1
|
Irannejadrankouhi S, Mivehchi H, Eskandari-Yaghbastlo A, Nejati ST, Emrahoglu S, Nazarian M, Zahedi F, Madani SM, Nabi-Afjadi M. Innovative nanoparticle strategies for treating oral cancers. Med Oncol 2025; 42:182. [PMID: 40285805 DOI: 10.1007/s12032-025-02728-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Conventional therapies for oral squamous cell carcinoma (OSCC), a serious worldwide health problem, are frequently constrained by inadequate targeting and serious side effects. Drug delivery systems (DDS) based on nanoparticles provide a possible substitute by improving drug stability, target accuracy, and lowering toxicity. By addressing issues like irregular vasculature and thick tumor matrices, these methods allow for more effective medication administration. For instance, the delivery of cisplatin via liposomes, as opposed to free drug formulations, results in a 40% improvement in tumor suppression. Likewise, compared to traditional techniques, poly (lactic-co-glycolic acid) (PLGA) nanoparticles can produce up to 2.3 times more intertumoral drug accumulation. These platforms have effectively administered natural substances like curcumin and chemotherapeutics like paclitaxel, enhancing therapeutic results while reducing adverse effects. Despite their promise, several types of nanoparticles have drawbacks. For example, PLGA nanoparticles have scaling issues because of their complicated production, whereas liposomes are quickly removed from circulation. In preclinical investigations, functionalized nanoparticles-like EGFR-targeted gold nanoparticles-improve selectivity and effectiveness by obtaining up to 90% receptor binding. By preferentially accumulating in tumors via the increased permeability and retention (EPR) effect, nanoparticles also improve immunotherapy and radiation. Mechanistically, they increase the death of cancer cells by causing DNA damage, interfering with cell division, and producing reactive oxygen species (ROS). There are still issues with toxicity (such as the buildup of metallic nanoparticles in the liver) and large-scale manufacturing. Nevertheless, developments in multifunctional platforms and stimuli-responsive nanoparticles show promise for getting over these obstacles. These developments open the door to more individualized and successful OSCC therapies.
Collapse
Affiliation(s)
| | - Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | | | - Sahand Emrahoglu
- School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Mohammad Nazarian
- Faculty of Dentistry, Belarusion State Medical University, Minsk, Belarus
| | - Farhad Zahedi
- Institute of Molecular Biophysics, Florida State University, 91 Chieftan Way, Tallahassee, FL, 32306, USA
| | - Seyed Mahdi Madani
- Faculty of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, University of Tarbiat Modares, Tehran, Iran.
| |
Collapse
|
2
|
Ye J, Cui H, Liu E, Pei X, Chai M, Sun L, Wang D, Yang VC, Yu F. Temperature switchable linkers suitable for triggered drug release in cancer thermo-chemotherapy. Int J Pharm 2024; 666:124757. [PMID: 39332459 DOI: 10.1016/j.ijpharm.2024.124757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/11/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
In drug delivery systems, a stimuli-responsive linker that attaches a targeting carrier and a cytotoxic payload can be dissociated to release the payload on the target over the action of a stimuli, thereby it would harden the selectivity, specificity and potency of the cytotoxic agent against targeted tissues whilst sparing the drug-induced toxicity on normal cells. Oligonucleotide duplexes can unwind and be separated into single-stranded random coils under a defined temperature, and this property makes the oligonucleotide an appealing thermo-responsive linker. In this work, we studied the melting temperatures of different DNA linkers with various lengths and mismatches inserted in the double helix with either different numbers or positions. We further chose the DNA linkers that can unwind at the hyperthermia temperature and used them in the construction of four different drug delivery systems both in vitro and in vivo. Results showed that the chosen DNA linkers in all of the constructed delivery systems can successfully unwind and release cargos or drugs after application of heat compared to control groups. This research demonstrated the potential applications of DNA duplexes as temperature-sensitive linkers of drug delivery systems for cancer therapy.
Collapse
Affiliation(s)
- Junxiao Ye
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; School of Pharmaceutical Sciences , Tsinghua University, Beijing 100084, China
| | - Hui Cui
- YUGEN MEDCH (Tianjin) Co., Ltd, Tianjin 300450, China
| | - Ergang Liu
- Zhongshan Institute for Drug Discovery, The Institutes of Drug Discovery and Development, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Xing Pei
- Tianjin Key Laboratory of Food and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Meihong Chai
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Lu Sun
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Dongmei Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Victor C Yang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Department of Pharmaceutical Sciences, College of Pharmacy, The University of Michigan, Ann Arbor, MI 48109-1065, USA
| | - Fei Yu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
3
|
Reviansyah FH, Putra DRD, Supriatna JA, Takarini V, Komariah M. Green Dentistry in Oral Cancer Treatment Using Biosynthesis Superparamagnetic Iron Oxide Nanoparticles: A Systematic Review. Cancer Manag Res 2024; 16:1231-1245. [PMID: 39282609 PMCID: PMC11402364 DOI: 10.2147/cmar.s477791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
Oral cancer is a worldwide health issue with high incidence and mortality, demands an effective treatment to improve patient prognosis. Superparamagnetic iron oxide nanoparticles (SPIONs) emerged as a candidate for oral cancer treatment due to their unique attributes, enabling a synergistic combination with its drug-delivery capabilities and hyperthermia when exposed to magnetic fields. SPIONs can be synthesized using biopolymers from agricultural waste like lignin from paddy, which produce biogenic nano iron oxide with superparamagnetic and antioxidant effects. In addition, lignin also acts as a stabilizing agent in creating SPIONs. This study aimed to explore how agricultural waste could be used to prepare SPIONs using the green synthesis method and to evaluate its potential for oral cancer specifically focusing on its effectiveness, side effects, biocompatibility, and toxicity. A systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses protocol. PubMed, EBSCO, and Scopus databases were exploited in the selection of articles published within the last decade. This study quality assessment uses OHAT for critical appraisal tools. Only 10 studies met the inclusion criteria. The findings suggest that the use of agricultural waste in the preparation of SPIONs not only holds potency for oral cancer treatment through drug delivery and hyperthermia but also aligns with the concept of green dentistry. SPIONs as a treatment modality for oral cancer have demonstrated notable effectiveness and versatility. This study provides robust evidence supporting green dentistry by using agricultural waste in the preparation and formulation of SPIONs for managing oral cancer. Its multifunctional nature and ability to enhance treatment efficacy while minimizing adverse effects on healthy tissues highlights the potency of SPION-based oral cancer treatments.
Collapse
Affiliation(s)
| | | | | | - Veni Takarini
- Department of Dental Materials and Technology, Faculty of Dentistry, Padjadjaran University, Bandung, 40132, Indonesia
- Oral Biomaterials Research Centre, Faculty of Dentistry, Padjadjaran University, Bandung, 40132, Indonesia
| | - Maria Komariah
- Department of Fundamental Nursing, Faculty of Nursing, Padjadjaran University, Bandung, 40132, Indonesia
| |
Collapse
|
4
|
Akimoto T, Islam MR, Nagasako A, Kishi K, Nakakaji R, Ohtake M, Hasumi H, Yamaguchi T, Yamada S, Yamamoto T, Ishikawa Y, Umemura M. Alternative magnetic field exposure suppresses tumor growth via metabolic reprogramming. Cancer Sci 2024; 115:2686-2700. [PMID: 38877783 PMCID: PMC11309929 DOI: 10.1111/cas.16243] [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: 02/07/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024] Open
Abstract
Application of physical forces, ranging from ultrasound to electric fields, is recommended in various clinical practice guidelines, including those for treating cancers and bone fractures. However, the mechanistic details of such treatments are often inadequately understood, primarily due to the absence of comprehensive study models. In this study, we demonstrate that an alternating magnetic field (AMF) inherently possesses a direct anti-cancer effect by enhancing oxidative phosphorylation (OXPHOS) and thereby inducing metabolic reprogramming. We observed that the proliferation of human glioblastoma multiforme (GBM) cells (U87 and LN229) was inhibited upon exposure to AMF within a specific narrow frequency range, including around 227 kHz. In contrast, this exposure did not affect normal human astrocytes (NHA). Additionally, in mouse models implanted with human GBM cells in the brain, daily exposure to AMF for 30 min over 21 days significantly suppressed tumor growth and prolonged overall survival. This effect was associated with heightened reactive oxygen species (ROS) production and increased manganese superoxide dismutase (MnSOD) expression. The anti-cancer efficacy of AMF was diminished by either a mitochondrial complex IV inhibitor or a ROS scavenger. Along with these observations, there was a decrease in the extracellular acidification rate (ECAR) and an increase in the oxygen consumption rate (OCR). This suggests that AMF-induced metabolic reprogramming occurs in GBM cells but not in normal cells. Our results suggest that AMF exposure may offer a straightforward strategy to inhibit cancer cell growth by leveraging oxidative stress through metabolic reprogramming.
Collapse
Affiliation(s)
- Taisuke Akimoto
- Department of NeurosurgeryYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Md Rafikul Islam
- Department of Biochemistry and Molecular BiologyWinthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences (UAMS)Little RockArkansasUSA
| | - Akane Nagasako
- Cardiovascular Research InstituteYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | | | - Rina Nakakaji
- Cardiovascular Research InstituteYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Makoto Ohtake
- Department of NeurosurgeryYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Hisashi Hasumi
- Department of Urology, Yokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | | | - Shigeki Yamada
- Department of Materials System Science, Yokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Tetsuya Yamamoto
- Department of NeurosurgeryYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Yoshihiro Ishikawa
- Cardiovascular Research InstituteYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Masanari Umemura
- Cardiovascular Research InstituteYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| |
Collapse
|
5
|
Elzoghby AO, Samir O, Emam HE, Soliman A, Abdelgalil RM, Elmorshedy YM, Elkhodairy KA, Nasr ML. Engineering nanomedicines for immunogenic eradication of cancer cells: Recent trends and synergistic approaches. Acta Pharm Sin B 2024; 14:2475-2504. [PMID: 38828160 PMCID: PMC11143780 DOI: 10.1016/j.apsb.2024.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/07/2024] [Accepted: 03/09/2024] [Indexed: 06/05/2024] Open
Abstract
Resistance to cancer immunotherapy is mainly attributed to poor tumor immunogenicity as well as the immunosuppressive tumor microenvironment (TME) leading to failure of immune response. Numerous therapeutic strategies including chemotherapy, radiotherapy, photodynamic, photothermal, magnetic, chemodynamic, sonodynamic and oncolytic therapy, have been developed to induce immunogenic cell death (ICD) of cancer cells and thereby elicit immunogenicity and boost the antitumor immune response. However, many challenges hamper the clinical application of ICD inducers resulting in modest immunogenic response. Here, we outline the current state of using nanomedicines for boosting ICD of cancer cells. Moreover, synergistic approaches used in combination with ICD inducing nanomedicines for remodeling the TME via targeting immune checkpoints, phagocytosis, macrophage polarization, tumor hypoxia, autophagy and stromal modulation to enhance immunogenicity of dying cancer cells were analyzed. We further highlight the emerging trends of using nanomaterials for triggering amplified ICD-mediated antitumor immune responses. Endoplasmic reticulum localized ICD, focused ultrasound hyperthermia, cell membrane camouflaged nanomedicines, amplified reactive oxygen species (ROS) generation, metallo-immunotherapy, ion modulators and engineered bacteria are among the most innovative approaches. Various challenges, merits and demerits of ICD inducer nanomedicines were also discussed with shedding light on the future role of this technology in improving the outcomes of cancer immunotherapy.
Collapse
Affiliation(s)
- Ahmed O. Elzoghby
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Omar Samir
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Hagar E. Emam
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Ahmed Soliman
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Riham M. Abdelgalil
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Yomna M. Elmorshedy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Kadria A. Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mahmoud L. Nasr
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| |
Collapse
|
6
|
Uthman A, AL-Rawi N, Saeed MH, Eid B, Al-Rawi NH. Tunable theranostics: innovative strategies in combating oral cancer. PeerJ 2024; 12:e16732. [PMID: 38188167 PMCID: PMC10771769 DOI: 10.7717/peerj.16732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/06/2023] [Indexed: 01/09/2024] Open
Abstract
OBJECTIVE This study aims to assess and compare the potential of advanced nano/micro delivery systems, including quantum dots, carbon nanotubes, magnetic nanoparticles, dendrimers, and microneedles, as theranostic platforms for oral cancer. Furthermore, we seek to evaluate their respective advantages and disadvantages over the past decade. MATERIALS AND METHODS A comprehensive literature search was performed using Google Scholar and PubMed, with a focus on articles published between 2013 and 2023. Search queries included the specific advanced delivery system as the primary term, followed by oral cancer as the secondary term (e.g., "quantum dots AND oral cancer," etc.). RESULTS The advanced delivery platforms exhibited notable diagnostic and therapeutic advantages when compared to conventional techniques or control groups. These benefits encompassed improved tumor detection and visualization, enhanced precision in targeting tumors with reduced harm to neighboring tissues, and improved drug solubility and distribution, leading to enhanced drug absorption and tumor uptake. CONCLUSION The findings suggest that advanced nano/micro delivery platforms hold promise for addressing numerous challenges associated with chemotherapy. By enabling precise targeting of cancerous cells, these platforms have the potential to mitigate adverse effects on surrounding healthy tissues, thus encouraging the development of innovative diagnostic and therapeutic strategies for oral cancer.
Collapse
Affiliation(s)
- Asmaa Uthman
- Department of Diagnostic and Surgical Dental Sciences, College of Dentistry, Gulf Medical University, Ajman, United Arab Emirates
| | - Noor AL-Rawi
- Department of Pharmaceutics and Pharmaceutical Technology, University of Sharjah, Sharjah, United Arab Emirates
| | - Musab Hamed Saeed
- Department of Clinical Sciences, College of Dentistry, Ajman University, Ajman, United Arab Emirates
- Ajman University, Centre of Medical and Bio-allied Health Sciences Research,, Ajman, United Arab Emirates
| | - Bassem Eid
- Department of Restorative Dental Sciences, College of Dentistry, Gulf Medical University, Ajman, Ajman, United Arab Emirates
| | - Natheer H. Al-Rawi
- University of Sharjah, Sharjah Institute of Medical Research, Sharjah, United Arab Emirates
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
7
|
Srivastava N, Chudasama B, Baranwal M. Advancement in magnetic hyperthermia-based targeted therapy for cancer treatment. Biointerphases 2023; 18:060801. [PMID: 38078795 DOI: 10.1116/6.0003079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Magnetic hyperthermia utilizing magnetic nanoparticles (MNPs) and an alternating magnetic field (AMF) represents a promising approach in the field of cancer treatment. Active targeting has emerged as a valuable strategy to enhance the effectiveness and specificity of drug delivery. Active targeting utilizes specific biomarkers that are predominantly found in abundance on cancer cells while being minimally expressed on healthy cells. Current comprehensive review provides an overview of several cancer-specific biomarkers, including human epidermal growth factor, transferrin, folate, luteinizing hormone-releasing hormone, integrin, cluster of differentiation (CD) receptors such as CD90, CD95, CD133, CD20, and CD44 also CXCR4 and vascular endothelial growth factor, these biomarkers bind to ligands present on the surface of MNPs, enabling precise targeting. Additionally, this review touches various combination therapies employed to combat cancer. Magnetic hyperthermia synergistically enhances the efficacy of conventional cancer treatments such as targeted chemotherapy, radiation therapy, gene therapy, and immunotherapy.
Collapse
Affiliation(s)
- Neha Srivastava
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Bhupendra Chudasama
- School of Physics and Materials Science, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| |
Collapse
|
8
|
Zoń A, Bednarek I. Cisplatin in Ovarian Cancer Treatment-Known Limitations in Therapy Force New Solutions. Int J Mol Sci 2023; 24:ijms24087585. [PMID: 37108749 PMCID: PMC10146189 DOI: 10.3390/ijms24087585] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Cisplatin is one of the most commonly used anticancer drugs worldwide. It is mainly used in the treatment of ovarian cancer, but also used in testicular, bladder and lung cancers. The significant advantage of this drug is the multidirectional mechanism of its anticancer action, with the most important direction being damaging the DNA of cancer cells. Unfortunately, cisplatin displays a number of serious disadvantages, including toxicity to the most important organs, such as kidneys, heart, liver and inner ear. Moreover, a significant problem among patients with ovarian cancer, treated with cisplatin, is the development of numerous resistance mechanisms during therapy, including changes in the processes of cellular drug import and export, changes in the DNA damage repair mechanisms, as well as numerous changes in the processes of apoptosis and autophagy. Due to all of the mentioned problems, strategies to increase the effectiveness of cisplatin in the treatment of ovarian cancer are intensively sought. The most important strategy includes the development of less toxic cisplatin analogs. Another important direction is combination therapy, involving the simultaneous use of cisplatin with different anticancer drugs, substances derived from plants, temperature or radiotherapy. Many years of observations accompanying the presence of cisplatin in the therapy made it possible to provide a series of verifiable, statistically significant data, but also to show how, over time, with the new information and scientific discoveries, it is possible to describe and understand the therapeutic problems observed in practice, such as the acquisition of drug resistance by tumor cells or induction of changes in the tumor microenvironment. According to the authors, confronting what we knew so far with what new trends offer has a profound meaning. This paper presents information on the history of cisplatin and describes the molecular mechanisms of its action and the development of resistance by cancer cells. In addition, our goal was to highlight a number of therapeutic strategies to increase the effectiveness of cisplatin in the treatment of ovarian cancer, as well as to identify methods to eliminate problems associated with the use of cisplatin.
Collapse
Affiliation(s)
- Aleksandra Zoń
- Department of Biotechnology and Genetic Engineering, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland
| | - Ilona Bednarek
- Department of Biotechnology and Genetic Engineering, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland
| |
Collapse
|
9
|
Spoială A, Ilie CI, Motelica L, Ficai D, Semenescu A, Oprea OC, Ficai A. Smart Magnetic Drug Delivery Systems for the Treatment of Cancer. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13050876. [PMID: 36903753 PMCID: PMC10004758 DOI: 10.3390/nano13050876] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 06/01/2023]
Abstract
Cancer remains the most devastating disease, being one of the main factors of death and morbidity worldwide since ancient times. Although early diagnosis and treatment represent the correct approach in the fight against cancer, traditional therapies, such as chemotherapy, radiotherapy, targeted therapy, and immunotherapy, have some limitations (lack of specificity, cytotoxicity, and multidrug resistance). These limitations represent a continuous challenge for determining optimal therapies for the diagnosis and treatment of cancer. Cancer diagnosis and treatment have seen significant achievements with the advent of nanotechnology and a wide range of nanoparticles. Due to their special advantages, such as low toxicity, high stability, good permeability, biocompatibility, improved retention effect, and precise targeting, nanoparticles with sizes ranging from 1 nm to 100 nm have been successfully used in cancer diagnosis and treatment by solving the limitations of conventional cancer treatment, but also overcoming multidrug resistance. Additionally, choosing the best cancer diagnosis, treatment, and management is extremely important. The use of nanotechnology and magnetic nanoparticles (MNPs) represents an effective alternative in the simultaneous diagnosis and treatment of cancer using nano-theranostic particles that facilitate early-stage detection and selective destruction of cancer cells. The specific properties, such as the control of the dimensions and the specific surface through the judicious choice of synthesis methods, and the possibility of targeting the target organ by applying an internal magnetic field, make these nanoparticles effective alternatives for the diagnosis and treatment of cancer. This review discusses the use of MNPs in cancer diagnosis and treatment and provides future perspectives in the field.
Collapse
Affiliation(s)
- Angela Spoială
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gh Polizu Street, 011061 Bucharest, Romania
- National Centre for Micro and Nanomaterials, and National Centre for Food Safety, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 313 Spl. Independentei, 060042 Bucharest, Romania
| | - Cornelia-Ioana Ilie
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gh Polizu Street, 011061 Bucharest, Romania
- National Centre for Micro and Nanomaterials, and National Centre for Food Safety, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 313 Spl. Independentei, 060042 Bucharest, Romania
| | - Ludmila Motelica
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gh Polizu Street, 011061 Bucharest, Romania
- National Centre for Micro and Nanomaterials, and National Centre for Food Safety, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 313 Spl. Independentei, 060042 Bucharest, Romania
| | - Denisa Ficai
- National Centre for Micro and Nanomaterials, and National Centre for Food Safety, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 313 Spl. Independentei, 060042 Bucharest, Romania
- Department of Inorganic Chemistry, Physical Chemistry and Electrochemistry, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gh Polizu Street, 050054 Bucharest, Romania
| | - Augustin Semenescu
- Departament of Engineering and Management for Transports, Faculty of Transports, University Politehnica of Bucharest, 313 Spl. Independentei, 060042 Bucharest, Romania
- Academy of Romanian Scientists, 3 Street Ilfov, 050045 Bucharest, Romania
| | - Ovidiu-Cristian Oprea
- National Centre for Micro and Nanomaterials, and National Centre for Food Safety, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 313 Spl. Independentei, 060042 Bucharest, Romania
- Department of Inorganic Chemistry, Physical Chemistry and Electrochemistry, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gh Polizu Street, 050054 Bucharest, Romania
- Academy of Romanian Scientists, 3 Street Ilfov, 050045 Bucharest, Romania
| | - Anton Ficai
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gh Polizu Street, 011061 Bucharest, Romania
- National Centre for Micro and Nanomaterials, and National Centre for Food Safety, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 313 Spl. Independentei, 060042 Bucharest, Romania
- Academy of Romanian Scientists, 3 Street Ilfov, 050045 Bucharest, Romania
| |
Collapse
|
10
|
Hyperthermia Treatment as a Promising Anti-Cancer Strategy: Therapeutic Targets, Perspective Mechanisms and Synergistic Combinations in Experimental Approaches. Antioxidants (Basel) 2022; 11:antiox11040625. [PMID: 35453310 PMCID: PMC9030926 DOI: 10.3390/antiox11040625] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 02/04/2023] Open
Abstract
Despite recent developments in diagnosis and treatment options, cancer remains one of the most critical threats to health. Several anti-cancer therapies have been identified, but further research is needed to provide more treatment options that are safe and effective for cancer. Hyperthermia (HT) is a promising treatment strategy for cancer because of its safety and cost-effectiveness. This review summarizes studies on the anti-cancer effects of HT and the detailed mechanisms. In addition, combination therapies with anti-cancer drugs or natural products that can effectively overcome the limitations of HT are reviewed because HT may trigger protective events, such as an increase of heat shock proteins (HSPs). In the 115 reports included, the mechanisms related to apoptosis, cell cycle, reactive oxygen species, mitochondrial membrane potential, DNA damage, transcription factors and HSPs were considered important. This review shows that HT is an effective inducer of apoptosis. Moreover, the limitations of HT may be overcome using combined therapy with anti-cancer drugs or natural products. Therefore, appropriate combinations of such agents with HT will exert maximal effects to treat cancer.
Collapse
|
11
|
The development and progress of nanomedicine for esophageal cancer diagnosis and treatment. Semin Cancer Biol 2022; 86:873-885. [DOI: 10.1016/j.semcancer.2022.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/22/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023]
|
12
|
Li Y, Liu X, Zhang X, Pan W, Li N, Tang B. Immunogenic cell death inducers for enhanced cancer immunotherapy. Chem Commun (Camb) 2021; 57:12087-12097. [PMID: 34714302 DOI: 10.1039/d1cc04604g] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Inducing the immunogenic cell death (ICD) of cancer cells is an important method to improve the immunogenicity of tumor cells for enhanced cancer immunotherapy. Therefore, we discuss the ICD process and then highlight various ICD inducers and strategies for triggering the ICD of cancer cells. We hope that this Feature Article will inspire readers to develop more effective ICD inducers.
Collapse
Affiliation(s)
- Yanhua Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Xiaohan Liu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Xia Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
13
|
Gavilán H, Avugadda SK, Fernández-Cabada T, Soni N, Cassani M, Mai BT, Chantrell R, Pellegrino T. Magnetic nanoparticles and clusters for magnetic hyperthermia: optimizing their heat performance and developing combinatorial therapies to tackle cancer. Chem Soc Rev 2021; 50:11614-11667. [PMID: 34661212 DOI: 10.1039/d1cs00427a] [Citation(s) in RCA: 200] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Magnetic hyperthermia (MHT) is a therapeutic modality for the treatment of solid tumors that has now accumulated more than 30 years of experience. In the ongoing MHT clinical trials for the treatment of brain and prostate tumors, iron oxide nanoparticles are employed as intra-tumoral MHT agents under a patient-safe 100 kHz alternating magnetic field (AMF) applicator. Although iron oxide nanoparticles are currently approved by FDA for imaging purposes and for the treatment of anemia, magnetic nanoparticles (MNPs) designed for the efficient treatment of MHT must respond to specific physical-chemical properties in terms of magneto-energy conversion, heat dose production, surface chemistry and aggregation state. Accordingly, in the past few decades, these requirements have boosted the development of a new generation of MNPs specifically aimed for MHT. In this review, we present an overview on MNPs and their assemblies produced via different synthetic routes, focusing on which MNP features have allowed unprecedented heating efficiency levels to be achieved in MHT and highlighting nanoplatforms that prevent magnetic heat loss in the intracellular environment. Moreover, we review the advances on MNP-based nanoplatforms that embrace the concept of multimodal therapy, which aims to combine MHT with chemotherapy, radiotherapy, immunotherapy, photodynamic or phototherapy. Next, for a better control of the therapeutic temperature at the tumor, we focus on the studies that have optimized MNPs to maintain gold-standard MHT performance and are also tackling MNP imaging with the aim to quantitatively assess the amount of nanoparticles accumulated at the tumor site and regulate the MHT field conditions. To conclude, future perspectives with guidance on how to advance MHT therapy will be provided.
Collapse
Affiliation(s)
- Helena Gavilán
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genoa, Italy.
| | | | | | - Nisarg Soni
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genoa, Italy.
| | - Marco Cassani
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genoa, Italy.
| | - Binh T Mai
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genoa, Italy.
| | - Roy Chantrell
- Department of Physics, University of York, York YO10 5DD, UK
| | | |
Collapse
|
14
|
Chien LH, Wu CT, Deng JS, Jiang WP, Huang WC, Huang GJ. Salvianolic Acid C Protects against Cisplatin-Induced Acute Kidney Injury through Attenuation of Inflammation, Oxidative Stress and Apoptotic Effects and Activation of the CaMKK-AMPK-Sirt1-Associated Signaling Pathway in Mouse Models. Antioxidants (Basel) 2021; 10:antiox10101620. [PMID: 34679755 PMCID: PMC8533075 DOI: 10.3390/antiox10101620] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/23/2021] [Accepted: 10/13/2021] [Indexed: 12/28/2022] Open
Abstract
Acute kidney injury (AKI) is a sudden reduction in kidney activity and has a high mortality rate. Salvianolic acid C (SAC), one of the main polyphenolic components of Salvia miltiorrhiza, displays significant pharmacologically active effects. An animal model of cisplatin-induced kidney injury was used to study the potential of SAC to improve AKI. First, SAC was administered intraperitoneally in mice for 10 consecutive days, and then cisplatin was administered intraperitoneally on day 7 to establish a nephrotoxicity mouse model. SAC mitigated renal histological changes, blood creatinine (CRE) and blood urea nitrogen (BUN) production and the levels of inflammatory mediators in the cisplatin-induced AKI. Furthermore, malondialdehyde (MDA) levels were reduced and glutathione (GSH) was increased after intraperitoneal injection (i.p.) administration of SAC. In addition, based on Western blot data, SAC reduced the expression of inducible NO synthase (iNOS), cyclooxygenase-2 (COX-2), nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) activation in mouse renal tissues. Finally, SAC diminished the level of TLR-4 expression and enhanced the production of several antioxidative enzymes (superoxidase dismutase (SOD1), glutathione peroxidase (GPx3), catalase, nuclear-factor-erythroid-2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1)), Sirtuin 1 (Sirt1), p-AMP-activated protein kinase (AMPK) and p-Ca2+/calmodulin-dependent protein kinase kinase (CaMKK). In addition, Sirt1 inhibition (EX 527) inverted the effect of SAC against cisplatin-induced nephrotoxicity. Collectively, SAC provides a therapeutic target with promising clinical potential after cisplatin treatment by attenuating oxidative stress and inflammation.
Collapse
Affiliation(s)
- Liang-Hsuan Chien
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan;
| | - Chien-Ta Wu
- Faculty of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Jeng-Shyan Deng
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 413, Taiwan;
| | - Wen-Ping Jiang
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan;
| | - Wen-Chin Huang
- Graduate Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404, Taiwan;
- International Master’s Program of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404, Taiwan
| | - Guan-Jhong Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan;
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 413, Taiwan;
- Correspondence: ; Tel.: +886-4-2205-3366 (ext. 5508)
| |
Collapse
|
15
|
|
16
|
Mamani JB, Souza TKF, Nucci MP, Oliveira FA, Nucci LP, Alves AH, Rego GNA, Marti L, Gamarra LF. In Vitro Evaluation of Hyperthermia Magnetic Technique Indicating the Best Strategy for Internalization of Magnetic Nanoparticles Applied in Glioblastoma Tumor Cells. Pharmaceutics 2021; 13:1219. [PMID: 34452180 PMCID: PMC8399657 DOI: 10.3390/pharmaceutics13081219] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 11/16/2022] Open
Abstract
This in vitro study aims to evaluate the magnetic hyperthermia (MHT) technique and the best strategy for internalization of magnetic nanoparticles coated with aminosilane (SPIONAmine) in glioblastoma tumor cells. SPIONAmine of 50 and 100 nm were used for specific absorption rate (SAR) analysis, performing the MHT with intensities of 50, 150, and 300 Gauss and frequencies varying between 305 and 557 kHz. The internalization strategy was performed using 100, 200, and 300 µgFe/mL of SPIONAmine, with or without Poly-L-Lysine (PLL) and filter, and with or without static or dynamic magnet field. The cell viability was evaluated after determination of MHT best condition of SPIONAmine internalization. The maximum SAR values of SPIONAmine (50 nm) and SPIONAmine (100 nm) identified were 184.41 W/g and 337.83 W/g, respectively, using a frequency of 557 kHz and intensity of 300 Gauss (≈23.93 kA/m). The best internalization strategy was 100 µgFe/mL of SPIONAmine (100 nm) using PLL with filter and dynamic magnet field, submitted to MHT for 40 min at 44 °C. This condition displayed 70.0% decreased in cell viability by flow cytometry and 68.1% by BLI. We can conclude that our study is promising as an antitumor treatment, based on intra- and extracellular MHT effects. The optimization of the nanoparticles internalization process associated with their magnetic characteristics potentiates the extracellular acute and late intracellular effect of MHT achieving greater efficiency in the therapeutic process.
Collapse
Affiliation(s)
- Javier B. Mamani
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (J.B.M.); (T.K.F.S.); (M.P.N.); (F.A.O.); (A.H.A.); (G.N.A.R.); (L.M.)
| | - Taylla K. F. Souza
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (J.B.M.); (T.K.F.S.); (M.P.N.); (F.A.O.); (A.H.A.); (G.N.A.R.); (L.M.)
| | - Mariana P. Nucci
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (J.B.M.); (T.K.F.S.); (M.P.N.); (F.A.O.); (A.H.A.); (G.N.A.R.); (L.M.)
- LIM44-Hospital das Clínicas da Faculdade Medicina da Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | - Fernando A. Oliveira
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (J.B.M.); (T.K.F.S.); (M.P.N.); (F.A.O.); (A.H.A.); (G.N.A.R.); (L.M.)
| | - Leopoldo P. Nucci
- Centro Universitário do Planalto Central, Brasília 72445-020, DF, Brazil;
| | - Arielly H. Alves
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (J.B.M.); (T.K.F.S.); (M.P.N.); (F.A.O.); (A.H.A.); (G.N.A.R.); (L.M.)
| | - Gabriel N. A. Rego
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (J.B.M.); (T.K.F.S.); (M.P.N.); (F.A.O.); (A.H.A.); (G.N.A.R.); (L.M.)
| | - Luciana Marti
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (J.B.M.); (T.K.F.S.); (M.P.N.); (F.A.O.); (A.H.A.); (G.N.A.R.); (L.M.)
| | - Lionel F. Gamarra
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (J.B.M.); (T.K.F.S.); (M.P.N.); (F.A.O.); (A.H.A.); (G.N.A.R.); (L.M.)
| |
Collapse
|
17
|
Amatya R, Hwang S, Park T, Min KA, Shin MC. In Vitro and In Vivo Evaluation of PEGylated Starch-Coated Iron Oxide Nanoparticles for Enhanced Photothermal Cancer Therapy. Pharmaceutics 2021; 13:871. [PMID: 34204840 PMCID: PMC8231641 DOI: 10.3390/pharmaceutics13060871] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 01/22/2023] Open
Abstract
Iron oxide nanoparticles (IONPs) possess versatile utility in cancer theranostics, thus, they have drawn enormous interest in the cancer research field. Herein, we prepared polyethylene glycol (PEG)-conjugated and starch-coated IONPs ("PEG-starch-IONPs"), and assessed their applicability for photothermal treatment (PTT) of cancer. The prepared PEG-starch-IONPs were investigated for their physical properties by transmission electron microscopy (TEM), energy dispersive spectroscopy (EDS), X-ray diffraction (XRD), Fourier transform infrared (FT-IR) spectroscopy, and dynamic light scattering (DLS). The pharmacokinetic study results showed a significant extension in the plasma half-life by PEGylation, which led to a markedly increased (5.7-fold) tumor accumulation. When PEG-starch-IONPs were evaluated for their photothermal activity, notably, they displayed marked and reproducible heating effects selectively on the tumor site with laser irradiation. Lastly, efficacy studies demonstrated that PEG-starch-IONPs-based PTT may be a promising mode of cancer therapy.
Collapse
Affiliation(s)
- Reeju Amatya
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju 52828, Gyeongnam, Korea; (R.A.); (T.P.)
| | - Seungmi Hwang
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea;
| | - Taehoon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju 52828, Gyeongnam, Korea; (R.A.); (T.P.)
| | - Kyoung Ah Min
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea;
| | - Meong Cheol Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju 52828, Gyeongnam, Korea; (R.A.); (T.P.)
| |
Collapse
|
18
|
Zheng W, Zhou Q, Yuan C. Nanoparticles for Oral Cancer Diagnosis and Therapy. Bioinorg Chem Appl 2021; 2021:9977131. [PMID: 33981334 PMCID: PMC8088384 DOI: 10.1155/2021/9977131] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Oral cancer is the sixth most common malignant cancer, affecting the health of people with an unacceptably high mortality rate. Despite numerous clinical methods in the diagnosis and therapy of oral cancer (e.g., magnetic resonance imaging, computed tomography, surgery, and chemoradiotherapy), they still remain far from optimal. Therefore, an urgent need exists for effective and practical techniques of early diagnosis and effective therapy of oral cancer. Currently, various types of nanoparticles have aroused wide public concern, representing a promising tool for diagnostic probes and therapeutic devices. Their inherent physicochemical features, including ultrasmall size, high reactivity, and tunable surface modification, enable them to overcome some of the limitations and achieve the expected diagnostic and therapeutic effect. In this review, we introduce different types of nanoparticles that emerged for the diagnosis and therapy of oral cancers. Then, the challenges and future perspectives for nanoparticles applied in oral cancer diagnosis and therapy are presented. The objective of this review is to help researchers better understand the effect of nanoparticles on oral cancer diagnosis and therapy and may accelerate breakthroughs in this field.
Collapse
Affiliation(s)
- Weiping Zheng
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
- School of Stomatology, Qingdao University, Qingdao 266003, China
| | - Qihui Zhou
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
- School of Stomatology, Qingdao University, Qingdao 266003, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266021, China
| | - Changqing Yuan
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
- School of Stomatology, Qingdao University, Qingdao 266003, China
| |
Collapse
|
19
|
Carter TJ, Agliardi G, Lin FY, Ellis M, Jones C, Robson M, Richard-Londt A, Southern P, Lythgoe M, Zaw Thin M, Ryzhov V, de Rosales RTM, Gruettner C, Abdollah MRA, Pedley RB, Pankhurst QA, Kalber TL, Brandner S, Quezada S, Mulholland P, Shevtsov M, Chester K. Potential of Magnetic Hyperthermia to Stimulate Localized Immune Activation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2005241. [PMID: 33734595 DOI: 10.1002/smll.202005241] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/20/2021] [Indexed: 05/27/2023]
Abstract
Magnetic hyperthermia (MH) harnesses the heat-releasing properties of superparamagnetic iron oxide nanoparticles (SPIONs) and has potential to stimulate immune activation in the tumor microenvironment whilst sparing surrounding normal tissues. To assess feasibility of localized MH in vivo, SPIONs are injected intratumorally and their fate tracked by Zirconium-89-positron emission tomography, histological analysis, and electron microscopy. Experiments show that an average of 49% (21-87%, n = 9) of SPIONs are retained within the tumor or immediately surrounding tissue. In situ heating is subsequently generated by exposure to an externally applied alternating magnetic field and monitored by thermal imaging. Tissue response to hyperthermia, measured by immunohistochemical image analysis, reveals specific and localized heat-shock protein expression following treatment. Tumor growth inhibition is also observed. To evaluate the potential effects of MH on the immune landscape, flow cytometry is used to characterize immune cells from excised tumors and draining lymph nodes. Results show an influx of activated cytotoxic T cells, alongside an increase in proliferating regulatory T cells, following treatment. Complementary changes are found in draining lymph nodes. In conclusion, results indicate that biologically reactive MH is achievable in vivo and can generate localized changes consistent with an anti-tumor immune response.
Collapse
Affiliation(s)
- Thomas J Carter
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Giulia Agliardi
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Fang-Yu Lin
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
| | - Matthew Ellis
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Cancer Sciences Unit, Cancer Research UK Centre, University of Southampton, Somers Building, Southampton, SO16 6YD, UK
| | - Clare Jones
- School of Biomedical Engineering and Imaging Sciences, King's College London (KCL), St Thomas' Hospital, London, SE1 7EH, UK
| | - Mathew Robson
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Angela Richard-Londt
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Paul Southern
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
- Resonant Circuits Limited (RCL), London, W1S 4BS, UK
| | - Mark Lythgoe
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - May Zaw Thin
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Vyacheslav Ryzhov
- NRC "Kurchatov Institute", Petersburg Nuclear Physics Institute, Gatchina, 188300, Russia
| | - Rafael T M de Rosales
- School of Biomedical Engineering and Imaging Sciences, King's College London (KCL), St Thomas' Hospital, London, SE1 7EH, UK
| | - Cordula Gruettner
- Micromod Partikeltechnologie GmbH, Friedrich-Barnewitz-Str. 4, Rostock, D-18119, Germany
| | - Maha R A Abdollah
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt (BUE), El Shorouk City, Misr- Ismalia Desert Road, 11873, Cairo, Egypt
| | - R Barbara Pedley
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Quentin A Pankhurst
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
- Resonant Circuits Limited (RCL), London, W1S 4BS, UK
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Sebastian Brandner
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Sergio Quezada
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Paul Mulholland
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Maxim Shevtsov
- NRC "Kurchatov Institute", Petersburg Nuclear Physics Institute, Gatchina, 188300, Russia
- Technical University of Munich, Klinikum Rechts der Isar, Ismaninger str. 22, Munich, 81675, Germany
| | - Kerry Chester
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| |
Collapse
|
20
|
Liu C, Zhang T, Chen L, Chen Y. The choice of anti-tumor strategies based on micromolecules or drug loading function of biomaterials. Cancer Lett 2020; 487:45-52. [PMID: 32474154 DOI: 10.1016/j.canlet.2020.05.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/08/2020] [Accepted: 05/15/2020] [Indexed: 01/22/2023]
Abstract
With advances in modern medicine, diverse tumor therapies have been developed. However, because of a lack of effective methods, the delivery of drugs or micromolecules in the human body has many limitations. Biomaterials are natural or synthetic functional materials that are prone to contact or interact with living systems. Therefore, the application of biomaterials provides innovative anti-tumor strategies, especially in tumor targeting, chemotherapy sensitization, tumor immunotherapy. The combination of biomaterials and drugs provides a promising strategy to overcome the biological barriers of drug delivery. Nanomaterials can target specific tumor sites to enhance the efficiency of tumor therapies and decrease the toxicity of drug through passive targeting, active targeting and direct targeting. Additionally, biomaterials can be used to enhance the sensitivity of tumor cells to chemotherapy drugs. Furthermore, modifiable biomaterials can induce effective anti-tumor immune response. Currently, the developmental trend of biomaterial for drug delivery is motivated by the combination and diversification of different therapies. With interdisciplinary development, a variety of anti-tumor strategies will emerge in an endless stream to bring great hope for tumor therapy. In this review, we will discuss the anti-tumor strategies based on nanoparticles and injectable scaffolds.
Collapse
Affiliation(s)
- Chengyi Liu
- Department of Urology, The Second Hospital of TianJin Medical University, TianJin Institute of Urology, Tianjin, 300211, China; Department of Urology, Lu'an Affiliated Hospital of Anhui Medical University, 237000, Anhui, China
| | - Tianke Zhang
- Department of Urology, The Second Hospital of TianJin Medical University, TianJin Institute of Urology, Tianjin, 300211, China; Department of Anorectal Surgery, Tianjin Union Medical Center, 300121, Tianjin, China
| | - Liqun Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, 300072, Tianjin, China
| | - Yue Chen
- Department of Urology, The Second Hospital of TianJin Medical University, TianJin Institute of Urology, Tianjin, 300211, China.
| |
Collapse
|
21
|
In vitro thermo-triggered drug release from magnetic polyurethane-urea nanocomposite. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101564] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
22
|
Mukherjee S, Liang L, Veiseh O. Recent Advancements of Magnetic Nanomaterials in Cancer Therapy. Pharmaceutics 2020; 12:pharmaceutics12020147. [PMID: 32053995 PMCID: PMC7076668 DOI: 10.3390/pharmaceutics12020147] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/05/2020] [Accepted: 02/08/2020] [Indexed: 12/16/2022] Open
Abstract
Magnetic nanomaterials belong to a class of highly-functionalizable tools for cancer therapy owing to their intrinsic magnetic properties and multifunctional design that provides a multimodal theranostics platform for cancer diagnosis, monitoring, and therapy. In this review article, we have provided an overview of the various applications of magnetic nanomaterials and recent advances in the development of these nanomaterials as cancer therapeutics. Moreover, the cancer targeting, potential toxicity, and degradability of these nanomaterials has been briefly addressed. Finally, the challenges for clinical translation and the future scope of magnetic nanoparticles in cancer therapy are discussed.
Collapse
|
23
|
Song Y, Li D, Lu Y, Jiang K, Yang Y, Xu Y, Dong L, Yan X, Ling D, Yang X, Yu SH. Ferrimagnetic mPEG-b-PHEP copolymer micelles loaded with iron oxide nanocubes and emodin for enhanced magnetic hyperthermia–chemotherapy. Natl Sci Rev 2020; 7:723-736. [PMID: 34692091 PMCID: PMC8289054 DOI: 10.1093/nsr/nwz201] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/18/2019] [Accepted: 12/02/2019] [Indexed: 02/01/2023] Open
Abstract
As a non-invasive therapeutic method without penetration-depth limitation, magnetic hyperthermia therapy (MHT) under alternating magnetic field (AMF) is a clinically promising thermal therapy. However, the poor heating conversion efficiency and lack of stimulus–response obstruct the clinical application of magnetofluid-mediated MHT. Here, we develop a ferrimagnetic polyethylene glycol-poly(2-hexoxy-2-oxo-1,3,2-dioxaphospholane) (mPEG-b-PHEP) copolymer micelle loaded with hydrophobic iron oxide nanocubes and emodin (denoted as EMM). Besides an enhanced magnetic resonance (MR) contrast ability (r2 = 271 mM−1 s−1) due to the high magnetization, the specific absorption rate (2518 W/g at 35 kA/m) and intrinsic loss power (6.5 nHm2/kg) of EMM are dozens of times higher than the clinically available iron oxide nanoagents (Feridex and Resovist), indicating the high heating conversion efficiency. Furthermore, this composite micelle with a flowable core exhibits a rapid response to magnetic hyperthermia, leading to an AMF-activated supersensitive drug release. With the high magnetic response, thermal sensitivity and magnetic targeting, this supersensitive ferrimagnetic nanocomposite realizes an above 70% tumor cell killing effect at an extremely low dosage (10 μg Fe/mL), and the tumors on mice are completely eliminated after the combined MHT–chemotherapy.
Collapse
Affiliation(s)
- Yonghong Song
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230009, China
| | - Dongdong Li
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Yang Lu
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230009, China
| | - Kun Jiang
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230009, China
| | - Yi Yang
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230009, China
| | - Yunjun Xu
- Division of Nanomaterials & Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center of CAS, Department of Chemistry, Institute of Biomimetic Materials & Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Liang Dong
- Division of Nanomaterials & Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center of CAS, Department of Chemistry, Institute of Biomimetic Materials & Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Xu Yan
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230009, China
| | - Daishun Ling
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Key Laboratory of Biomedical Engineering of the Ministry of Education, Zhejiang University, Hangzhou 310058, China
| | - Xianzhu Yang
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Shu-Hong Yu
- Division of Nanomaterials & Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center of CAS, Department of Chemistry, Institute of Biomimetic Materials & Chemistry, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
24
|
Liu X, Zheng J, Sun W, Zhao X, Li Y, Gong N, Wang Y, Ma X, Zhang T, Zhao LY, Hou Y, Wu Z, Du Y, Fan H, Tian J, Liang XJ. Ferrimagnetic Vortex Nanoring-Mediated Mild Magnetic Hyperthermia Imparts Potent Immunological Effect for Treating Cancer Metastasis. ACS NANO 2019; 13:8811-8825. [PMID: 31328922 DOI: 10.1021/acsnano.9b01979] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Cancer metastasis is a serious concern and a major reason for treatment failure. Herein, we have reported the development of an effective and safe nanotherapeutic strategy that can eradicate primary tumors, inhibit metastasizing to lung, and control the metastasis and growth of distant tumors. Briefly, ferrimagnetic vortex-domain iron oxide nanoring (FVIO)-mediated mild magnetic hyperthermia caused calreticulin (CRT) expression on the 4T1 breast cancer cells. The CRT expression transmitted an "eat-me" signal and promoted phagocytic uptake of cancer cells by the immune system to induce an efficient immunogenic cell death, further leading to the macrophage polarization. This mild thermotherapy promoted 88% increase of CD8+ cytotoxic T lymphocyte infiltration in distant tumors and triggered immunotherapy by effectively sensitizing tumors to the PD-L1 checkpoint blockade. The percentage of CD8+ cytotoxic T lymphocytes can be further increased from 55.4% to 64.5% after combining with PD-L1 blockade. Moreover, the combination treatment also inhibited the immunosuppressive response of the tumor, evidenced by significant down-regulation of myeloid-derived suppressor cells (MDSCs). Our results revealed that the FVIO-mediated mild magnetic hyperthermia can activate the host immune systems and efficiently cooperate with PD-L1 blockade to inhibit the potential metastatic spreading as well as the growth of distant tumors.
Collapse
Affiliation(s)
- Xiaoli Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
- The College of Life Sciences , Northwest University , Xi'an , Shaanxi 710069 , China
| | - Jianjun Zheng
- Department of Radiology, Hwa Mei Hospital , University of Chinese Academy of Sciences , Ningbo No.2 Hospital, Ningbo , Zhejiang 315010 , China
| | - Wei Sun
- Department of Radiology, Hwa Mei Hospital , University of Chinese Academy of Sciences , Ningbo No.2 Hospital, Ningbo , Zhejiang 315010 , China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
| | - Yao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
| | - Ningqiang Gong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
| | - Yanyun Wang
- The College of Life Sciences , Northwest University , Xi'an , Shaanxi 710069 , China
| | - Xiaowei Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
| | - Tingbin Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science , Northwest University , Xi'an 710127 , China
| | - Ling-Yun Zhao
- Key Laboratory of Advanced Materials, School of Material Science & Engineering , Tsinghua University , Beijing 100084 , China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School , Nanjing University , Nanjing 210093 , China
- Jiangsu Key Laboratory of Molecular Medicine , Nanjing University , Nanjing , 210093 , China
| | - Zhibing Wu
- Department of Radiation Oncology , Zhejiang Hospital , Hangzhou , Zhejiang 310013 , China
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems , Institute of Automation, Chinese Academy of Sciences , Beijing 100190 , China
| | - Haiming Fan
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science , Northwest University , Xi'an 710127 , China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems , Institute of Automation, Chinese Academy of Sciences , Beijing 100190 , China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
25
|
Umemura M, Islam MR, Fukumura H, Sato I, Kawabata Y, Matsuo K, Nakakaji R, Nagasako A, Ohtake M, Takayuki F, Yokoyama U, Nakayama T, Eguchi H, Ishikawa Y. Simultaneous hyperthermia-chemotherapy effect by arterial injection of Fe(Salen) for femur tumor. Cancer Sci 2018; 110:356-365. [PMID: 30375142 PMCID: PMC6317948 DOI: 10.1111/cas.13851] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/12/2018] [Accepted: 10/23/2018] [Indexed: 12/14/2022] Open
Abstract
We previously identified a novel nanomagnetic particle, N,N′‐bis(salicylidene)ethylenediamine iron [Fe(Salen)]. Fe(Salen) not only shows antitumor effects but also magnetic properties. We found that Fe(Salen) can be used for magnet‐guided drug delivery and visualization of accumulated drug by magnetic resonance imaging (MRI) because of its magnetism. In addition, Fe(Salen) can generate heat by itself when exposed to an alternating current magnetic field (AMF), resulting in a hyperthermia effect. Herein, we partly elucidated the antitumor mechanism of Fe(Salen) and carried out an i.v. repeated dose toxicity study to decide the therapeutic amount. Furthermore, we evaluated the antitumor effect of selective intra‐arterial injection or i.v. injection of Fe(Salen) by catheter and the hyperthermia effect of Fe(Salen) when exposed to AMF in vivo. We used a rabbit model grafted with VX2 cells (rabbit squamous cell carcinoma) on the right leg. Intra‐arterial injection of Fe(Salen) showed a greater antitumor effect than did i.v. injection. The combination of Fe(Salen) intra‐arterial injection and AMF exposure showed a greater antitumor effect than did either Fe(Salen) or methotrexate (MTX) without AMF exposure, suggesting that AMF exposure greatly enhanced the antitumor effect of Fe(Salen) by arterial injection by catheter. This is the first report that the effectiveness of Fe(Salen) was evaluated in the point of administration route; that is, selective intra‐arterial injection by catheter. Taken together, these results indicate a new administration route; that is, selective arterial injection of Fe(Salen) by catheter, and the development of a new strategy of simultaneous hyperthermia‐chemotherapy in the future.
Collapse
Affiliation(s)
- Masanari Umemura
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Md Rafikul Islam
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | | | - Itaru Sato
- Sato Oral and Maxillofacial Surgery Clinic, Zushi, Japan
| | - Yusuke Kawabata
- Department of Orthopedic Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kousuke Matsuo
- Department of Orthopedic Surgery, Fujisawa City Hospital, Fujisawa, Japan
| | - Rina Nakakaji
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan.,Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akane Nagasako
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Makoto Ohtake
- Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Fujita Takayuki
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Utako Yokoyama
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Tomohiro Nakayama
- Department of Veterinary Medicine, Nihon University, Fujisawa, Japan
| | - Haruki Eguchi
- Research Laboratory, Corporate Research & Development, IHI Corporation, Yokohama, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| |
Collapse
|
26
|
Akimoto T, Umemura M, Nagasako A, Ohtake M, Fujita T, Yokoyama U, Eguchi H, Yamamoto T, Ishikawa Y. Alternating magnetic field enhances cytotoxicity of Compound C. Cancer Sci 2018; 109:3483-3493. [PMID: 30155931 PMCID: PMC6215876 DOI: 10.1111/cas.13781] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 12/22/2022] Open
Abstract
We previously reported the efficacy of anti-cancer therapy with hyperthermia using an alternating magnetic field (AMF) and a magnetic compound. In the course of the study, unexpectedly, we found that an AMF enhances the cytotoxicity of Compound C, an activated protein kinase (AMPK) inhibitor, although this compound is not magnetic. Therefore, we examined the cellular mechanism of AMF-induced cytotoxicity of Compound C in cultured human glioblastoma (GB) cells. An AMF (280 kHz, 250 Arms) for 30 minutes significantly enhanced the cytotoxicity of Compound C and promoted apoptosis towards several human GB cell lines in vitro. The AMF also increased Compound C-induced cell-cycle arrest of GB cells at the G2 phase and, thus, inhibited cell proliferation. The AMF increased Compound C-induced reactive oxygen species production. Furthermore, the AMF decreased ERK phosphorylation in the presence of Compound C and suppressed the protective autophagy induced by this compound. The application of an AMF in cancer chemotherapy may be a simple and promising method, which might reduce the doses of drugs used in future cancer treatment and, therefore, the associated side effects.
Collapse
Affiliation(s)
- Taisuke Akimoto
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan.,Department of Neurosurgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Masanari Umemura
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Akane Nagasako
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Makoto Ohtake
- Department of Neurosurgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Takayuki Fujita
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Utako Yokoyama
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Haruki Eguchi
- Research Laboratory, Corporate Research & Development, IHI Corporation, Yokohama, Japan
| | - Tetsuya Yamamoto
- Department of Neurosurgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| |
Collapse
|
27
|
El Hajj Diab D, Clerc P, Serhan N, Fourmy D, Gigoux V. Combined Treatments of Magnetic Intra-Lysosomal Hyperthermia with Doxorubicin Promotes Synergistic Anti-Tumoral Activity. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E468. [PMID: 29954075 PMCID: PMC6071107 DOI: 10.3390/nano8070468] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/14/2018] [Accepted: 06/19/2018] [Indexed: 11/25/2022]
Abstract
Doxorubicin is a cytotoxic drug used for the treatment of many cancer types. However, its significant dose-related adverse effects including cardiotoxicity may hamper its efficiency. Moreover, the multidrug resistance that appears during treatments limits anti-cancer therapies. Hyperthermia has been introduced as an adjuvant anti-cancer therapy and presents promising opportunities especially in combination with chemotherapy. However, hyperthermia methods including standard magnetic hyperthermia do not discriminate between the target and the surrounding normal tissues and can lead to side effects. In this context, a Magnetic Intra-Lysosomal Hyperthermia (MILH) approach, which occurs without perceptible temperature rise, has been developed. We previously showed that minute amounts of iron oxide magnetic nanoparticles targeting the gastrin receptor (CCK2R) are internalized by cancer cells through a CCK2R-dependent physiological process, accumulated into their lysosomes and kill cancer cells upon high frequency alternating magnetic field (AMF) application through lysosomal cell death. Here, we show that the combination of MILH with doxorubicin increases the efficiency of the eradication of endocrine tumor cells with synergism. We also demonstrate that these two treatments activate two different cell death pathways that are respectively dependent on Caspase-1 and Caspase-3 activation. These findings will result in the development of new anti-tumoral, intra-lysosomal-thermo/chemotherapy with better curative effects than chemotherapy alone and that are devoid of adverse effects linked to standard hyperthermia approaches.
Collapse
Affiliation(s)
- Darine El Hajj Diab
- INSERM ERL1226-Receptology and Therapeutic Targeting of Cancers, Laboratoire de Physique et Chimie des Nano-Objets, CNRS UMR5215-INSA, Université de Toulouse III, F-31432 Toulouse, France.
| | - Pascal Clerc
- INSERM ERL1226-Receptology and Therapeutic Targeting of Cancers, Laboratoire de Physique et Chimie des Nano-Objets, CNRS UMR5215-INSA, Université de Toulouse III, F-31432 Toulouse, France.
| | - Nizar Serhan
- INSERM ERL1226-Receptology and Therapeutic Targeting of Cancers, Laboratoire de Physique et Chimie des Nano-Objets, CNRS UMR5215-INSA, Université de Toulouse III, F-31432 Toulouse, France.
| | - Daniel Fourmy
- INSERM ERL1226-Receptology and Therapeutic Targeting of Cancers, Laboratoire de Physique et Chimie des Nano-Objets, CNRS UMR5215-INSA, Université de Toulouse III, F-31432 Toulouse, France.
| | - Véronique Gigoux
- INSERM ERL1226-Receptology and Therapeutic Targeting of Cancers, Laboratoire de Physique et Chimie des Nano-Objets, CNRS UMR5215-INSA, Université de Toulouse III, F-31432 Toulouse, France.
| |
Collapse
|
28
|
Abstract
N,N’-Bis(salicylidene)ethylenediamine iron (Fe(Salen)) is an anti-cancer agent with intrinsic magnetic property. Here, we covalently linked Fe(Salen) to paclitaxel (PTX), a widely used anti-cancer drug, to obtain a magnetized paclitaxel conjugate (M-PTX), which exhibited magnetic characteristics for magnet-guided drug delivery and MRI visualization. M-PTX increased apoptosis and G2/M arrest of cultured human oral cancer cell lines in the same manner as PTX. Furthermore, marked contrast intensity was obtained in magnetic resonance imaging (MRI) of M-PTX. In a mouse oral cancer model, a permanent magnet placed on the body surface adjacent to the tumor resulted in distinct accumulation of M-PTX, and the anti-cancer effect was greater than that of M-PTX without the magnet. We believe that this strategy may improve future cancer chemotherapy by providing conventional anti-cancer drugs with novel functionalities such as magnet-guided drug delivery or MRI-based visualization/quantitation of drug distribution.
Collapse
|
29
|
Falone S, Santini S, Cordone V, Di Emidio G, Tatone C, Cacchio M, Amicarelli F. Extremely Low-Frequency Magnetic Fields and Redox-Responsive Pathways Linked to Cancer Drug Resistance: Insights from Co-Exposure-Based In Vitro Studies. Front Public Health 2018. [PMID: 29527520 PMCID: PMC5829633 DOI: 10.3389/fpubh.2018.00033] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Electrical devices currently used in clinical practice and common household equipments generate extremely low-frequency magnetic fields (ELF-MF) that were classified by the International Agency for Research on Cancer as “possible carcinogenic.” Assuming that ELF-MF plays a role in the carcinogenic process without inducing direct genomic alterations, ELF-MF may be involved in the promotion or progression of cancers. In particular, ELF-MF-induced responses are suspected to activate redox-responsive intracellular signaling or detoxification scavenging systems. In fact, improved protection against oxidative stress and redox-active xenobiotics is thought to provide critical proliferative and survival advantage in tumors. On this basis, an ever-growing research activity worldwide is attempting to establish whether tumor cells may develop multidrug resistance through the activation of essential cytoprotective networks in the presence of ELF fields, and how this might trigger relevant changes in tumor phenotype. This review builds a framework around how the activity of redox-responsive mediators may be controlled by co-exposure to ELF-MF and reactive oxygen species-generating agents in tumor and cancer cells, in order to clarify whether and how such potential molecular targets could help to minimize or neutralize the functional interaction between ELF-MF and malignancies.
Collapse
Affiliation(s)
- Stefano Falone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Silvano Santini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Valeria Cordone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giovanna Di Emidio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Carla Tatone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marisa Cacchio
- Department of Neurosciences, Imaging and Clinical Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Fernanda Amicarelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Institute of Translational Pharmacology (IFT)-National Research Council (CNR), L'Aquila, Italy
| |
Collapse
|
30
|
Gao S, Zheng M, Ren X, Tang Y, Liang X. Local hyperthermia in head and neck cancer: mechanism, application and advance. Oncotarget 2018; 7:57367-57378. [PMID: 27384678 PMCID: PMC5302995 DOI: 10.18632/oncotarget.10350] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 06/16/2016] [Indexed: 02/05/2023] Open
Abstract
Local hyperthermia (HT), particularly in conjunction with surgery, radiotherapy and chemotherapy was useful for the treatment of human malignant tumors including head and neck cancer. However, at present it suffered from many limitations such as thermal dose control, target treatment regions and discrimination between healthy and cancer cells. Recent developments in nanotechnology have introduced novel and smart therapeutic nanomaterials to local HT of head and neck cancer that basically take advantage of various targeting approaches. The aim of this paper is to give a brief review of the mechanism, methods and clinical applications of local HT in head and neck cancer, mainly focusing on photothermal therapy (PTT) and nanoparticle-based hyperthermia.
Collapse
Affiliation(s)
- Shiyu Gao
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Min Zheng
- Department of Stomatology, Zhoushan Hospital, Zhoushan, China
| | - Xiaohua Ren
- Department of Stomatology, Sichuan Medical Science Academy and Sichuan Provincial People's Hospital, Chengdu, China
| | - Yaling Tang
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinhua Liang
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Browning RJ, Reardon PJT, Parhizkar M, Pedley RB, Edirisinghe M, Knowles JC, Stride E. Drug Delivery Strategies for Platinum-Based Chemotherapy. ACS NANO 2017; 11:8560-8578. [PMID: 28829568 DOI: 10.1021/acsnano.7b04092] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Few chemotherapeutics have had such an impact on cancer management as cis-diamminedichloridoplatinum(II) (CDDP), also known as cisplatin. The first member of the platinum-based drug family, CDDP's potent toxicity in disrupting DNA replication has led to its widespread use in multidrug therapies, with particular benefit in patients with testicular cancers. However, CDDP also produces significant side effects that limit the maximum systemic dose. Various strategies have been developed to address this challenge including encapsulation within micro- or nanocarriers and the use of external stimuli such as ultrasound to promote uptake and release. The aim of this review is to look at these strategies and recent scientific and clinical developments.
Collapse
Affiliation(s)
- Richard J Browning
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford , Oxford OX1 2JD, United Kingdom
| | | | | | | | | | - Jonathan C Knowles
- Department of Nanobiomedical Science and BK21 Plus NBM, Global Research Center for Regenerative Medicine, Dankook University , 518-10 Anseo-dong, Dongnam-gu, Cheonan, Chungcheongnam-do, Republic of Korea
- The Discoveries Centre for Regenerative and Precision Medicine, UCL Campus , Gower Street, London WC1E 6BT, United Kingdom
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford , Oxford OX1 2JD, United Kingdom
| |
Collapse
|
32
|
Ota T, Nishida Y, Ikuta K, Kato R, Kozawa E, Hamada S, Sakai T, Ishiguro N. Heat-stimuli-enhanced osteogenesis using clinically available biomaterials. PLoS One 2017; 12:e0181404. [PMID: 28719650 PMCID: PMC5515435 DOI: 10.1371/journal.pone.0181404] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/02/2017] [Indexed: 11/18/2022] Open
Abstract
A recent study reported that heat stress stimulates osteogenesis in an in vivo rat model using alginate gel and magnetite cationic liposomes. However, for clinical use, the efficacy for promoting osteogenesis needs to be investigated using clinically approved materials, and preferably with animals larger than rats. The aim of this study was to evaluate multiple heat stimuli-triggered osteogenesis in rat tibial defect models using already clinically applicable materials (Resovist® and REGENOS®) and determine the efficacy also in the rabbit. Fifty-eight rats and 10 rabbits were divided into two groups, respectively, with or without hyperthermia treatment at 45°C for 15 min. (hyperthermia; 20 rats once a week, 8 rats three times a week, 5 rabbits once a week, control; 30 rats and 5 rabbits). Micro-CT assessment at 4 weeks revealed that a significantly stimulated osteogenesis was observed in the once a week group of both rats and rabbits as compared to the control group (p = 0.018 and 0.036, respectively). In contrast, the three times a week group did not show enhanced osteogenesis. Histological examination and image analysis showed consistent results in which the area of mineralized bone formation in the once a week hyperthermia group was significantly increased compared with that in the control group at four weeks (rat; p = 0.026, rabbit; p = 0.031). Newly formed bone was observed in the grafted materials from the periphery toward the center, and more osteoclasts were found in the once a week group. Heat stress also induced enhanced alkaline phosphatase expression in cultured osteoblastic cells, MC3T3, in vitro (p = 0.03). On the other hand, heat stress had no obvious effects on chondrogenic differentiation using ATDC5 cells. Our study demonstrates that heat-stimuli with clinically applicable novel heating materials can promote significant osteogenesis, and may thus be a promising treatment option for diseases associated with bone defects.
Collapse
Affiliation(s)
- Takehiro Ota
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| | - Yoshihiro Nishida
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
- * E-mail:
| | - Kunihiro Ikuta
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| | - Ryuji Kato
- Department of Basic Medical Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Eiji Kozawa
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| | - Shunsuke Hamada
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| | - Tomohisa Sakai
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| | - Naoki Ishiguro
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| |
Collapse
|
33
|
Hyperthermia and chemotherapy using Fe(Salen) nanoparticles might impact glioblastoma treatment. Sci Rep 2017; 7:42783. [PMID: 28218292 PMCID: PMC5316938 DOI: 10.1038/srep42783] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 01/16/2017] [Indexed: 12/03/2022] Open
Abstract
We previously reported that μ-oxo N,N’-bis(salicylidene)ethylenediamine iron [Fe(Salen)], a magnetic organic compound, has direct anti-tumor activity, and generates heat in an alternating magnetic field (AMF). We showed that Fe(Salen) nanoparticles are useful for combined hyperthermia-chemotherapy of tongue cancer. Here, we have examined the effect of Fe(Salen) on human glioblastoma (GB). Fe(Salen) showed in vitro anti-tumor activity towards several human GB cell lines. It inhibited cell proliferation, and its apoptosis-inducing activity was greater than that of clinically used drugs. Fe(Salen) also showed in vivo anti-tumor activity in the mouse brain. We evaluated the drug distribution and systemic side effects of intracerebrally injected Fe(Salen) nanoparticles in rats. Further, to examine whether hyperthermia, which was induced by exposing Fe(Salen) nanoparticles to AMF, enhanced the intrinsic anti-tumor effect of Fe(Salen), we used a mouse model grafted with U251 cells on the left leg. Fe(Salen), BCNU, or normal saline was injected into the tumor in the presence or absence of AMF exposure. The combination of Fe(Salen) injection and AMF exposure showed a greater anti-tumor effect than did either Fe(Salen) or BCNU alone. Our results indicate that hyperthermia and chemotherapy with single-drug nanoparticles could be done for GB treatment.
Collapse
|
34
|
Karponis D, Azzawi M, Seifalian A. An arsenal of magnetic nanoparticles; perspectives in the treatment of cancer. Nanomedicine (Lond) 2016; 11:2215-32. [DOI: 10.2217/nnm-2016-0113] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Nanomedicine is an emerging field, which constitutes a new direction in the treatment of cancer. Magnetic nanoparticles (MNPs) can circumvent vascular tissue to concentrate at the site of the tumor. Under the influence of an external, alternating magnetic field, MNPs generate high temperatures within the tumor and ablate malignant cells while inflicting minimal damage to healthy host tissue. Due to their theranostic properties, they constitute a promising candidate for the treatment of cancer. A critical review of the type, size and therapeutic effect of different MNPs is presented, following an appraisal of the literature in the last 5 years. This is a multibillion dollar industry, with a few studies moving to clinical trials within the next 5 years.
Collapse
Affiliation(s)
| | - May Azzawi
- School of Healthcare Science, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Alexander Seifalian
- Center for Nanotechnology & Regenerative Medicine, University College London, London, UK
- NanoRegMed Ltd, The London BioScience Innovation Center, London, UK
| |
Collapse
|
35
|
Sato I, Umemura M, Mitsudo K, Fukumura H, Kim JH, Hoshino Y, Nakashima H, Kioi M, Nakakaji R, Sato M, Fujita T, Yokoyama U, Okumura S, Oshiro H, Eguchi H, Tohnai I, Ishikawa Y. Simultaneous hyperthermia-chemotherapy with controlled drug delivery using single-drug nanoparticles. Sci Rep 2016; 6:24629. [PMID: 27103308 PMCID: PMC4840378 DOI: 10.1038/srep24629] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/31/2016] [Indexed: 11/09/2022] Open
Abstract
We previously investigated the utility of μ-oxo N,N'- bis(salicylidene)ethylenediamine iron (Fe(Salen)) nanoparticles as a new anti-cancer agent for magnet-guided delivery with anti-cancer activity. Fe(Salen) nanoparticles should rapidly heat up in an alternating magnetic field (AMF), and we hypothesized that these single-drug nanoparticles would be effective for combined hyperthermia-chemotherapy. Conventional hyperthermic particles are usually made of iron oxide, and thus cannot exhibit anti-cancer activity in the absence of an AMF. We found that Fe(Salen) nanoparticles induced apoptosis in cultured cancer cells, and that AMF exposure enhanced the apoptotic effect. Therefore, we evaluated the combined three-fold strategy, i.e., chemotherapy with Fe(Salen) nanoparticles, magnetically guided delivery of the nanoparticles to the tumor, and AMF-induced heating of the nanoparticles to induce local hyperthermia, in a rabbit model of tongue cancer. Intravenous administration of Fe(Salen) nanoparticles per se inhibited tumor growth before the other two modalities were applied. This inhibition was enhanced when a magnet was used to accumulate Fe(Salen) nanoparticles at the tongue. When an AMF was further applied (magnet-guided chemotherapy plus hyperthermia), the tumor masses were dramatically reduced. These results indicate that our strategy of combined hyperthermia-chemotherapy using Fe(Salen) nanoparticles specifically delivered with magnetic guidance represents a powerful new approach for cancer treatment.
Collapse
Affiliation(s)
- Itaru Sato
- Cardiovascular Research Institute, Yokohama City University, Graduate School of Medicine, Yokohama, 236-0004, Japan
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Masanari Umemura
- Cardiovascular Research Institute, Yokohama City University, Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Kenji Mitsudo
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hidenobu Fukumura
- Department of Orthopedic Surgery, Yokohama Touhoukai Hospital, Yokohama, 236-0031, Japan
| | - Jeong-Hwan Kim
- Cardiovascular Research Institute, Yokohama City University, Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Yujiro Hoshino
- Department of Environment and Natural Sciences, Graduate School of Environment and Information Sciences, Yokohama National University, Yokohama, 240-8501, Japan
| | - Hideyuki Nakashima
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Mitomu Kioi
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Rina Nakakaji
- Cardiovascular Research Institute, Yokohama City University, Graduate School of Medicine, Yokohama, 236-0004, Japan
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Motohiko Sato
- Department of Physiology, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Takayuki Fujita
- Cardiovascular Research Institute, Yokohama City University, Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Utako Yokoyama
- Cardiovascular Research Institute, Yokohama City University, Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Satoshi Okumura
- Tsurumi University School of Dental Medicine, Tsurumi, 230-8501, Japan
| | - Hisashi Oshiro
- Department of Pathology, Jichi Medical Universityepartment of Pathology, Tochigi, 329-2111, Japan
| | - Haruki Eguchi
- Advanced Applied Science Department, Research Laboratory, IHI Corporation, Yokohama, 235-8501, Japan
| | - Iwai Tohnai
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University, Graduate School of Medicine, Yokohama, 236-0004, Japan
| |
Collapse
|
36
|
Shiozawa K, Ohnuki Y, Mototani Y, Umeki D, Ito A, Saeki Y, Hanada N, Okumura S. Effects of food diameter on bite size per mouthful and chewing behavior. J Physiol Sci 2016; 66:93-8. [PMID: 26493202 PMCID: PMC10717241 DOI: 10.1007/s12576-015-0411-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/30/2015] [Indexed: 10/22/2022]
Abstract
Obesity is well known to be associated with a wide variety of illnesses, and is an increasing problem not only in developed countries but also in developing countries. It is well known that large bite size contributes to excess energy intake and obesity, whereas an increased number of chews before swallowing the food bolus is associated with suppression of obesity. However, the effect of food diameter on bite size per mouthful and on chewing behavior remains poorly understood. Here, we examined the effects of food diameter on bite size and chewing behavior using a masticatory counter during the mastication of stick-type biscuits having the same length (10 cm) and ingredients, but with four different diameters (3.0, 3.5, 4.0, and 8.0 mm). Bite length and bite weight per mouthful were similar among the 3.0, 3.5, and 4.0 mm groups. However, bite length in the 8.0 mm group was significantly smaller, whereas bite weight was significantly greater than in the 3.0/3.5 mm groups. Further, the number of chews gradually increased, whereas the number of chews per bite weight gradually decreased, with an increase of biscuit diameter. These results indicate that a smaller biscuit diameter is associated with a smaller bite weight per mouthful and a greater number of chews per bite weight. This is the first report to quantity the effect of food diameter on bite weight per mouthful and on chewing behavior; these results should be helpful in the design of effective, safe, and low-cost behavioral modification therapy to combat obesity.
Collapse
Affiliation(s)
- Kouichi Shiozawa
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501 Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501 Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501 Japan
| | - Daisuke Umeki
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501 Japan
- Department of Orthodontics, Tsurumi University of Dental Medicine, Yokohama, Japan
| | - Aiko Ito
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501 Japan
- Department of Orthodontics, Tsurumi University of Dental Medicine, Yokohama, Japan
| | - Yasutake Saeki
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501 Japan
| | - Nobuhiro Hanada
- Department of Translational Research, Tsurumi University of Dental Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501 Japan
| |
Collapse
|
37
|
Eguchi H, Umemura M, Kurotani R, Fukumura H, Sato I, Kim JH, Hoshino Y, Lee J, Amemiya N, Sato M, Hirata K, Singh DJ, Masuda T, Yamamoto M, Urano T, Yoshida K, Tanigaki K, Yamamoto M, Sato M, Inoue S, Aoki I, Ishikawa Y. A magnetic anti-cancer compound for magnet-guided delivery and magnetic resonance imaging. Sci Rep 2015; 5:9194. [PMID: 25779357 PMCID: PMC4361848 DOI: 10.1038/srep09194] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/04/2015] [Indexed: 01/07/2023] Open
Abstract
Research on controlled drug delivery for cancer chemotherapy has focused mainly on ways to deliver existing anti-cancer drug compounds to specified targets, e.g., by conjugating them with magnetic particles or encapsulating them in micelles. Here, we show that an iron-salen, i.e., μ-oxo N,N'- bis(salicylidene)ethylenediamine iron (Fe(Salen)), but not other metal salen derivatives, intrinsically exhibits both magnetic character and anti-cancer activity. X-Ray crystallographic analysis and first principles calculations based on the measured structure support this. It promoted apoptosis of various cancer cell lines, likely, via production of reactive oxygen species. In mouse leg tumor and tail melanoma models, Fe(Salen) delivery with magnet caused a robust decrease in tumor size, and the accumulation of Fe(Salen) was visualized by magnetic resonance imaging. Fe(Salen) is an anti-cancer compound with magnetic property, which is suitable for drug delivery and imaging. We believe such magnetic anti-cancer drugs have the potential to greatly advance cancer chemotherapy for new theranostics and drug-delivery strategies.
Collapse
Affiliation(s)
- Haruki Eguchi
- Cardiovascular Research Institute, Yokohama City University, Graduate School of Medicine, Yokohama, Japan,Advanced Applied Science Department, Research Laboratory, IHI Corporation, Yokohama, Japan,
| | - Masanari Umemura
- Cardiovascular Research Institute, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Reiko Kurotani
- Biochemical Engineering, Faculty of Engineering, Yamagata University, Yonezawa, Yamagata, Japan
| | | | - Itaru Sato
- Department of Oral Surgery, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Jeong-Hwan Kim
- Cardiovascular Research Institute, Yokohama City University, Graduate School of Medicine, Yokohama, Japan,Nanoparticles by Design Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Japan
| | - Yujiro Hoshino
- Department of Environment and Natural Sciences, Graduate School of Environment and Information Sciences, Yokohama National University, Yokohama, Japan
| | - Jin Lee
- Department of Radiology, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Naoyuki Amemiya
- Department of Electrical Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Motohiko Sato
- Department of Physiology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Kunio Hirata
- RIKEN, Research Infrastructure Group, SR Life Science Instrumentation Unit, Hyogo, Japan
| | - David J. Singh
- Materials Science and Technology Division, Oak Ridge National Laboratory, TN, USA
| | - Takatsugu Masuda
- Tokyo Neutron Science Laboratory, Institute for Solid State Physics, the University of Tokyo, Shirakata, Tokai, Japan
| | - Masahiro Yamamoto
- Department of Chemistry of Functional Molecules, Faculty of Science and Engineering, Konan University, Kobe, Japan
| | - Tsutomu Urano
- Yokohama City University, Graduate School, Yokohama, Japan
| | - Keiichiro Yoshida
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Katsumi Tanigaki
- WPI-AIMR & Department of Physics, Graduate School of Science, Tohoku University, Sendai, Japan
| | - Masaki Yamamoto
- RIKEN, Research Infrastructure Group, SR Life Science Instrumentation Unit, Hyogo, Japan
| | - Mamoru Sato
- Structural Biology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Seiichi Inoue
- Department of Environment and Natural Sciences, Graduate School of Environment and Information Sciences, Yokohama National University, Yokohama, Japan
| | - Ichio Aoki
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University, Graduate School of Medicine, Yokohama, Japan,
| |
Collapse
|