechnology Research Center, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Abdolghaffar Ebadi
- Department of Agriculture, Jouybar Branch, Islamic Azad University, Jouybar, Iran
| |
Collapse
|
32
|
Liu J, Tian B, Liu Y, Wan JB. Cyclodextrin-Containing Hydrogels: A Review of Preparation Method, Drug Delivery, and Degradation Behavior. Int J Mol Sci 2021; 22:13516. [PMID: 34948312 PMCID: PMC8703588 DOI: 10.3390/ijms222413516] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/06/2021] [Accepted: 12/14/2021] [Indexed: 02/01/2023] Open
Abstract
Hydrogels possess porous structures, which are widely applied in the field of materials and biomedicine. As a natural oligosaccharide, cyclodextrin (CD) has shown remarkable application prospects in the synthesis and utilization of hydrogels. CD can be incorporated into hydrogels to form chemically or physically cross-linked networks. Furthermore, the unique cavity structure of CD makes it an ideal vehicle for the delivery of active ingredients into target tissues. This review describes useful methods to prepare CD-containing hydrogels. In addition, the potential biomedical applications of CD-containing hydrogels are reviewed. The release and degradation process of CD-containing hydrogels under different conditions are discussed. Finally, the current challenges and future research directions on CD-containing hydrogels are presented.
Collapse
Affiliation(s)
- Jiayue Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China;
| | - Bingren Tian
- School of Chemical Engineering and Technology, Xinjiang University, Urumqi 830046, China;
| | - Yumei Liu
- School of Chemical Engineering and Technology, Xinjiang University, Urumqi 830046, China;
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China;
| |
Collapse
|
33
|
Ahmad AM, Mohammed HA, Faris TM, Hassan AS, Mohamed HB, El Dosoky MI, Aboubakr EM. Nano-Structured Lipid Carrier-Based Oral Glutathione Formulation Mediates Renoprotection against Cyclophosphamide-Induced Nephrotoxicity, and Improves Oral Bioavailability of Glutathione Confirmed through RP-HPLC Micellar Liquid Chromatography. Molecules 2021; 26:7491. [PMID: 34946570 PMCID: PMC8706828 DOI: 10.3390/molecules26247491] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 12/28/2022] Open
Abstract
The study aimed to develop a new glutathione (GSH) oral formulation to enhance the delivery of GSH and counter the nephrotoxicity of the anticancer drug, cyclophosphamide (CP). A nanostructured lipid carrier glutathione formulation (GSH-NLCs) composed of glutathione (500 mg), stearic and oleic acid (300 mg, each), and Tween® 80 (2%, w/v) was prepared through the emulsification-solvent-evaporation technique, which exhibited a 452.4 ± 33.19 nm spheroidal-sized particulate material with narrow particle size distributions, -38.5 ± 1.4 mV zeta potential, and an entrapment efficiency of 79.8 ± 1.9%. The GSH formulation was orally delivered, and biologically tested to ameliorate the CP-induced renal toxicity in a rat model. Detailed renal morphology, before and after the GSH-NLCs administration, including the histopathological examinations, confirmed the ameliorating effects of the prepared glutathione formulation together with its safe oral delivery. CP-induced oxidative stress, superoxide dismutase depletion, elevation of malondialdehyde levels, depletion of Bcl-2 concentration levels, and upregulated NF-KB levels were observed and were controlled within the recommended and near normal/control levels. Additionally, the inflammatory mediator marker, IL-1β, serum levels were marginally normalized by delivery of the GHS-NLCs formulation. Oral administration of the pure glutathione did not exhibit any ameliorating effects on the renal tissues, which suggested that the pure glutathione is reactive and is chemically transformed during the oral delivery, which affected its pharmacological action at the renal site. The protective effects of the GSH-NLCs formulation through its antioxidant and anti-inflammatory effects suggested its prominent role in containing CP-induced renal toxicity and renal tissue damage, together with the possibility of administrating higher doses of the anticancer drug, cyclophosphamide, to achieve higher and effective anticancer action in combination with the GSH-NLCs formulation.
Collapse
Affiliation(s)
- Adel M. Ahmad
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt
| | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo 11371, Egypt
| | - Tarek M. Faris
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Al-Azhar University, Cairo 11371, Egypt;
| | - Abeer S. Hassan
- Department of Pharmaceutics, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt; (A.S.H.); (H.B.M.)
| | - Hebatallah B. Mohamed
- Department of Pharmaceutics, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt; (A.S.H.); (H.B.M.)
| | - Mahmoud I. El Dosoky
- Department of Pathology, Faculty of Medicine, South Valley University, Qena 83523, Egypt;
| | - Esam M. Aboubakr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt;
| |
Collapse
|
34
|
Pérez Quiñones J, Roschger C, Zierer A, Peniche-Covas C, Brüggemann O. Self-Assembled Silk Fibroin-Based Aggregates for Delivery of Camptothecin. Polymers (Basel) 2021; 13:polym13213804. [PMID: 34771362 PMCID: PMC8587969 DOI: 10.3390/polym13213804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/24/2022] Open
Abstract
A water-soluble hydrolysate of silk fibroin (SF) (~30 kDa) was esterified with tocopherol, ergocalciferol, and testosterone to form SF aggregates for the controlled delivery of the anticancer drug camptothecin (CPT). Elemental analysis and 1H NMR spectroscopy showed a degree of substitution (DS) on SF of 0.4 to 3.8 mol %. Yields of 58 to 71% on vitamins- and testosterone-grafted SF conjugates were achieved. CPT was efficiently incorporated into the lipophilic core of SF aggregates using a dialysis-precipitation method, achieving drug contents of 6.3-8.5 wt %. FTIR spectra and DSC thermograms showed that tocopherol- and testosterone-grafted SF conjugates predominantly adopted a β-sheet conformation. After the esterification of tyrosine residues on SF chains with the vitamin or testosterone, the hydrodynamic diameters almost doubled or tripled that of SF. The zeta potential values after esterification increased to about -30 mV, which favors the stability of aggregates in aqueous medium. Controlled and almost quantitative release of CPT was achieved after 6 days in PBS at 37 °C, with almost linear release during the first 8 h. MCF-7 cancer cells exhibited good uptake of CPT-loaded SF aggregates after 6 h, causing cell death and cell cycle arrest in the G2/M phase. Substantial uptake of the CPT-loaded aggregates into MCF-7 spheroids was shown after 3 days. Furthermore, all CPT-loaded SF aggregates demonstrated superior toxicity to MCF-7 spheroids compared with parent CPT. Blank SF aggregates induced no hemolysis at pH 6.2 and 7.4, while CPT-loaded SF aggregates provoked hemolysis at pH 6.2 but not at pH 7.4. In contrast, parent CPT caused hemolysis at both pH tested. Therefore, CPT-loaded SF aggregates are promising candidates for chemotherapy.
Collapse
Affiliation(s)
- Javier Pérez Quiñones
- Institute of Polymer Chemistry, Johannes Kepler University Linz, Altenberger Straße 69, 4040 Linz, Austria;
- Correspondence: or ; Tel.: +43-670-4039820
| | - Cornelia Roschger
- Department for Cardiac-, Vascular- and Thoracic Surgery, Johannes Kepler University Linz, Kepler University Hospital GmBH, Altenberger Straße 69, 4040 Linz and Krankenhausstraße 7a, 4020 Linz, Austria; (C.R.); or (A.Z.)
| | - Andreas Zierer
- Department for Cardiac-, Vascular- and Thoracic Surgery, Johannes Kepler University Linz, Kepler University Hospital GmBH, Altenberger Straße 69, 4040 Linz and Krankenhausstraße 7a, 4020 Linz, Austria; (C.R.); or (A.Z.)
| | - Carlos Peniche-Covas
- Facultad de Química, Universidad de La Habana, Zapata S/N entre G y Carlitos Aguirre, La Habana 10400, Cuba;
| | - Oliver Brüggemann
- Institute of Polymer Chemistry, Johannes Kepler University Linz, Altenberger Straße 69, 4040 Linz, Austria;
| |
Collapse
|
35
|
Single-Shot Local Injection of Microfragmented Fat Tissue Loaded with Paclitaxel Induces Potent Growth Inhibition of Hepatocellular Carcinoma in Nude Mice. Cancers (Basel) 2021; 13:cancers13215505. [PMID: 34771667 PMCID: PMC8583409 DOI: 10.3390/cancers13215505] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is poorly beneficiated by intravenous chemotherapy due to inadequate availability of drugs at the tumor site. We previously demonstrated that human micro-fragmented adipose tissue (MFAT) and its devitalized counterpart (DMFAT) could be effective natural scaffolds to deliver Paclitaxel (PTX) to tumors in both in vitro and in vivo tests, affecting cancer growth relapse. Here we tested the efficacy of DMFAT-PTX in a well-established HCC in nude mice. MFAT-PTX and DMFAT-PTX preparations were tested for anti-cancer activity in 2D and 3D assays using Hep-3B tumor cells. The efficacy of DMFAT-PTX was evaluated after a single-shot subcutaneous injection near a Hep-3B growing tumor by assessing tumor volumes, apoptosis rate, and drug pharmacokinetics in an in vivo model. Potent antiproliferative activity was seen in both in vitro 2D and 3D tests. Mice treated with DMFAT-PTX (10 mg/kg) produced potent Hep-3B growth inhibition with 33% complete tumor regressions. All treated animals experienced tumor ulceration at the site of DMFAT-PTX injection, which healed spontaneously. Lowering the drug concentration (5 mg/kg) prevented the formation of ulcers, maintaining statistically significant efficacy. Histology revealed a higher number of apoptotic cancer cells intratumorally, suggesting prolonged presence of PTX that was confirmed by the pharmacokinetic analysis. DMFAT may be a potent and valid new tool for local chemotherapy of HCC in an advanced stage of progression, also suggesting potential effectiveness in other human primary inoperable cancers.
Collapse
|
36
|
Moku G, Vangala S, Yakati V, Gali CC, Saha S, Madamsetty VS, Vyas A. Novel Suberoylanilide Hydroxamic Acid Analogs Inhibit Angiogenesis and Induce Apoptosis in Breast Cancer Cells. Anticancer Agents Med Chem 2021; 22:914-925. [PMID: 34488592 DOI: 10.2174/1871520621666210901102425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/31/2021] [Accepted: 06/05/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Histone deacetylases (HDACs) are the enzymes that catalyze the removal of the acetyl group from lysine residues and regulate several biological processes. Suberoylanilide hydroxamic acid (SAHA) is a notable HDAC inhibitor that exhibited remarkable anti-proliferative efficiency by alleviating gene regulation against solid and hematologic cancers. AIM The aim of this study was to develop new chemotherapeutic agents for breast cancer treatment, therefore, a novel series of Suberoylanilide hydroxamic acid (SAHA) analogs were investigated as anticancer agents. METHODS We designed and synthesized a novel series of analogs derived from SAHA by substituting alkyl, alkoxy, halo, and benzyl groups at different positions of the phenyl ring. The newly synthesized analogs were assessed for their cytotoxic potential against four human cancer cell lines in comparison with healthy cell lines, using several biological assays. RESULTS SAHA analogs displayed significant cytotoxic potential with IC50 values ranging from 1.6 to 19.2 µM in various tumor cell lines. Among these analogs, 2d (containing 3-chloro, 4-floro substitutions on phenyl moiety), 2h (containing 3,4-di chloro substitutions on phenyl moiety), and 2j (containing 4-chloro, 3-methyl substitutions on phenyl moiety) showed significant cytotoxic potential with IC50 values ranging from 1.6 to 1.8 µM in MCF-7 (breast carcinoma) cell line. More importantly, these analogs were found to be non-toxic towards healthy primary human hepatocytes (PHH) and mouse fibroblast cells (NIH3T3), which represent their tumor selectivity. These analogs were further analyzed for their effect on cell migration, BrdU incorporation, Annexin V-FITC and cell cycle arrest (Sub-G1 phase). Remarkably, analogs 2d, 2h, and 2j displayed significant HDAC inhibition than the parent SAHA molecule. Further studies also confirmed that these SAHA analogs are efficient in inducing apoptosis, as they regulated the expression of several proteins involved in mitochondrial or intrinsic apoptosis pathways. Findings in the Chick Chorioallantoic Membrane (CAM) assay studies revealed anti-angiogenic properties of the currently described SAHA analogs. CONCLUSION From anti-proliferative study results, it is clearly evident that 3,4-substitution at the SAHA phenyl ring improves the anti-proliferative activity of SAHA. Based on these findings, we presume that the synthesized novel SAHA analogs could be potential therapeutic agents in treating breast cancer.
Collapse
Affiliation(s)
- Gopikrishna Moku
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Uppal Road, Hyderabad 500 007. India
| | - Swathi Vangala
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Uppal Road, Hyderabad 500 007. India
| | - Venu Yakati
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Uppal Road, Hyderabad 500 007. India
| | - Chaitanya Chakravarthi Gali
- Institute of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz 8032. Austria
| | - Soumen Saha
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Uppal Road, Hyderabad 500 007. India
| | - Vijay Sagar Madamsetty
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Uppal Road, Hyderabad 500 007. India
| | - Amber Vyas
- Department of Pharmaceutics, University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492 010. India
| |
Collapse
|
37
|
Narayanan M, Deepika M, Ma Y, Nasif O, Alharbi SA, Srinivasan R, Natarajan D. Phyto-fabrication, characterization, and biomedical activity of silver nanoparticles mediated from an epiphytic plant Luisia tenuifolia Blume. APPLIED NANOSCIENCE 2021. [DOI: 10.1007/s13204-021-02022-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
38
|
Caputo T, Cusano AM, Ruvo M, Aliberti A, Cusano A. Human Serum Albumin Nanoparticles as a Carrier for On-Demand Sorafenib Delivery. Curr Pharm Biotechnol 2021; 23:1214-1225. [PMID: 34445947 DOI: 10.2174/1389201022666210826152311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/04/2021] [Accepted: 04/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Drug delivery systems based on Human Serum Albumin (HSA) have been widely investigated due to their capability to interact with several molecules together with their nontoxicity, non-immunogenicity and biocompatibility. Sorafenib (SOR) is a kinase inhibitor used as the first-line treatment in hepatic cancer. However, because of its several intrinsic drawbacks (low solubility and bioavailability), there is a growing need for discovering new carriers able to overcome the current limitations. OBJECTIVE To study HSA particles loaded with SOR as a thermal responsive drug delivery system. METHOD A detailed spectroscopy analysis of the HSA and SOR interaction in solution was carried out in order to characterize the temperature dependence of the complex. Based on this study, the synthesis of HSA particles loaded with SOR was optimized. Particles were characterized by Dynamic Light Scattering, Atomic Force Microscopy and by spectrofluorometer. Encapsulation efficiency and in vitro drug release were quantified by RP-HPLC. RESULTS HSA particles were monodispersed in size (≈ 200 nm); encapsulation efficiency ranged from 25% to 58%. Drug release studies that were performed at 37 °C and 50 °C showed that HS5 particles achieved a drug release of 0.430 µM in 72 hours at 50 °C in PBS buffer, accomplishing a 4.6-fold overall SOR release enhancement following a temperature increase from 37 °C to 50 °C. CONCLUSION The system herein presented has the potential to exert a therapeutic action (in the nM range) triggering a sustained temperature-controllable release of relevant drugs.
Collapse
Affiliation(s)
- Tania Caputo
- CeRICT scrl Regional Center Information Communication Technology, Benevento. Italy
| | - Angela Maria Cusano
- CeRICT scrl Regional Center Information Communication Technology, Benevento. Italy
| | - Menotti Ruvo
- Institute of Biostructure and Bioimaging, National Research Council, I-80134, Napoli. Italy
| | - Anna Aliberti
- Optoelectronics Group, Department of Engineering, University of Sannio, I-82100, Benevento. Italy
| | - Andrea Cusano
- CeRICT scrl Regional Center Information Communication Technology, Benevento. Italy
| |
Collapse
|
39
|
Taheri-Ledari R, Zhang W, Radmanesh M, Cathcart N, Maleki A, Kitaev V. Plasmonic photothermal release of docetaxel by gold nanoparticles incorporated onto halloysite nanotubes with conjugated 2D8-E3 antibodies for selective cancer therapy. J Nanobiotechnology 2021; 19:239. [PMID: 34380469 PMCID: PMC8359560 DOI: 10.1186/s12951-021-00982-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/28/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Applied nanomaterials in targeted drug delivery have received increased attention due to tangible advantages, including enhanced cell adhesion and internalization, controlled targeted release, convenient detection in the body, enhanced biodegradation, etc. Furthermore, conjugation of the biologically active ingredients with the drug-containing nanocarriers (nanobioconjugates) has realized impressive opportunities in targeted therapy. Among diverse nanostructures, halloysite nanotubes (NHTs) with a rolled multilayer structure offer great possibilities for drug encapsulation and controlled release. The presence of a strong hydrogen bond network between the rolled HNT layers enables the controlled release of the encapsulated drug molecules through the modulation of hydrogen bonding either in acidic conditions or at higher temperatures. The latter can be conveniently achieved through the photothermal effect via the incorporation of plasmonic nanoparticles. RESULTS The developed nanotherapeutic integrated natural halloysite nanotubes (HNTs) as a carrier; gold nanoparticles (AuNPs) for selective release; docetaxel (DTX) as a cytotoxic anticancer agent; human IgG1 sortilin 2D8-E3 monoclonal antibody (SORT) for selective targeting; and 3-chloropropyltrimethoxysilane as a linker for antibody attachment that also enhances the hydrophobicity of DTX@HNT/Au-SORT and minimizes DTX leaching in body's internal environment. HNTs efficiently store DTX at room temperature and release it at higher temperatures via disruption of interlayer hydrogen bonding. The role of the physical expansion and disruption of the interlayer hydrogen bonding in HNTs for the controlled DTX release has been studied by dynamic light scattering (DLS), electron microscopy (EM), and differential scanning calorimetry (DSC) at different pH conditions. HNT interlayer bond disruption has been confirmed to take place at a much lower temperature (44 °C) at low pH vs. 88 °C, at neutral pH thus enabling the effective drug release by DTX@HNT/Au-SORT through plasmonic photothermal therapy (PPTT) by light interaction with localized plasmon resonance (LSPR) of AuNPs incorporated into the HNT pores. CONCLUSIONS Selective ovarian tumor targeting was accomplished, demonstrating practical efficiency of the designed nanocomposite therapeutic, DTX@HNT/Au-SORT. The antitumor activity of DTX@HNT/Au-SORT (apoptosis of 90 ± 0.3%) was confirmed by in vitro experiments using a caov-4 (ATCC HTB76) cell line (sortilin expression > 70%) that was successfully targeted by the sortilin 2D8-E3 mAb, tagged on the DTX@HNT/Au.
Collapse
Affiliation(s)
- Reza Taheri-Ledari
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Wenjie Zhang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, People's Republic of China
| | - Maral Radmanesh
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Nicole Cathcart
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, 75 University Ave. W., Waterloo, ON, Canada
| | - Ali Maleki
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran.
| | - Vladimir Kitaev
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, 75 University Ave. W., Waterloo, ON, Canada.
| |
Collapse
|
40
|
Chen XX, Hou MJ, Mao GJ, Wang WX, Xu F, Li Y, Li CY. ATP-responsive near-infrared fluorescence MOF nanoprobe for the controlled release of anticancer drug. Mikrochim Acta 2021; 188:287. [PMID: 34350511 DOI: 10.1007/s00604-021-04953-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/23/2021] [Indexed: 01/31/2023]
Abstract
A near-infrared (NIR) fluorescence nanoprobe named RhI-DOX@ZIF-90 has been synthesized by wrapping the guest molecule (RhI and DOX) into ZIF-90 framework. The nanoprobe itself is non-fluorescent and the drug (DOX) is inactive. Upon the addition of ATP, the structure of RhI-DOX@ZIF-90 is degraded. The fluorescence of RhI is recovered and DOX is released. The nanoprobe can detect ATP with high sensitivity and selectivity. There is good linear relationship between the nanoprobe and ATP concentration from 0.25 to 10 mM and the detection limit is 0.10 mM. The nanoprobe has the ability to monitor the change of ATP level in living cells and DOX is released inducing apoptosis of cancer cells. RhI-DOX@ZIF-90 is capable of targeting mitochondria, which provides a basis for improving the efficiency of drug delivery by mitochondrial administration. In particular, the nanoprobe is preferentially accumulated in the tumor sites and detect ATP in tumor mice by fluorescence imaging using near-infrared fluorescence. At the same time, DOX can be released accurately in tumor sites and have good anti-tumor efficiency. So, this nanoprobe is a reliable tool to realize early diagnosis of cancer and improve effect of anticancer drug.
Collapse
Affiliation(s)
- Xi-Xi Chen
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Mei-Jia Hou
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Guo-Jiang Mao
- Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, People's Republic of China
| | - Wen-Xin Wang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Fen Xu
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China.
| | - Yongfei Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China.,College of Chemical Engineering, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Chun-Yan Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China.
| |
Collapse
|
41
|
Local Delivery of Pirfenidone by PLA Implants Modifies Foreign Body Reaction and Prevents Fibrosis. Biomedicines 2021; 9:biomedicines9080853. [PMID: 34440057 PMCID: PMC8389617 DOI: 10.3390/biomedicines9080853] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/19/2021] [Accepted: 07/19/2021] [Indexed: 01/04/2023] Open
Abstract
Peri-implant fibrosis (PIF) increases the postsurgical risks after implantation and limits the efficacy of the implantable drug delivery systems (IDDS). Pirfenidone (PF) is an oral anti-fibrotic drug with a short (<3 h) circulation half-life and strong adverse side effects. In the current study, disk-shaped IDDS prototype combining polylactic acid (PLA) and PF, PLA@PF, with prolonged (~3 days) PF release (in vitro) was prepared. The effects of the PLA@PF implants on PIF were examined in the rabbit ear skin pocket model on postoperative days (POD) 30 and 60. Matching blank PLA implants (PLA0) and PLA0 with an equivalent single-dose PF injection performed on POD0 (PLA0+injPF) served as control. On POD30, the intergroup differences were observed in α-SMA, iNOS and arginase-1 expressions in PLA@PF and PLA0+injPF groups vs. PLA0. On POD60, PIF was significantly reduced in PLA@PF group. The peri-implant tissue thickness decreased (532 ± 98 μm vs. >1100 μm in control groups) approaching the intact derma thickness value (302 ± 15 μm). In PLA@PF group, the implant biodegradation developed faster, while arginase-1 expression was suppressed in comparison with other groups. This study proves the feasibility of the local control of fibrotic response on implants via modulation of foreign body reaction with slowly biodegradable PF-loaded IDDS.
Collapse
|
42
|
Prasad SR, Kumar TSS, Jayakrishnan A. Nanocarrier-based drug delivery systems for bone cancer therapy: a review. Biomed Mater 2021; 16. [PMID: 33853043 DOI: 10.1088/1748-605x/abf7d5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/14/2021] [Indexed: 02/07/2023]
Abstract
Bone cancer is a malignant tumor that originates in the bone and destroys the healthy bone tissues. Of the various types of bone tumors, osteosarcoma is the most commonly diagnosed primary bone malignancy. The standard treatment for primary malignant bone tumors comprises surgery, chemotherapy and radiotherapy. Owing to the lack of proven treatments, different forms of alternative therapeutic approaches have been examined in recent decades. Among the new therapeutic methodologies, nanotechnology-based anticancer therapy has paved the way for new targeted strategies for bone cancer treatment and bone regeneration. They include approaches such as the co-delivery of multiple drug cargoes, the enhancement of their biodistribution and transport properties, normalizing accumulation and the optimization of drug release profiles to overcome shortcomings of the existing therapy. This review examines the standard treatments for osteosarcoma, their lacunae, and the evolving therapeutic strategies based on nanocarrier-mediated combinational drug delivery systems, and future perspectives for osteosarcoma therapy.
Collapse
Affiliation(s)
- S Ram Prasad
- Rajiv Gandhi Centre for Biotechnology, Jagathy, Trivandrum 695 014 Kerala, India
| | - T S Sampath Kumar
- Medical Materials Laboratory, Department of Metallurgical and Materials Engineering, Indian Institute of Technology Madras, Chennai 600 036 Tamil Nadu, India
| | - A Jayakrishnan
- Rajiv Gandhi Centre for Biotechnology, Jagathy, Trivandrum 695 014 Kerala, India
| |
Collapse
|
43
|
Redox-responsive hyaluronic acid-based nanoparticles for targeted photodynamic therapy/chemotherapy against breast cancer. J Colloid Interface Sci 2021; 598:213-228. [PMID: 33901847 DOI: 10.1016/j.jcis.2021.04.056] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 12/24/2022]
Abstract
Specific cellular uptake and sufficient drug release in tumor tissues are important for effective cancer therapy. Hyaluronic acid (HA), a skeleton material, could specifically bind to cluster determinant 44 (CD44) receptors highly expressed on the surface of tumor cells to realize active targeting. Cystamine (cys) is sensitive highly reductive environment inside tumor cells and was used as a connecting arm to connect docosahexaenoic acid (DHA) and chlorin e6 (Ce6) to the HA skeleton to obtain redox-sensitive polymer HA-cys-DHA/Ce6 (CHD). Nanoparticles were fabricated and loaded with chemotherapeutic drug docetaxel (DTX) by physical encapsulation. The prepared nanoparticles had significantly increased uptake by MCF-7 cells that overexpressed CD44 receptors, and DTX was effectively released at high reducing condition. Compared with mono-photodynamic therapy (PDT) or mono-chemotherapy, the prepared nanoparticles exhibited superior anti-tumor effect by inhibiting microtubule depolymerization, blocking cell cycle and generating reactive oxygen species (ROS). In vivo anti-tumor experiments proved that DTX/CHD nanoparticles had the best antitumor response versus DTX and CHD nanoparticles under near-infrared (NIR) irradiation. These studies revealed that redox-responsive DTX-loaded CHD nanoparticles held great potential for the treatment of breast cancer.
Collapse
|
44
|
Functional ferrocene polymer multilayer coatings for implantable medical devices: Biocompatible, antifouling, and ROS-sensitive controlled release of therapeutic drugs. Acta Biomater 2021; 125:242-252. [PMID: 33657454 DOI: 10.1016/j.actbio.2021.02.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 02/02/2023]
Abstract
Bacterial infections and the formation of biofilms on the surface of implantable medical devices are critical issues that cause device failure. Implantable medical devices, such as drug delivery technologies, offer promising benefits for targeted and prolonged drug release, but a number of common disadvantages arise that include inadequate release and side effects. Organic film coatings for antifouling and drug delivery are expected to overcome these challenges. Ferrocene polymer-based multifunctional multilayer films were prepared to control the reactive oxygen species (ROS)-responsive release of therapeutic agents while maintaining an antifouling effect and improving biocompatibility. Polymers based on ferrocene and polyethylene glycol were prepared by controlling the molar ratio of carboxylate and amine groups. Layer-by-layer deposition was optimized to achieve the linear growth and self-assembly of dense and stable films. Outstanding anti-biofilm activity (~91% decrease) could be achieved and the films were found to be blood compatible. Importantly, the films effectively incorporated hydrophobic drugs and exhibited dual-responsive drug release at low pH and under ROS conditions at physiological pH. Drug delivery to MCF-7 breast cancer cells was achieved using a Paclitaxel loaded film, which exhibited an anticancer efficacy of 62%. STATEMENT OF SIGNIFICANCE: Healthcare associated infection is caused by the formation of a biofilm by bacteria on the surface of a medical device. In order to solve this, extensive research has been conducted on many coating technologies. Also, a method of chemical treatment by releasing the drug when it enters the body by loading the drug into the coating film is being studied. However, there is still a lack of technology that can achieve both functions of preventing biofilm production and drug delivery. Therefore, in this study, a multilayer thin film that supports drug and inhibits biofilm formation was prepared through Layer-by-Layer coating of a polymer containing PEG to prevent adsorption. As such, it helps the design of multifunctional coatings for implantable medical devices.
Collapse
|
45
|
Wu C, Yi X, Xu R, Zhang M, Xu Y, Ma Y, Gao L, Zha Z. Biodistribution of etoposide via intratumoral chemotherapy with etoposide-loaded implants. Drug Deliv 2021; 27:974-982. [PMID: 32611260 PMCID: PMC8216434 DOI: 10.1080/10717544.2020.1787558] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Etoposide (VP16) is the traditional antitumor agent which has been widely used in a variety of cancers. However, intravenous administration of VP16 was limited in clinical application because of its low aqueous solubility, poor bioavailability and dose-limiting adverse effects. Local chemotherapy with VP16-loaded drug delivery systems could provide a continuous release of drug at the target site, while minimizing the systemic toxicity. In this study, we prepared the poly-l-lactic acid (PLLA) based VP16-loaded implants (VP16 implants) by the direct compression method. The VP16 implants were characterized with regards to drug content, micromorphology, drug release profiles, differential scanning calorimetry (DSC) and Fourier transform infrared spectroscopy (FTIR) analyses. Furthermore, the biodistribution of VP16 via intratumoral chemotherapy with VP16 implants was investigated using the murine Lewis lung carcinoma model. Our results showed that VP16 dispersed homogenously in the polymeric matrix. Both in vitro and in vivo drug release profiles of the implants were characterized by high initial burst release followed by sustained release of VP16. The VP16 implants showed good compatibility between VP16 and the excipients. Intratumoral chemotherapy with VP16 implants resulted in significantly higher concentration and longer duration of VP16 in tumor tissues compared with single intraperitoneal injection of VP16 solution. Moreover, we found the low level of VP16 in plasma and normal organ tissues. These results suggested that intratumoral chemotherapy with VP16 implants enabled high drug concentration at the target site and has the potential to be used as a novel method to treat cancer.
Collapse
Affiliation(s)
- Chunsheng Wu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Xiangting Yi
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Renzhi Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Maokuan Zhang
- Laboratory of Pharmaceutical Research, Anhui Zhongren Science and Technology Co., Ltd, Hefei, PR China
| | - Yan Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Yan Ma
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Li Gao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| |
Collapse
|
46
|
Yuan J, Yuan X, Wu K, Gao J, Li L. A Local and Low-Dose Chemotherapy/Autophagy-Enhancing Regimen Treatment Markedly Inhibited the Growth of Established Solid Tumors Through a Systemic Antitumor Immune Response. Front Oncol 2021; 11:658254. [PMID: 33859948 PMCID: PMC8042230 DOI: 10.3389/fonc.2021.658254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy is one of the main options for the treatment of a variety of malignant tumors. However, the severe side effects resulting from the killing of normal proliferating cells limit the application of cancer-targeting chemotherapeutic drugs. To improve the efficacy of classic systemic chemotherapy, the local delivery of high-dose chemotherapeutic drugs was developed as a method to enhance local drug concentrations and minimize systemic toxicity. Studies have demonstrated that chemotherapy is often accompanied by cancer-associated immunogenic cell death (ICD) and that autophagy is involved in the induction of ICD. To improve the efficacy of local cancer chemotherapy, we hypothesized that the local delivery of chemotherapeutic plus autophagy-enhancing agents would enhance the promotive effects of ICD on the antitumor immune response. Here, we report that a low-dose chemotherapy/autophagy enhancing regimen (CAER) not only resulted in the increased death of B16F10 and 4T1 tumor cells, but also induced higher levels of autophagy in vitro. Importantly, the local delivery of the CARE drugs significantly inhibited tumor growth in B16F10 and 4T1 tumor-bearing mice. Systemic antitumor T-cell immunity was observed in vivo, including neoantigen-specific T-cell responses. Furthermore, bioinformatic analysis of human breast cancer and melanoma tissues showed that autophagy-associated gene expression was upregulated in tumor samples. Increased autophagy and immune cell infiltration in tumor tissues were positively correlated with good prognosis of tumor patients. This work highlights a new approach to improve the effects of local chemotherapy and enhance systemic antitumor immunity.
Collapse
Affiliation(s)
- Jia Yuan
- Institute of Clinical Oncology, Research Center of Cancer Diagnosis and Therapy, and Department of Clinical Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xianlin Yuan
- Institute of Clinical Oncology, Research Center of Cancer Diagnosis and Therapy, and Department of Clinical Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Kunlong Wu
- Institute of Clinical Oncology, Research Center of Cancer Diagnosis and Therapy, and Department of Clinical Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Junxia Gao
- Institute of Clinical Oncology, Research Center of Cancer Diagnosis and Therapy, and Department of Clinical Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Liangping Li
- Institute of Clinical Oncology, Research Center of Cancer Diagnosis and Therapy, and Department of Clinical Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
47
|
pH-Responsive Release of Ruthenium Metallotherapeutics from Mesoporous Silica-Based Nanocarriers. Pharmaceutics 2021; 13:pharmaceutics13040460. [PMID: 33800647 PMCID: PMC8067187 DOI: 10.3390/pharmaceutics13040460] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 01/04/2023] Open
Abstract
Ruthenium complexes are attracting interest in cancer treatment due to their potent cytotoxic activity. However, as their high toxicity may also affect healthy tissues, efficient and selective drug delivery systems to tumour tissues are needed. Our study focuses on the construction of such drug delivery systems for the delivery of cytotoxic Ru(II) complexes upon exposure to a weakly acidic environment of tumours. As nanocarriers, mesoporous silica nanoparticles (MSN) are utilized, whose surface is functionalized with two types of ligands, (2-thienylmethyl)hydrazine hydrochloride (H1) and (5,6-dimethylthieno[2,3-d]pyrimidin-4-yl)hydrazine (H2), which were attached to MSN through a pH-responsive hydrazone linkage. Further coordination to ruthenium(II) center yielded two types of nanomaterials MSN-H1[Ru] and MSN-H2[Ru]. Spectrophotometric measurements of the drug release kinetics at different pH (5.0, 6.0 and 7.4) confirm the enhanced release of Ru(II) complexes at lower pH values, which is further supported by inductively coupled plasma optical emission spectrometry (ICP-OES) measurements. Furthermore, the cytotoxicity effect of the released metallotherapeutics is evaluated in vitro on metastatic B16F1 melanoma cells and enhanced cancer cell-killing efficacy is demonstrated upon exposure of the nanomaterials to weakly acidic conditions. The obtained results showcase the promising capabilities of the designed MSN nanocarriers for the pH-responsive delivery of metallotherapeutics and targeted treatment of cancer.
Collapse
|
48
|
Kong FH, Ye QF, Miao XY, Liu X, Huang SQ, Xiong L, Wen Y, Zhang ZJ. Current status of sorafenib nanoparticle delivery systems in the treatment of hepatocellular carcinoma. Am J Cancer Res 2021; 11:5464-5490. [PMID: 33859758 PMCID: PMC8039945 DOI: 10.7150/thno.54822] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer and one of the leading causes of cancer-related death worldwide. Advanced HCC displays strong resistance to chemotherapy, and traditional chemotherapy drugs do not achieve satisfactory therapeutic efficacy. Sorafenib is an oral kinase inhibitor that inhibits tumor cell proliferation and angiogenesis and induces cancer cell apoptosis. It also improves the survival rates of patients with advanced liver cancer. However, due to its poor solubility, fast metabolism, and low bioavailability, clinical applications of sorafenib have been substantially restricted. In recent years, various studies have been conducted on the use of nanoparticles to improve drug targeting and therapeutic efficacy in HCC. Moreover, nanoparticles have been extensively explored to improve the therapeutic efficacy of sorafenib, and a variety of nanoparticles, such as polymer, lipid, silica, and metal nanoparticles, have been developed for treating liver cancer. All these new technologies have improved the targeted treatment of HCC by sorafenib and promoted nanomedicines as treatments for HCC. This review provides an overview of hot topics in tumor nanoscience and the latest status of treatments for HCC. It further introduces the current research status of nanoparticle drug delivery systems for treatment of HCC with sorafenib.
Collapse
|
49
|
Mameri A, Bournine L, Mouni L, Bensalem S, Iguer-Ouada M. Oxidative stress as an underlying mechanism of anticancer drugs cytotoxicity on human red blood cells' membrane. Toxicol In Vitro 2021; 72:105106. [PMID: 33539984 DOI: 10.1016/j.tiv.2021.105106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/17/2021] [Accepted: 01/30/2021] [Indexed: 11/29/2022]
Abstract
The aim of this study is to investigate the direct in vitro effects of anticancer drugs on red blood cells (RBCs) and to explore the underlying mechanism, mainly by measuring RBCs oxidative stress (OS) status. After RBCs direct contact with fourteen (14) anticancer drugs, several parameters were assessed including: cellular turbidity, methemoglobin (metHb) generation, released Hb and Hb stability. Moreover, intracellular Hb, considered as new molecular target of anticancer drugs, was quantified inside RBCs. MDA level, the main biomarker of OS, was simultaneously measured. The cellular turbidity reveled severe (docetaxel "TXT", 0.03 ± 0.002), moderate (methotrexate "MTX", 0.49 ± 0.009), or none (5-fluorouracil "5-FU", 0.76 ± 0.029) membrane cytotoxicity (MC). An inverse relationship between cell concentration, released Hb and metHb content was obtained. High metHb generation, revealing intense OS, was also mostly expressed in paclitaxel "TXL" and etoposide "VP16". Further, epirubicin "EPI" and "TXT" induced important oxidation of membrane lipids with 0.32 ± 0.014 and 0.26 ± 0.004, respectively. Also, MTX (0.17 ± 0.006) and doxorubicin "DOX" (0.32 ± 0.034) affected significantly Hb stability by a direct contact with molecule. These findings demonstrated that anticancer drugs have the ability to induce membrane damages by the exacerbation of OS through membrane lipid peroxidation and Hb oxidation even inside RBCs.
Collapse
Affiliation(s)
- Amal Mameri
- Laboratoire de Gestion et Valorisation des Ressources Naturelles et Assurances Qualités (LGVRNAQ), Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, 10000 Bouira, Algeria
| | - Lamine Bournine
- Département des Sciences Biologiques, Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, 10000 Bouira, Algeria; Laboratoire de Biotechnologie Végétales et Ethnobotanique (LBVEB), Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, 06000 Bejaia, Algeria.
| | - Lotfi Mouni
- Laboratoire de Gestion et Valorisation des Ressources Naturelles et Assurances Qualités (LGVRNAQ), Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, 10000 Bouira, Algeria; Département des Sciences Biologiques, Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, 10000 Bouira, Algeria
| | - Sihem Bensalem
- Laboratoire de Biotechnologie Végétales et Ethnobotanique (LBVEB), Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, 06000 Bejaia, Algeria
| | - Mokrane Iguer-Ouada
- Laboratoire Associé en Ecosystèmes Marins et Aquacoles (LAEMA), Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, 06000 Bejaia, Algeria
| |
Collapse
|
50
|
Kim WJ, Kang HG, Kim SJ. Dehydroabietic acid inhibits the gastric cancer cell growth via induced apoptosis and cell cycle arrest. Mol Cell Toxicol 2021. [DOI: 10.1007/s13273-021-00118-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|