1
|
Sabri ME, Moghaddasi L, Wilson P, Saran F, Bezak E. Targeted Alpha Therapy for Glioblastoma: Review on In Vitro, In Vivo and Clinical Trials. Target Oncol 2024; 19:511-531. [PMID: 38836953 PMCID: PMC11230998 DOI: 10.1007/s11523-024-01071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2024] [Indexed: 06/06/2024]
Abstract
Glioblastoma (GB), a prevalent and highly malignant primary brain tumour with a very high mortality rate due to its resistance to conventional therapies and invasive nature, resulting in 5-year survival rates of only 4-17%. Despite recent advancements in cancer management, the survival rates for GB patients have not significantly improved over the last 10-20 years. Consequently, there exists a critical unmet need for innovative therapies. One promising approach for GB is Targeted Alpha Therapy (TAT), which aims to selectively deliver potentially therapeutic radiation doses to malignant cells and the tumour microenvironment while minimising radiation exposure to surrounding normal tissue with or without conventional external beam radiation. This approach has shown promise in both pre-clinical and clinical settings. A review was conducted following PRISMA 2020 guidelines across Medline, SCOPUS, and Embase, identifying 34 relevant studies out of 526 initially found. In pre-clinical studies, TAT demonstrated high binding specificity to targeted GB cells, with affinity rates between 60.0% and 84.2%, and minimal binding to non-targeted cells (4.0-5.6%). This specificity significantly enhanced cytotoxic effects and improved biodistribution when delivered intratumorally. Mice treated with TAT showed markedly higher median survival rates compared to control groups. In clinical trials, TAT applied to recurrent GB (rGB) displayed varying success rates in extending overall survival (OS) and progression-free survival. Particularly effective when integrated into treatment regimens for both newly diagnosed and recurrent cases, TAT increased the median OS by 16.1% in newly diagnosed GB and by 36.4% in rGB, compared to current standard therapies. Furthermore, it was generally well tolerated with minimal adverse effects. These findings underscore the potential of TAT as a viable therapeutic option in the management of GB.
Collapse
Affiliation(s)
- Maram El Sabri
- Allied Health and Human Performance, University of South Australia, University of South Australia City East Campus, Adelaide, SA, 5001, Australia.
| | - Leyla Moghaddasi
- Department of Medical Physics, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
| | - Puthenparampil Wilson
- UniSA STEM, University of South Australia, Adelaide, SA, 5001, Australia
- Department of Medical Physics, Royal Adelaide Hospital, Adelaide, Australia
| | - Frank Saran
- Allied Health and Human Performance, University of South Australia, University of South Australia City East Campus, Adelaide, SA, 5001, Australia
- Australian Bragg Centre for Proton Therapy and Research, Adelaide, SA, 5000, Australia
- Department of Radiotherapy, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Eva Bezak
- Allied Health and Human Performance, University of South Australia, University of South Australia City East Campus, Adelaide, SA, 5001, Australia
| |
Collapse
|
2
|
Toro-González M, Akingbesote N, Bible A, Pal D, Sanders B, Ivanov AS, Jansone-Popova S, Popovs I, Benny P, Perry R, Davern S. Development of 225Ac-doped biocompatible nanoparticles for targeted alpha therapy. J Nanobiotechnology 2024; 22:306. [PMID: 38825717 PMCID: PMC11145892 DOI: 10.1186/s12951-024-02520-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/01/2024] [Indexed: 06/04/2024] Open
Abstract
Targeted alpha therapy (TAT) relies on chemical affinity or active targeting using radioimmunoconjugates as strategies to deliver α-emitting radionuclides to cancerous tissue. These strategies can be affected by transmetalation of the parent radionuclide by competing ions in vivo and the bond-breaking recoil energy of decay daughters. The retention of α-emitting radionuclides and the dose delivered to cancer cells are influenced by these processes. Encapsulating α-emitting radionuclides within nanoparticles can help overcome many of these challenges. Poly(lactic-co-glycolic acid) (PLGA) nanoparticles are a biodegradable and biocompatible delivery platform that has been used for drug delivery. In this study, PLGA nanoparticles are utilized for encapsulation and retention of actinium-225 ([225Ac]Ac3+). Encapsulation of [225Ac]Ac3+ within PLGA nanoparticles (Zave = 155.3 nm) was achieved by adapting a double-emulsion solvent evaporation method. The encapsulation efficiency was affected by both the solvent conditions and the chelation of [225Ac]Ac3+. Chelation of [225Ac]Ac3+ to a lipophilic 2,9-bis-lactam-1,10-phenanthroline ligand ([225Ac]AcBLPhen) significantly decreased its release (< 2%) and that of its decay daughters (< 50%) from PLGA nanoparticles. PLGA nanoparticles encapsulating [225Ac]AcBLPhen significantly increased the delivery of [225Ac]Ac3+ to murine (E0771) and human (MCF-7 and MDA-MB-231) breast cancer cells with a concomitant increase in cell death over free [225Ac]Ac3+ in solution. These results demonstrate that PLGA nanoparticles have potential as radionuclide delivery platforms for TAT to advance precision radiotherapy for cancer. In addition, this technology offers an alternative use for ligands with poor aqueous solubility, low stability, or low affinity, allowing them to be repurposed for TAT by encapsulation within PLGA nanoparticles.
Collapse
Affiliation(s)
- Miguel Toro-González
- Isotope Science and Engineering Directorate, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN, 37830, USA
| | - Ngozi Akingbesote
- Isotope Science and Engineering Directorate, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN, 37830, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Amber Bible
- Biological and Environmental Systems Science Directorate, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN, 37830, USA
| | - Debjani Pal
- Isotope Science and Engineering Directorate, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN, 37830, USA
| | - Brian Sanders
- Biological and Environmental Systems Science Directorate, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN, 37830, USA
| | - Alexander S Ivanov
- Physical Sciences Directorate, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN, 37830, USA
| | - Santa Jansone-Popova
- Physical Sciences Directorate, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN, 37830, USA
| | - Ilja Popovs
- Physical Sciences Directorate, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN, 37830, USA
| | - Paul Benny
- Isotope Science and Engineering Directorate, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN, 37830, USA
| | - Rachel Perry
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Sandra Davern
- Isotope Science and Engineering Directorate, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN, 37830, USA.
| |
Collapse
|
3
|
Pinto CIG, Branco ADM, Bucar S, Fonseca A, Abrunhosa AJ, da Silva CL, Guerreiro JF, Mendes F. Evaluation of the theranostic potential of [ 64Cu]CuCl 2 in glioblastoma spheroids. EJNMMI Res 2024; 14:26. [PMID: 38453813 PMCID: PMC10920519 DOI: 10.1186/s13550-024-01084-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Glioblastoma is an extremely aggressive malignant tumor with a very poor prognosis. Due to the increased proliferation rate of glioblastoma, there is the development of hypoxic regions, characterized by an increased concentration of copper (Cu). Considering this, 64Cu has attracted attention as a possible theranostic radionuclide for glioblastoma. In particular, [64Cu]CuCl2 accumulates in glioblastoma, being considered a suitable agent for positron emission tomography. Here, we explore further the theranostic potential of [64Cu]CuCl2, by studying its therapeutic effects in advanced three-dimensional glioblastoma cellular models. First, we established spheroids from three glioblastoma (T98G, U373, and U87) and a non-tumoral astrocytic cell line. Then, we evaluated the therapeutic responses of spheroids to [64Cu]CuCl2 exposure by analyzing spheroids' growth, viability, and cells' proliferative capacity. Afterward, we studied possible mechanisms responsible for the therapeutic outcomes, including the uptake of 64Cu, the expression levels of a copper transporter (CTR1), the presence of a cancer stem cell population, and the production of reactive oxygen species (ROS). RESULTS Results revealed that [64Cu]CuCl2 is able to significantly reduce spheroids' growth and viability, while also affecting cells' proliferation capacity. The uptake of 64Cu, the presence of cancer stem-like cells and the production of ROS were in accordance with the therapeutic response. However, expression levels of CTR1 were not in agreement with uptake levels, revealing that other mechanisms could be involved in the uptake of 64Cu. CONCLUSIONS Overall, our results further support [64Cu]CuCl2 potential as a theranostic agent for glioblastoma, unveiling potential mechanisms that could be involved in the therapeutic response.
Collapse
Affiliation(s)
- Catarina I G Pinto
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - André D M Branco
- Department of Bioengineering, iBB - Institute for Bioengineering and Biosciences, Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Bucar
- Department of Bioengineering, iBB - Institute for Bioengineering and Biosciences, Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Alexandra Fonseca
- CIBIT/ICNAS Instituto de Ciências Nucleares Aplicadas à Saúde, Universidade de Coimbra, Coimbra, Portugal
- ICNAS PHARMA, Universidade de Coimbra, Coimbra, Portugal
| | - Antero J Abrunhosa
- CIBIT/ICNAS Instituto de Ciências Nucleares Aplicadas à Saúde, Universidade de Coimbra, Coimbra, Portugal
- ICNAS PHARMA, Universidade de Coimbra, Coimbra, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering, iBB - Institute for Bioengineering and Biosciences, Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Joana F Guerreiro
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa and Laboratório Associado Para Ciência Animal e Veterinária (AL4AnimalS), Lisbon, Portugal
| | - Filipa Mendes
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
4
|
Zhang S, Shen J, Wang X, Sun X, Wu Y, Zhang M, Wang R, Hu K. Integration of organoids in peptide drug discovery: Rise of the high‐throughput screening. VIEW 2023; 4. [DOI: 10.1002/viw.20230010] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/13/2023] [Indexed: 04/05/2025] Open
Abstract
AbstractOrganoids are three‐dimensional cell aggregates with near‐physiologic cell behaviors and can undergo long‐term expansion in vitro. They are amenable to high‐throughput drug screening processes, which renders them a viable preclinical model for drug development. The procedure of organoid‐based high‐throughput screening has been extensively employed to discover small‐molecule drugs, encompassing the steps of generating organoids, examining efficient drugs in organoid cultures, and data assessment. Compared to small molecules, peptides are more straightforward to synthesize, can be modified chemically, and demonstrate high target specificity and low cytotoxicity. Therefore, they have emerged as promising carriers to deliver drugs to disease‐associated targets and could be efficient therapeutic drugs for various diseases. To date, organoids have been used to evaluate the efficacy of certain peptide agents; however, no organoid‐based high‐throughput screening of peptide drugs has been reported. Given the advantages of peptide drugs, there is an urgent need to establish organoid‐based peptide high‐throughput screening platforms. In this review, we discuss the typical approach of screening small‐molecular drugs with the use of organoid cultures, as well as provide an overview of the studies that have incorporated organoids in peptide research. Drawing on the knowledge from small molecular screens, we explore the difficulties and potential avenues for creating new platforms to identify peptide agents using organoid models.
Collapse
Affiliation(s)
- Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jieting Shen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Xiaona Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Yuxuan Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Ming‐Rong Zhang
- Department of Advanced Nuclear Medicine Sciences Institute of Quantum Medical Science National Institutes for Quantum Science and Technology Chiba Japan
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| |
Collapse
|
5
|
Hassan M, Bokhari TH, Lodhi NA, Khosa MK, Usman M. A review of recent advancements in Actinium-225 labeled compounds and biomolecules for therapeutic purposes. Chem Biol Drug Des 2023; 102:1276-1292. [PMID: 37715360 DOI: 10.1111/cbdd.14311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/03/2023] [Accepted: 07/17/2023] [Indexed: 09/17/2023]
Abstract
In nuclear medicine, cancers that cannot be cured or can only be treated partially by traditional techniques like surgery or chemotherapy are killed by ionizing radiation as a form of therapeutic treatment. Actinium-225 is an alpha-emitting radionuclide that is highly encouraging as a therapeutic approach and more promising for targeted alpha therapy (TAT). Actinium-225 is the best candidate for tumor cells treatment and has physical characteristics such as high (LET) linear energy transfer (150 keV per μm), half-life (t1/2 = 9.92d), and short ranges (400-100 μm) which prevent the damage of normal healthy tissues. The introduction of various new radiopharmaceuticals and radioisotopes has significantly assisted the advancement of nuclear medicine. Ac-225 radiopharmaceuticals continuously demonstrate their potential as targeted alpha therapeutics. 225 Ac-labeled radiopharmaceuticals have confirmed their importance in medical and clinical areas by introducing [225 Ac]Ac-PSMA-617, [225 Ac]Ac-DOTATOC, [225 Ac]Ac-DOTA-substance-P, reported significantly improved response in patients with prostate cancer, neuroendocrine, and glioma, respectively. The development of these radiopharmaceuticals required a suitable buffer, incubation time, optimal pH, and reaction temperature. There is a growing need to standardize quality control (QC) testing techniques such as radiochemical purity (RCP). This review aims to summarize the development of the Ac-225 labeled compounds and biomolecules. The current state of their reported resulting clinical applications is also summarized as well.
Collapse
Affiliation(s)
- Maria Hassan
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| | | | - Nadeem Ahmed Lodhi
- Isotope Production Division, Pakistan institute of Nuclear Science & Technology (PINSTECH), Islamabad, Pakistan
| | | | - Muhammad Usman
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| |
Collapse
|
6
|
Nayak P, Bentivoglio V, Varani M, Signore A. Three-Dimensional In Vitro Tumor Spheroid Models for Evaluation of Anticancer Therapy: Recent Updates. Cancers (Basel) 2023; 15:4846. [PMID: 37835541 PMCID: PMC10571930 DOI: 10.3390/cancers15194846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/25/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
Advanced tissue engineering processes and regenerative medicine provide modern strategies for fabricating 3D spheroids. Several different 3D cancer models are being developed to study a variety of cancers. Three-dimensional spheroids can correctly replicate some features of solid tumors (such as the secretion of soluble mediators, drug resistance mechanisms, gene expression patterns and physiological responses) better than 2D cell cultures or animal models. Tumor spheroids are also helpful for precisely reproducing the three-dimensional organization and microenvironmental factors of tumors. Because of these unique properties, the potential of 3D cell aggregates has been emphasized, and they have been utilized in in vitro models for the detection of novel anticancer drugs. This review discusses applications of 3D spheroid models in nuclear medicine for diagnosis and therapy, immunotherapy, and stem cell and photodynamic therapy and also discusses the establishment of the anticancer activity of nanocarriers.
Collapse
Affiliation(s)
- Pallavi Nayak
- Nuclear Medicine Unit, University Hospital Sant’Andrea, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, “Sapienza” University of Rome, 00189 Roma, Italy; (V.B.); (M.V.); (A.S.)
| | | | | | | |
Collapse
|
7
|
Zaid NRR, Kletting P, Beer AJ, Stallons TAR, Torgue JJ, Glatting G. Mathematical Modeling of In Vivo Alpha Particle Generators and Chelator Stability. Cancer Biother Radiopharm 2023; 38:528-535. [PMID: 33481653 DOI: 10.1089/cbr.2020.4112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Targeted α particle therapy using long-lived in vivo α particle generators is cytotoxic to target tissues. However, the redistribution of released radioactive daughters through the circulation should be considered. A mathematical model was developed to describe the physicochemical kinetics of 212Pb-labeled pharmaceuticals and its radioactive daughters. Materials and Methods: A bolus of 212Pb-labeled pharmaceuticals injected in a developed compartmental model was simulated. The contributions of chelated and free radionuclides to the total released energy were investigated for different dissociation fractions of 212Bi for different chelators, for example, 36% for DOTA. The compartmental model was applied to describe a 212Bi retention study and to assess the stability of the 212Bi-1,4,7,10-tetrakis(carbamoylmethyl)-1,4,7,10-tetraazacyclododecane (212Bi-DOTAM) complex after β- decay of 212Pb. Results: The simulation of the injection showed that α emissions contribute 75% to the total released energy, mostly from 212Po (72%). The simulation of the 212Bi retention study showed that (16 ± 5)% of 212Bi atoms dissociate from the 212Bi-DOTAM complexes. The fractions of energies released by free radionuclides were 21% and 38% for DOTAM and DOTA chelators, respectively. Conclusion: The developed α particle generator model allows for simulating the radioactive kinetics of labeled and unlabeled pharmaceuticals being released from the chelating system due to a preceding disintegration.
Collapse
Affiliation(s)
- Nouran R R Zaid
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany
- Department of Biomedical Sciences, Biophysics and Medical Imaging Program, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Peter Kletting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | | | | | - Gerhard Glatting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| |
Collapse
|
8
|
Ahmed T. Biomaterial-based in vitro 3D modeling of glioblastoma multiforme. CANCER PATHOGENESIS AND THERAPY 2023; 1:177-194. [PMID: 38327839 PMCID: PMC10846340 DOI: 10.1016/j.cpt.2023.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/24/2022] [Accepted: 01/04/2023] [Indexed: 02/09/2024]
Abstract
Adult-onset brain cancers, such as glioblastomas, are particularly lethal. People with glioblastoma multiforme (GBM) do not anticipate living for more than 15 months if there is no cure. The results of conventional treatments over the past 20 years have been underwhelming. Tumor aggressiveness, location, and lack of systemic therapies that can penetrate the blood-brain barrier are all contributing factors. For GBM treatments that appear promising in preclinical studies, there is a considerable rate of failure in phase I and II clinical trials. Unfortunately, access becomes impossible due to the intricate architecture of tumors. In vitro, bioengineered cancer models are currently being used by researchers to study disease development, test novel therapies, and advance specialized medications. Many different techniques for creating in vitro systems have arisen over the past few decades due to developments in cellular and tissue engineering. Later-stage research may yield better results if in vitro models that resemble brain tissue and the blood-brain barrier are used. With the use of 3D preclinical models made available by biomaterials, researchers have discovered that it is possible to overcome these limitations. Innovative in vitro models for the treatment of GBM are possible using biomaterials and novel drug carriers. This review discusses the benefits and drawbacks of 3D in vitro glioblastoma modeling systems.
Collapse
Affiliation(s)
- Tanvir Ahmed
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| |
Collapse
|
9
|
Lin J, Yin M, Liu X, Meng F, Luo L. Nanomaterials Based on Functional Polymers for Sensitizing Cancer Radiotherapy. Macromol Rapid Commun 2022; 43:e2200194. [PMID: 35578790 DOI: 10.1002/marc.202200194] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/21/2022] [Indexed: 11/12/2022]
Abstract
Despite being the mainstay treatment for many types of cancer in clinic, radiotherapy is undertaking great challenges in overcoming a series of limitations. Radiosensitizers are promising agents capable of depositing irradiation energy and generating free radicals to enhance the radiosensitivity of tumor cells. Combining radiosensitizers with functional polymer-based nanomaterials holds great potential to improve biodistribution, circulation time, and stability in vivo. The derived polymeric nano-radiosensitizers can significantly improve the efficiency of tumor targeting and radiotherapy, and reduce the side effect to healthy tissues. In this review, we provide an overview of functional polymer-based nanomaterials for radiosensitization in recent years. Particular emphases are given to the action mechanisms, drug loading methods, targeting efficiencies, the impact on therapeutic effects and biocompatibility of various radiosensitizing polymers, which are classified as polymeric micelles, dendrimers, polymeric nanospheres, nanoscale coordination polymers, polymersomes, and nanogels. The challenges and outlooks of polymeric nano-radiosensitizers are also discussed. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jinfeng Lin
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Mingming Yin
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaoming Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Fanling Meng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Liang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
10
|
Mapanao AK, Che PP, Sarogni P, Sminia P, Giovannetti E, Voliani V. Tumor grafted - chick chorioallantoic membrane as an alternative model for biological cancer research and conventional/nanomaterial-based theranostics evaluation. Expert Opin Drug Metab Toxicol 2021; 17:947-968. [PMID: 33565346 DOI: 10.1080/17425255.2021.1879047] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/18/2021] [Indexed: 02/09/2023]
Abstract
Introduction: Advancements in cancer management and treatment are associated with strong preclinical research data, in which reliable cancer models are demanded. Indeed, inconsistent preclinical findings and stringent regulations following the 3Rs principle of reduction, refinement, and replacement of conventional animal models currently pose challenges in the development and translation of efficient technologies. The chick embryo chorioallantoic membrane (CAM) is a system for the evaluation of treatment effects on the vasculature, therefore suitable for studies on angiogenesis. Apart from vascular effects, the model is now increasingly employed as a preclinical cancer model following tumor-grafting procedures.Areas covered: The broad application of CAM tumor model is highlighted along with the methods for analyzing the neoplasm and vascular system. The presented and cited investigations focus on cancer biology and treatment, encompassing both conventional and emerging nanomaterial-based modalities.Expert opinion: The CAM tumor model finds increased significance given the influences of angiogenesis and the tumor microenvironment in cancer behavior, then providing a qualified miniature system for oncological research. Ultimately, the establishment and increased employment of such a model may resolve some of the limitations present in the standard preclinical tumor models, thereby redefining the preclinical research workflow.
Collapse
Affiliation(s)
- Ana Katrina Mapanao
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
- NEST-Scuola Normale Superiore, Pisa, Italy
| | - Pei Pei Che
- Department of Radiation Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, The Netherlands
| | - Patrizia Sarogni
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
| | - Peter Sminia
- Department of Radiation Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per La Scienza, Pisa, Italy
| | - Valerio Voliani
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
| |
Collapse
|
11
|
Trujillo-Nolasco M, Morales-Avila E, Cruz-Nova P, Katti KV, Ocampo-García B. Nanoradiopharmaceuticals Based on Alpha Emitters: Recent Developments for Medical Applications. Pharmaceutics 2021; 13:1123. [PMID: 34452084 PMCID: PMC8398190 DOI: 10.3390/pharmaceutics13081123] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 12/02/2022] Open
Abstract
The application of nanotechnology in nuclear medicine offers attractive therapeutic opportunities for the treatment of various diseases, including cancer. Indeed, nanoparticles-conjugated targeted alpha-particle therapy (TAT) would be ideal for localized cell killing due to high linear energy transfer and short ranges of alpha emitters. New approaches in radiolabeling are necessary because chemical radiolabeling techniques are rendered sub-optimal due to the presence of recoil energy generated by alpha decay, which causes chemical bonds to break. This review attempts to cover, in a concise fashion, various aspects of physics, radiobiology, and production of alpha emitters, as well as highlight the main problems they present, with possible new approaches to mitigate those problems. Special emphasis is placed on the strategies proposed for managing recoil energy. We will also provide an account of the recent studies in vitro and in vivo preclinical investigations of α-particle therapy delivered by various nanosystems from different materials, including inorganic nanoparticles, liposomes, and polymersomes, and some carbon-based systems are also summarized.
Collapse
Affiliation(s)
- Maydelid Trujillo-Nolasco
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Carretera México-Toluca S/N, Ocoyoacac 52750, Mexico; (M.T.-N.); (P.C.-N.)
- Facultad de Química, Universidad Autónoma del Estado de México, Paseo Tollocan S/N, Toluca 50120, Mexico;
| | - Enrique Morales-Avila
- Facultad de Química, Universidad Autónoma del Estado de México, Paseo Tollocan S/N, Toluca 50120, Mexico;
| | - Pedro Cruz-Nova
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Carretera México-Toluca S/N, Ocoyoacac 52750, Mexico; (M.T.-N.); (P.C.-N.)
| | - Kattesh V. Katti
- Department of Radiology, Institute of Green Nanotechnology, University of Missouri, Columbia, MO 65212, USA;
| | - Blanca Ocampo-García
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Carretera México-Toluca S/N, Ocoyoacac 52750, Mexico; (M.T.-N.); (P.C.-N.)
| |
Collapse
|
12
|
Stanković T, Ranđelović T, Dragoj M, Stojković Burić S, Fernández L, Ochoa I, Pérez-García VM, Pešić M. In vitro biomimetic models for glioblastoma-a promising tool for drug response studies. Drug Resist Updat 2021; 55:100753. [PMID: 33667959 DOI: 10.1016/j.drup.2021.100753] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
The poor response of glioblastoma to current treatment protocols is a consequence of its intrinsic drug resistance. Resistance to chemotherapy is primarily associated with considerable cellular heterogeneity, and plasticity of glioblastoma cells, alterations in gene expression, presence of specific tumor microenvironment conditions and blood-brain barrier. In an attempt to successfully overcome chemoresistance and better understand the biological behavior of glioblastoma, numerous tri-dimensional (3D) biomimetic models were developed in the past decade. These novel advanced models are able to better recapitulate the spatial organization of glioblastoma in a real time, therefore providing more realistic and reliable evidence to the response of glioblastoma to therapy. Moreover, these models enable the fine-tuning of different tumor microenvironment conditions and facilitate studies on the effects of the tumor microenvironment on glioblastoma chemoresistance. This review outlines current knowledge on the essence of glioblastoma chemoresistance and describes the progress achieved by 3D biomimetic models. Moreover, comprehensive literature assessment regarding the influence of 3D culturing and microenvironment mimicking on glioblastoma gene expression and biological behavior is also provided. The contribution of the blood-brain barrier as well as the blood-tumor barrier to glioblastoma chemoresistance is also reviewed from the perspective of 3D biomimetic models. Finally, the role of mathematical models in predicting 3D glioblastoma behavior and drug response is elaborated. In the future, technological innovations along with mathematical simulations should create reliable 3D biomimetic systems for glioblastoma research that should facilitate the identification and possibly application in preclinical drug testing and precision medicine.
Collapse
Affiliation(s)
- Tijana Stanković
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Teodora Ranđelović
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Miodrag Dragoj
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Sonja Stojković Burić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Luis Fernández
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Centro Investigación Biomédica en Red. Bioingenieria, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Centro Investigación Biomédica en Red. Bioingenieria, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Victor M Pérez-García
- Departamento de Matemáticas, E.T.S.I. Industriales and Instituto de Matemática Aplicada a la Ciencia y la Ingeniería (IMACI), Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia.
| |
Collapse
|
13
|
Tailoring the Radionuclide Encapsulation and Surface Chemistry of La(223Ra)VO4 Nanoparticles for Targeted Alpha Therapy. JOURNAL OF NANOTHERANOSTICS 2021. [DOI: 10.3390/jnt2010003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The development of targeted alpha therapy (TAT) as a viable cancer treatment requires innovative solutions to challenges associated with radionuclide retention to enhance local tumor cytotoxicity and to minimize off-target effects. Nanoparticles (NPs) with high encapsulation and high retention of radionuclides have shown potential in overcoming these issues. This article shows the influence of pH on the structure of lanthanum vanadate (LaVO4) NPs and its impact on the radiochemical yield of 223Ra and subsequent retention of its decay daughters, 211Pb and 211Bi. An acidic pH (4.9) results in a high fraction of La(223Ra)VO4 NPs with tetragonal structure (44.6–66.1%) and a 223Ra radiochemical yield <40%. Adjusting the pH to 11 yields >80% of La(223Ra)VO4 NPs with monoclinic structure and increases the 223Ra radiochemical yield >85%. The leakage of decay daughters from La(223Ra)VO4 NPs (pH 11) was <5% and <0.5% when exposed to deionized water and phosphate-buffered saline, respectively. Altering the surface chemistry of La(223Ra)VO4 NPs with carboxylate and phosphate compounds resulted in a threefold decrease in hydrodynamic diameter and a 223Ra radiochemical yield between 74.7% and 99.6%. These results show the importance of tailoring the synthesis parameters and surface chemistry of LaVO4 NPs to obtain high encapsulation and retention of radionuclides.
Collapse
|
14
|
Doctor A, Seifert V, Ullrich M, Hauser S, Pietzsch J. Three-Dimensional Cell Culture Systems in Radiopharmaceutical Cancer Research. Cancers (Basel) 2020; 12:cancers12102765. [PMID: 32993034 PMCID: PMC7600608 DOI: 10.3390/cancers12102765] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
In preclinical cancer research, three-dimensional (3D) cell culture systems such as multicellular spheroids and organoids are becoming increasingly important. They provide valuable information before studies on animal models begin and, in some cases, are even suitable for reducing or replacing animal experiments. Furthermore, they recapitulate microtumors, metastases, and the tumor microenvironment much better than monolayer culture systems could. Three-dimensional models show higher structural complexity and diverse cell interactions while reflecting (patho)physiological phenomena such as oxygen and nutrient gradients in the course of their growth or development. These interactions and properties are of great importance for understanding the pathophysiological importance of stromal cells and the extracellular matrix for tumor progression, treatment response, or resistance mechanisms of solid tumors. Special emphasis is placed on co-cultivation with tumor-associated cells, which further increases the predictive value of 3D models, e.g., for drug development. The aim of this overview is to shed light on selected 3D models and their advantages and disadvantages, especially from the radiopharmacist's point of view with focus on the suitability of 3D models for the radiopharmacological characterization of novel radiotracers and radiotherapeutics. Special attention is paid to pancreatic ductal adenocarcinoma (PDAC) as a predestined target for the development of new radionuclide-based theranostics.
Collapse
Affiliation(s)
- Alina Doctor
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
| | - Verena Seifert
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
| | - Martin Ullrich
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
- Correspondence: ; Tel.: +49-351-260-2622
| |
Collapse
|
15
|
Toro-González M, Dame AN, Mirzadeh S, Rojas JV. Encapsulation and retention of 225Ac, 223Ra, 227Th, and decay daughters in zircon-type gadolinium vanadate nanoparticles. RADIOCHIM ACTA 2020. [DOI: 10.1515/ract-2019-3206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Unwanted targeting of healthy organs caused by the relocation of radionuclides from the target site has been one of the limiting factors in the widespread application of targeted alpha therapy in patient regimens. GdVO4 nanoparticles (NPs) were developed as platforms to encapsulate α-emitting radionuclides 223Ra, 225Ac, and 227Th, and retain their decay daughters at the target site. Polycrystalline GdVO4 NPs with different morphologies and a zircon-type tetragonal crystal structure were obtained by precipitation of GdCl3 and Na3VO4 in aqueous media at room temperature. The ability of GdVO4 crystals to host multivalent ions was initially assessed using La, Cs, Bi, Ba, and Pb as surrogates of the radionuclides under investigation. A decrease in Ba encapsulation was obtained after increasing the concentration of surrogate ions, whereas the encapsulation of La cations in GdVO4 NPs was quantitative (∼100%). Retention of radionuclides was assessed in vitro by dialyzing the radioactive GdVO4 NPs against deionized water. While 227Th was quantitatively encapsulated (100%), a partial encapsulation of 223Ra (∼75%) and 225Ac (>60%) was observed in GdVO4 NPs. The maximum leakage of 221Fr (1st decay daughter of 225Ac) was 55.4 ± 3.6%, whereas for 223Ra (1st decay daughter of 227Th) the maximum leakage was 73.0 ± 4.0%. These results show the potential of GdVO4 NPs as platforms of α-emitting radionuclides for their application in targeted alpha therapy.
Collapse
Affiliation(s)
- Miguel Toro-González
- Department of Mechanical and Nuclear Engineering , Virginia Commonwealth University , Richmond , VA , USA
- Isotope and Fuel Cycle Technology Division , Oak Ridge National Laboratory , Oak Ridge , TN , USA
| | - Ashley N. Dame
- Isotope and Fuel Cycle Technology Division , Oak Ridge National Laboratory , Oak Ridge , TN , USA
| | - Saed Mirzadeh
- Isotope and Fuel Cycle Technology Division , Oak Ridge National Laboratory , Oak Ridge , TN , USA
| | - Jessika V. Rojas
- Department of Mechanical and Nuclear Engineering , Virginia Commonwealth University , Richmond , VA , USA
| |
Collapse
|
16
|
Majkowska-Pilip A, Gawęda W, Żelechowska-Matysiak K, Wawrowicz K, Bilewicz A. Nanoparticles in Targeted Alpha Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1366. [PMID: 32668687 PMCID: PMC7408031 DOI: 10.3390/nano10071366] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 02/01/2023]
Abstract
Recent advances in the field of nanotechnology application in nuclear medicine offer the promise of better therapeutic options. In recent years, increasing efforts have been made on developing nanoconstructs that can be used as carriers for immobilising alpha (α)-emitters in targeted drug delivery. In this publication, we provide a comprehensive overview of available information on functional nanomaterials for targeted alpha therapy. The first section describes why nanoconstructs are used for the synthesis of α-emitting radiopharmaceuticals. Next, we present the synthesis and summarise the recent studies demonstrating therapeutic applications of α-emitting labelled radiobioconjugates in targeted therapy. Finally, future prospects and the emerging possibility of therapeutic application of radiolabelled nanomaterials are discussed.
Collapse
Affiliation(s)
- Agnieszka Majkowska-Pilip
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (W.G.); (K.Ż.-M.); (K.W.); (A.B.)
| | | | | | | | | |
Collapse
|
17
|
Xu T, Liu T, Li G, Dugal C, Aydemir NA, Liu Y, Roeske JC. Technical Note: The development of a multi-physics simulation tool to estimate the background dose by systemic targeted alpha therapy. Med Phys 2020; 47:2550-2557. [PMID: 32129888 PMCID: PMC7384137 DOI: 10.1002/mp.14111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/20/2020] [Accepted: 02/18/2020] [Indexed: 11/09/2022] Open
Abstract
PURPOSE To predict biological effects of targeted alpha therapy (TAT) in preclinical studies, dosimetry calculations based on the micro-level distributions of emitters are essential. Due to the saturation of the tumor antigenic sites and bonding breaks by decay, some of Alpha-immuno-conjugate and decay daughters may inevitably be transported by convection and diffusion along with blood or lymphatic circulation. This results in highly nonuniform and unsteady distributions of irradiation sources. Since the micro-level distribution of emitters cannot be measured and obtained in patients with current technology, a modeling toolset to give more insight of the internal dose could be an alternative. METHODS A multi-physics model based on a Monte Carlo microdosimetry technique and computational fluid dynamics (CFD) modeling was developed and applied to multiple internal irradiation sources. The CFD model tracks the path of the radionuclides and the dose model is capable of evaluating the time-dependent absorbed dose to the target. RESULTS The conceptual model is capable of handling complex nonuniform irradiation sources in vasculature. The results from the simulations indicate that the assumption of homogeneous and motionless distribution of the administered activity used in the conventional dose calculation tends to significantly underestimate or overestimate the absorbed dose to the vascular system in various scenarios. CONCLUSION Modeling the in vivo transport of radionuclides has the potential to improve the accuracy of TAT dose estimates. It could be the first step to develop a simulation tool set for assessing absorbed dose to tumor or normal tissues and predict the corresponding biological responses in the future.
Collapse
Affiliation(s)
- T. Xu
- Canadian Nuclear LaboratoriesChalk RiverONK0J 1J0Canada
| | - T. Liu
- Canadian Nuclear LaboratoriesChalk RiverONK0J 1J0Canada
| | - G. Li
- Canadian Nuclear LaboratoriesChalk RiverONK0J 1J0Canada
| | - C. Dugal
- Canadian Nuclear LaboratoriesChalk RiverONK0J 1J0Canada
| | - N. A. Aydemir
- Canadian Nuclear LaboratoriesChalk RiverONK0J 1J0Canada
| | - Y. Liu
- Department of Electrical and Computer EngineeringClarkson UniversityPotsdamNY13699USA
| | - J. C. Roeske
- Department of Radiation OncologyLoyola University Medical CenterMaywoodIL60153USA
| |
Collapse
|
18
|
Toro-González M, Dame AN, Foster CM, Millet LJ, Woodward JD, Rojas JV, Mirzadeh S, Davern SM. Quantitative encapsulation and retention of 227Th and decay daughters in core-shell lanthanum phosphate nanoparticles. NANOSCALE 2020; 12:9744-9755. [PMID: 32324185 DOI: 10.1039/d0nr01172j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Targeted alpha therapy (TAT) offers great promise for treating recalcitrant tumors and micrometastatic cancers. One drawback of TAT is the potential damage to normal tissues and organs due to the relocation of decay daughters from the treatment site. The present study evaluates La(227Th)PO4 core (C) and core +2 shells (C2S) nanoparticles (NPs) as a delivery platform of 227Th to minimize systemic distribution of decay daughters, 223Ra and 211Pb. In vitro retention of decay daughters within La(227Th)PO4 C NPs was influenced by the concentration of reagents used during synthesis, in which the leakage of 223Ra was between 0.4 ± 0.2% and 20.3 ± 1.1% in deionized water. Deposition of two nonradioactive LaPO4 shells onto La(227Th)PO4 C NPs increased the retention of decay daughters to >99.75%. The toxicity of the nonradioactive LaPO4 C and C2S NP delivery platforms was examined in a mammalian breast cancer cell line, BT-474. No significant decrease in cell viability was observed for a monolayer of BT-474 cells for NP concentrations below 233.9 μg mL-1, however cell viability decreased below 60% when BT-474 spheroids were incubated with either LaPO4 C or C2S NPs at concentrations exceeding 29.2 μg mL-1. La(227Th)PO4 C2S NPs exhibit a high encapsulation and in vitro retention of radionuclides with limited contribution to cellular cytotoxicity for TAT applications.
Collapse
Affiliation(s)
- M Toro-González
- Department of Mechanical and Nuclear Engineering, Virginia Commonwealth University, , Richmond 23284, USA. and Isotope and Fuel Cycle Technology Division, Oak Ridge National Laboratory, Oak Ridge 37830, USA.
| | - A N Dame
- Isotope and Fuel Cycle Technology Division, Oak Ridge National Laboratory, Oak Ridge 37830, USA.
| | - C M Foster
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge 37830, USA
| | - L J Millet
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge 37830, USA and Joint Research Activity, The Bredesen Center, University of Tennessee, Knoxville 37996, USA
| | - J D Woodward
- Isotope and Fuel Cycle Technology Division, Oak Ridge National Laboratory, Oak Ridge 37830, USA.
| | - J V Rojas
- Department of Mechanical and Nuclear Engineering, Virginia Commonwealth University, , Richmond 23284, USA.
| | - S Mirzadeh
- Isotope and Fuel Cycle Technology Division, Oak Ridge National Laboratory, Oak Ridge 37830, USA.
| | - S M Davern
- Isotope and Fuel Cycle Technology Division, Oak Ridge National Laboratory, Oak Ridge 37830, USA.
| |
Collapse
|
19
|
Cędrowska E, Pruszyński M, Gawęda W, Żuk M, Krysiński P, Bruchertseifer F, Morgenstern A, Karageorgou MA, Bouziotis P, Bilewicz A. Trastuzumab Conjugated Superparamagnetic Iron Oxide Nanoparticles Labeled with 225Ac as a Perspective Tool for Combined α-Radioimmunotherapy and Magnetic Hyperthermia of HER2-Positive Breast Cancer. Molecules 2020; 25:molecules25051025. [PMID: 32106568 PMCID: PMC7179151 DOI: 10.3390/molecules25051025] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/23/2020] [Accepted: 02/24/2020] [Indexed: 12/18/2022] Open
Abstract
It has been proven and confirmed in numerous repeated tests, that the use of a combination of several therapeutic methods gives much better treatment results than in the case of separate therapies. Particularly promising is the combination of ionizing radiation and magnetic hyperthermia in one drug. To achieve this objective, magnetite nanoparticles have been modified in their core with α emitter 225Ac, in an amount affecting only slightly their magnetic properties. By 3-phosphonopropionic acid (CEPA) linker nanoparticles were conjugated covalently with trastuzumab (Herceptin®), a monoclonal antibody that recognizes ovarian and breast cancer cells overexpressing the HER2 receptors. The synthesized bioconjugates were characterized by transmission electron microscopy (TEM), Dynamic Light Scattering (DLS) measurement, thermogravimetric analysis (TGA) and application of 131I-labeled trastuzumab for quantification of the bound biomolecule. The obtained results show that one 225Ac@Fe3O4-CEPA-trastuzumab bioconjugate contains an average of 8–11 molecules of trastuzumab. The labeled nanoparticles almost quantitatively retain 225Ac (>98%) in phosphate-buffered saline (PBS) and physiological salt, and more than 90% of 221Fr and 213Bi over 10 days. In human serum after 10 days, the fraction of 225Ac released from 225Ac@Fe3O4 was still less than 2%, but the retention of 221Fr and 213Bi decreased to 70%. The synthesized 225Ac@Fe3O4-CEPA-trastuzumab bioconjugates have shown a high cytotoxic effect toward SKOV-3 ovarian cancer cells expressing HER2 receptor in-vitro. The in-vivo studies indicate that this bioconjugate exhibits properties suitable for the treatment of cancer cells by intratumoral or post-resection injection. The intravenous injection of the 225Ac@Fe3O4-CEPA-trastuzumab radiobioconjugate is excluded due to its high accumulation in the liver, lungs and spleen. Additionally, the high value of a specific absorption rate (SAR) allows its use in a new very perspective combination of α radionuclide therapy with magnetic hyperthermia.
Collapse
Affiliation(s)
- Edyta Cędrowska
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (E.C.); (W.G.)
| | - Marek Pruszyński
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (E.C.); (W.G.)
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089 Warsaw, Poland
- Correspondence: (M.P.); (A.B.); Tel.: +48-22-5041357 (A.B.)
| | - Weronika Gawęda
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (E.C.); (W.G.)
| | - Michał Żuk
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland; (M.Ż.); (P.K.)
| | - Paweł Krysiński
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland; (M.Ż.); (P.K.)
| | - Frank Bruchertseifer
- Department for Nuclear Safety and Security, Joint Research Centre, European Commission, 76125 Karlsruhe, Germany; (F.B.); (A.M.)
| | - Alfred Morgenstern
- Department for Nuclear Safety and Security, Joint Research Centre, European Commission, 76125 Karlsruhe, Germany; (F.B.); (A.M.)
| | - Maria-Argyro Karageorgou
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, N.C.S.R. ‘Demokritos’, Aghia Paraskevi, 15341 Athens, Greece; (M.-A.K.); (P.B.)
- Department of Physics, National and Kapodistrian University of Athens, Zografou Panepistimioupolis, 15784 Athens, Greece
| | - Penelope Bouziotis
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, N.C.S.R. ‘Demokritos’, Aghia Paraskevi, 15341 Athens, Greece; (M.-A.K.); (P.B.)
| | - Aleksander Bilewicz
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (E.C.); (W.G.)
- Correspondence: (M.P.); (A.B.); Tel.: +48-22-5041357 (A.B.)
| |
Collapse
|
20
|
Roobol SJ, Hartjes TA, Slotman JA, de Kruijff RM, Torrelo G, Abraham TE, Bruchertseifer F, Morgenstern A, Kanaar R, van Gent DC, Houtsmuller AB, Denkova AG, van Royen ME, Essers J. Uptake and subcellular distribution of radiolabeled polymersomes for radiotherapy. Nanotheranostics 2020; 4:14-25. [PMID: 31911891 PMCID: PMC6940201 DOI: 10.7150/ntno.37080] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 11/06/2019] [Indexed: 12/17/2022] Open
Abstract
Polymersomes have the potential to be applied in targeted alpha radionuclide therapy, while in addition preventing release of recoiling daughter isotopes. In this study, we investigated the cellular uptake, post uptake processing and intracellular localization of polymersomes. Methods: High-content microscopy was used to validate polymersome uptake kinetics. Confocal (live cell) microscopy was used to elucidate the uptake mechanism and DNA damage induction. Intracellular distribution of polymersomes in 3-D was determined using super-resolution microscopy. Results: We found that altering polymersome size and concentration affects the initial uptake and overall uptake capacity; uptake efficiency and eventual plateau levels varied between cell lines; and mitotic cells show increased uptake. Intracellular polymersomes were transported along microtubules in a fast and dynamic manner. Endocytic uptake of polymersomes was evidenced through co-localization with endocytic pathway components. Finally, we show the intracellular distribution of polymersomes in 3-D and DNA damage inducing capabilities of 213Bi labeled polymersomes. Conclusion: Polymersome size and concentration affect the uptake efficiency, which also varies for different cell types. In addition, we present advanced assays to investigate uptake characteristics in detail, a necessity for optimization of nano-carriers. Moreover, by elucidating the uptake mechanism, as well as uptake extent and geometrical distribution of radiolabeled polymersomes we provide insight on how to improve polymersome design.
Collapse
Affiliation(s)
- Stefan J. Roobol
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
- Oncode Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Thomas A. Hartjes
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Optical Imaging Centre (OIC), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Johan A. Slotman
- Optical Imaging Centre (OIC), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robin M. de Kruijff
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| | - Guzman Torrelo
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| | - Tsion E. Abraham
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Optical Imaging Centre (OIC), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Frank Bruchertseifer
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - Alfred Morgenstern
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - Roland Kanaar
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
- Oncode Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dik C. van Gent
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
- Oncode Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Adriaan B. Houtsmuller
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Optical Imaging Centre (OIC), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Antonia G. Denkova
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| | - Martin E. van Royen
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Optical Imaging Centre (OIC), Erasmus University Medical Center, Rotterdam, The Netherlands
- Cancer Treatment Screening Facility (CTSF), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiation Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
21
|
Preclinical evaluation of binimetinib (MEK162) delivered via polymeric nanocarriers in combination with radiation and temozolomide in glioma. J Neurooncol 2019; 146:239-246. [PMID: 31875307 PMCID: PMC6971148 DOI: 10.1007/s11060-019-03365-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/09/2019] [Indexed: 12/27/2022]
Abstract
Background and purpose Glioblastoma multiforme (GBM) is the most aggressive subtype of malignant gliomas, with an average survival rate of 15 months after diagnosis. More than 90% of all GBMs have activating mutations in the MAPK/ERK pathway. Recently, we showed the allosteric MEK1/2 inhibitor binimetinib (MEK162) to inhibit cell proliferation and to enhance the effect of radiation in preclinical human GBM models. Because the free drug cannot pass the blood–brain barrier (BBB), we investigated the use of nanocarriers for transport of the drug through the BBB and its efficacy when combined with radiotherapy and temozolomide (TMZ) in glioma spheroids. Methods In vitro studies were performed using multicellular U87 human GBM spheroids. Polymeric nanocarriers (polymersomes) were loaded with MEK162. The interaction between nanocarrier delivered MEK162, irradiation and TMZ was studied on the kinetics of spheroid growth and on protein expression in the MAPK/ERK pathway. BBB passaging was evaluated in a transwell system with human cerebral microvascular endothelial (hCMEC/D3) cells. Results MEK162 loaded polymersomes inhibited spheroid growth. A synergistic effect was found in combination with fractionated irradiation and an additive effect with TMZ on spheroid volume reduction. Fluorescent labeled polymersomes were taken up by human cerebral microvascular endothelial cells and passed the BBB in vitro. Conclusion MEK162 loaded polymersomes are taken up by multicellular spheroids. The nanocarrier delivered drug reduced spheroid growth and inhibited its molecular target. MEK162 delivered via polymersomes showed interaction with irradiation and TMZ. The polymersomes crossed the in vitro BBB model and therewith offer exciting challenges ahead for delivery of therapeutics agents to brain tumours.
Collapse
|
22
|
Loya J, Zhang C, Cox E, Achrol AS, Kesari S. Biological intratumoral therapy for the high-grade glioma part II: vector- and cell-based therapies and radioimmunotherapy. CNS Oncol 2019; 8:CNS40. [PMID: 31747784 PMCID: PMC6880300 DOI: 10.2217/cns-2019-0002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Management of high-grade gliomas (HGGs) remains a complex challenge with an overall poor prognosis despite aggressive multimodal treatment. New translational research has focused on maximizing tumor cell eradication through improved tumor cell targeting while minimizing collateral systemic side effects. In particular, biological intratumoral therapies have been the focus of novel translational research efforts due to their inherent potential to be both dynamically adaptive and target specific. This two part review will provide an overview of biological intratumoral therapies that have been evaluated in human clinical trials in HGGs, and summarize key advances and remaining challenges in the development of these therapies as a potential new paradigm in the management of HGGs. Part II discusses vector-based therapies, cell-based therapies and radioimmunotherapy.
Collapse
Affiliation(s)
- Joshua Loya
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Charlie Zhang
- Buffalo School of Medicine, State University of New York, Buffalo, NY 14202, USA
| | - Emily Cox
- Providence Medical Research Center, Spokane, WA 99204, USA
| | - Achal S Achrol
- John Wayne Cancer Institute, Pacific Neuroscience Institute, Santa Monica, CA 90404, USA
| | - Santosh Kesari
- John Wayne Cancer Institute, Pacific Neuroscience Institute, Santa Monica, CA 90404, USA
| |
Collapse
|
23
|
Kruijff RMD, Raavé R, Kip A, Molkenboer-Kuenen J, Morgenstern A, Bruchertseifer F, Heskamp S, Denkova AG. The in vivo fate of 225Ac daughter nuclides using polymersomes as a model carrier. Sci Rep 2019; 9:11671. [PMID: 31406320 PMCID: PMC6690960 DOI: 10.1038/s41598-019-48298-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/01/2019] [Indexed: 12/19/2022] Open
Abstract
Increasing attention is given to personalized tumour therapy, where α-emitters can potentially play an important role. Alpha particles are ideal for localized cell killing because of their high linear energy transfer and short ranges. However, upon the emission of an α particle the daughter nuclide experiences a recoil energy large enough to ensure decoupling from any chemical bond. These 'free' daughter nuclides are no longer targeted to the tumour and can accumulate in normal tissue. In this paper, we used polymersomes as model carrier to evaluate the retention of recoiling daughters of 225Ac in vivo, and assessed their suitability as therapeutic agents. Vesicles containing 225Ac were injected intravenously in healthy mice, and intratumourally in tumour-bearing mice, and the relocation of free 213Bi was assessed in different organs upon the injection [225Ac]Ac-polymersomes. The therapeutic effect of 225Ac-containing vesicles was studied upon intratumoural injection, where treatment groups experienced no tumour-related deaths over a 115 day period. While polymersomes containing 225Ac could be suitable agents for long-term irradiation of tumours without causing significant renal toxicity, there is still a significant re-distribution of daughter nuclides throughout the body, signifying the importance of careful evaluation of the effect of daughter nuclides in targeted alpha therapy.
Collapse
Affiliation(s)
- R M de Kruijff
- Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands.
| | - R Raavé
- Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - A Kip
- Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - J Molkenboer-Kuenen
- Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - A Morgenstern
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - F Bruchertseifer
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - S Heskamp
- Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - A G Denkova
- Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
24
|
de Kruijff RM, Raavé R, Kip A, Molkenboer-Kuenen J, Roobol SJ, Essers J, Heskamp S, Denkova AG. Elucidating the Influence of Tumor Presence on the Polymersome Circulation Time in Mice. Pharmaceutics 2019; 11:E241. [PMID: 31137479 PMCID: PMC6572275 DOI: 10.3390/pharmaceutics11050241] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 11/16/2022] Open
Abstract
The use of nanoparticles as tumor-targeting agents is steadily increasing, and the influence of nanoparticle characteristics such as size and stealthiness have been established for a large number of nanocarrier systems. However, not much is known about the impact of tumor presence on nanocarrier circulation times. This paper reports on the influence of tumor presence on the in vivo circulation time and biodistribution of polybutadiene-polyethylene oxide (PBd-PEO) polymersomes. For this purpose, polymersomes were loaded with the gamma-emitter 111In and administered intravenously, followed by timed ex vivo biodistribution. A large reduction in circulation time was observed for tumor-bearing mice, with a circulation half-life of merely 5 min (R2 = 0.98) vs 117 min (R2 = 0.95) in healthy mice. To determine whether the rapid polymersome clearance observed in tumor-bearing mice was mediated by macrophages, chlodronate liposomes were administered to both healthy and tumor-bearing mice prior to the intravenous injection of radiolabeled polymersomes to deplete their macrophages. Pretreatment with chlodronate liposomes depleted macrophages in the spleen and liver and restored the circulation time of the polymersomes with no significant difference in circulation time between healthy mice and tumor-bearing mice pretreated with clodronate liposomes (15.2 ± 1.2% ID/g and 13.6 ± 2.7% ID/g, respectively, at 4 h p.i. with p = 0.3). This indicates that activation of macrophages due to tumor presence indeed affected polymersome clearance rate. Thus, next to particle design, the presence of a tumor can also greatly impact circulation times and should be taken into account when designing studies to evaluate the distribution of polymersomes.
Collapse
Affiliation(s)
- Robin M de Kruijff
- Radiation Science and Technology, Delft University of Technology, 2629 JB Delft, The Netherlands.
| | - René Raavé
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Annemarie Kip
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Janneke Molkenboer-Kuenen
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Stefan J Roobol
- Molecular Genetics, Oncode Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands.
- Radiology and Nuclear Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands.
| | - Jeroen Essers
- Molecular Genetics, Oncode Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands.
| | - Sandra Heskamp
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Antonia G Denkova
- Radiation Science and Technology, Delft University of Technology, 2629 JB Delft, The Netherlands.
| |
Collapse
|
25
|
Jeon J. Review of Therapeutic Applications of Radiolabeled Functional Nanomaterials. Int J Mol Sci 2019; 20:E2323. [PMID: 31083402 PMCID: PMC6539387 DOI: 10.3390/ijms20092323] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 01/10/2023] Open
Abstract
In the last two decades, various nanomaterials have attracted increasing attention in medical science owing to their unique physical and chemical characteristics. Incorporating radionuclides into conventionally used nanomaterials can confer useful additional properties compared to the original material. Therefore, various radionuclides have been used to synthesize functional nanomaterials for biomedical applications. In particular, several α- or β-emitter-labeled organic and inorganic nanoparticles have been extensively investigated for efficient and targeted cancer treatment. This article reviews recent progress in cancer therapy using radiolabeled nanomaterials including inorganic, polymeric, and carbon-based materials and liposomes. We first provide an overview of radiolabeling methods for preparing anticancer agents that have been investigated recently in preclinical studies. Next, we discuss the therapeutic applications and effectiveness of α- or β-emitter-incorporated nanomaterials in animal models and the emerging possibilities of these nanomaterials in cancer therapy.
Collapse
Affiliation(s)
- Jongho Jeon
- Department of Applied Chemistry, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Korea.
| |
Collapse
|
26
|
Kleynhans J, Grobler AF, Ebenhan T, Sathekge MM, Zeevaart JR. Radiopharmaceutical enhancement by drug delivery systems: A review. J Control Release 2018; 287:177-193. [DOI: 10.1016/j.jconrel.2018.08.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022]
|