1
|
Ma L, Jiang X, Gao J. Revolutionizing rheumatoid arthritis therapy: harnessing cytomembrane biomimetic nanoparticles for novel treatment strategies. Drug Deliv Transl Res 2025; 15:66-83. [PMID: 38758497 DOI: 10.1007/s13346-024-01605-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2024] [Indexed: 05/18/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic immune disease with severe implications for joint health. The issue of non-specific drug distribution potentially limits the therapeutic efficacy and increases the risk associated with RA treatment. Researchers employed cytomembrane-coated biomimetic nanoparticles (NPs) to enhance the targeting delivery efficacy to meet the demand for drug accumulation within the affected joints. Furthermore, distinct cytomembranes offer unique functionalities, such as immune cell activation and augmented NP biocompatibility. In this review, the current strategies of RA treatments were summarized in detail, and then an overview of RA's pathogenesis and the methodologies for producing cytomembrane-coated biomimetic NPs was provided. The application of cytomembrane biomimetic NPs derived from various cell sources in RA therapy is explored, highlighting the distinctive attributes of individual cytomembranes as well as hybrid membrane configurations. Through this comprehensive assessment of cytomembrane biomimetic NPs, we elucidate the prospective applications and challenges in the realm of RA therapy, and the strategy of combined therapy is proposed. In the future, cytomembrane biomimetic NPs have a broad therapeutic prospect for RA.
Collapse
Affiliation(s)
- Lan Ma
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
- College of Pharmacy, Inner Mongolia Medical University, Chilechuan dairy economic development zone, Hohhot, Inner Mongolia Autonomous Region, 010110, China
| | - Xinchi Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Jianqing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
2
|
Dinakaran D, Moore-Palhares D, Yang F, Hill JB. Precision radiotherapy with molecular-profiling of CNS tumours. J Neurooncol 2024:10.1007/s11060-024-04911-z. [PMID: 39699761 DOI: 10.1007/s11060-024-04911-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
Diagnoses of CNS malignancies in the primary and metastatic setting have significantly advanced in the last decade with the advent of molecular pathology. Using a combination of immunohistochemistry, next-generation sequencing, and methylation profiling integrated with traditional histopathology, patient prognosis and disease characteristics can be understood to a much greater extent. This has recently manifested in predicting response to targeted drug therapies that are redefining management practices of CNS tumours. Radiotherapy, along with surgery, still remains an integral part of treating the majority of CNS tumours. However, the rapid advances in CNS molecular diagnostics have not yet been effectively translated into improving CNS radiotherapy. We explore several promising strategies under development to integrate molecular oncology into radiotherapy, and explore future directions that can serve to use molecular diagnostics to personalize radiotherapy. Evolving the management of CNS tumours with molecular profiling will be integral to supporting the future of precision radiotherapy.
Collapse
Affiliation(s)
- Deepak Dinakaran
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada.
- Department of Medical Biophysics and Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, 149 College Street, Suite 504, Toronto, ON, M5T 1P5, Canada.
| | - Daniel Moore-Palhares
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Fan Yang
- Radiation Oncology, Mayo Clinic Arizona, 5881 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Jordan B Hill
- Banner MD Anderson Cancer Center, 925 E. McDowell Rd, Phoenix, AZ, 85006, USA
| |
Collapse
|
3
|
Jasim SA, Salahdin OD, Malathi H, Sharma N, Rab SO, Aminov Z, Pramanik A, Mohammed IH, Jawad MA, Gabel BC. Targeting Hepatic Cancer Stem Cells (CSCs) and Related Drug Resistance by Small Interfering RNA (siRNA). Cell Biochem Biophys 2024; 82:3031-3051. [PMID: 39060914 DOI: 10.1007/s12013-024-01423-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 07/28/2024]
Abstract
Tumor recurrence after curative therapy and hepatocellular carcinoma (HCC) cells' resistance to conventional therapies is the reasons for the worse clinical results of HCC patients. A tiny population of cancer cells with a strong potential for self-renewal, differentiation, and tumorigenesis has been identified as cancer stem cells (CSCs). The discovery of CSC surface markers and the separation of CSC subpopulations from HCC cells have been made possible by recent developments in the study of hepatic (liver) CSCs. Hepatic CSC surface markers include epithelial cell adhesion molecules (EpCAM), CD133, CD90, CD13, CD44, OV-6, ALDH, and K19. CSCs have a significant influence on the development of cancer, invasiveness, self-renewal, metastasis, and drug resistance in HCC, and thus provide a therapeutic chance to treat HCC and avoid its recurrence. Therefore, it is essential to develop treatment approaches that specifically and effectively target hepatic stem cells. Given this, one potential treatment approach is to use particular small interfering RNA (siRNA) to target CSC, disrupting their behavior and microenvironment as well as changing their epigenetic state. The characteristics of CSCs in HCC are outlined in this study, along with new treatment approaches based on siRNA that may be used to target hepatic CSCs and overcome HCC resistance to traditional therapies.
Collapse
Affiliation(s)
| | | | - H Malathi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University, Bangalore, Karnataka, India
| | - Neha Sharma
- Chandigarh Pharmacy College, Chandigarh group of Colleges, Jhanjeri, 140307, Mohali, Punjab, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Zafar Aminov
- Department of Public Health and Healthcare management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Israa Hussein Mohammed
- College of nursing, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Mohammed Abed Jawad
- Department of Medical Laboratories Technology, Al-Nisour University College, Baghdad, Iraq
| | - Benien C Gabel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq
- Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
4
|
Schäfer M, Hildenbrand G, Hausmann M. Impact of Gold Nanoparticles and Ionizing Radiation on Whole Chromatin Organization as Detected by Single-Molecule Localization Microscopy. Int J Mol Sci 2024; 25:12843. [PMID: 39684554 DOI: 10.3390/ijms252312843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
In radiation tumor therapy, irradiation, on one hand, should cause cell death to the tumor. On the other hand, the surrounding non-tumor tissue should be maintained unaffected. Therefore, methods of local dose enhancements are highly interesting. Gold nanoparticles, which are preferentially uptaken by very-fast-proliferating tumor cells, may enhance damaging. However, the results in the literature obtained from cell culture and animal tissue experiments are very contradictory, i.e., only some experiments reveal increased cell killing but others do not. Thus, a better understanding of cellular mechanisms is required. Using the breast cancer cell model SkBr3, the effects of gold nanoparticles in combination with ionizing radiation on chromatin network organization were investigated by Single-Molecule Localization Microscopy (SMLM) and applications of mathematical topology calculations (e.g., Persistent Homology, Principal Component Analysis, etc.). The data reveal a dose and nanoparticle dependent re-organization of chromatin, although colony forming assays do not show a significant reduction of cell survival after the application of gold nanoparticles to the cells. In addition, the spatial organization of γH2AX clusters was elucidated, and characteristic changes were obtained depending on dose and gold nanoparticle application. The results indicate a complex response of ALU-related chromatin and heterochromatin organization correlating to ionizing radiation and gold nanoparticle incorporation. Such complex whole chromatin re-organization is usually associated with changes in genome function and supports the hypothesis that, with the application of gold nanoparticles, not only is DNA damage increasing but also the efficiency of DNA repair may be increased. The understanding of complex chromatin responses might help to improve the gold nanoparticle efficiency in radiation treatment.
Collapse
Affiliation(s)
- Myriam Schäfer
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
- Faculty of Engineering, University of Applied Sciences Aschaffenburg, Würzburger Str. 45, 63743 Aschaffenburg, Germany
| | - Georg Hildenbrand
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
- Faculty of Engineering, University of Applied Sciences Aschaffenburg, Würzburger Str. 45, 63743 Aschaffenburg, Germany
| | - Michael Hausmann
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
| |
Collapse
|
5
|
Schick U, Bourbonne V, Lucia F, Verry C. Use of nanoparticles in radiation oncology. Cancer Radiother 2024; 28:618-622. [PMID: 39379182 DOI: 10.1016/j.canrad.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 10/10/2024]
Abstract
Radiotherapy is a major therapeutic strategy for cancer treatment. Despite many technology advances in the last two decades, local control remains often suboptimal, especially in locally advanced tumours, which are often hypoxic, and radioresistant. In addition, irradiation of surrounding tissues and organs at risk usually precludes further dose escalation to minimize acute and late toxicities. Radiosensitizing agents such as chemotherapies targeting the DNA repair, or targeted monoclonal antibodies (cetuximab) have been shown to improve local control in many tumour types. More recently, radioenhancers have emerged as a new way to overcome the limitations of radiation. Here, we review the state of the art in this field and will focus on the past and ongoing clinical trials with the nanoparticles NBTXR3 and AGuIX®.
Collapse
Affiliation(s)
- Ulrike Schick
- Radiation Oncology Department, centre hospitalier universitaire de Brest, Brest, France; Inserm, LaTim UMR 1101, université de Bretagne occidentale, Brest, France.
| | - Vincent Bourbonne
- Radiation Oncology Department, centre hospitalier universitaire de Brest, Brest, France; Inserm, LaTim UMR 1101, université de Bretagne occidentale, Brest, France
| | - François Lucia
- Radiation Oncology Department, centre hospitalier universitaire de Brest, Brest, France; Inserm, LaTim UMR 1101, université de Bretagne occidentale, Brest, France
| | - Camille Verry
- Radiotherapy Department, Grenoble Alps University Hospital, Grenoble, France; Inserm UA7, University of Grenoble Alps, Synchrotron Radiation for Biomedical Research (ESRF), Grenoble, France
| |
Collapse
|
6
|
Khosravi Z, Kiani F. The Effect of the Continuous Care Model on Oral Health, Self-Efficacy, and Self-Care in Patients With Head and Neck Cancer Undergoing Radiotherapy: A Study Protocol for a Quasi-Experimental Study. Health Sci Rep 2024; 7:e70175. [PMID: 39524485 PMCID: PMC11544031 DOI: 10.1002/hsr2.70175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 09/27/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Background and Aims Head and neck cancer, with its unique challenges, often involves radiotherapy as a primary treatment. This can lead to complications affecting patients' well-being. A continuous care model offers potential benefits, but its effectiveness requires validation through rigorous studies. This protocol aims to evaluate its impact on patients undergoing head and neck cancer radiotherapy, contributing to improved care strategies and well-being. Methods The study is a double-arm and parallel-group quasi experimental study in which a 4-week intervention will be compared with usual care. A total of 70 eligible patients with head and neck cancer who are undergoing radiotherapy will be recruited to the intervention or control group. The patients in the intervention group will receive a continues care model designed for head and neck cancer patients in four phases of Orientation, Sensitization, Control, and Evaluation. The primary outcomes include dental plaque and gingival index, which will be measured by an approved dentist using The Gingival and Quigley-Hein index. The secondary outcomes are Oral hygiene self-efficacy and Dental and Oral Health Self-Care Behavior which will be assessed by the Oral and Dental Health Self-Efficacy and Dental and Oral Health Self-Care Behavior Questionnaires. Descriptive statistics will be used to describe variables. According to the types of variables, appropriate statistical tests, including two-sample t-tests, Χ 2, analysis of covariance, or linear regression will be performed. Conclusion In conclusion, this study aims to evaluate the impact of a continuous care model on head and neck cancer patients undergoing radiotherapy. Findings will contribute to enhancing care strategies and well-being in this population. Ethics and Dissemination The trial has been approved by the Research Ethics Committees of Zahedan University Of Medical Sciences. In this study, written consent will be obtained from all participants. The results will be presented to representative groups and published in peer-reviewed journals.
Collapse
Affiliation(s)
- Zohre Khosravi
- Department of Nursing, Nursing and Midwifery SchoolZahedan University of Medical SciencesZahedanIran
| | - Fatemeh Kiani
- Department of Nursing, Nursing and Midwifery SchoolZahedan University of Medical SciencesZahedanIran
| |
Collapse
|
7
|
Kot Y, Klochkov V, Prokopiuk V, Sedyh O, Tryfonyuk L, Grygorova G, Karpenko N, Tomchuk O, Kot K, Onishchenko A, Yefimova S, Tkachenko A. GdVO 4:Eu 3+ and LaVO 4:Eu 3+ Nanoparticles Exacerbate Oxidative Stress in L929 Cells: Potential Implications for Cancer Therapy. Int J Mol Sci 2024; 25:11687. [PMID: 39519237 PMCID: PMC11546343 DOI: 10.3390/ijms252111687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
The therapeutic potential of redox-active nanoscale materials as antioxidant- or reactive oxygen species (ROS)-inducing agents was intensely studied. Herein, we demonstrate that the synthesized and characterized GdVO4:Eu3+ and LaVO4:Eu3+ nanoparticles, which have been already shown to have redox-active, anti-inflammatory, antibacterial, and wound healing properties, both in vitro and in vivo, worsen oxidative stress of L929 cells triggered by hydrogen peroxide or tert-butyl hydroperoxide (tBuOOH) at the concentrations that are safe for intact L929 cells. This effect was observed upon internalization of the investigated nanosized materials and is associated with the cleavage of caspase-3 and caspase-9 without recruitment of caspase-8. Such changes in the caspase cascade indicate activation of the intrinsic caspase-9-dependent mitochondrial but not the extrinsic death, receptor-mediated, and caspase-8-dependent apoptotic pathway. The GdVO4:Eu3+ and LaVO4:Eu3+ nanoparticle-induced apoptosis of oxidatively compromised L929 cells is mediated by ROS overgeneration, Ca2+ overload, endoplasmic reticulum stress-associated JNK (c-Jun N-terminal kinase), and DNA damage-inducible transcript 3 (DDIT3). Our findings demonstrate that GdVO4:Eu3+ and LaVO4:Eu3+ nanoparticles aggravate the oxidative stress-induced damage to L929 cells, indicating that they might potentially be applied as anti-cancer agents.
Collapse
Affiliation(s)
- Yuriy Kot
- Department of Biochemistry, V.N. Karazin Kharkiv National, 4 Svobody Sq, 61022 Kharkiv, Ukraine; (Y.K.); (K.K.)
| | - Vladimir Klochkov
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky Ave, 61072 Kharkiv, Ukraine; (V.K.); (O.S.); (G.G.); (N.K.)
| | - Volodymyr Prokopiuk
- Department of Cryobiochemistry, Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, 23 Pereyaslavskaya Str., 61015 Kharkiv, Ukraine; (V.P.); (A.O.)
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, 4 Nauky Ave, 61022 Kharkiv, Ukraine
| | - Olha Sedyh
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky Ave, 61072 Kharkiv, Ukraine; (V.K.); (O.S.); (G.G.); (N.K.)
| | - Liliya Tryfonyuk
- Institute of Health, National University of Water and Environmental Engineering, 11 Soborna Str., 33028 Rivne, Ukraine;
| | - Ganna Grygorova
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky Ave, 61072 Kharkiv, Ukraine; (V.K.); (O.S.); (G.G.); (N.K.)
| | - Nina Karpenko
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky Ave, 61072 Kharkiv, Ukraine; (V.K.); (O.S.); (G.G.); (N.K.)
| | - Oleksandr Tomchuk
- ISIS Neutron and Muon Source, Rutherford Appleton Laboratory, Harwell Oxford, Didcot OX11 0QX, UK;
| | - Kateryna Kot
- Department of Biochemistry, V.N. Karazin Kharkiv National, 4 Svobody Sq, 61022 Kharkiv, Ukraine; (Y.K.); (K.K.)
| | - Anatolii Onishchenko
- Department of Cryobiochemistry, Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, 23 Pereyaslavskaya Str., 61015 Kharkiv, Ukraine; (V.P.); (A.O.)
| | - Svetlana Yefimova
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky Ave, 61072 Kharkiv, Ukraine; (V.K.); (O.S.); (G.G.); (N.K.)
| | - Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Průmyslová 595, 25250 Vestec, Czech Republic
| |
Collapse
|
8
|
Fadlallah H, El Masri J, Fakhereddine H, Youssef J, Chemaly C, Doughan S, Abou-Kheir W. Colorectal cancer: Recent advances in management and treatment. World J Clin Oncol 2024; 15:1136-1156. [PMID: 39351451 PMCID: PMC11438855 DOI: 10.5306/wjco.v15.i9.1136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 06/11/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide, and the second most common cause of cancer-related death. In 2020, the estimated number of deaths due to CRC was approximately 930000, accounting for 10% of all cancer deaths worldwide. Accordingly, there is a vast amount of ongoing research aiming to find new and improved treatment modalities for CRC that can potentially increase survival and decrease overall morbidity and mortality. Current management strategies for CRC include surgical procedures for resectable cases, and radiotherapy, chemotherapy, and immunotherapy, in addition to their combination, for non-resectable tumors. Despite these options, CRC remains incurable in 50% of cases. Nonetheless, significant improvements in research techniques have allowed for treatment approaches for CRC to be frequently updated, leading to the availability of new drugs and therapeutic strategies. This review summarizes the most recent therapeutic approaches for CRC, with special emphasis on new strategies that are currently being studied and have great potential to improve the prognosis and lifespan of patients with CRC.
Collapse
Affiliation(s)
- Hiba Fadlallah
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Jad El Masri
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Hiam Fakhereddine
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Joe Youssef
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Chrystelle Chemaly
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Samer Doughan
- Department of Surgery, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| |
Collapse
|
9
|
Arcambal A, Septembre-Malaterre A, Pesnel S, Morel AL, Gasque P, Begue M, Slama Y. The Potential of Human Pulmonary Mesenchymal Stem Cells as Vectors for Radiosensitizing Metallic Nanoparticles: An In Vitro Study. Cancers (Basel) 2024; 16:3239. [PMID: 39335210 PMCID: PMC11430180 DOI: 10.3390/cancers16183239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/08/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVES Metallic nanoparticles (NPs) exhibit interesting radiosensitizing effects, and finding a way to accurately deliver them appears to be crucial. Due to their tumor tropism, mesenchymal stem cells (MSCs) represent a strategic approach. Therefore, we aimed to evaluate the impact of core-shell Fe3O4@Au NPs on the functionality of human pulmonary MSCs (HPMSCs). METHODS/RESULTS The results showed that 100 µg/mL Fe3O4@Au NPs, accumulated in HPMSCs (revealed by Prussian blue staining), did not alter cell viability as assessed by cell counting, MTT, and LDH assays. However, caspase 9 and Bcl2 gene expression, evaluated by RT-qPCR, was regulated 72 h after exposure to the NPs. Moreover, the NPs also decreased proinflammatory cytokine/chemokine secretions, except for CXCL8 (ELISA). These modulations were associated with the downregulation of AMPK gene expression at 24 h. In contrast, the NPs did not modulate VEGF, PI3K, or PDGF gene expression. Nevertheless, a decrease in VEGF secretion was observed after 24 h of exposure to the NPs. Interestingly, the Fe3O4@Au NPs did not modulate Nrf2 gene expression, but they did regulate the expression of the genes encoding Nox4 and HMOX-1. Additionally, the NPs increased ROS production, suggesting a redox imbalance. CONCLUSIONS Finally, the Fe3O4@Au NPs did not affect the HPMSCs' viability or proangiogenic/tumorigenic markers. These findings are encouraging for investigating the effects of Fe3O4@Au NPs delivered by HPMSCs to tumor sites in combination with radiation.
Collapse
Affiliation(s)
- Angélique Arcambal
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Sainte-Clotilde Clinic, 127 Route de Bois de Nèfles, 97400 Saint-Denis, Reunion Island, France
| | - Axelle Septembre-Malaterre
- Unité de Recherche Etudes Pharmaco-Immunologiques (EPI), University of La Réunion, CHU of La Réunion, Felix Guyon Site, Allée des Topazes, SC11021, 97400 Saint-Denis, Reunion Island, France
| | - Sabrina Pesnel
- Torskal Nanosciences, 2 Rue Maxime Rivière, 97490 Sainte-Clotilde, Reunion Island, France
| | - Anne-Laure Morel
- Torskal Nanosciences, 2 Rue Maxime Rivière, 97490 Sainte-Clotilde, Reunion Island, France
| | - Philippe Gasque
- Unité de Recherche Etudes Pharmaco-Immunologiques (EPI), University of La Réunion, CHU of La Réunion, Felix Guyon Site, Allée des Topazes, SC11021, 97400 Saint-Denis, Reunion Island, France
| | - Mickael Begue
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Sainte-Clotilde Clinic, 127 Route de Bois de Nèfles, 97400 Saint-Denis, Reunion Island, France
- Department of Radiotherapy, Sainte-Clotilde Clinic, Clinifutur Group, 127 Route de Bois de Nèfles, 97400 Saint-Denis, Reunion Island, France
| | - Youssef Slama
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Sainte-Clotilde Clinic, 127 Route de Bois de Nèfles, 97400 Saint-Denis, Reunion Island, France
- Unité de Recherche Etudes Pharmaco-Immunologiques (EPI), University of La Réunion, CHU of La Réunion, Felix Guyon Site, Allée des Topazes, SC11021, 97400 Saint-Denis, Reunion Island, France
- Department of Radiotherapy, Sainte-Clotilde Clinic, Clinifutur Group, 127 Route de Bois de Nèfles, 97400 Saint-Denis, Reunion Island, France
| |
Collapse
|
10
|
Talaee O, Faghihi R, Rastegari B, Sina S. Enhanced radio-photodynamic therapy potential of advanced gold-based nanoclusters for breast cancer treatment. Radiol Phys Technol 2024; 17:703-714. [PMID: 39014282 DOI: 10.1007/s12194-024-00824-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/31/2024] [Accepted: 06/23/2024] [Indexed: 07/18/2024]
Abstract
The purpose of current study was to assess the impact of ALA-coated gold nanoclusters (Au NPs) on the combined therapeutic effects of radiotherapy (RT) and photodynamic therapy (PDT) on healthy MCF-10A and MCF-7 breast cancer cells. The Au NPs were covered with ALA using PEG polymer, resulting in the synthesis of Au@ALA NPs. The successful synthesis of the final NPs was confirmed through FTIR, XRD, TEM, and UV-Vis tests. MCF-10A and MCF-7 cell lines were treated with different concentrations of Au@ALA NPs and exposed to irradiation of 2 and 4 Gy (using MV X-ray) and 630 nm laser light irradiation. Cytotoxicity was assessed using a multifaceted approach involving the MTT assay, real-time PCR, and colony forming assay. The findings revealed that the damage inflicted by Au@ALA NPs on cancerous tissue was significantly greater than that on normal tissue. The cytotoxic effects of all experimental groups exhibited a direct correlation with increasing concentrations and radiation doses. The combination of Au@ALA NPs with RT doses of 2 and 4 Gy resulted in a reduction in cell viability by a factor of 1.58 (P = 0.001) and 1.73 (P = 0.004), respectively. Furthermore, the simultaneous intervention of NPs with PDT and RT at doses of 2 and 4 Gy led to a decrease in cell viability by a factor of 2.10 (P = 0.001) and 3.08 (P = 0.001) in turn. Furthermore, the real-time PCR and colonogenic assay results demonstrated that the combined treatment significantly increased phosphorylation of ATM and expression of TP53, indicating an adequate synergistic effect on breast cancer cells. The concurrent application of Au@ALA NPs in RT and PDT successfully enhanced the radiosensitization of breast cancer cells to megavoltage RT and PDT.
Collapse
Affiliation(s)
- Omid Talaee
- Nuclear Engineering Department, Shiraz University, Shiraz, Iran
| | - Reza Faghihi
- Nuclear Engineering Department, Shiraz University, Shiraz, Iran.
- Radiation Research Center, Shiraz University, Shiraz, Iran.
| | - Banafsheh Rastegari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sedigheh Sina
- Nuclear Engineering Department, Shiraz University, Shiraz, Iran
- Radiation Research Center, Shiraz University, Shiraz, Iran
| |
Collapse
|
11
|
Qiao R, Yuan Z, Yang M, Tang Z, He L, Chen T. Selenium-Doped Nanoheterojunctions for Highly Efficient Cancer Radiosensitization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402039. [PMID: 38828705 PMCID: PMC11304322 DOI: 10.1002/advs.202402039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/30/2024] [Indexed: 06/05/2024]
Abstract
Exploring efficient and low-toxicity radiosensitizers to break through the bottleneck of radiation tolerance, immunosuppression and poor prognosis remains one of the critical developmental challenges in radiotherapy. Nanoheterojunctions, due to their unique physicochemical properties, have demonstrated excellent radiosensitization effects in radiation energy deposition and in lifting tumor radiotherapy inhibition. Herein, they doped selenium (Se) into prussian blue (PB) to construct a nano-heterojunction (Se@PB), which could promote the increase of Fe2+/Fe3+ ratio and conversion of Se to a high valence state with Se introduction. The Fe2+-Se-Fe3+ electron transfer chain accelerates the rate of electron transfer on the surface of the nanoparticles, which in turn endows it with efficient X-ray energy transfer and electron transport capability, and enhances radiotherapy physical sensitivity. Furthermore, Se@PB induces glutathione (GSH) depletion and Fe2+ accumulation through pro-Fenton reaction, thereby disturbs the redox balance in tumor cells and enhances biochemical sensitivity of radiotherapy. As an excellent radiosensitizer, Se@PB effectively enhances X-ray induced mitochondrial dysfunction and DNA damage, thereby promotes cell apoptosis and synergistic cervical cancer radiotherapy. This study elucidates the radiosensitization mechanism of Se-doped nanoheterojunction from the perspective of the electron transfer chain and biochemistry reaction, which provides an efficient and low-toxic strategy in radiotherapy.
Collapse
Affiliation(s)
- Rui Qiao
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Zhongwen Yuan
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Meijin Yang
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Zhiying Tang
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Lizhen He
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Tianfeng Chen
- College of Chemistry and Materials ScienceDepartment of Oncology of The First Affiliated HospitalJinan UniversityGuangzhou510632China
| |
Collapse
|
12
|
Branco F, Cunha J, Mendes M, Vitorino C, Sousa JJ. Peptide-Hitchhiking for the Development of Nanosystems in Glioblastoma. ACS NANO 2024; 18:16359-16394. [PMID: 38861272 PMCID: PMC11223498 DOI: 10.1021/acsnano.4c01790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Glioblastoma (GBM) remains the epitome of aggressiveness and lethality in the spectrum of brain tumors, primarily due to the blood-brain barrier (BBB) that hinders effective treatment delivery, tumor heterogeneity, and the presence of treatment-resistant stem cells that contribute to tumor recurrence. Nanoparticles (NPs) have been used to overcome these obstacles by attaching targeting ligands to enhance therapeutic efficacy. Among these ligands, peptides stand out due to their ease of synthesis and high selectivity. This article aims to review single and multiligand strategies critically. In addition, it highlights other strategies that integrate the effects of external stimuli, biomimetic approaches, and chemical approaches as nanocatalytic medicine, revealing their significant potential in treating GBM with peptide-functionalized NPs. Alternative routes of parenteral administration, specifically nose-to-brain delivery and local treatment within the resected tumor cavity, are also discussed. Finally, an overview of the significant obstacles and potential strategies to overcome them are discussed to provide a perspective on this promising field of GBM therapy.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Joana Cunha
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria Mendes
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - João J. Sousa
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
13
|
Gonnelli A, Gerbé de Thoré M, Ermini ML, Frusca V, Zamborlin A, Signolle N, Bawa O, Clémenson C, Meziani L, Bergeron P, El-Azrak I, Sarogni P, Mugnaioli E, Giannini N, Drava G, Deutsch E, Paiar F, Mondini M, Voliani V. Nonpersistent Nanoarchitectures Enhance Concurrent Chemoradiotherapy in an Immunocompetent Orthotopic Model of HPV+ Head/Neck Carcinoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400949. [PMID: 38761135 DOI: 10.1002/adma.202400949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/13/2024] [Indexed: 05/20/2024]
Abstract
Cisplatin chemoradiotherapy (CRT) is the established standard of care for managing locally advanced human papillomavirus-positive head/neck carcinoma. The typically young patients may suffer serious and long-time side effects caused by the treatment, such as dysphagia, and hearing loss. Thus, ensuring a satisfactory post-treatment quality of life is paramount. One potential replacing approach to the classical CRT involves the combination of standard-dose radiotherapy and radiosensitizers such as noble metal nanoparticles (NPs). However, several concerns about size, shape, and biocompatibility limit the translation of metal nanomaterials to the clinical practice. Here, it is demonstrated that a new model of nonpersistent gold nanoarchitectures containing cisplatin (NAs-Cluster-CisPt) generates, in combination with radiotherapy, a significant in vivo tumor-reducing effect compared to the standard CRT, achieving a complete tumor clearance in 25% of the immunocompetent models that persist for 60 days. These findings, together with the negligible amount of metals recognized in the excretory organs, highlight that the concurrent administration of NAs-Cluster-CisPt and radiotherapy has the potential to overcome some clinical limitations associated to NP-based approaches while enhancing the treatment outcome with respect to standard CRT. Overall, despite further mechanistic investigations being essential, these data support the exploiting of nonpersistent metal-nanomaterial-mediated approaches for oral cancer management.
Collapse
Affiliation(s)
- Alessandra Gonnelli
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, Pisa, 56126, Italy
| | - Marine Gerbé de Thoré
- Gustave Roussy, INSERM U1030 Molecular Radiotherapy and Therapeutic Innovation, Université Paris Saclay, 114, rue Edouard Vaillant, Villejuif Cedex, 94805, France
| | - Maria Laura Ermini
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
| | - Valentina Frusca
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
- Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, Pisa, 56127, Italy
| | - Agata Zamborlin
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
- NEST-Scuola Normale Superiore, Piazza San Silvestro 12, Pisa, 56127, Italy
| | - Nicolas Signolle
- Gustave Roussy, Plateforme de pathologie expérimentale et translationnelle, UMS AMMICA, 114, rue Edouard Vaillant, Villejuif Cedex, 94805, France
| | - Olivia Bawa
- Gustave Roussy, Plateforme de pathologie expérimentale et translationnelle, UMS AMMICA, 114, rue Edouard Vaillant, Villejuif Cedex, 94805, France
| | - Céline Clémenson
- Gustave Roussy, INSERM U1030 Molecular Radiotherapy and Therapeutic Innovation, Université Paris Saclay, 114, rue Edouard Vaillant, Villejuif Cedex, 94805, France
| | - Lydia Meziani
- Gustave Roussy, INSERM U1030 Molecular Radiotherapy and Therapeutic Innovation, Université Paris Saclay, 114, rue Edouard Vaillant, Villejuif Cedex, 94805, France
| | - Paul Bergeron
- Gustave Roussy, INSERM U1030 Molecular Radiotherapy and Therapeutic Innovation, Université Paris Saclay, 114, rue Edouard Vaillant, Villejuif Cedex, 94805, France
| | - Ismail El-Azrak
- Gustave Roussy, INSERM U1030 Molecular Radiotherapy and Therapeutic Innovation, Université Paris Saclay, 114, rue Edouard Vaillant, Villejuif Cedex, 94805, France
| | - Patrizia Sarogni
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
| | - Enrico Mugnaioli
- Department of Earth Sciences, University of Pisa, Via S. Maria 53, Pisa, 56126, Italy
| | - Noemi Giannini
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, Pisa, 56126, Italy
| | - Giuliana Drava
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, Genoa, 16148, Italy
| | - Eric Deutsch
- Gustave Roussy, INSERM U1030 Molecular Radiotherapy and Therapeutic Innovation, Université Paris Saclay, 114, rue Edouard Vaillant, Villejuif Cedex, 94805, France
| | - Fabiola Paiar
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, Pisa, 56126, Italy
| | - Michele Mondini
- Gustave Roussy, INSERM U1030 Molecular Radiotherapy and Therapeutic Innovation, Université Paris Saclay, 114, rue Edouard Vaillant, Villejuif Cedex, 94805, France
| | - Valerio Voliani
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, Genoa, 16148, Italy
| |
Collapse
|
14
|
Mahmud MM, Pandey N, Winkles JA, Woodworth GF, Kim AJ. Toward the scale-up production of polymeric nanotherapeutics for cancer clinical trials. NANO TODAY 2024; 56:102314. [PMID: 38854931 PMCID: PMC11155436 DOI: 10.1016/j.nantod.2024.102314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Nanotherapeutics have gained significant attention for the treatment of numerous cancers, primarily because they can accumulate in and/or selectively target tumors leading to improved pharmacodynamics of encapsulated drugs. The flexibility to engineer the nanotherapeutic characteristics including size, morphology, drug release profiles, and surface properties make nanotherapeutics a unique platform for cancer drug formulation. Polymeric nanotherapeutics including micelles and dendrimers represent a large number of formulation strategies developed over the last decade. However, compared to liposomes and lipid-based nanotherapeutics, polymeric nanotherapeutics have had limited clinical translation from the laboratory. One of the key limitations of polymeric nanotherapeutics formulations for clinical translation has been the reproducibility in preparing consistent and homogeneous large-scale batches. In this review, we describe polymeric nanotherapeutics and discuss the most common laboratory and scale-up formulation methods, specifically those proposed for clinical cancer therapies. We also provide an overview of the major challenges and opportunities for scaling polymeric nanotherapeutics to clinical-grade formulations. Finally, we will review the regulatory requirements and challenges in advancing nanotherapeutics to the clinic.
Collapse
Affiliation(s)
- Md Musavvir Mahmud
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, MD, 20742, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nikhil Pandey
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jeffrey A. Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Graeme F. Woodworth
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, MD, 20742, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Anthony J. Kim
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, MD, 20742, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| |
Collapse
|
15
|
Garrigós MM, de Oliveira FA, Costa CJS, Rodrigues LR, Nucci MP, Alves ADH, Mamani JB, Rego GNDA, Munoz JM, Gamarra LF. Assessing the toxicity of one-step-synthesized PEG-coated gold nanoparticles: in vitro and in vivo studies. EINSTEIN-SAO PAULO 2024; 22:eAO0764. [PMID: 38775605 PMCID: PMC11081025 DOI: 10.31744/einstein_journal/2024ao0764] [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: 09/11/2023] [Accepted: 12/18/2023] [Indexed: 05/25/2024] Open
Abstract
OBJECTIVE To evaluate the in vitro and in vivo toxicities of polyethylene glycol-coated gold nanoparticles synthesized using a one-step process. METHODS Gold nanoparticles were prepared via a co-precipitation method using polyethylene glycol, and the synthesis product was characterized. For the in vitro evaluation, a flow cytometry analysis with Annexin V and iodide propidium staining was used to assess cytotoxicity in MG-63 cells labeled with 10, 50, and 100µg/mL of nanoparticle concentration. For the in vivo evaluation, nanoparticles were administered intraperitoneally at a dose of 10mg/kg dose in 10-week-old mice. Toxicity was assessed 24 hours and 7 days after administration via histopathological analysis of various tissues, as well as through renal, hepatic, and hematopoietic evaluations. RESULTS Synthesized nanoparticles exhibited different hydrodynamic sizes depending on the medium: 51.27±1.62nm in water and 268.12±28.45nm (0 hour) in culture medium. They demonstrated a maximum absorbance at 520nm and a zeta potential of -8.419mV. Cellular viability exceeded 90%, with less than 3% early apoptosis, 6% late apoptosis, and 1% necrosis across all labeling conditions, indicating minimal cytotoxicity differences. Histopathological analysis highlighted the accumulation of nanoparticles in the mesentery; however, no lesions or visible agglomeration was observed in the remaining tissues. Renal, hepatic, and hematopoietic analyses showed no significant differences at any time point. CONCLUSION Polyethylene glycol-coated gold nanoparticles exhibit extremely low toxicity and high biocompatibility, showing promise for future studies.
Collapse
Affiliation(s)
- Murilo Montenegro Garrigós
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | | | - Cícero Júlio Silva Costa
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Lucas Renan Rodrigues
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Mariana Penteado Nucci
- Hospital das ClínicasFaculdade MedicinaUniversidade de São PauloSão PauloSPBrazil LIM44 - Hospital das Clínicas, Faculdade Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Arielly da Hora Alves
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Javier Bustamante Mamani
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | | | - Juan Matheus Munoz
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Lionel Fernel Gamarra
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| |
Collapse
|
16
|
Bala VM, Lampropoulou DI, Grammatikaki S, Kouloulias V, Lagopati N, Aravantinos G, Gazouli M. Nanoparticle-Mediated Hyperthermia and Cytotoxicity Mechanisms in Cancer. Int J Mol Sci 2023; 25:296. [PMID: 38203467 PMCID: PMC10779099 DOI: 10.3390/ijms25010296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Hyperthermia has the potential to damage cancerous tissue by increasing the body temperature. However, targeting cancer cells whilst protecting the surrounding tissues is often challenging, especially when implemented in clinical practice. In this direction, there are data showing that the combination of nanotechnology and hyperthermia offers more successful penetration of nanoparticles in the tumor environment, thus allowing targeted hyperthermia in the region of interest. At the same time, unlike radiotherapy, the use of non-ionizing radiation makes hyperthermia an attractive therapeutic option. This review summarizes the existing literature regarding the use of hyperthermia and nanoparticles in cancer, with a focus on nanoparticle-induced cytotoxicity mechanisms.
Collapse
Affiliation(s)
| | | | - Stamatiki Grammatikaki
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (N.L.)
| | - Vassilios Kouloulias
- Radiation Oncology Unit, 2nd Department of Radiology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nefeli Lagopati
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (N.L.)
| | | | - Maria Gazouli
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (N.L.)
| |
Collapse
|
17
|
Najafi A, Keykhaee M, Kazemi MH, Karimi MY, Khorramdelazad H, Aghamohamadi N, Bolouri MR, Ghaffari-Nazari H, Mirsharif ES, Karimi M, Dehghan Manshadi HR, Mahdavi SR, Safari E, Jalali SA, Falak R, Khoobi M. Catalase-gold nanoaggregates manipulate the tumor microenvironment and enhance the effect of low-dose radiation therapy by reducing hypoxia. Biomed Pharmacother 2023; 167:115557. [PMID: 37757491 DOI: 10.1016/j.biopha.2023.115557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Radiotherapy as a standard method for cancer treatment faces tumor recurrence and antitumoral unresponsiveness. Suppressive tumor microenvironment (TME) and hypoxia are significant challenges affecting efficacy of radiotherapy. Herein, a versatile method is introduced for the preparation of pH-sensitive catalase-gold cross-linked nanoaggregate (Au@CAT) having acceptable stability and selective activity in tumor microenvironment. Combining Au@CAT with low-dose radiotherapy enhanced radiotherapy effects via polarizing protumoral immune cells to the antitumoral landscape. This therapeutic approach also attenuated hypoxia, confirmed by downregulating hypoxia hallmarks, such as hypoxia-inducible factor α-subunits (HIF-α), vascular endothelial growth factor (VEGF), and EGF. Catalase stability against protease digestion was improved significantly in Au@CAT compared to the free catalase. Moreover, minimal toxicity of Au@CAT on normal cells and increased reactive oxygen species (ROS) were confirmed in vitro compared with radiotherapy. Using the nanoaggregates combined with radiotherapy led to a significant reduction of immunosuppressive infiltrating cells such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (T-regs) compared to the other groups. While, this combined therapy could significantly increase the frequency of CD8+ cells as well as M1 to M2 macrophages (MQs) ratio. The combination therapy also reduced the tumor size and increased survival rate in mice models of colorectal cancer (CRC). Our results indicate that this innovative nanocomposite could be an excellent system for catalase delivery, manipulating the TME and providing a potential therapeutic strategy for treating CRC.
Collapse
Affiliation(s)
- Alireza Najafi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Keykhaee
- Department of Pharmaceutical Biomaterials and Medical Biomaterial Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Kazemi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Khorramdelazad
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Nazanin Aghamohamadi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Bolouri
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haniyeh Ghaffari-Nazari
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Milad Karimi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Seied Rabi Mahdavi
- Radiation Biology Research Center& Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Safari
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Jalali
- Immunology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mehdi Khoobi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Sell M, Lopes AR, Escudeiro M, Esteves B, Monteiro AR, Trindade T, Cruz-Lopes L. Application of Nanoparticles in Cancer Treatment: A Concise Review. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2887. [PMID: 37947732 PMCID: PMC10650201 DOI: 10.3390/nano13212887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023]
Abstract
Timely diagnosis and appropriate antitumoral treatments remain of utmost importance, since cancer remains a leading cause of death worldwide. Within this context, nanotechnology offers specific benefits in terms of cancer therapy by reducing its adverse effects and guiding drugs to selectively target cancer cells. In this comprehensive review, we have summarized the most relevant novel outcomes in the range of 2010-2023, covering the design and application of nanosystems for cancer therapy. We have established the general requirements for nanoparticles to be used in drug delivery and strategies for their uptake in tumor microenvironment and vasculature, including the reticuloendothelial system uptake and surface functionalization with protein corona. After a brief review of the classes of nanovectors, we have covered different classes of nanoparticles used in cancer therapies. First, the advances in the encapsulation of drugs (such as paclitaxel and fisetin) into nanoliposomes and nanoemulsions are described, as well as their relevance in current clinical trials. Then, polymeric nanoparticles are presented, namely the ones comprising poly lactic-co-glycolic acid, polyethylene glycol (and PEG dilemma) and dendrimers. The relevance of quantum dots in bioimaging is also covered, namely the systems with zinc sulfide and indium phosphide. Afterwards, we have reviewed gold nanoparticles (spheres and anisotropic) and their application in plasmon-induced photothermal therapy. The clinical relevance of iron oxide nanoparticles, such as magnetite and maghemite, has been analyzed in different fields, namely for magnetic resonance imaging, immunotherapy, hyperthermia, and drug delivery. Lastly, we have covered the recent advances in the systems using carbon nanomaterials, namely graphene oxide, carbon nanotubes, fullerenes, and carbon dots. Finally, we have compared the strategies of passive and active targeting of nanoparticles and their relevance in cancer theranostics. This review aims to be a (nano)mark on the ongoing journey towards realizing the remarkable potential of different nanoparticles in the realm of cancer therapeutics.
Collapse
Affiliation(s)
- Mariana Sell
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
| | - Ana Rita Lopes
- Faculty of Dental Medicine, Portuguese Catholic University, 3504-505 Viseu, Portugal;
| | - Maria Escudeiro
- Abel Salazar Biomedical Institute, University of Porto, 4050-313 Porto, Portugal;
| | - Bruno Esteves
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
- Centre for Natural Resources, Environment and Society-CERNAS-IPV Research Centre, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal
| | - Ana R. Monteiro
- Centro de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain;
| | - Tito Trindade
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Luísa Cruz-Lopes
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
- Centre for Natural Resources, Environment and Society-CERNAS-IPV Research Centre, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal
| |
Collapse
|
19
|
Babu B, Stoltz SA, Mittal A, Pawar S, Kolanthai E, Coathup M, Seal S. Inorganic Nanoparticles as Radiosensitizers for Cancer Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2873. [PMID: 37947718 PMCID: PMC10647410 DOI: 10.3390/nano13212873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
Nanotechnology has expanded what can be achieved in our approach to cancer treatment. The ability to produce and engineer functional nanoparticle formulations to elicit higher incidences of tumor cell radiolysis has resulted in substantial improvements in cancer cell eradication while also permitting multi-modal biomedical functionalities. These radiosensitive nanomaterials utilize material characteristics, such as radio-blocking/absorbing high-Z atomic number elements, to mediate localized effects from therapeutic irradiation. These materials thereby allow subsequent scattered or emitted radiation to produce direct (e.g., damage to genetic materials) or indirect (e.g., protein oxidation, reactive oxygen species formation) damage to tumor cells. Using nanomaterials that activate under certain physiologic conditions, such as the tumor microenvironment, can selectively target tumor cells. These characteristics, combined with biological interactions that can target the tumor environment, allow for localized radio-sensitization while mitigating damage to healthy cells. This review explores the various nanomaterial formulations utilized in cancer radiosensitivity research. Emphasis on inorganic nanomaterials showcases the specific material characteristics that enable higher incidences of radiation while ensuring localized cancer targeting based on tumor microenvironment activation. The aim of this review is to guide future research in cancer radiosensitization using nanomaterial formulations and to detail common approaches to its treatment, as well as their relations to commonly implemented radiotherapy techniques.
Collapse
Affiliation(s)
- Balaashwin Babu
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
| | - Samantha Archer Stoltz
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Agastya Mittal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Shreya Pawar
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
| | - Melanie Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA;
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Nanoscience Technology Center, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
20
|
Li J, Wang Q, Xia G, Adilijiang N, Li Y, Hou Z, Fan Z, Li J. Recent Advances in Targeted Drug Delivery Strategy for Enhancing Oncotherapy. Pharmaceutics 2023; 15:2233. [PMID: 37765202 PMCID: PMC10534854 DOI: 10.3390/pharmaceutics15092233] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/04/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Targeted drug delivery is a precise and effective strategy in oncotherapy that can accurately deliver drugs to tumor cells or tissues to enhance their therapeutic effect and, meanwhile, weaken their undesirable side effects on normal cells or tissues. In this research field, a large number of researchers have achieved significant breakthroughs and advances in oncotherapy. Typically, nanocarriers as a promising drug delivery strategy can effectively deliver drugs to the tumor site through enhanced permeability and retention (EPR) effect-mediated passive targeting and various types of receptor-mediated active targeting, respectively. Herein, we review recent targeted drug delivery strategies and technologies for enhancing oncotherapy. In addition, we also review two mainstream drug delivery strategies, passive and active targeting, based on various nanocarriers for enhancing tumor therapy. Meanwhile, a comparison and combination of passive and active targeting are also carried out. Furthermore, we discuss the associated challenges of passive and active targeted drug delivery strategies and the prospects for further study.
Collapse
Affiliation(s)
- Jianmin Li
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| | - Qingluo Wang
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| | - Guoyu Xia
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| | - Nigela Adilijiang
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| | - Ying Li
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Zhenqing Hou
- College of Materials, Xiamen University, Xiamen 361002, China;
| | - Zhongxiong Fan
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| | - Jinyao Li
- College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (J.L.); (Q.W.); (G.X.); (N.A.)
| |
Collapse
|
21
|
Wu W, Wang Y, Xie J, Fan S. Empagliflozin: a potential anticancer drug. Discov Oncol 2023; 14:127. [PMID: 37436535 DOI: 10.1007/s12672-023-00719-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/06/2023] [Indexed: 07/13/2023] Open
Abstract
Empagliflozin, a sodium-glucose cotransporter 2 (SGLT2) inhibitor, is a highly effective and well-tolerated antidiabetic drug. In addition to hypoglycemic effects, empagliflozin has many other effects, such as being hypotensive and cardioprotective. It also has anti-inflammatory and antioxidative stress effects in diabetic nephropathy. Several studies have shown that empagliflozin has anticancer effects. SGLT2 is expressed in a variety of cancer cell lines. The SGLT2 inhibitor empagliflozin has significant inhibitory effects on certain types of tumor cells, such as inhibition of proliferation, migration and induction of apoptosis. In conclusion, empagliflozin has promising applications in cancer therapy as a drug for the treatment of diabetes and heart failure. This article provides a brief review of the anticancer effects of empagliflozin.
Collapse
Affiliation(s)
- Wenwen Wu
- School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Yanyan Wang
- Department of Ultrasonic Medicine, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, 221000, China
| | - Jun Xie
- School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China.
| | - Shaohua Fan
- School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China.
| |
Collapse
|