1
|
Marsh IR, Li C, Grudzinski J, Jeffery J, Longhurst C, Adam DP, Hernandez R, Weichert JP, Harari PM, Bednarz BP. Targeting of Head and Neck Cancer by Radioiodinated CLR1404 in Murine Xenograft Tumor Models with Partial Volume Corrected Theranostic Dosimetry. Cancer Biother Radiopharm 2023; 38:458-467. [PMID: 37022739 PMCID: PMC10516227 DOI: 10.1089/cbr.2022.0084] [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] [Indexed: 04/07/2023] Open
Abstract
Background: Delivery of radiotherapeutic dose to recurrent head and neck cancer (HNC) is primarily limited by locoregional toxicity in conventional radiotherapy. As such, HNC patients stand to benefit from the conformal targeting of primary and remnant disease achievable with radiopharmaceutical therapies. In this study, the authors investigated the tumor targeting capacity of 131I-CLR1404 (iopofosine I-131) in various HNC xenograft mouse models and the impact of partial volume correction (PVC) on theranostic dosimetry based on 124I-CLR1404 (CLR 124) positron emission tomography (PET)/computed tomography (CT) imaging. Methods: Mice bearing flank tumor xenograft models of HNC (six murine cell line and six human patient derived) were intravenously administered 6.5-9.1 MBq of CLR 124 and imaged five times over the course of 6 d using microPET/CT. In vivo tumor uptake of CLR 124 was assessed and PVC for 124I was applied using a novel preclinical phantom. Using subject-specific theranostic dosimetry estimations for iopofosine I-131 based on CLR 124 imaging, a discrete radiation dose escalation study (2, 4, 6, and 8 Gy) was performed to evaluate tumor growth response to iopofosine I-131 relative to a single fraction of external beam radiation therapy (6 Gy). Results: PET imaging demonstrated consistent tumor selective uptake and retention of CLR 124 across all HNC xenograft models. Peak uptake of 4.4% ± 0.8% and 4.2% ± 0.4% was observed in squamous cell carcinoma-22B and UW-13, respectively. PVC application increased uptake measures by 47%-188% and reduced absolute differences between in vivo and ex vivo uptake measurements from 3.3% to 1.0 percent injected activity per gram. Tumor dosimetry averaged over all HNC models was 0.85 ± 0.27 Gy/MBq (1.58 ± 0.46 Gy/MBq with PVC). Therapeutic iopofosine I-131 studies demonstrated a variable, but linear relationship between iopofosine I-131 radiation dose and tumor growth delay (p < 0.05). Conclusions: Iopofosine I-131 demonstrated tumoricidal capacity in preclinical HNC tumor models and the theranostic pairing with CLR 124 presents a promising new treatment approach for personalizing administration of iopofosine I-131.
Collapse
Affiliation(s)
- Ian R. Marsh
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Chunrong Li
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joseph Grudzinski
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Justin Jeffery
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Colin Longhurst
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David P. Adam
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Reinier Hernandez
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jamey P. Weichert
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Paul M. Harari
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Bryan P. Bednarz
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
2
|
Allen KJH, Kwon O, Hutcheson MR, Grudzinski JJ, Cain SM, Cruz FA, Vinayakamoorthy RM, Sun YS, Fairley L, Prabaharan CB, Dickinson R, MacDonald-Dickinson V, Uppalapati M, Bednarz BP, Dadachova E. Image-Based Dosimetry in Dogs and Cross-Reactivity with Human Tissues of IGF2R-Targeting Human Antibody. Pharmaceuticals (Basel) 2023; 16:979. [PMID: 37513891 PMCID: PMC10384855 DOI: 10.3390/ph16070979] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) represents the most common primary bone tumor in humans and in companion dogs, being practically phenotypically identical. There is a need for effective treatments to extend the survival of patients with OS. Here, we examine the dosimetry in beagle dogs and cross-reactivity with human tissues of a novel human antibody, IF3, that targets the insulin growth factor receptor type 2 (IGF2R), which is overexpressed on OS cells, making it a candidate for radioimmunotherapy of OS. METHODS [89Zr]Zr-DFO-IF3 was injected into three healthy beagle dogs. PET/CT was conducted at 4, 24, 48, and 72 h. RAPID analysis was used to determine the dosimetry of [177Lu]Lu-CHXA"-IF3 for a clinical trial in companion dogs with OS. IF3 antibody was biotinylated, and a multitude of human tissues were assessed with immunohistochemistry. RESULTS PET/CT revealed that only the liver, bone marrow, and adrenal glands had high uptake. Clearance was initially through renal and hepatobiliary excretion in the first 72 h followed by primarily physical decay. RAPID analysis showed bone marrow to be the dose-limiting organ with a therapeutic range for 177Lu calculated to be 0.487-0.583 GBq. Immunohistochemistry demonstrated the absence of IGF2R expression on the surface of healthy human cells, thus suggesting that radioimmunotherapy with [177Lu]Lu-CHXA"-IF3 will be well tolerated. CONCLUSIONS Image-based dosimetry has defined a safe therapeutic range for canine clinical trials, while immunohistochemistry has suggested that the antibody will not cross-react with healthy human tissues.
Collapse
Affiliation(s)
- Kevin J H Allen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Ohyun Kwon
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Joseph J Grudzinski
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Stuart M Cain
- adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada
| | | | | | - Ying S Sun
- adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada
| | | | - Chandra B Prabaharan
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Ryan Dickinson
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Valerie MacDonald-Dickinson
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Maruti Uppalapati
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Bryan P Bednarz
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
3
|
Hoffman SLV, Mixdorf JC, Kwon O, Johnson TR, Makvandi M, Lee H, Aluicio-Sarduy E, Barnhart TE, Jeffery JJ, Patankar MS, Engle JW, Bednarz BP, Ellison PA. Preclinical studies of a PARP targeted, Meitner-Auger emitting, theranostic radiopharmaceutical for metastatic ovarian cancer. Nucl Med Biol 2023; 122-123:108368. [PMID: 37490805 PMCID: PMC10529069 DOI: 10.1016/j.nucmedbio.2023.108368] [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: 05/09/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 07/27/2023]
Abstract
Advanced ovarian cancer currently has few therapeutic options. Poly(ADP-ribose) polymerase (PARP) inhibitors bind to nuclear PARP and trap the protein-inhibitor complex to DNA. This work investigates a theranostic PARP inhibitor for targeted radiopharmaceutical therapy of ovarian cancer in vitro and PET imaging of healthy mice in vivo. METHODS [77Br]RD1 was synthesized and assessed for pharmacokinetics and cytotoxicity in human and murine ovarian cancer cell lines. [76Br]RD1 biodistribution and organ uptake in healthy mice were quantified through longitudinal PET/CT imaging and ex vivo radioactivity measurements. Organ-level dosimetry following [76/77Br]RD1 administration was calculated using RAPID, an in-house platform for absorbed dose in mice, and OLINDA for equivalent and effective dose in human. RESULTS The maximum specific binding (Bmax), equilibrium dissociation constant (Kd), and nonspecific binding slope (NS) were calculated for each cell line. These values were used to calculate the cell specific activity uptake for cell viability studies. The half maximal effective concentration (EC50) was measured as 0.17 (95 % CI: 0.13-0.24) nM and 0.46 (0.13-0.24) nM for PARP(+) and PARP(-) expressing cell lines, respectively. The EC50 was 0.27 (0.21-0.36) nM and 0.30 (0.22-0.41) nM for BRCA1(-) and BRCA1(+) expressing cell lines, respectively. When measuring the EC50 as a function of cellular activity uptake and nuclear dose, the EC50 ranges from 0.020 to 0.039 Bq/cell and 3.3-9.2 Gy, respectively. Excretion through the hepatobiliary and renal pathways were observed in mice, with liver uptake of 2.3 ± 0.4 %ID/g after 48 h, contributing to estimated absorbed dose values in mice of 19.3 ± 0.3 mGy/MBq and 290 ± 10 mGy/MBq for [77Br]RD1 and [76Br]RD1, respectively. CONCLUSION [77Br]RD1 cytotoxicity was dependent on PARP expression and independent of BRCA1 status. The in vitro results suggest that [77Br]RD1 cytotoxicity is driven by the targeted Meitner-Auger electron (MAe) radiotherapeutic effect of the agent. Further studies investigating the theranostic potential, organ dose, and tumor uptake of [76/77Br]RD1 are warranted.
Collapse
Affiliation(s)
- S L V Hoffman
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J C Mixdorf
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - O Kwon
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - T R Johnson
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - M Makvandi
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - H Lee
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - E Aluicio-Sarduy
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - T E Barnhart
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J J Jeffery
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - M S Patankar
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J W Engle
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - B P Bednarz
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - P A Ellison
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
4
|
Omidian H, Mfoafo K. Exploring the Potential of Nanotechnology in Pediatric Healthcare: Advances, Challenges, and Future Directions. Pharmaceutics 2023; 15:1583. [PMID: 37376032 DOI: 10.3390/pharmaceutics15061583] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
The utilization of nanotechnology has brought about notable advancements in the field of pediatric medicine, providing novel approaches for drug delivery, disease diagnosis, and tissue engineering. Nanotechnology involves the manipulation of materials at the nanoscale, resulting in improved drug effectiveness and decreased toxicity. Numerous nanosystems, including nanoparticles, nanocapsules, and nanotubes, have been explored for their therapeutic potential in addressing pediatric diseases such as HIV, leukemia, and neuroblastoma. Nanotechnology has also shown promise in enhancing disease diagnosis accuracy, drug availability, and overcoming the blood-brain barrier obstacle in treating medulloblastoma. It is important to acknowledge that while nanotechnology offers significant opportunities, there are inherent risks and limitations associated with the use of nanoparticles. This review provides a comprehensive summary of the existing literature on nanotechnology in pediatric medicine, highlighting its potential to revolutionize pediatric healthcare while also recognizing the challenges and limitations that need to be addressed.
Collapse
Affiliation(s)
- Hossein Omidian
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Kwadwo Mfoafo
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
5
|
Regenold M, Kaneko K, Wang X, Peng HB, Evans JC, Bannigan P, Allen C. Triggered release from thermosensitive liposomes improves tumor targeting of vinorelbine. J Control Release 2023; 354:19-33. [PMID: 36503069 DOI: 10.1016/j.jconrel.2022.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/28/2022]
Abstract
Triggered drug delivery strategies have been shown to enhance drug accumulation at target diseased sites in comparison to administration of free drug. In particular, many studies have demonstrated improved targetability of chemotherapeutics when delivered via thermosensitive liposomes. However, most studies continue to focus on encapsulating doxorubicin while many other drugs would benefit from this targeted and localized delivery approach. The proposed study explores the therapeutic potential of a thermosensitive liposome formulation of the commonly used chemotherapy drug vinorelbine in combination with mild hyperthermia (39-43 °C) in a murine model of rhabdomyosarcoma. Rhabdomyosarcoma, the most common soft tissue sarcoma in children, is largely treated using conventional chemotherapy which is associated with significant adverse long-term sequelae. In this study, mild hyperthermia was pursued as a non-invasive, non-toxic means to improve the efficacy and safety profiles of vinorelbine. Thorough assessment of the pharmacokinetics, biodistribution, efficacy and toxicity of vinorelbine administered in the thermosensitive liposome formulation was compared to administration in a traditional, non-thermosensitive liposome formulation. This study shows the potential of an advanced formulation technology in combination with mild hyperthermia as a means to target an untargeted therapeutic agent and result in a significant improvement in its therapeutic index.
Collapse
Affiliation(s)
- Maximilian Regenold
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Kan Kaneko
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Xuehan Wang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - H Benson Peng
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - James C Evans
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Pauric Bannigan
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
6
|
Potluri HK, Ferreira CA, Grudzinski J, Massey C, Aluicio-Sarduy E, Engle JW, Kwon O, Marsh IR, Bednarz BP, Hernandez R, Weichert JP, McNeel DG. Antitumor efficacy of 90Y-NM600 targeted radionuclide therapy and PD-1 blockade is limited by regulatory T cells in murine prostate tumors. J Immunother Cancer 2022; 10:jitc-2022-005060. [PMID: 36002185 PMCID: PMC9413196 DOI: 10.1136/jitc-2022-005060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2022] [Indexed: 12/14/2022] Open
Abstract
Background Systemic radiation treatments that preferentially irradiate cancer cells over normal tissue, known as targeted radionuclide therapy (TRT), have shown significant potential for treating metastatic prostate cancer. Preclinical studies have demonstrated the ability of external beam radiation therapy (EBRT) to sensitize tumors to T cell checkpoint blockade. Combining TRT approaches with immunotherapy may be more feasible than combining with EBRT to treat widely metastatic disease, however the effects of TRT on the prostate tumor microenvironment alone and in combinfation with checkpoint blockade have not yet been studied. Methods C57BL/6 mice-bearing TRAMP-C1 tumors and FVB/NJ mice-bearing Myc-CaP tumors were treated with a single intravenous administration of either low-dose or high-dose 90Y-NM600 TRT, and with or without anti-PD-1 therapy. Groups of mice were followed for tumor growth while others were used for tissue collection and immunophenotyping of the tumors via flow cytometry. Results 90Y-NM600 TRT was safe at doses that elicited a moderate antitumor response. TRT had multiple effects on the tumor microenvironment including increasing CD8 +T cell infiltration, increasing checkpoint molecule expression on CD8 +T cells, and increasing PD-L1 expression on myeloid cells. However, PD-1 blockade with TRT treatment did not improve antitumor efficacy. Tregs remained functional up to 1 week following TRT, but CD8 +T cells were not, and the suppressive function of Tregs increased when anti-PD-1 was present in in vitro studies. The combination of anti-PD-1 and TRT was only effective in vivo when Tregs were depleted. Conclusions Our data suggest that the combination of 90Y-NM600 TRT and PD-1 blockade therapy is ineffective in these prostate cancer models due to the activating effect of anti-PD-1 on Tregs. This finding underscores the importance of thorough understanding of the effects of TRT and immunotherapy combinations on the tumor immune microenvironment prior to clinical investigation.
Collapse
Affiliation(s)
- Hemanth K Potluri
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Carolina A Ferreira
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joseph Grudzinski
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Christopher Massey
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Jonathan W Engle
- Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ohyun Kwon
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ian R Marsh
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bryan P Bednarz
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Reinier Hernandez
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jamey P Weichert
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
7
|
Nanotheranostics for Image-Guided Cancer Treatment. Pharmaceutics 2022; 14:pharmaceutics14050917. [PMID: 35631503 PMCID: PMC9144228 DOI: 10.3390/pharmaceutics14050917] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 12/13/2022] Open
Abstract
Image-guided nanotheranostics have the potential to represent a new paradigm in the treatment of cancer. Recent developments in modern imaging and nanoparticle design offer an answer to many of the issues associated with conventional chemotherapy, including their indiscriminate side effects and susceptibility to drug resistance. Imaging is one of the tools best poised to enable tailoring of cancer therapies. The field of image-guided nanotheranostics has the potential to harness the precision of modern imaging techniques and use this to direct, dictate, and follow site-specific drug delivery, all of which can be used to further tailor cancer therapies on both the individual and population level. The use of image-guided drug delivery has exploded in preclinical and clinical trials although the clinical translation is incipient. This review will focus on traditional mechanisms of targeted drug delivery in cancer, including the use of molecular targeting, as well as the foundations of designing nanotheranostics, with a focus on current clinical applications of nanotheranostics in cancer. A variety of specially engineered and targeted drug carriers, along with strategies of labeling nanoparticles to endow detectability in different imaging modalities will be reviewed. It will also introduce newer concepts of image-guided drug delivery, which may circumvent many of the issues seen with other techniques. Finally, we will review the current barriers to clinical translation of image-guided nanotheranostics and how these may be overcome.
Collapse
|
8
|
Nemes K, Johann PD, Tüchert S, Melchior P, Vokuhl C, Siebert R, Furtwängler R, Frühwald MC. Current and Emerging Therapeutic Approaches for Extracranial Malignant Rhabdoid Tumors. Cancer Manag Res 2022; 14:479-498. [PMID: 35173482 PMCID: PMC8841298 DOI: 10.2147/cmar.s289544] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Extracranial malignant rhabdoid tumors (extracranial MRT) are rare, highly aggressive malignancies affecting mainly infants and children younger than 3 years. Common anatomic sites comprise the kidneys (RTK – rhabdoid tumor of kidney) and other soft tissues (eMRT – extracranial, extrarenal malignant rhabdoid tumor). The genetic origin of these diseases is linked to biallelic pathogenic variants in the genes SMARCB1, or rarely SMARCA4, encoding subunits of the SWI/SNF chromatin-remodeling complex. Even if extracranial MRT seem to be quite homogeneous, recent epigenome analyses reveal a certain degree of epigenetic heterogeneity. Use of intensified therapies has modestly improved survival for extracranial MRT. Patients at standard risk profit from conventional therapies; most high-risk patients still experience a dismal course and often therapy resistance. Discoveries of clinical and molecular hallmarks and the exploration of experimental therapeutic approaches open exciting perspectives for clinical and molecularly stratified experimental treatment approaches. To ultimately improve the outcome of patients with extracranial MRTs, they need to be characterized and stratified clinically and molecularly. High-risk patients need novel therapeutic approaches including selective experimental agents in phase I/II clinical trials.
Collapse
Affiliation(s)
- Karolina Nemes
- Paediatrics and Adolescent Medicine, Swabian Children's Cancer Center, University Medical Center Augsburg, Augsburg, Germany
| | - Pascal D Johann
- Paediatrics and Adolescent Medicine, Swabian Children's Cancer Center, University Medical Center Augsburg, Augsburg, Germany.,Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefanie Tüchert
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Augsburg, Germany
| | - Patrick Melchior
- Department of Radiation Oncology, University of Saarland, Homburg, Germany
| | - Christian Vokuhl
- Section of Pediatric Pathology, Department of Pathology, University Hospital Bonn, Bonn, Germany
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University & Ulm University Medical Center, Ulm, Germany
| | - Rhoikos Furtwängler
- Department of Pediatric Hematology and Oncology, University of Saarland, Homburg, Germany
| | - Michael C Frühwald
- Paediatrics and Adolescent Medicine, Swabian Children's Cancer Center, University Medical Center Augsburg, Augsburg, Germany
| |
Collapse
|
9
|
Development of a Compartmental Pharmacokinetic Model for Molecular Radiotherapy with 131I-CLR1404. Pharmaceutics 2021; 13:pharmaceutics13091497. [PMID: 34575575 PMCID: PMC8465706 DOI: 10.3390/pharmaceutics13091497] [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: 07/20/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 11/25/2022] Open
Abstract
Pharmacokinetic modeling of the radiopharmaceuticals used in molecular radiotherapy is an important step towards accurate radiation dosimetry of such therapies. In this paper, we present a pharmacokinetic model for CLR1404, a phospholipid ether analog that, labeled with 124I/131I, has emerged as a promising theranostic agent. We follow a systematic approach for the model construction based on a decoupling process applied to previously published experimental data, and using the goodness-of-fit, Sobol’s sensitivity analysis, and the Akaike Information Criterion to construct the optimal form of the model, investigate potential simplifications, and study factor prioritization. This methodology was applied to previously published experimental human time-activity curves for 9 organs. The resulting model consists of 17 compartments involved in the CLR1404 metabolism. Activity dynamics in most tissues are well described by a blood contribution plus a two-compartment system, describing fast and slow uptakes. The model can fit both clinical and pre-clinical kinetic data of 124I/131I. In addition, we have investigated how simple fits (exponential and biexponential) differ from the complete model. Such fits, despite providing a less accurate description of time-activity curves, may be a viable alternative when limited data is available in a practical case.
Collapse
|
10
|
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: 16] [Impact Index Per Article: 5.3] [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
|
11
|
Mittra E, Bodei L. Landscape Analysis of Phase 2 and 3 Clinical Trials for Targeted Radionuclide Therapy. J Nucl Med 2021; 62:1031-1032. [PMID: 33579808 PMCID: PMC9364763 DOI: 10.2967/jnumed.120.258103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/19/2021] [Indexed: 11/16/2022] Open
Affiliation(s)
- Erik Mittra
- Division of Nuclear Medicine and Molecular Imaging, Oregon Health & Science University, Portland, Oregon; and
| | - Lisa Bodei
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
12
|
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
|
13
|
Rafael MS, Cohen-Gogo S, Irwin MS, Vali R, Shammas A, Morgenstern DA. Theranostics in Neuroblastoma. PET Clin 2021; 16:419-427. [PMID: 34053585 DOI: 10.1016/j.cpet.2021.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Theranostics combines diagnosis and targeted therapy, achieved by the use of the same or similar molecules labeled with different radiopharmaceuticals or identical with different dosages. One of the best examples is the use of metaiodobenzylguanidine (MIBG). In the management of neuroblastoma-the most common extracranial solid tumor in children. MIBG has utility not only for diagnosis, risk-stratification, and response monitoring but also for cancer therapy, particularly in the setting of relapsed/refractory disease. Improved techniques and new emerging radiopharmaceuticals likely will strengthen the role of nuclear medicine in the management of neuroblastoma.
Collapse
Affiliation(s)
- Margarida Simao Rafael
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, 555 University Ave, Toronto, ON M5G 1X8, Canada
| | - Sarah Cohen-Gogo
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, 555 University Ave, Toronto, ON M5G 1X8, Canada
| | - Meredith S Irwin
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, 555 University Ave, Toronto, ON M5G 1X8, Canada
| | - Reza Vali
- Division of Nuclear Medicine, Department of Diagnostic Imaging, The Hospital for Sick Children, University of Toronto, 555 University Ave, Toronto, ON M5G 1X8, Canada.
| | - Amer Shammas
- Division of Nuclear Medicine, Department of Diagnostic Imaging, The Hospital for Sick Children, University of Toronto, 555 University Ave, Toronto, ON M5G 1X8, Canada
| | - Daniel A Morgenstern
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, 555 University Ave, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
14
|
Katal S, Maldonado A, Carrascoso J, Assadi M, Gholamrezanezhad A. Theranostic Agents in Musculoskeletal Disorders. PET Clin 2021; 16:441-448. [PMID: 34053587 DOI: 10.1016/j.cpet.2021.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Theranostic-based strategies, combining therapeutic and diagnostic properties of a single agent, have gained enormous attention in the past few years. Today, various multifunctional theranostic modalities have been examined, using different bioactive targeting, for the detection, quantifying, and monitoring of therapy response in different pathologies. Herein we review the newly emerging approaches in theranostic nanomedicine for the detection and therapy for musculoskeletal disorders to provide valuable insights for developing more efficient agents for clinical use. Some potential preclinical applications of radionuclide nanotheranostic agents are described in rheumatoid arthritis, osteoarthrosis, multiple myeloma, and neoplastic diseases.
Collapse
Affiliation(s)
- Sanaz Katal
- Department of Nuclear Medicine, Kowsar Hospital, Shiraz, Iran
| | - Antonio Maldonado
- Department of Nuclear Medicine, Quironsalud Madrid University Hospital, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Javier Carrascoso
- Department of Radiology, Quironsalud Madrid University Hospital, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Majid Assadi
- Department of Molecular Imaging and Radionuclide Therapy (MIRT), The Persian Gulf Nuclear Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ali Gholamrezanezhad
- Department of Diagnostic Radiology, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA.
| |
Collapse
|
15
|
Jagodinsky JC, Jin WJ, Bates AM, Hernandez R, Grudzinski JJ, Marsh IR, Chakravarty I, Arthur IS, Zangl LM, Brown RJ, Nystuen EJ, Emma SE, Kerr C, Carlson PM, Sriramaneni RN, Engle JW, Aluicio-Sarduy E, Barnhart TE, Le T, Kim K, Bednarz BP, Weichert JP, Patel RB, Morris ZS. Temporal analysis of type 1 interferon activation in tumor cells following external beam radiotherapy or targeted radionuclide therapy. Theranostics 2021; 11:6120-6137. [PMID: 33995649 PMCID: PMC8120207 DOI: 10.7150/thno.54881] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/26/2021] [Indexed: 12/15/2022] Open
Abstract
Rationale: Clinical interest in combining targeted radionuclide therapies (TRT) with immunotherapies is growing. External beam radiation therapy (EBRT) activates a type 1 interferon (IFN1) response mediated via stimulator of interferon genes (STING), and this is critical to its therapeutic interaction with immune checkpoint blockade. However, little is known about the time course of IFN1 activation after EBRT or whether this may be induced by decay of a TRT source. Methods: We examined the IFN1 response and expression of immune susceptibility markers in B78 and B16 melanomas and MOC2 head and neck cancer murine models using qPCR and western blot. For TRT, we used 90Y chelated to NM600, an alkylphosphocholine analog that exhibits selective uptake and retention in tumor cells including B78 and MOC2. Results: We observed significant IFN1 activation in all cell lines, with peak activation in B78, B16, and MOC2 cell lines occurring 7, 7, and 1 days, respectively, following RT for all doses. This effect was STING-dependent. Select IFN response genes remained upregulated at 14 days following RT. IFN1 activation following STING agonist treatment in vitro was identical to RT suggesting time course differences between cell lines were mediated by STING pathway kinetics and not DNA damage susceptibility. In vivo delivery of EBRT and TRT to B78 and MOC2 tumors resulted in a comparable time course and magnitude of IFN1 activation. In the MOC2 model, the combination of 90Y-NM600 and dual checkpoint blockade therapy reduced tumor growth and prolonged survival compared to single agent therapy and cumulative dose equivalent combination EBRT and dual checkpoint blockade therapy. Conclusions: We report the time course of the STING-dependent IFN1 response following radiation in multiple murine tumor models. We show the potential of TRT to stimulate IFN1 activation that is comparable to that observed with EBRT and this may be critical to the therapeutic integration of TRT with immunotherapies.
Collapse
MESH Headings
- Animals
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/physiopathology
- Carcinoma, Squamous Cell/radiotherapy
- Cell Line, Tumor
- Combined Modality Therapy
- Dose-Response Relationship, Radiation
- Female
- Gene Expression Regulation, Neoplastic/radiation effects
- Gene Knockout Techniques
- Head and Neck Neoplasms/pathology
- Immune Checkpoint Inhibitors
- Interferon Type I/biosynthesis
- Interferon Type I/genetics
- Interferon Type I/physiology
- Lymphocytes/drug effects
- Lymphocytes/radiation effects
- Melanoma, Experimental/immunology
- Melanoma, Experimental/physiopathology
- Melanoma, Experimental/radiotherapy
- Membrane Proteins/agonists
- Membrane Proteins/deficiency
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Mice
- Mice, Inbred C57BL
- Neoplasm Proteins/agonists
- Neoplasm Proteins/physiology
- Radiopharmaceuticals/pharmacokinetics
- Radiopharmaceuticals/therapeutic use
- Time Factors
- Tumor Protein, Translationally-Controlled 1
- Tumor Stem Cell Assay
- Up-Regulation
- Yttrium Radioisotopes/pharmacokinetics
- Yttrium Radioisotopes/therapeutic use
Collapse
Affiliation(s)
- Justin C. Jagodinsky
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Won Jong Jin
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Amber M. Bates
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Reinier Hernandez
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Joseph J. Grudzinski
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Ian R. Marsh
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Ishan Chakravarty
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Ian S. Arthur
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Luke M. Zangl
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Ryan J. Brown
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Erin J. Nystuen
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Sarah E. Emma
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Caroline Kerr
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Peter M. Carlson
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Raghava N. Sriramaneni
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Jonathan W. Engle
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Eduardo Aluicio-Sarduy
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Todd E. Barnhart
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Trang Le
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Bryan P. Bednarz
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Jamey P. Weichert
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Ravi B. Patel
- Department of Radiation Oncology, University of Pittsburgh School Hillman Cancer Center, Pittsburgh, PA
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
16
|
Shahi A, Weiss GE, Bhattacharya S, Baiu DC, Marino R, Pula T, Callander NS, Asimakopoulos F, Otto M. Targeted treatment of multiple myeloma with a radioiodinated small molecule radiopharmaceutical. Leuk Lymphoma 2021; 62:1518-1521. [PMID: 33502270 DOI: 10.1080/10428194.2021.1876858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Ankita Shahi
- Department of Pediatrics, School of Medicine and Public Health, Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Gerald E Weiss
- Department of Pediatrics, School of Medicine and Public Health, Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Saswati Bhattacharya
- Department of Medicine, Division of Hematology/Oncology, School of Medicine and Public Health, Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Dana C Baiu
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Roberta Marino
- Department of Therapeutics Production and Quality, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Taner Pula
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Natalie S Callander
- Department of Medicine, Division of Hematology/Oncology, School of Medicine and Public Health, Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Fotis Asimakopoulos
- Department of Medicine, Division of Hematology/Oncology, School of Medicine and Public Health, Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.,Department of Medicine, Division of Blood and Marrow Transplantation, Moores Cancer Center, University of California-San Diego, CA, USA
| | - Mario Otto
- Department of Pediatrics, School of Medicine and Public Health, Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
17
|
Anderson PM. Radiopharmaceuticals for Treatment of Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1257:45-53. [PMID: 32483729 DOI: 10.1007/978-3-030-43032-0_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although trace amounts of radioactivity are routinely used to detect osteosarcoma, the use of larger therapeutic amounts of radiation is often an unrecognized opportunity to treat metastatic osteosarcoma. This chapter will review a number of approaches to use ionizing radiation in the form of injectable radiopharmaceuticals. Since bone metastases are a common pattern of metastatic spread of cancer in general, a number of bone-seeking radiopharmaceuticals have been developed and FDA approved for treatment of bone metastases. Although osteosarcoma, a bone-forming cancer, would seem ideally suited to be treated with bone seekers, patterns of relapse involving non-ossifying metastases remain a major problem to be overcome. Thus, this review will not only describe experience using a number of bone-seeking radiopharmaceuticals such as 153-samarium-EDTMP, 153-samarium-DOTMP, and 223-radium against osteosarcoma, but also approaches to identify patients who may benefit as well as some means to the improve overall efficacy including combination therapy with routine agents and using nuclear imaging to develop best strategy for use. These include imaging with not only 99mTc-MDP standard bone scans, but also 99mTc-MDP bone scans with SPECT CT, bone-specific sodium fluoride PET-CT (Na18F), and 18FDG-PET-CT. Accurate knowledge of oligometastatic active disease can facilitate more effective use of combination therapy, including radiosensitizers and local control measures, for example, stereotactic body radiotherapy (SBRT) and/or cryoablation to reduce disease burden as well as manage and prevent micrometastatic disease from growing and metastasizing. Finally, a new tumor-specific radiopharmaceutical, CLR 131, may also provide another radiopharmaceutical to treat both osteoblastic and non-ossifying areas of osteosarcoma.
Collapse
Affiliation(s)
- Peter M Anderson
- Pediatric and Taussig Cancer Institutes, Department of Pediatric Hematology, Oncology and BMT, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|