1
|
Muralidhar A, Hernandez R, Morris ZS, Comas Rojas H, Bio Idrissou M, Weichert JP, McNeel DG. Myeloid-derived suppressor cells attenuate the antitumor efficacy of radiopharmaceutical therapy using 90Y-NM600 in combination with androgen deprivation therapy in murine prostate tumors. J Immunother Cancer 2024; 12:e008760. [PMID: 38663936 PMCID: PMC11043705 DOI: 10.1136/jitc-2023-008760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
RATIONALE Androgen deprivation therapy (ADT) is pivotal in treating recurrent prostate cancer and is often combined with external beam radiation therapy (EBRT) for localized disease. However, for metastatic castration-resistant prostate cancer, EBRT is typically only used in the palliative setting, because of the inability to radiate all sites of disease. Systemic radiation treatments that preferentially irradiate cancer cells, known as radiopharmaceutical therapy or targeted radionuclide therapy (TRT), have demonstrable benefits for treating metastatic prostate cancer. Here, we explored the use of a novel TRT, 90Y-NM600, specifically in combination with ADT, in murine prostate tumor models. METHODS 6-week-old male FVB mice were implanted subcutaneously with Myc-CaP tumor cells and given a single intravenous injection of 90Y-NM600, in combination with ADT (degarelix). The combination and sequence of administration were evaluated for effect on tumor growth and infiltrating immune populations were analyzed by flow cytometry. Sera were assessed to determine treatment effects on cytokine profiles. RESULTS ADT delivered prior to TRT (ADT→TRT) resulted in significantly greater antitumor response and overall survival than if delivered after TRT (TRT→ADT). Studies conducted in immunodeficient NRG mice failed to show a difference in treatment sequence, suggesting an immunological mechanism. Myeloid-derived suppressor cells (MDSCs) significantly accumulated in tumors following TRT→ADT treatment and retained immune suppressive function. However, CD4+ and CD8+ T cells with an activated and memory phenotype were more prevalent in the ADT→TRT group. Depletion of Gr1+MDSCs led to greater antitumor response following either treatment sequence. Chemotaxis assays suggested that tumor cells secreted chemokines that recruited MDSCs, notably CXCL1 and CXCL2. The use of a selective CXCR2 antagonist, reparixin, further improved antitumor responses and overall survival when used in tumor-bearing mice treated with TRT→ADT. CONCLUSION The combination of ADT and TRT improved antitumor responses in murine models of prostate cancer, however, this was dependent on the order of administration. This was found to be associated with one treatment sequence leading to an increase in infiltrating MDSCs. Combining treatment with a CXCR2 antagonist improved the antitumor effect of this combination, suggesting a possible approach for treating advanced human prostate cancer.
Collapse
Affiliation(s)
| | | | - Zachary S Morris
- Human Oncology, University of Wisconsin Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Hansel Comas Rojas
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Malick Bio Idrissou
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jamey P Weichert
- Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Douglas G McNeel
- Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
2
|
Becker KV, Aluicio-Sarduy E, Bradshaw T, Hurley SA, Olson AP, Barrett KE, Batterton J, Ellison PA, Barnhart TE, Pirasteh A, Engle JW. Cyclotron production of 43Sc and 44gSc from enriched 42CaO, 43CaO, and 44CaO targets. Front Chem 2023; 11:1167783. [PMID: 37179772 PMCID: PMC10169720 DOI: 10.3389/fchem.2023.1167783] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Introduction: 43Sc and 44gSc are both positron-emitting radioisotopes of scandium with suitable half-lives and favorable positron energies for clinical positron emission tomography (PET) imaging. Irradiation of isotopically enriched calcium targets has higher cross sections compared to titanium targets and higher radionuclidic purity and cross sections than natural calcium targets for reaction routes possible on small cyclotrons capable of accelerating protons and deuterons. Methods: In this work, we investigate the following production routes via proton and deuteron bombardment on CaCO3 and CaO target materials: 42Ca(d,n)43Sc, 43Ca(p,n)43Sc, 43Ca(d,n)44gSc, 44Ca(p,n)44gSc, and 44Ca(p,2n)43Sc. Radiochemical isolation of the produced radioscandium was performed with extraction chromatography using branched DGA resin and apparent molar activity was measured with the chelator DOTA. The imaging performance of 43Sc and 44gSc was compared with 18F, 68Ga, and 64Cu on two clinical PET/CT scanners. Discussion: The results of this work demonstrate that proton and deuteron bombardment of isotopically enriched CaO targets produce high yield and high radionuclidic purity 43Sc and 44gSc. Laboratory capabilities, circumstances, and budgets are likely to dictate which reaction route and radioisotope of scandium is chosen.
Collapse
Affiliation(s)
- Kaelyn V. Becker
- Department of Medical Physics, University of Wisconsin, Madison, WI, United States
| | | | - Tyler Bradshaw
- Department of Radiology, University of Wisconsin, Madison, WI, United States
| | - Samuel A. Hurley
- Department of Radiology, University of Wisconsin, Madison, WI, United States
| | - Aeli P. Olson
- Department of Medical Physics, University of Wisconsin, Madison, WI, United States
| | - Kendall E. Barrett
- Department of Medical Physics, University of Wisconsin, Madison, WI, United States
| | - Jeanine Batterton
- Department of Radiology, University of Wisconsin, Madison, WI, United States
| | - Paul A. Ellison
- Department of Medical Physics, University of Wisconsin, Madison, WI, United States
| | - Todd E. Barnhart
- Department of Medical Physics, University of Wisconsin, Madison, WI, United States
| | - Ali Pirasteh
- Department of Radiology, University of Wisconsin, Madison, WI, United States
| | - Jonathan W. Engle
- Department of Medical Physics, University of Wisconsin, Madison, WI, United States
- Department of Radiology, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
3
|
A High Separation Factor for 165Er from Ho for Targeted Radionuclide Therapy. Molecules 2021; 26:molecules26247513. [PMID: 34946596 PMCID: PMC8707915 DOI: 10.3390/molecules26247513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Radionuclides emitting Auger electrons (AEs) with low (0.02–50 keV) energy, short (0.0007–40 µm) range, and high (1–10 keV/µm) linear energy transfer may have an important role in the targeted radionuclide therapy of metastatic and disseminated disease. Erbium-165 is a pure AE-emitting radionuclide that is chemically matched to clinical therapeutic radionuclide 177Lu, making it a useful tool for fundamental studies on the biological effects of AEs. This work develops new biomedical cyclotron irradiation and radiochemical isolation methods to produce 165Er suitable for targeted radionuclide therapeutic studies and characterizes a new such agent targeting prostate-specific membrane antigen. Methods: Biomedical cyclotrons proton-irradiated spot-welded Ho(m) targets to produce 165Er, which was isolated via cation exchange chromatography (AG 50W-X8, 200–400 mesh, 20 mL) using alpha-hydroxyisobutyrate (70 mM, pH 4.7) followed by LN2 (20–50 µm, 1.3 mL) and bDGA (50–100 µm, 0.2 mL) extraction chromatography. The purified 165Er was radiolabeled with standard radiometal chelators and used to produce and characterize a new AE-emitting radiopharmaceutical, [165Er]PSMA-617. Results: Irradiation of 80–180 mg natHo targets with 40 µA of 11–12.5 MeV protons produced 165Er at 20–30 MBq·µA−1·h−1. The 4.9 ± 0.7 h radiochemical isolation yielded 165Er in 0.01 M HCl (400 µL) with decay-corrected (DC) yield of 64 ± 2% and a Ho/165Er separation factor of (2.8 ± 1.1) · 105. Radiolabeling experiments synthesized [165Er]PSMA-617 at DC molar activities of 37–130 GBq·µmol−1. Conclusions: A 2 h biomedical cyclotron irradiation and 5 h radiochemical separation produced GBq-scale 165Er suitable for producing radiopharmaceuticals at molar activities satisfactory for investigations of targeted radionuclide therapeutics. This will enable fundamental radiation biology experiments of pure AE-emitting therapeutic radiopharmaceuticals such as [165Er]PSMA-617, which will be used to understand the impact of AEs in PSMA-targeted radionuclide therapy of prostate cancer.
Collapse
|
4
|
Choiński J, Łyczko M. Prospects for the production of radioisotopes and radiobioconjugates for theranostics. BIO-ALGORITHMS AND MED-SYSTEMS 2021. [DOI: 10.1515/bams-2021-0136] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Abstract
The development of diagnostic methods in medicine as well as the progress in the synthesis of biologically active compounds allows the use of selected radioisotopes for the simultaneous diagnosis and treatment of diseases, especially cancerous ones, in patients. This approach is called theranostic. This review article includes chemical and physical characterization of chosen theranostic radioisotopes and their compounds that are or could be useful in nuclear medicine.
Collapse
Affiliation(s)
| | - Monika Łyczko
- Institute of Nuclear Chemistry and Technology , Warsaw , Poland
| |
Collapse
|
5
|
Magee K, Marsh IR, Turek MM, Grudzinski J, Aluicio-Sarduy E, Engle JW, Kurzman ID, Zuleger CL, Oseid EA, Jaskowiak C, Albertini MR, Esbona K, Bednarz B, Sondel PM, Weichert JP, Morris ZS, Hernandez R, Vail DM. Safety and feasibility of an in situ vaccination and immunomodulatory targeted radionuclide combination immuno-radiotherapy approach in a comparative (companion dog) setting. PLoS One 2021; 16:e0255798. [PMID: 34383787 PMCID: PMC8360580 DOI: 10.1371/journal.pone.0255798] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/23/2021] [Indexed: 12/18/2022] Open
Abstract
Rationale Murine syngeneic tumor models have revealed efficacious systemic antitumor responses following primary tumor in situ vaccination combined with targeted radionuclide therapy to secondary or metastatic tumors. Here we present studies on the safety and feasibility of this approach in a relevant translational companion dog model (n = 17 dogs) with advanced cancer. Methods The three component of the combination immuno-radiotherapy approach were employed either separately or in combination in companion dogs with advanced stage cancer. In situ vaccination was achieved through the administration of hypofractionated external beam radiotherapy and intratumoral hu14.18-IL2 fusion immunocytokine injections to the index tumor. In situ vaccination was subsequently combined with targeted radionuclide therapy using a theranostic pairing of IV 86Y-NM600 (for PET imaging and subject-specific dosimetry) and IV 90Y-NM600 (therapeutic radionuclide) prescribed to deliver an immunomodulatory 2 Gy dose to all metastatic sites in companion dogs with metastatic melanoma or osteosarcoma. In a subset of dogs, immunologic parameters preliminarily assessed. Results The components of the immuno-radiotherapy combination were well tolerated either alone or in combination, resulting in only transient low grade (1 or 2) adverse events with no dose-limiting events observed. In subject-specific dosimetry analyses, we observed 86Y-NM600 tumor:bone marrow absorbed-dose differential uptakes ≥2 in 4 of 5 dogs receiving the combination, which allowed subsequent safe delivery of at least 2 Gy 90Y-NM600 TRT to tumors. NanoString gene expression profiling and immunohistochemistry from pre- and post-treatment biopsy specimens provide evidence of tumor microenvironment immunomodulation by 90Y-NM600 TRT. Conclusions The combination of external beam radiotherapy, intratumoral immunocytokine, and targeted radionuclide immuno-radiotherapy known to have activity against syngeneic melanoma in murine models is feasible and well tolerated in companion dogs with advanced stage, spontaneously arising melanoma or osteosarcoma and has immunomodulatory potential. Further studies evaluating the dose-dependent immunomodulatory effects of this immuno-radiotherapy combination are currently ongoing.
Collapse
Affiliation(s)
- Kara Magee
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ian R. Marsh
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Michelle M. Turek
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Joseph Grudzinski
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Eduardo Aluicio-Sarduy
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jonathan W. Engle
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ilene D. Kurzman
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Cindy L. Zuleger
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Elizabeth A. Oseid
- Office of Environment, Health and Safety, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Christine Jaskowiak
- Department of Radiology, School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Mark R. Albertini
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- The Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
| | - Karla Esbona
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Bryan Bednarz
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Radiology, School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Paul M. Sondel
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Human Oncology, School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Department of Pediatrics, School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Jamey P. Weichert
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Radiology, School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Zachary S. Morris
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Reinier Hernandez
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Radiology, School of Medicine and Public Health, Madison, Wisconsin, United States of America
- * E-mail: (RH); . (DMV)
| | - David M. Vail
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail: (RH); . (DMV)
| |
Collapse
|
6
|
Clark PA, Sriramaneni RN, Bates AM, Jin WJ, Jagodinsky JC, Hernandez R, Le T, Jeffery JJ, Marsh IR, Grudzinski JJ, Aluicio-Sarduy E, Barnhart TE, Anderson BR, Chakravarty I, Arthur IS, Kim K, Engle JW, Bednarz BP, Weichert JP, Morris ZS. Low-Dose Radiation Potentiates the Propagation of Anti-Tumor Immunity against Melanoma Tumor in the Brain after In Situ Vaccination at a Tumor outside the Brain. Radiat Res 2021; 195:522-540. [PMID: 33826741 DOI: 10.1667/rade-20-00237.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/11/2021] [Indexed: 01/02/2023]
Abstract
Brain metastases develop in over 60% of advanced melanoma patients and negatively impact quality of life and prognosis. In a murine melanoma model, we previously showed that an in situ vaccination (ISV) regimen, combining radiation treatment and intratumoral (IT) injection of immunocytokine (IC: anti-GD2 antibody fused to IL2), along with the immune checkpoint inhibitor anti-CTLA-4, robustly eliminates peripheral flank tumors but only has modest effects on co-occurring intracranial tumors. In this study, we investigated the ability of low-dose radiation to the brain to potentiate anti-tumor immunity against a brain tumor when combined with ISV + anti-CTLA-4. B78 (GD2+, immunologically "cold") melanoma tumor cells were implanted into the flank and the right striatum of the brain in C57BL/6 mice. Flank tumors (50-150 mm3) were treated following a previously optimized ISV regimen [radiation (12 Gy × 1, treatment day 1), IT-IC (50 µg daily, treatment days 6-10), and anti-CTLA-4 (100 µg, treatment days 3, 6, 9)]. Mice that additionally received whole-brain radiation treatment (WBRT, 4 Gy × 1) on day 15 demonstrated significantly increased survival compared to animals that received ISV + anti-CTLA-4 alone, WBRT alone or no treatment (control) (P < 0.001, log-rank test). Timing of WBRT was critical, as WBRT administration on day 1 did not significantly enhance survival compared to ISV + anti-CTLA-4, suggesting that the effect of WBRT on survival might be mediated through immune modulation and not just direct tumor cell cytotoxicity. Modest increases in T cells (CD8+ and CD4+) and monocytes/macrophages (F4/80+) but no changes in FOXP3+ regulatory T cells (Tregs), were observed in brain melanoma tumors with addition of WBRT (on day 15) to ISV + anti-CTLA-4. Cytokine multiplex immunoassay revealed distinct changes in both intracranial melanoma and contralateral normal brain with addition of WBRT (day 15) to ISV + anti-CTLA-4, with notable significant changes in pro-inflammatory (e.g., IFNγ, TNFα and LIX/CXCL5) and suppressive (e.g., IL10, IL13) cytokines as well as chemokines (e.g., IP-10/CXCL10 and MIG/CXCL9). We tested the ability of the alkylphosphocholine analog, NM600, to deliver immunomodulatory radiation to melanoma brain tumors as a targeted radionuclide therapy (TRT). Yttrium-86 (86Y) chelated to NM600 was delivered intravenously by tail vein to mice harboring flank and brain melanoma tumors, and PET imaging demonstrated specific accumulation up to 72 h at each tumor site (∼12:1 brain tumor/brain and ∼8:1 flank tumor/muscle). When NM600 was chelated to therapeutic β-particle-emitting 90Y and administered on treatment day 13, T-cell infiltration and cytokine profiles were altered in melanoma brain tumor, like that observed for WBRT. Overall, our results demonstrate that addition of low-dose radiation, timed appropriately with ISV administration to tumors outside the brain, significantly increases survival in animals co-harboring melanoma brain tumors. This observation has potentially important translational implications as a treatment strategy for increasing the response of tumors in the brain to systemically administered immunotherapies.
Collapse
Affiliation(s)
- Paul A Clark
- Department of a Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Raghava N Sriramaneni
- Department of a Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Amber M Bates
- Department of a Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Won Jong Jin
- Department of a Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Justin C Jagodinsky
- Department of a Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Reinier Hernandez
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Trang Le
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Justin J Jeffery
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Ian R Marsh
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Joseph J Grudzinski
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Eduardo Aluicio-Sarduy
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Todd E Barnhart
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Bryce R Anderson
- Department of a Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Ishan Chakravarty
- Department of a Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Ian S Arthur
- Department of a Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jonathan W Engle
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.,Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Bryan P Bednarz
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jamey P Weichert
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Zachary S Morris
- Department of a Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
7
|
Patel RB, Hernandez R, Carlson P, Grudzinski J, Bates AM, Jagodinsky JC, Erbe A, Marsh IR, Arthur I, Aluicio-Sarduy E, Sriramaneni RN, Jin WJ, Massey C, Rakhmilevich AL, Vail D, Engle JW, Le T, Kim K, Bednarz B, Sondel PM, Weichert J, Morris ZS. Low-dose targeted radionuclide therapy renders immunologically cold tumors responsive to immune checkpoint blockade. Sci Transl Med 2021; 13:eabb3631. [PMID: 34261797 PMCID: PMC8449934 DOI: 10.1126/scitranslmed.abb3631] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 02/10/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022]
Abstract
Molecular and cellular effects of radiotherapy on tumor microenvironment (TME) can help prime and propagate antitumor immunity. We hypothesized that delivering radiation to all tumor sites could augment response to immunotherapies. We tested an approach to enhance response to immune checkpoint inhibitors (ICIs) by using targeted radionuclide therapy (TRT) to deliver radiation semiselectively to tumors. NM600, an alkylphosphocholine analog that preferentially accumulates in most tumor types, chelates a radioisotope and semiselectively delivers it to the TME for therapeutic or diagnostic applications. Using serial 86Y-NM600 positron emission tomography (PET) imaging, we estimated the dosimetry of 90Y-NM600 in immunologically cold syngeneic murine models that do not respond to ICIs alone. We observed strong therapeutic efficacy and reported optimal dose (2.5 to 5 gray) and sequence for 90Y-NM600 in combination with ICIs. After combined treatment, 45 to 66% of mice exhibited complete response and tumor-specific T cell memory, compared to 0% with 90Y-NM600 or ICI alone. This required expression of STING in tumor cells. Combined TRT and ICI activated production of proinflammatory cytokines in the TME, promoted tumor infiltration by and clonal expansion of CD8+ T cells, and reduced metastases. In mice bearing multiple tumors, combining TRT with moderate-dose (12 gray) external beam radiotherapy (EBRT) targeting a single tumor augmented response to ICIs compared to combination of ICIs with either TRT or EBRT alone. The safety of TRT was confirmed in a companion canine study. Low-dose TRT represents a translatable approach to promote response to ICIs for many tumor types, regardless of location.
Collapse
Affiliation(s)
- Ravi B Patel
- Department of Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA 15213, USA.
| | - Reinier Hernandez
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Peter Carlson
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Joseph Grudzinski
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Amber M Bates
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Justin C Jagodinsky
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Amy Erbe
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Ian R Marsh
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Ian Arthur
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | | | - Raghava N Sriramaneni
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Won Jong Jin
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Christopher Massey
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | | | - David Vail
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53792, USA
- Barbara A. Suran Comparative Oncology Institute, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Johnathan W Engle
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Trang Le
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Bryan Bednarz
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53792, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Jamey Weichert
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53792, USA.
| |
Collapse
|
8
|
Hu A, Aluicio-Sarduy E, Brown V, MacMillan SN, Becker KV, Barnhart TE, Radchenko V, Ramogida CF, Engle JW, Wilson JJ. Py-Macrodipa: A Janus Chelator Capable of Binding Medicinally Relevant Rare-Earth Radiometals of Disparate Sizes. J Am Chem Soc 2021; 143:10429-10440. [PMID: 34190542 DOI: 10.1021/jacs.1c05339] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Nuclear medicine leverages different types of radiometals for disease diagnosis and treatment, but these applications usually require them to be stably chelated. Given the often-disparate chemical properties of these radionuclides, it is challenging to find a single chelator that binds all of them effectively. Toward addressing this problem, we recently reported a macrocyclic chelator macrodipa with an unprecedented "dual-size-selectivity" pattern for lanthanide (Ln3+) ions, characterized by its high affinity for both the large and the small Ln3+ ( J. Am. Chem. Soc, 2020, 142, 13500). Here, we describe a second-generation "macrodipa-type" ligand, py-macrodipa. Its coordination chemistry with Ln3+ was thoroughly investigated experimentally and computationally. These studies reveal that the Ln3+-py-macrodipa complexes exhibit enhanced thermodynamic and kinetic stabilities compared to Ln3+-macrodipa, while retaining the unusual dual-size selectivity. Nuclear medicine applications of py-macrodipa for chelating radiometals with disparate chemical properties were assessed using the therapeutic 135La3+ and diagnostic 44Sc3+ radiometals representing the two size extremes within the rare-earth series. Radiolabeling and stability studies demonstrate that the rapidly formed complexes of these radionuclides with py-macrodipa are highly stable in human serum. Thus, in contrast to gold standard chelators like DOTA and macropa, py-macrodipa can be harnessed for the simultaneous, efficient binding of radiometals with disparate ionic radii like La3+ and Sc3+, signifying a substantial achievement in nuclear medicine. This concept could enable the facile incorporation of a breadth of medicinally relevant radiometals into chemically identical radiopharmaceutical agents. The fundamental coordination chemistry learned from py-macrodipa provides valuable insight for future chelator development.
Collapse
Affiliation(s)
- Aohan Hu
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Eduardo Aluicio-Sarduy
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Victoria Brown
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Samantha N MacMillan
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Kaelyn V Becker
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States.,Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Todd E Barnhart
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, Vancouver, British Columbia V6T 2A3, Canada.,Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Caterina F Ramogida
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada.,Life Sciences Division, TRIUMF, Vancouver, British Columbia V6T 2A3, Canada
| | - Jonathan W Engle
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States.,Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
9
|
Production of a broad palette of positron emitting radioisotopes using a low-energy cyclotron: Towards a new success story in cancer imaging? Appl Radiat Isot 2021; 176:109860. [PMID: 34284216 DOI: 10.1016/j.apradiso.2021.109860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 06/28/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022]
Abstract
Over the last several years, positron emission tomography (PET) has matured as an indispensable component of cancer diagnostics. Owing to the large variability observed among the cancer patients and the need to personalize individual patient's diagnosis and treatment, the need for new positron emitting radioisotopes has continued to grow. This mini review opens with a brief introduction to the criteria for radioisotope selection for PET imaging. Subsequently, positron emitting radioisotopes are categorized as: established, emerging and futuristic, based on the stages of their advancement. The production methodologies and the radiochemical separation procedures for obtaining the important radioisotopes in a form suitable for preparation of radiopharmaceuticals for PET imaging are briefly discussed.
Collapse
|
10
|
Rabiei M, Khorshidi A, Soltani-Nabipour J. Production of Yttrium-86 radioisotope using genetic algorithm and neural network. Biomed Signal Process Control 2021. [DOI: 10.1016/j.bspc.2021.102449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
11
|
Assessing the potential of online ICP–MS analysis to optimize Ca/matrix separation using DGA Resin for subsequent isotopic analysis. MONATSHEFTE FUR CHEMIE 2021. [DOI: 10.1007/s00706-021-02754-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
AbstractCa isotopes have gained increasing interest as a diagnostic tool for bone diseases due to the variations in abundances as a consequence of changes in bone-mineral balance. Optimized Ca/matrix separation prior to analysis is a prerequisite for reliable isotope ratio measurements in complex biological matrices such as blood, serum, or urine. The online analysis of analyte/matrix separation by ICP-MS enables direct assessment of elution profiles supporting the optimization process. The integration of transient signals and signal suppression challenge the quantification and interpretation of the elution profiles. Mn and Co remain unretained by the DGA Resin (TrisKem International) from nitric acid. Hence, in the present study, these elements were investigated for their application as standards to monitor signal suppression. Successful analyte quantification was accomplished using a dynamic correction strategy applying a linear gradient of a suppression factor based on Mn and Co intensities. An optimized Ca/matrix separation procedure using DGA Resin is proposed based on the results during online ICP-MS analysis.
Graphic abstract
Collapse
|
12
|
Abstract
Yttrium-86 is a non-standard positron emitter that can provide dosimetry information prior to therapy with yttrium-90 radiopharmaceuticals and be used to follow biochemical processes. In this chapter, we discuss the production, purification and applications of 86Y for PET imaging. More specifically, 86Y radiolabeling is highlighted and protocols to determine the radiochemical purity of 86Y-DOTA and 86Y-DTPA are presented.
Collapse
Affiliation(s)
- Mariane Le Fur
- The Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States.
| | - Peter Caravan
- The Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
13
|
Coenen HH, Ermert J. Expanding PET-applications in life sciences with positron-emitters beyond fluorine-18. Nucl Med Biol 2021; 92:241-269. [PMID: 32900582 DOI: 10.1016/j.nucmedbio.2020.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 07/09/2020] [Indexed: 12/20/2022]
Abstract
Positron-emission-tomography (PET) has become an indispensable diagnostic tool in modern nuclear medicine. Its outstanding molecular imaging features allow repetitive studies on one individual and with high sensitivity, though no interference. Rather few positron-emitters with near favourable physical properties, i.e. carbon-11 and fluorine-18, furnished most studies in the beginning, preferably if covalently bound as isotopic label of small molecules. With the advancement of PET-devices the scope of in vivo research in life sciences and especially that of medical applications expanded, and other than "standard" PET-nuclides received increasing significance, like the radiometals copper-64 and gallium-68. Especially during the last decades, positron-emitters of other chemical elements have gotten into the focus of interest, concomitant with the technical advancements in imaging and radionuclide production. With known nuclear imaging properties and main production methods of emerging positron-emitters their usefulness for medical application is promising and even proven for several ones already. Unfortunate decay properties could be corrected for, and β+-emitters, especially with a longer half-life, provided new possibilities for application where slower processes are of importance. Further on, (bio)chemical features of positron-emitters of other elements, among there many metals, not only expanded the field of classical clinical investigations, but also opened up new fields of application. Appropriately labelled peptides, proteins and nanoparticles lend itself as newer probes for PET-imaging, e.g. in theragnostic or PET/MR hybrid imaging. Furthermore, the potential of non-destructive in-vivo imaging with positron-emission-tomography directs the view on further areas of life sciences. Thus, exploiting the excellent methodology for basic research on molecular biochemical functions and processes is increasingly encouraged as well in areas outside of health, such as plant and environmental sciences.
Collapse
Affiliation(s)
- Heinz H Coenen
- Institut für Neurowissenschaften und Medizin, INM-5, Nuklearchemie, Forschungszentrum Jülich GmbH, D-52425 Jülich, Germany.
| | - Johannes Ermert
- Institut für Neurowissenschaften und Medizin, INM-5, Nuklearchemie, Forschungszentrum Jülich GmbH, D-52425 Jülich, Germany.
| |
Collapse
|
14
|
Uddin MS, Scholten B, Basunia MS, Sudár S, Spellerberg S, Voyles AS, Morrell JT, Zaneb H, Rios JA, Spahn I, Bernstein LA, Neumaier B, Qaim SM. Accurate determination of production data of the non-standard positron emitter 86Y via the 86Sr(p,n)-reaction. RADIOCHIM ACTA 2020. [DOI: 10.1515/ract-2020-0021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Abstract
In view of several significant discrepancies in the excitation function of the 86Sr(p,n)86g+xmY reaction which is the method of choice for the production of the non-standard positron emitter 86Y for theranostic application, we carried out a careful measurement of the cross sections of this reaction from its threshold up to 16.2 MeV at Forschungszentrum Jülich (FZJ) and from 14.3 to 24.5 MeV at LBNL. Thin samples of 96.4% enriched 86SrCO3 were prepared by sedimentation and, after irradiation with protons in a stacked-form, the induced radioactivity was measured by high-resolution γ-ray spectrometry. The projectile flux was determined by using the monitor reactions natCu(p,xn)62,63,65Zn and natTi(p,x)48V, and the calculated proton energy for each sample was verified by considering the ratios of two reaction products of different thresholds. The experimental cross section data obtained agreed well with the results of a nuclear model calculation based on the code TALYS. From the cross section data, the integral yield of 86Y was calculated. Over the optimum production energy range Ep = 14 → 7 MeV the yield of 86Y amounts to 291 MBq/μA for 1 h irradiation time. This value is appreciably lower than the previous literature values calculated from measured and evaluated excitation functions. It is, however, more compatible with the experimental yields of 86Y obtained in clinical scale production runs. The levels of the isotopic impurities 87mY, 87gY, and 88Y were also estimated and found to be <2% in sum.
Collapse
Affiliation(s)
- M. Shuza Uddin
- Institut für Neurowissenschaften und Medizin, INM-5: Nuklearchemie , Forschungszentrum Jülich , 52425 Jülich , Germany
- Nuclear Science Division , Lawrence Berkeley National Laboratory , Berkeley , CA , 94720 , USA
- Tandem Accelerator Facilities , INST , Atomic Energy Research Establishment , Savar , Dhaka , Bangladesh
| | - Bernhard Scholten
- Institut für Neurowissenschaften und Medizin, INM-5: Nuklearchemie , Forschungszentrum Jülich , 52425 Jülich , Germany
| | - M. Shamsuzzhoha Basunia
- Nuclear Science Division , Lawrence Berkeley National Laboratory , Berkeley , CA , 94720 , USA
| | - Sandor Sudár
- Institute of Experimental Physics , Debrecen University , Debrecen , 4001 , Hungary
| | - Stefan Spellerberg
- Institut für Neurowissenschaften und Medizin, INM-5: Nuklearchemie , Forschungszentrum Jülich , 52425 Jülich , Germany
| | - Andrew S. Voyles
- Nuclear Science Division , Lawrence Berkeley National Laboratory , Berkeley , CA , 94720 , USA
| | - Jonathan T. Morrell
- Nuclear Science Division , Lawrence Berkeley National Laboratory , Berkeley , CA , 94720 , USA
| | - Haleema Zaneb
- Nuclear Science Division , Lawrence Berkeley National Laboratory , Berkeley , CA , 94720 , USA
| | - Jesus A. Rios
- Nuclear Science Division , Lawrence Berkeley National Laboratory , Berkeley , CA , 94720 , USA
| | - Ingo Spahn
- Institut für Neurowissenschaften und Medizin, INM-5: Nuklearchemie , Forschungszentrum Jülich , 52425 Jülich , Germany
| | - Lee A. Bernstein
- Nuclear Science Division , Lawrence Berkeley National Laboratory , Berkeley , CA , 94720 , USA
| | - Bernd Neumaier
- Institut für Neurowissenschaften und Medizin, INM-5: Nuklearchemie , Forschungszentrum Jülich , 52425 Jülich , Germany
| | - Syed M. Qaim
- Institut für Neurowissenschaften und Medizin, INM-5: Nuklearchemie , Forschungszentrum Jülich , 52425 Jülich , Germany
| |
Collapse
|
15
|
Imaging using radiolabelled targeted proteins: radioimmunodetection and beyond. EJNMMI Radiopharm Chem 2020; 5:16. [PMID: 32577943 PMCID: PMC7311618 DOI: 10.1186/s41181-020-00094-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/14/2020] [Indexed: 12/18/2022] Open
Abstract
The use of radiolabelled antibodies was proposed in 1970s for staging of malignant tumours. Intensive research established chemistry for radiolabelling of proteins and understanding of factors determining biodistribution and targeting properties. The use of radioimmunodetection for staging of cancer was not established as common practice due to approval and widespread use of [18F]-FDG, which provided a more general diagnostic use than antibodies or their fragments. Expanded application of antibody-based therapeutics renewed the interest in radiolabelled antibodies. RadioimmunoPET emerged as a powerful tool for evaluation of pharmacokinetics of and target engagement by biotherapeutics. In addition to monoclonal antibodies, new radiolabelled engineered proteins have recently appeared, offering high-contrast imaging of expression of therapeutic molecular targets in tumours shortly after injection. This creates preconditions for noninvasive determination of a target expression level and stratification of patients for targeted therapies. Radiolabelled proteins hold great promise to play an important role in development and implementation of personalised targeted treatment of malignant tumours. This article provides an overview of biodistribution and tumour-seeking features of major classes of targeting proteins currently utilized for molecular imaging. Such information might be useful for researchers entering the field of the protein-based radionuclide molecular imaging.
Collapse
|
16
|
Li L, de Guadalupe Jaraquemada-Peláez M, Aluicio-Sarduy E, Wang X, Barnhart TE, Cai W, Radchenko V, Schaffer P, Engle JW, Orvig C. Coordination chemistry of [Y(pypa)] - and comparison immuno-PET imaging of [ 44Sc]Sc- and [ 86Y]Y-pypa-phenyl-TRC105. Dalton Trans 2020; 49:5547-5562. [PMID: 32270167 PMCID: PMC7222037 DOI: 10.1039/d0dt00437e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Both scandium-44 and yttrium-86 are popular PET isotopes with appropriate half-lives for immuno-positron emission tomography (immuno-PET) imaging. Herein, a new bifunctional H4pypa ligand, H4pypa-phenyl-NCS, is synthesized, conjugated to a monoclonal antibody, TRC105, and labeled with both radionuclides to investigate the long-term in vivo stability of each complex. While the 44Sc-labeled radiotracer exhibited promising pharmacokinetics and stability in 4T1-xenograft mice (n = 3) even upon prolonged interactions with blood serum proteins, the progressive bone uptake of the 86Y-counterpart indicated in vivo demetallation, obviating H4pypa as a suitable chelator for Y3+ ion in vivo. The solution chemistry of [natY(pypa)]- was studied in detail and the complex found to be thermodynamically stable in solution with a pM value 22.0, ≥3 units higher than those of the analogous DOTA- and CHX-A''-DTPA-complexes; the 86Y-result in vivo was therefore most unexpected. To explore further this in vivo lability, Density Functional Theory (DFT) calculation was performed to predict the geometry of [Y(pypa)]- and the results were compared with those for the analogous Sc- and Lu-complexes; all three adopted the same coordination geometry (i.e. distorted capped square antiprism), but the metal-ligand bonds were much longer in [Y(pypa)]- than in [Lu(pypa)]- and [Sc(pypa)]-, which could indicate that the size of the binding cavity is too small for the Y3+ ion, but suitable for both the Lu3+ and Sc3+ ions. Considered along with results from [86Y][Y(pypa-phenyl-TRC105)], it is noted that when matching chelators with radionuclides, chemical data such as the thermodynamic stability and in vitro inertness, albeit useful and necessary, do not always translate to in vivo inertness, especially with the prolonged blood circulation of the radiotracer bound to a monoclonal antibody. Although H4pypa is a nonadentate chelator, which theoretically matches the coordination number of the Y3+ ion, we show herein that its binding cavity, in fact, favors smaller metal ions such as Sc3+ and Lu3+ and further exploitation of the Sc-pypa combination is desired.
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.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
18
|
Hernandez R, Grudzinski JJ, Aluicio-Sarduy E, Massey CF, Pinchuk AN, Bitton AN, Patel R, Zhang R, Rao AV, Iyer G, Engle JW, Weichert JP. 177Lu-NM600 Targeted Radionuclide Therapy Extends Survival in Syngeneic Murine Models of Triple-Negative Breast Cancer. J Nucl Med 2019; 61:1187-1194. [PMID: 31862799 PMCID: PMC7413241 DOI: 10.2967/jnumed.119.236265] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/03/2019] [Indexed: 11/23/2022] Open
Abstract
There is a clinically unmet need for effective treatments for triple-negative breast cancer (TNBC), as it remains the most aggressive subtype of breast cancer. Herein, we demonstrate a promising strategy using a tumor-targeting alkylphosphocholine (NM600) for targeted radionuclide therapy of TNBC. Methods: NM600 was radiolabeled with 86Y for PET imaging and 177Lu for targeted radionuclide therapy. 86Y-NM600 PET imaging was performed on female BALB/C mice bearing syngeneic 4T07 (nonmetastatic) and 4T1 (metastatic) TNBC tumor grafts (n = 3–5). Quantitative data derived from a PET-image region-of-interest analysis, which was corroborated by ex vivo biodistribution, were used to estimate the dosimetry of 177Lu-NM600 treatments. Weight measurement, complete blood counts, and histopathology analysis were performed to determine 177Lu-NM600 toxicity in naïve BALB/C mice administered 9.25 or 18.5 MBq. Groups of mice bearing 4T07 or 4T1 grafts (n = 5–6) received excipient or 9.25 or 18.5 MBq of 177Lu-NM600 as a single or fractionated schedule, and tumor growth and overall survival were monitored. Results: Excellent tumor targeting and rapid normal-tissue clearance of 86Y-NM600 were noted in both 4T07 and 4T1 murine models. Ex vivo biodistribution corroborated the accuracy of the PET data and validated 86Y-NM600 as a surrogate for 177Lu-NM600. 177Lu-NM600 dosimetry showed absorbed doses of 2.04 ± 0.32 and 1.68 ± 0.06 Gy/MBq to 4T07 and 4T1 tumors, respectively, which were larger than those delivered to liver (1.28 ± 0.09 Gy/MBq) and to bone marrow (0.31 ± 0.05 Gy/MBq). The 177Lu-NM600 injected activities used for treatment were well tolerated and resulted in significant tumor growth inhibition and prolonged overall survival in both tested TNBC models. A complete response was attained in 60% of treated mice bearing 4T07 grafts. Conclusion: Overall, our results suggest that 177Lu-NM600 targeted radionuclide therapy has potential for TNBC and merits further exploration in a clinical setting.
Collapse
Affiliation(s)
- Reinier Hernandez
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Joseph J Grudzinski
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | | | | | - Anatoly N Pinchuk
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ariana N Bitton
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ravi Patel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Ray Zhang
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Aakarsha V Rao
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Gopal Iyer
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Jonathan W Engle
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jamey P Weichert
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin.,UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
19
|
Mikolajczak R, van der Meulen NP, Lapi SE. Radiometals for imaging and theranostics, current production, and future perspectives. J Labelled Comp Radiopharm 2019; 62:615-634. [PMID: 31137083 DOI: 10.1002/jlcr.3770] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/10/2019] [Accepted: 05/15/2019] [Indexed: 02/06/2023]
Abstract
The aim of this review is to make the reader familiar with currently available radiometals, their production modes, capacities, and quality concerns related to their medical use, as well as new emerging radiometals and irradiation technologies from the perspective of their diagnostic and theranostic applications. Production methods of 177 Lu serve as an example of various issues related to the production yield, specific activity, radionuclidic and chemical purity, and production economy. Other radiometals that are currently used or explored for potential medical applications, with particular focus on their theranostic value, are discussed. Using radiometals for diagnostic imaging and therapy is on the rise. The high demand for radiometals for medical use prompts investigations towards using alternative irradiation reactions, while using existing nuclear reactors and accelerator facilities. This review discusses these production capacities and what is necessary to cover the growing demand for theranostic nuclides.
Collapse
Affiliation(s)
- Renata Mikolajczak
- Radioisotope Centre POLATOM, National Centre for Nuclear Research, Otwock, Poland
| | | | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
20
|
Production and in vivo PET/CT imaging of the theranostic pair 132/135La. Sci Rep 2019; 9:10658. [PMID: 31337833 PMCID: PMC6650468 DOI: 10.1038/s41598-019-47137-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 07/11/2019] [Indexed: 11/23/2022] Open
Abstract
The present study describes a novel method for the low energy cyclotron production and radiochemical isolation of no-carrier-added 132/135La3+ from bulk natBa. This separation strategy combines precipitation and single-column extraction chromatography to afford an overall radiochemical yield (92 ± 2%) and apparent molar activity (22 ± 4 Mbq/nmol) suitable for the radiolabeling of DOTA-conjugated vectors. The produced 132/135La3+ has a radiochemical and radionuclidic purity amenable for 132La/135La-based cancer theranostic applications. Longitudinal PET/CT images acquired using the positron-emitting 132La and ex vivo biodistribution data separately corroborated the accumulation of unchelated 132/135La3+ ions in bone and the liver.
Collapse
|
21
|
Grudzinski JJ, Hernandez R, Marsh I, Patel RB, Aluicio-Sarduy E, Engle J, Morris Z, Bednarz B, Weichert J. Preclinical Characterization of 86/90Y-NM600 in a Variety of Murine and Human Cancer Tumor Models. J Nucl Med 2019; 60:1622-1628. [PMID: 30954941 DOI: 10.2967/jnumed.118.224808] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/26/2019] [Indexed: 11/16/2022] Open
Abstract
We characterize the in vivo biodistribution and tumor selectivity of 86Y-NM600, a theranostic alkylphosphocholine radiometal chelate with broad tumor selectivity, in a variety of preclinical cancer models. Methods: Mice bearing flank tumors (representative of lung, pancreatic, prostate, liver, skin, and lymphoid cancers) were injected intravenously with 9.25 MBq of 86Y-NM600 and imaged longitudinally over 4-5 d using small-animal PET/CT. Percentage injected activity per gram (%IA/g) for each volume of interest was measured at each time point for the organs of interest. Mice were euthanized after the final time point, and the tumor and organs of interest were counted with an automatic γ-counter. Absorbed doses delivered by 90Y-NM600 per injected activity (Gy/MBq) were estimated. Mice bearing B78 flank tumors were injected with a prescription of 90Y-NM600 that delivered 2.5 Gy of absorbed tumor dose and was compared with an equivalent absorbed dose delivered via external-beam radiotherapy using tumor volume as a measure of response. Histology and complete blood counts were analyzed in naïve C57BL/6 mice that were injected with 9.25 MBq of 90Y-NM600 at 5, 10, and 28 d after injection. Results: PET imaging showed consistent tumor accumulation and retention across all tumor models investigated, with little off-target retention of NM600 except in the liver, as is characteristic of hepatobiliary metabolism. The tumor uptake was highest in the pancreatic and lymphoid cancer models, reaching peak concentrations of 9.34 ± 2.66 %IA/g (n = 3) and 9.10 ± 0.13 %IA/g (n = 3), respectively, at approximately 40-48 h after injection. These corresponded to tumor dose estimates of 2.72 ± 0.33 Gy/MBq and 2.67 ± 0.32 Gy/MBq, respectively. In the toxicity study, there were no visible signs of acute toxicity by histology, and perturbation of hematologic parameters was transient when observed, returning to pretherapy levels after 28 d. Conclusion: NM600 is a theranostic agent with a unique ability to selectively target a variety of cancer types, presenting a unique opportunity for PET image-guided targeted radionuclide therapy and combination with immunotherapies.
Collapse
Affiliation(s)
- Joseph J Grudzinski
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Reinier Hernandez
- Department of Radiology, University of Wisconsin, Madison, Wisconsin
| | - Ian Marsh
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ravi B Patel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin; and
| | | | - Jon Engle
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Radiology, University of Wisconsin, Madison, Wisconsin
| | - Zachary Morris
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Bryan Bednarz
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Radiology, University of Wisconsin, Madison, Wisconsin.,Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Jamey Weichert
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Radiology, University of Wisconsin, Madison, Wisconsin.,University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| |
Collapse
|
22
|
90Y-NM600 targeted radionuclide therapy induces immunologic memory in syngeneic models of T-cell Non-Hodgkin's Lymphoma. Commun Biol 2019; 2:79. [PMID: 30820474 PMCID: PMC6391402 DOI: 10.1038/s42003-019-0327-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 01/25/2019] [Indexed: 12/29/2022] Open
Abstract
Finding improved therapeutic strategies against T-cell Non-Hodgkin’s Lymphoma (NHL) remains an unmet clinical need. We implemented a theranostic approach employing a tumor-targeting alkylphosphocholine (NM600) radiolabeled with 86Y for positron emission tomography (PET) imaging and 90Y for targeted radionuclide therapy (TRT) of T-cell NHL. PET imaging and biodistribution performed in mouse models of T-cell NHL showed in vivo selective tumor uptake and retention of 86Y-NM600. An initial toxicity assessment examining complete blood counts, blood chemistry, and histopathology of major organs established 90Y-NM600 safety. Mice bearing T-cell NHL tumors treated with 90Y-NM600 experienced tumor growth inhibition, extended survival, and a high degree of cure with immune memory toward tumor reestablishment. 90Y-NM600 treatment was also effective against disseminated tumors, improving survival and cure rates. Finally, we observed a key role for the adaptive immune system in potentiating a durable anti-tumor response to TRT, especially in the presence of microscopic disease. Hernandez et al. show the effectiveness of 90Y for targeted radionucleotide therapy of T-cell Non-Hodgkin’s Lymphoma (NHL). This study suggests that delivering radiation to all NHL disease sites elicits minimal toxicity and induces a memory T-cell response, inviting combination therapies with immune activating agents.
Collapse
|