1
|
Reilly RM, Georgiou CJ, Brown MK, Cai Z. Radiation nanomedicines for cancer treatment: a scientific journey and view of the landscape. EJNMMI Radiopharm Chem 2024; 9:37. [PMID: 38703297 PMCID: PMC11069497 DOI: 10.1186/s41181-024-00266-y] [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: 01/31/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Radiation nanomedicines are nanoparticles labeled with radionuclides that emit α- or β-particles or Auger electrons for cancer treatment. We describe here our 15 years scientific journey studying locally-administered radiation nanomedicines for cancer treatment. We further present a view of the radiation nanomedicine landscape by reviewing research reported by other groups. MAIN BODY Gold nanoparticles were studied initially for radiosensitization of breast cancer to X-radiation therapy. These nanoparticles were labeled with 111In to assess their biodistribution after intratumoural vs. intravenous injection. Intravenous injection was limited by high liver and spleen uptake and low tumour uptake, while intratumoural injection provided high tumour uptake but low normal tissue uptake. Further, [111In]In-labeled gold nanoparticles modified with trastuzumab and injected iintratumourally exhibited strong tumour growth inhibition in mice with subcutaneous HER2-positive human breast cancer xenografts. In subsequent studies, strong tumour growth inhibition in mice was achieved without normal tissue toxicity in mice with human breast cancer xenografts injected intratumourally with gold nanoparticles labeled with β-particle emitting 177Lu and modified with panitumumab or trastuzumab to specifically bind EGFR or HER2, respectively. A nanoparticle depot (nanodepot) was designed to incorporate and deliver radiolabeled gold nanoparticles to tumours using brachytherapy needle insertion techniques. Treatment of mice with s.c. 4T1 murine mammary carcinoma tumours with a nanodepot incorporating [90Y]Y-labeled gold nanoparticles inserted into one tumour arrested tumour growth and caused an abscopal growth-inhibitory effect on a distant second tumour. Convection-enhanced delivery of [177Lu]Lu-AuNPs to orthotopic human glioblastoma multiforme (GBM) tumours in mice arrested tumour growth without normal tissue toxicity. Other groups have explored radiation nanomedicines for cancer treatment in preclinical animal tumour xenograft models using gold nanoparticles, liposomes, block copolymer micelles, dendrimers, carbon nanotubes, cellulose nanocrystals or iron oxide nanoparticles. These nanoparticles were labeled with radionuclides emitting Auger electrons (111In, 99mTc, 125I, 103Pd, 193mPt, 195mPt), β-particles (177Lu, 186Re, 188Re, 90Y, 198Au, 131I) or α-particles (225Ac, 213Bi, 212Pb, 211At, 223Ra). These studies employed intravenous or intratumoural injection or convection enhanced delivery. Local administration of these radiation nanomedicines was most effective and minimized normal tissue toxicity. CONCLUSIONS Radiation nanomedicines have shown great promise for treating cancer in preclinical studies. Local intratumoural administration avoids sequestration by the liver and spleen and is most effective for treating tumours, while minimizing normal tissue toxicity.
Collapse
Affiliation(s)
- Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.
- Princess Margaret Cancer Centre, Toronto, ON, Canada.
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.
- Joint Department of Medical Imaging, University Health Network, Toronto, ON, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| | | | - Madeline K Brown
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
Munekane M, Fuchigami T, Ogawa K. Recent advances in the development of 225Ac- and 211At-labeled radioligands for radiotheranostics. ANAL SCI 2024; 40:803-826. [PMID: 38564087 PMCID: PMC11035452 DOI: 10.1007/s44211-024-00514-w] [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: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 04/04/2024]
Abstract
Radiotheranostics utilizes a set of radioligands incorporating diagnostic or therapeutic radionuclides to achieve both diagnosis and therapy. Imaging probes using diagnostic radionuclides have been used for systemic cancer imaging. Integration of therapeutic radionuclides into the imaging probes serves as potent agents for radionuclide therapy. Among them, targeted alpha therapy (TAT) is a promising next-generation cancer therapy. The α-particles emitted by the radioligands used in TAT result in a high linear energy transfer over a short range, inducing substantial damage to nearby cells surrounding the binding site. Therefore, the key to successful cancer treatment with minimal side effects by TAT depends on the selective delivery of radioligands to their targets. Recently, TAT agents targeting biomolecules highly expressed in various cancer cells, such as sodium/iodide symporter, norepinephrine transporter, somatostatin receptor, αvβ3 integrin, prostate-specific membrane antigen, fibroblast-activation protein, and human epidermal growth factor receptor 2 have been developed and have made remarkable progress toward clinical application. In this review, we focus on two radionuclides, 225Ac and 211At, which are expected to have a wide range of applications in TAT. We also introduce recent fundamental and clinical studies of radiopharmaceuticals labeled with these radionuclides.
Collapse
Affiliation(s)
- Masayuki Munekane
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Takeshi Fuchigami
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
| | - Kazuma Ogawa
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
| |
Collapse
|
3
|
Sakmár M, Ondrák L, Fialová K, Vlk M, Kozempel J, Bruchertseifer F, Morgenstern A. In vitro studies of 223Ra- and 225Ac-labelled α-zirconium phosphate as potential carrier for alpha targeted therapy. J Radioanal Nucl Chem 2023. [DOI: 10.1007/s10967-022-08742-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
AbstractIn this study suitability of α-ZrP nanoparticles as a 223Ra and 225Ac carriers for TAT was investigated. The yields of radiolabelling were higher than 98% in both cases. Subsequently, in vitro stability studies were carried out in various biological matrices during 48 h period. Measurements of released radioactivity showed the highest stability in saline. Released activity of 223Ra, 225Ac and their daughter radionuclides was around 0.5%. On the other hand, the lowest stability was shown in plasma and serum. Released activity for 223Ra, 225Ac and their progeny atoms was from 15 to 32%.
Collapse
|
4
|
Kozempel J, Sakmár M, Janská T, Vlk M. Study of 213Bi and 211Pb Recoils Release from 223Ra Labelled TiO 2 Nanoparticles. MATERIALS (BASEL, SWITZERLAND) 2022; 16:343. [PMID: 36614682 PMCID: PMC9821810 DOI: 10.3390/ma16010343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 06/17/2023]
Abstract
Nanoparticles of various materials were proposed as carriers of nuclides in targeted alpha particle therapy to at least partially eliminate the nuclear recoil effect causing the unwanted release of radioactive progeny originating in nuclear decay series of so-called in vivo generators. Here, we report on the study of 211Pb and 211Bi recoils release from the 223Ra surface-labelled TiO2 nanoparticles in the concentration range of 0.01-1 mg/mL using two phase separation methods different in their kinetics in order to test the ability of progeny resorption. We have found significant differences between the centrifugation and the dialysis used for labelled NPs separation as well as that the release of 211Pb and 211Bi from the nanoparticles also depends on the NPs dispersion concentration. These findings support our previously proposed recoils-retaining mechanism of the progeny by their resorption on the NPs surface. At the 24 h time-point, the highest overall released progeny fractions were observed using centrifugation (4.0% and 13.5% for 211Pb and 211Bi, respectively) at 0.01 mg/mL TiO2 concentration. The lowest overall released fractions at the 24 h time-point (1.5% and 2.5% for 211Pb and 211Bi respectively) were observed using dialysis at 1 mg/mL TiO2 concentration. Our findings also indicate that the in vitro stability tests of such radionuclide systems designed to retain recoil-progeny may end up with biased results and particular care needs to be given to in vitro stability test experimental setup to mimic in vivo dynamic conditions. On the other hand, controlled and well-defined progeny release may enhance the alpha-emitter radiation therapy of some tumours.
Collapse
|
5
|
Katugampola S, Wang J, Prasad A, Sofou S, Howell RW. Predicting response of micrometastases with MIRDcell V3: proof of principle with 225Ac-DOTA encapsulating liposomes that produce different activity distributions in tumor spheroids. Eur J Nucl Med Mol Imaging 2022; 49:3989-3999. [PMID: 35802160 PMCID: PMC9529908 DOI: 10.1007/s00259-022-05878-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/14/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE The spatial distribution of radiopharmaceuticals within multicellular clusters is known to have a significant effect on their biological response. Most therapeutic radiopharmaceuticals distribute nonuniformly in tissues which makes predicting responses of micrometastases challenging. The work presented here analyzes published temporally dependent nonuniform activity distributions within tumor spheroids treated with actinium-225-DOTA encapsulating liposomes (225Ac-liposomes) and uses these data in MIRDcell V3.11 to calculate absorbed dose distributions and predict biological response. The predicted responses are compared with experimental responses. METHODS Four types of liposomes were prepared having membranes with different combinations of release (R) and adhesion (A) properties. The combinations were R-A-, R-A+, R+A-, and R+A+. These afford different penetrating properties into tissue. The liposomes were loaded with either carboxyfluorescein diacetate succinimidyl ester (CFDA-SE) or 225Ac. MDA-MB-231 spheroids were treated with the CFDA-SE-liposomes, harvested at different times, and the time-integrated CFDA-SE concentration at each radial position within the spheroid was determined. This was translated into mean 225Ac decays/cell versus radial position, uploaded to MIRDcell, and the surviving fraction of cells in spherical multicellular clusters was simulated. The MIRDcell-predicted surviving fractions were compared with experimental fractional-outgrowths of the spheroids following treatment with 225Ac-liposomes. RESULTS The biological responses of the multicellular clusters treated with 225Ac-liposomes with physicochemical properties R+A+, R-A+, and R-A- were predicted by MIRDcell with statistically significant accuracy. The prediction for R+A- was not predicted accurately. CONCLUSION In most instances, MIRDcell predicts responses of spheroids treated with 225Ac-liposomes that result in different tissue-penetrating profiles of the delivered radionuclides.
Collapse
Affiliation(s)
- Sumudu Katugampola
- Division of Radiation Research, Department of Radiology and Center for Cell Signaling, New Jersey Medical School, Rutgers University, 205 S. Orange Avenue, Newark, NJ, 07103, USA
| | - Jianchao Wang
- Division of Radiation Research, Department of Radiology and Center for Cell Signaling, New Jersey Medical School, Rutgers University, 205 S. Orange Avenue, Newark, NJ, 07103, USA
| | - Aprameya Prasad
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Stavroula Sofou
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Roger W Howell
- Division of Radiation Research, Department of Radiology and Center for Cell Signaling, New Jersey Medical School, Rutgers University, 205 S. Orange Avenue, Newark, NJ, 07103, USA.
| |
Collapse
|
6
|
Cytryniak A, Żelechowska-Matysiak K, Nazaruk E, Bilewicz R, Walczak R, Majka E, Mames A, Bruchertseifer F, Morgenstern A, Bilewicz A, Majkowska-Pilip A. Cubosomal Lipid Formulation for Combination Cancer Treatment: Delivery of a Chemotherapeutic Agent and Complexed α-Particle Emitter 213Bi. Mol Pharm 2022; 19:2818-2831. [PMID: 35849547 PMCID: PMC9346610 DOI: 10.1021/acs.molpharmaceut.2c00182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Here, we propose tailored lipid liquid-crystalline carriers (cubosomes), which incorporate an anticancer drug (doxorubicin) and complexed short-lived α-emitter (bismuth-213), as a strategy to obtain more effective action toward the cancer cells. Cubosomes were formulated with doxorubicin (DOX) and an amphiphilic ligand (DOTAGA-OA), which forms stable complexes with 213Bi radionuclide. The behavior of DOX incorporated into the carrier together with the chelating agent was investigated, and the drug liberation profile was determined. The experiments revealed that the presence of the DOTAGA-OA ligand affects the activity of DOX when they are incorporated into the same carrier. This unexpected influence was explained based on the results of release studies, which proved the contribution of electrostatics in molecular interactions between the positively charged DOX and negatively charged DOTAGA-OA in acidic and neutral solutions. A significant decrease in the viability of HeLa cancer cells was achieved using sequential cell exposure: first to the radiolabeled cubosomes containing 213Bi complex and next to DOX-doped cubosomes. Therefore, the sequential procedure for the delivery of both drugs encapsulated in cubosomes is suggested for further biological and in vivo studies.
Collapse
Affiliation(s)
- Adrianna Cytryniak
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Kinga Żelechowska-Matysiak
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland
| | - Ewa Nazaruk
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Renata Bilewicz
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Rafał Walczak
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland
| | - Emilia Majka
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland
| | - Adam Mames
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Frank Bruchertseifer
- Directorate for Nuclear Safety and Security, European Commission, Joint Research Centre, Postfach 2340, 76125 Karlsruhe, Germany
| | - Alfred Morgenstern
- Directorate for Nuclear Safety and Security, European Commission, Joint Research Centre, Postfach 2340, 76125 Karlsruhe, Germany
| | - Aleksander Bilewicz
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland
| | - Agnieszka Majkowska-Pilip
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland
| |
Collapse
|
7
|
225Ac-rHDL Nanoparticles: A Potential Agent for Targeted Alpha-Particle Therapy of Tumors Overexpressing SR-BI Proteins. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072156. [PMID: 35408554 PMCID: PMC9000893 DOI: 10.3390/molecules27072156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 11/25/2022]
Abstract
Actinium-225 and other alpha-particle-emitting radionuclides have shown high potential for cancer treatment. Reconstituted high-density lipoproteins (rHDL) specifically recognize the scavenger receptor B type I (SR-BI) overexpressed in several types of cancer cells. Furthermore, after rHDL-SR-BI recognition, the rHDL content is injected into the cell cytoplasm. This research aimed to prepare a targeted 225Ac-delivering nanosystem by encapsulating the radionuclide into rHDL nanoparticles. The synthesis of rHDL was performed in two steps using the microfluidic synthesis method for the subsequent encapsulation of 225Ac, previously complexed to a lipophilic molecule (225Ac-DOTA-benzene-p-SCN, CLog P = 3.42). The nanosystem (13 nm particle size) showed a radiochemical purity higher than 99% and stability in human serum. In vitro studies in HEP-G2 and PC-3 cancer cells (SR-BI positive) demonstrated that 225Ac was successfully internalized into the cytoplasm of cells, delivering high radiation doses to cell nuclei (107 Gy to PC-3 and 161 Gy to HEP-G2 nuclei at 24 h), resulting in a significant decrease in cell viability down to 3.22 ± 0.72% for the PC-3 and to 1.79 ± 0.23% for HEP-G2 at 192 h after 225Ac-rHDL treatment. After intratumoral 225Ac-rHDL administration in mice bearing HEP-G2 tumors, the biokinetic profile showed significant retention of radioactivity in the tumor masses (90.16 ± 2.52% of the injected activity), which generated ablative radiation doses (649 Gy/MBq). The results demonstrated adequate properties of rHDL as a stable carrier for selective deposition of 225Ac within cancer cells overexpressing SR-BI. The results obtained in this research justify further preclinical studies, designed to evaluate the therapeutic efficacy of the 225Ac-rHDL system for targeted alpha-particle therapy of tumors that overexpress the SR-BI receptor.
Collapse
|
8
|
Abstract
8-Hydroxyquinoline (8-HQ, oxine) is a small, monoprotic, bicyclic aromatic compound and its relative donor group orientation imparts impressive bidentate metal chelating abilities that have been exploited in a vast array of applications over decades. 8-HQ and its derivatives have been explored in medicinal applications including anti-neurodegeneration, anticancer properties, and antimicrobial activities. One long established use of 8-HQ in medicinal inorganic chemistry is the coordination of radioactive isotopes of metal ions in nuclear medicine. The metal-oxine complex with the single photon emission computed tomography (SPECT) imaging isotope [111In]In3+ was developed in the 1970s and 1980s to radiolabel leukocytes for inflammation and infection imaging. The [111In][In(oxine)3] complex functions as an ionophore: a moderately stable lipophilic complex to enter cells; however, inside the cell environment [111In]In3+ undergoes exchange and remains localized. As new developments have progressed towards radiopharmaceuticals capable of both imaging and therapy (theranostics), 8-HQ has been re-explored in recent years to investigate its potential to chelate larger radiometal ions with longer half-lives and different indications. Further, metal-oxine complexes have been used to study liposomes and other nanomaterials by tracking these nanomedicines in vivo. Expanding 8-HQ to multidentate ligands for highly thermodynamically stable and kinetically inert complexes has increased the possibilities of this small molecule in nuclear medicine. This article outlines the historic use of metal-oxine complexes in inorganic radiopharmaceutical chemistry, with a focus on recent advances highlighting the possibilities of developing higher denticity, targeted bifunctional chelators with 8-HQ.
Collapse
Affiliation(s)
- Lily Southcott
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia, V6T 2A3, Canada.,Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.
| |
Collapse
|
9
|
Deblonde GJP, Zavarin M, Kersting AB. The coordination properties and ionic radius of actinium: A 120-year-old enigma. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
10
|
Kleynhans J, Sathekge M, Ebenhan T. Obstacles and Recommendations for Clinical Translation of Nanoparticle System-Based Targeted Alpha-Particle Therapy. MATERIALS (BASEL, SWITZERLAND) 2021; 14:4784. [PMID: 34500873 PMCID: PMC8432563 DOI: 10.3390/ma14174784] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/31/2022]
Abstract
The rationale for application of nanotechnology in targeted alpha therapy (TAT) is sound. However, the translational strategy requires attention. Formulation of TAT in nanoparticulate drug delivery systems has the potential to resolve many of the issues currently experienced. As α-particle emitters are more cytotoxic compared to beta-minus-emitting agents, the results of poor biodistribution are more dangerous. Formulation in nanotechnology is also suggested to be the ideal solution for containing the recoil daughters emitted by actinium-225, radium-223, and thorium-227. Nanoparticle-based TAT is likely to increase stability, enhance radiation dosimetry profiles, and increase therapeutic efficacy. Unfortunately, nanoparticles have their own unique barriers towards clinical translation. A major obstacle is accumulation in critical organs such as the spleen, liver, and lungs. Furthermore, inflammation, necrosis, reactive oxidative species, and apoptosis are key mechanisms through which nanoparticle-mediated toxicity takes place. It is important at this stage of the technology's readiness level that focus is shifted to clinical translation. The relative scarcity of α-particle emitters also contributes to slow-moving research in the field of TAT nanotechnology. This review describes approaches and solutions which may overcome obstacles impeding nanoparticle-based TAT and enhance clinical translation. In addition, an in-depth discussion of relevant issues and a view on technical and regulatory barriers are presented.
Collapse
Affiliation(s)
- Janke Kleynhans
- Division of Nuclear Medicine, Tygerberg Hospital, Stellenbosch University, Cape Town 8000, South Africa;
| | - Mike Sathekge
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Nuclear Medicine Research Infrastructure NPC, Pretoria 0001, South Africa
| | - Thomas Ebenhan
- Nuclear Medicine Research Infrastructure NPC, Pretoria 0001, South Africa
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0001, South Africa
| |
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
|
Choi JS, Park JW, Seu YB, Doh KO. Enhanced efficacy of folate-incorporated cholesteryl doxorubicin liposome in folate receptor abundant cancer cell. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
13
|
Muslimov AR, Antuganov D, Tarakanchikova YV, Karpov TE, Zhukov MV, Zyuzin MV, Timin AS. An investigation of calcium carbonate core-shell particles for incorporation of 225Ac and sequester of daughter radionuclides: in vitro and in vivo studies. J Control Release 2021; 330:726-737. [DOI: 10.1016/j.jconrel.2021.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/04/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023]
|
14
|
Pellico J, Gawne PJ, T M de Rosales R. Radiolabelling of nanomaterials for medical imaging and therapy. Chem Soc Rev 2021; 50:3355-3423. [PMID: 33491714 DOI: 10.1039/d0cs00384k] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanomaterials offer unique physical, chemical and biological properties of interest for medical imaging and therapy. Over the last two decades, there has been an increasing effort to translate nanomaterial-based medicinal products (so-called nanomedicines) into clinical practice and, although multiple nanoparticle-based formulations are clinically available, there is still a disparity between the number of pre-clinical products and those that reach clinical approval. To facilitate the efficient clinical translation of nanomedicinal-drugs, it is important to study their whole-body biodistribution and pharmacokinetics from the early stages of their development. Integrating this knowledge with that of their therapeutic profile and/or toxicity should provide a powerful combination to efficiently inform nanomedicine trials and allow early selection of the most promising candidates. In this context, radiolabelling nanomaterials allows whole-body and non-invasive in vivo tracking by the sensitive clinical imaging techniques positron emission tomography (PET), and single photon emission computed tomography (SPECT). Furthermore, certain radionuclides with specific nuclear emissions can elicit therapeutic effects by themselves, leading to radionuclide-based therapy. To ensure robust information during the development of nanomaterials for PET/SPECT imaging and/or radionuclide therapy, selection of the most appropriate radiolabelling method and knowledge of its limitations are critical. Different radiolabelling strategies are available depending on the type of material, the radionuclide and/or the final application. In this review we describe the different radiolabelling strategies currently available, with a critical vision over their advantages and disadvantages. The final aim is to review the most relevant and up-to-date knowledge available in this field, and support the efficient clinical translation of future nanomedicinal products for in vivo imaging and/or therapy.
Collapse
Affiliation(s)
- Juan Pellico
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK.
| | | | | |
Collapse
|
15
|
Wang LZ, Lim TL, Padakanti PK, Carlin SD, Alavi A, Mach RH, Prud’homme RK. Kinetics of Nanoparticle Radiolabeling of Metalloporphyrin with 64Cu for Positron Emission Tomography (PET) Imaging. Ind Eng Chem Res 2020. [DOI: 10.1021/acs.iecr.0c03671] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Leon Z. Wang
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Tristan L. Lim
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Prashanth K. Padakanti
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sean D. Carlin
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Abass Alavi
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Robert H. Mach
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Robert K. Prud’homme
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
16
|
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
|
17
|
Silindir-Gunay M, Karpuz M, Ozer AY. Targeted Alpha Therapy and Nanocarrier Approach. Cancer Biother Radiopharm 2020; 35:446-458. [DOI: 10.1089/cbr.2019.3213] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Mine Silindir-Gunay
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Merve Karpuz
- Department of Radiopharmacy, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| | - A. Yekta Ozer
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Jadvar H. Targeted α-Therapy in Cancer Management: Synopsis of Preclinical and Clinical Studies. Cancer Biother Radiopharm 2020; 35:475-484. [PMID: 32202923 DOI: 10.1089/cbr.2019.3340] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The approval of 223Ra dichloride (223RaCl2) in 2013 was a principal event in introducing targeted α-therapy as a form of safe and effective management strategy in cancer. There is an increasing interest in research and development of new targeted α-therapy agents spearheaded by advancements in cancer biology, radiochemistry, and availability of clinically relevant α particles. There are active clinical studies on sequencing or combining 223RaCl2 with other drug regimens in the setting of metastatic prostate cancer and in other cancers such as osteosarcoma and bone-dominant breast cancer. Targeted α-therapy strategy is also being actively explored through many preclinical and few early clinical studies using 225Ac, 213Bi, 211At, 227Th, and 212Pb. Investigations incorporating 225Ac are more robust and active at this time with promising results. The author provide a brief synopsis of the preclinical and clinical studies in the rapidly evolving field of targeted α-therapy in cancer management.
Collapse
Affiliation(s)
- Hossein Jadvar
- Division of Nuclear Medicine, Department of Radiology, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
21
|
Tafreshi NK, Doligalski ML, Tichacek CJ, Pandya DN, Budzevich MM, El-Haddad G, Khushalani NI, Moros EG, McLaughlin ML, Wadas TJ, Morse DL. Development of Targeted Alpha Particle Therapy for Solid Tumors. Molecules 2019; 24:molecules24234314. [PMID: 31779154 PMCID: PMC6930656 DOI: 10.3390/molecules24234314] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022] Open
Abstract
Targeted alpha-particle therapy (TAT) aims to selectively deliver radionuclides emitting α-particles (cytotoxic payload) to tumors by chelation to monoclonal antibodies, peptides or small molecules that recognize tumor-associated antigens or cell-surface receptors. Because of the high linear energy transfer (LET) and short range of alpha (α) particles in tissue, cancer cells can be significantly damaged while causing minimal toxicity to surrounding healthy cells. Recent clinical studies have demonstrated the remarkable efficacy of TAT in the treatment of metastatic, castration-resistant prostate cancer. In this comprehensive review, we discuss the current consensus regarding the properties of the α-particle-emitting radionuclides that are potentially relevant for use in the clinic; the TAT-mediated mechanisms responsible for cell death; the different classes of targeting moieties and radiometal chelators available for TAT development; current approaches to calculating radiation dosimetry for TATs; and lead optimization via medicinal chemistry to improve the TAT radiopharmaceutical properties. We have also summarized the use of TATs in pre-clinical and clinical studies to date.
Collapse
Affiliation(s)
- Narges K. Tafreshi
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Michael L. Doligalski
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Christopher J. Tichacek
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Darpan N. Pandya
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA; (D.N.P.); (T.J.W.)
| | - Mikalai M. Budzevich
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Ghassan El-Haddad
- Depts. of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Nikhil I. Khushalani
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Eduardo G. Moros
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Department of Physics, University of South Florida, Tampa, FL 33612, USA
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Mark L. McLaughlin
- Department of Pharmaceutical Sciences, West Virginia University, Health Sciences Center, Morgantown, WV & Modulation Therapeutics Inc., 64 Medical Center Drive, Morgantown, WV 26506, USA;
| | - Thaddeus J. Wadas
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA; (D.N.P.); (T.J.W.)
| | - David L. Morse
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
- Department of Physics, University of South Florida, Tampa, FL 33612, USA
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
- Correspondence: ; Tel.: +1-813-745-8948; Fax: +1-813-745-8375
| |
Collapse
|
22
|
Alpha-Emitters and Targeted Alpha Therapy in Oncology: from Basic Science to Clinical Investigations. Target Oncol 2019; 13:189-203. [PMID: 29423595 DOI: 10.1007/s11523-018-0550-9] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Alpha-emitters are radionuclides that decay through the emission of high linear energy transfer α-particles and possess favorable pharmacologic profiles for cancer treatment. When coupled with monoclonal antibodies, peptides, small molecules, or nanoparticles, the excellent cytotoxic capability of α-particle emissions has generated a strong interest in exploring targeted α-therapy in the pre-clinical setting and more recently in clinical trials in oncology. Multiple obstacles have been overcome by researchers and clinicians to accelerate the development of targeted α-therapies, especially with the recent improvement in isotope production and purification, but also with the development of innovative strategies for optimized targeting. Numerous studies have demonstrated the in vitro and in vivo efficacy of the targeted α-therapy. Radium-223 (223Ra) dichloride (Xofigo®) is the first α-emitter to have received FDA approval for the treatment of prostate cancer with metastatic bone lesions. There is a significant increase in the number of clinical trials in oncology using several radionuclides such as Actinium-225 (225Ac), Bismuth-213 (213Bi), Lead-212 (212Pb), Astatine (211At) or Radium-223 (223Ra) assessing their safety and preliminary activity. This review will cover their therapeutic application as well as summarize the investigations that provide the foundation for further clinical development.
Collapse
|
23
|
Hu G, Guo M, Xu J, Wu F, Fan J, Huang Q, Yang G, Lv Z, Wang X, Jin Y. Nanoparticles Targeting Macrophages as Potential Clinical Therapeutic Agents Against Cancer and Inflammation. Front Immunol 2019; 10:1998. [PMID: 31497026 PMCID: PMC6712945 DOI: 10.3389/fimmu.2019.01998] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/07/2019] [Indexed: 12/11/2022] Open
Abstract
With the development of nanotechnology, significant progress has been made in the design, and manufacture of nanoparticles (NPs) for use in clinical treatments. Recent increases in our understanding of the central role of macrophages in the context of inflammation and cancer have reinvigorated interest in macrophages as drug targets. Macrophages play an integral role in maintaining the steady state of the immune system and are involved in cancer and inflammation processes. Thus, NPs tailored to accurately target macrophages have the potential to transform disease treatment. Herein, we first present a brief background information of NPs as drug carriers, including but not limited to the types of nanomaterials, their biological properties and their advantages in clinical application. Then, macrophage effector mechanisms and recent NPs-based strategies aimed at targeting macrophages by eliminating or re-educating macrophages in inflammation and cancer are summarized. Additionally, the development of nanocarriers targeting macrophages for disease diagnosis is also discussed. Finally, the significance of macrophage-targeting nanomedicine is highlighted, with the goal of facilitating future clinical translation.
Collapse
Affiliation(s)
- Guorong Hu
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Mengfei Guo
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Juanjuan Xu
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wu
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jinshuo Fan
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Huang
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Guanghai Yang
- Department of Thoracic Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhilei Lv
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wang
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Jin
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
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: 41] [Impact Index Per Article: 6.8] [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
|
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
|
Kowalik M, Masternak J, Barszcz B. Recent Research Trends on Bismuth Compounds in Cancer Chemoand Radiotherapy. Curr Med Chem 2019; 26:729-759. [DOI: 10.2174/0929867324666171003113540] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 12/15/2022]
Abstract
Background:Application of coordination chemistry in nanotechnology is a rapidly developing research field in medicine. Bismuth complexes have been widely used in biomedicine with satisfactory therapeutic effects, mostly in Helicobacter pylori eradication, but also as potential antimicrobial and anti-leishmanial agents. Additionally, in recent years, application of bismuth-based compounds as potent anticancer drugs has been studied extensively.Methods:Search for data connected with recent trends on bismuth compounds in cancer chemo- and radiotherapy was carried out using web-based literature searching tools such as ScienceDirect, Springer, Royal Society of Chemistry, American Chemical Society and Wiley. Pertinent literature is covered up to 2016.Results:In this review, based on 213 papers, we highlighted a number of current problems connected with: (i) characterization of bismuth complexes with selected thiosemicarbazone, hydrazone, and dithiocarbamate classes of ligands as potential chemotherapeutics. Literature results derived from 50 papers show that almost all bismuth compounds inhibit growth and proliferation of breast, colon, ovarian, lung, and other tumours; (ii) pioneering research on application of bismuth-based nanoparticles and nanodots for radiosensitization. Results show great promise for improvement in therapeutic efficacy of ionizing radiation in advanced radiotherapy (described in 36 papers); and (iii) research challenges in using bismuth radionuclides in targeted radioimmunotherapy, connected with choice of adequate radionuclide, targeting vector, proper bifunctional ligand and problems with 213Bi recoil daughters toxicity (derived from 92 papers).Conclusion:This review presents recent research trends on bismuth compounds in cancer chemo- and radiotherapy, suggesting directions for future research.
Collapse
Affiliation(s)
- Mateusz Kowalik
- Institute of Chemistry, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Joanna Masternak
- Institute of Chemistry, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Barbara Barszcz
- Institute of Chemistry, Jan Kochanowski University in Kielce, Kielce, Poland
| |
Collapse
|
27
|
Man F, Gawne PJ, T M de Rosales R. Nuclear imaging of liposomal drug delivery systems: A critical review of radiolabelling methods and applications in nanomedicine. Adv Drug Deliv Rev 2019; 143:134-160. [PMID: 31170428 PMCID: PMC6866902 DOI: 10.1016/j.addr.2019.05.012] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/25/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022]
Abstract
The integration of nuclear imaging with nanomedicine is a powerful tool for efficient development and clinical translation of liposomal drug delivery systems. Furthermore, it may allow highly efficient imaging-guided personalised treatments. In this article, we critically review methods available for radiolabelling liposomes. We discuss the influence that the radiolabelling methods can have on their biodistribution and highlight the often-overlooked possibility of misinterpretation of results due to decomposition in vivo. We stress the need for knowing the biodistribution/pharmacokinetics of both the radiolabelled liposomal components and free radionuclides in order to confidently evaluate the images, as they often share excretion pathways with intact liposomes (e.g. phospholipids, metallic radionuclides) and even show significant tumour uptake by themselves (e.g. some radionuclides). Finally, we describe preclinical and clinical studies using radiolabelled liposomes and discuss their impact in supporting liposomal drug development and clinical translation in several diseases, including personalised nanomedicine approaches.
Collapse
Affiliation(s)
- Francis Man
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Peter J Gawne
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom; London Centre for Nanotechnology, King's College London, Strand Campus, London WC2R 2LS, United Kingdom.
| |
Collapse
|
28
|
Farzin L, Sheibani S, Moassesi ME, Shamsipur M. An overview of nanoscale radionuclides and radiolabeled nanomaterials commonly used for nuclear molecular imaging and therapeutic functions. J Biomed Mater Res A 2018; 107:251-285. [PMID: 30358098 DOI: 10.1002/jbm.a.36550] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/08/2018] [Accepted: 09/03/2018] [Indexed: 02/06/2023]
Abstract
Recent advances in the field of nanotechnology applications in nuclear medicine offer the promise of better diagnostic and therapeutic options. In recent years, increasing efforts have been focused on developing nanoconstructs that can be used as core platforms for attaching medical radionuclides with different strategies for the purposes of molecular imaging and targeted drug delivery. This review article presents an introduction to some commonly used nanomaterials with zero-dimensional, one-dimensional, two-dimensional, and three-dimensional structures, describes the various methods applied to radiolabeling of nanomaterials, and provides illustrative examples of application of the nanoscale radionuclides or radiolabeled nanocarriers in nuclear nanomedicine. Especially, the passive and active nanotargeting delivery of radionuclides with illustrating examples for tumor imaging and therapy was reviewed and summarized. The accurate and early diagnosis of cancer can lead to increased survival rates for different types of this disease. Although, the conventional single-modality diagnostic methods such as positron emission tomography/single photon emission computed tomography or MRI used for such purposes are powerful means; most of these are limited by sensitivity or resolution. By integrating complementary signal reporters into a single nanoparticulate contrast agent, multimodal molecular imaging can be performed as scalable images with high sensitivity, resolution, and specificity. The advent of radiolabeled nanocarriers or radioisotope-loaded nanomaterials with magnetic, plasmonic, or fluorescent properties has stimulated growing interest in the developing multimodality imaging probes. These new developments in nuclear nanomedicine are expected to introduce a paradigm shift in multimodal molecular imaging and thereby opening up an era of new diagnostic medical imaging agents. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 251-285, 2019.
Collapse
Affiliation(s)
- Leila Farzin
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| | - Shahab Sheibani
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| | - Mohammad Esmaeil Moassesi
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| | | |
Collapse
|
29
|
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]
|
30
|
Arms L, Smith DW, Flynn J, Palmer W, Martin A, Woldu A, Hua S. Advantages and Limitations of Current Techniques for Analyzing the Biodistribution of Nanoparticles. Front Pharmacol 2018; 9:802. [PMID: 30154715 PMCID: PMC6102329 DOI: 10.3389/fphar.2018.00802] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/03/2018] [Indexed: 12/22/2022] Open
Abstract
Nanomedicines are typically submicrometer-sized carrier materials (nanoparticles) encapsulating therapeutic and/or imaging compounds that are used for the prevention, diagnosis and treatment of diseases. They are increasingly being used to overcome biological barriers in the body to improve the way we deliver compounds to specific tissues and organs. Nanomedicine technology aims to improve the balance between the efficacy and the toxicity of therapeutic compounds. Nanoparticles, one of the key technologies of nanomedicine, can exhibit a combination of physical, chemical and biological characteristics that determine their in vivo behavior. A key component in the translational assessment of nanomedicines is determining the biodistribution of the nanoparticles following in vivo administration in animals and humans. There are a range of techniques available for evaluating nanoparticle biodistribution, including histology, electron microscopy, liquid scintillation counting (LSC), indirectly measuring drug concentrations, in vivo optical imaging, computed tomography (CT), magnetic resonance imaging (MRI), and nuclear medicine imaging. Each technique has its own advantages and limitations, as well as capabilities for assessing real-time, whole-organ and cellular accumulation. This review will address the principles and methodology of each technique and their advantages and limitations for evaluating in vivo biodistribution of nanoparticles.
Collapse
Affiliation(s)
- Lauren Arms
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Doug W. Smith
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Jamie Flynn
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - William Palmer
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Antony Martin
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Ameha Woldu
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Susan Hua
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
31
|
Abstract
α-Particle irradiation of cancerous tissue is increasingly recognized as a potent therapeutic option. We briefly review the physics, radiobiology, and dosimetry of α-particle emitters, as well as the distinguishing features that make them unique for radiopharmaceutical therapy. We also review the emerging clinical role of α-particle therapy in managing cancer and recent studies on in vitro and preclinical α-particle therapy delivered by antibodies, other small molecules, and nanometer-sized particles. In addition to their unique radiopharmaceutical characteristics, the increased availability and improved radiochemistry of α-particle radionuclides have contributed to the growing recent interest in α-particle radiotherapy. Targeted therapy strategies have presented novel possibilities for the use of α-particles in the treatment of cancer. Clinical experience has already demonstrated the safe and effective use of α-particle emitters as potent tumor-selective drugs for the treatment of leukemia and metastatic disease.
Collapse
Affiliation(s)
- Michael R McDevitt
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - George Sgouros
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Stavroula Sofou
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| |
Collapse
|
32
|
Holzwarth U, Ojea Jimenez I, Calzolai L. A random walk approach to estimate the confinement of α-particle emitters in nanoparticles for targeted radionuclide therapy. EJNMMI Radiopharm Chem 2018; 3:9. [PMID: 29888318 PMCID: PMC5976682 DOI: 10.1186/s41181-018-0042-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 03/28/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Targeted radionuclide therapy is a highly efficient but still underused treatment modality for various types of cancers that uses so far mainly readily available β-emitting radionuclides. By using α-particle emitters several shortcomings due to hypoxia, cell proliferation and in the selected treatment of small volumes such as micrometastasis could be overcome. To enable efficient targeting longer-lived α-particle emitters are required. These are the starting point of decay chains emitting several α-particles delivering extremely high radiation doses into small treatment volumes. However, as a consequence of the α-decay the daughter nuclides receive high recoil energies that cannot be managed by chemical radiolabelling techniques. By safe encapsulation of all α-emitters in the decay chain in properly sized nanocarriers their release may be avoided. RESULTS The encapsulation of small core nanoparticles loaded with the radionuclide in a shell structure that safely confines the recoiling daughter nuclides promises good tumour targeting, penetration and uptake, provided these nanostructures can be kept small enough. A model for spherical nanoparticles is proposed that allows an estimate of the fraction of recoiling α-particle emitters that may escape from the nanoparticles as a function of their size. The model treats the recoil ranges of the daughter nuclides as approximately equidistant steps with arbitrary orientation in a three-dimensional random walk model. CONCLUSIONS The presented model allows an estimate of the fraction of α-particles that are emitted from outside the nanoparticle when its size is reduced below the radius that guarantees complete confinement of all radioactive daughter nuclides. Smaller nanoparticle size with reduced retention of daughter radionuclides might be tolerated when the effects can be compensated by fast internalisation of the nanoparticles by the target cells.
Collapse
Affiliation(s)
- Uwe Holzwarth
- European Commission, Joint Research Centre, Via Enrico Fermi 2749, 21027 Ispra, VA Italy
| | - Isaac Ojea Jimenez
- European Commission, Joint Research Centre, Via Enrico Fermi 2749, 21027 Ispra, VA Italy
| | - Luigi Calzolai
- European Commission, Joint Research Centre, Via Enrico Fermi 2749, 21027 Ispra, VA Italy
| |
Collapse
|
33
|
Cędrowska E, Pruszynski M, Majkowska-Pilip A, Męczyńska-Wielgosz S, Bruchertseifer F, Morgenstern A, Bilewicz A. Functionalized TiO 2 nanoparticles labelled with 225Ac for targeted alpha radionuclide therapy. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2018; 20:83. [PMID: 29576738 PMCID: PMC5861168 DOI: 10.1007/s11051-018-4181-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/28/2018] [Indexed: 06/08/2023]
Abstract
The 225Ac radioisotope exhibits very attractive nuclear properties for application in radionuclide therapy. Unfortunately, the major challenge for radioconjugates labelled with 225Ac is that traditional chelating moieties are unable to sequester the radioactive daughters in the bioconjugate which is critical to minimize toxicity to healthy, non-targeted tissues. In the present work, we propose to apply TiO2 nanoparticles (NPs) as carrier for 225Ac and its decay products. The surface of TiO2 nanoparticles with 25 nm diameter was modified with Substance P (5-11), a peptide fragment which targets NK1 receptors on the glioma cells, through the silan-PEG-NHS linker. Nanoparticles functionalized with Substance P (5-11) were synthesized with high yield in a two-step procedure, and the products were characterized by transmission electron microscopy (TEM), dynamic light scattering (DLS) and thermogravimetric analysis (TGA). The obtained results show that one TiO2-bioconjugate nanoparticle contains in average 80 peptide molecules on its surface. The synthesized TiO2-PEG-SP(5-11) conjugates were labelled with 225Ac by ion-exchange reaction on hydroxyl (OH) functional groups on the TiO2 surface. The labelled bioconjugates almost quantitatively retain 225Ac in phosphate-buffered saline (PBS), physiological salt and cerebrospinal fluid (CSF) for up to 10 days. The leaching of 221Fr, a first decay daughter of 225Ac, in an amount of 30% was observed only in CSF after 10 days. The synthesized 225Ac-TiO2-PEG-SP(5-11) has shown high cytotoxic effect in vitro in T98G glioma cells; therefore, it is a promising new radioconjugate for targeted radionuclide therapy of brain tumours.
Collapse
Affiliation(s)
- Edyta Cędrowska
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland
| | - Marek Pruszynski
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland
| | | | | | - Frank Bruchertseifer
- European Commission, Joint Research Centre, Department for Nuclear Safety and Security, 76125 Karlsruhe, Germany
| | - Alfred Morgenstern
- European Commission, Joint Research Centre, Department for Nuclear Safety and Security, 76125 Karlsruhe, Germany
| | - Aleksander Bilewicz
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland
| |
Collapse
|
34
|
Progress in Targeted Alpha-Particle Therapy. What We Learned about Recoils Release from In Vivo Generators. Molecules 2018; 23:molecules23030581. [PMID: 29510568 PMCID: PMC6017877 DOI: 10.3390/molecules23030581] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 12/02/2022] Open
Abstract
This review summarizes recent progress and developments as well as the most important pitfalls in targeted alpha-particle therapy, covering single alpha-particle emitters as well as in vivo alpha-particle generators. It discusses the production of radionuclides like 211At, 223Ra, 225Ac/213Bi, labelling and delivery employing various targeting vectors (small molecules, chelators for alpha-emitting nuclides and their biomolecular targets as well as nanocarriers), general radiopharmaceutical issues, preclinical studies, and clinical trials including the possibilities of therapy prognosis and follow-up imaging. Special attention is given to the nuclear recoil effect and its impacts on the possible use of alpha emitters for cancer treatment, proper dose estimation, and labelling chemistry. The most recent and important achievements in the development of alpha emitters carrying vectors for preclinical and clinical use are highlighted along with an outlook for future developments.
Collapse
|
35
|
Lu HD, Wang LZ, Wilson BK, McManus SA, Jumai'an J, Padakanti PK, Alavi A, Mach RH, Prud'homme RK. Copper Loading of Preformed Nanoparticles for PET-Imaging Applications. ACS APPLIED MATERIALS & INTERFACES 2018; 10:3191-3199. [PMID: 29272577 DOI: 10.1021/acsami.7b07242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Nanoparticles (NP) are promising contrast agents for positron emission tomography (PET) radionuclide imaging that can increase signal intensity by localizing clusters of PET radionuclides together. However, methods to load NPs with PET radionuclides suffer from harsh loading conditions or poor loading efficacies or result in NP surface modifications that alter targeting in vivo. We present the formation of water-dispersible, polyethylene glycol coated NPs that encapsulate phthalocyanines into NP cores at greater than 50 wt % loading, using the self-assembly technique Flash NanoPrecipitation. Particles from 70 to 160 nm are produced. Phthalocyanine NPs rapidly and spontaneously chelate metals under mild conditions and can act as sinks for PET radionuclides such as 64-Cu to produce PET-active NPs. NPs chelate copper(II) with characteristic rates of 1845 M-1 h-1 at pH 6 and 37 °C, which produced >90% radionuclide chelation within 1 h. NP physical properties, such as core composition, core fluidity, and size, can be tuned to modulate chelation kinetics. These NPs retain 64Cu even in the presence of the strong chelator ethylene diamine tetraacetic acid. The development of these constructs for rapid and facile radionuclide labeling expands the applications of NP-based PET imaging.
Collapse
Affiliation(s)
- Hoang D Lu
- Department of Chemical and Biological Engineering, Princeton University , Princeton, New Jersey 08544, United States
| | - Leon Z Wang
- Department of Chemical and Biological Engineering, Princeton University , Princeton, New Jersey 08544, United States
| | - Brian K Wilson
- Department of Chemical and Biological Engineering, Princeton University , Princeton, New Jersey 08544, United States
| | - Simon A McManus
- Department of Chemical and Biological Engineering, Princeton University , Princeton, New Jersey 08544, United States
| | - Jenny Jumai'an
- Department of Chemical and Biological Engineering, Princeton University , Princeton, New Jersey 08544, United States
| | - Prashanth K Padakanti
- Department of Radiology, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Abass Alavi
- Department of Radiology, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Robert H Mach
- Department of Radiology, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Robert K Prud'homme
- Department of Chemical and Biological Engineering, Princeton University , Princeton, New Jersey 08544, United States
| |
Collapse
|
36
|
Toro-González M, Copping R, Mirzadeh S, Rojas JV. Multifunctional GdVO4:Eu core–shell nanoparticles containing 225Ac for targeted alpha therapy and molecular imaging. J Mater Chem B 2018; 6:7985-7997. [DOI: 10.1039/c8tb02173b] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Development of actinium-225 doped Gd0.8Eu0.2VO4 core–shell nanoparticles as multifunctional platforms for multimodal molecular imaging and targeted radionuclide therapy.
Collapse
Affiliation(s)
- M. Toro-González
- Department of Mechanical and Nuclear Engineering
- Virginia Commonwealth University
- Richmond
- USA
| | - R. Copping
- Nuclear Security and Isotope Technology Division
- Oak Ridge National Laboratory
- Oak Ridge
- USA
| | - S. Mirzadeh
- Nuclear Security and Isotope Technology Division
- Oak Ridge National Laboratory
- Oak Ridge
- USA
| | - J. V. Rojas
- Department of Mechanical and Nuclear Engineering
- Virginia Commonwealth University
- Richmond
- USA
| |
Collapse
|
37
|
de Kruijff RM, Drost K, Thijssen L, Morgenstern A, Bruchertseifer F, Lathouwers D, Wolterbeek HT, Denkova AG. Improved 225Ac daughter retention in InPO 4 containing polymersomes. Appl Radiat Isot 2017; 128:183-189. [PMID: 28734193 DOI: 10.1016/j.apradiso.2017.07.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 01/19/2023]
Abstract
Alpha-emitting radionuclides like actinium-225 (225Ac) are ideal candidates for the treatment of small metastasised tumours, where the long half-life of 225Ac enables it to also reach less accessible tumours. The main challenge lies in retaining the recoiled alpha-emitting daughter nuclides, which are decoupled from targeting agents upon emission of an alpha particle and can subsequently cause unwanted toxicity to healthy tissue. Polymersomes, vesicles composed of amphiphilic block copolymers, are capable of transporting (radio)pharmaceuticals to tumours, and are ideal candidates for the retention of these daughter nuclides. In this study, the Geant4 Monte Carlo simulation package was used to simulate ideal vesicle designs. Vesicles containing an InPO4 nanoparticle in the core were found to have the highest recoil retention, and were subsequently synthesized in the lab. The recoil retention of two of the daughter nuclides, namely francium-221 (221Fr) and bismuth-213 (213Bi) was determined at different vesicle sizes. Recoil retention was found to have improved significantly, from 37 ± 4% and 22 ± 1% to 57 ± 5% and 40 ± 2% for 221Fr and 213Bi respectively for 100nm polymersomes, as compared to earlier published results by Wang et al. where 225Ac was encapsulated using a hydrophilic chelate (Wang et al. 2014). To better understand the different parameters influencing daughter retention, simulation data was expanded to include vesicle polydispersity and nanoparticle position within the polymersome. The high retention of the recoiling daughters and the 225Ac itself makes this vesicle design very suitable for future in vivo verification.
Collapse
Affiliation(s)
- R M de Kruijff
- Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629 JB Delft, The Netherlands.
| | - K Drost
- Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629 JB Delft, The Netherlands
| | - L Thijssen
- SynerScope, Kastanjelaan 14, 5268 CA Helvoirt, The Netherlands
| | - A Morgenstern
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, P.O. Box 2340, 76125 Karlsruhe, Germany
| | - F Bruchertseifer
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, P.O. Box 2340, 76125 Karlsruhe, Germany
| | - D Lathouwers
- Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629 JB Delft, The Netherlands
| | - H T Wolterbeek
- Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629 JB Delft, The Netherlands
| | - A G Denkova
- Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629 JB Delft, The Netherlands.
| |
Collapse
|
38
|
Alpha-particle radiotherapy: For large solid tumors diffusion trumps targeting. Biomaterials 2017; 130:67-75. [DOI: 10.1016/j.biomaterials.2017.03.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/20/2017] [Accepted: 03/23/2017] [Indexed: 12/29/2022]
|
39
|
Pant K, Sedláček O, Nadar RA, Hrubý M, Stephan H. Radiolabelled Polymeric Materials for Imaging and Treatment of Cancer: Quo Vadis? Adv Healthc Mater 2017; 6. [PMID: 28218487 DOI: 10.1002/adhm.201601115] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/24/2016] [Indexed: 12/15/2022]
Abstract
Owing to their tunable blood circulation time and suitable plasma stability, polymer-based nanomaterials hold a great potential for designing and utilising multifunctional nanocarriers for efficient imaging and effective treatment of cancer. When tagged with appropriate radionuclides, they may allow for specific detection (diagnosis) as well as the destruction of tumours (therapy) or even customization of materials, aiming to both diagnosis and therapy (theranostic approach). This review provides an overview of recent developments of radiolabelled polymeric nanomaterials (natural and synthetic polymers) for molecular imaging of cancer, specifically, applying nuclear techniques such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT). Different approaches to radiolabel polymers are evaluated from the methodical radiochemical point of view. This includes new bifunctional chelating agents (BFCAs) for radiometals as well as novel labelling methods. Special emphasis is given to eligible strategies employed to evade the mononuclear phagocytic system (MPS) in view of efficient targeting. The discussion encompasses promising strategies currently employed as well as emerging possibilities in radionuclide-based cancer therapy. Key issues involved in the clinical translation of radiolabelled polymers and future scopes of this intriguing research field are also discussed.
Collapse
Affiliation(s)
- Kritee Pant
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| | - Ondřej Sedláček
- Institute of Macromolecular Chemistry; The Academy of Sciences of the Czech Republic; Heyrovského námeˇstí 2 16206 Prague 6 Czech Republic
| | - Robin A. Nadar
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| | - Martin Hrubý
- Institute of Macromolecular Chemistry; The Academy of Sciences of the Czech Republic; Heyrovského námeˇstí 2 16206 Prague 6 Czech Republic
| | - Holger Stephan
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| |
Collapse
|
40
|
van der Geest T, Laverman P, Metselaar JM, Storm G, Boerman OC. Radionuclide imaging of liposomal drug delivery. Expert Opin Drug Deliv 2016; 13:1231-42. [PMID: 27351233 DOI: 10.1080/17425247.2016.1205584] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Ever since their discovery, liposomes have been radiolabeled to monitor their fate in vivo. Despite extensive preclinical studies, only a limited number of radiolabeled liposomal formulations have been examined in patients. Since they can play a crucial role in patient management, it is of importance to enable translation of radiolabeled liposomes into the clinic. AREAS COVERED Liposomes have demonstrated substantial advantages as drug delivery systems and can be efficiently radiolabeled. Potentially, radiolabeled drug-loaded liposomes form an elegant theranostic system, which can be tracked in vivo using single-photon emission computed tomography (SPECT) or positron emission tomography (PET) imaging. In this review, we discuss important aspects of liposomal research with a focus on the use of radiolabeled liposomes and their potential role in drug delivery and monitoring therapeutic effects. EXPERT OPINION Radiolabeled drug-loaded liposomes have been poorly investigated in patients and no radiolabeled liposomes have been approved for use in clinical practice. Evaluation of the risks, pharmacokinetics, pharmacodynamics and toxicity is necessary to meet pharmaceutical and commercial requirements. It remains to be demonstrated whether the results found in animal studies translate to humans before radiolabeled liposomes can be implemented into clinical practice.
Collapse
Affiliation(s)
- Tessa van der Geest
- a Department of Radiology and Nuclear Medicine , Radboud University Medical Center , Nijmegen , The Netherlands
| | - Peter Laverman
- a Department of Radiology and Nuclear Medicine , Radboud University Medical Center , Nijmegen , The Netherlands
| | - Josbert M Metselaar
- b Department of Experimental Molecular Imaging , University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH - Aachen University , Aachen , Germany.,c Department of Targeted Therapeutics , MIRA Institute, University of Twente , Enschede , The Netherlands
| | - Gert Storm
- c Department of Targeted Therapeutics , MIRA Institute, University of Twente , Enschede , The Netherlands.,d Department of Pharmaceutics , Utrecht Institute for Pharmaceutical Sciences, Utrecht University , Utrecht , The Netherlands
| | - Otto C Boerman
- a Department of Radiology and Nuclear Medicine , Radboud University Medical Center , Nijmegen , The Netherlands
| |
Collapse
|
41
|
Kumar C, Shetake N, Desai S, Kumar A, Samuel G, Pandey BN. Relevance of radiobiological concepts in radionuclide therapy of cancer. Int J Radiat Biol 2016; 92:173-86. [PMID: 26917443 DOI: 10.3109/09553002.2016.1144944] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Radionuclide therapy (RNT) is a rapidly growing area of clinical nuclear medicine, wherein radionuclides are employed to deliver cytotoxic dose of radiation to the diseased cells/tissues. During RNT, radionuclides are either directly administered or delivered through biomolecules targeting the diseased site. RNT has been clinically used for diverse range of diseases including cancer, which is the focus of the review. CONCLUSIONS The major emphasis in RNT has so far been given towards developing peptides/antibodies and other molecules to conjugate a variety of therapeutic radioisotopes for improved targeting/delivery of radiation dose to the tumor cells. Despite that, many of the RNT approaches have not achieved their desired therapeutic success probably due to poor knowledge about complex and dynamic (i) fate of radiolabeled molecules; (ii) radiation dose delivered; (iii) cellular heterogeneity in tumor mass; and (iv) cellular radiobiological response. Based on understanding gathered during recent years, it may be stated that besides the absorbed dose, the net radiobiological response of tumor/normal cells also determines the clinical response of radiotherapeutic modalities including RNT. The radiosensitivity of tumor/normal cells is governed by radiobiological phenomenon such as radiation-induced bystander effect, genomic instability, adaptive response and low dose hyper-radiosensitivity. These concepts have been well investigated in the context of external beam radiotherapy, but their clinical implications during RNT have received meagre attention. In this direction, a few studies performed using in vitro and in vivo models envisage the possibilities of exploiting the radiobiological knowledge for improved therapeutic outcome of RNT.
Collapse
Affiliation(s)
- Chandan Kumar
- a Radiopharmaceutical Chemistry Section , Bhabha Atomic Research Centre , Mumbai
| | - Neena Shetake
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai
| | - Sejal Desai
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| | - Amit Kumar
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| | - Grace Samuel
- c Isotope Production and Applications Division , Bhabha Atomic Research Centre , Mumbai
| | - Badri N Pandey
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| |
Collapse
|
42
|
Yook S, Cai Z, Lu Y, Winnik MA, Pignol JP, Reilly RM. Radiation Nanomedicine for EGFR-Positive Breast Cancer: Panitumumab-Modified Gold Nanoparticles Complexed to the β-Particle-Emitter, (177)Lu. Mol Pharm 2015; 12:3963-72. [PMID: 26402157 DOI: 10.1021/acs.molpharmaceut.5b00425] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Our objective was to construct a novel radiation nanomedicine for treatment of breast cancer (BC) expressing epidermal growth factor receptors (EGFR), particularly triple-negative tumors (TNBC). Gold nanoparticles (AuNP; 30 nm) were modified with polyethylene glycol (PEG) chains (4 kDa) derivatized with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelators for complexing the β-emitter, (177)Lu and with PEG chains (5 kDa) linked to panitumumab for targeting BC cells expressing EGFR. The AuNP were further coated with PEG chains (2 kDa) to stabilize the particles to aggregation. The binding and internalization of EGFR-targeted AuNP ((177)Lu-T-AuNP) into BC cells was studied and compared to nontargeted (177)Lu-NT-AuNP. The cytotoxicity of (177)Lu-T-AuNP and (177)Lu-NT-AuNP was measured in clonogenic assays using BC cells with widely different EGFR densities: MDA-MB-468 (10(6) receptors/cell), MDA-MB-231 (10(5) receptors/cell), and MCF-7 cells (10(4) receptors/cell). Radiation absorbed doses to the cell nucleus of MDA-MB-468 cells were estimated based on subcellular distribution. Darkfield and fluorescence microscopy as well as radioligand binding assays revealed that (177)Lu-T-AuNP were specifically bound by BC cells dependent on their EGFR density whereas the binding and internalization of (177)Lu-NT-AuNP was significantly lower. The affinity of binding of (177)Lu-T-AuNP to MDA-MB-468 cells was reduced by 2-fold compared to (123)I-labeled panitumumab (KD = 1.3 ± 0.2 nM vs 0.7 ± 0.4 nM, respectively). The cytotoxicity of (177)Lu-T-AuNP was dependent on the amount of radioactivity incubated with BC cells, their EGFR density and the radiosensitivity of the cells. The clonogenic survival (CS) of MDA-MB-468 cells overexpressing EGFR was reduced to <0.001% at the highest amount of (177)Lu-T-AuNP tested (4.5 MBq; 6 × 10(11) AuNP per 2.5 × 10(4)-1.2 × 10(5) cells). (177)Lu-T-AuNP were less effective for killing MDA-MB-231 cells or MCF-7 cells with moderate or low EGFR density (CS = 33.8 ± 1.6% and 25.8 ± 1.2%, respectively). Because the β-particles emitted by (177)Lu have a 2 mm range, (177)Lu-NT-AuNP were also cytotoxic to BC cells due to a cross-fire effect but (177)Lu-T-AuNP were significantly more potent for killing MDA-MB-468 cells overexpressing EGFR than (177)Lu-NT-AuNP at all amounts tested. The cross-fire effect of the β-particles emitted by (177)Lu may be valuable for eradicating BC cells in tumors that have low or moderate EGFR expression or cells that are not targeted by (177)Lu-T-AuNP as a consequence of heterogeneous intratumoral distribution. The radiation dose to the nucleus of a single MDA-MB-468 cell was 73.2 ± 6.7 Gy, whereas (177)Lu-NT-AuNP delivered 5.6 ± 0.6 Gy. We conclude that (177)Lu-T-AuNP is a promising novel radiation nanomedicine with potential application for treatment of TNBC, in which EGFR are often overexpressed.
Collapse
Affiliation(s)
- Simmyung Yook
- Department of Pharmaceutical Sciences, ‡Department of Chemistry, §Department of Medical Biophysics, University of Toronto , Toronto, Ontario M5S 3M2, Canada.,Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands, ⊥Department of Medical Imaging, University of Toronto , ∇Toronto General Research Institute, and #Joint Department of Medical Imaging, University Health Network, Toronto, Ontario M5S 3M2, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, ‡Department of Chemistry, §Department of Medical Biophysics, University of Toronto , Toronto, Ontario M5S 3M2, Canada.,Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands, ⊥Department of Medical Imaging, University of Toronto , ∇Toronto General Research Institute, and #Joint Department of Medical Imaging, University Health Network, Toronto, Ontario M5S 3M2, Canada
| | - Yijie Lu
- Department of Pharmaceutical Sciences, ‡Department of Chemistry, §Department of Medical Biophysics, University of Toronto , Toronto, Ontario M5S 3M2, Canada.,Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands, ⊥Department of Medical Imaging, University of Toronto , ∇Toronto General Research Institute, and #Joint Department of Medical Imaging, University Health Network, Toronto, Ontario M5S 3M2, Canada
| | - Mitchell A Winnik
- Department of Pharmaceutical Sciences, ‡Department of Chemistry, §Department of Medical Biophysics, University of Toronto , Toronto, Ontario M5S 3M2, Canada.,Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands, ⊥Department of Medical Imaging, University of Toronto , ∇Toronto General Research Institute, and #Joint Department of Medical Imaging, University Health Network, Toronto, Ontario M5S 3M2, Canada
| | - Jean-Philippe Pignol
- Department of Pharmaceutical Sciences, ‡Department of Chemistry, §Department of Medical Biophysics, University of Toronto , Toronto, Ontario M5S 3M2, Canada.,Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands, ⊥Department of Medical Imaging, University of Toronto , ∇Toronto General Research Institute, and #Joint Department of Medical Imaging, University Health Network, Toronto, Ontario M5S 3M2, Canada
| | - Raymond M Reilly
- Department of Pharmaceutical Sciences, ‡Department of Chemistry, §Department of Medical Biophysics, University of Toronto , Toronto, Ontario M5S 3M2, Canada.,Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands, ⊥Department of Medical Imaging, University of Toronto , ∇Toronto General Research Institute, and #Joint Department of Medical Imaging, University Health Network, Toronto, Ontario M5S 3M2, Canada
| |
Collapse
|
43
|
Sandoval S, Mendez N, Alfaro JG, Yang J, Aschemeyer S, Liberman A, Trogler WC, Kummel AC. Quantification of endocytosis using a folate functionalized silica hollow nanoshell platform. JOURNAL OF BIOMEDICAL OPTICS 2015; 20:88003. [PMID: 26315280 PMCID: PMC5996829 DOI: 10.1117/1.jbo.20.8.088003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/27/2015] [Indexed: 06/04/2023]
Abstract
A quantification method to measure endocytosis was designed to assess cellular uptake and specificity of a targeting nanoparticle platform. A simple N -hydroxysuccinimide ester conjugation technique to functionalize 100-nm hollow silica nanoshell particles with fluorescent reporter fluorescein isothiocyanate and folate or polyethylene glycol (PEG) was developed. Functionalized nanoshells were characterized using scanning electron microscopy and transmission electron microscopy and the maximum amount of folate functionalized on nanoshell surfaces was quantified with UV-Vis spectroscopy. The extent of endocytosis by HeLa cervical cancer cells and human foreskin fibroblast (HFF-1) cells was investigated in vitro using fluorescence and confocal microscopy. A simple fluorescence ratio analysis was developed to quantify endocytosis versus surface adhesion. Nanoshells functionalized with folate showed enhanced endocytosis by cancer cells when compared to PEG functionalized nanoshells. Fluorescence ratio analyses showed that 95% of folate functionalized silica nanoshells which adhered to cancer cells were endocytosed, while only 27% of PEG functionalized nanoshells adhered to the cell surface and underwent endocytosis when functionalized with 200 and 900 μg , respectively. Additionally, the endocytosis of folate functionalized nanoshells proved to be cancer cell selective while sparing normal cells. The developed fluorescence ratio analysis is a simple and rapid verification/validation method to quantify cellular uptake between datasets by using an internal control for normalization.
Collapse
Affiliation(s)
- Sergio Sandoval
- University of California, San Diego, Moores Cancer Center, Department of Bioengineering, CalIT Nanomedicine Laboratory, La Jolla, California 92093, United States
| | - Natalie Mendez
- University of California, San Diego, Department of Nanoengineering, Chemical Engineering, and Material Science, La Jolla, California 92093, United States
| | - Jesus G. Alfaro
- University of California, San Diego, Department of Nanoengineering, Chemical Engineering, and Material Science, La Jolla, California 92093, United States
| | - Jian Yang
- University of California, San Diego, Department of Nanoengineering, Chemical Engineering, and Material Science, La Jolla, California 92093, United States
| | - Sharraya Aschemeyer
- University of California, San Diego, Department of Chemistry and Biochemistry, La Jolla, California 92093, United States
| | - Alex Liberman
- University of California, San Diego, Department of Nanoengineering, Chemical Engineering, and Material Science, La Jolla, California 92093, United States
| | - William C. Trogler
- University of California, San Diego, Department of Chemistry and Biochemistry, La Jolla, California 92093, United States
| | - Andrew C. Kummel
- University of California, San Diego, Department of Chemistry and Biochemistry, La Jolla, California 92093, United States
| |
Collapse
|
44
|
|
45
|
Abstract
α-particle-emitting radionuclides are highly cytotoxic and are thus promising candidates for use in targeted radioimmunotherapy of cancer. Due to their high linear energy transfer (LET) combined with a short path length in tissue, α-particles cause severe DNA double-strand breaks that are repaired inaccurately and finally trigger cell death. For radioimmunotherapy, α-emitters such as 225Ac, 211At, 212Bi/212Pb, 213Bi and 227Th are coupled to antibodies via appropriate chelating agents. The α-emitter immunoconjugates preferably target proteins that are overexpressed or exclusively expressed on cancer cells. Application of α-emitter immunoconjugates seems particularly promising in treatment of disseminated cancer cells and small tumor cell clusters that are released during the resection of a primary tumor. α-emitter immunoconjugates have been successfully administered in numerous experimental studies for therapy of ovarian, colon, gastric, blood, breast and bladder cancer. Initial clinical trials evaluating α-emitter immunoconjugates in terms of toxicity and therapeutic efficacy have also shown positive results in patients with melanoma, ovarian cancer, acute myeloid lymphoma and glioma. The present problems in terms of availability of therapeutically effiective α-emitters will presumably be solved by use of alternative production routes and installation of additional production facilities in the near future. Therefore, clinical establishment of targeted α-emitter radioimmunotherapy as one part of a multimodal concept for therapy of cancer is a promising, middle-term concept.
Collapse
Affiliation(s)
- Christof Seidl
- Technische Universität München, Department of Nuclear Medicine, Ismaninger Strasse 22, 81675 Munich, Germany
| |
Collapse
|
46
|
Wang G, de Kruijff R, Rol A, Thijssen L, Mendes E, Morgenstern A, Bruchertseifer F, Stuart M, Wolterbeek H, Denkova A. Retention studies of recoiling daughter nuclides of 225Ac in polymer vesicles. Appl Radiat Isot 2014; 85:45-53. [DOI: 10.1016/j.apradiso.2013.12.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 12/05/2013] [Accepted: 12/05/2013] [Indexed: 10/25/2022]
|
47
|
Bandekar A, Zhu C, Jindal R, Bruchertseifer F, Morgenstern A, Sofou S. Anti-prostate-specific membrane antigen liposomes loaded with 225Ac for potential targeted antivascular α-particle therapy of cancer. J Nucl Med 2013; 55:107-14. [PMID: 24337602 DOI: 10.2967/jnumed.113.125476] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED This study evaluates targeted liposomes loaded with the α-particle generator (225)Ac to selectively kill prostate-specific membrane antigen (PSMA)-expressing cells with the aim to assess their potential for targeted antivascular radiotherapy. METHODS In this study, PEGylated liposomes were loaded with (225)Ac and labeled with the mouse antihuman PSMA J591 antibody or with the A10 PSMA aptamer. The targeting selectivity, extent of internalization, and killing efficacy of liposomes were evaluated on monolayers of prostate cancer cells intrinsically expressing PSMA (human LNCaP and rat Mat-Lu cells) and on monolayers of HUVEC induced to express PSMA (induced HUVEC). RESULTS The loading efficiency of (225)Ac into preformed liposomes ranged from 58.0% ± 4.6% to 85.6% ± 11.7% of introduced radioactivity. The conjugation reactions resulted in approximately 17 ± 2 J591 antibodies and 9 ± 2 A10 aptamers per liposome. The average size of liposomes, 107 ± 2 nm in diameter, was not affected by conjugation or loading. LNCaP cells exhibit 2:1:0.5 relative PSMA expression, compared with MatLu and induced HUVEC, respectively, based on flow cytometry detecting association of the J591 antibody. J591-labeled liposomes display higher levels of total specific binding to all cell lines than A10 aptamer-labeled liposomes. Specific cell association of targeted liposomes increases with incubation time. Cytotoxicity studies demonstrate that radiolabeled J591-labeled liposomes are most cytotoxic, with median lethal dose values, after 24 h of incubation, equal to 1.96 (5.3 × 10(-5)), 2.92 × 10(2) (7.9 × 10(-3)), and 2.33 × 10(1) Bq/mL (6.3 × 10(-4) μCi/mL) for LNCaP, Mat-Lu, and induced HUVEC, respectively, which are comparable to the values for the radiolabeled J591 antibody. For A10 aptamer-labeled liposomes, the corresponding values are 3.70 × 10(1) (1.0 × 10(-3)), 1.85 × 10(3) (5.0 × 10(-2)), and 4.07 × 10(3) Bq/mL (1.1 × 10(-1) μCi/mL), respectively. CONCLUSION Our studies demonstrate that anti-PSMA-targeted liposomes loaded with (225)Ac selectively bind, become internalized, and kill PSMA-expressing cells including endothelial cells induced to express PSMA. These findings-combined with the unique ability of liposomes to be easily tuned, in terms of size and surface modification, for optimizing biodistributions-suggest the potential of PSMA-targeting liposomes encapsulating α-particle emitters for selective antivascular α radiotherapy.
Collapse
Affiliation(s)
- Amey Bandekar
- Departments of Chemical and Biochemical Engineering and Biomedical Engineering, Rutgers University, Piscataway, New Jersey; and
| | | | | | | | | | | |
Collapse
|
48
|
Scheinberg DA, McDevitt MR. Actinium-225 in targeted alpha-particle therapeutic applications. Curr Radiopharm 2012; 4:306-20. [PMID: 22202153 DOI: 10.2174/1874471011104040306] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 03/31/2011] [Accepted: 04/19/2011] [Indexed: 01/22/2023]
Abstract
Alpha particle-emitting isotopes are being investigated in radioimmunotherapeutic applications because of their unparalleled cytotoxicity when targeted to cancer and their relative lack of toxicity towards untargeted normal tissue. Actinium- 225 has been developed into potent targeting drug constructs and is in clinical use against acute myelogenous leukemia. The key properties of the alpha particles generated by 225Ac are the following: i) limited range in tissue of a few cell diameters; ii) high linear energy transfer leading to dense radiation damage along each alpha track; iii) a 10 day halflife; and iv) four net alpha particles emitted per decay. Targeting 225Ac-drug constructs have potential in the treatment of cancer.
Collapse
Affiliation(s)
- David A Scheinberg
- Department of Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue,New York, NY 10065, USA
| | | |
Collapse
|
49
|
Polymersomes as nano-carriers to retain harmful recoil nuclides in alpha radionuclide therapy: a feasibility study. RADIOCHIM ACTA 2012. [DOI: 10.1524/ract.2012.1935] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Abstract
Targeted alpha therapy has shown promising pre-clinical and clinical results in the fight against cancer. The use of in vivo generators, generating a highly cytotoxic cascade of alpha particles, is attracting increasing interest for clinical application. 225Ac is one of the nuclides that can serve as an in vivo generator. It is commercially available and provides four alpha particles with a total energy of 28 MeV per 225Ac decay. However, its alpha emitting daughter nuclides may escape from the target region due to recoil and cause unwanted toxicity in other parts of the body. In this paper, we investigate the feasibility of designing spherical, block-copolymer based nano-carriers (polymersomes) to retain the recoiling daughter nuclides. A Monte Carlo code, called NANVES, has been developed to simulate the range distributions of recoil atoms in different materials and to determine the optimum nano-carriers design. Recoil ranges in planar polystyrene films were determined experimentally and compared to simulations of the experiment, indicating that NANVES may provide accurate results. Simulations of various nano-carriers designs indicate that double-layered polymersomes with a diameter of 800 nm are capable of completely retaining the first daughter nuclide 221Fr, while the escape fraction of the third radioactive daughter 213Bi is reduced to 20% and the percentage of alpha particles emitted from escaped daughter products outside the nano-carriers is less than 10%.
Collapse
|
50
|
Papagiannaros A, Upponi J, Hartner W, Mongayt D, Levchenko T, Torchilin V. Quantum dot loaded immunomicelles for tumor imaging. BMC Med Imaging 2010; 10:22. [PMID: 20955559 PMCID: PMC2975645 DOI: 10.1186/1471-2342-10-22] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 10/18/2010] [Indexed: 01/23/2023] Open
Abstract
Background Optical imaging is a promising method for the detection of tumors in animals, with speed and minimal invasiveness. We have previously developed a lipid coated quantum dot system that doubles the fluorescence of PEG-grafted quantum dots at half the dose. Here, we describe a tumor-targeted near infrared imaging agent composed of cancer-specific monoclonal anti-nucleosome antibody 2C5, coupled to quantum dot (QD)-containing polymeric micelles, prepared from a polyethylene glycol/phosphatidylethanolamine (PEG-PE) conjugate. Its production is simple and involves no special equipment. Its imaging potential is great since the fluorescence intensity in the tumor is twofold that of non-targeted QD-loaded PEG-PE micelles at one hour after injection. Methods Para-nitrophenol-containing (5%) PEG-PE quantum dot micelles were produced by the thin layer method. Following hydration, 2C5 antibody was attached to the PEG-PE micelles and the QD-micelles were purified using dialysis. 4T1 breast tumors were inoculated subcutaneously in the flank of the animals. A lung pseudometastatic B16F10 melanoma model was developed using tail vein injection. The contrast agents were injected via the tail vein and mice were depilated, anesthetized and imaged on a Kodak Image Station. Images were taken at one, two, and four hours and analyzed using a methodology that produces normalized signal-to-noise data. This allowed for the comparison between different subjects and time points. For the pseudometastatic model, lungs were removed and imaged ex vivo at one and twenty four hours. Results The contrast agent signal intensity at the tumor was double that of the passively targeted QD-micelles with equally fast and sharply contrasted images. With the side views of the animals only tumor is visible, while in the dorsal view internal organs including liver and kidney are visible. Ex vivo results demonstrated that the agent detects melanoma nodes in a lung pseudometastatic model after a 24 hours wash-out period, while at one hour, only a uniform signal is detected. Conclusions The targeted agent produces ultrabright tumor images and double the fluorescence intensity, as rapidly and at the same low dose as the passively targeted agents. It represents a development that may potentially serve to enhance early detection for metastases.
Collapse
Affiliation(s)
- Aristarchos Papagiannaros
- Department of Pharmaceutical Sciences and Center for Pharmaceutical Biotechnology and Nanomedicine, 312 Mugar Life Sciences Building, Northeastern University, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|