1
|
Zhang R, Guo L, Li Q, Liang Y, Liao Y, Xu H, Liu C, Zhou G, Wang L, Xu S, Yuan M. Biodegradable Carrier-Free Nanomedicine via Self-Assembly of Pure Drug Molecules for Triple Sensitization of Radiotherapy. ACS NANO 2025; 19:16355-16371. [PMID: 40265972 DOI: 10.1021/acsnano.4c15736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Radiotherapy (RT) has been one of the most widely applied cancer treatments, while radiotherapy resistance remains a major limitation. Herein, we synthesized a biodegradable AID nanomedicine incorporating atovaquone (ATO), new indocyanine green (IR820), and doxorubicin (DOX) via π-π stacking and hydrophobic interactions, along with high drug loading efficiency and long-term stability. The AID nanomedicine effectively reduces the activity of mitochondrial electron transport chain complexes I/II/III/IV/V, disrupts the mitochondrial oxidative respiratory chain, and decreases oxygen consumption, thereby alleviating the hypoxic microenvironment within the tumor. Moreover, mild hyperthermia induced by IR820 improves intratumor blood flow, thereby enhancing the radiotherapeutic efficiency. Additionally, DOX-triggered chemotherapy further sensitizes the tumor to radiotherapy, achieving triple sensitization. Our findings demonstrate that AID nanomedicine, combined with near-infrared (NIR) and radiotherapy, significantly suppresses tumor growth in vivo without noticeable side effects. In conclusion, our work presents a self-assembling nanomedicine with excellent biocompatibility, showing great potential for future development in triple radiotherapy sensitization.
Collapse
Affiliation(s)
- Rongjun Zhang
- Department of Laboratory Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, Hangzhou 310003, China
- Institute of Laboratory Medicine, Zhejiang University, Hangzhou 310003, China
| | - Lihao Guo
- Precision Research Center for Refractory Diseases, Shanghai Jiao Tong University Pioneer Research Institute for Molecular and Cell Therapies, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Qingjiao Li
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China
| | - Yin Liang
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China
| | - Yingying Liao
- Precision Research Center for Refractory Diseases, Shanghai Jiao Tong University Pioneer Research Institute for Molecular and Cell Therapies, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Huibin Xu
- Precision Research Center for Refractory Diseases, Shanghai Jiao Tong University Pioneer Research Institute for Molecular and Cell Therapies, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Chutong Liu
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China
| | - Gandong Zhou
- Precision Research Center for Refractory Diseases, Shanghai Jiao Tong University Pioneer Research Institute for Molecular and Cell Therapies, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Li Wang
- Precision Research Center for Refractory Diseases, Shanghai Jiao Tong University Pioneer Research Institute for Molecular and Cell Therapies, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Shuxiang Xu
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Miaomiao Yuan
- Precision Research Center for Refractory Diseases, Shanghai Jiao Tong University Pioneer Research Institute for Molecular and Cell Therapies, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China
| |
Collapse
|
2
|
Valable S, Césaire M, Lecrosnier K, Gilbert A, Tudor M, Vares G, Hamdi DH, Diouf OB, Nguyen Pham T, Coupey J, Thariat J, Lesueur P, Pérès EA, Aury-Landas J, Nikitaki Z, Haghdoost S, Laurent C, Poully JC, Balosso J, Bernaudin M, Savu DI, Chevalier F. Particle Therapy to Overcome Cancer Radiation Resistance: "ARCHADE" Consortium Updates in Radiation Biology. Cancers (Basel) 2025; 17:1580. [PMID: 40361506 PMCID: PMC12071746 DOI: 10.3390/cancers17091580] [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: 03/31/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Radiation therapy is a medical treatment that uses high doses of radiation to kill or damage cancer cells. It works by damaging the DNA within the cancer cells, ultimately causing cell death. Radiotherapy can be used as a primary treatment, adjuvant treatment in combination with surgery or chemotherapy or palliative treatment to relieve symptoms in advanced cancer stages. Radiation therapy is constantly improving in order to enhance the effect on cancer cells and reduce the side effects on healthy tissues. Our results clearly demonstrate that proton therapy and, even more, carbon ion therapy appear as promising alternatives to overcome the radioresistance of various tumors thanks to less dependency on oxygen and a better ability to kill cancer stem cells. Interestingly, hadrons also retain the advantages of radiosensitization approaches. These data confirm the great ability of hadrons to spare healthy tissue near the tumor via various mechanisms (reduced lymphopenia, bystander effect, etc.). Technology and machine improvements such as image-guided radiotherapy or particle therapies can improve treatment quality and efficacy (dose deposition and biological effect) in tumors while increasingly sparing healthy tissues. Radiation biology can help to understand how cancer cells resist radiation (hypoxia, DNA repair mechanisms, stem cell status, cell cycle position, etc.), how normal tissues may display sensitivity to radiation and how radiation effects can be increased with either radiosensitizers or accelerated particles. All these research topics are under investigation within the ARCHADE research community in France. By focusing on these areas, radiotherapy can become more effective, targeted and safe, enhancing the overall treatment experience and outcomes for cancer patients. Our goal is to provide biological evidence of the therapeutic advantages of hadrontherapy, according to the tumor characteristics. This article aims to give an updated view of our research in radiation biology within the frame of the French "ARCHADE association" and new perspectives on research and treatment with the C400 multi-ions accelerator prototype.
Collapse
Affiliation(s)
- Samuel Valable
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Mathieu Césaire
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| | - Kilian Lecrosnier
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| | - Antoine Gilbert
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| | - Mihaela Tudor
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania;
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania
| | - Guillaume Vares
- Autorité de Sûreté Nucléaire et de Radioprotection (ASNR), PSE-SANTE/SESANE/LRTOX, 92260 Fontenay-aux-Roses, France;
| | - Dounia Houria Hamdi
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| | - Ousseynou Ben Diouf
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
- Mixed Research Exploration and Diagnosis (UMRED), UFR-Healthy, Iba Der THIAM University of Thies, Thies BP A967, Senegal
| | - Thao Nguyen Pham
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Julie Coupey
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Juliette Thariat
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Radiation Oncology Department, Centre François Baclesse, 14000 Caen, France
| | - Paul Lesueur
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Radiation Oncology Department, Centre Guillaume Le Conquérant, 76600 Le Havre, France
| | - Elodie Anne Pérès
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Juliette Aury-Landas
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Zacharenia Nikitaki
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
- Normandie University, UNICAEN, UNIROUEN, ABTE UR4651, Cancer Center François Baclesse, 14000 Caen, France
| | - Siamak Haghdoost
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
- Normandie University, UNICAEN, UNIROUEN, ABTE UR4651, Cancer Center François Baclesse, 14000 Caen, France
| | - Carine Laurent
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Normandie University, UNICAEN, UNIROUEN, ABTE UR4651, Cancer Center François Baclesse, 14000 Caen, France
| | - Jean-Christophe Poully
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| | - Jacques Balosso
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Radiation Oncology Department, Centre François Baclesse, 14000 Caen, France
| | - Myriam Bernaudin
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Diana I. Savu
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania;
| | - François Chevalier
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| |
Collapse
|
3
|
Ma R, Yang Z, Miao X, Hu J, Zhang T, Ma LT, Lin JY, Zhao LN. Dual-Mode Radiosensitization of Esophageal Squamous Cell Carcinoma via SOCS6-Loaded Virus-Inspired Manganese-Bismuth Bimetallic Oxide Nanoparticles. Adv Healthc Mater 2025; 14:e2404737. [PMID: 40159874 DOI: 10.1002/adhm.202404737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/21/2025] [Indexed: 04/02/2025]
Abstract
Radioresistance poses a significant obstacle to controlling the recurrence of esophageal squamous cell carcinoma (ESCC) during radiotherapy. It is urgent to develop innovative radiosensitization strategies to improve the prognosis of patients with ESCC. Here, a novel dual-mode radiosensitizer: a virus-inspired hollow mesoporous manganese-bismuth bimetallic oxide nanoparticles (vHMMn-Bi) encapsulating the radiosensitizing plasmids (suppressor of cytokine signaling 6, SOCS6) is developed, designed to significantly amplify ESCC radiotherapy under hypoxic conditions. After intravenous injection, the SOCS6@vHMMn-Bi nanoparticles can be efficiently delivered to the tumor site and rapidly invade tumor cells by virus-like surface-assisted adhesion. Under X-ray irradiation, the nanoparticles exhibits a unique dual-mode sensitization effect, encompassing exogenous and endogenous mechanisms, thereby significantly augmenting the ESCC radiotherapeutic effectiveness. First, the Bi2O3 within the shell can enhance the radiosensitivity owing to its robust X-ray attenuation characteristics. Second, the SOCS6 released from the interior can inhibit both HIF-1α and JAK2/STAT3 signaling pathways, triggering ROS upregulation and intensifying radiation-mediated DNA damage inside ESCC cells. Furthermore, the shell employs MnO2 to catalyze the decomposition of endogenous H2O2 to increase oxygen generation, alleviating hypoxia within the tumor microenvironment. These nanoparticles demonstrates considerable potential as dual-mode radiosensitizers with no systemic toxicity and low immunogenicity for amplifying radiotherapeutic efficacy in ESCC.
Collapse
Affiliation(s)
- Rui Ma
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Zhi Yang
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Xia Miao
- Department of Radiation Medicine, The Faculty of Preventive Medicine, The Fourth Military Medical University, Xi'an, 710000, P. R. China
| | - Jing Hu
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Te Zhang
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Li-Tian Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710000, P. R. China
| | - Jin-Yan Lin
- Department of Occupational and Environmental Health, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710000, P. R. China
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, P. R. China
| | - Li-Na Zhao
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| |
Collapse
|
4
|
Ryzhov V, Marchenko Y, Deriglazov V, Yudintceva N, Smirnov O, Arutyunyan A, Shtam T, Ivanov E, Combs SE, Shevtsov M. Nonlinear Magnetic Response Measurements in Study of Magnetic Nanoparticles Uptake by Mesenchymal Stem Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:675. [PMID: 40358293 PMCID: PMC12073591 DOI: 10.3390/nano15090675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
Stem cells therapies offer a promising approach in translational oncology, as well as in regenerative medicine due to the tropism of these cells to the damage site. To track the distribution of stem cells, the latter could be labeled by MRI-sensitive superparamagnetic (SPM) iron oxide nanoparticles. In the current study, magnetic properties of the magnetic nanoparticles (MNPs) incorporated into the bone marrow-derived fetal mesenchymal stem cells (FetMSCs) were evaluated employing nonlinear magnetic response measurements. Synthesized dextran-coated iron oxide nanoparticles were additionally characterized by X-ray diffraction, transmission electron microscopy, and dynamic light scattering. The MNP uptake by the FetMSCs 24 h following coincubation was studied by longitudinal nonlinear response to weak alternating magnetic field with registration of the second harmonic of magnetization. Subsequent data processing using a formalism based on the numerical solution of the Fokker-Planck kinetic equation allowed us to determine magnetic and dynamic parameters and the state of MNPs in the cells, as well as in the culture medium. It was found that MNPs formed aggregates in the culture medium; they were absorbed by the cells during coincubation. The aggregates exhibited SPM regime in the medium, and the parameters of the MNP aggregates remained virtually unchanged in the cells, indicating the preservation of the aggregation state of MNPs inside the cells. This implies also the preservation of the organic shell of the nanoparticles inside FetMSCs. The accumulation of MNPs by mesenchymal stem cells gradually increased with the concentration of MNPs. Thus, the study confirmed that the labeling of MSCs with MNPs is an effective method for subsequent cell tracking as incorporated nanoparticles retain their magnetic properties.
Collapse
Affiliation(s)
- Vyacheslav Ryzhov
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova Roscha 1, 188300 Gatchina, Russia; (Y.M.); (V.D.); (O.S.); (A.A.); (T.S.)
| | - Yaroslav Marchenko
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova Roscha 1, 188300 Gatchina, Russia; (Y.M.); (V.D.); (O.S.); (A.A.); (T.S.)
| | - Vladimir Deriglazov
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova Roscha 1, 188300 Gatchina, Russia; (Y.M.); (V.D.); (O.S.); (A.A.); (T.S.)
| | - Natalia Yudintceva
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 St. Petersburg, Russia;
| | - Oleg Smirnov
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova Roscha 1, 188300 Gatchina, Russia; (Y.M.); (V.D.); (O.S.); (A.A.); (T.S.)
| | - Alexandr Arutyunyan
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova Roscha 1, 188300 Gatchina, Russia; (Y.M.); (V.D.); (O.S.); (A.A.); (T.S.)
| | - Tatiana Shtam
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova Roscha 1, 188300 Gatchina, Russia; (Y.M.); (V.D.); (O.S.); (A.A.); (T.S.)
| | - Evgenii Ivanov
- LLC “SPF “HELIX”, Sampsonievsky B. Prospect, 20 Litera A, 194044 St. Petersburg, Russia;
| | - Stephanie E. Combs
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany;
| | - Maxim Shevtsov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 St. Petersburg, Russia;
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany;
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 St. Petersburg, Russia
| |
Collapse
|
5
|
Zhang R, Chen M, Zhou H, Liu Y, Wang Y, Chen C, Li Y, Zeng J, Cui J, Duan R, Gao M. Eliminating Radioresistance With a Magnetic Ion-Generator by Simultaneously Augmenting DNA Damage and Diminishing Immunosuppression. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2406378. [PMID: 39996275 DOI: 10.1002/adma.202406378] [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: 05/04/2024] [Revised: 01/23/2025] [Indexed: 02/26/2025]
Abstract
Radiotherapy (RT) hinges on DNA damage-induced cancer cell death and the subsequent anti-tumor immunity. However, the efficacy of RT is curtailed by cell cycle heterogeneity and an immunosuppressive tumor microenvironment, which foster radioresistance. Here an ion generator-based RT enhancement strategy is demonstrated in a mouse model of the radioresistant tumor. The ion generator is degraded in the tumor microenvironment, resulting in iron-triggered ferroptosis that enhanced immunogenic cell death and a manganese-activated stimulator of interferon gene that reversed the immunosuppressive environment. As a result, the proposed strategy promotes dendritic cells maturity, augmentes CD8+ T cell infiltration of tumors, suppresses intratumoral myeloid-derived suppressor cells, and limits the M2 macrophages polarization, indicating the formation of an immunoreactive microenvironment. Significantly, this approach impedes the growth of not just primary, but also distal metastatic tumors. It is thus believed that the current ion generator provides a robust and enduring countermeasure to radioresistant cancer and its metastasis, with potential implications for enhancing the efficacy of RT in clinically resistant tumors.
Collapse
Affiliation(s)
- Ruru Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Mei Chen
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Hui Zhou
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Yan Liu
- The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yi Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Can Chen
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Yueping Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Jiabin Cui
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Ruixue Duan
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Mingyuan Gao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
- The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| |
Collapse
|
6
|
Fagundes DA, Leonel LV, Fernandez-Outon LE, Ardisson JD, dos Santos RG. Radiosensitization Induced by Magnetic Hyperthermia of PEGylated Nickel Ferrite Nanoparticles on Breast Cancer Cells. Int J Mol Sci 2025; 26:2706. [PMID: 40141347 PMCID: PMC11942335 DOI: 10.3390/ijms26062706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 03/28/2025] Open
Abstract
Magnetic hyperthermia can complement traditional cancer treatments by exploiting the greater heat sensitivity of tumor cells. This approach allows for localized action, increasing its therapeutic effectiveness. In this study, MCF-7 breast cancer cell radiosensitization, induced by the magnetic hyperthermia of PEGylated nickel ferrite magnetic nanoparticles (PEG-NiF MNPs), was evaluated by exposing the cells in the presence of MNPs to an alternating magnetic field followed by 60Co gamma irradiation. Superparamagnetic PEG-NiF MNPs (25.6 ± 0.5 nm) synthesized via the hydrothermal method exhibited a hydrodynamic size below 150 nm, a saturation magnetization of 53 emu·g-1, biocompatibility of up to 100 µg·mL-1, selectivity for breast cancer cells, and an up-to-fivefold increase in therapeutic efficacy of radiation. When combined with magnetic hyperthermia, this increase reached up-to-sevenfold. These results indicate that PEG-NiF MNPs are suitable thermal radiosensitization agents for breast cancer cells.
Collapse
Affiliation(s)
- Daniele A. Fagundes
- Centro de Desenvolvimento da Tecnologia Nuclear, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, Minas Gerais, Brazil; (L.V.L.); (J.D.A.); (R.G.d.S.)
| | - Liliam V. Leonel
- Centro de Desenvolvimento da Tecnologia Nuclear, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, Minas Gerais, Brazil; (L.V.L.); (J.D.A.); (R.G.d.S.)
| | - Luis E. Fernandez-Outon
- Departamento de Física, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, Minas Gerais, Brazil;
| | - José D. Ardisson
- Centro de Desenvolvimento da Tecnologia Nuclear, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, Minas Gerais, Brazil; (L.V.L.); (J.D.A.); (R.G.d.S.)
| | - Raquel G. dos Santos
- Centro de Desenvolvimento da Tecnologia Nuclear, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, Minas Gerais, Brazil; (L.V.L.); (J.D.A.); (R.G.d.S.)
| |
Collapse
|
7
|
Ding X, Zhang J, Wan S, Wang X, Wang Z, Pu K, Wang M, Cao Y, Weng L, Zhu H, Peng F, Chao J, Pei R, Leong DT, Wang L. Non-discriminating engineered masking of immuno-evasive ligands on tumour-derived extracellular vesicles enhances tumour vaccination outcomes. NATURE NANOTECHNOLOGY 2025; 20:156-166. [PMID: 39327512 DOI: 10.1038/s41565-024-01783-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 08/08/2024] [Indexed: 09/28/2024]
Abstract
The success of personalized cancer immunotherapy depends on the initial tumour antigenic presentation to dendritic cells and macrophages. Tumour-derived extracellular vesicles (TEVs) contain abundant tumour antigenic molecules. The presence of anti-phagocytotic signals such as cluster of differentiation 47 (CD47) on the surface of the TEVs, however, leads to evasion of the same dendritic cells and macrophages. Here we show that iron oxide hydroxide nanocomposites can successfully mask TEV surfaces and unblock phagocytosis without affecting extracellular vesicles' elicited immune goals. After internalization, the mask disintegrates in the lysosome, releasing the tumour antigenic cargo. This triggers antigen presentation and promotes dendritic cell activation and maturation and macrophage reprogramming in animal models, leading to a drastic reduction of tumour volume and metastasis, and in human malignant pleural effusion clinical samples. This straightforward masking strategy eliminates the ubiquitous anti-phagocytosis block found in clinical samples and can be applied universally across all patient-specific TEVs as tumour antigenic agents for enhanced immunotherapy.
Collapse
Affiliation(s)
- Xianguang Ding
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China.
| | - JingJing Zhang
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Shuangshuang Wan
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Xu Wang
- Department of Anesthesiology and Intensive Care Unit Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiyu Wang
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Kefeng Pu
- CAS Key Laboratory of Nano-Bio Interface, Key Laboratory of Nanodevices and Applications, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Mao Wang
- CAS Key Laboratory of Nano-Bio Interface, Key Laboratory of Nanodevices and Applications, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Yi Cao
- CAS Key Laboratory of Nano-Bio Interface, Key Laboratory of Nanodevices and Applications, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Lixing Weng
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Houjuan Zhu
- Agency for Science Technology and Research, Singapore, Singapore
| | - Fei Peng
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Jie Chao
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Renjun Pei
- CAS Key Laboratory of Nano-Bio Interface, Key Laboratory of Nanodevices and Applications, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, Singapore.
| | - Lianhui Wang
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China.
| |
Collapse
|
8
|
Song M, Ivkov R, Korangath P. Dendritic cell activation by iron oxide nanoparticles depends on the extracellular environment. NANOSCALE ADVANCES 2024; 7:209-218. [PMID: 39569333 PMCID: PMC11575603 DOI: 10.1039/d4na00561a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024]
Abstract
Nanoparticles can exert immune modulating effects in a host depending on composition, mode of administration, and type of disease. Although the specific mechanisms of nanoparticle-induced immune responses remain unclear, their uptake by macrophages and other phagocytic innate immune cells is considered to be a key event. Our objective here was to ascertain if nanoparticle-mediated activation of dendritic cells (DCs) occurs in vitro or in vivo when exposed to hydroxyethyl starch-coated iron oxide nanoparticles. For the present studies, our choice of nanoparticles, animal model, and experimental design is motivated by our previously published observations that systemic exposure can induce antitumor adaptive immune responses in mouse models of metastatic breast cancer. Here, we began by assessing the potential toxicity of systemic exposure to commercially available starch-coated Bionized Nanoferrite® nanoparticles (BP) by measuring body weight, complete blood count, and enzyme parameters in healthy FVB/NJ mice after repeated BP dosing. We observed no evidence of toxicity at doses up to 25 mg Fe per mouse, five-fold higher than those used in subsequent in vivo experiments. We then measured the expression of surface maturation markers (CD86, MHC II) in DCs incubated with BP in vitro. Although DCs cultured with BP revealed high levels of nanoparticle uptake, neither JAWSII dendritic cells nor bone marrow derived dendritic cells (BMDCs) showed significant changes in marker expression to indicate stimulation of maturation and effector function. To assess whether BP interactions in vivo produced different effects, we analyzed CD80, CD86, and MHC II expression of DCs recovered from the livers, spleens, bone marrows, and lymph nodes of mice injected once with BP (5 mg Fe). Interestingly, only DCs in spleens and bone marrow cells responded to BP exposure. DCs recovered from other organs showed no evidence of increased activation. These findings highlight complex interactions between living systems and nanoparticles, and their potential to mediate context-specific and selective activation of innate immune cells. Our study also emphasizes that results obtained from in vitro experiments must be interpreted with caution, as they may not faithfully represent responses in living systems.
Collapse
Affiliation(s)
- Mason Song
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University Baltimore 21218 USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University 1550 Orleans Street, Cancer Research Building - II, Rm 416 Baltimore MD 21231 USA
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University Baltimore MD 21231 USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University Baltimore 21218 USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University Baltimore 21218 USA
| | - Preethi Korangath
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University 1550 Orleans Street, Cancer Research Building - II, Rm 416 Baltimore MD 21231 USA
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University Baltimore MD 21231 USA
| |
Collapse
|
9
|
Acter S, Ngema LM, Moreau M, China D, Viswanathan A, Ding K, Choonara YE, Yasmin-Karim S, Ngwa W. PRIMERS: Polydopamine Radioimmunotherapy with Image-Guided Monitoring and Enhanced Release System. Pharmaceutics 2024; 16:1481. [PMID: 39598603 PMCID: PMC11597857 DOI: 10.3390/pharmaceutics16111481] [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: 10/25/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: To overcome the side effects of conventional cancer treatment, multifunctional nanoparticles with image-guidance properties are increasingly desired to obtain enhanced therapeutic efficacy without any toxicity of the treatment. Herein, we introduce the potential of Polydopamine Radioimmunotherapy with Image-guided Monitoring and Enhanced (drug) Release System (PRIMERS) to meet the challenges of currently used cancer therapy. Methods: The PDA nanobowls were synthesized using an emulsion-induced interfacial anisotropic assembly method followed by surface modification with high-Z material to obtained the final product PRIMERS. Results: The engineered multifunctional nanosystem "PRIMERS" could serve as fiducial markers with the potential for use in combination cancer therapy. By leveraging the advantages of the excellent surface functionalization capability of PDA, the anisotropic nanostructure (PDA nanobowls) has been successfully functionalized with gadolinium, which shows strong MRI contrast signal both in vitro in phantom and in vivo in animals. The results of anti-cancer drug loading and releasing efficiency of these functionalized nanobowls are presented. Moreover, the gadolinium-coated PDA nanobowls demonstrate the capacity for loading immunotherapy drugs (Anti-CD40) with activated release in acidic pH levels characteristic of the tumor microenvironment, with enhanced release following administration of radiation therapy in vitro. Conclusions: Overall, the results highlight the potential of this new technology for combining radiotherapy with activated image-guided drug delivery, which offers broad opportunities to overcome current challenges in cancer treatment.
Collapse
Affiliation(s)
- Shahinur Acter
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA; (L.M.N.); (M.M.); (A.V.); (K.D.)
| | - Lindokuhle M. Ngema
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA; (L.M.N.); (M.M.); (A.V.); (K.D.)
- WITS Advanced Drug Delivery Platform Research Unit, Department of Pharmacy & Pharmacology, University of Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa;
| | - Michele Moreau
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA; (L.M.N.); (M.M.); (A.V.); (K.D.)
| | - Debarghya China
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA;
| | - Akila Viswanathan
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA; (L.M.N.); (M.M.); (A.V.); (K.D.)
| | - Kai Ding
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA; (L.M.N.); (M.M.); (A.V.); (K.D.)
| | - Yahya E. Choonara
- WITS Advanced Drug Delivery Platform Research Unit, Department of Pharmacy & Pharmacology, University of Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa;
| | - Sayeda Yasmin-Karim
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Boston, MA 02115, USA;
| | - Wilfred Ngwa
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA; (L.M.N.); (M.M.); (A.V.); (K.D.)
| |
Collapse
|
10
|
Bal T, Anjrini N, Zeroual M. Recent Advances and Challenges in Targeted Drug Delivery Using Biofunctional Coatings. MEDICAL APPLICATIONS FOR BIOCOMPATIBLE SURFACES AND COATINGS 2024:41-75. [DOI: 10.1039/9781837675555-00041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Globally, clinics are overwhelmed by drugs targeting undesired cells and organs, causing adverse systemic effects on the body. This shortfall in targeting specificity, safety, and efficiency has noticeably contributed to the failure of the bench-to-bedside transition. Activation or impairment of immune activity due to a misdirected drug and its carrier fuels complications, extending the range of destruction which can convert the course of disease into a life-threatening route. To address these great challenges, advanced coatings as indispensable components of future medicine have been investigated over the last few decades for precisely targeted drug delivery to achieve favorable prognoses in the treatment of a broad spectrum of diseases. Complemented by advancements in the pharmacological parameters, these systems hold great promise for the field. This chapter aims to discuss recent progress on new coatings for targeted drug delivery and the parameters for manufacturing these platforms for their cargo based on major determinants such as biocompatibility and bioactivity. A brief overview of the various applications of targeted drug delivery with functional coatings is also provided to offer a new perspective on the field.
Collapse
Affiliation(s)
- Tugba Bal
- aDepartment of Bioengineering, Graduate School of Sciences, Uskudar University, 34662, Istanbul, Turkiye
- bDepartment of Bioengineering, Faculty of Engineering and Natural Sciences, Uskudar University, 34662, Istanbul, Turkiye
| | - Nasma Anjrini
- aDepartment of Bioengineering, Graduate School of Sciences, Uskudar University, 34662, Istanbul, Turkiye
| | - Meryem Zeroual
- aDepartment of Bioengineering, Graduate School of Sciences, Uskudar University, 34662, Istanbul, Turkiye
| |
Collapse
|
11
|
Qiao K, Pan Y, Zhang S, Shi G, Yang J, Zhang Z, Wang K, Chen X, Ning S. Cold Exposure Therapy Sensitizes Nanodrug-Mediated Radioimmunotherapy of Breast Cancer. ACS NANO 2024; 18:29689-29703. [PMID: 39401104 DOI: 10.1021/acsnano.4c09021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Cold exposure (CE) therapy can quickly induce tumor starvation by brown adipose tissue (BAT) thermogenesis. Exploring the combined antitumor mechanism of CE and traditional therapies (such as radiotherapy (RT)) is exciting and promising. In this study, we investigated the effect of CE in combination with nitric oxide (NO) gas therapy on sensitizing tumors to RT and promoting tumor radio-immunotherapy. We first constructed a liposome (SL) loaded with the NO prodrug S-nitroso-N-acetylpenicillamine (SNAP). When SL is injected, the glutathione (GSH) within the tumor region promotes the release of NO from SNAP. Subsequently, the superoxide anion produced by RT reacts with NO to generate peroxynitrite (ONOO-), which has strong oxidative properties and induces cell death. Meanwhile, the mice were exposed to a CE environment of 4 °C. CE-mediated BAT thermogenesis induced tumor starvation, which led to a decrease in ATP and GSH content within the tumor as well as an improvement in the hypoxic microenvironment and a decrease in myeloid-derived suppressor cells. All of the above have promoted the effectiveness of RT and activated the systemic antitumor immunity. In the bilateral tumor experiment, treatment of the primary tumor inhibited the growth of the distant tumor and promoted the infiltration of CD8+ T cells into the tumor. These findings reveal that the synergy of CE, NO gas therapy, and RT could confer high effective anticancer effects, providing possibilities in personalized cancer treatment.
Collapse
Affiliation(s)
- Kun Qiao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - You Pan
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Shiyuan Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Guangfu Shi
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Jinglin Yang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Zhenlin Zhang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Kaiyuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| |
Collapse
|
12
|
Tudor M, Popescu RC, Irimescu IN, Rzyanina A, Tarba N, Dinischiotu A, Craciun L, Esanu TR, Vasile E, Hotnog AT, Radu M, Mytsin G, Mihailescu M, Savu DI. Enhancing Proton Radiosensitivity of Chondrosarcoma Using Nanoparticle-Based Drug Delivery Approaches: A Comparative Study of High- and Low-Energy Protons. Int J Mol Sci 2024; 25:11481. [PMID: 39519034 PMCID: PMC11546389 DOI: 10.3390/ijms252111481] [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: 09/16/2024] [Revised: 10/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
To overcome chondrosarcoma's (CHS) high chemo- and radioresistance, we used polyethylene glycol-encapsulated iron oxide nanoparticles (IONPs) for the controlled delivery of the chemotherapeutic doxorubicin (IONPDOX) to amplify the cytotoxicity of proton radiation therapy. Human 2D CHS SW1353 cells were treated with protons (linear energy transfer (LET): 1.6 and 12.6 keV/µm) with and without IONPDOX. Cell survival was assayed using a clonogenic test, and genotoxicity was tested through the formation of micronuclei (MN) and γH2AX foci, respectively. Morphology together with spectral fingerprints of nuclei were measured using enhanced dark-field microscopy (EDFM) assembled with a hyperspectral imaging (HI) module and an axial scanning fluorescence module, as well as scanning electron microscopy (SEM) coupled with energy-dispersive X-Ray spectroscopy (EDX). Cell survival was also determined in 3D SW3153 spheroids following treatment with low-LET protons with/without the IONPDOX compound. IONPDOX increased radiosensitivity following proton irradiation at both LETs in correlation with DNA damage expressed as MN or γH2AX. The IONPDOX-low-LET proton combination caused a more lethal effect compared to IONPDOX-high-LET protons. CHS cell biological alterations were reflected by the modifications in the hyperspectral images and spectral profiles, emphasizing new possible spectroscopic markers of cancer therapy effects. Our findings show that the proposed treatment combination has the potential to improve the management of CHS.
Collapse
Affiliation(s)
- Mihaela Tudor
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, Reactorului 30, P.O. Box MG-6, 077125 Magurele, Romania; (M.T.); (R.C.P.); (M.R.)
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania;
| | - Roxana Cristina Popescu
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, Reactorului 30, P.O. Box MG-6, 077125 Magurele, Romania; (M.T.); (R.C.P.); (M.R.)
- Department of Bioengineering and Biotechnology, Faculty of Medical Engineering, National University for Science and Technology Politehnica of Bucharest, Gheorghe Polizu Street, 1-7, 011061 Bucharest, Romania
| | - Ionela N. Irimescu
- Applied Sciences Doctoral School, National University for Science and Technology Politehnica of Bucharest, 060042 Bucharest, Romania;
| | - Ann Rzyanina
- Laboratory of Nuclear Problems, Joint Institute for Nuclear Research, 6 Joliot-Curie Street, 141980 Dubna, Moscow Region, Russia; (A.R.); (G.M.)
| | - Nicolae Tarba
- Doctoral School of Computer Sciences, National University for Science and Technology Politehnica of Bucharest, 060042 Bucharest, Romania;
| | - Anca Dinischiotu
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania;
| | - Liviu Craciun
- Radiopharmaceutical Research Centre, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania; (L.C.); (T.R.E.)
| | - Tiberiu Relu Esanu
- Radiopharmaceutical Research Centre, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania; (L.C.); (T.R.E.)
| | - Eugeniu Vasile
- Faculty of Applied Physics, National University for Science and Technology Politehnica of Bucharest, 060042 Bucharest, Romania;
| | - Andrei Theodor Hotnog
- Applied Nuclear Physics Department, Horia Hulubei National Institute of Physics and Nuclear Engineering, Reactorului 30, P.O. Box MG-6, 077125 Magurele, Romania;
| | - Mihai Radu
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, Reactorului 30, P.O. Box MG-6, 077125 Magurele, Romania; (M.T.); (R.C.P.); (M.R.)
| | - Gennady Mytsin
- Laboratory of Nuclear Problems, Joint Institute for Nuclear Research, 6 Joliot-Curie Street, 141980 Dubna, Moscow Region, Russia; (A.R.); (G.M.)
| | - Mona Mihailescu
- Holographic Imaging and Processing Laboratory, Physics Department, National University for Science and Technology Politehnica of Bucharest, 060042 Bucharest, Romania;
- Centre for Research in Fundamental Sciences Applied in Engineering, National University for Science and Technology Politehnica of Bucharest, 060042 Bucharest, Romania
| | - Diana Iulia Savu
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, Reactorului 30, P.O. Box MG-6, 077125 Magurele, Romania; (M.T.); (R.C.P.); (M.R.)
| |
Collapse
|
13
|
Heo D, Lokeshwar BL, Barrett JT, Mostafaei F, Kwon SH, Huh C. Quantitative analysis of radiosensitizing effect for magnetic hyperthermia-radiation combined therapy on prostate cancer cells. Med Phys 2024; 51:7606-7618. [PMID: 38923579 DOI: 10.1002/mp.17248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/17/2024] [Accepted: 05/19/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Magnetic hyperthermia (MHT) has emerged as a promising therapeutic approach in the field of radiation oncology due to its superior precision in controlling temperature and managing the heating area compared to conventional hyperthermia. Recent studies have proposed solutions to address clinical safety concerns associated with MHT, which arise from the use of highly concentrated magnetic nanoparticles and the strong magnetic field needed to induce hyperthermic effects. Despite these efforts, challenges remain in quantifying therapeutic outcomes and developing treatment plan systems for combining MHT with radiation therapy (RT). PURPOSE This study aims to quantitatively measure the therapeutic effect, including radiation dose enhancement (RDE) in the magnetic hyperthermia-radiation combined therapy (MHRT), using the equivalent radiation dose (EQD) estimation method. METHODS To conduct EQD estimation for MHRT, we compared the therapeutic effects between the conventional hyperthermia-radiation combined therapy (HTRT) and MHRT in human prostate cancer cell lines, PC3 and LNCaP. We adopted a clonogenic assay to validate RDE and the radiosensitizing effect induced by MHT. The data on survival fractions were analyzed using both the linear-quadradic model and Arrhenius model to estimate the biological parameters describing RDE and radiosensitizing effect of MHRT for both cell lines through maximum likelihood estimation. Based on these parameters, a new survival fraction model was suggested for EQD estimation of MHRT. RESULTS The newly designed model describing the MHRT effect, effectively captures the variations in thermal and radiation dose for both cell lines (R2 > 0.95), and its suitability was confirmed through the normality test of residuals. This model appropriately describes the survival fractions up to 10 Gy for PC3 cells and 8 Gy for LNCaP cells under RT-only conditions. Furthermore, using the newly defined parameter r, the RDE effect was calculated as 29% in PC3 cells and 23% in LNCaP cells. EQDMHRT calculated through this model was 9.47 Gy for PC3 and 4.71 Gy for LNCaP when given 2 Gy and MHT for 30 min. Compared to EQDHTRT, EQDMHRT showed a 26% increase for PC3 and a 20% increase for LNCaP. CONCLUSIONS The proposed model effectively describes the changes of the survival fraction induced by MHRT in both cell lines and adequately represents actual data values through residual analysis. Newly suggested parameter r for RDE effect shows potential for quantitative comparisons between HTRT and MHRT, and optimizing therapeutic outcomes in MHRT for prostate cancer.
Collapse
Affiliation(s)
- Dan Heo
- Georgia Cancer Center, Department of Radiation Oncology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Bal L Lokeshwar
- Georgia Cancer Center, Department of Medicine, Augusta University, Augusta, Georgia, USA
| | - John T Barrett
- Georgia Cancer Center, Department of Radiation Oncology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Farshad Mostafaei
- Georgia Cancer Center, Department of Radiation Oncology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Sang-Ho Kwon
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Chulhaeng Huh
- Georgia Cancer Center, Department of Radiation Oncology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
14
|
Liu M, Li T, Zhao M, Qian C, Wang R, Liu L, Xiao Y, Xiao H, Tang X, Liu H. Nanoradiosensitizers in glioblastoma treatment: recent advances and future perspectives. Nanomedicine (Lond) 2024; 19:2229-2249. [PMID: 39311492 PMCID: PMC11487349 DOI: 10.1080/17435889.2024.2395238] [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: 06/12/2024] [Accepted: 08/19/2024] [Indexed: 10/16/2024] Open
Abstract
Glioblastoma (GBM), a highly invasive type of brain tumor located within the central nervous system, manifests a median survival time of merely 14.6 months. Radiotherapy kills tumor cells through focused high-energy radiation and has become a crucial treatment strategy for GBM, especially in cases where surgical resection is not viable. However, the presence of radioresistant tumor cells limits its clinical effectiveness. Radioresistance is a key factor of treatment failure, prompting the development of various therapeutic strategies to overcome this challenge. With the rapid development of nanomedicine, nanoradiosensitizers provide a novel approach to enhancing the effectiveness of radiotherapy. In this review, we discuss the reasons behind GBM radio-resistance and the mechanisms of radiotherapy sensitization. Then we summarize the primary types of nanoradiosensitizers and recent progress in their application for the radiosensitization of GBM. Finally, we elucidate the factors influencing their practical implementation, along with the challenges and promising prospects associated with multifunctional nanoradiosensitizers.
Collapse
Affiliation(s)
- Mingxi Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Taiping Li
- Department of Neuro-Psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Mengjie Zhao
- Department of Neuro-Psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Chunfa Qian
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Ran Wang
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Yong Xiao
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Hong Xiao
- Department of Neuro-Psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Xianglong Tang
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
- Department of Neuro-Psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Hongyi Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Patel KD, Keskin-Erdogan Z, Sawadkar P, Nik Sharifulden NSA, Shannon MR, Patel M, Silva LB, Patel R, Chau DYS, Knowles JC, Perriman AW, Kim HW. Oxidative stress modulating nanomaterials and their biochemical roles in nanomedicine. NANOSCALE HORIZONS 2024; 9:1630-1682. [PMID: 39018043 DOI: 10.1039/d4nh00171k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Many pathological conditions are predominantly associated with oxidative stress, arising from reactive oxygen species (ROS); therefore, the modulation of redox activities has been a key strategy to restore normal tissue functions. Current approaches involve establishing a favorable cellular redox environment through the administration of therapeutic drugs and redox-active nanomaterials (RANs). In particular, RANs not only provide a stable and reliable means of therapeutic delivery but also possess the capacity to finely tune various interconnected components, including radicals, enzymes, proteins, transcription factors, and metabolites. Here, we discuss the roles that engineered RANs play in a spectrum of pathological conditions, such as cancer, neurodegenerative diseases, infections, and inflammation. We visualize the dual functions of RANs as both generator and scavenger of ROS, emphasizing their profound impact on diverse cellular functions. The focus of this review is solely on inorganic redox-active nanomaterials (inorganic RANs). Additionally, we deliberate on the challenges associated with current RANs-based approaches and propose potential research directions for their future clinical translation.
Collapse
Affiliation(s)
- Kapil D Patel
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
| | - Zalike Keskin-Erdogan
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
- Department of Chemical Engineering, Imperial College London, Exhibition Rd, South Kensington, SW7 2BX, London, UK
| | - Prasad Sawadkar
- Division of Surgery and Interventional Science, UCL, London, UK
- The Griffin Institute, Northwick Park Institute for Medical Research, Northwick Park and St Mark's Hospitals, London, HA1 3UJ, UK
| | - Nik Syahirah Aliaa Nik Sharifulden
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Mark Robert Shannon
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Women University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Lady Barrios Silva
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Rajkumar Patel
- Energy & Environment Sciences and Engineering (EESE), Integrated Sciences and Engineering Division (ISED), Underwood International College, Yonsei University, 85 Songdongwahak-ro, Yeonsungu, Incheon 21938, Republic of Korea
| | - David Y S Chau
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Jonathan C Knowles
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Adam W Perriman
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
16
|
Grancharova T, Zagorchev P, Pilicheva B. Iron Oxide Nanoparticles: Parameters for Optimized Photoconversion Efficiency in Synergistic Cancer Treatment. J Funct Biomater 2024; 15:207. [PMID: 39194645 DOI: 10.3390/jfb15080207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Photothermal therapy (PTT) can overcome cancer treatment resistance by enhancing the cell membrane permeability, facilitating drug accumulation, and promoting drug release within the tumor tissue. Iron oxide nanoparticles (IONPs) have emerged as effective agents for PTT due to their unique properties and biocompatibility. Approved for the treatment of anemia, as MRI contrast agents, and as magnetic hyperthermia mediators, IONPs also offer excellent light-to-heat conversion and can be manipulated using external magnetic fields for targeted accumulation in specific tissue. Optimizing parameters such as the laser wavelength, power density, shape, size, iron oxidation state, functionalization, and concentration is crucial for IONPs' effectiveness. In addition to PTT, IONPs enhance other cancer treatment modalities. They improve tumor oxygenation, enhancing the efficacy of radiotherapy and photodynamic therapy. IONPs can also trigger ferroptosis, a programmed cell death pathway mediated by iron-dependent lipid peroxidation. Their magneto-mechanical effect allows them to exert a mechanical force on cancer cells to destroy tumors, minimizing the damage to healthy tissue. This review outlines strategies for the management of the photothermal performance and PTT efficiency with iron oxide nanoparticles, as well as synergies with other cancer therapies.
Collapse
Affiliation(s)
- Tsenka Grancharova
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Plamen Zagorchev
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Bissera Pilicheva
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| |
Collapse
|
17
|
Mahdavi Niyaki Z, Salehzadeh A, Peymani M, Zaefizadeh M. Exploring the Therapeutic Potential of Fe 3O 4@Glu-Oleuropein Nanoparticles in Targeting KRAS Pathway-Regulating lncRNAs in Colorectal Cancer Cells. Biol Trace Elem Res 2024; 202:3073-3085. [PMID: 37792268 DOI: 10.1007/s12011-023-03892-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/25/2023] [Indexed: 10/05/2023]
Abstract
Cancer, the leading cause of death worldwide, has witnessed significant advancements in treatment through targeted therapies. Among the proto-oncogenes prevalent in human cancers, KRAS stands out, and recent research has focused on long noncoding RNAs (lncRNAs) as regulators of miRNAs targeting the KRAS oncogene. This study specifically explores lncRNAs involved in the KRAS pathway in colorectal cancer (CRC). To investigate this, researchers employed iron oxide nanoparticles coated with glucose and conjugated with Oleuropein (Fe3O4@Glu-Oleuropein NPs) to evaluate their impact on candidate lncRNAs associated with KRAS pathway deregulation. The study utilized TCGA data to identify genes affected by KRAS mutation and lncRNAs linked to KRAS in CRC. Enrichr and MsigDB databases helped identify relevant pathways. Genes with a correlation coefficient above 0.5 and a P-value less than 0.01 with candidate lncRNAs were selected. MTT and flow cytometry assays determined the anti-proliferative and apoptotic effects of Fe3O4@Glu-Oleuropein NPs on CRC cells (SW480) and normal cells (HEK293). The findings showed that increased expression of FEZF1-AS1, GAS6-AS1, and LINC00920 correlated with mutated KRAS, and co-expressed genes were significantly involved in hypoxia, KRAS signaling, DNA repair, and IL-2/STAT5 signaling pathways. Fe3O4@Glu-Oleuropein NPs exhibited higher toxicity toward cancer cells, with IC50 values of 92 μg/ml for SW480 and 281 μg/ml for HEK293. Flow cytometry analysis revealed a substantial increase in necrotic and apoptotic cells when treated with Fe3O4@Glu-Oleuropein, along with down-regulation of GAS6-AS1, LINC00920, and FEZF1-AS1 lncRNAs in treated cells. In conclusion, this study highlights the therapeutic potential of Fe3O4@Glu-Oleuropein on colon cancer cells in vitro. The identification of lncRNAs involved in the KRAS pathway provides insights into the underlying mechanisms and offers avenues for further research in targeted cancer therapies.
Collapse
Affiliation(s)
| | - Ali Salehzadeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran.
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Mohammad Zaefizadeh
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| |
Collapse
|
18
|
Tang C, Wang Y, Wu M, Wang Z, Zhou Y, Lin Y, Wang Y, Xu H. Cancer cell membrane-camouflaged biomimetic nanoparticles for enhancing chemo-radiation therapy efficacy in glioma. J Biomed Res 2024; 39:87-102. [PMID: 38812286 PMCID: PMC11873587 DOI: 10.7555/jbr.38.20240100] [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: 04/08/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and lethal brain tumor with limited treatment options. To improve therapeutic efficacy, we developed a novel multifunctional nanoplatform, GM@P(T/S), comprised of polymeric nanoparticles coated with GBM cell membranes as well as co-loaded with temozolomide (TMZ) and superparamagnetic iron oxide (SPIO) nanoparticles. The successful preparation was confirmed in terms of particle size, morphology, stability, the in vitro drug release, and cellular uptake assays. We demonstrated that GM@P(T/S) exhibited the enhanced homotypic targeting, the prolonged blood circulation, and efficient blood-brain barrier penetration in both in vitro and in vivo studies. The combination of TMZ and SPIO nanoparticles within GM@P(T/S) synergistically improved chemo-radiation therapy, leading to a reduced tumor growth, an increased survival, and minimal systemic toxicity in the orthotopic GBM mouse models. Our findings suggest that GM@P(T/S) holds a great promise as a targeted and efficient therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Chunming Tang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yanling Wang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Min Wu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Zhiji Wang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yupeng Zhou
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ya Lin
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yijun Wang
- Department of Pharmacy, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210003, China
| | - Huae Xu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
19
|
Wang Y, Wang L, Li T, Ouyang M, Xiong H, Zhou D. Bimetallic nanoparticles as cascade sensitizing amplifiers for low-dose and robust cancer radio-immunotherapy. Acta Pharm Sin B 2024; 14:1787-1800. [PMID: 38572091 PMCID: PMC10985033 DOI: 10.1016/j.apsb.2023.11.028] [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: 08/31/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 04/05/2024] Open
Abstract
Radiotherapy (RT) is one of the most feasible and routinely used therapeutic modalities for treating malignant tumors. In particular, immune responses triggered by RT, known as radio-immunotherapy, can partially inhibit the growth of distantly spreading tumors and recurrent tumors. However, the safety and efficacy of radio-immunotherapy is impeded by the radio-resistance and poor immunogenicity of tumor. Herein, we report oxaliplatin (IV)-iron bimetallic nanoparticles (OXA/Fe NPs) as cascade sensitizing amplifiers for low-dose and robust radio-immunotherapy. The OXA/Fe NPs exhibit tumor-specific accumulation and activation of OXA (II) and Fe2+ in response to the reductive and acidic microenvironment within tumor cells. The cascade reactions of the released metallic drugs can sensitize RT by inducing DNA damage, increasing ROS and O2 levels, and amplifying the immunogenic cell death (ICD) effect after RT to facilitate potent immune activation. As a result, OXA/Fe NPs-based low-dose RT triggered a robust immune response and inhibited the distant and metastatic tumors effectively by a strong abscopal effect. Moreover, a long-term immunological memory effect to protect mice from tumor rechallenging is observed. Overall, the bimetallic NPs-based cascade sensitizing amplifier system offers an efficient radio-immunotherapy regimen that addresses the key challenges.
Collapse
Affiliation(s)
- Yupeng Wang
- Department of Ultrasonic Diagnosis, Zhujiang Hospital, Key Laboratory of Mental Health of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lina Wang
- Testing and Analysis Center, Hebei Normal University, Shijiazhuang 050024, China
| | - Tao Li
- Department of Ultrasonic Diagnosis, Zhujiang Hospital, Key Laboratory of Mental Health of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Min Ouyang
- Department of Ultrasonic Diagnosis, Zhujiang Hospital, Key Laboratory of Mental Health of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hejian Xiong
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Dongfang Zhou
- Department of Ultrasonic Diagnosis, Zhujiang Hospital, Key Laboratory of Mental Health of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
20
|
Ko MJ, Yoo W, Min S, Zhang YS, Joo J, Kang H, Kim DH. Photonic control of image-guided ferroptosis cancer nanomedicine. Coord Chem Rev 2024; 500:215532. [PMID: 38645709 PMCID: PMC11027759 DOI: 10.1016/j.ccr.2023.215532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Photonic nanomaterials, characterized by their remarkable photonic tunability, empower a diverse range of applications, including cutting-edge advances in cancer nanomedicine. Recently, ferroptosis has emerged as a promising alternative strategy for effectively killing cancer cells with minimizing therapeutic resistance. Novel design of photonic nanomaterials that can integrate photoresponsive-ferroptosis inducers, -diagnostic imaging, and -synergistic components provide significant benefits to effectively trigger local ferroptosis. This review provides a comprehensive overview of recent advancements in photonic nanomaterials for image-guided ferroptosis cancer nanomedicine, offering insights into their strengths, constraints, and their potential as a future paradigm in cancer treatment.
Collapse
Affiliation(s)
- Min Jun Ko
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Woojung Yoo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital Harvard Medical School, Cambridge, MA 02139, USA
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
- College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, USA
| |
Collapse
|
21
|
Jenkins SV, Jung S, Jamshidi-Parsian A, Borrelli MJ, Dings RPM, Griffin RJ. Morphological Effects and In Vitro Biological Mechanisms of Radiation-Induced Cell Killing by Gold Nanomaterials. ACS APPLIED MATERIALS & INTERFACES 2023; 15:58241-58250. [PMID: 38059477 DOI: 10.1021/acsami.3c15358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Gold nanomaterials have been shown to augment radiation therapy both in vitro and in vivo. However, studies on these materials are mostly phenomenological due to nanoparticle heterogeneity and the complexity of biological systems. Even accurate quantification of the particle dose still results in bulk average biases; the effect on individual cells is not measured but rather the effect on the overall population. To perform quantitative nanobiology, we coated glass coverslips uniformly at varying densities with Au nanoparticle preparations with different morphologies (45 nm cages, 25 nm spheres, and 30 nm rods). Consequently, the effect of a specific number of particles per unit area in contact with breast cancer cells growing on the coated surfaces was ascertained. Gold nanocages showed the highest degree of radiosensitization on a per particle basis, followed by gold nanospheres and gold nanorods, respectively. All three materials showed little cytotoxic effect at 0 Gy, but clonogenic survival decreased proportionally with the radiation dose and particle coverage density. A similar trend was seen in vivo in the combined treatment antitumor response in 4T1 tumor-bearing animals. The presence of gold affected the type and quantity of reactive oxygen species generated, specifically superoxide and hydroxyl radicals, and the concentration of nanocages correlated with the development of more numerous double-stranded DNA breaks and increased protein oxidation as measured by carbonylation. This work demonstrates the dependence on morphology and concentration of radiation enhancement by gold nanomaterials and may lead to a novel method to differentiate intra- and extracellular functionalities of gold nanomedicine treatment strategies. It further provides insights that can guide the rational development of gold nanomaterial-based radiosensitizers for clinical use.
Collapse
Affiliation(s)
- Samir V Jenkins
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Seunghyun Jung
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Azemat Jamshidi-Parsian
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Michael J Borrelli
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| |
Collapse
|
22
|
Afifi MM, El-Gebaly RH, Abdelrahman IY, Rageh MM. Efficacy of iron-silver bimetallic nanoparticles to enhance radiotherapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3647-3657. [PMID: 37289284 PMCID: PMC10643307 DOI: 10.1007/s00210-023-02556-9] [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: 01/02/2023] [Accepted: 05/26/2023] [Indexed: 06/09/2023]
Abstract
Radiotherapy (RT) is one of the primary cancer treatment methods. Radiosensitizers are used to enhance RT and protect healthy tissue. Heavy metals have been studied as radiosensitizers. Thus, iron oxide and iron oxide/silver nanoparticles have been the main subjects of this investigation. A simple honey-based synthesis of iron (IONPs) and iron-silver bimetallic nanoparticles (IO@AgNPs) were prepared followed by characterization with transmission electron microscope (TEM), absorption spectra, vibrating sample magnetometer (VSM), and X-ray diffraction (XRD). Additionally, Ehrlich carcinoma was induced in 30 adult BALB/c mice and divided into 6 groups. Mice of group G1 were not treated with nanoparticles or exposed to irradiation (control group), and group G2 and G3 were treated with IONPs and IO@AgNPs respectively. Mice of group G4 were exposed to a high dose of gamma radiation (HRD) (12 Gy). Groups G5 and G6 were treated with IONPs and IO@AgNPs followed by exposure to a low dose of gamma radiation (LRD) (6 Gy) respectively. The impact of NP on the treatment protocol was evaluated by checking tumor growth, DNA damage, and level of oxidative stress in addition to investigating tumor histopathology. Additional research on the toxicity of this protocol was also evaluated by looking at the liver's cytotoxicity. When compared to HRD therapy, combination therapy (bimetallic NPs and LRD) significantly increased DNA damage by about 75% while having a stronger efficacy in slowing Ehrlich tumor growth (at the end of treatment protocol) by about 45%. Regarding the biosafety concern, mice treated with combination therapy showed lower alanine aminotransferase (ALT) levels in their liver tissues by about half the value of HRD. IO@AgNPs enhanced the therapeutic effect of low-dose radiation and increased the efficacy of treating Ehrlich tumors with the least amount of harm to normal tissues as compared to high radiation dosage therapy.
Collapse
Affiliation(s)
- Marwa M Afifi
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt.
| | - Reem H El-Gebaly
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Ibrahim Y Abdelrahman
- Egyptian Atomic Energy Authority, National Center for Radiation Research and Technology, Cairo, Egypt
| | - Monira M Rageh
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| |
Collapse
|
23
|
Shirvalilou S, Khoee S, Khoei S, Karimi MR, Sadri E, Shirvaliloo M. Targeted magnetochemotherapy modified by 5-Fu-loaded thermally on/off switching nanoheaters for the eradication of CT26 murine colon cancer by inducing apoptotic and autophagic cell death. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00164-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
AbstractDespite significant breakthroughs in diagnosis and treatment of colorectal cancer (CRC), the extent of morbidity and mortality secondary to CRC is still concerning. In this study, we evaluated the efficacy of our new tumor-selective nanoplatforms at induction of apoptosis and autophagy, which was tested using active 5-fluorouracil (5-Fu)-based targeting of tumor cells in a BALB/c murine model of CRC combined with magnetic thermal therapy. Nanoparticles were synthesized and characterized by zeta sizer, transmission electron microscopy (TEM), and scanning electron microscopy (SEM). The cytotoxicity and tissue uptake of 5-Fu-loaded folic acid (Fa)-modified magnetic nanoparticles (5-Fu/MNPs-Fa) was assessed using MTT, ICP-OES, and HPLC. The rate of apoptosis and autophagy, as two major indicators of antitumor activity, was measured based on protein expression of Bax, Bcl2, Caspase 3, mTOR, P-mTOR, Beclin-1, and LC3B in CT-26 murine CRC, along with tumor volume and survival time. The spherical 5-Fu/MNPs-Fa exhibited sustained thermal on/off switching drug release and higher therapeutic index compared to free 5-Fu. Our de novo synthetized magnetic nanoheaters successfully delivered the therapeutic agent to the tumor site, enhanced the conversion of radio frequency energy to heat in tumor cells, exhibited higher antitumor efficiency based on Bax/Bcl2 ratio and overexpression of Beclin-1 and LC3B, increased the survival time, and decreased the tumor volume (P < 0.05). Our findings indicated that magnetochemotherapy (MHC) was substantially more effective than hyperthermia and/or chemotherapy alone. From a translational standpoint, the 5-Fu/MNPs-Fa would be a promising candidate sustained drug targeting system that could improve cancer cell therapy via inducing apoptosis and autophagy.
Graphical Abstract
Collapse
|
24
|
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
|
25
|
Zhao B, Ma Z, Ding S, Cao Y, Du J, Zeng L, Hu Y, Zhou J, Zhang X, Bian X, Tian G. Catalytic MnWO 4 Nanorods for Chemodynamic Therapy Synergized Radiotherapy of Triple Negative Breast Cancer. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202306328] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Indexed: 01/12/2025]
Abstract
AbstractNanomedicine‐based synergy of chemodynamic therapy (CDT) and radiotherapy (RT) modulated by tumor microenvironment enables rapid tumor ablation, which holds great hope for the refractory and recurrent cancers, such as triple negative breast cancer (TNBC). The clinical translation of hafnium oxide (HfO2), commercially named as NBTXR3, has aroused new research focus on single‐component inorganic nanomedicines as clinical candidates. Herein, the single‐component MnWO4 is first reported as a new kind of Fenton‐like agent yet radiosensitizer for TNBC treatment undergoing the synergistic CDT/RT mechanism. MnWO4 nanorods are synthesized via a simple one‐pot hydrothermal method and then undergo a layer‐by‐layer PEGylation to obtain bioavailable MnWO4‐PEG (MWP). MWP‐based Fenton‐like reaction efficacy depends on reaction time, temperatures, pH values, and MWP concentrations. Mn‐triggered chemodynamic effect delays RT‐induced DNA damage repair and sorts cell cycles distribution toward radiosensitive phases, while W‐mediated radiosensitization improves the tumoral H2O2 overexpression to enhance CDT, remarkably amplifying of the intracellular oxidative stress to boost 4T1 cell apoptosis. In vitro and in vivo evaluations further demonstrate the effectiveness and biosafety of MWP‐based synergistic therapy. Considering the potential magnetic resonance and computed tomography imaging capabilities, MWP can be expected as an intelligent cancer theranostics for imaging‐guided cancer therapy in clinic in the future.
Collapse
Affiliation(s)
- Bin Zhao
- College of Basic Medicine and College of Pharmacy Shanxi Medical University Jinzhong Shanxi 030619 P. R. China
- Institute of Pathology and Southwest Cancer Center The First Affiliated Hospital Third Military Medical University (Army Medical University) Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 400038 P. R. China
- Chongqing Institute of Advanced Pathology Jinfeng Laboratory Chongqing 401329 P. R. China
| | - Zhili Ma
- College of Basic Medicine and College of Pharmacy Shanxi Medical University Jinzhong Shanxi 030619 P. R. China
- Institute of Pathology and Southwest Cancer Center The First Affiliated Hospital Third Military Medical University (Army Medical University) Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 400038 P. R. China
- Chongqing Institute of Advanced Pathology Jinfeng Laboratory Chongqing 401329 P. R. China
| | - Shuaishuai Ding
- Institute of Pathology and Southwest Cancer Center The First Affiliated Hospital Third Military Medical University (Army Medical University) Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 400038 P. R. China
- Chongqing Institute of Advanced Pathology Jinfeng Laboratory Chongqing 401329 P. R. China
| | - Yuhua Cao
- Institute of Pathology and Southwest Cancer Center The First Affiliated Hospital Third Military Medical University (Army Medical University) Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 400038 P. R. China
| | - Jiangfeng Du
- College of Basic Medicine and College of Pharmacy Shanxi Medical University Jinzhong Shanxi 030619 P. R. China
| | - Lijuan Zeng
- Institute of Pathology and Southwest Cancer Center The First Affiliated Hospital Third Military Medical University (Army Medical University) Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 400038 P. R. China
| | - Yunping Hu
- Institute of Pathology and Southwest Cancer Center The First Affiliated Hospital Third Military Medical University (Army Medical University) Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 400038 P. R. China
| | - Jingrong Zhou
- Institute of Pathology and Southwest Cancer Center The First Affiliated Hospital Third Military Medical University (Army Medical University) Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 400038 P. R. China
| | - Xiao Zhang
- Institute of Pathology and Southwest Cancer Center The First Affiliated Hospital Third Military Medical University (Army Medical University) Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 400038 P. R. China
- Chongqing Institute of Advanced Pathology Jinfeng Laboratory Chongqing 401329 P. R. China
| | - Xiu‐wu Bian
- Institute of Pathology and Southwest Cancer Center The First Affiliated Hospital Third Military Medical University (Army Medical University) Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 400038 P. R. China
- Chongqing Institute of Advanced Pathology Jinfeng Laboratory Chongqing 401329 P. R. China
| | - Gan Tian
- Institute of Pathology and Southwest Cancer Center The First Affiliated Hospital Third Military Medical University (Army Medical University) Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 400038 P. R. China
- Chongqing Institute of Advanced Pathology Jinfeng Laboratory Chongqing 401329 P. R. China
| |
Collapse
|
26
|
Tran NH, Ryzhov V, Volnitskiy A, Amerkanov D, Pack F, Golubev AM, Arutyunyan A, Spitsyna A, Burdakov V, Lebedev D, Konevega AL, Shtam T, Marchenko Y. Radiosensitizing Effect of Dextran-Coated Iron Oxide Nanoparticles on Malignant Glioma Cells. Int J Mol Sci 2023; 24:15150. [PMID: 37894830 PMCID: PMC10606998 DOI: 10.3390/ijms242015150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/21/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
The potential of standard methods of radiation therapy is limited by the dose that can be safely delivered to the tumor, which could be too low for radical treatment. The dose efficiency can be increased by using radiosensitizers. In this study, we evaluated the sensitizing potential of biocompatible iron oxide nanoparticles coated with a dextran shell in A172 and Gl-Tr glioblastoma cells in vitro. The cells preincubated with nanoparticles for 24 h were exposed to ionizing radiation (X-ray, gamma, or proton) at doses of 0.5-6 Gy, and their viability was assessed by the Resazurin assay and by staining of the surviving cells with crystal violet. A statistically significant effect of radiosensitization by nanoparticles was observed in both cell lines when cells were exposed to 35 keV X-rays. A weak radiosensitizing effect was found only in the Gl-Tr line for the 1.2 MeV gamma irradiation and there was no radiosensitizing effect in both lines for the 200 MeV proton irradiation at the Bragg peak. A slight (ca. 10%) increase in the formation of additional reactive oxygen species after X-ray irradiation was found when nanoparticles were present. These results suggest that the nanoparticles absorbed by glioma cells can produce a significant radiosensitizing effect, probably due to the action of secondary electrons generated by the magnetite core, whereas the dextran shell of the nanoparticles used in these experiments appears to be rather stable under radiation exposure.
Collapse
Affiliation(s)
- Nhan Hau Tran
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Politehnicheskaya 29, St. Petersburg 195251, Russia
| | - Vyacheslav Ryzhov
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
| | - Andrey Volnitskiy
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
| | - Dmitry Amerkanov
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
- National Research Center “Kurchatov Institute”, Akademika Kurchatova pl. 1, Moscow 123182, Russia
| | - Fedor Pack
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
- National Research Center “Kurchatov Institute”, Akademika Kurchatova pl. 1, Moscow 123182, Russia
| | - Aleksander M. Golubev
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
| | - Alexandr Arutyunyan
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
| | - Anastasiia Spitsyna
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
| | - Vladimir Burdakov
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
| | - Dmitry Lebedev
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
- National Research Center “Kurchatov Institute”, Akademika Kurchatova pl. 1, Moscow 123182, Russia
| | - Andrey L. Konevega
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Politehnicheskaya 29, St. Petersburg 195251, Russia
- National Research Center “Kurchatov Institute”, Akademika Kurchatova pl. 1, Moscow 123182, Russia
| | - Tatiana Shtam
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
- National Research Center “Kurchatov Institute”, Akademika Kurchatova pl. 1, Moscow 123182, Russia
| | - Yaroslav Marchenko
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Orlova roscha 1, Gatchina 188300, Russia; (N.H.T.); (A.V.); (D.A.); (F.P.); (A.M.G.); (A.A.); (A.S.); (V.B.); (D.L.); (A.L.K.); (T.S.)
| |
Collapse
|
27
|
Li J, Lin C, Zhu Y, Shao C, Wang T, Chen B. Colorectal cancer cell membrane biomimetic ferroferric oxide nanomaterials for homologous bio-imaging and chemotherapy application. Med Oncol 2023; 40:322. [PMID: 37801170 DOI: 10.1007/s12032-023-02175-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023]
Abstract
The research of nanomaterials for bio-imaging and theranostic are very active nowadays with unprecedented advantages in nanomedicine. Homologous targeting and bio-imaging greatly improve the ability of targeted drug delivery and enhance active targeting and treatment ability of nanomedicine for the tumor. In this work, lycorine hydrochloride (LH) and magnetic iron oxide nanoparticles coated with a colorectal cancer (CRC) cell membrane (LH-Fe3O4@M) were prepared, for homologous targeting, magnetic resonance imaging (MRI), and chemotherapy. Results showed that the LH-Fe3O4@M and Fe3O4@M intensity at HT29 tumor was significantly higher than that Fe3O4@PEG, proving the superior selectivity of cancer cell membrane-camouflaged nanomedicine for homologous tumors and the MRI effect of darkening contrast enhancement were remarkable at HT29 tumor. The LH-Fe3O4@M exhibited excellent chemotherapy effect in CRC models as well as LH alone and achieved a high tumor ablation rate but no damage to normal tissues and cells. Therefore, our biomimetic system achieved a homologous targeting, bio-imaging, and efficient therapeutic effect of CRC.
Collapse
Affiliation(s)
- Jun Li
- The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Chenyu Lin
- The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yuqian Zhu
- The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
| | - Tiegong Wang
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China.
| | - Bingdi Chen
- The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
28
|
Shestovskaya MV, Luss AL, Bezborodova OA, Makarov VV, Keskinov AA. Iron Oxide Nanoparticles in Cancer Treatment: Cell Responses and the Potency to Improve Radiosensitivity. Pharmaceutics 2023; 15:2406. [PMID: 37896166 PMCID: PMC10610190 DOI: 10.3390/pharmaceutics15102406] [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: 08/11/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
The main concept of radiosensitization is making the tumor tissue more responsive to ionizing radiation, which leads to an increase in the potency of radiation therapy and allows for decreasing radiation dose and the concomitant side effects. Radiosensitization by metal oxide nanoparticles is widely discussed, but the range of mechanisms studied is not sufficiently codified and often does not reflect the ability of nanocarriers to have a specific impact on cells. This review is focused on the magnetic iron oxide nanoparticles while they occupied a special niche among the prospective radiosensitizers due to unique physicochemical characteristics and reactivity. We collected data about the possible molecular mechanisms underlying the radiosensitizing effects of iron oxide nanoparticles (IONPs) and the main approaches to increase their therapeutic efficacy by variable modifications.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| | - Anna L. Luss
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
- The Department of Technology of Chemical, Pharmaceutical and Cosmetic Products Mendeleev of University of Chemical Technology of Russia, Miusskaya sq. 9, Moscow 125047, Russia
| | - Olga A. Bezborodova
- P. Hertsen Moscow Oncology Research Institute of the National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinskiy p. 3, Moscow 125284, Russia;
| | - Valentin V. Makarov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| | - Anton A. Keskinov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| |
Collapse
|
29
|
Li Z, Bai R, Yi J, Zhou H, Xian J, Chen C. Designing Smart Iron Oxide Nanoparticles for MR Imaging of Tumors. CHEMICAL & BIOMEDICAL IMAGING 2023; 1:315-339. [PMID: 37501794 PMCID: PMC10369497 DOI: 10.1021/cbmi.3c00026] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 07/29/2023]
Abstract
Iron oxide nanoparticles (IONPs) possess unique magnetism and good biocompatibility, and they have been widely applied as contrast agents (CAs) for magnetic resonance imaging (MRI). Traditional CAs typically show a fixed enhanced signal, thus exhibiting the limitations of low sensitivity and a lack of specificity. Nowadays, the progress of stimulus-responsive IONPs allows alteration of the relaxation signal in response to internal stimuli of the tumor, or external stimuli, thus providing an opportunity to overcome those limitations. This review summarizes the current status of smart IONPs as tumor imaging MRI CAs that exhibit responsiveness to endogenous stimuli, such as pH, hypoxia, glutathione, and enzymes, or exogenous stimuli, such as magnets, light, and so on. We discuss the challenges and future opportunities for IONPs as MRI CAs and comprehensively illustrate the applications of these stimuli-responsive IONPs. This review will help provide guidance for designing IONPs as MRI CAs and further promote the reasonable design of magnetic nanoparticles and achieve early and accurate tumor detection.
Collapse
Affiliation(s)
- Zhenzhen Li
- CAS
Key Laboratory for Biomedical Effects of Nanoparticles and Nanosafety
& CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Department
of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Ru Bai
- CAS
Key Laboratory for Biomedical Effects of Nanoparticles and Nanosafety
& CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- Research
Unit of Nanoscience and Technology, Chinese
Academy of Medical Sciences, Beijing 100021, China
| | - Jia Yi
- Guangdong
Provincial Development and Reform Commission, Guangzhou 510031, China
| | - Huige Zhou
- CAS
Key Laboratory for Biomedical Effects of Nanoparticles and Nanosafety
& CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- Research
Unit of Nanoscience and Technology, Chinese
Academy of Medical Sciences, Beijing 100021, China
| | - Junfang Xian
- Department
of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Chunying Chen
- CAS
Key Laboratory for Biomedical Effects of Nanoparticles and Nanosafety
& CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- Research
Unit of Nanoscience and Technology, Chinese
Academy of Medical Sciences, Beijing 100021, China
- The
GBA National Institute for Nanotechnology Innovation, Guangzhou 510700, China
| |
Collapse
|
30
|
Grissi C, Taverna Porro M, Perona M, Atia M, Negrin L, Moreno MS, Sacanell J, Olivera MS, Del Grosso M, Durán H, Ibañez IL. Superparamagnetic iron oxide nanoparticles induce persistent large foci of DNA damage in human melanoma cells post-irradiation. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2023:10.1007/s00411-023-01037-0. [PMID: 37452828 DOI: 10.1007/s00411-023-01037-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 06/25/2023] [Indexed: 07/18/2023]
Abstract
The synergy of superparamagnetic iron oxide nanoparticles (SPIONs) and ionizing radiation (IR), attributed to reactive oxygen species (ROS) and DNA double-strand breaks (DSBs) increase, was widely investigated in different cancers, but scarcely in melanoma. Herein, SPIONs were evaluated as radiosensitizers in A-375 human melanoma cells. Moreover, the effect of the combined treatment of SPIONs and gamma irradiation (SPIONs-IR) was assessed at the DNA level, where DSBs induction and their repair capacity were studied. SPIONs were synthesized, stabilized by poly(ethylene glycol) methyl ether and physicochemically characterized by high resolution-transmission electron microscopy (HR-TEM), X-ray diffraction and magnetometry and dynamic light scattering. The obtained nanoparticles showing superparamagnetic behavior and low dispersion in shape and sizes were tested in A-375 cells. The intracellular internalization of SPIONs was verified by HR-TEM and quantified by inductively coupled plasma atomic emission spectroscopy. Cells treated with SPIONs exhibited high ROS levels without associated cytotoxicity. Next, a significant radiosensitization in SPIONs-IR vs. control (IR) cells was demonstrated at 1 Gy of gamma radiation. Furthermore, a decreased DSBs repair capacity in SPIONs-IR vs. IR-treated cells was evidenced by the size increase of persistent phosphorylated H2AX foci at 24 h post-irradiation. In conclusion, these nanoparticles show the potential to radiosensitize melanoma cells by the induction of unrepairable DNA damage.
Collapse
Affiliation(s)
- Cecilia Grissi
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Marisa Taverna Porro
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos Aires, Junín 954 (C1113AAD), Ciudad Autónoma de Buenos Aires, Argentina
- Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Campus Miguelete (B1650KNA), San Martín, Provincia de Buenos Aires, Argentina
| | - Marina Perona
- División Bioquímica Nuclear, Departamento de Radiobiología, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. General Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Mariel Atia
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Lara Negrin
- Laboratorio de Radiobiología y Biodosimetría, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Centro de Medicina Nuclear y Radioterapia - Instituto de Tecnologías Nucleares Para La Salud (INTECNUS), Av. Bustillo Km. 9,5 (R8402AGP), S.C. de Bariloche, Río Negro, Argentina
| | - M Sergio Moreno
- Instituto de Nanociencia y Nanotecnología (INN), Comisión Nacional de Energía Atómica (CNEA) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Bariloche, Centro Atómico Bariloche, Av. Bustillo Km. 9,5 (R8402AGP), S.C. de Bariloche, Río Negro, Argentina
| | - Joaquín Sacanell
- Departamento de Física de la Materia Condensada, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - María Silvina Olivera
- Departamento Coordinación BNCT, Comisión Nacional de Energía Atómica (CNEA), Centro Atómico Constituyentes, Av. General Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Mariela Del Grosso
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Hebe Durán
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina.
- Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Campus Miguelete (B1650KNA), San Martín, Provincia de Buenos Aires, Argentina.
| | - Irene L Ibañez
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina.
| |
Collapse
|
31
|
Girigoswami A, Girigoswami K. Potential Applications of Nanoparticles in Improving the Outcome of Lung Cancer Treatment. Genes (Basel) 2023; 14:1370. [PMID: 37510275 PMCID: PMC10379962 DOI: 10.3390/genes14071370] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/20/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Lung cancer is managed using conventional therapies, including chemotherapy, radiation therapy, or a combination of both. Each of these therapies has its own limitations, such as the indiscriminate killing of normal as well as cancer cells, the solubility of the chemotherapeutic drugs, rapid clearance of the drugs from circulation before reaching the tumor site, the resistance of cancer cells to radiation, and over-sensitization of normal cells to radiation. Other treatment modalities include gene therapy, immunological checkpoint inhibitors, drug repurposing, and in situ cryo-immune engineering (ICIE) strategy. Nanotechnology has come to the rescue to overcome many shortfalls of conventional therapies. Some of the nano-formulated chemotherapeutic drugs, as well as nanoparticles and nanostructures with surface modifications, have been used for effective cancer cell killing and radio sensitization, respectively. Nano-enabled drug delivery systems act as cargo to deliver the sensitizer molecules specifically to the tumor cells, thereby enabling the radiation therapy to be more effective. In this review, we have discussed the different conventional chemotherapies and radiation therapies used for inhibiting lung cancer. We have also discussed the improvement in chemotherapy and radiation sensitization using nanoparticles.
Collapse
Affiliation(s)
- Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, Chennai 603103, India
| | - Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, Chennai 603103, India
| |
Collapse
|
32
|
Ding Y, Xiao X, Bai L, Yang B, Lin G, Zeng L, Xie L, Li L, Duan X, Shen J, Zhou J, Pan Y. Enhanced radiosensitivity and chemoradiation efficacy in nasopharyngeal carcinoma via a dual-targeted SPION@polymer hybrid nanosensitizer. NPG ASIA MATERIALS 2023; 15:37. [DOI: 10.1038/s41427-023-00484-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 01/22/2025]
Abstract
AbstractCisplatin-based nanoparticles show good potential in enhancing the effect of nasopharynx carcinoma (NPC) therapy but are still limited by their low radiation sensitization and poor tumor targeting ability. Herein, an ingenious design of multifunctional superparamagnetic iron oxide nanoparticle (SPION)@polymer hybrid nanosensitizer (SPHN) with enhanced radiosensitization and dual-targeting capability is described. SPHN have a core-shell structure, in which radiosensitizer superparamagnetic iron oxide particle (SPION) and cis-platinum (CDDP) are encapsulated in RGD-conjugated amphiphilic block copolymers. These unique structures endow SPHN with outstanding radiosensitization and tumor targeting abilities. When combined with X-rays, SPHN showed strong promotion of the apoptosis of CNE-1 cells in vitro. In addition, RNA-seq and KEGG enrichment analyses indicated that the PI3K-Akt and TNF signaling pathways were closely related to the molecular mechanism of SPHN in chemoradiotherapy. Furthermore, gene set enrichment analysis (GSEA) revealed that SPHN + X-rays treatment decreased translation initiation pathways and the cytoplasmic translation pathway. Through a combination of radiation and chemotherapy, SPHN can achieve remarkable inhibition of tumor growth in vivo, making this nanotechnology a general platform for the chemoradiation therapy of NPC in the future.
Collapse
|
33
|
Misra SK, Rosenholm JM, Pathak K. Functionalized and Nonfunctionalized Nanosystems for Mitochondrial Drug Delivery with Metallic Nanoparticles. Molecules 2023; 28:4701. [PMID: 37375256 DOI: 10.3390/molecules28124701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/04/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Background: The application of metallic nanoparticles as a novel therapeutic tool has significant potential to facilitate the treatment and diagnosis of mitochondria-based disorders. Recently, subcellular mitochondria have been trialed to cure pathologies that depend on their dysfunction. Nanoparticles made from metals and their oxides (including gold, iron, silver, platinum, zinc oxide, and titanium dioxide) have unique modi operandi that can competently rectify mitochondrial disorders. Materials: This review presents insight into the recent research reports on exposure to a myriad of metallic nanoparticles that can alter the dynamic ultrastructure of mitochondria (via altering metabolic homeostasis), as well as pause ATP production, and trigger oxidative stress. The facts and figures have been compiled from more than a hundred PubMed, Web of Science, and Scopus indexed articles that describe the essential functions of mitochondria for the management of human diseases. Result: Nanoengineered metals and their oxide nanoparticles are targeted at the mitochondrial architecture that partakes in the management of a myriad of health issues, including different cancers. These nanosystems not only act as antioxidants but are also fabricated for the delivery of chemotherapeutic agents. However, the biocompatibility, safety, and efficacy of using metal nanoparticles is contested among researchers, which will be discussed further in this review.
Collapse
Affiliation(s)
- Shashi Kiran Misra
- School of Pharmaceutical Sciences, CSJM University Kanpur, Kanpur 208024, India
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, BioCity (3rd Floor), Tykistökatu, 6A, 20520 Turku, Finland
| | - Kamla Pathak
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah 206130, India
| |
Collapse
|
34
|
Zhen W, Weichselbaum RR, Lin W. Nanoparticle-Mediated Radiotherapy Remodels the Tumor Microenvironment to Enhance Antitumor Efficacy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2206370. [PMID: 36524978 PMCID: PMC10213153 DOI: 10.1002/adma.202206370] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/12/2022] [Indexed: 05/26/2023]
Abstract
Radiotherapy (RT) uses ionizing radiation to eradicate localized tumors and, in rare cases, control tumors outside of the irradiated fields via stimulating an antitumor immune response (abscopal effect). However, the therapeutic effect of RT is often limited by inherent physiological barriers of the tumor microenvironment (TME), such as hypoxia, abnormal vasculature, dense extracellular matrix (ECM), and an immunosuppressive TME. Thus, it is critical to develop new RT strategies that can remodel the TME to overcome radio-resistance and immune suppression. In the past decade, high-Z-element nanoparticles have been developed to increase radiotherapeutic indices of localized tumors by reducing X-ray doses and side effects to normal tissues and enhance abscopal effects by activating the TME to elicit systemic antitumor immunity. In this review, the principles of RT and radiosensitization, the mechanisms of radio-resistance and immune suppression, and the use of various nanoparticles to sensitize RT and remodel TMEs for enhanced antitumor efficacy are discussed. The challenges in clinical translation of multifunctional TME-remodeling nanoradiosensitizers are also highlighted.
Collapse
Affiliation(s)
- Wenyao Zhen
- Department of Chemistry, Department of Radiation and Cellular Oncology, and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Wenbin Lin
- Department of Chemistry, Department of Radiation and Cellular Oncology, and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
35
|
Varzandeh M, Sabouri L, Mansouri V, Gharibshahian M, Beheshtizadeh N, Hamblin MR, Rezaei N. Application of nano-radiosensitizers in combination cancer therapy. Bioeng Transl Med 2023; 8:e10498. [PMID: 37206240 PMCID: PMC10189501 DOI: 10.1002/btm2.10498] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 11/08/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
Radiosensitizers are compounds or nanostructures, which can improve the efficiency of ionizing radiation to kill cells. Radiosensitization increases the susceptibility of cancer cells to radiation-induced killing, while simultaneously reducing the potentially damaging effect on the cellular structure and function of the surrounding healthy tissues. Therefore, radiosensitizers are therapeutic agents used to boost the effectiveness of radiation treatment. The complexity and heterogeneity of cancer, and the multifactorial nature of its pathophysiology has led to many approaches to treatment. The effectiveness of each approach has been proven to some extent, but no definitive treatment to eradicate cancer has been discovered. The current review discusses a broad range of nano-radiosensitizers, summarizing possible combinations of radiosensitizing NPs with several other types of cancer therapy options, focusing on the benefits and drawbacks, challenges, and future prospects.
Collapse
Affiliation(s)
- Mohammad Varzandeh
- Department of Materials EngineeringIsfahan University of TechnologyIsfahanIran
| | - Leila Sabouri
- AmitisGen TECH Dev GroupTehranIran
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Vahid Mansouri
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical SciencesTehranIran
| | - Maliheh Gharibshahian
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Student Research CommitteeSchool of Medicine, Shahroud University of Medical SciencesShahroudIran
| | - Nima Beheshtizadeh
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Department of Tissue EngineeringSchool of Advanced Technologies in Medicine, Tehran University of Medical SciencesTehranIran
| | - Michael R. Hamblin
- Laser Research Center, Faculty of Health ScienceUniversity of JohannesburgDoornfonteinSouth Africa
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran
- Research Center for ImmunodeficienciesChildren's Medical Center, Tehran University of Medical SciencesTehranIran
- Department of ImmunologySchool of Medicine, Tehran University of Medical SciencesTehranIran
| |
Collapse
|
36
|
Uzhytchak M, Smolková B, Lunova M, Frtús A, Jirsa M, Dejneka A, Lunov O. Lysosomal nanotoxicity: Impact of nanomedicines on lysosomal function. Adv Drug Deliv Rev 2023; 197:114828. [PMID: 37075952 DOI: 10.1016/j.addr.2023.114828] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Although several nanomedicines got clinical approval over the past two decades, the clinical translation rate is relatively small so far. There are many post-surveillance withdrawals of nanomedicines caused by various safety issues. For successful clinical advancement of nanotechnology, it is of unmet need to realize cellular and molecular foundation of nanotoxicity. Current data suggest that lysosomal dysfunction caused by nanoparticles is emerging as the most common intracellular trigger of nanotoxicity. This review analyzes prospect mechanisms of lysosomal dysfunction-mediated toxicity induced by nanoparticles. We summarized and critically assessed adverse drug reactions of current clinically approved nanomedicines. Importantly, we show that physicochemical properties have great impact on nanoparticles interaction with cells, excretion route and kinetics, and subsequently on toxicity. We analyzed literature on adverse reactions of current nanomedicines and hypothesized that adverse reactions might be linked with lysosomal dysfunction caused by nanomedicines. Finally, from our analysis it becomes clear that it is unjustifiable to generalize safety and toxicity of nanoparticles, since different particles possess distinct toxicological properties. We propose that the biological mechanism of the disease progression and treatment should be central in the optimization of nanoparticle design.
Collapse
Affiliation(s)
- Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Adam Frtús
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic.
| |
Collapse
|
37
|
Chuang YC, Wu PH, Shen YA, Kuo CC, Wang WJ, Chen YC, Lee HL, Chiou JF. Recent Advances in Metal-Based NanoEnhancers for Particle Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1011. [PMID: 36985905 PMCID: PMC10056155 DOI: 10.3390/nano13061011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 06/18/2023]
Abstract
Radiotherapy is one of the most common therapeutic regimens for cancer treatment. Over the past decade, proton therapy (PT) has emerged as an advanced type of radiotherapy (RT) that uses proton beams instead of conventional photon RT. Both PT and carbon-ion beam therapy (CIBT) exhibit excellent therapeutic results because of the physical characteristics of the resulting Bragg peaks, which has been exploited for cancer treatment in medical centers worldwide. Although particle therapies show significant advantages to photon RT by minimizing the radiation damage to normal tissue after the tumors, they still cause damage to normal tissue before the tumor. Since the physical mechanisms are different from particle therapy and photon RT, efforts have been made to ameliorate these effects by combining nanomaterials and particle therapies to improve tumor targeting by concentrating the radiation effects. Metallic nanoparticles (MNPs) exhibit many unique properties, such as strong X-ray absorption cross-sections and catalytic activity, and they are considered nano-radioenhancers (NREs) for RT. In this review, we systematically summarize the putative mechanisms involved in NRE-induced radioenhancement in particle therapy and the experimental results in in vitro and in vivo models. We also discuss the potential of translating preclinical metal-based NP-enhanced particle therapy studies into clinical practice using examples of several metal-based NREs, such as SPION, Abraxane, AGuIX, and NBTXR3. Furthermore, the future challenges and development of NREs for PT are presented for clinical translation. Finally, we propose a roadmap to pursue future studies to strengthen the interplay of particle therapy and nanomedicine.
Collapse
Affiliation(s)
- Yao-Chen Chuang
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
| | - Ping-Hsiu Wu
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Proton Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110301, Taiwan
| | - Yao-An Shen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Chia-Chun Kuo
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
- Proton Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110301, Taiwan
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei 110301, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Jun Wang
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
- Proton Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110301, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Yu-Chen Chen
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Hsin-Lun Lee
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Proton Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110301, Taiwan
| | - Jeng-Fong Chiou
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan; (Y.-C.C.)
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Proton Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110301, Taiwan
| |
Collapse
|
38
|
Vangijzegem T, Lecomte V, Ternad I, Van Leuven L, Muller RN, Stanicki D, Laurent S. Superparamagnetic Iron Oxide Nanoparticles (SPION): From Fundamentals to State-of-the-Art Innovative Applications for Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15010236. [PMID: 36678868 PMCID: PMC9861355 DOI: 10.3390/pharmaceutics15010236] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/01/2023] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
Despite significant advances in cancer therapy over the years, its complex pathological process still represents a major health challenge when seeking effective treatment and improved healthcare. With the advent of nanotechnologies, nanomedicine-based cancer therapy has been widely explored as a promising technology able to handle the requirements of the clinical sector. Superparamagnetic iron oxide nanoparticles (SPION) have been at the forefront of nanotechnology development since the mid-1990s, thanks to their former role as contrast agents for magnetic resonance imaging. Though their use as MRI probes has been discontinued due to an unfavorable cost/benefit ratio, several innovative applications as therapeutic tools have prompted a renewal of interest. The unique characteristics of SPION, i.e., their magnetic properties enabling specific response when submitted to high frequency (magnetic hyperthermia) or low frequency (magneto-mechanical therapy) alternating magnetic field, and their ability to generate reactive oxygen species (either intrinsically or when activated using various stimuli), make them particularly adapted for cancer therapy. This review provides a comprehensive description of the fundamental aspects of SPION formulation and highlights various recent approaches regarding in vivo applications in the field of cancer therapy.
Collapse
Affiliation(s)
- Thomas Vangijzegem
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
- Correspondence: (T.V.); (S.L.)
| | - Valentin Lecomte
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
| | - Indiana Ternad
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
| | - Levy Van Leuven
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
| | - Robert N. Muller
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
- Center for Microscopy and Molecular Imaging (CMMI), Non-Ionizing Molecular Imaging Unit, 6041 Gosselies, Belgium
| | - Dimitri Stanicki
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
| | - Sophie Laurent
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
- Center for Microscopy and Molecular Imaging (CMMI), Non-Ionizing Molecular Imaging Unit, 6041 Gosselies, Belgium
- Correspondence: (T.V.); (S.L.)
| |
Collapse
|
39
|
Ternad I, Penninckx S, Lecomte V, Vangijzegem T, Conrard L, Lucas S, Heuskin AC, Michiels C, Muller RN, Stanicki D, Laurent S. Advances in the Mechanistic Understanding of Iron Oxide Nanoparticles' Radiosensitizing Properties. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:201. [PMID: 36616111 PMCID: PMC9823929 DOI: 10.3390/nano13010201] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/16/2022] [Accepted: 12/26/2022] [Indexed: 06/17/2023]
Abstract
Among the plethora of nanosystems used in the field of theranostics, iron oxide nanoparticles (IONPs) occupy a central place because of their biocompatibility and magnetic properties. In this study, we highlight the radiosensitizing effect of two IONPs formulations (namely 7 nm carboxylated IONPs and PEG5000-IONPs) on A549 lung carcinoma cells when exposed to 225 kV X-rays after 6 h, 24 h and 48 h incubation. The hypothesis that nanoparticles exhibit their radiosensitizing effect by weakening cells through the inhibition of detoxification enzymes was evidenced by thioredoxin reductase activity monitoring. In particular, a good correlation between the amplification effect at 2 Gy and the residual activity of thioredoxin reductase was observed, which is consistent with previous observations made for gold nanoparticles (NPs). This emphasizes that NP-induced radiosensitization does not result solely from physical phenomena but also results from biological events.
Collapse
Affiliation(s)
- Indiana Ternad
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), B-7000 Mons, Belgium
| | - Sebastien Penninckx
- Medical Physics Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), B-1070 Brussels, Belgium
| | - Valentin Lecomte
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), B-7000 Mons, Belgium
| | - Thomas Vangijzegem
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), B-7000 Mons, Belgium
| | - Louise Conrard
- Center for Microscopy and Molecular Imaging (CMMI), B-6041 Gosselies, Belgium
| | - Stéphane Lucas
- Namur Research Institute for Life Sciences (NARILIS), University of Namur, B-5000 Namur, Belgium
| | - Anne-Catherine Heuskin
- Namur Research Institute for Life Sciences (NARILIS), University of Namur, B-5000 Namur, Belgium
| | - Carine Michiels
- Namur Research Institute for Life Sciences (NARILIS), University of Namur, B-5000 Namur, Belgium
| | - Robert N. Muller
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), B-7000 Mons, Belgium
- Center for Microscopy and Molecular Imaging (CMMI), B-6041 Gosselies, Belgium
| | - Dimitri Stanicki
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), B-7000 Mons, Belgium
| | - Sophie Laurent
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), B-7000 Mons, Belgium
- Center for Microscopy and Molecular Imaging (CMMI), B-6041 Gosselies, Belgium
| |
Collapse
|
40
|
Majeed S, Mohd Rozi NAB, Danish M, Mohamad Ibrahim M, Joel EL. Invitro apoptosis and molecular response of engineered green iron oxide nanoparticles with l-arginine in MD-MBA 231 breast cancer cells. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
41
|
Improve the cytotoxic effects of megavoltage radiation treatment by Fe3O4@Cus–PEG nanoparticles as a novel radiosensitizer in colorectal cancer cells. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00131-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
To enhance the performance of radiotherapy, emerging nanoparticles that can professionally enhance X-ray irradiation to destruct cancer cells are extremely necessary. Here, we examined the potential of PEG-coated magnetite copper sulfide hetero-nanoparticles (Fe3O4@Cus–PEG) as a radiosensitizer agent.
Methods
Fe3O4@Cus–PEG nanoparticles were synthesized and characterized. The toxicity of nanoparticles on HT-29 colorectal cancer cells was assessed by the MTT assay. The radio-sensitizing effects of Fe3O4@Cus–PEG nanoparticles on HT-29 cancer cells were investigated by the MTT and colony formation assays. Moreover, the underlying mechanisms for Fe3O4@Cus–PEG nanoparticles to improve the radiation sensitivity of cells were evaluated.
Results
The results demonstrated that nanoparticles enhanced the effects of X-ray irradiation in a dose-dependent manner. The effects of combined treatments (nanoparticles and X-ray radiation) were strongly synergistic. The sensitizing enhancement ratio (SER) of nanoparticles was 2.02. Our in vitro assays demonstrated that the nitric oxide production, the intracellular hydrogen peroxide concentration, and the expression level of Bax and Caspase-3 genes significantly increased in the cells treated with the combination of nanoparticles and radiation. Whereas, the Glutathione peroxidase enzyme activity and the expression level of the Bcl-2 gene in the combined treatment significantly decreased compared to the radiation alone.
Conclusions
Our study suggests that Fe3O4@Cus–PEG nanoparticles are the promising nano radio-sensitizing agents for the treatment of cancer cells to enhance the efficacy of radiation therapy through increasing the reactive oxygen species generation, nitric oxide production, and inducing apoptosis.
Graphical Abstract
Collapse
|
42
|
Varzandeh M, Labbaf S, Varshosaz J, Laurent S. An overview of the intracellular localization of high-Z nanoradiosensitizers. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 175:14-30. [PMID: 36029849 DOI: 10.1016/j.pbiomolbio.2022.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/17/2022] [Accepted: 08/19/2022] [Indexed: 06/15/2023]
Abstract
Radiation therapy (RT) is a method commonly used for cancer treatment worldwide. Commonly, RT utilizes two routes for combating cancers: 1) high-energy radiation to generate toxic reactive oxygen species (ROS) (through the dissociation of water molecules) for damaging the deoxyribonucleic acid (DNA) inside the nucleus 2) direct degradation of the DNA. However, cancer cells have mechanisms to survive under intense RT, which can considerably decrease its therapeutic efficacy. Excessive radiation energy damages healthy tissues, and hence, low doses are applied for cancer treatment. Additionally, different radiosensitizers were used to sensitize cancer cells towards RT through individual mechanisms. Following this route, nanoparticle-based radiosensitizers (herein called nanoradiosensitizers) have recently gained attention owing to their ability to produce massive electrons which leads to the production of a huge amount of ROS. The success of the nanoradiosensitizer effect is closely correlated to its interaction with cells and its localization within the cells. In other words, tumor treatment is affected from the chain of events which is started from cell-nanoparticle interaction followed by the nanoparticles direction and homing inside the cell. Therefore, passive or active targeting of the nanoradiosensitizers in the subcellular level and the cell-nano interaction would determine the efficacy of the radiation therapy. The importance of the nanoradiosensitizer's targeting is increased while the organelles beyond nucleus are recently recognized as the mediators of the cancer cell death or resistance under RT. In this review, the principals of cell-nanomaterial interactions and which dominate nanoradiosensitizer efficiency in cancer therapy, are thoroughly discussed.
Collapse
Affiliation(s)
- Mohammad Varzandeh
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Sheyda Labbaf
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center and Department of Pharmaceutics, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Sophie Laurent
- Laboratory of NMR and Molecular Imaging, Department of General, Organic Chemistry and Biomedical, University of Mons, Mons, Belgium.
| |
Collapse
|
43
|
Aram E, Moeni M, Abedizadeh R, Sabour D, Sadeghi-Abandansari H, Gardy J, Hassanpour A. Smart and Multi-Functional Magnetic Nanoparticles for Cancer Treatment Applications: Clinical Challenges and Future Prospects. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12203567. [PMID: 36296756 PMCID: PMC9611246 DOI: 10.3390/nano12203567] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/16/2022] [Accepted: 09/27/2022] [Indexed: 05/14/2023]
Abstract
Iron oxide nanoparticle (IONPs) have become a subject of interest in various biomedical fields due to their magnetism and biocompatibility. They can be utilized as heat mediators in magnetic hyperthermia (MHT) or as contrast media in magnetic resonance imaging (MRI), and ultrasound (US). In addition, their high drug-loading capacity enabled them to be therapeutic agent transporters for malignancy treatment. Hence, smartening them allows for an intelligent controlled drug release (CDR) and targeted drug delivery (TDD). Smart magnetic nanoparticles (SMNPs) can overcome the impediments faced by classical chemo-treatment strategies, since they can be navigated and release drug via external or internal stimuli. Recently, they have been synchronized with other modalities, e.g., MRI, MHT, US, and for dual/multimodal theranostic applications in a single platform. Herein, we provide an overview of the attributes of MNPs for cancer theranostic application, fabrication procedures, surface coatings, targeting approaches, and recent advancement of SMNPs. Even though MNPs feature numerous privileges over chemotherapy agents, obstacles remain in clinical usage. This review in particular covers the clinical predicaments faced by SMNPs and future research scopes in the field of SMNPs for cancer theranostics.
Collapse
Affiliation(s)
- Elham Aram
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
- Department of Polymer Engineering, Faculty of Engineering, Golestan University, Gorgan 49188-88369, Iran
| | - Masome Moeni
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK
| | - Roya Abedizadeh
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
| | - Davood Sabour
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
| | - Hamid Sadeghi-Abandansari
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Jabbar Gardy
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: (J.G.); (A.H.)
| | - Ali Hassanpour
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: (J.G.); (A.H.)
| |
Collapse
|
44
|
Zhang Y, Sriramaneni RN, Clark PA, Jagodinsky JC, Ye M, Jin W, Wang Y, Bates A, Kerr CP, Le T, Allawi R, Wang X, Xie R, Havighurst TC, Chakravarty I, Rakhmilevich AL, O'Leary KA, Schuler LA, Sondel PM, Kim K, Gong S, Morris ZS. Multifunctional nanoparticle potentiates the in situ vaccination effect of radiation therapy and enhances response to immune checkpoint blockade. Nat Commun 2022; 13:4948. [PMID: 35999216 PMCID: PMC9399096 DOI: 10.1038/s41467-022-32645-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
Radiation therapy (RT) activates an in situ vaccine effect when combined with immune checkpoint blockade (ICB), yet this effect may be limited because RT does not fully optimize tumor antigen presentation or fully overcome suppressive mechanisms in the tumor-immune microenvironment. To overcome this, we develop a multifunctional nanoparticle composed of polylysine, iron oxide, and CpG (PIC) to increase tumor antigen presentation, increase the ratio of M1:M2 tumor-associated macrophages, and enhance stimulation of a type I interferon response in conjunction with RT. In syngeneic immunologically "cold" murine tumor models, the combination of RT, PIC, and ICB significantly improves tumor response and overall survival resulting in cure of many mice and consistent activation of tumor-specific immune memory. Combining RT with PIC to elicit a robust in situ vaccine effect presents a simple and readily translatable strategy to potentiate adaptive anti-tumor immunity and augment response to ICB or potentially other immunotherapies.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Biomedical Engineering and Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Paul A Clark
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Justin C Jagodinsky
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Mingzhou Ye
- Department of Biomedical Engineering and Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Wonjong Jin
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Yuyuan Wang
- Department of Biomedical Engineering and Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Amber Bates
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Caroline P Kerr
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, USA
| | - Trang Le
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Raad Allawi
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Xiuxiu Wang
- Department of Biomedical Engineering and Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ruosen Xie
- Department of Biomedical Engineering and Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Thomas C Havighurst
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Ishan Chakravarty
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Kathleen A O'Leary
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - Kyungmann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Shaoqin Gong
- Department of Biomedical Engineering and Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
45
|
Barbaro D, Di Bari L, Gandin V, Marzano C, Ciaramella A, Malventi M, Evangelisti C. Glucose-coated superparamagnetic iron oxide nanoparticles prepared by metal vapor synthesis can target GLUT1 overexpressing tumors: In vitro tests and in vivo preliminary assessment. PLoS One 2022; 17:e0269603. [PMID: 35704647 PMCID: PMC9200296 DOI: 10.1371/journal.pone.0269603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 05/24/2022] [Indexed: 11/21/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) coated with glucose (Glc-SPIONs) were prepared by a new approach called Metal Vapor Synthesis (MVS) and their morphological/structural features were investigated by transmission electron microscopy (TEM) and dynamic light scattering. TEM analysis revealed the presence of small roundish crystalline iron oxide nanoparticles in the organic amorphous phase of glucose, The particles were distributed in a narrow range (1.5 nm—3.5 nm) with a mean diameter of 2.7 nm. The hydrodynamic mean diameter of the Glc-SPIONs, was 15.5 nm. From 4 mg/mL onwards, there was a constant level of positive contrast in a T1-weighted sequence. In vitro experiments were performed in three cell lines: pancreatic cancer (PSN-1), human thyroid cancer (BCPAP), and human embryonic kidney non-tumor cells. We evaluated GLUT1 expression in each cell line and demonstrated that the exposure time and concentration of the Glc-SPIONs we used did not affect cell viability. PSN-1 cells were the most effective at internalizing Glc-SPIONs. Although significantly higher than the control cells, a lower Fe content was detected BCPAP cells treated with Glc-SPIONs. To confirm the involvement of GLUT1 in Glc-SPIONs internalization, cellular uptake experiments were also conducted by pre-treating cancer cells with specific GLUT1 inhibitors, All the inhibitors reduced the cancer cell uptake of Glc-SPIONs In vivo tests were performed on mice inoculated with Lewis lung carcinoma. Mice were treated with a single i.v. injection of Glc-SPION and our results showed a great bioavailability to the malignant tissue by the i.v. administration of Glc-SPIONs. Glc-SPIONs were efficiently eliminated by the kidney. To the best of our knowledge, our study demonstrates for the first time that Glc-SPIONs prepared with MVS can be electively internalized by tumor cells both in vitro and in vivo by exploiting one of the most universal metabolic anomalies of cancer.
Collapse
Affiliation(s)
- Daniele Barbaro
- U.O. Endocrinology, General Hospital, Livorno, Livorno, Italy
- * E-mail:
| | - Lorenzo Di Bari
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Valentina Gandin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Cristina Marzano
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | | | - Claudio Evangelisti
- Institute for the Chemistry of OrganoMetallic Compounds, Italian National Council for Research (ICCOM-CNR), Pisa, Italy
| |
Collapse
|
46
|
Zhao Z, Li M, Zeng J, Huo L, Liu K, Wei R, Ni K, Gao J. Recent advances in engineering iron oxide nanoparticles for effective magnetic resonance imaging. Bioact Mater 2022; 12:214-245. [PMID: 35310380 PMCID: PMC8897217 DOI: 10.1016/j.bioactmat.2021.10.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/27/2021] [Accepted: 10/10/2021] [Indexed: 02/09/2023] Open
Abstract
Iron oxide nanoparticle (IONP) with unique magnetic property and high biocompatibility have been widely used as magnetic resonance imaging (MRI) contrast agent (CA) for long time. However, a review which comprehensively summarizes the recent development of IONP as traditional T2 CA and its new application for different modality of MRI, such as T1 imaging, simultaneous T2/T1 or MRI/other imaging modality, and as environment responsive CA is rare. This review starts with an investigation of direction on the development of high-performance MRI CA in both T2 and T1 modal based on quantum mechanical outer sphere and Solomon–Bloembergen–Morgan (SBM) theory. Recent rational attempts to increase the MRI contrast of IONP by adjusting the key parameters, including magnetization, size, effective radius, inhomogeneity of surrounding generated magnetic field, crystal phase, coordination number of water, electronic relaxation time, and surface modification are summarized. Besides the strategies to improve r2 or r1 values, strategies to increase the in vivo contrast efficiency of IONP have been reviewed from three different aspects, those are introducing second imaging modality to increase the imaging accuracy, endowing IONP with environment response capacity to elevate the signal difference between lesion and normal tissue, and optimizing the interface structure to improve the accumulation amount of IONP in lesion. This detailed review provides a deep understanding of recent researches on the development of high-performance IONP based MRI CAs. It is hoped to trigger deep thinking for design of next generation MRI CAs for early and accurate diagnosis. T2 contrast capacity of iron oxide nanoparticles (IONPs) could be improved based on quantum mechanical outer sphere theory. IONPs could be expand to be used as effective T1 CAs by improving q value, extending τs, and optimizing interface structure. Environment responsive MRI CAs have been developed to improve the diagnosis accuracy. Introducing other imaging contrast moiety into IONPs could increase the contrast efficiency. Optimizing in vivo behavior of IONPs have been proved to enlarge the signal difference between normal tissue and lesion.
Collapse
|
47
|
Delille F, Pu Y, Lequeux N, Pons T. Designing the Surface Chemistry of Inorganic Nanocrystals for Cancer Imaging and Therapy. Cancers (Basel) 2022; 14:2456. [PMID: 35626059 PMCID: PMC9139368 DOI: 10.3390/cancers14102456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/27/2022] Open
Abstract
Inorganic nanocrystals, such as gold, iron oxide and semiconductor quantum dots, offer promising prospects for cancer diagnostics, imaging and therapy, due to their specific plasmonic, magnetic or fluorescent properties. The organic coating, or surface ligands, of these nanoparticles ensures their colloidal stability in complex biological fluids and enables their functionalization with targeting functions. It also controls the interactions of the nanoparticle with biomolecules in their environment. It therefore plays a crucial role in determining nanoparticle biodistribution and, ultimately, the imaging or therapeutic efficiency. This review summarizes the various strategies used to develop optimal surface chemistries for the in vivo preclinical and clinical application of inorganic nanocrystals. It discusses the current understanding of the influence of the nanoparticle surface chemistry on its colloidal stability, interaction with proteins, biodistribution and tumor uptake, and the requirements to develop an optimal surface chemistry.
Collapse
Affiliation(s)
- Fanny Delille
- Laboratoire de Physique et d’Etude des Matériaux, Ecole Supérieure de Physique et Chimie Industrielle, Université PSL (Paris Sciences & Lettres), Centre National de Recherche Scientifique, 75005 Paris, France; (F.D.); (Y.P.); (N.L.)
- Laboratoire de Physique et d’Etude des Matériaux, Centre National de Recherche Scientifique, Sorbonne Université, 75005 Paris, France
| | - Yuzhou Pu
- Laboratoire de Physique et d’Etude des Matériaux, Ecole Supérieure de Physique et Chimie Industrielle, Université PSL (Paris Sciences & Lettres), Centre National de Recherche Scientifique, 75005 Paris, France; (F.D.); (Y.P.); (N.L.)
- Laboratoire de Physique et d’Etude des Matériaux, Centre National de Recherche Scientifique, Sorbonne Université, 75005 Paris, France
| | - Nicolas Lequeux
- Laboratoire de Physique et d’Etude des Matériaux, Ecole Supérieure de Physique et Chimie Industrielle, Université PSL (Paris Sciences & Lettres), Centre National de Recherche Scientifique, 75005 Paris, France; (F.D.); (Y.P.); (N.L.)
- Laboratoire de Physique et d’Etude des Matériaux, Centre National de Recherche Scientifique, Sorbonne Université, 75005 Paris, France
| | - Thomas Pons
- Laboratoire de Physique et d’Etude des Matériaux, Ecole Supérieure de Physique et Chimie Industrielle, Université PSL (Paris Sciences & Lettres), Centre National de Recherche Scientifique, 75005 Paris, France; (F.D.); (Y.P.); (N.L.)
- Laboratoire de Physique et d’Etude des Matériaux, Centre National de Recherche Scientifique, Sorbonne Université, 75005 Paris, France
| |
Collapse
|
48
|
Zhang Z, Zhou F, Davies G, Williams GR. Theranostics for MRI‐guided therapy: Recent developments. VIEW 2022. [DOI: 10.1002/viw.20200134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Ziwei Zhang
- UCL School of Pharmacy University College London London UK
- UCL Department of Chemistry University College London London UK
| | - Feng‐Lei Zhou
- Department of Medical Physics and Biomedical Engineering University College London London UK
- College of Textiles and Clothing Qingdao University Qingdao PR China
| | | | | |
Collapse
|
49
|
Yoshida A, Kitayama Y, Hayakawa N, Mizukawa Y, Nishimura Y, Takano E, Sunayama H, Takeuchi T. Biocompatible polymer-modified gold nanocomposites of different shapes as radiation sensitizers. Biomater Sci 2022; 10:2665-2672. [PMID: 35420601 DOI: 10.1039/d2bm00174h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Radiation therapy is a powerful approach for cancer treatment due to its low invasiveness. The development of radiation sensitizers is of great importance as they assist in providing radiation therapy at a low dose. In this study, poly(2-methacryloyloxyethyl phosphorylcholine) (PMPC)-modified gold nanocomposites of different shapes were created using the grafting-to approach to serve as a novel radiation sensitizer with high cellular uptake. The effect of the shape of the nanocomposite on cellular uptake by the breast cancer cell line MCF-7 was also investigated. The PMPC-modified gold nanostars showed the highest cellular uptake compared to the other gold nanocomposites (spheres and rods), whereas cell cytotoxicity was negligible among all candidates. Furthermore, the therapeutic effect of radiation of PMPC-modified nanostars was the highest among all the gold nanocomposites. These results clearly indicate that the shape of the gold nanocomposite is an important parameter for cellular uptake and radiation sensitizing effects in breast cancer cells.
Collapse
Affiliation(s)
- Aoi Yoshida
- Graduate School of Engineering, Kobe University, Nada-ku, Kobe 657-8501, Japan.
| | - Yukiya Kitayama
- Graduate School of Engineering, Osaka Prefecture University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan.
| | - Natsuki Hayakawa
- Graduate School of Engineering, Kobe University, Nada-ku, Kobe 657-8501, Japan.
| | - Yuki Mizukawa
- Graduate School of Engineering, Kobe University, Nada-ku, Kobe 657-8501, Japan.
| | - Yuya Nishimura
- Graduate School of Science, Technology and Innovation, Kobe University, Nada-ku, Kobe 657-8501, Japan
| | - Eri Takano
- Graduate School of Engineering, Kobe University, Nada-ku, Kobe 657-8501, Japan.
| | - Hirobumi Sunayama
- Graduate School of Engineering, Kobe University, Nada-ku, Kobe 657-8501, Japan.
| | - Toshifumi Takeuchi
- Graduate School of Engineering, Kobe University, Nada-ku, Kobe 657-8501, Japan. .,Center for Advanced Medical Engineering Research & Development (CAMED), Kobe University, Chuo-ku, Kobe 650-0047, Japan
| |
Collapse
|
50
|
Askar MA, El-Nashar HA, Al-Azzawi MA, Rahman SSA, Elshawi OE. Synergistic Effect of Quercetin Magnetite Nanoparticles and Targeted Radiotherapy in Treatment of Breast Cancer. BREAST CANCER: BASIC AND CLINICAL RESEARCH 2022; 16:11782234221086728. [PMID: 35359610 PMCID: PMC8961357 DOI: 10.1177/11782234221086728] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/17/2022] [Indexed: 01/16/2023] Open
Abstract
Quercetin is a potent cancer therapeutic agent present in fruits and vegetables. The pharmaceutical uses of quercetin are limited due to many problems associated with low solubility, bioavailability, permeability, and instability. In addition, the high doses of quercetin show toxic effects in clinical and experimental studies. Therefore, a new strategy is warranted to overcome these problems without the use of toxic doses. The iron oxide nanoparticles can be used as a drug delivery system. This study aimed to prepare quercetin-conjugated magnetite nanoparticles (QMNPs) using biological simple nanoprecipitation and mediated by fungus Aspergillus oryzae. Also, we initiated in vitro and in vivo studies to determine whether QMNPs might sensitize breast cancer to radiotherapy treatment. The structural, morphological, and magnetic properties of the prepared nanoparticles were studied. The results indicated that QMNPs were spherical in shape and 40 nm in diameter. The in vitro studies showed that the incubation of MCF-7, HePG-2, and A459 cancer cells with QMNPs for 24 h effectively inhibited the growth of cancer cell lines in a concentration-dependent manner with IC50 values of 11, 77.5, and104 nmol/mL, respectively. The combination of QMNPs with irradiation (IR) potently blocked MCF-7 cancer cell proliferation and showed significant changes in the morphology of these cells as observed by bright-field inverted light microscopy. Focusing on the long-term toxicity of QMNPs (20 ml/kg), the assessment of hematological, hepatic, and renal markers indicated no toxic effect. Besides, QMNPs inhibited tumor growth and potently enhanced the lateral radiotherapy treatment in N-methyl-N-nitrosourea (MNU)-induced breast cancer in female white albino rats. These anticancer and radiosensitizing activities were ascribed to cytotoxicity, cell cycle arrest, immunomodulation, and efficiency through induction of apoptosis. In a conclusion, these observations suggest that the QMNPs combined with LRT could act as a potential targeted therapy in breast cancer.
Collapse
Affiliation(s)
- Mostafa A Askar
- Department of Radiation Biology, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Heba As El-Nashar
- Department of Pharmacognosy and Center of Drug Discovery Research and Development, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mahmood A Al-Azzawi
- Department of Pathological Analysis Techniques, College of Medical & Health Technologies, Ahl Al Bayt University, Karbala, Iraq
| | - Sahar S Abdel Rahman
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Omama E Elshawi
- Department of Health and Radiation Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|