1
|
Aubrun C, Doussineau T, Carmès L, Meyzaud A, Boux F, Dufort S, Delfour A, De Beaumont O, Mirjolet C, Le Duc G. Mechanisms of Action of AGuIX as a Pan-Cancer Nano-Radiosensitizer: A Comprehensive Review. Pharmaceuticals (Basel) 2025; 18:519. [PMID: 40283954 PMCID: PMC12030438 DOI: 10.3390/ph18040519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Objective: This review provides an overview of the current knowledge regarding the mechanisms of action of AGuIX, a clinical-stage theranostic nano-radiosensitizer composed of gadolinium. It covers the steps following the administration, from the internalization in tumor cells to the interaction with X-rays and the subsequent physical, chemical, biological, and immunological events. Results: After intravenous injection, AGuIX accumulates in tumors through the enhanced permeability and retention (EPR) effect, and its specific retention properties allow its persistence in tumors for several days. At the cellular level, the nanomedicine is internalized by endocytic processes and mainly located in the cytoplasm, especially in lysosomes. AGuIX enhances the effects of radiotherapy (RT) at several levels, starting from radiation-matter interactions to a chemical stage of reactive oxygen species (ROS) production, followed by a cascade of biological events leading to tumor cell death and immune response. Indeed, AGuIX induces a local increase in radiation dose deposition through the emission of Auger electrons, leading to a subsequent increase in ROS generation. AGuIX also impacts RT-induced biological mechanisms, including DNA damage and cell death mechanisms such as apoptosis, autophagic cell death, and ferroptosis. Last, the combination of AGuIX and RT stimulates an antitumor immune response through the induction of immunogenic cell death (ICD), the activation of dendritic and T cells, and the reprogramming of tumor-associated macrophages (TAMs) into a pro-inflammatory phenotype. Conclusions: AGuIX is a clinical-stage nanoparticle (NP) intravenously administered with pan-cancer potential due to its specific biodistribution properties and a strong ability to amplify RT-induced mechanisms.
Collapse
Affiliation(s)
- Clémentine Aubrun
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Tristan Doussineau
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Léna Carmès
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Aurélien Meyzaud
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Fabien Boux
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Sandrine Dufort
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Adeline Delfour
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
- X-Rain: Research Unit in Radiotherapy Combined with Immunotherapies and Nanoparticles, IMATHERA, Radiation Therapy Department, Centre Georges-François Leclerc, 21000 Dijon, France;
- TIReCS Team, CTM (Center for Translational and Molecular Medicine), INSERM UMR 1231, 21000 Dijon, France
| | - Olivier De Beaumont
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Céline Mirjolet
- X-Rain: Research Unit in Radiotherapy Combined with Immunotherapies and Nanoparticles, IMATHERA, Radiation Therapy Department, Centre Georges-François Leclerc, 21000 Dijon, France;
- TIReCS Team, CTM (Center for Translational and Molecular Medicine), INSERM UMR 1231, 21000 Dijon, France
| | - Géraldine Le Duc
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| |
Collapse
|
2
|
Blind S, Lerouge L, Gries M, Retif P, Thomas N, Barberi-Heyob M, Daouk J. An alternate model to describe the radio-potentializing effects of metal-based nanoparticles in radiation therapy. Comput Biol Med 2025; 188:109861. [PMID: 39970825 DOI: 10.1016/j.compbiomed.2025.109861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND AND OBJECTIVES The use of numerical models to predict radiosensitizing properties induced by metal-based nanoparticles (NPs) remains a real challenge in oncology. As most of the interactions due to radiation in biological environments originate from the secondary particles produced, we aim to formalize the relationship between these secondary particles, the irradiation dose and the NP concentration, to optimize mathematical and numerical tools for assessing NP-induced radiosensitization. METHODS GATE simulations were carried out to demonstrate a linear and affine relationship between specific radiophysical quantities, the irradiation dose and NP concentration. This research has led to an effective new method for predicting radiophysical events and the proposal of a new model for predicting cell death. This model was confirmed by experimental biological results obtained from a clonogenic assay performed on U251 and U87 glioblastoma cells after exposure to different concentrations of metal-based NPs. RESULTS We achieved an efficient method for quantifying certain radiophysical species (number of ionizations, photo- and compton electrons, bremsstrahlung and deposited dose) in the presence of NPs and at different irradiation doses. These findings have enabled us to suggest an extension of the linear quadratic (LQ) cell survival model. The LQ extension model was compared with experimental data both obtained in the laboratory and extracted from the literature. CONCLUSIONS Radiophysical events provide valuable information for predicting the radiobiological and radiosensitizing effects of metal-based NPs in the context of X-ray photon irradiation. The extension of the LQ model we developed enables cell death to be predicted for different NP concentrations based on concentration effects alone.
Collapse
Affiliation(s)
- Sarah Blind
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France
| | - Lucie Lerouge
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France
| | - Mickaël Gries
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Paul Retif
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; Department of Medical Physics, Mercy Hospital, CHR Metz-Thionville, F-57530 Ars-Laquenexy, France
| | - Noémie Thomas
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France
| | | | - Joël Daouk
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France.
| |
Collapse
|
3
|
Bennett S, Verry C, Kaza E, Miao X, Dufort S, Boux F, Crémillieux Y, de Beaumont O, Le Duc G, Berbeco R, Sudhyadhom A. Quantifying gadolinium-based nanoparticle uptake distributions in brain metastases via magnetic resonance imaging. Sci Rep 2024; 14:11959. [PMID: 38796495 PMCID: PMC11128019 DOI: 10.1038/s41598-024-62389-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/16/2024] [Indexed: 05/28/2024] Open
Abstract
AGuIX, a novel gadolinium-based nanoparticle, has been deployed in a pioneering double-blinded Phase II clinical trial aiming to assess its efficacy in enhancing radiotherapy for tumor treatment. This paper moves towards this goal by analyzing AGuIX uptake patterns in 23 patients. A phantom was designed to establish the relationship between AGuIX concentration and longitudinal ( T 1 ) relaxation. A 3T MRI and MP2RAGE sequence were used to generate patient T 1 maps. AGuIX uptake in tumors was determined based on longitudinal relaxivity. AGuIX (or placebo) was administered to 23 patients intravenously at 100 mg/kg 1-5 hours pre-imaging. Each of 129 brain metastases across 23 patients were captured in T 1 maps and examined for AGuIX uptake and distribution. Inferred AGuIX recipients had average tumor uptakes between 0.012 and 0.17 mg/ml, with a mean of 0.055 mg/ml. Suspected placebo recipients appeared to have no appreciable uptake. Tumors presented with varying spatial AGuIX uptake distributions, suspected to be related to differences in accumulation time and patient-specific bioaccumulation factors. This research demonstrates AGuIX's ability to accumulate in brain metastases, with quantifiable uptake via T 1 mapping. Future analyses will extend these methods to complete clinical trial data (~ 134 patients) to evaluate the potential relationship between nanoparticle uptake and possible tumor response following radiotherapy.Clinical Trial Registration Number: NCT04899908.Clinical Trial Registration Date: 25/05/2021.
Collapse
Affiliation(s)
- Stephanie Bennett
- Brigham and Women's Hospital|Dana-Farber Cancer Institute|Harvard Medical School, Boston, MA, USA.
| | - Camille Verry
- Université Grenoble Alpes, CHU Grenoble Alpes, Service de Radiothérapie, Inserm UA7, Grenoble, France
| | - Evangelia Kaza
- Brigham and Women's Hospital|Dana-Farber Cancer Institute|Harvard Medical School, Boston, MA, USA
| | - Xin Miao
- Siemens Medical Solutions USA Inc., Malvern, PA, USA
| | | | | | - Yannick Crémillieux
- NH TherAguix SA, Meylan, France
- Institut des Sciences Moléculaires, UMR5255, Université de Bordeaux, Bordeaux, France
| | | | | | - Ross Berbeco
- Brigham and Women's Hospital|Dana-Farber Cancer Institute|Harvard Medical School, Boston, MA, USA
| | - Atchar Sudhyadhom
- Brigham and Women's Hospital|Dana-Farber Cancer Institute|Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Kouri MA, Spyratou E, Kalkou ME, Patatoukas G, Angelopoulou E, Tremi I, Havaki S, Gorgoulis VG, Kouloulias V, Platoni K, Efstathopoulos EP. Nanoparticle-Mediated Radiotherapy: Unraveling Dose Enhancement and Apoptotic Responses in Cancer and Normal Cell Lines. Biomolecules 2023; 13:1720. [PMID: 38136591 PMCID: PMC10742116 DOI: 10.3390/biom13121720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Cervical cancer remains a pressing global health concern, necessitating advanced therapeutic strategies. Radiotherapy, a fundamental treatment modality, has faced challenges such as targeted dose deposition and radiation exposure to healthy tissues, limiting optimal outcomes. To address these hurdles, nanomaterials, specifically gold nanoparticles (AuNPs), have emerged as a promising avenue. This study delves into the realm of cervical cancer radiotherapy through the meticulous exploration of AuNPs' impact. Utilizing ex vivo experiments involving cell lines, this research dissected intricate radiobiological interactions. Detailed scrutiny of cell survival curves, dose enhancement factors (DEFs), and apoptosis in both cancer and normal cervical cells revealed profound insights. The outcomes showcased the substantial enhancement of radiation responses in cancer cells following AuNP treatment, resulting in heightened cell death and apoptotic levels. Significantly, the most pronounced effects were observed 24 h post-irradiation, emphasizing the pivotal role of timing in AuNPs' efficacy. Importantly, AuNPs exhibited targeted precision, selectively impacting cancer cells while preserving normal cells. This study illuminates the potential of AuNPs as potent radiosensitizers in cervical cancer therapy, offering a tailored and efficient approach. Through meticulous ex vivo experimentation, this research expands our comprehension of the complex dynamics between AuNPs and cells, laying the foundation for their optimized clinical utilization.
Collapse
Affiliation(s)
- Maria Anthi Kouri
- 2nd Department of Radiology, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.A.K.); (E.S.); (G.P.); (V.K.); (K.P.)
- Medical Physics Program, Department of Physics and Applied Physics, Kennedy College of Sciences, University of Massachusetts Lowell, 265 Riverside St., Lowell, MA 01854, USA
| | - Ellas Spyratou
- 2nd Department of Radiology, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.A.K.); (E.S.); (G.P.); (V.K.); (K.P.)
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Iroon Polytechniou 9, 15780 Athens, Greece
| | - Maria-Eleni Kalkou
- Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Georgios Patatoukas
- 2nd Department of Radiology, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.A.K.); (E.S.); (G.P.); (V.K.); (K.P.)
| | - Evangelia Angelopoulou
- 2nd Department of Pathology, School of Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Ioanna Tremi
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.T.); (S.H.); (V.G.G.)
| | - Sophia Havaki
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.T.); (S.H.); (V.G.G.)
| | - Vassilis G. Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.T.); (S.H.); (V.G.G.)
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
- Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4GJ, UK
- Faculty of Health and Medical Sciences, University of Surrey, Surrey GU2 7YH, UK
| | - Vassilis Kouloulias
- 2nd Department of Radiology, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.A.K.); (E.S.); (G.P.); (V.K.); (K.P.)
| | - Kalliopi Platoni
- 2nd Department of Radiology, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.A.K.); (E.S.); (G.P.); (V.K.); (K.P.)
| | - Efstathios P. Efstathopoulos
- 2nd Department of Radiology, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.A.K.); (E.S.); (G.P.); (V.K.); (K.P.)
| |
Collapse
|
5
|
Gerken LRH, Gerdes ME, Pruschy M, Herrmann IK. Prospects of nanoparticle-based radioenhancement for radiotherapy. MATERIALS HORIZONS 2023; 10:4059-4082. [PMID: 37555747 PMCID: PMC10544071 DOI: 10.1039/d3mh00265a] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/02/2023] [Indexed: 08/10/2023]
Abstract
Radiotherapy is a key pillar of solid cancer treatment. Despite a high level of conformal dose deposition, radiotherapy is limited due to co-irradiation of organs at risk and subsequent normal tissue toxicities. Nanotechnology offers an attractive opportunity for increasing the efficacy and safety of cancer radiotherapy. Leveraging the freedom of design and the growing synthetic capabilities of the nanomaterial-community, a variety of engineered nanomaterials have been designed and investigated as radiosensitizers or radioenhancers. While research so far has been primarily focused on gold nanoparticles and other high atomic number materials to increase the absorption cross section of tumor tissue, recent studies are challenging the traditional concept of high-Z nanoparticle radioenhancers and highlight the importance of catalytic activity. This review provides a concise overview on the knowledge of nanoparticle radioenhancement mechanisms and their quantification. It critically discusses potential radioenhancer candidate materials and general design criteria for different radiation therapy modalities, and concludes with research priorities in order to advance the development of nanomaterials, to enhance the efficacy of radiotherapy and to increase at the same time the therapeutic window.
Collapse
Affiliation(s)
- Lukas R H Gerken
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering (IEPE), Department of Mechanical and Process Engineering (D-MAVT), ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland.
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Maren E Gerdes
- Karolinska Institutet, Solnavägen 1, 171 77 Stockholm, Sweden
| | - Martin Pruschy
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Inge K Herrmann
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering (IEPE), Department of Mechanical and Process Engineering (D-MAVT), ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland.
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| |
Collapse
|
6
|
Brown N, Rocchi P, Carmès L, Guthier R, Iyer M, Seban L, Morris T, Bennett S, Lavelle M, Penailillo J, Carrasco R, Williams C, Huynh E, Han Z, Kaza E, Doussineau T, Toprani SM, Qin X, Nagel ZD, Sarosiek KA, Hagège A, Dufort S, Bort G, Lux F, Tillement O, Berbeco R. Tuning ultrasmall theranostic nanoparticles for MRI contrast and radiation dose amplification. Theranostics 2023; 13:4711-4729. [PMID: 37771768 PMCID: PMC10526655 DOI: 10.7150/thno.85663] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/07/2023] [Indexed: 09/30/2023] Open
Abstract
Background: The introduction of magnetic resonance (MR)-guided radiation treatment planning has opened a new space for theranostic nanoparticles to reduce acute toxicity while improving local control. In this work, second-generation AGuIX® nanoparticles (AGuIX-Bi) are synthesized and validated. AGuIX-Bi are shown to maintain MR positive contrast while further amplifying the radiation dose by the replacement of some Gd3+ cations with higher Z Bi3+. These next-generation nanoparticles are based on the AGuIX® platform, which is currently being evaluated in multiple Phase II clinical trials in combination with radiotherapy. Methods: In this clinically scalable methodology, AGuIX® is used as an initial chelation platform to exchange Gd3+ for Bi3+. AGuIX-Bi nanoparticles are synthesized with three ratios of Gd/Bi, each maintaining MR contrast while further amplifying radiation dose relative to Bi3+. Safety, efficacy, and theranostic potential of the nanoparticles were evaluated in vitro and in vivo in a human non-small cell lung cancer model. Results: We demonstrated that increasing Bi3+ in the nanoparticles is associated with more DNA damage and improves in vivo efficacy with a statistically significant delay in tumor growth and 33% complete regression for the largest Bi/Gd ratio tested. The addition of Bi3+ by our synthetic method leads to nanoparticles that present slightly altered pharmacokinetics and lengthening of the period of high tumor accumulation with no observed evidence of toxicity. Conclusions: We confirmed the safety and enhanced efficacy of AGuIX-Bi with radiation therapy at the selected ratio of 30Gd/70Bi. These results provide crucial evidence towards patient translation.
Collapse
Affiliation(s)
- Needa Brown
- Department of Physics, Northeastern University, Boston 02115, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
| | - Paul Rocchi
- NH TherAguix, Meylan 38240, France
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex 69100, France
| | - Léna Carmès
- NH TherAguix, Meylan 38240, France
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex 69100, France
| | - Romy Guthier
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
- Department of Physics and Applied Physics, University of Massachusetts Lowell, Lowell 01854, USA
| | - Meghna Iyer
- Department of Physics, Northeastern University, Boston 02115, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
| | - Léa Seban
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
| | - Toby Morris
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
- Department of Physics and Applied Physics, University of Massachusetts Lowell, Lowell 01854, USA
| | - Stephanie Bennett
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
| | - Michael Lavelle
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
- Department of Physics and Applied Physics, University of Massachusetts Lowell, Lowell 01854, USA
| | - Johany Penailillo
- Department of Pathology, Harvard Medical School and Dana-Farber Cancer Institute, Boston 02115, USA
| | - Ruben Carrasco
- Department of Pathology, Harvard Medical School and Dana-Farber Cancer Institute, Boston 02115, USA
| | - Chris Williams
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
| | - Elizabeth Huynh
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
| | - Zhaohui Han
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
| | - Evangelia Kaza
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
| | | | - Sneh M. Toprani
- John B. Little Center for Radiation Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Xingping Qin
- John B. Little Center for Radiation Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
- Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute/ Harvard Cancer Center, Boston, MA, 02115, USA
| | - Zachary D. Nagel
- John B. Little Center for Radiation Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Kristopher A. Sarosiek
- John B. Little Center for Radiation Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
- Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute/ Harvard Cancer Center, Boston, MA, 02115, USA
| | - Agnès Hagège
- Institut des Sciences Analytiques, Université de Lyon, CNRS, Université Claude Bernard Lyon 1, UMR 5280, 69100, Villeurbanne, France
| | | | - Guillaume Bort
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex 69100, France
| | - François Lux
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex 69100, France
- Institut Universitaire de France (IUF), Paris 75005, France
| | - Olivier Tillement
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex 69100, France
| | - Ross Berbeco
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston 02115, USA
| |
Collapse
|
7
|
Freitas SC, Sanderson D, Caspani S, Magalhães R, Cortés-Llanos B, Granja A, Reis S, Belo JH, Azevedo J, Gómez-Gaviro MV, de Sousa CT. New Frontiers in Colorectal Cancer Treatment Combining Nanotechnology with Photo- and Radiotherapy. Cancers (Basel) 2023; 15:383. [PMID: 36672333 PMCID: PMC9856291 DOI: 10.3390/cancers15020383] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/19/2022] [Accepted: 12/24/2022] [Indexed: 01/11/2023] Open
Abstract
Colorectal cancer is the third most common cancer worldwide. Despite recent advances in the treatment of this pathology, which include a personalized approach using radio- and chemotherapies in combination with advanced surgical techniques, it is imperative to enhance the performance of these treatments and decrease their detrimental side effects on patients' health. Nanomedicine is likely the pathway towards solving this challenge by enhancing both the therapeutic and diagnostic capabilities. In particular, plasmonic nanoparticles show remarkable potential due to their dual therapeutic functionalities as photothermal therapy agents and as radiosensitizers in radiotherapy. Their dual functionality, high biocompatibility, easy functionalization, and targeting capabilities make them potential agents for inducing efficient cancer cell death with minimal side effects. This review aims to identify the main challenges in the diagnosis and treatment of colorectal cancer. The heterogeneous nature of this cancer is also discussed from a single-cell point of view. The most relevant works in photo- and radiotherapy using nanotechnology-based therapies for colorectal cancer are addressed, ranging from in vitro studies (2D and 3D cell cultures) to in vivo studies and clinical trials. Although the results using nanoparticles as a photo- and radiosensitizers in photo- and radiotherapy are promising, preliminary studies showed that the possibility of combining both therapies must be explored to improve the treatment efficiency.
Collapse
Affiliation(s)
- Sara C. Freitas
- IFIMUP-Institute of Physics for Advanced Materials, Nanotechnology and Photonics of University of Porto, LaPMET-Laboratory of Physics for Materials and Emergent Technologies, Departamento de Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Daniel Sanderson
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Doctor Esquerdo 46, 28007 Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, 28911 Leganés, Spain
| | - Sofia Caspani
- IFIMUP-Institute of Physics for Advanced Materials, Nanotechnology and Photonics of University of Porto, LaPMET-Laboratory of Physics for Materials and Emergent Technologies, Departamento de Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Ricardo Magalhães
- IFIMUP-Institute of Physics for Advanced Materials, Nanotechnology and Photonics of University of Porto, LaPMET-Laboratory of Physics for Materials and Emergent Technologies, Departamento de Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | | | - Andreia Granja
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Salette Reis
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - João Horta Belo
- IFIMUP-Institute of Physics for Advanced Materials, Nanotechnology and Photonics of University of Porto, LaPMET-Laboratory of Physics for Materials and Emergent Technologies, Departamento de Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - José Azevedo
- Colorectal Surgery—Champalimaud Foundation, Champalimaud Centre for the Unknown, Avenida Brasília, 1400-038 Lisboa, Portugal
| | - Maria Victoria Gómez-Gaviro
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Doctor Esquerdo 46, 28007 Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, 28911 Leganés, Spain
| | - Célia Tavares de Sousa
- IFIMUP-Institute of Physics for Advanced Materials, Nanotechnology and Photonics of University of Porto, LaPMET-Laboratory of Physics for Materials and Emergent Technologies, Departamento de Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
- Departamento de Física Aplicada, Facultad de Ciencias, Universidad Autonoma de Madrid (UAM), Campus de Cantoblanco, C/ Francisco Tomas y Valiente, 7, 28049 Madrid, Spain
| |
Collapse
|
8
|
Sharma G, Razeghi Kondelaji MH, Sharma GP, Hansen C, Parchur AK, Shafiee S, Jagtap JM, Fish B, Bergom C, Paulson E, Hall WA, Himburg HA, Joshi A. X-ray and MR Contrast Bearing Nanoparticles Enhance the Therapeutic Response of Image-Guided Radiation Therapy for Oral Cancer. Technol Cancer Res Treat 2023; 22:15330338231189593. [PMID: 37469184 PMCID: PMC10363893 DOI: 10.1177/15330338231189593] [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: 10/18/2022] [Revised: 05/09/2023] [Accepted: 06/27/2023] [Indexed: 07/21/2023] Open
Abstract
INTRODUCTION Radiation therapy for head and neck squamous cell carcinoma is constrained by radiotoxicity to normal tissue. We demonstrate 100 nm theranostic nanoparticles for image-guided radiation therapy planning and enhancement in rat head and neck squamous cell carcinoma models. METHODS PEG conjugated theranostic nanoparticles comprising of Au nanorods coated with Gadolinium oxide layers were tested for radiation therapy enhancement in 2D cultures of OSC-19-GFP-luc cells, and orthotopic tongue xenografts in male immunocompromised Salt sensitive or SS rats via both intratumoral and intravenous delivery. The radiation therapy enhancement mechanism was investigated. RESULTS Theranostic nanoparticles demonstrated both X-ray/magnetic resonance contrast in a dose-dependent manner. Magnetic resonance images depicted optimal tumor-to-background uptake at 4 h post injection. Theranostic nanoparticle + Radiation treated rats experienced reduced tumor growth compared to controls, and reduction in lung metastasis. CONCLUSIONS Theranostic nanoparticles enable preprocedure radiotherapy planning, as well as enhance radiation treatment efficacy for head and neck tumors.
Collapse
Affiliation(s)
- Gayatri Sharma
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | | | - Guru P. Sharma
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christopher Hansen
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Abdul K. Parchur
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shayan Shafiee
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Brian Fish
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Carmen Bergom
- Department of Radiation Oncology, Washington University, St Louis, MO, USA
| | - Eric Paulson
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - William A. Hall
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Heather A. Himburg
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amit Joshi
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
9
|
Sun H, Cai H, Xu C, Zhai H, Lux F, Xie Y, Feng L, Du L, Liu Y, Sun X, Wang Q, Song H, He N, Zhang M, Ji K, Wang J, Gu Y, Leduc G, Doussineau T, Wang Y, Liu Q, Tillement O. AGuIX nanoparticles enhance ionizing radiation-induced ferroptosis on tumor cells by targeting the NRF2-GPX4 signaling pathway. J Nanobiotechnology 2022; 20:449. [PMID: 36242003 PMCID: PMC9569109 DOI: 10.1186/s12951-022-01654-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/25/2022] [Indexed: 11/10/2022] Open
Abstract
In the frame of radiotherapy treatment of cancer, radioresistance remains a major issue that still needs solutions to be overcome. To effectively improve the radiosensitivity of tumors and reduce the damage of radiation to neighboring normal tissues, radiosensitizers have been given increasing attention in recent years. As nanoparticles based on the metal element gadolinium, AGuIX nanoparticles have been shown to increase the radiosensitivity of cancers. Although it is a rare nanomaterial that has entered preclinical trials, the unclear biological mechanism hinders its further clinical application. In this study, we demonstrated the effectiveness of AGuIX nanoparticles in the radiosensitization of triple-negative breast cancer. We found that AGuIX nanoparticles increased the level of DNA damage by compromising the homologous recombination repair pathway instead of the non-homologous end joining pathway. Moreover, the results showed that AGuIX nanoparticles induced apoptosis, but the degree of apoptosis ability was very low, which cannot fully explain their strong radiosensitizing effect. Ferroptosis, the other mode of cell death, was also discovered to play a significant role in radiation sensitization, and AGuIX nanoparticles may regulate the anti-ferroptosis system by inhibiting the NRF2-GSH-GPX4 signaling pathway.
Collapse
Affiliation(s)
- Hao Sun
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Hui Cai
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Chang Xu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Hezheng Zhai
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China.,School of Precision Instruments and OPTO-Electronics Engineering, Tianjin University, Tianjin, 300072, China
| | - François Lux
- Institute Light and Mater, UMR5306, Lyon1 University-CNRS, Lyon University, 69100, Villeurbanne, France.,Institut Universitaire de France (IUF), 75231, Paris, France
| | - Yi Xie
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Li Feng
- Department of Medical Ultrasound, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, Jinan, 250014, China
| | - Liqing Du
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Yang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Xiaohui Sun
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Qin Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Huijuan Song
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Ningning He
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Manman Zhang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Kaihua Ji
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Jinhan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Yeqing Gu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Géraldine Leduc
- NH TherAguix S.A.S, 29 chemin du Vieux Chêne, 38240, Meylan, France
| | | | - Yan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China.
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China.
| | - Olivier Tillement
- Institute Light and Mater, UMR5306, Lyon1 University-CNRS, Lyon University, 69100, Villeurbanne, France
| |
Collapse
|
10
|
Clinical Feasibility Study of Gold Nanoparticles as Theragnostic Agents for Precision Radiotherapy. Biomedicines 2022; 10:biomedicines10051214. [PMID: 35625950 PMCID: PMC9139134 DOI: 10.3390/biomedicines10051214] [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: 04/30/2022] [Revised: 05/18/2022] [Accepted: 05/21/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Gold nanoparticles (AuNP) may be useful in precision radiotherapy and disease monitoring as theragnostic agents. In diagnostics, they can be detected by computerized tomography (CT) because of their higher atomic number. AuNP may also improve the treatment results in radiotherapy due to a higher cross-section, locally improving the physically absorbed dose. Methods: Key parameters values involved in the use of AuNP were imposed to be optimal in the clinical scenario. Mass concentration of AuNP as an efficient contrast agent in clinical CT was found and implemented in a Monte Carlo simulation method for dose calculation under different proposed therapeutic beams. The radiosensitization effect was determined in irradiated cells with AuNP. Results: an AuNP concentration was found for a proper contrast level and enhanced therapeutic effect under a beam typically used for image-guided therapy and monitoring. This lower energetic proposed beam showed potential use for treatment monitoring in addition to absorbed dose enhancement and higher radiosensitization at the cellular level. Conclusion: the results obtained show the use of AuNP concentration around 20 mg Au·mL−1 as an efficient tool for diagnosis, treatment planning, and monitoring treatment. Simultaneously, the delivered prescription dose provides a higher radiobiological effect on the cancer cell for achieving precision radiotherapy.
Collapse
|
11
|
Piccolo O, Lincoln JD, Melong N, Orr BC, Fernandez NR, Borsavage J, Berman JN, Robar J, Ha MN. Radiation dose enhancement using gold nanoparticles with a diamond linear accelerator target: a multiple cell type analysis. Sci Rep 2022; 12:1559. [PMID: 35091583 PMCID: PMC8799734 DOI: 10.1038/s41598-022-05339-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 01/04/2022] [Indexed: 01/14/2023] Open
Abstract
Radiotherapy (RT) is an effective cancer treatment modality, but standard RT often causes collateral damage to nearby healthy tissues. To increase therapeutic ratio, radiosensitization via gold nanoparticles (GNPs) has been shown to be effective. One challenge is that megavoltage beams generated by clinical linear accelerators are poor initiators of the photoelectric effect. Previous computer models predicted that a diamond target beam (DTB) will yield 400% more low-energy photons, increasing the probability of interacting with GNPs to enhance the radiation dose by 7.7-fold in the GNP vicinity. After testing DTB radiation coupled with GNPs in multiple cell types, we demonstrate decreased head-and-neck cancer (HNC) cell viability in vitro and enhanced cell-killing in zebrafish xenografts compared to standard RT. HNC cell lines also displayed increased double-stranded DNA breaks with DTB irradiation in the presence of GNPs. This study presents preclinical responses to GNP-enhanced radiotherapy with the novel DTB, providing the first functional data to support the theoretical evidence for radiosensitization via GNPs in this context, and highlighting the potential of this approach to optimize the efficacy of RT in anatomically difficult-to-treat tumors.
Collapse
Affiliation(s)
- Olivia Piccolo
- Department of Biology, Dalhousie University, Halifax, NS, Canada
- Department of Pediatrics, IWK Health Centre/Dalhousie University, Halifax, NS, Canada
| | - John D Lincoln
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, NS, Canada
| | - Nicole Melong
- Children's Hospital of Eastern Ontario Research Institute/Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - Benno C Orr
- Department of Pediatrics, IWK Health Centre/Dalhousie University, Halifax, NS, Canada
| | - Nicholas R Fernandez
- Department of Pediatrics, IWK Health Centre/Dalhousie University, Halifax, NS, Canada
| | - Jennifer Borsavage
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, NS, Canada
| | - Jason N Berman
- Department of Pediatrics, IWK Health Centre/Dalhousie University, Halifax, NS, Canada
- Children's Hospital of Eastern Ontario Research Institute/Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - James Robar
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, NS, Canada
- Department of Radiation Oncology, Dalhousie University, Halifax, NS, Canada
| | - Michael N Ha
- Department of Radiation Oncology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
12
|
Thomas E, Mathieu C, Moreno‐Gaona P, Mittelheisser V, Lux F, Tillement O, Pivot X, Ghoroghchian PP, Detappe A. Anti-BCMA Immuno-NanoPET Radiotracers for Improved Detection of Multiple Myeloma. Adv Healthc Mater 2022; 11:e2101565. [PMID: 34710281 PMCID: PMC11469021 DOI: 10.1002/adhm.202101565] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/09/2021] [Indexed: 11/07/2022]
Abstract
Current clinical imaging modalities for the sensitive and specific detection of multiple myeloma (MM) rely on nonspecific imaging contrast agents based on gadolinium chelates for magnetic resonance imaging (MRI) or for 18 F-FDG-directed and combined positron emission tomography (PET) and computed tomography (CT) scans. These tracers are not, however, able to detect minute plasma cell populations in the tumor niche, leading to false negative results. Here, a novel PET-based anti-BCMA nanoplatform labeled with 64 Cu is developed to improve the monitoring of these cells in both the spine and femur and to compare its sensitivity and specificity to more conventional immunoPET (64 Cu labeled anti-BCMA antibody) and passively targeted PET radiotracers (64 CuCl2 and 18 F-FDG). This proof-of-concept preclinical study confirmed that by conjugating up to four times more radioisotopes per antibody with the immuno-nanoPET platform, an improvement in the sensitivity and in the specificity of PET to detect tumor cells in an orthotopic model of MM is observed when compared to the traditional immunoPET approach. It is anticipated that when combined with tumor biopsy, this immuno-nanoPET platform may improve the management of patients with MM.
Collapse
Affiliation(s)
- Eloise Thomas
- LAGEPP Université Claude Bernard Lyon 1CNRS UMR5007VilleurbanneFrance
| | - Clélia Mathieu
- Université Paris‐SaclayCNRS UMR 8612Institut Galien Paris‐SaclayFrance
| | | | | | - François Lux
- Institut Lumière‐MatièreUniversité Claude Bernard Lyon 1CNRS UMR5306VilleurbanneFrance
- Institut Universitaire de France (IUF)ParisFrance
| | - Olivier Tillement
- Institut Lumière‐MatièreUniversité Claude Bernard Lyon 1CNRS UMR5306VilleurbanneFrance
| | - Xavier Pivot
- Institut de Cancérologie Strasbourg Europe (ICANS)StrasbourgFrance
| | - Paiman Peter Ghoroghchian
- David H Koch Institute for Integrative Cancer ResearchMITCambridgeMAUSA
- Dana Farber Cancer InstituteBostonMAUSA
| | - Alexandre Detappe
- Institut de Cancérologie Strasbourg Europe (ICANS)StrasbourgFrance
- Strasbourg Drug Discovery and Development Institute (IMS)StrasbourgFrance
| |
Collapse
|
13
|
Tremi I, Spyratou E, Souli M, Efstathopoulos EP, Makropoulou M, Georgakilas AG, Sihver L. Requirements for Designing an Effective Metallic Nanoparticle (NP)-Boosted Radiation Therapy (RT). Cancers (Basel) 2021; 13:cancers13133185. [PMID: 34202342 PMCID: PMC8269428 DOI: 10.3390/cancers13133185] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Recent advances in nanotechnology gave rise to trials with various types of metallic nanoparticles (NPs) to enhance the radiosensitization of cancer cells while reducing or maintaining the normal tissue complication probability during radiation therapy. This work reviews the physical and chemical mechanisms leading to the enhancement of ionizing radiation’s detrimental effects on cells and tissues, as well as the plethora of experimental procedures to study these effects of the so-called “NPs’ radiosensitization”. The paper presents the need to a better understanding of all the phases of actions before applying metallic-based NPs in clinical practice to improve the effect of IR therapy. More physical and biological experiments especially in vivo must be performed and simulation Monte Carlo or mathematical codes based on more accurate models for all phases must be developed. Abstract Many different tumor-targeted strategies are under development worldwide to limit the side effects and improve the effectiveness of cancer therapies. One promising method is to enhance the radiosensitization of the cancer cells while reducing or maintaining the normal tissue complication probability during radiation therapy using metallic nanoparticles (NPs). Radiotherapy with MV photons is more commonly available and applied in cancer clinics than high LET particle radiotherapy, so the addition of high-Z NPs has the potential to further increase the efficacy of photon radiotherapy in terms of NP radiosensitization. Generally, when using X-rays, mainly the inner electron shells are ionized, which creates cascades of both low and high energy Auger electrons. When using high LET particles, mainly the outer shells are ionized, which give electrons with lower energies than when using X-rays. The amount of the produced low energy electrons is higher when exposing NPs to heavy charged particles than when exposing them to X-rays. Since ions traverse the material along tracks, and therefore give rise to a much more inhomogeneous dose distributions than X-rays, there might be a need to introduce a higher number of NPs when using ions compared to when using X-rays to create enough primary and secondary electrons to get the desired dose escalations. This raises the questions of toxicity. This paper provides a review of the fundamental processes controlling the outcome of metallic NP-boosted photon beam and ion beam radiation therapy and presents some experimental procedures to study the biological effects of NPs’ radiosensitization. The overview shows the need for more systematic studies of the behavior of NPs when exposed to different kinds of ionizing radiation before applying metallic-based NPs in clinical practice to improve the effect of IR therapy.
Collapse
Affiliation(s)
- Ioanna Tremi
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
| | - Ellas Spyratou
- 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11517 Athens, Greece; (E.S.); (E.P.E.)
| | - Maria Souli
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
- Atominstitut, Technische Universität Wien, Stadionallee 2, 1020 Vienna, Austria
| | - Efstathios P. Efstathopoulos
- 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11517 Athens, Greece; (E.S.); (E.P.E.)
| | - Mersini Makropoulou
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
| | - Alexandros G. Georgakilas
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
- Correspondence: (A.G.G.); (L.S.)
| | - Lembit Sihver
- Atominstitut, Technische Universität Wien, Stadionallee 2, 1020 Vienna, Austria
- Department of Physics, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
- Correspondence: (A.G.G.); (L.S.)
| |
Collapse
|
14
|
Schuemann J, Bagley AF, Berbeco R, Bromma K, Butterworth KT, Byrne HL, Chithrani BD, Cho SH, Cook JR, Favaudon V, Gholami YH, Gargioni E, Hainfeld JF, Hespeels F, Heuskin AC, Ibeh UM, Kuncic Z, Kunjachan S, Lacombe S, Lucas S, Lux F, McMahon S, Nevozhay D, Ngwa W, Payne JD, Penninckx S, Porcel E, Prise KM, Rabus H, Ridwan SM, Rudek B, Sanche L, Singh B, Smilowitz HM, Sokolov KV, Sridhar S, Stanishevskiy Y, Sung W, Tillement O, Virani N, Yantasee W, Krishnan S. Roadmap for metal nanoparticles in radiation therapy: current status, translational challenges, and future directions. Phys Med Biol 2020; 65:21RM02. [PMID: 32380492 DOI: 10.1088/1361-6560/ab9159] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This roadmap outlines the potential roles of metallic nanoparticles (MNPs) in the field of radiation therapy. MNPs made up of a wide range of materials (from Titanium, Z = 22, to Bismuth, Z = 83) and a similarly wide spectrum of potential clinical applications, including diagnostic, therapeutic (radiation dose enhancers, hyperthermia inducers, drug delivery vehicles, vaccine adjuvants, photosensitizers, enhancers of immunotherapy) and theranostic (combining both diagnostic and therapeutic), are being fabricated and evaluated. This roadmap covers contributions from experts in these topics summarizing their view of the current status and challenges, as well as expected advancements in technology to address these challenges.
Collapse
Affiliation(s)
- Jan Schuemann
- Department of Radiation Oncology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Mittelheisser V, Banerjee M, Pivot X, Charbonnière LJ, Goetz J, Detappe A. Leveraging Immunotherapy with Nanomedicine. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Vincent Mittelheisser
- Centre Paul Strauss Strasbourg 67000 France
- INSERM UMR_S1109 Strasbourg 67000 France
- Université de Strasbourg Strasbourg 67000 France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg 67000 France
| | - Mainak Banerjee
- Centre Paul Strauss Strasbourg 67000 France
- Institut de Cancérologie Strasbourg Europe Strasbourg 67000 France
- Institut Pluridisciplinaire Hubert Curien CNRS UMR‐7178 Strasbourg 67087 France
| | - Xavier Pivot
- Institut de Cancérologie Strasbourg Europe Strasbourg 67000 France
| | - Loïc J. Charbonnière
- Université de Strasbourg Strasbourg 67000 France
- Institut Pluridisciplinaire Hubert Curien CNRS UMR‐7178 Strasbourg 67087 France
| | - Jacky Goetz
- INSERM UMR_S1109 Strasbourg 67000 France
- Université de Strasbourg Strasbourg 67000 France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg 67000 France
| | - Alexandre Detappe
- Centre Paul Strauss Strasbourg 67000 France
- Université de Strasbourg Strasbourg 67000 France
- Institut de Cancérologie Strasbourg Europe Strasbourg 67000 France
- Institut Pluridisciplinaire Hubert Curien CNRS UMR‐7178 Strasbourg 67087 France
| |
Collapse
|
16
|
Virani NA, Kelada OJ, Kunjachan S, Detappe A, Kwon J, Hayashi J, Vazquez-Pagan A, Biancur DE, Ireland T, Kumar R, Sridhar S, Makrigiorgos GM, Berbeco RI. Noninvasive imaging of tumor hypoxia after nanoparticle-mediated tumor vascular disruption. PLoS One 2020; 15:e0236245. [PMID: 32706818 PMCID: PMC7380644 DOI: 10.1371/journal.pone.0236245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/01/2020] [Indexed: 01/09/2023] Open
Abstract
We have previously demonstrated that endothelial targeting of gold nanoparticles followed by external beam irradiation can cause specific tumor vascular disruption in mouse models of cancer. The induced vascular damage may lead to changes in tumor physiology, including tumor hypoxia, thereby compromising future therapeutic interventions. In this study, we investigate the dynamic changes in tumor hypoxia mediated by targeted gold nanoparticles and clinical radiation therapy (RT). By using noninvasive whole-body fluorescence imaging, tumor hypoxia was measured at baseline, on day 2 and day 13, post-tumor vascular disruption. A 2.5-fold increase (P<0.05) in tumor hypoxia was measured two days after combined therapy, resolving by day 13. In addition, the combination of vascular-targeted gold nanoparticles and radiation therapy resulted in a significant (P<0.05) suppression of tumor growth. This is the first study to demonstrate the tumor hypoxic physiological response and recovery after delivery of vascular-targeted gold nanoparticles followed by clinical radiation therapy in a human non-small cell lung cancer athymic Foxn1nu mouse model.
Collapse
Affiliation(s)
- Needa A. Virani
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Olivia J. Kelada
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sijumon Kunjachan
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alexandre Detappe
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston Massachusetts, United States of America
| | - Jihun Kwon
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Radiation Oncology, Hokkaido University, Sapporo, Japan
| | - Jennifer Hayashi
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Nanomedicine Innovation Center and Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - Ana Vazquez-Pagan
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Nanomedicine Innovation Center and Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - Douglas E. Biancur
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston Massachusetts, United States of America
| | - Thomas Ireland
- LA-ICP-MS and ICP-ES Laboratories, Boston University, Boston, Massachusetts, United States of America
| | - Rajiv Kumar
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Nanomedicine Innovation Center and Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - Srinivas Sridhar
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Nanomedicine Innovation Center and Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - G. Mike Makrigiorgos
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ross I. Berbeco
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
17
|
Detappe A, Mathieu C, Jin C, Agius MP, Diringer MC, Tran VL, Pivot X, Lux F, Tillement O, Kufe D, Ghoroghchian PP. Anti-MUC1-C Antibody-Conjugated Nanoparticles Potentiate the Efficacy of Fractionated Radiation Therapy. Int J Radiat Oncol Biol Phys 2020; 108:1380-1389. [PMID: 32634545 DOI: 10.1016/j.ijrobp.2020.06.069] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE Heavy-metal chelators and inorganic nanoparticles (NPs) have been examined as potential radioenhancers to increase the efficacy of external beam radiation therapy for various cancers. Most of these agents have, unfortunately, displayed relatively poor pharmacokinetic properties, which limit the percentage of injected dose (%ID/g) that localizes to tumors and which shorten the window for effective radiation enhancement due to rapid tumor washout. METHODS AND MATERIALS To address these challenges, we sought to conjugate gadolinium-based ultrasmall (<5 nm) NPs to an antibody directed against the oncogenic MUC1-C subunit that is overexpressed on the surface of many different human cancer types. The binding of the anti-MUC1-C antibody 3D1 to MUC1-C on the surface of a cancer cell is associated with its internalization and, thereby, to effective intracellular delivery of the antibody-associated payload, promoting its effective tumor retention. As such, we examined whether systemically administered anti-MUC1-C antibody-conjugated, gadolinium-based NPs (anti-MUC1-C/NPs) could accumulate within cell-line xenograft models of MUC1-C-expressing (H460) lung and (E0771) breast cancers to improve the efficacy of radiation therapy (XRT). RESULTS The %ID/g of anti-MUC1-C/NPs that accumulated within tumors was found to be similar to that of their unconjugated counterparts (6.6 ± 1.4 vs 5.9 ± 1.7 %ID/g, respectively). Importantly, the anti-MUC1-C/NPs demonstrated prolonged retention in in vivo tumor microenvironments; as a result, the radiation boost was maintained during the course of fractionated therapy (3 × 5.2 Gy). We found that by administering anti-MUC1-C/NPs with XRT, it was possible to significantly augment tumor growth inhibition and to prolong the animals' overall survival (46.2 ± 3.1 days) compared with the administration of control NPs with XRT (31.1 ± 2.4 days) or with XRT alone (27.3 ± 1.6 days; P < .01, log-rank). CONCLUSIONS These findings suggest that anti-MUC1-C/NPs could be used to enhance the effectiveness of radiation therapy and potentially to improve clinical outcomes.
Collapse
Affiliation(s)
- Alexandre Detappe
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts; Centre Paul Strauss, Strasbourg, France.
| | - Clélia Mathieu
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | - Caining Jin
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | - Michael P Agius
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | | | - Vu-Long Tran
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex, France
| | - Xavier Pivot
- Institut du Cancer Strasbourg, Strasbourg, France
| | - Francois Lux
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex, France; Institut Universitaire de France, Paris, France
| | - Olivier Tillement
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex, France
| | - Donald Kufe
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | - Peter P Ghoroghchian
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
18
|
Nakayama M, Akasaka H, Geso M, Morita K, Yada R, Uehara K, Sasaki R. Utilisation of the chemiluminescence method to measure the radiation dose enhancement caused by gold nanoparticles: A phantom-based study. RADIAT MEAS 2020. [DOI: 10.1016/j.radmeas.2020.106317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
19
|
Detappe A, Reidy M, Yu Y, Mathieu C, Nguyen HVT, Coroller TP, Lam F, Jarolim P, Harvey P, Protti A, Nguyen QD, Johnson JA, Cremillieux Y, Tillement O, Ghobrial IM, Ghoroghchian PP. Antibody-targeting of ultra-small nanoparticles enhances imaging sensitivity and enables longitudinal tracking of multiple myeloma. NANOSCALE 2019; 11:20485-20496. [PMID: 31650133 DOI: 10.1039/c9nr06512a] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Monitoring malignant progression and disease recurrence post-therapy are central challenges to improving the outcomes of patients with multiple myeloma (MM). Whereas current detection methods that rely upon bone marrow examination allow for precise monitoring of minimal residual disease and can help to elucidate clonal evolution, they do not take into account the spatial heterogeneity of the tumor microenvironment. As such, they are uninformative as to the localization of malignant plasma cells and may lead to false negative results. With respect to the latter challenge, clinically-available imaging agents are neither sufficiently sensitive nor specific enough to detect minute plasma cell populations. Here, we sought to explore methods by which to improve detection of MM cells within their natural bone marrow environment, using whole-animal magnetic resonance imaging to longitudinally monitor early-stage disease as well as to enhance tumor detection after systemic therapy. We conducted a proof-of-concept study to demonstrate that ultra-small (<5 nm) gadolinium-containing nanoparticles bound to full-length antibodies against the B-cell maturation antigen (BCMA) exhibit rapid tumor uptake followed by renal clearance, improving the signal-to-noise ratio for MM detection beyond levels that are currently afforded by other FDA-approved clinical imaging modalities. We anticipate that when combined with bone marrow or blood biopsy, such imaging constructs could help to augment the effective management of patients with MM.
Collapse
Affiliation(s)
- Alexandre Detappe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. and Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA. and Centre Paul Strauss, 3 rue de la porte de l'hôpital, 67000 Strasbourg, France
| | - Mairead Reidy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. and Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Yingjie Yu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| | - Clelia Mathieu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. and Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Hung V-T Nguyen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Thibaud P Coroller
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA and Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Fred Lam
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA. and Clinical Scholar Program, Division of Neurosurgery, McMaster University, 237 Barton St East, Hamilton General Hospital, Hamilton ON, L8L 2X2, Canada
| | - Petr Jarolim
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Peter Harvey
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Andrea Protti
- Lurie Family Imaging Center, Department of Radiology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Quang-De Nguyen
- Lurie Family Imaging Center, Department of Radiology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Jeremiah A Johnson
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yannick Cremillieux
- Institut des Sciences Moléculaires, Université de Bordeaux, UMR CNRS 5255, 33076 Bordeaux, France
| | - Olivier Tillement
- Institut Lumière Matière, UMR 5306 Université Lyon1-CNRS, Université de Lyon, 69622 Villeurbanne Cedex, France
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. and Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - P Peter Ghoroghchian
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. and Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| |
Collapse
|
20
|
Hashemi S, Aghamiri MR, Kahani M, Jaberi R. Investigation of gold nanoparticle effects in brachytherapy by an electron emitter ophthalmic plaque. Int J Nanomedicine 2019; 14:4157-4165. [PMID: 31239674 PMCID: PMC6560204 DOI: 10.2147/ijn.s205814] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/04/2019] [Indexed: 12/17/2022] Open
Abstract
Background: During decades, all improvements and developments in radiation therapy technologies have been focused on its main goal: maximize the dose in the tumor and minimize it in surrounding normal tissues. Recently, scientists have some approaches to nanoparticles, especially gold nanoparticles (GNPs), for dose localization. Purpose: Herein, the effect of GNPs in combination with electron brachytherapy in a model of eye tumor has been investigated. Materials and methods: Monte Carlo simulation was utilized and a complete anatomical model of the eye, a tumor with 5 mm thick, and a type of Ruthenium-106 beta emitter ophthalmic plaque were simulated. Simulation results have been validated by a Plexiglas eye phantom and film dosimetry, experimentally. Results: The results showed using GNPs causes the dose amplification in 2 mm from the plaque surface which the higher concentration has the higher enhancement. At more distances, Dose Enhancement Factors (DEFs) have the negative amounts, so that total delivered dose to the tumor has decreased with increasing of Au concentrations and the dose of organ at risk like sclera has increased. Conclusion: Therefore, using of GNPs along with a 106Ru/106Rh ocular plaque, as an electron emitter source, is a good choice only for superficial lesions, and it is not recommended for deeper tumors due to the parameters of radiation treatment and delivered dose to the tissues.
Collapse
Affiliation(s)
- S Hashemi
- Radiation Medicine Engineering Department, Shahid Beheshti University, Tehran, Iran
| | - MR Aghamiri
- Radiation Medicine Engineering Department, Shahid Beheshti University, Tehran, Iran
| | - M Kahani
- Radiation Medicine Engineering Department, Shahid Beheshti University, Tehran, Iran
| | - R Jaberi
- Cancer Institute, Imam Khomeini Hospital, Tehran, Iran
| |
Collapse
|
21
|
Nguyen HVT, Detappe A, Gallagher NM, Zhang H, Harvey P, Yan C, Mathieu C, Golder MR, Jiang Y, Ottaviani MF, Jasanoff A, Rajca A, Ghobrial I, Ghoroghchian PP, Johnson JA. Triply Loaded Nitroxide Brush-Arm Star Polymers Enable Metal-Free Millimetric Tumor Detection by Magnetic Resonance Imaging. ACS NANO 2018; 12:11343-11354. [PMID: 30387988 PMCID: PMC6320246 DOI: 10.1021/acsnano.8b06160] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Nitroxides occupy a privileged position among plausible metal-free magnetic resonance imaging (MRI) contrast agents (CAs) due to their inherently low-toxicity profiles; nevertheless, their translational development has been hindered by a lack of appropriate contrast sensitivity. Nanostructured materials with high nitroxide densities, where each individual nitroxide within a macromolecular construct contributes to the image contrast, could address this limitation, but the synthesis of such materials remains challenging. Here, we report a modular and scalable synthetic approach to nitroxide-based brush-arm star polymer (BASP) organic radical CAs (ORCAs) with high nitroxide loadings. The optimized ∼30 nm diameter "BASP-ORCA3" displays outstanding T2 sensitivity with a very high molecular transverse relaxivity ( r2 > 1000 mM-1 s-1). BASP-ORCA3 further exhibits excellent stability in vivo, no acute toxicity, and highly desirable pharmacokinetic and biodistribution profiles for longitudinal detection of tumors by MRI. When injected intravenously into mice bearing subcutaneous plasmacytomas, BASP-ORCA3 affords distinct in vivo visualization of tumors on translationally relevant time scales. Leveraging its high sensitivity, BASP-ORCA3 enables efficient mapping of tumor necrosis, which is an important biomarker to predict therapeutic outcomes. Moreover, BASP-ORCA3 allows for detection of millimetric tumor implants in a disseminated murine model of advanced-stage human ovarian cancer that possess genetic, histological, and vascular characteristics that are similar to those seen in patients. This work establishes BASP-ORCA3 as a promising metal-free spin contrast agent for MRI.
Collapse
Affiliation(s)
- Hung V.-T. Nguyen
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts 02115, United States
| | - Alexandre Detappe
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts 02115, United States
| | - Nolan M. Gallagher
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Hui Zhang
- Department of Chemistry, University of Nebraska, Lincoln, Nebraska 68588, United States
| | - Peter Harvey
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Changcun Yan
- Department of Chemistry, University of Nebraska, Lincoln, Nebraska 68588, United States
| | - Clelia Mathieu
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
| | - Matthew R. Golder
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Yivan Jiang
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | | | - Alan Jasanoff
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Nuclear Science and Engineering Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Andrzej Rajca
- Department of Chemistry, University of Nebraska, Lincoln, Nebraska 68588, United States
| | - Irene Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts 02115, United States
| | - P. Peter Ghoroghchian
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts 02115, United States
| | - Jeremiah A. Johnson
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
22
|
Kohout C, Santi C, Polito L. Anisotropic Gold Nanoparticles in Biomedical Applications. Int J Mol Sci 2018; 19:E3385. [PMID: 30380664 PMCID: PMC6274885 DOI: 10.3390/ijms19113385] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/22/2018] [Accepted: 10/24/2018] [Indexed: 02/07/2023] Open
Abstract
Gold nanoparticles (AuNPs) play a crucial role in the development of nanomedicine, principally due to their unique photophysical properties and high biocompatibility. The possibility to tune and customize the localized surface plasmon resonance (LSPR) toward near-infrared region by modulating the AuNP shape is one of the reasons for the huge widespread use of AuNPs. The controlled synthesis of no-symmetrical nanoparticles, named anisotropic, is an exciting goal achieved by the scientific community which explains the exponential increase of the number of publications related to the synthesis and use of such type of AuNPs. Even with such steps forward and the AuNP translation in clinic being done, some key issues are still remain and they are related to a reliable and scalable production, a full characterization, and to the development of nanotoxicology studies on the long run. In this review we highlight the very recent advances on the synthesis of the main classes of anisotropic AuNPs (nanorods, nanourchins and nanocages) and their use in the biomedical fields, in terms of diagnosis and therapeutics.
Collapse
Affiliation(s)
- Claudia Kohout
- Department of Chemistry, University of Milan, via C. Golgi 19, 20131 Milan, Italy.
| | - Cristina Santi
- Department of Chemistry, University of Milan, via C. Golgi 19, 20131 Milan, Italy.
| | - Laura Polito
- ISTM-CNR, Nanotechnology Lab., via G. Fantoli 16/15, 20138 Milan, Italy.
| |
Collapse
|
23
|
Lux F, Tran VL, Thomas E, Dufort S, Rossetti F, Martini M, Truillet C, Doussineau T, Bort G, Denat F, Boschetti F, Angelovski G, Detappe A, Crémillieux Y, Mignet N, Doan BT, Larrat B, Meriaux S, Barbier E, Roux S, Fries P, Müller A, Abadjian MC, Anderson C, Canet-Soulas E, Bouziotis P, Barberi-Heyob M, Frochot C, Verry C, Balosso J, Evans M, Sidi-Boumedine J, Janier M, Butterworth K, McMahon S, Prise K, Aloy MT, Ardail D, Rodriguez-Lafrasse C, Porcel E, Lacombe S, Berbeco R, Allouch A, Perfettini JL, Chargari C, Deutsch E, Le Duc G, Tillement O. AGuIX ® from bench to bedside-Transfer of an ultrasmall theranostic gadolinium-based nanoparticle to clinical medicine. Br J Radiol 2018; 92:20180365. [PMID: 30226413 PMCID: PMC6435081 DOI: 10.1259/bjr.20180365] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
AGuIX® are sub-5 nm nanoparticles made of a polysiloxane matrix and gadolinium chelates. This nanoparticle has been recently accepted in clinical trials in association with radiotherapy. This review will summarize the principal preclinical results that have led to first in man administration. No evidence of toxicity has been observed during regulatory toxicity tests on two animal species (rodents and monkeys). Biodistributions on different animal models have shown passive uptake in tumours due to enhanced permeability and retention effect combined with renal elimination of the nanoparticles after intravenous administration. High radiosensitizing effect has been observed with different types of irradiations in vitro and in vivo on a large number of cancer types (brain, lung, melanoma, head and neck…). The review concludes with the second generation of AGuIX nanoparticles and the first preliminary results on human.
Collapse
Affiliation(s)
- François Lux
- NH TherAguix SAS, Villeurbanne, France.,Univ Lyon Université Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, LYON, France
| | - Vu Long Tran
- Univ Lyon Université Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, LYON, France.,Nano-H SAS, Saint-Quentin-Fallavier, France
| | - Eloïse Thomas
- Univ Lyon Université Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, LYON, France
| | | | - Fabien Rossetti
- Univ Lyon Université Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, LYON, France
| | - Matteo Martini
- Univ Lyon Université Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, LYON, France
| | - Charles Truillet
- Imagerie Moléculaire In Vivo, Inserm, CEA, CNRS, Univ Paris Sud, Université Paris Saclay - Service Hospitalier Frédéric Joliot, Orsay, France
| | | | - Guillaume Bort
- Univ Lyon Université Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, LYON, France
| | - Franck Denat
- Institut de Chimie Moléculaire, Université de Bourgogne, Dijon, France
| | | | - Goran Angelovski
- MR Neuroimaging Agents, Max Planck Institute for Biological Cybernetics, Tuebingen, Germany
| | - Alexandre Detappe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, MA, USA
| | - Yannick Crémillieux
- Centre de Résonance Magnétique des Systèmes Biologiques, CNRS UMR, Université Bordeaux, Bordeaux, France
| | - Nathalie Mignet
- Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France.,CNRS, UTCBS UMR , Paris, France.,Université Paris Descartes Sorbonne-Paris-Cité, Paris, France.,INSERM, UTCBS U 1022, Paris, France
| | - Bich-Thuy Doan
- Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France.,CNRS, UTCBS UMR , Paris, France.,Université Paris Descartes Sorbonne-Paris-Cité, Paris, France.,INSERM, UTCBS U 1022, Paris, France
| | - Benoit Larrat
- NeuroSpin, CEA Saclay, Gif-sur-Yvette, France.,Université Paris-Saclay, Orsay, France
| | - Sébastien Meriaux
- NeuroSpin, CEA Saclay, Gif-sur-Yvette, France.,Université Paris-Saclay, Orsay, France
| | - Emmanuel Barbier
- INSERM, Univ. Grenoble Alpes, Grenoble Institut des Neurosciences , Grenoble, France
| | - Stéphane Roux
- Institut UTINAM, UMR CNRS 6213-Université de Bourgogne Franche-Comté, Besançon, France
| | - Peter Fries
- Clinic of Diagnostic and Interventional Radiology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Andreas Müller
- Clinic of Diagnostic and Interventional Radiology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Marie-Caline Abadjian
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Carolyn Anderson
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Emmanuelle Canet-Soulas
- Univ Lyon, CarMeN Laboratory Institut National de la Santé et de la Recherche Médicale U1060,INRA U1397, Université Lyon 1, INSA Lyon, Oullins, France
| | - Penelope Bouziotis
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center forScientific Research "Demokritos", Aghia Paraskevi, Athens, Greece
| | | | - Céline Frochot
- Laboratoire Réactions et Génie des Procédés, UMR, Université de Lorraine-CNRS, Nancy, France
| | - Camille Verry
- Radiotherapy department, CHU de Grenoble, Grenoble cedex 9, France
| | - Jacques Balosso
- Radiotherapy department, CHU de Grenoble, Grenoble cedex 9, France
| | - Michael Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, USA
| | | | - Marc Janier
- UNIV Lyon - Université Claude Bernard Lyon 1, Villeurbanne, France.,Hospices Civils de Lyon, plateforme Imthernat, Hôpital Edouard Herriot, Lyon, France
| | - Karl Butterworth
- Centre for Cancer Research and Cell Biology Queen's University Belfast,, Belfast BT9 7AE, UK
| | - Stephen McMahon
- Centre for Cancer Research and Cell Biology Queen's University Belfast,, Belfast BT9 7AE, UK
| | - Kevin Prise
- Centre for Cancer Research and Cell Biology Queen's University Belfast,, Belfast BT9 7AE, UK
| | - Marie-Thérèse Aloy
- IPNL, PRISME, Laboratoire de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Université Lyon 1; Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Dominique Ardail
- IPNL, PRISME, Laboratoire de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Université Lyon 1; Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Claire Rodriguez-Lafrasse
- IPNL, PRISME, Laboratoire de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Université Lyon 1; Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Erika Porcel
- ISMO UMR, Université Paris Saclay, Université Paris Sud, CNRS, Orsay cedex, France
| | - Sandrine Lacombe
- ISMO UMR, Université Paris Saclay, Université Paris Sud, CNRS, Orsay cedex, France
| | - Ross Berbeco
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, MA, USA
| | - Awatef Allouch
- Cell death and Aging team, Gustave Roussy, rue Edouard Vaillant, Villejuif, France.,Laboratory of Molecular Radiotherapy INSERM, Gustave Roussy, rue Edouard Vaillant, Villejuif, France.,Gustave Roussy, rue Edouard Vaillant, Villejuif, France.,Université Paris Sud - Paris , rue Edouard Vaillant, Villejuif, France
| | - Jean-Luc Perfettini
- Cell death and Aging team, Gustave Roussy, rue Edouard Vaillant, Villejuif, France.,Laboratory of Molecular Radiotherapy INSERM, Gustave Roussy, rue Edouard Vaillant, Villejuif, France.,Gustave Roussy, rue Edouard Vaillant, Villejuif, France.,Université Paris Sud - Paris , rue Edouard Vaillant, Villejuif, France
| | - Cyrus Chargari
- French Military Health Academy, Ecole du Val-de-Grâce, Paris, France.,Institut de Recherche Biomédicale des Armées, Bretigny-sur-Orge, France.,Radiotherapy Department, Gustave Roussy, Villejuif, France.,Brachytherapy Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Eric Deutsch
- Radiotherapy Department, Gustave Roussy, Villejuif, France.,Brachytherapy Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Olivier Tillement
- NH TherAguix SAS, Villeurbanne, France.,Univ Lyon Université Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, LYON, France
| |
Collapse
|
24
|
Zhu S, Gu Z, Zhao Y. Harnessing Tumor Microenvironment for Nanoparticle-Mediated Radiotherapy. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800050] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 China
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 China
- College of Materials Science and Optoelectronic Technology; University of Chinese Academy of Sciences; Beijing 100049 China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 China
- CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology of China; Chinese Academy of Sciences; Beijing 100190 China
- College of Materials Science and Optoelectronic Technology; University of Chinese Academy of Sciences; Beijing 100049 China
| |
Collapse
|
25
|
Cheng K, Sano M, Jenkins CH, Zhang G, Vernekohl D, Zhao W, Wei C, Zhang Y, Zhang Z, Liu Y, Cheng Z, Xing L. Synergistically Enhancing the Therapeutic Effect of Radiation Therapy with Radiation Activatable and Reactive Oxygen Species-Releasing Nanostructures. ACS NANO 2018; 12:4946-4958. [PMID: 29689158 DOI: 10.1021/acsnano.8b02038] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Nanoparticle-based radio-sensitizers can amplify the effects of radiation therapy on tumor tissue even at relatively low concentrations while reducing the potential side effects to healthy surrounding tissues. In this study, we investigated a hybrid anisotropic nanostructure, composed of gold (Au) and titanium dioxide (TiO2), as a radio-sensitizer for radiation therapy of triple-negative breast cancer (TNBC). In contrast to other gold-based radio sensitizers, dumbbell-like Au-TiO2 nanoparticles (DATs) show a synergistic therapeutic effect on radiation therapy, mainly because of strong asymmetric electric coupling between the high atomic number metals and dielectric oxides at their interfaces. The generation of secondary electrons and reactive oxygen species (ROS) from DATs triggered by X-ray irradiation can significantly enhance the radiation effect. After endocytosed by cancer cells, DATs can generate a large amount of ROS under X-ray irradiation, eventually inducing cancer cell apoptosis. Significant tumor growth suppression and overall improvement in survival rate in a TNBC tumor model have been successfully demonstrated under DAT uptake for a radio-sensitized radiation therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Chenxi Wei
- Stanford Synchrotron Radiation Light Source (SSRL) , SLAC National Accelerator Laboratory , Menlo Park , California 94025 , United States
| | - Yan Zhang
- Stanford Synchrotron Radiation Light Source (SSRL) , SLAC National Accelerator Laboratory , Menlo Park , California 94025 , United States
| | | | - Yijin Liu
- Stanford Synchrotron Radiation Light Source (SSRL) , SLAC National Accelerator Laboratory , Menlo Park , California 94025 , United States
| | | | | |
Collapse
|
26
|
Liu Y, Zhang P, Li F, Jin X, Li J, Chen W, Li Q. Metal-based NanoEnhancers for Future Radiotherapy: Radiosensitizing and Synergistic Effects on Tumor Cells. Theranostics 2018; 8:1824-1849. [PMID: 29556359 PMCID: PMC5858503 DOI: 10.7150/thno.22172] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/05/2018] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is one of the major therapeutic strategies for cancer treatment. In the past decade, there has been growing interest in using high Z (atomic number) elements (materials) as radiosensitizers. New strategies in nanomedicine could help to improve cancer diagnosis and therapy at cellular and molecular levels. Metal-based nanoparticles usually exhibit chemical inertness in cellular and subcellular systems and may play a role in radiosensitization and synergistic cell-killing effects for radiation therapy. This review summarizes the efficacy of metal-based NanoEnhancers against cancers in both in vitro and in vivo systems for a range of ionizing radiations including gamma-rays, X-rays, and charged particles. The potential of translating preclinical studies on metal-based nanoparticles-enhanced radiation therapy into clinical practice is also discussed using examples of several metal-based NanoEnhancers (such as CYT-6091, AGuIX, and NBTXR3). Also, a few general examples of theranostic multimetallic nanocomposites are presented, and the related biological mechanisms are discussed.
Collapse
Affiliation(s)
- Yan Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Pengcheng Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Feifei Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
| | - Jin Li
- State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
| |
Collapse
|
27
|
King RB, McMahon SJ, Hyland WB, Jain S, Butterworth KT, Prise KM, Hounsell AR, McGarry CK. An overview of current practice in external beam radiation oncology with consideration to potential benefits and challenges for nanotechnology. Cancer Nanotechnol 2017; 8:3. [PMID: 28217177 PMCID: PMC5291831 DOI: 10.1186/s12645-017-0027-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 01/20/2017] [Indexed: 12/24/2022] Open
Abstract
Over the past two decades, there has been a significant evolution in the technologies and techniques employed within the radiation oncology environment. Over the same period, extensive research into the use of nanotechnology in medicine has highlighted a range of potential benefits to its incorporation into clinical radiation oncology. This short communication describes key tools and techniques that have recently been introduced into specific stages of a patient’s radiotherapy pathway, including diagnosis, external beam treatment and subsequent follow-up. At each pathway stage, consideration is given towards how nanotechnology may be combined with clinical developments to further enhance their benefit, with some potential opportunities for future research also highlighted. Prospective challenges that may influence the introduction of nanotechnology into clinical radiotherapy are also discussed, indicating the need for close collaboration between academic and clinical staff to realise the full clinical benefit of this exciting technology.
Collapse
Affiliation(s)
- Raymond B King
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE UK.,Radiotherapy Physics, Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, BT9 7AB UK
| | - Stephen J McMahon
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE UK
| | - Wendy B Hyland
- Radiotherapy Physics, North West Cancer Centre, Western Health and Social Care Trust, Londonderry, BT47 6SB UK
| | - Suneil Jain
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE UK.,Clinical Oncology, Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, BT9 7AB UK
| | - Karl T Butterworth
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE UK
| | - Kevin M Prise
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE UK
| | - Alan R Hounsell
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE UK.,Radiotherapy Physics, Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, BT9 7AB UK
| | - Conor K McGarry
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE UK.,Radiotherapy Physics, Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, BT9 7AB UK
| |
Collapse
|