1
|
Nair RR, Sarkar A, Hariharan P, Gabrielson KL, Wu T, Liu C, Sharma A, Liyanage W, Bhujwalla ZM, Vidaver MFP, Kannan RM, Sofou S. Low-dose temozolomide selectively increases glioblastoma's vascular permeability, tumor microenvironment penetration and the killing potential of systemic actinium-225 α-particle dendrimer-radioconjugates improving treatment efficacy. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07332-w. [PMID: 40366390 DOI: 10.1007/s00259-025-07332-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: 03/08/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
PURPOSE The poor prognosis of glioblastoma is mostly due to the relatively low tumor vascular permeability to therapeutics, the tumor's vicinity to the brain, that limits treatment aggressiveness, and/or drug resistance. METHODS In this study, the efficacy of systemically injected actinium-225 dendrimer-radioconjugates was evaluated in an immune-competent orthotopic GL261-C57BL/6 mouse model after administration of low-dose, standard-of-care temozolomide, that selectively increased the tumor vascular permeability to dendrimer-radioconjugates. Alpha-particles' short range in tissue combined with the dendrimers' selective uptake by glioblastomas, could limit the irradiation of the neighboring brain, while the complex double-strand DNA breaks caused by α-particles were expected to be largely impervious to resistance by cancer cells. RESULTS On mice bearing 9.7 ± 5.7mm3 brain tumors, at activities that did not cause long-term (11-months) toxicities, dendrimer-radioconjugates, that were systemically-administered 24-hours after injection of temozolomide, significantly improved survival compared to dendrimer-radioconjugates alone (44 vs. 39 days mean survival, p = 0.0017) and/or compared to temozolomide alone and/or to non-treated animals (31 and 30 days, p < 0.001). This was attributed to: (1) the noteworthy increase (by 33%) in tumor absorbed doses delivered by dendrimer-radioconjugates when injected after chemotherapy, without altering normal organ (including the brain's) dosimetry; (2) the potentially deeper tumor penetration of dendrimer-radioconjugates, suggested by the enhanced dendrimer penetration within GL261-spheroids, employed as model tumor-avascular regions; and/or (3) the formation of a more lethal cocktail when both modalities acted on same cancer cells. CONCLUSIONS This study demonstrates the potential and safety of actinium-225 dendrimer-radioconjugates as a systemic α-particle radiotherapy for glioblastoma enhanced by low-dose temozolomide.
Collapse
Affiliation(s)
- Rajiv Ranjit Nair
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology (INBT), Johns Hopkins University, Baltimore, MD, USA
| | - Aira Sarkar
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology (INBT), Johns Hopkins University, Baltimore, MD, USA
| | - Pooja Hariharan
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology (INBT), Johns Hopkins University, Baltimore, MD, USA
| | - Kathleen L Gabrielson
- Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, USA
| | - Tony Wu
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Chang Liu
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Anjali Sharma
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Wathsala Liyanage
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Marie-France Penet Vidaver
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Stavroula Sofou
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology (INBT), Johns Hopkins University, Baltimore, MD, USA.
- Sidney Kimmel Comprehensive Cancer Center, Cancer Invasion & Metastasis Program, Department of Oncology, Johns Hopkins University, Baltimore, MD, USA.
- ChemBE, Johns Hopkins University, 3400 North Charles Street Maryland Hall 116, Baltimore, MD, 21218, USA.
| |
Collapse
|
2
|
Shoshan Y, Gomori MJ, Moss L, Bari SE, Edery N, Den RB, Arazi L, Popovtzer A, Feldman J, Moscovici S. Stereotactic implantation of diffusing alpha-emitters radiation therapy sources in the swine brain: a potential new focal therapy for brain tumors. J Neurooncol 2025; 172:387-396. [PMID: 39747715 PMCID: PMC11937107 DOI: 10.1007/s11060-024-04919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE Diffusing alpha-emitters Radiation Therapy ("Alpha DaRT") is a new cancer treatment modality that employs radium-224-loaded metal sources implanted in solid tumors to disperse alpha-emitting atoms within a therapeutic "kill-zone" of a few millimeters around each source. Preclinical studies have demonstrated tumor growth delay in various cancer types, including glioblastoma multiforme, and the method is used in clinical trials for patients with skin and head and neck cancer. This study aims to assess the safety and feasibility of implementing Alpha DaRT for brain tumor treatment in a large animal model. METHODS Alpha-DaRT sources were delivered via image-guided stereotactic implantation into both hemispheres of eight swine. 1-3 layers of radial deployment of 7 sources were delivered through a single penetration point into each hemisphere. A 90-day follow-up period included clinical evaluation, brain MRI, head CT, blood, CSF, urine, and feces sampling, and an analysis of source location over time. Brain tissue pathology was performed on termination. RESULTS Alpha-DaRT sources were reproducibly and efficiently delivered to the brain cortex and subcortex. No unexpected abnormalities were detected in blood or CSF samples. MRI and CT scans revealed no evidence of major bleeding or infection. Measurements of 212Pb in blood and CSF exhibited the expected exponential decay from day 7 to day 14 post-source implantation. Minimal spatial and temporal movements of the sources were noted. Histopathological analysis demonstrated locally confined findings in brain parenchyma in a very close proximity to the sources. CONCLUSION Alpha-DaRT sources can be safely delivered into a large animal brain using image-guided stereotactic implantation. These findings support further exploration of Alpha DaRT as a potential treatment modality for brain tumors.
Collapse
Affiliation(s)
- Yigal Shoshan
- Department of Neurosurgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| | - Moshe J Gomori
- Department of Radiology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Lior Moss
- Department of Pathology, Kimron Veterinary Institute, Bet Dagan, Israel
| | - Saleem Eben Bari
- Department of Pathology, Kimron Veterinary Institute, Bet Dagan, Israel
| | - Nir Edery
- Department of Pathology, Kimron Veterinary Institute, Bet Dagan, Israel
| | | | - Lior Arazi
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
| | - Aron Popovtzer
- Sharett Institute of Oncology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Jon Feldman
- Sharett Institute of Oncology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Samuel Moscovici
- Department of Neurosurgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
3
|
Bakht MK, Beltran H. Biological determinants of PSMA expression, regulation and heterogeneity in prostate cancer. Nat Rev Urol 2025; 22:26-45. [PMID: 38977769 PMCID: PMC11841200 DOI: 10.1038/s41585-024-00900-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 07/10/2024]
Abstract
Prostate-specific membrane antigen (PSMA) is an important cell-surface imaging biomarker and therapeutic target in prostate cancer. The PSMA-targeted theranostic 177Lu-PSMA-617 was approved in 2022 for men with PSMA-PET-positive metastatic castration-resistant prostate cancer. However, not all patients respond to PSMA-radioligand therapy, in part owing to the heterogeneity of PSMA expression in the tumour. The PSMA regulatory network is composed of a PSMA transcription complex, an upstream enhancer that loops to the FOLH1 (PSMA) gene promoter, intergenic enhancers and differentially methylated regions. Our understanding of the PSMA regulatory network and the mechanisms underlying PSMA suppression is evolving. Clinically, molecular imaging provides a unique window into PSMA dynamics that occur on therapy and with disease progression, although challenges arise owing to the limited resolution of PET. PSMA regulation and heterogeneity - including intertumoural and inter-patient heterogeneity, temporal changes, lineage dynamics and the tumour microenvironment - affect PSMA theranostics. PSMA response and resistance to radioligand therapy are mediated by a number of potential mechanisms, and complementary biomarkers beyond PSMA are under development. Understanding the biological determinants of cell surface target regulation and heterogeneity can inform precision medicine approaches to PSMA theranostics as well as other emerging therapies.
Collapse
Affiliation(s)
- Martin K Bakht
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Alcocer-Ávila M, Levrague V, Delorme R, Testa É, Beuve M. Biophysical modeling of low-energy ion irradiations with NanOx. Med Phys 2024; 51:9358-9371. [PMID: 39287463 DOI: 10.1002/mp.17407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/22/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Targeted radiotherapies with low-energy ions show interesting possibilities for the selective irradiation of tumor cells, a strategy particularly appropriate for the treatment of disseminated cancer. Two promising examples are boron neutron capture therapy (BNCT) and targeted radionuclide therapy with α $\alpha$ -particle emitters (TAT). The successful clinical translation of these radiotherapies requires the implementation of accurate radiation dosimetry approaches able to take into account the impact on treatments of the biological effectiveness of ions and the heterogeneity in the therapeutic agent distribution inside the tumor cells. To this end, biophysical models can be applied to translate the interactions of radiations with matter into biological endpoints, such as cell survival. PURPOSE The NanOx model was initially developed for predicting the cell survival fractions resulting from irradiations with the high-energy ion beams encountered in hadrontherapy. We present in this work a new implementation of the model that extends its application to irradiations with low-energy ions, as the ones found in TAT and BNCT. METHODS The NanOx model was adapted to consider the energy loss of primary ions within the sensitive volume (i.e., the cell nucleus). Additional assumptions were introduced to simplify the practical implementation of the model and reduce computation time. In particular, for low-energy ions the narrow-track approximation allowed to neglect the energy deposited by secondary electrons outside the sensitive volume, increasing significantly the performance of simulations. Calculations were performed to compare the original hadrontherapy implementation of the NanOx model with the present one in terms of the inactivation cross sections of human salivary gland cells as a function of the kinetic energy of incident α $\alpha$ -particles. RESULTS The predictions of the previous and current versions of NanOx agreed for incident energies higher than 1 MeV/n. For lower energies, the new NanOx implementation predicted a decrease in the inactivation cross sections that depended on the length of the sensitive volume. CONCLUSIONS We reported in this work an extension of the NanOx biophysical model to consider irradiations with low-energy ions, such as the ones found in TAT and BNCT. The excellent agreement observed at intermediate and high energies between the original hadrontherapy implementation and the present one showed that NanOx offers a consistent, self-integrated framework for describing the biological effects induced by ion irradiations. Future work will focus on the application of the latest version of NanOx to cases closer to the clinical setting.
Collapse
Affiliation(s)
- Mario Alcocer-Ávila
- Université Claude Bernard Lyon 1, CNRS/IN2P3, IP2I Lyon, UMR 5822, Villeurbanne, France
| | - Victor Levrague
- University of Grenoble Alpes, CNRS, Grenoble INP, LPSC-IN2P3, Grenoble, France
| | - Rachel Delorme
- University of Grenoble Alpes, CNRS, Grenoble INP, LPSC-IN2P3, Grenoble, France
| | - Étienne Testa
- Université Claude Bernard Lyon 1, CNRS/IN2P3, IP2I Lyon, UMR 5822, Villeurbanne, France
| | - Michaël Beuve
- Université Claude Bernard Lyon 1, CNRS/IN2P3, IP2I Lyon, UMR 5822, Villeurbanne, France
| |
Collapse
|
5
|
Heger G, Dumančić M, Luz I, Vatarescu M, Weizman N, Miller BW, Cooks T, Arazi L. First measurements of radon-220 diffusion in mice tumors, towards treatment planning in diffusing alpha-emitters radiation therapy. Med Phys 2024; 51:5045-5058. [PMID: 38507254 DOI: 10.1002/mp.17020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 01/22/2024] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Diffusing alpha-emitters radiation therapy ("Alpha-DaRT") is a new method for treating solid tumors with alpha particles, relying on the release of the short-lived alpha-emitting daughter atoms of radium-224 from interstitial sources inserted into the tumor. Alpha-DaRT tumor dosimetry is governed by the spread of radium's progeny around the source, as described by an approximate framework called the "diffusion-leakage model". The most important model parameters are the diffusion lengths of radon-220 and lead-212, and their estimation is therefore essential for treatment planning. PURPOSE Previous works have provided initial estimates for the dominant diffusion length, by measuring the activity spread inside mice-borne tumors several days after the insertion of an Alpha-DaRT source. The measurements, taken when lead-212 was in secular equilibrium with radium-224, were interpreted as representing the lead-212 diffusion length. The aim of this work is to provide first experimental estimates for the diffusion length of radon-220, using a new methodology. METHODS The diffusion length of radon-220 was estimated from autoradiography measurements of histological sections taken from 24 mice-borne subcutaneous tumors of five different types. Unlike previous studies, the source dwell time inside the tumor was limited to 30 min, to prevent the buildup of lead-212. To investigate the contribution of potential non-diffusive processes, experiments were done in two sets: fourteen in vivo tumors, where during the treatment the tumors were still carried by the mice with active blood supply, and 10 ex-vivo tumors, where the tumors were excised before source insertion and kept in a medium at37 ∘ C $37^\circ {\text{C}}$ with the source inside. RESULTS The measured diffusion lengths of radon-220, extracted by fitting the recorded activity pattern up to 1.5 mm from the source, lie in the range0.25 - 0.6 mm ${0.25-0.6}\nobreakspace {\text{mm}}$ , with no significant difference between the average values measured in in-vivo and ex-vivo tumors:L R n i n - v i v o = 0.40 ± 0.08 mm $L_{Rn}^{in-vivo}=0.40{\pm }0.08\nobreakspace {\text{mm}}$ versusL R n e x - v i v o = 0.39 ± 0.07 mm $L_{Rn}^{ex-vivo}=0.39{\pm }0.07\nobreakspace {\text{mm}}$ . However, in-vivo tumors display an enhanced spread of activity 2-3 mm away from the source. This effect is not explained by the current model and is much less pronounced in ex-vivo tumors. CONCLUSIONS The average measured radon-220 diffusion lengths in both in-vivo and ex-vivo tumors are consistent with published data on the diffusion length of radon in water and lie close to the upper limit of the previously estimated range of0.2 - 0.4 mm $0.2-0.4\nobreakspace {\text{mm}}$ . The observation that close to the source there is no apparent difference between in-vivo and ex-vivo tumors, and the good agreement with the theoretical model in this region suggest that the spread of radon-220 is predominantly diffusive in this region. The departure from the model prediction in in-vivo tumors at large radial distances may hint at potential vascular contribution, which will be the subject of future works.
Collapse
Affiliation(s)
- Guy Heger
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Mirta Dumančić
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- Now at Gerald Bronfman Department of Oncology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Ishai Luz
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Maayan Vatarescu
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Noam Weizman
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- Oncology Department, Radiation Therapy Unit, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Brian W Miller
- College of Medicine, Department of Radiation Oncology, Department of Medical Imaging, The University of Arizona, Tucson, Arizona, USA
| | - Tomer Cooks
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Lior Arazi
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
6
|
Li F, Wang H, Ye T, Guo P, Lin X, Hu Y, Wei W, Wang S, Ma G. Recent Advances in Material Technology for Leukemia Treatments. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313955. [PMID: 38547845 DOI: 10.1002/adma.202313955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/11/2024] [Indexed: 04/13/2024]
Abstract
Leukemia is a widespread hematological malignancy characterized by an elevated white blood cell count in both the blood and the bone marrow. Despite notable advancements in leukemia intervention in the clinic, a large proportion of patients, especially acute leukemia patients, fail to achieve long-term remission or complete remission following treatment. Therefore, leukemia therapy necessitates optimization to meet the treatment requirements. In recent years, a multitude of materials have undergone rigorous study to serve as delivery vectors or direct intervention agents to bolster the effectiveness of leukemia therapy. These materials include liposomes, protein-based materials, polymeric materials, cell-derived materials, and inorganic materials. They possess unique characteristics and are applied in a broad array of therapeutic modalities, including chemotherapy, gene therapy, immunotherapy, radiotherapy, hematopoietic stem cell transplantation, and other evolving treatments. Here, an overview of these materials is presented, describing their physicochemical properties, their role in leukemia treatment, and the challenges they face in the context of clinical translation. This review inspires researchers to further develop various materials that can be used to augment the efficacy of multiple therapeutic modalities for novel applications in leukemia treatment.
Collapse
Affiliation(s)
- Feng Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huaiji Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tong Ye
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peilin Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoyun Lin
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yuxing Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
7
|
Naka S, Ooe K, Shirakami Y, Kurimoto K, Sakai T, Takahashi K, Toyoshima A, Wang Y, Haba H, Kato H, Tomiyama N, Watabe T. Production of [ 211At]NaAt solution under GMP compliance for investigator-initiated clinical trial. EJNMMI Radiopharm Chem 2024; 9:29. [PMID: 38619655 PMCID: PMC11018728 DOI: 10.1186/s41181-024-00257-z] [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: 01/26/2024] [Accepted: 03/21/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND The alpha emitter astatine-211 (211At) is garnering attention as a novel targeted alpha therapy for patients with refractory thyroid cancer resistant to conventional therapy using beta emitter radioiodine (131I). Herein, we aimed to establish a robust method for the manufacturing and quality control of [211At]NaAt solution for intravenous administration under the good manufacturing practice guidelines for investigational products to conduct an investigator-initiated clinical trial. RESULTS 211At was separated and purified via dry distillation using irradiated Bi plates containing 211At obtained by the nuclear reaction of 209Bi(4He, 2n)211At. After purification, the 211At trapped in the cold trap was collected in a reaction vessel using 15 mL recovery solution (1% ascorbic acid and 2.3% sodium hydrogen carbonate). After stirring the 211At solution for 1 h inside a closed system, the reaction solution was passed through a sterile 0.22 μm filter placed in a Grade A controlled area and collected in a product vial to prepare the [211At]NaAt solution. According to the 3-lot tests, decay collected radioactivity and radiochemical yield of [211At]NaAt were 78.8 ± 6.0 MBq and 40 ± 3%, respectively. The radiochemical purity of [211At]At- obtained via ion-pair chromatography at the end of synthesis (EOS) was 97 ± 1%, and remained > 96% 6 h after EOS; it was detected at a retention time (RT) 3.2-3.3 min + RT of I-. LC-MS analysis indicated that this principal peak corresponded with an astatide ion (m/z = 210.988046). In gamma-ray spectrometry, the 211At-related peaks were identified (X-ray: 76.9, 79.3, 89.3, 89.8, and 92.3 keV; γ-ray: 569.7 and 687.0 keV), whereas the peak at 245.31 keV derived from 210At was not detected during the 22 h continuous measurement. The target material, Bi, was below the 9 ng/mL detection limit in all lots of the finished product. The pH of the [211At]NaAt solution was 7.9-8.6; the concentration of ascorbic acid was 9-10 mg/mL. Other quality control tests, including endotoxin and sterility tests, confirmed that the [211At]NaAt solution met all quality standards. CONCLUSIONS We successfully established a stable method of [211At]NaAt solution that can be administered to humans intravenously as an investigational product.
Collapse
Affiliation(s)
- Sadahiro Naka
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Pharmacy, Osaka University Hospital, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazuhiro Ooe
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshifumi Shirakami
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kenta Kurimoto
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Pharmacy, Osaka University Hospital, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toshihiro Sakai
- Hanwa Intelligent Medical Center, Hanwa Daini Senboku Hospital, 3176 Fukaikitamachi, Naka- ku, Sakai, Osaka, 599-8271, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima, 960-1295, Japan
| | - Atsushi Toyoshima
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yang Wang
- Nishina Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Hiromitsu Haba
- Nishina Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Hiroki Kato
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Noriyuki Tomiyama
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Radiology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
8
|
Epstein L, Heger G, Roy A, Gannot I, Kelson I, Arazi L. The low-LET radiation contribution to the tumor dose in diffusing alpha-emitters radiation therapy. Med Phys 2024; 51:3020-3033. [PMID: 38096442 DOI: 10.1002/mp.16885] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Diffusing alpha-emitters Radiation Therapy ("Alpha DaRT") is a new technique that enables the use of alpha particles for the treatment of solid tumors. Alpha DaRT employs interstitial sources carrying a few μ $\mu$ Ci of224 $^{224}$ Ra below their surface, designed to release a chain of short-lived atoms (progeny of224 $^{224}$ Ra) which emit alpha particles, along with beta, Auger, and conversion electrons, x- and gamma rays. These atoms diffuse around the source and create-primarily through their alpha decays-a lethal high-dose region measuring a few millimeters in diameter. PURPOSE While previous studies focused on the dose from the alpha emissions alone, this work addresses the electron and photon dose contributed by the diffusing atoms and by the atoms remaining on the source surface, for both a single Alpha DaRT source and multi-source lattices. This allows to evaluate the low-LET contribution to the tumor dose and tumor cell survival, and demonstrate the sparing of surrounding healthy tissue. METHODS The low-LET dose is calculated using the EGSnrc and FLUKA Monte Carlo (MC) codes. We compare the results of a simple line-source approximation with no diffusion to those of a full simulation, which implements a realistic source geometry and the spread of diffusing atoms. We consider two opposite scenarios: one with low diffusion and high212 $^{212}$ Pb leakage, and the other with high diffusion and low leakage. The low-LET dose in source lattices is calculated by superposition of single-source contributions. Its effect on cell survival is estimated with the linear quadratic model in the limit of low dose rate. RESULTS For sources carrying 3 μ $\umu$ Ci/cm224 $^{224}$ Ra arranged in a hexagonal lattice with 4 mm spacing, the minimal low-LET dose between sources is∼ 18 - 30 $\sim 18-30$ Gy for the two test cases and is dominated by the beta contribution. The low-LET dose drops below 5 Gy∼ 3 $\sim 3$ mm away from the outermost source in the lattice with an effective maximal dose rate of< 0.04 $<0.04$ Gy/h. The accuracy of the line-source/no-diffusion approximation is∼ 15 % $\sim 15\%$ for the total low-LET dose over clinically relevant distances (2-4 mm). The low-LET dose reduces tumor cell survival by a factor of∼ 2 - 200 $\sim 2-200$ . CONCLUSIONS The low-LET dose in Alpha DaRT can be modeled by conventional MC techniques with appropriate leakage corrections to the source activity. For 3 μ $\umu$ Ci/cm224 $^{224}$ Ra sources, the contribution of the low-LET dose can reduce cell survival inside the tumor by up to two orders of magnitude. The low-LET dose to surrounding healthy tissue is negligible. Increasing source activities by a factor of 5 can bring the low-LET dose itself to therapeutic levels, in addition to the high-LET dose contributed by alpha particles, leading to a "self-boosted" Alpha DaRT configuration, and potentially allowing to increase the lattice spacing.
Collapse
Affiliation(s)
- Lior Epstein
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Soreq Nuclear Research Center, Yavne, Israel
| | - Guy Heger
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
| | - Arindam Roy
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
| | - Israel Gannot
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Itzhak Kelson
- School of Physics and Astronomy, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Lior Arazi
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
| |
Collapse
|
9
|
Zhou L, Liu Y, Wu Y, Yang X, Spring Kong FM, Lu Y, Xue J. Low-dose radiation therapy mobilizes antitumor immunity: New findings and future perspectives. Int J Cancer 2024; 154:1143-1157. [PMID: 38059788 DOI: 10.1002/ijc.34801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023]
Abstract
Radiotherapy has unique immunostimulatory and immunosuppressive effects. Although high-dose radiotherapy has been found to have systemic antitumor effects, clinically significant abscopal effects were uncommon on the basis of irradiating single lesion. Low-dose radiation therapy (LDRT) emerges as a novel approach to enhance the antitumor immune response due to its role as a leverage to reshape the tumor immune microenvironment (TIME). In this article, from bench to bedside, we reviewed the possible immunomodulatory role of LDRT on TIME and systemic tumor immune environment, and outlined preclinical evidence and clinical application. We also discussed the current challenges when LDRT is used as a combination therapy, including the optimal dose, fraction, frequency, and combination of drugs. The advantage of low toxicity makes LDRT potential to be applied in multiple lesions to amplify antitumor immune response in polymetastatic disease, and its intersection with other disciplines might also make it a direction for radiotherapy-combined modalities.
Collapse
Affiliation(s)
- Laiyan Zhou
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Disaster Medical Center, Sichuan University, Chengdu, China
| | - Yuanxin Liu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanjun Wu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Yang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feng-Ming Spring Kong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Rahman AKMR, Babu MH, Ovi MK, Zilani MM, Eithu IS, Chakraborty A. Actinium-225 in Targeted Alpha Therapy. J Med Phys 2024; 49:137-147. [PMID: 39131433 PMCID: PMC11309130 DOI: 10.4103/jmp.jmp_22_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/20/2024] [Accepted: 04/14/2024] [Indexed: 08/13/2024] Open
Abstract
The utilization of actinium-225 (225Ac) radionuclides in targeted alpha therapy for cancer was initially outlined in 1993. Over the past two decades, substantial research has been conducted, encompassing the establishment of 225Ac production methods, various preclinical investigations, and several clinical studies. Currently, there is a growing number of compounds labeled with 225Ac that are being developed and tested in clinical trials. In response to the increasing demand for this nuclide, production facilities are either being built or have already been established. This article offers a concise summary of the present state of clinical advancements in compounds labeled with 225Ac. It outlines various processes involved in the production and purification of 225Ac to cater to the growing demand for this radionuclide. The article examines the merits and drawbacks of different procedures, delves into preclinical trials, and discusses ongoing clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Amit Chakraborty
- Department of Physics, University of Chittagong, Chittagong, Bangladesh
| |
Collapse
|
11
|
Kaizuka Y, Suzuki H, Watabe T, Ooe K, Toyoshima A, Takahashi K, Sawada K, Iimori T, Masuda Y, Uno T, Kannaka K, Uehara T. Neopentyl glycol-based radiohalogen-labeled amino acid derivatives for cancer radiotheranostics. EJNMMI Radiopharm Chem 2024; 9:17. [PMID: 38407647 PMCID: PMC10897087 DOI: 10.1186/s41181-024-00244-4] [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: 01/11/2024] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND L-type amino acid transporter 1 (LAT1) is overexpressed in various cancers; therefore, radiohalogen-labeled amino acid derivatives targeting LAT1 have emerged as promising candidates for cancer radiotheranostics. However, 211At-labeled amino acid derivatives exhibit instability against deastatination in vivo, making it challenging to use 211At for radiotherapy. In this study, radiohalogen-labeled amino acid derivatives with high dehalogenation stability were developed. RESULTS We designed and synthesized new radiohalogen-labeled amino acid derivatives ([211At]At-NpGT, [125I]I-NpGT, and [18F]F-NpGT) in which L-tyrosine was introduced into the neopentyl glycol (NpG) structure. The radiolabeled amino acid derivatives were recognized as substrates of LAT1 in the in vitro studies using C6 glioma cells. In a biodistribution study using C6 glioma-bearing mice, these agents exhibited high stability against in vivo dehalogenation and similar biodistributions. The similarity of [211At]At-NpGT and [18F]F-NpGT indicated that these pairs of radiolabeled compounds would be helpful in radiotheranostics. Moreover, [211At]At-NpGT exhibited a dose-dependent inhibitory effect on the growth of C6 glioma-bearing mice. CONCLUSIONS [211At]At-NpGT exhibited a dose-dependent inhibitory effect on the tumor growth of glioma-bearing mice, and its biodistribution was similar to that of other radiohalogen-labeled amino acid derivatives. These findings suggest that radiotheranostics using [18F]F-NpGT and [123/131I]I-NpGT for diagnostic applications and [211At]At-NpGT and [131I]I-NpGT for therapeutic applications are promising.
Collapse
Affiliation(s)
- Yuta Kaizuka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8675, Japan
| | - Hiroyuki Suzuki
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8675, Japan.
| | - Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Kazuhiro Ooe
- Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Atsushi Toyoshima
- Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Koichi Sawada
- Department of Radiology, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8677, Japan
| | - Takashi Iimori
- Department of Radiology, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8677, Japan
| | - Yoshitada Masuda
- Department of Radiology, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8677, Japan
| | - Takashi Uno
- Department of Radiology, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8677, Japan
- Diagnostic Radiology and Radiation Oncology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8670, Japan
| | - Kento Kannaka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8675, Japan
| | - Tomoya Uehara
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8675, Japan.
| |
Collapse
|
12
|
Jalloul W, Ghizdovat V, Stolniceanu CR, Ionescu T, Grierosu IC, Pavaleanu I, Moscalu M, Stefanescu C. Targeted Alpha Therapy: All We Need to Know about 225Ac's Physical Characteristics and Production as a Potential Theranostic Radionuclide. Pharmaceuticals (Basel) 2023; 16:1679. [PMID: 38139806 PMCID: PMC10747780 DOI: 10.3390/ph16121679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
The high energy of α emitters, and the strong linear energy transfer that goes along with it, lead to very efficient cell killing through DNA damage. Moreover, the degree of oxygenation and the cell cycle state have no impact on these effects. Therefore, α radioisotopes can offer a treatment choice to individuals who are not responding to β- or gamma-radiation therapy or chemotherapy drugs. Only a few α-particle emitters are suitable for targeted alpha therapy (TAT) and clinical applications. The majority of available clinical research involves 225Ac and its daughter nuclide 213Bi. Additionally, the 225Ac disintegration cascade generates γ decays that can be used in single-photon emission computed tomography (SPECT) imaging, expanding the potential theranostic applications in nuclear medicine. Despite the growing interest in applying 225Ac, the restricted global accessibility of this radioisotope makes it difficult to conduct extensive clinical trials for many radiopharmaceutical candidates. To boost the availability of 225Ac, along with its clinical and potential theranostic applications, this review attempts to highlight the fundamental physical properties of this α-particle-emitting isotope, as well as its existing and possible production methods.
Collapse
Affiliation(s)
- Wael Jalloul
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Vlad Ghizdovat
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Teodor Ionescu
- Department of Morpho-Functional Sciences (Pathophysiology), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Irena Cristina Grierosu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ioana Pavaleanu
- Department of Mother and Child, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| |
Collapse
|
13
|
Liatsou I, Josefsson A, Yu J, Li Z, Davis K, Brayton C, Wang H, Hobbs RF, Sgouros G. Early Normal Tissue Effects and Bone Marrow Relative Biological Effectiveness for an Actinium 225-Labeled HER2/neu-Targeting Antibody. Int J Radiat Oncol Biol Phys 2023; 117:1028-1037. [PMID: 37331568 DOI: 10.1016/j.ijrobp.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/16/2023] [Accepted: 06/11/2023] [Indexed: 06/20/2023]
Abstract
PURPOSE In this study we determined the dose-independent relative biological effectiveness (RBE2) of bone marrow for an anti-HER2/neu antibody labeled with the alpha-particle emitter actinium 225 (225Ac). Hematologic toxicity is often a consequence of radiopharmaceutical therapy (RPT) administration, and dosimetric guidance to the bone marrow is required to limit toxicity. METHODS AND MATERIALS Female neu/N transgenic mice (MMTV-neu) were intravenously injected with 0 to 16.65 kBq of the alpha-particle emitter labeled antibody, 225Ac-DOTA-7.16.4, and euthanized at 1 to 9 days after treatment. Complete blood counts were performed. Femurs and tibias were collected, and bone marrow was isolated from 1 femur and tibia and counted for radioactivity. Contralateral intact femurs were fixed, decalcified, and assessed by histology. Marrow cellularity was the biologic endpoint selected for RBE2 determination. For the reference radiation, both femurs of the mice were photon irradiated with 0 to 5 Gy using a small animal radiation research platform. RESULTS Response as measured by cellularity for the alpha-particle emitter RPT (αRPT) RPT and the external beam radiation therapy were linear and linear quadratic, respectively, as a function of absorbed dose. The resulting dose-independent RBE2 for bone marrow was 6. CONCLUSIONS As αRPT gains prominence, preclinical studies evaluating RBE in vivo will be important in relating to human experience with beta-particle emitter RPT. Such normal tissue RBE evaluations will help mitigate unexpected toxicity in αRPT.
Collapse
Affiliation(s)
- Ioanna Liatsou
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Anders Josefsson
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jing Yu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zhi Li
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kaori Davis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hao Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert F Hobbs
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - George Sgouros
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
14
|
Heger G, Roy A, Dumančić M, Arazi L. Alpha dose modeling in diffusing alpha-emitters radiation therapy-Part I: single-seed calculations in one and two dimensions. Med Phys 2023; 50:1793-1811. [PMID: 36464914 DOI: 10.1002/mp.16145] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 09/01/2022] [Accepted: 09/29/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Diffusing alpha-emitters Radiation Therapy ("DaRT") is a new method, presently in clinical trials, which allows treating solid tumors by alpha particles. DaRT relies on interstitial seeds carrying μCi-level 224 Ra activity below their surface, which release a chain of short-lived alpha emitters that spread throughout the tumor volume primarily by diffusion. Alpha dose calculations in DaRT are based on describing the transport of alpha emitting atoms, requiring new modeling techniques. PURPOSE A previous study introduced a simplified framework, the "Diffusion-Leakage (DL) model", for DaRT alpha dose calculations, and employed it to a point source, as a basic building block of arbitrary configurations of line sources. The aim of this work, which is divided into two parts, is to extend the model to realistic seed geometries (in Part I), and to employ single-seed calculations to study the properties of DaRT seed lattices (Part II). Such calculations can serve as a pragmatic guide for treatment planning in future clinical trials. METHODS We derive a closed-form asymptotic solution for an infinitely long cylindrical source, and extend it to an approximate time-dependent expression that assumes a uniform temporal profile at all radial distances from the source. We then develop a finite-element one-dimensional numerical scheme for a complete time-dependent solution of this geometry and validate it against the closed-form expressions. Finally, we discuss a two-dimensional axisymmetric scheme for a complete time-dependent solution for a seed of finite diameter and length. Different solutions are compared over the relevant parameter space, providing guidelines on their usability and limitations. RESULTS We show that approximating the seed as a finite line source comprised of point-like segments significantly underestimates the correct alpha dose, as predicted by the full two-dimensional calculation. The time-dependent one-dimensional solution is shown to coincide to sub-percent-level with the two-dimensional calculation in the seed midplane, and maintains an accuracy of a few percent up to ∼2 mm from the seed edge. CONCLUSIONS For actual treatment plans, the full two-dimensional solution should be used to generate dose lookup tables, similarly to the TG-43 format employed in conventional brachytherapy. Given the accuracy of the one-dimensional solution up to ∼2 mm from the seed edge it can be used for efficient parametric studies of DaRT seed lattices.
Collapse
Affiliation(s)
- Guy Heger
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
| | - Arindam Roy
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
| | - Mirta Dumančić
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
| | - Lior Arazi
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
| |
Collapse
|
15
|
Kerr CP, Grudzinski JJ, Nguyen TP, Hernandez R, Weichert JP, Morris ZS. Developments in Combining Targeted Radionuclide Therapies and Immunotherapies for Cancer Treatment. Pharmaceutics 2022; 15:128. [PMID: 36678756 PMCID: PMC9865370 DOI: 10.3390/pharmaceutics15010128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
Targeted radionuclide therapy (TRT) and immunotherapy are rapidly growing classes of cancer treatments. Basic, translational, and clinical research are now investigating therapeutic combinations of these agents. In comparison to external beam radiation therapy (EBRT), TRT has the unique advantage of treating all disease sites following intravenous injection and selective tumor uptake and retention-a particularly beneficial property in metastatic disease settings. The therapeutic value of combining radiation therapy with immune checkpoint blockade to treat metastases has been demonstrated in preclinical studies, whereas results of clinical studies have been mixed. Several clinical trials combining TRT and immune checkpoint blockade have been initiated based on preclinical studies combining these with EBRT and/or TRT. Despite the interest in translation of TRT and immunotherapy combinations, many questions remain surrounding the mechanisms of interaction and the optimal approach to clinical implementation of these combinations. This review highlights the mechanisms of interaction between anti-tumor immunity and radiation therapy and the status of basic and translational research and clinical trials investigating combinations of TRT and immunotherapies.
Collapse
Affiliation(s)
- Caroline P. Kerr
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Joseph J. Grudzinski
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Thanh Phuong Nguyen
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Reinier Hernandez
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jamey P. Weichert
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachary S. Morris
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
16
|
Nishri Y, Vatarescu M, Luz I, Epstein L, Dumančić M, Del Mare S, Shai A, Schmidt M, Deutsch L, Den RB, Kelson I, Keisari Y, Arazi L, Cooks T, Domankevich V. Diffusing alpha-emitters radiation therapy in combination with temozolomide or bevacizumab in human glioblastoma multiforme xenografts. Front Oncol 2022; 12:888100. [PMID: 36237307 PMCID: PMC9552201 DOI: 10.3389/fonc.2022.888100] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is at present an incurable disease with a 5-year survival rate of 5.5%, despite improvements in treatment modalities such as surgery, radiation therapy, chemotherapy [e.g., temozolomide (TMZ)], and targeted therapy [e.g., the antiangiogenic agent bevacizumab (BEV)]. Diffusing alpha-emitters radiation therapy (DaRT) is a new modality that employs radium-224-loaded seeds that disperse alpha-emitting atoms inside the tumor. This treatment was shown to be effective in mice bearing human-derived GBM tumors. Here, the effect of DaRT in combination with standard-of-care therapies such as TMZ or BEV was investigated. In a viability assay, the combination of alpha radiation with TMZ doubled the cytotoxic effect of each of the treatments alone in U87 cultured cells. A colony formation assay demonstrated that the surviving fraction of U87 cells treated by TMZ in combination with alpha irradiation was lower than was achieved by alpha- or x-ray irradiation as monotherapies, or by x-ray combined with TMZ. The treatment of U87-bearing mice with DaRT and TMZ delayed tumor development more than the monotherapies. Unlike other radiation types, alpha radiation did not increase VEGF secretion from U87 cells in culture. BEV treatment introduced several days after DaRT implantation improved tumor control, compared to BEV or DaRT as monotherapies. The combination was also shown to be superior when starting BEV administration prior to DaRT implantation in large tumors relative to the seed size. BEV induced a decrease in CD31 staining under DaRT treatment, increased the diffusive spread of 224Ra progeny atoms in the tumor tissue, and decreased their clearance from the tumor through the blood. Taken together, the combinations of DaRT with standard-of-care chemotherapy or antiangiogenic therapy are promising approaches, which may improve the treatment of GBM patients.
Collapse
Affiliation(s)
- Yossi Nishri
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
| | - Maayan Vatarescu
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Ishai Luz
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Lior Epstein
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Radiation Protection Department, Soreq Nuclear Research Center, Yavne, Israel
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Mirta Dumančić
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sara Del Mare
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
| | - Amit Shai
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
| | | | - Lisa Deutsch
- Biostatistics Department, BioStats Statistical Consulting Ltd., Maccabim, Israel
| | - Robert B. Den
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
- Department of Radiation Oncology, Urology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Itzhak Kelson
- School of Physics and Astronomy, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yona Keisari
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lior Arazi
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- *Correspondence: Lior Arazi, ; Tomer Cooks, ; Vered Domankevich,
| | - Tomer Cooks
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
- *Correspondence: Lior Arazi, ; Tomer Cooks, ; Vered Domankevich,
| | - Vered Domankevich
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
- *Correspondence: Lior Arazi, ; Tomer Cooks, ; Vered Domankevich,
| |
Collapse
|
17
|
Zhang J, Li F, Yin Y, Liu N, Zhu M, Zhang H, Liu W, Yang M, Qin S, Fan X, Yang Y, Zhang K, Yu F. Alpha radionuclide-chelated radioimmunotherapy promoters enable local radiotherapy/chemodynamic therapy to discourage cancer progression. Biomater Res 2022; 26:44. [PMID: 36076298 PMCID: PMC9461185 DOI: 10.1186/s40824-022-00290-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/28/2022] [Indexed: 12/07/2022] Open
Abstract
BACKGROUND Astatine-211 is an α-emitter with high-energy α-ray and high cytotoxicity for cancer cells. However, the targeted alpha therapy (TAT) also suffers from insufficient systematic immune activation, resulting in tumor metastasis and relapse. Combined immune checkpoint blockade (ICB) with chemodynamic therapy (CDT) could boost antitumor immunity, which may magnify the immune responses of TAT. This study aims to discourage tumor metastasis and relapse by tri-model TAT-CDT-ICB strategy. METHODS We successfully designed Mn-based radioimmunotherapy promoters (211At-ATE-MnO2-BSA), which are consisting of 211At, MnO2 and bovine serum albumin (BSA). The efficacy of 211At-ATE-MnO2-BSA was studied as monotherapy or in combination with anti-PD-L1 in both metastatic and relapse models. The immune effects of radioimmunotherapy promoters on cytotoxic T lymphocytes and dendritic cells (DCs) were analyzed by flow cytometry. Enzyme-linked immunosorbent assay and immunofluorescence were used to explore the underlying mechanism. RESULTS Such radioimmunotherapy promoters could not only enhance the therapeutic outcomes of TAT and CDT, but also induce robust anti-cancer immune activity by activating dendritic cells. More intriguingly, 211At-ATE-MnO2-BSA could effectively suppress the growths of primary tumors and distant tumors when combined with immune checkpoint inhibitors. CONCLUSIONS The tri-model TAT-CDT-ICB strategy provides a long-term immunological memory, which can protect against tumor rechallenge after eliminating original tumors. Therefore, this work presents a novel approach for TAT-CDT-ICB tri-modal cancer therapy with repressed metastasis and relapse in clinics.
Collapse
Affiliation(s)
- Jiajia Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China.,Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China.,Department of Medical Ultrasound and Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China
| | - Feize Li
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu, 610064, People's Republic of China
| | - Yuzhen Yin
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China.,Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China
| | - Ning Liu
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu, 610064, People's Republic of China
| | - Mengqin Zhu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China.,Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China
| | - Han Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China.,Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China
| | - Weihao Liu
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu, 610064, People's Republic of China
| | - Mengdie Yang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China.,Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China
| | - Shanshan Qin
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China.,Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China
| | - Xin Fan
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China.,Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China
| | - Yuanyou Yang
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu, 610064, People's Republic of China.
| | - Kun Zhang
- Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China. .,Department of Medical Ultrasound and Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China.
| | - Fei Yu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China. .,Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, People's Republic of China.
| |
Collapse
|
18
|
Salih S, Alkatheeri A, Alomaim W, Elliyanti A. Radiopharmaceutical Treatments for Cancer Therapy, Radionuclides Characteristics, Applications, and Challenges. Molecules 2022; 27:molecules27165231. [PMID: 36014472 PMCID: PMC9415873 DOI: 10.3390/molecules27165231] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Advances in the field of molecular biology have had an impact on biomedical applications, which provide greater hope for both imaging and therapeutics. Work has been intensified on the development of radionuclides and their application in radiopharmaceuticals (RPS) which will certainly influence and expand therapeutic approaches in the future treatment of patients. Alpha or beta particles and Auger electrons are used for therapy purposes, and each has advantages and disadvantages. The radionuclides labeled drug delivery system will deliver the particles to the specific targeting cell. Different radioligands can be chosen to uniquely target molecular receptors or intracellular components, making them suitable for personal patient-tailored therapy in modern cancer therapy management. Advances in nanotechnology have enabled nanoparticle drug delivery systems that can allow for specific multivalent attachment of targeted molecules of antibodies, peptides, or ligands to the surface of nanoparticles for therapy and imaging purposes. This review presents fundamental radionuclide properties with particular reference to tumor biology and receptor characteristic of radiopharmaceutical targeted therapy development.
Collapse
Affiliation(s)
- Suliman Salih
- Radiology and Medical Imaging Department, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates
- National Cancer Institute, University of Gezira, Wad Madani 2667, Sudan
| | - Ajnas Alkatheeri
- Radiology and Medical Imaging Department, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates
| | - Wijdan Alomaim
- Radiology and Medical Imaging Department, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates
| | - Aisyah Elliyanti
- Nuclear Medicine Division of Radiology Department, Faculty of Medicine, Universitas Andalas, Padang 25163, Indonesia
- Correspondence:
| |
Collapse
|
19
|
Howe A, Bhatavdekar O, Salerno D, Josefsson A, Pacheco-Torres J, Bhujwalla ZM, Gabrielson KL, Sgouros G, Sofou S. Combination of Carriers with Complementary Intratumoral Microdistributions of Delivered α-Particles May Realize the Promise for 225Ac in Large, Solid Tumors. J Nucl Med 2022; 63:1223-1230. [PMID: 34795012 PMCID: PMC9364351 DOI: 10.2967/jnumed.121.262992] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/09/2021] [Indexed: 02/03/2023] Open
Abstract
α-particle radiotherapy has already been shown to be impervious to most resistance mechanisms. However, in established (i.e., large, vascularized) soft-tissue lesions, the diffusion-limited penetration depths of radiolabeled antibodies or nanocarriers (≤50-80 μm) combined with the short range of α-particles (4-5 cell diameters) may result in only partial tumor irradiation, potentially limiting treatment efficacy. To address this challenge, we combined carriers with complementary intratumoral microdistributions of the delivered α-particles. We used the α-particle generator 225Ac, and we combined a tumor-responsive liposome (which, on tumor uptake, releases into the interstitium a highly diffusing form of its radioactive payload [225Ac-DOTA], potentially penetrating the deeper parts of tumors where antibodies do not reach) with a separately administered, less-penetrating radiolabeled antibody (irradiating the tumor perivascular regions where liposome contents clear too quickly). Methods: In a murine model with orthotopic human epidermal growth factor receptor 2-positive BT474 breast cancer xenografts, the biodistributions of each carrier were evaluated, and the control of tumor growth was monitored after administration of the same total radioactivity of 225Ac delivered by the 225Ac-DOTA-encapsulating liposomes, by the 225Ac-DOTA-SCN--labeled trastuzumab, and by both carriers at equally split radioactivities. Results: Tumor growth was significantly more inhibited when the same total injected radioactivity was divided between the 2 separate carriers than when delivered by either of the carriers alone. The combined carriers enabled more uniform intratumoral microdistributions of α-particles, at a tumor dose that was lower than the dose delivered by the antibody alone. Conclusion: This strategy demonstrates that more uniform microdistributions of the delivered α-particles within established solid tumors improve efficacy even at lower tumor doses. Augmentation of antibody-targeted α-particle therapies with tumor-responsive liposomes may address partial tumor irradiation, improving therapeutic effects.
Collapse
Affiliation(s)
- Alaina Howe
- Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Omkar Bhatavdekar
- Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Dominick Salerno
- Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Anders Josefsson
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Jesus Pacheco-Torres
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Zaver M. Bhujwalla
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Kathleen L. Gabrielson
- Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, Maryland; and
| | - George Sgouros
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Stavroula Sofou
- Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland;,Sidney Kimmel Comprehensive Cancer Center, Cancer Invasion and Metastasis Program, Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
20
|
Kiess AP, Hobbs RF, Bednarz B, Knox SJ, Meredith R, Escorcia FE. ASTRO's Framework for Radiopharmaceutical Therapy Curriculum Development for Trainees. Int J Radiat Oncol Biol Phys 2022; 113:719-726. [PMID: 35367328 DOI: 10.1016/j.ijrobp.2022.03.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 10/18/2022]
Abstract
In 2017, the American Society for Radiation Oncology (ASTRO) board of directors prioritized radiopharmaceutical therapy (RPT) as a leading area for new therapeutic development, and the ASTRO RPT workgroup was created. Herein, the workgroup has developed a framework for RPT curriculum development upon which education leaders can build to integrate this modality into radiation oncology resident education. Through this effort, the workgroup aims to provide a guide to ensure robust training in an emerging therapeutic area within the context of existing radiation oncology training in radiation biology, medical physics, and clinical radiation oncology. The framework first determines the core RPT knowledge required to select patients, prescribe, safely administer, and manage related adverse events. Then, it defines the most important topics for preparing residents for clinical RPT planning and delivery. This framework is designed as a tool to supplement the current training that exists for radiation oncology residents. The final document was approved by the ASTRO board of directors in the fall of 2021.
Collapse
Affiliation(s)
- Ana P Kiess
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland.
| | - Robert F Hobbs
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Bryan Bednarz
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Susan J Knox
- Department of Radiation Oncology, Stanford University Medical Center, Stanford, California
| | - Ruby Meredith
- Department of Radiation Oncology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Freddy E Escorcia
- Molecular Imaging Branch, Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
21
|
The risk of increasing tumor malignancy after PET diagnosis. CURRENT ISSUES IN PHARMACY AND MEDICAL SCIENCES 2022. [DOI: 10.2478/cipms-2022-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
This manuscript reviews evidences underlying the estimation of risk of malignancy enhancement of advanced aggressive cancers as a result of the gamma radiation emitted by tracers used in PET diagnostics. We conclude that among many cancers, such a phenomenon likely occurs, particularly in tumor cells with an aggressive biology in the advanced stages of their development, e.g. prostate cancer, melanoma and colorectal cancer. Moreover, we surmise based on gathered evidence that fluorine -18 (18F) labeled pharmaceuticals (18F-deoxyglucose and 18F-choline), commonly used in positron emission tomography (PET) can lead to malignancy enhancement of diagnosed cancer, manifesting as accelerated infiltration of the neighboring tissue, accelerated metastasis and/or radio- and chemotherapy resistance. In this review, some suggestions on future studies verifying this concept are also proposed. If our concerns are justified, it might be appropriate in the future to consider this assumption at the stage of deciding whether to undertake PET monitoring in some patients with advanced aggressive cancer.
Collapse
|
22
|
Du Y, Cortez A, Josefsson A, Zarisfi M, Krimins R, Liapi E, Nedrow JR. Preliminary evaluation of alpha-emitting radioembolization in animal models of hepatocellular carcinoma. PLoS One 2022; 17:e0261982. [PMID: 35061763 PMCID: PMC8782514 DOI: 10.1371/journal.pone.0261982] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 12/14/2021] [Indexed: 12/03/2022] Open
Abstract
Hepatocellular carcinoma is the most common primary liver cancer and the fifth most frequently diagnosed cancer worldwide. Most patients with advanced disease are offered non-surgical palliative treatment options. This work explores the first alpha-particle emitting radioembolization for the treatment and monitoring of hepatic tumors. Furthermore, this works demonstrates the first in vivo simultaneous multiple-radionuclide SPECT-images of the complex decay chain of an [225Ac]Ac-labeled agent using a clinical SPECT system to monitor the temporal distribution. A DOTA chelator was modified with a lipophilic moiety and radiolabeled with the α-particle emitter Actinium-225. The resulting agent, [225Ac]Ac-DOTA-TDA, was emulsified in ethiodized oil and evaluated in vivo in mouse model and the VX2 rabbit technical model of liver cancer. SPECT imaging was performed to monitor distribution of the TAT agent and the free daughters. The [225Ac]Ac-DOTA-TDA emulsion was shown to retain within the HEP2G tumors and VX2 tumor, with minimal uptake within normal tissue. In the mouse model, significant improvements in overall survival were observed. SPECT-imaging was able to distinguish between the Actinium-225 agent (Francium-221) and the loss of the longer lived daughter, Bismuth-213. An α-particle emitting TARE agent is capable of targeting liver tumors with minimal accumulation in normal tissue, providing a potential therapeutic agent for the treatment of hepatocellular carcinoma as well as a variety of hepatic tumors. In addition, SPECT-imaging presented here supports the further development of imaging methodology and protocols that can be incorporated into the clinic to monitor Actinium-225-labeled agents.
Collapse
Affiliation(s)
- Yong Du
- Russell H. Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Angel Cortez
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Anders Josefsson
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Mohammadreza Zarisfi
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Rebecca Krimins
- Russell H. Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Eleni Liapi
- Russell H. Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Jessie R. Nedrow
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| |
Collapse
|
23
|
Radiobiology of Targeted Alpha Therapy. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00093-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
24
|
King AP, Lin FI, Escorcia FE. Why bother with alpha particles? Eur J Nucl Med Mol Imaging 2021; 49:7-17. [PMID: 34175980 DOI: 10.1007/s00259-021-05431-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022]
Abstract
The approval of 223RaCl2 for cancer therapy in 2013 has heralded a resurgence of interest in the development of α-particle emitting radiopharmaceuticals. In the last decade, over a dozen α-emitting radiopharmaceuticals have entered clinical trials, spawned by strong preclinical studies. In this article, we explore the potential role of α-particle therapy in cancer treatment. We begin by providing a background for the basic principles of therapy with α-emitters, and we explore recent breakthroughs in therapy with α-emitting radionuclides, including conjugates with small molecules and antibodies. Finally, we discuss some outstanding challenges to the clinical adoption of α-therapies and potential strategies to address them.
Collapse
Affiliation(s)
- A Paden King
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20817, USA
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20817, USA
| | - Frank I Lin
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20817, USA
| | - Freddy E Escorcia
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20817, USA.
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20817, USA.
| |
Collapse
|
25
|
Hanaoka H, Ohshima Y, Suzuki H, Sasaki I, Watabe T, Ooe K, Watanabe S, Ishioka NS. Enhancing the Therapeutic Effect of 2- 211At-astato-α-methyl-L-phenylalanine with Probenecid Loading. Cancers (Basel) 2021; 13:cancers13215514. [PMID: 34771676 PMCID: PMC8583516 DOI: 10.3390/cancers13215514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 10/28/2021] [Indexed: 11/24/2022] Open
Abstract
Simple Summary To enhance the therapeutic effect of 2-211At-astato-α-methyl-L-phenylalanine (2-211At-AAMP), a radiopharmaceutical for targeted alpha therapy, we evaluated the effect of probenecid loading on its biodistribution and therapeutic effect in mice. Probenecid preloading significantly delayed the clearance of 2-211At-AAMP from the blood, increasing its accumulation in tumors. Consequently, the therapeutic effect of 2-211At-AAMP markedly improved. These results indicate that 2-211At-AAMP with probenecid loading is useful for the treatment of various types of cancers. Abstract L-type amino acid transporter 1 (LAT1) might be a useful target for tumor therapy since it is highly expressed in various types of cancers. We previously developed an astatine-211 (211At)-labeled amino acid derivative, 2-211At-astato-α-methyl-L-phenylalanine (2-211At-AAMP), and demonstrated its therapeutic potential for LAT1-positive cancers. However, the therapeutic effect of 2-211At-AAMP was insufficient, probably due to its low tumor retention. The preloading of probenecid, an organic anion transporter inhibitor, can delay the clearance of some amino acid tracers from the blood and consequently increase their accumulation in tumors. In this study, we evaluated the effect of probenecid preloading on the biodistribution and therapeutic effect of 2-211At-AAMP in mice. In biodistribution studies, the blood radioactivity of 2-211At-AAMP significantly increased with probenecid preloading. Consequently, the accumulation of 2-211At-AAMP in tumors was significantly higher with probenecid than without probenecid loading. In a therapeutic study, tumor growth was suppressed by 2-211At-AAMP with probenecid, and the tumor volume was significantly lower in the treatment group than in the untreated control group from day 2 to day 30 (end of the follow-up period) after treatment. These results indicate that probenecid loading could improve the therapeutic effect of 2-211At-AAMP by increasing its accumulation in tumors.
Collapse
Affiliation(s)
- Hirofumi Hanaoka
- Faculty of Medicine, Kansai Medical University, 2-5-1 Shin-machi, Hirakata 573-1010, Osaka, Japan
- Department of Radiotheranostics, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi 371-8511, Gunma, Japan
- Correspondence: ; Tel.: +81-72-804-2452
| | - Yasuhiro Ohshima
- Department of Radiation-Applied Biology Research, Quantum Beam Science Research Directorate, National Institute for Quantum Science and Technology, 1233 Watanuki-machi, Takasaki 370-1292, Gunma, Japan; (Y.O.); (I.S.); (S.W.); (N.S.I.)
| | - Hiroyuki Suzuki
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Science, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Chiba, Japan;
| | - Ichiro Sasaki
- Department of Radiation-Applied Biology Research, Quantum Beam Science Research Directorate, National Institute for Quantum Science and Technology, 1233 Watanuki-machi, Takasaki 370-1292, Gunma, Japan; (Y.O.); (I.S.); (S.W.); (N.S.I.)
| | - Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, 1-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (T.W.); (K.O.)
| | - Kazuhiro Ooe
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, 1-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (T.W.); (K.O.)
| | - Shigeki Watanabe
- Department of Radiation-Applied Biology Research, Quantum Beam Science Research Directorate, National Institute for Quantum Science and Technology, 1233 Watanuki-machi, Takasaki 370-1292, Gunma, Japan; (Y.O.); (I.S.); (S.W.); (N.S.I.)
| | - Noriko S. Ishioka
- Department of Radiation-Applied Biology Research, Quantum Beam Science Research Directorate, National Institute for Quantum Science and Technology, 1233 Watanuki-machi, Takasaki 370-1292, Gunma, Japan; (Y.O.); (I.S.); (S.W.); (N.S.I.)
| |
Collapse
|
26
|
Synthesis and Properties of Targeted Radioisotope Carriers Based on Poly(Acrylic Acid) Nanogels. Pharmaceutics 2021; 13:pharmaceutics13081240. [PMID: 34452201 PMCID: PMC8400054 DOI: 10.3390/pharmaceutics13081240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/01/2022] Open
Abstract
Radiation crosslinking was employed to obtain nanocarriers based on poly(acrylic acid)—PAA—for targeted delivery of radioactive isotopes. These nanocarriers are internally crosslinked hydrophilic macromolecules—nanogels—bearing carboxylic groups to facilitate functionalization. PAA nanogels were conjugated with an engineered bombesin-derivative—oligopeptide combined with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetate chelating moiety, aimed to provide selective radioligand transport. 4-(4,6-Dimethoxy-1,3,5-triazin-2-yl)-4-methylmorpholinium (DMTMM) toluene-4-sulfonate was used as the coupling agent. After tests on a model amine—p-toluidine—both commercial and home-synthesized DOTA-bombesin were successfully coupled to the nanogels and the obtained products were characterized. The radiolabeling efficiency of nanocarriers with 177Lu, was chromatographically tested. The results provide a proof of concept for the synthesis of radiation-synthesized nanogel-based radioisotope nanocarriers for theranostic applications.
Collapse
|
27
|
Zaid NRR, Kletting P, Winter G, Beer AJ, Glatting G. A Whole-Body Physiologically Based Pharmacokinetic Model for Alpha Particle Emitting Bismuth in Rats. Cancer Biother Radiopharm 2021; 37:41-46. [PMID: 34185608 DOI: 10.1089/cbr.2021.0028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction/Aim: α particle emitting bismuth (212Bi) as decay product of 212Pb-labeled pharmaceuticals has been effective in targeted α particle therapy (TAT). Estimating the contribution of 212Bi released from its chelator to the absorbed doses in nontarget tissues is challenging in TAT. Physiologically based pharmacokinetic (PBPK) modeling can help overcome this limitation. Therefore, a whole-body 212Bi-PBPK model was developed to describe the pharmacokinetics (PKs) of 212Bi in rats. Materials and Methods: The rat 212Bi-PBPK model was implemented using the modeling software SAAM II with data and parameter values from the literature. Besides other mechanisms, 212Bi interactions with red blood cells, high molecular weight plasma protein, and intracellular biological thiols are described. Important PK parameters were fitted to time-activity data. Absorbed dose coefficients (ADCs) were calculated for injecting 0.774 fmol of 212Bi. Results: 212Bi uptake rates of liver, bone, small intestine, bone marrow, skin, and muscle were (0.86 ± 0.13), (3.85 ± 0.63), (0.27 ± 0.05), (1.44 ± 0.29), (0.04 ± 0.01), and (0.007 ± 0.007) per min with corresponding ADCs of 0.09, 0.03, 0.03, 0.07, 0.01, and 0.003 mGy/kBq, respectively. An ADC of 0.70 mGy/kBq was determined for kidneys. Conclusion: Kidneys are the dose-limiting organs in 212Bi-based TAT. The 212Bi-PBPK model is an effective tool to investigate the 212Bi biodistribution in murine models. Integrating the 212Bi-PBPK model into other murine and human PBPK models of α particle generators can help study the efficacy and safety of TAT.
Collapse
Affiliation(s)
- Nouran R R Zaid
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.,Department of Biomedical Sciences, Biophysics and Medical Imaging Program, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Peter Kletting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.,Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Gordon Winter
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Gerhard Glatting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.,Department of Nuclear Medicine, Ulm University, Ulm, Germany
| |
Collapse
|
28
|
Prasad A, Nair R, Bhatavdekar O, Howe A, Salerno D, Sempkowski M, Josefsson A, Pacheco-Torres J, Bhujwalla ZM, Gabrielson KL, Sgouros G, Sofou S. Transport-driven engineering of liposomes for delivery of α-particle radiotherapy to solid tumors: effect on inhibition of tumor progression and onset delay of spontaneous metastases. Eur J Nucl Med Mol Imaging 2021; 48:4246-4258. [PMID: 34117896 DOI: 10.1007/s00259-021-05406-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/10/2021] [Indexed: 12/31/2022]
Abstract
PURPOSE Highly cytotoxic α-particle radiotherapy delivered by tumor-selective nanocarriers is evaluated on metastatic Triple Negative Breast Cancer (TNBC). On vascularized tumors, the limited penetration of nanocarriers (<50-80 μm) combined with the short range of α-particles (40-100 μm) may, however, result in only partial tumor irradiation, compromising efficacy. Utilizing the α-particle emitter Actinium-225 (225Ac), we studied how the therapeutic potential of a general delivery strategy using nanometer-sized engineered liposomes was affected by two key transport-driven properties: (1) the release from liposomes, when in the tumor interstitium, of the highly diffusing 225Ac-DOTA that improves the uniformity of tumor irradiation by α-particles and (2) the adhesion of liposomes on the tumors' ECM that increases liposomes' time-integrated concentrations within tumors and, therefore, the tumor-delivered radioactivities. METHODS On an orthotopic MDA-MB-231 TNBC murine model forming spontaneous metastases, we evaluated the maximum tolerated dose (MTD), biodistributions, and control of tumor growth and/or spreading after administration of 225Ac-DOTA-encapsulating liposomes, with different combinations of the two transport-driven properties. RESULTS At 83% of MTD, 225Ac-DOTA-encapsulating liposomes with both properties (1) eliminated formation of spontaneous metastases and (2) best inhibited the progression of orthotopic xenografts, compared to liposomes lacking one or both properties. These findings were primarily affected by the extent of uniformity of the intratumoral microdistributions of 225Ac followed by the overall tumor uptake of radioactivity. At the MTD, long-term toxicities were not detected 9.5 months post administration. CONCLUSION Our findings demonstrate the potential of a general, transport-driven strategy enabling more uniform and prolonged solid tumor irradiation by α-particles without cell-specific targeting.
Collapse
Affiliation(s)
- Aprameya Prasad
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Rajiv Nair
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Omkar Bhatavdekar
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Alaina Howe
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Dominick Salerno
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Anders Josefsson
- The Russell H. Morgan Department of Radiology and Radiological Science, Cancer Invasion & Metastasis Program, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jesus Pacheco-Torres
- The Russell H. Morgan Department of Radiology and Radiological Science, Cancer Invasion & Metastasis Program, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- The Russell H. Morgan Department of Radiology and Radiological Science, Cancer Invasion & Metastasis Program, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kathleen L Gabrielson
- Molecular and Comparative Pathobiology, Cancer Invasion & Metastasis Program, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - George Sgouros
- The Russell H. Morgan Department of Radiology and Radiological Science, Cancer Invasion & Metastasis Program, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Stavroula Sofou
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA. .,Sidney Kimmel Comprehensive Cancer Center, Cancer Invasion & Metastasis Program, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA. .,ChemBE, Johns Hopkins University, 3400 North Charles Street, Maryland Hall 221, Baltimore, MD, 21218, USA.
| |
Collapse
|
29
|
Iqbal Z, Arafa ESA, Kanwal Z, Murtaza G. Smart solution of severe problems: Radiolabeled nanocarriers for cancer imaging and therapy. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
30
|
Affiliation(s)
- John M Buatti
- University of Iowa, Department of Radiation Oncology, Iowa,.
| | - Ana P Kiess
- Johns Hopkins University, Department of Radiation Oncology and Molecular Radiation Sciences, Baltimore, Maryland
| |
Collapse
|
31
|
Ohshima Y, Suzuki H, Hanaoka H, Sasaki I, Watanabe S, Haba H, Arano Y, Tsushima Y, Ishioka NS. Preclinical evaluation of new α-radionuclide therapy targeting LAT1: 2-[ 211At]astato-α-methyl-L-phenylalanine in tumor-bearing model. Nucl Med Biol 2020; 90-91:15-22. [PMID: 32916470 DOI: 10.1016/j.nucmedbio.2020.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Targeted α-radionuclide therapy has attracted attention as a promising therapy for refractory cancers. However, the application is limited to certain types of cancer. Since L-type amino acid transporter 1 (LAT1) is highly expressed in various human cancers, we prepared an LAT1-selective α-radionuclide-labeled amino acid analog, 2-[211At]astato-α-methyl-L-phenylalanine (2-[211At]AAMP), and evaluated its potential as a therapeutic agent. METHODS 2-[211At]AAMP was prepared from the stannyl precursor. Stability of 2-[211At]AAMP was evaluated both in vitro and in vivo. In vitro studies using an LAT1-expressing human ovarian cancer cell line, SKOV3, were performed to evaluate cellular uptake and cytotoxicity of 2-[211At]AAMP. Biodistribution and therapeutic studies in SKOV3-bearing mice were performed after intravenous injection of 2-[211At]AAMP. RESULTS 2-[211At]AAMP was stable in murine plasma in vitro and excreted intact into urine. Cellular uptake of 2-[211At]AAMP was inhibited by treatment with an LAT1-selective inhibitor. After 24 h incubation, 2-[211At]AAMP suppressed clonogenic growth at 10 kBq/ml, and induced cell death and DNA double-strand breaks at 25 kBq/ml. When injected into mice, 2-[211At]AAMP exhibited peak accumulation in the tumor at 30 min postinjection, and radioactivity levels in the tumor were retained up to 60 min. The majority of the radioactivity was rapidly eliminated from the body into urine in an intact form immediately after injection. 2-[211At]AAMP significantly improved the survival of mice (P < 0.05) without serious side effects. CONCLUSION 2-[211At]AAMP showed α-radiation-dependent cellular growth inhibition after it was taken up via LAT1. In addition, 2-[211At]AAMP had a beneficial effect on survival in vivo. These findings suggest that 2-[211At]AAMP would be useful for the treatment of LAT1-positive cancer. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE This is the first report of an LAT1-targeting radiopharmaceutical for α-radionuclide therapy; this agent would be applicable for the treatment of various types of cancer.
Collapse
Affiliation(s)
- Yasuhiro Ohshima
- Department of Radiation-Applied Biology Research, Quantum Beam Science Research Directorate, National Institute for Quantum and Radiological Science and Technology, 1233 Watanuki-machi, Takasaki, Gunma 370-1292, Japan.
| | - Hiroyuki Suzuki
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Science, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8675, Japan
| | - Hirofumi Hanaoka
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Ichiro Sasaki
- Department of Radiation-Applied Biology Research, Quantum Beam Science Research Directorate, National Institute for Quantum and Radiological Science and Technology, 1233 Watanuki-machi, Takasaki, Gunma 370-1292, Japan
| | - Shigeki Watanabe
- Department of Radiation-Applied Biology Research, Quantum Beam Science Research Directorate, National Institute for Quantum and Radiological Science and Technology, 1233 Watanuki-machi, Takasaki, Gunma 370-1292, Japan
| | - Hiromitsu Haba
- Superheavy Element Production Team, Nishina Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yasushi Arano
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Science, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8675, Japan
| | - Yoshito Tsushima
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Noriko S Ishioka
- Department of Radiation-Applied Biology Research, Quantum Beam Science Research Directorate, National Institute for Quantum and Radiological Science and Technology, 1233 Watanuki-machi, Takasaki, Gunma 370-1292, Japan
| |
Collapse
|
32
|
Cheal SM, McDevitt MR, Santich BH, Patel M, Yang G, Fung EK, Veach DR, Bell M, Ahad A, Vargas DB, Punzalan B, Pillarsetty NVK, Xu H, Guo HF, Monette S, Michel AO, Piersigilli A, Scheinberg DA, Ouerfelli O, Cheung NKV, Larson SM. Alpha radioimmunotherapy using 225Ac-proteus-DOTA for solid tumors - safety at curative doses. Theranostics 2020; 10:11359-11375. [PMID: 33052220 PMCID: PMC7546012 DOI: 10.7150/thno.48810] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
This is the initial report of an α-based pre-targeted radioimmunotherapy (PRIT) using 225Ac and its theranostic pair, 111In. We call our novel tumor-targeting DOTA-hapten PRIT system "proteus-DOTA" or "Pr." Herein we report the first results of radiochemistry development, radiopharmacology, and stoichiometry of tumor antigen binding, including the role of specific activity, anti-tumor efficacy, and normal tissue toxicity with the Pr-PRIT approach (as α-DOTA-PRIT). A series of α-DOTA-PRIT therapy studies were performed in three solid human cancer xenograft models of colorectal cancer (GPA33), breast cancer (HER2), and neuroblastoma (GD2), including evaluation of chronic toxicity at ~20 weeks of select survivors. Methods: Preliminary biodistribution experiments in SW1222 tumor-bearing mice revealed that 225Ac could not be efficiently pretargeted with current DOTA-Bn hapten utilized for 177Lu or 90Y, leading to poor tumor uptake in vivo. Therefore, we synthesized Pr consisting of an empty DOTA-chelate for 225Ac, tethered via a short polyethylene glycol linker to a lutetium-complexed DOTA for picomolar anti-DOTA chelate single-chain variable fragment (scFv) binding. Pr was radiolabeled with 225Ac and its imaging surrogate, 111In. In vitro studies verified anti-DOTA scFv recognition of [225Ac]Pr, and in vivo biodistribution and clearance studies were performed to evaluate hapten suitability and in vivo targeting efficiency. Results: Intravenously (i.v.) administered 225Ac- or 111In-radiolabeled Pr in mice showed rapid renal clearance and minimal normal tissue retention. In vivo pretargeting studies show high tumor accumulation of Pr (16.71 ± 5.11 %IA/g or 13.19 ± 3.88 %IA/g at 24 h p.i. for [225Ac]Pr and [111In]Pr, respectively) and relatively low uptake in normal tissues (all average ≤ 1.4 %IA/g at 24 h p.i.). Maximum tolerated dose (MTD) was not reached for either [225Ac]Pr alone or pretargeted [225Ac]Pr at administered activities up to 296 kBq/mouse. Single-cycle treatment consisting of α-DOTA-PRIT with either huA33-C825 bispecific anti-tumor/anti-DOTA-hapten antibody (BsAb), anti-HER2-C825 BsAb, or hu3F8-C825 BsAb for targeting GPA33, HER2, or GD2, respectively, was highly effective. In the GPA33 model, no complete responses (CRs) were observed but prolonged overall survival of treated animals was 42 d for α-DOTA-PRIT vs. 25 d for [225Ac]Pr only (P < 0.0001); for GD2, CRs (7/7, 100%) and histologic cures (4/7, 57%); and for HER2, CRs (7/19, 37%) and histologic cures (10/19, 56%) with no acute or chronic toxicity. Conclusions: [225Ac]Pr and its imaging biomarker [111In]Pr demonstrate optimal radiopharmacologic behavior for theranostic applications of α-DOTA-PRIT. For this initial evaluation of efficacy and toxicity, single-cycle treatment regimens were performed in all three systems. Histologic toxicity was not observed, so MTD was not observed. Prolonged overall survival, CRs, and histologic cures were observed in treated animals. In comparison to RIT with anti-tumor IgG antibodies, [225Ac]Pr has a much improved safety profile. Ultimately, these data will be used to guide clinical development of toxicity and efficacy studies of [225Ac]Pr, with the goal of delivering massive lethal doses of radiation to achieve a high probability of cure without toxicity.
Collapse
|
33
|
Lee JY, Mushtaq S, Park JE, Shin HS, Lee SY, Jeon J. Radioanalytical Techniques to Quantitatively Assess the Biological Uptake and In Vivo Behavior of Hazardous Substances. Molecules 2020; 25:molecules25173985. [PMID: 32882977 PMCID: PMC7504758 DOI: 10.3390/molecules25173985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 12/25/2022] Open
Abstract
Concern about environmental exposure to hazardous substances has grown over the past several decades, because these substances have adverse effects on human health. Methods used to monitor the biological uptake of hazardous substances and their spatiotemporal behavior in vivo must be accurate and reliable. Recent advances in radiolabeling chemistry and radioanalytical methodologies have facilitated the quantitative analysis of toxic substances, and whole-body imaging can be achieved using nuclear imaging instruments. Herein, we review recent literature on the radioanalytical methods used to study the biological distribution, changes in the uptake and accumulation of hazardous substances, including industrial chemicals, nanomaterials, and microorganisms. We begin with an overview of the radioisotopes used to prepare radiotracers for in vivo experiments. We then summarize the results of molecular imaging studies involving radiolabeled toxins and their quantitative assessment. We conclude the review with perspectives on the use of radioanalytical methods for future environmental research.
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Environmental and Safety Engineering, Ajou University, Suwon 16499, Korea;
| | - Sajid Mushtaq
- Department of Nuclear Engineering, Pakistan Institute of Engineering and Applied Sciences, Islamabad 45650, Pakistan;
| | - Jung Eun Park
- Department of Applied Chemistry, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Korea;
| | - Hee Soon Shin
- Division of Functional Food Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Korea; (H.S.S.); (S.-Y.L.)
- Food Biotechnology Program, University of Science and Technology, Daejeon 34113, Korea
| | - So-Young Lee
- Division of Functional Food Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Korea; (H.S.S.); (S.-Y.L.)
- Food Biotechnology Program, University of Science and Technology, Daejeon 34113, Korea
| | - Jongho Jeon
- Department of Applied Chemistry, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Korea;
- Correspondence: ; Tel.: +82-53-950-5584
| |
Collapse
|
34
|
Reissig F, Zarschler K, Hübner R, Pietzsch H, Kopka K, Mamat C. Sub-10 nm Radiolabeled Barium Sulfate Nanoparticles as Carriers for Theranostic Applications and Targeted Alpha Therapy. ChemistryOpen 2020; 9:797-805. [PMID: 32775141 PMCID: PMC7397357 DOI: 10.1002/open.202000126] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/08/2020] [Indexed: 11/13/2022] Open
Abstract
The treatment of cancer patients with α-particle-emitting therapeutics continues to gain in importance and relevance. The range of radiopharmaceutically relevant α-emitters is limited to a few radionuclides, as stable chelators or carrier systems for safe transport of the radioactive cargo are often lacking. Encapsulation of α-emitters into solid inorganic systems can help to diversify the portfolio of candidate radionuclides, provided, that these nanomaterials effectively retain both the parent and the recoil daughters. We therefore focus on designing stable and defined nanocarrier-based systems for various clinically relevant radionuclides, including the promising α-emitting radionuclide 224Ra. Hence, sub-10 nm barium sulfate nanocontainers were prepared and different radiometals like 89Zr, 111In, 131Ba, 177Lu or 224Ra were incorporated. Our system shows stabilities of >90 % regarding the radiometal release from the BaSO4 matrix. Furthermore, we confirm the presence of surface-exposed amine functionalities as well as the formation of a biomolecular corona.
Collapse
Affiliation(s)
- Falco Reissig
- Institute of Radiopharmaceutical Cancer Research Helmholtz-ZentrumDresden-RossendorfBautzner Landstraße 40001328DresdenGermany
- Faculty of Chemistry and Food ChemistryTechnische Universität Dresden01062DresdenGermany
| | - Kristof Zarschler
- Institute of Radiopharmaceutical Cancer Research Helmholtz-ZentrumDresden-RossendorfBautzner Landstraße 40001328DresdenGermany
| | - René Hübner
- Institute of Ion Beam Physics and Materials ResearchHelmholtz-ZentrumDresden-RossendorfBautzner Landstraße 40001328DresdenGermany
| | - Hans‐Jürgen Pietzsch
- Institute of Radiopharmaceutical Cancer Research Helmholtz-ZentrumDresden-RossendorfBautzner Landstraße 40001328DresdenGermany
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research Helmholtz-ZentrumDresden-RossendorfBautzner Landstraße 40001328DresdenGermany
- Faculty of Chemistry and Food ChemistryTechnische Universität Dresden01062DresdenGermany
| | - Constantin Mamat
- Institute of Radiopharmaceutical Cancer Research Helmholtz-ZentrumDresden-RossendorfBautzner Landstraße 40001328DresdenGermany
- Faculty of Chemistry and Food ChemistryTechnische Universität Dresden01062DresdenGermany
| |
Collapse
|
35
|
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
|
36
|
Farzin A, Etesami SA, Quint J, Memic A, Tamayol A. Magnetic Nanoparticles in Cancer Therapy and Diagnosis. Adv Healthc Mater 2020; 9:e1901058. [PMID: 32196144 PMCID: PMC7482193 DOI: 10.1002/adhm.201901058] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/15/2020] [Indexed: 12/16/2022]
Abstract
There is urgency for the development of nanomaterials that can meet emerging biomedical needs. Magnetic nanoparticles (MNPs) offer high magnetic moments and surface-area-to-volume ratios that make them attractive for hyperthermia therapy of cancer and targeted drug delivery. Additionally, they can function as contrast agents for magnetic resonance imaging (MRI) and can improve the sensitivity of biosensors and diagnostic tools. Recent advancements in nanotechnology have resulted in the realization of the next generation of MNPs suitable for these and other biomedical applications. This review discusses methods utilized for the fabrication and engineering of MNPs. Recent progress in the use of MNPs for hyperthermia therapy, controlling drug release, MRI, and biosensing is also critically reviewed. Finally, challenges in the field and potential opportunities for the use of MNPs toward improving their properties are discussed.
Collapse
Affiliation(s)
- A. Farzin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA
| | - S. Alireza Etesami
- Department of Mechanical Engineering, The University of Memphis. Memphis, TN 38152, USA
| | - Jacob Quint
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Adnan Memic
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT, 06030, USA
| | - Ali Tamayol
- Division of Engineering in Medicine Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT, 06030, USA
| |
Collapse
|
37
|
Atomic Nanogenerators in Targeted Alpha Therapies: Curie's Legacy in Modern Cancer Management. Pharmaceuticals (Basel) 2020; 13:ph13040076. [PMID: 32340103 PMCID: PMC7243103 DOI: 10.3390/ph13040076] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Atomic in vivo nanogenerators such as actinium-225, thorium-227, and radium-223 are of increasing interest and importance in the treatment of patients with metastatic cancer diseases. This is due to their peculiar physical, chemical, and biological characteristics, leading to astonishing responses in otherwise resistant patients. Nevertheless, there are still a few obstacles and hurdles to be overcome that hamper the broader utilization in the clinical setting. Next to the limited supply and relatively high costs, the in vivo complex stability and the fate of the recoiling daughter radionuclides are substantial problems that need to be solved. In radiobiology, the mechanisms underlying treatment efficiency, possible resistance mechanisms, and late side effect occurrence are still far from being understood and need to be unraveled. In this review, the current knowledge on the scientific and clinical background of targeted alpha therapies is summarized. Furthermore, open issues and novel approaches with a focus on the future perspective are discussed. Once these are unraveled, targeted alpha therapies with atomic in vivo nanogenerators can be tailored to suit the needs of each patient when applying careful risk stratification and combination therapies. They have the potential to become one of the major treatment pillars in modern cancer management.
Collapse
|
38
|
Li L, Rousseau J, Jaraquemada-Peláez MDG, Wang X, Robertson A, Radchenko V, Schaffer P, Lin KS, Bénard F, Orvig C. 225Ac-H 4py4pa for Targeted Alpha Therapy. Bioconjug Chem 2020; 32:1348-1363. [PMID: 32216377 DOI: 10.1021/acs.bioconjchem.0c00171] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Herein, we present the syntheses and characterization of a new undecadendate chelator, H4py4pa, and its bifunctional analog H4py4pa-phenyl-NCS, conjugated to the monoclonal antibody, Trastuzumab, which targets the HER2+ cancer. H4py4pa possesses excellent affinity for 225Ac (α, t1/2 = 9.92 d) for targeted alpha therapy (TAT), where quantitative radiolabeling yield was achieved at ambient temperature, pH = 7, in 30 min at 10-6 M chelator concentration, leading to a complex highly stable in mouse serum for at least 9 d. To investigate the chelation of H4py4pa with large metal ions, lanthanum (La3+), which is the largest nonradioactive metal of the lanthanide series, was adopted as a surrogate for 225Ac to enable a series of nonradioactive chemical studies. In line with the 1H NMR spectrum, the DFT (density functional theory)-calculated structure of the [La(py4pa)]- anion possessed a high degree of symmetry, and the La3+ ion was secured by two distinct pairs of picolinate arms. Furthermore, the [La(py4pa)]- complex also demonstrated a superb thermodynamic stability (log K[La(py4pa)]- ∼ 20.33, pLa = 21.0) compared to those of DOTA (log K[La(DOTA)]- ∼ 24.25, pLa = 19.2) or H2macropa (log K[La(macropa)]- = 14.99, pLa ∼ 8.5). Moreover, the functional versatility offered by the bifunctional py4pa precursor permits facile incorporation of various linkers for bioconjugation through direct nucleophilic substitution. In this work, a short phenyl-NCS linker was incorporated to tether H4py4pa to Trastuzumab. Radiolabeling studies, in vitro serum stability, and animal studies were performed in parallel with the DOTA-benzyl-Trastuzumab. Both displayed excellent in vivo stability and tumor specificity.
Collapse
Affiliation(s)
- Lily Li
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.,Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada
| | - Julie Rousseau
- Department of Molecular Oncology, BC Cancer, 675 West 10th Avenue, Vancouver, British Columbia V5Z 1L3, Canada
| | - María de Guadalupe Jaraquemada-Peláez
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Xiaozhu Wang
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Andrew Robertson
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada.,Department of Physics and Astronomy, University of British Columbia, 325-6224 Agricultural Road, Vancouver, British Columbia V6T 1Z1, Canada
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada.,Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada.,Department of Radiology, University of British Columbia, 2775 Laurel Street, Vancouver, British Columbia V5Z 1M9, Canada.,Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer, 675 West 10th Avenue, Vancouver, British Columbia V5Z 1L3, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer, 675 West 10th Avenue, Vancouver, British Columbia V5Z 1L3, Canada
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| |
Collapse
|
39
|
Savastano M, Fiaschi M, Ferraro G, Gratteri P, Mariani P, Bianchi A, Bazzicalupi C. Sensing Zn 2+ in Aqueous Solution with a Fluorescent Scorpiand Macrocyclic Ligand Decorated with an Anthracene Bearing Tail. Molecules 2020; 25:E1355. [PMID: 32192025 PMCID: PMC7146481 DOI: 10.3390/molecules25061355] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/13/2020] [Accepted: 03/15/2020] [Indexed: 01/16/2023] Open
Abstract
Synthesis of the new scorpiand ligand L composed of a [9]aneN3 macrocyclic ring bearing a CH2CH2NHCH2-anthracene tail is reported. L forms both cation (Zn2+) and anion (phosphate, benzoate) complexes. In addition, the zinc complexes of L bind these anions. The equilibrium constants for ligand protonation and complex formation were determined in 0.1 M NaCl aqueous solution at 298.1 ± 0.1 K by means of potentiometric (pH-metric) titrations. pH Controlled coordination/detachment of the ligand tail to Zn2+ switch on and off the fluorescence emission from the anthracene fluorophore. Accordingly, L is able to sense Zn2+ in the pH range 6-10 down to nM concentrations of the metal ion. L can efficiently sense Zn2+ even in the presence of large excess of coordinating anions, such as cyanide, sulphide, phosphate and benzoate, despite their ability to bind the metal ion.
Collapse
Affiliation(s)
- Matteo Savastano
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia, 3-13, 50019 Sesto Fiorentino, Italy; (M.S.); (M.F.); (G.F.); (P.M.); (C.B.)
| | - Matteo Fiaschi
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia, 3-13, 50019 Sesto Fiorentino, Italy; (M.S.); (M.F.); (G.F.); (P.M.); (C.B.)
| | - Giovanni Ferraro
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia, 3-13, 50019 Sesto Fiorentino, Italy; (M.S.); (M.F.); (G.F.); (P.M.); (C.B.)
| | - Paola Gratteri
- Department of NEUROFARBA-Pharmaceutical and Nutraceutical section, and Laboratory of Molecular Modeling Cheminformatics & QSAR, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy;
| | - Palma Mariani
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia, 3-13, 50019 Sesto Fiorentino, Italy; (M.S.); (M.F.); (G.F.); (P.M.); (C.B.)
| | - Antonio Bianchi
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia, 3-13, 50019 Sesto Fiorentino, Italy; (M.S.); (M.F.); (G.F.); (P.M.); (C.B.)
| | - Carla Bazzicalupi
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia, 3-13, 50019 Sesto Fiorentino, Italy; (M.S.); (M.F.); (G.F.); (P.M.); (C.B.)
| |
Collapse
|
40
|
Nimmagadda S, Penet MF. Ovarian Cancer Targeted Theranostics. Front Oncol 2020; 9:1537. [PMID: 32039018 PMCID: PMC6985364 DOI: 10.3389/fonc.2019.01537] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/19/2019] [Indexed: 11/20/2022] Open
Abstract
Ovarian cancer is a leading cause of death from gynecological malignancies. Although the prognosis is quite favorable if detected at an early stage, the vast majority of cases are diagnosed at an advanced stage, when 5-year survival rates are only 30–40%. Most recurrent ovarian tumors are resistant to traditional therapies underscoring the need for new therapeutic options. Theranostic agents, that combine diagnostic and therapeutic capabilities, are being explored to better detect, diagnose and treat ovarian cancer. To minimize morbidity, improve survival rates, and eventually cure patients, new strategies are needed for early detection and for delivering specifically anticancer therapies to tumor sites. In this review we will discuss various molecular imaging modalities and targets that can be used for imaging, therapeutic and theranostic agent development for improved diagnosis and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Sridhar Nimmagadda
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Marie-France Penet
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
41
|
De Vincentis G, Gerritsen W, Gschwend JE, Hacker M, Lewington V, O'Sullivan JM, Oya M, Pacilio M, Parker C, Shore N, Sartor O. Advances in targeted alpha therapy for prostate cancer. Ann Oncol 2019; 30:1728-1739. [PMID: 31418764 PMCID: PMC6927314 DOI: 10.1093/annonc/mdz270] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Amongst therapeutic radiopharmaceuticals, targeted alpha therapy (TαT) can deliver potent and local radiation selectively to cancer cells as well as the tumor microenvironment and thereby control cancer while minimizing toxicity. In this review, we discuss the history, progress, and future potential of TαT in the treatment of prostate cancer, including dosimetry-individualized treatment planning, combinations with small-molecule therapies, and conjugation to molecules directed against antigens expressed by prostate cancer cells, such as prostate-specific membrane antigen (PSMA) or components of the tumor microenvironment. A clinical proof of concept that TαT is efficacious in treating bone-metastatic castration-resistant prostate cancer has been demonstrated by radium-223 via improved overall survival and long-term safety/tolerability in the phase III ALSYMPCA trial. Dosimetry calculation and pharmacokinetic measurements of TαT provide the potential for optimization and individualized treatment planning for a precision medicine-based cancer management paradigm. The ability to combine TαTs with other agents, including chemotherapy, androgen receptor-targeting agents, DNA repair inhibitors, and immuno-oncology agents, is under investigation. Currently, TαTs that specifically target prostate cancer cells expressing PSMA represents a promising therapeutic approach. Both PSMA-targeted actinium-225 and thorium-227 conjugates are under investigation. The described clinical benefit, safety and tolerability of radium-223 and the recent progress in TαT trial development suggest that TαT occupies an important new role in prostate cancer treatment. Ongoing studies with newer dosimetry methods, PSMA targeting, and novel approaches to combination therapies should expand the utility of TαT in prostate cancer treatment.
Collapse
Affiliation(s)
- G De Vincentis
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, Rome, Italy
| | - W Gerritsen
- Department of Medical Oncology, Radboud UMC, Nijmegen, The Netherlands
| | - J E Gschwend
- Department of Urology, Technical University of Munich, Rechts der Isar Medical Center, Munich, Germany
| | - M Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - V Lewington
- Department of Imaging Sciences and Biomedical Engineering, King's College, London, UK
| | - J M O'Sullivan
- Center for Cancer Research and Cell Biology, Queen's University Belfast, Northern Ireland Cancer Center, Belfast City Hospital, Belfast, Northern Ireland
| | - M Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - M Pacilio
- Medical Physics Department, "Policlinico Umberto I" University Hospital, Rome, Italy
| | - C Parker
- The Royal Marsden Hospital, Sutton, UK
| | - N Shore
- Carolina Urologic Research Center, Myrtle Beach
| | - O Sartor
- Tulane Cancer Center, Tulane University, New Orleans, USA.
| |
Collapse
|
42
|
Ferrier MG, Radchenko V. An Appendix of Radionuclides Used in Targeted Alpha Therapy. J Med Imaging Radiat Sci 2019; 50:S58-S65. [PMID: 31427258 DOI: 10.1016/j.jmir.2019.06.051] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Maryline G Ferrier
- Lawrence Livermore National Laboratory, Nuclear and Chemical Sciences Division, Livermore, California, USA.
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, Vancouver, British Columbia, Canada; Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
43
|
Ferrier MG, Radchenko V, Wilbur DS. Radiochemical aspects of alpha emitting radionuclides for medical application. RADIOCHIM ACTA 2019. [DOI: 10.1515/ract-2019-0005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Abstract
The use of α-emitting radionuclides in targeted alpha therapy (TAT) holds great potential for treatment of human diseases, such as cancer, due to the short pathlength and high potency of the α particle, which can localize damage to targeted cells while minimizing effects to healthy surrounding tissues. In this review several potential α-emitting radionuclides having emission properties applicable to TAT are discussed from a radiochemical point of view. Overviews of production, radiochemical separation and chelation aspects relative to developing TAT radiopharmaceuticals are provided for the α-emitting radionuclides (and their generator systems) 211At, 224Ra/212Pb/212Bi, 225Ac/213Bi, 227Th/223Ra, 230U/226Th, 149Tb and 255Fm.
Collapse
Affiliation(s)
- Maryline G. Ferrier
- Department of Radiation Oncology, Radiochemistry Division , University of Washington , Seattle, WA , USA
| | - Valery Radchenko
- Life Sciences Division, TRIUMF , Vancouver, BC , Canada
- Department of Chemistry , University of British Columbia , Vancouver, BC , Canada
| | - D. Scott Wilbur
- Department of Radiation Oncology, Radiochemistry Division , University of Washington , Seattle, WA , USA
| |
Collapse
|
44
|
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
|
45
|
Uccelli L, Martini P, Cittanti C, Carnevale A, Missiroli L, Giganti M, Bartolomei M, Boschi A. Therapeutic Radiometals: Worldwide Scientific Literature Trend Analysis (2008⁻2018). Molecules 2019; 24:molecules24030640. [PMID: 30759753 PMCID: PMC6385165 DOI: 10.3390/molecules24030640] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 11/21/2022] Open
Abstract
Academic journals have published a large number of papers in the therapeutic nuclear medicine (NM) research field in the last 10 years. Despite this, a literature analysis has never before been made to point out the research interest in therapeutic radionuclides (RNs). For this reason, the present study aims specifically to analyze the research output on therapeutic radiometals from 2008 to 2018, with intent to quantify and identify global trends in scientific literature and emphasize the interdisciplinary nature of this research field. The data search targeted conventional (131I, 90Y, 177Lu, 188Re, 186Re, 153Sm, 89Sr, 186Er) and emergent (67Cu, 47Sc, 223Ra, 166Ho, 161Tb, 149Tb, 212Pb/212Bi, 225Ac, 213Bi, 211At, 117mSn) RNs. Starting from this time frame, authors have analyzed and interpreted this scientific trend quantitatively first, and qualitatively after.
Collapse
Affiliation(s)
- Licia Uccelli
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Ludovico Ariosto, 35-44121 Ferrara, Italy.
- Nuclear Medicine Unit, University Hospital, Via Aldo Moro, 8-44124 Ferrara, Italy.
| | - Petra Martini
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Ludovico Ariosto, 35-44121 Ferrara, Italy.
- Legnaro National Laboratories, Italian National Institute for Nuclear Physics (LNL-INFN), Viale dell'Università, 2, 35020 Legnaro (PD), Italy.
| | - Corrado Cittanti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Ludovico Ariosto, 35-44121 Ferrara, Italy.
- Nuclear Medicine Unit, University Hospital, Via Aldo Moro, 8-44124 Ferrara, Italy.
| | - Aldo Carnevale
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Ludovico Ariosto, 35-44121 Ferrara, Italy.
- Radiology University Unit, University Hospital, Via Aldo Moro, 8-44124 Ferrara, Italy.
| | - Loretta Missiroli
- Bibliometric and Databases Unit, Research Office, University of Ferrara, Via Ludovico Ariosto, 35-44121 Ferrara, Italy.
| | - Melchiore Giganti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Ludovico Ariosto, 35-44121 Ferrara, Italy.
- Radiology University Unit, University Hospital, Via Aldo Moro, 8-44124 Ferrara, Italy.
| | - Mirco Bartolomei
- Nuclear Medicine Unit, University Hospital, Via Aldo Moro, 8-44124 Ferrara, Italy.
| | - Alessandra Boschi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Ludovico Ariosto, 35-44121 Ferrara, Italy.
| |
Collapse
|
46
|
Abstract
Radiometals possess an exceptional breadth of decay properties and have been applied to medicine with great success for several decades. The majority of current clinical use involves diagnostic procedures, which use either positron-emission tomography (PET) or single-photon imaging to detect anatomic abnormalities that are difficult to visualize using conventional imaging techniques (e.g., MRI and X-ray). The potential of therapeutic radiometals has more recently been realized and relies on ionizing radiation to induce irreversible DNA damage, resulting in cell death. In both cases, radiopharmaceutical development has been largely geared toward the field of oncology; thus, selective tumor targeting is often essential for efficacious drug use. To this end, the rational design of four-component radiopharmaceuticals has become popularized. This Review introduces fundamental concepts of drug design and applications, with particular emphasis on bifunctional chelators (BFCs), which ensure secure consolidation of the radiometal and targeting vector and are integral for optimal drug performance. Also presented are detailed accounts of production, chelation chemistry, and biological use of selected main group and rare earth radiometals.
Collapse
Affiliation(s)
- Thomas I Kostelnik
- Medicinal Inorganic Chemistry Group, Department of Chemistry , University of British Columbia , Vancouver , British Columbia V6T 1Z1 , Canada
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry , University of British Columbia , Vancouver , British Columbia V6T 1Z1 , Canada
| |
Collapse
|