1
|
Janowicz PW, Boele T, Maschmeyer RT, Gholami YH, Kempe EG, Stringer BW, Stoner SP, Zhang M, du Toit-Thompson T, Williams F, Touffu A, Munoz L, Kuncic Z, Brighi C, Waddington DEJ. Enhanced detection of glioblastoma vasculature with superparamagnetic iron oxide nanoparticles and MRI. Sci Rep 2025; 15:14283. [PMID: 40274951 PMCID: PMC12022243 DOI: 10.1038/s41598-025-97943-y] [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: 01/22/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Detecting glioblastoma infiltration in the brain is challenging due to limited MRI contrast beyond the enhancing tumour core. This study aims to investigate the potential of superparamagnetic iron oxide nanoparticles (SPIONs) as contrast agents for improved detection of diffuse brain cancer. We examine the distribution and pharmacokinetics of SPIONs in glioblastoma models with intact and disrupted blood-brain barriers. Using MRI, we imaged RN1-luc and U87MG mice injected with Gadovist and SPIONs, observing differences in blood-brain barrier permeability. Peripheral imaging showed strong uptake of nanoparticles in the liver and spleen, while vascular and renal signals were transient. Susceptibility gradient mapping enabled positive nanoparticle contrast within tumours and provided additional information on tumour angiogenesis. This approach offers a novel method for detecting diffuse brain cancer. Our findings demonstrate that SPIONs enhance glioblastoma detection beyond conventional MRI, providing insights into tumour angiogenesis and opening new avenues for early diagnosis and targeted treatment strategies.
Collapse
Affiliation(s)
- Phillip W Janowicz
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| | - Thomas Boele
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Richard T Maschmeyer
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
| | - Yaser H Gholami
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, Australia
| | - Emma G Kempe
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Brett W Stringer
- Institute for Biomedicine and Glycomics, Griffith University, Brisbane, Australia
| | - Shihani P Stoner
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Marie Zhang
- Imagion Biosystems Ltd, Melbourne, Australia
| | - Taymin du Toit-Thompson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Fern Williams
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Aude Touffu
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Lenka Munoz
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Zdenka Kuncic
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
- The Sydney Nano Institute, The University of Sydney, Sydney, Australia
| | - Caterina Brighi
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - David E J Waddington
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| |
Collapse
|
2
|
Li Y, Thamizhchelvan AM, Ma H, Padelford J, Zhang Z, Wu T, Gu Q, Wang Z, Mao H. A subtype specific probe for targeted magnetic resonance imaging of M2 tumor-associated macrophages in brain tumors. Acta Biomater 2025; 194:336-351. [PMID: 39805525 DOI: 10.1016/j.actbio.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/15/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Pro-tumoral M2 tumor-associated macrophages (TAMs) play a critical role in the tumor immune microenvironment (TIME), making them an important therapeutic target for cancer treatment. Approaches for imaging and monitoring M2 TAMs, as well as tracking their changes in response to tumor progression or treatment are highly sought-after but remain underdeveloped. Here, we report an M2-targeted magnetic resonance imaging (MRI) probe based on sub-5 nm ultrafine iron oxide nanoparticles (uIONP), featuring an anti-biofouling coating to prevent non-specific macrophage uptake and an M2-specific peptide ligand (M2pep) for active targeting of M2 TAMs. The targeting specificity of M2pep-uIONP was validated in vitro, using M0, M1, and M2 macrophages, and in vivo, using an orthotopic patient-tissue-derived xenograft (PDX) mouse model of glioblastoma (GBM). MRI of the mice revealed hypointense contrast in T2-weighted images of intracranial tumors 24 h after receiving intravenous (i.v.) injection of M2pep-uIONP. In contrast, no noticeable contrast change was observed in mice receiving scrambled-sequence M2pep-conjugated uIONP (scM2pep-uIONP) or the commercially available iron oxide nanoparticle formulation, Ferumoxytol. Measurement of nanoparticle-induced T2 value changes in tumors showed 38 %, 9 %, and 2 % decrease for M2pep-uIONP, scM2pep-uIONP, and Ferumoxytol, respectively. Moreover, M2pep-uIONP exhibited 88.7-fold higher intra-tumoral accumulation compared to co-injected Ferumoxytol at 24 h post-injection. Immunofluorescence-stained tumor sections showed that CD68+/CD163+ M2 TAMs were highly co-localized with Cy7-M2pep-uIONP, but not with Cy7-scM2pep-uIONP and Cy7-Ferumoxytol. Flow cytometry analysis revealed 26 ± 10 % of M2 TAMs were targeted by M2pep-uIONP, which was significantly higher than Ferumoxytol (16 ± 1 %) and scM2pep-uIONP (13 ± 4 %) with the same dosage (20 mg Fe/kg). These findings demonstrate that M2pep-uIONP functions as a ligand-mediated MRI probe for targeted imaging of M2 TAMs in GBM, with potential applications for imaging of M2 TAM in other cancer types. STATEMENT OF SIGNIFICANCE: Targeting the pro-tumoral M2 subtype of tumor-associated macrophages (TAMs) to modulate the tumor immune microenvironment (TIME) is an emerging strategy for developing novel cancer therapies and enhancing the efficacy of existing treatments. In this study, we have developed a magnetic resonance imaging (MRI) probe using sub-5 nm ultrafine iron oxide nanoparticles (uIONP), which are coated with an anti-biofouling polymer and conjugated to an M2-specific peptide ligand (M2pep). Our results demonstrate that M2pep-uIONP exhibits an 88.7-fold higher accumulation in intracranial tumors in an orthotopic patient-derived xenograft (PDX) model of glioblastoma compared to the commercial iron oxide nanoparticle, Ferumoxytol. This enhanced accumulation enables M2pep-uIONP to induce significant MRI contrast, providing a non-invasive imaging tool to visualize M2 TAMs and monitor changes in the TIME of brain tumors and potentially other cancers.
Collapse
Affiliation(s)
- Yuancheng Li
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA; 5M Biomed, LLC, Atlanta, Georgia 30303, USA
| | - Anbu Mozhi Thamizhchelvan
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Hedi Ma
- 5M Biomed, LLC, Atlanta, Georgia 30303, USA
| | | | - Zhaobin Zhang
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Tianhe Wu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Quanquan Gu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Zi Wang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA; Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| |
Collapse
|
3
|
Giordano C, Marrone L, Romano S, Della Pepa GM, Donzelli CM, Tufano M, Capasso M, Lasorsa VA, Quintavalle C, Guerri G, Martucci M, Auricchio A, Gessi M, Sala E, Olivi A, Romano MF, Gaudino S. The FKBP51s Splice Isoform Predicts Unfavorable Prognosis in Patients with Glioblastoma. CANCER RESEARCH COMMUNICATIONS 2024; 4:1296-1306. [PMID: 38651817 PMCID: PMC11097923 DOI: 10.1158/2767-9764.crc-24-0083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/21/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
The primary treatment for glioblastoma (GBM) is removing the tumor mass as defined by MRI. However, MRI has limited diagnostic and predictive value. Tumor-associated macrophages (TAM) are abundant in GBM tumor microenvironment (TME) and are found in peripheral blood (PB). FKBP51 expression, with its canonical and spliced isoforms, is constitutive in immune cells and aberrant in GBM. Spliced FKBP51s supports M2 polarization. To find an immunologic signature that combined with MRI could advance in diagnosis, we immunophenotyped the macrophages of TME and PB from 37 patients with GBM using FKBP51s and classical M1-M2 markers. We also determined the tumor levels of FKBP51s, PD-L1, and HLA-DR. Tumors expressing FKBP51s showed an increase in various M2 phenotypes and regulatory T cells in PB, indicating immunosuppression. Tumors expressing FKBP51s also activated STAT3 and were associated with reduced survival. Correlative studies with MRI and tumor/macrophages cocultures allowed to interpret TAMs. Tumor volume correlated with M1 infiltration of TME. Cocultures with spheroids produced M1 polarization, suggesting that M1 macrophages may infiltrate alongside cancer stem cells. Cocultures of adherent cells developed the M2 phenotype CD163/FKBP51s expressing pSTAT6, a transcription factor enabling migration and invasion. In patients with recurrences, increased counts of CD163/FKBP51s monocyte/macrophages in PB correlated with callosal infiltration and were accompanied by a concomitant decrease in TME-infiltrating M1 macrophages. PB PD-L1/FKBP51s connoted necrotic tumors. In conclusion, FKBP51s identifies a GBM subtype that significantly impairs the immune system. Moreover, FKBP51s marks PB macrophages associated with MRI features of glioma malignancy that can aid in patient monitoring. SIGNIFICANCE Our research suggests that by combining imaging with analysis of monocyte/macrophage subsets in patients with GBM, we can enhance our understanding of the disease and assist in its treatment. We discovered a similarity in the macrophage composition between the TME and PB, and through association with imaging, we could interpret macrophages. In addition, we identified a predictive biomarker that drew more attention to immune suppression of patients with GBM.
Collapse
Affiliation(s)
- Carolina Giordano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, Universitaà Cattolica del Sacro Cuore, Rome, Italy
| | - Laura Marrone
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
| | - Simona Romano
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
| | - Giuseppe Maria Della Pepa
- UOC Neurochirurgia, Istituto di Neurochirurgia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica S. Cuore, Roma, Italy
| | - Carlo Maria Donzelli
- UOC Neurochirurgia, Istituto di Neurochirurgia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica S. Cuore, Roma, Italy
| | - Martina Tufano
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
| | - Mario Capasso
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Vito Alessandro Lasorsa
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Cristina Quintavalle
- Istituto di Endocrinologia e Oncologia Sperimentale “Gaetano Salvatore” (IEOS), Consiglio Nazionale delle Ricerche (CNR), Napoli, Italia
| | - Giulia Guerri
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, Universitaà Cattolica del Sacro Cuore, Rome, Italy
| | - Matia Martucci
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, Universitaà Cattolica del Sacro Cuore, Rome, Italy
| | - Annamaria Auricchio
- UOC Neurochirurgia, Istituto di Neurochirurgia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica S. Cuore, Roma, Italy
| | - Marco Gessi
- UOS di Neuropatologia, UOC Anatomia Patologica, Fondazione Policlinico “A. Gemelli” IRCCS, Rome, Italy
| | - Evis Sala
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, Universitaà Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Olivi
- UOC Neurochirurgia, Istituto di Neurochirurgia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica S. Cuore, Roma, Italy
| | - Maria Fiammetta Romano
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
| | - Simona Gaudino
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, Universitaà Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
4
|
Wang DJJ, Hua J, Cao D, Ho ML. Neurofluids and the glymphatic system: anatomy, physiology, and imaging. Br J Radiol 2023; 96:20230016. [PMID: 37191063 PMCID: PMC10607419 DOI: 10.1259/bjr.20230016] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/05/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
First described in 2012, the glymphatic system is responsible for maintaining homeostasis within the central nervous system, including nutrient delivery, waste clearance, and consistency of the ionic microenvironment. It is comprised of glial cells and barrier systems that modulate neurofluid production, circulation, and exchange. Experimental interrogation of neurofluid dynamics is restricted to ex vivo and in vitro studies in animals and humans, therefore diagnostic imaging plays an important role in minimally invasive evaluation. This review article will synthesize current knowledge and theories regarding neurofluid circulation and implications for neuroimaging. First, we will discuss the anatomy of the neurogliovascular unit, including paravascular and perivascular pathways of fluid exchange. In addition, we will summarize the structure and function of barrier systems including the blood-brain, blood-cerebrospinal fluid, and brain-cerebrospinal fluid barriers. Next, we will mention physiologic factors that yield normal variations in neurofluid circulation, and how various disease pathologies can disrupt glymphatic drainage pathways. Lastly, we will cover the spectrum of diagnostic imaging and interventional techniques with relevance to glymphatic structure, flow, and function. We conclude by highlighting current barriers and future directions for translational imaging and applications to neurologic disorders.
Collapse
Affiliation(s)
- Danny JJ Wang
- Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, United States
| | | | | | - Mai-Lan Ho
- Nationwide Children’s Hospital and The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
5
|
Ravi H, Arias-Lorza AM, Costello JR, Han HS, Jeong DK, Klinz SG, Sachdev JC, Korn RL, Raghunand N. Pretherapy Ferumoxytol-enhanced MRI to Predict Response to Liposomal Irinotecan in Metastatic Breast Cancer. Radiol Imaging Cancer 2023; 5:e220022. [PMID: 36734848 PMCID: PMC10077095 DOI: 10.1148/rycan.220022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Purpose To investigate ferumoxytol (FMX)-enhanced MRI as a pretreatment predictor of response to liposomal irinotecan (nal-IRI) for thoracoabdominal and brain metastases in women with metastatic breast cancer (mBC). Materials and Methods In this phase 1 expansion trial (ClinicalTrials.gov identifier, NCT01770353; 27 participants), 49 thoracoabdominal (19 participants; mean age, 48 years ± 11 [SD]) and 19 brain (seven participants; mean age, 54 years ± 8) metastases were analyzed on MR images acquired before, 1-4 hours after, and 16-24 hours after FMX administration. In thoracoabdominal metastases, tumor transverse relaxation rate (R*2) was normalized to the mean R*2 in the spleen (rR*2), and the tumor histogram metric rR*2,N, representing the average of rR*2 in voxels above the nth percentile, was computed. In brain metastases, a novel compartmentation index was derived by applying the MRI signal equation to phantom-calibrated coregistered FMX-enhanced MRI brain scans acquired before, 1-4 hours after, and 16-24 hours after FMX administration. The fraction of voxels with an FMX compartmentation index greater than 1 was computed over the whole tumor (FCIGT1) and from voxels above the 90th percentile R*2 (FCIGT1 R*2,90). Results rR*2,90 computed from pretherapy MRI performed 16-24 hours after FMX administration, without reference to calibration phantoms, predicted response to nal-IRI in thoracoabdominal metastases (accuracy, 74%). rR*2,90 performance was robust to the inclusion of some peritumoral tissue within the tumor region of interest. FCIGT1 R*2,90 provided 79% accuracy on cross-validation in prediction of response in brain metastases. Conclusion This first in-human study focused on mBC suggests that FMX-enhanced MRI biologic markers can be useful for pretherapy prediction of response to nal-IRI in patients with mBC. Keywords: MRI Contrast Agent, MRI, Breast, Head/Neck, Tumor Response, Experimental Investigations, Brain/Brain Stem Clinical trial registration no. NCT01770353 Supplemental material is available for this article. © RSNA, 2023 See also commentary by Daldrup-Link in this issue.
Collapse
Affiliation(s)
- Harshan Ravi
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Andres M Arias-Lorza
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - James R Costello
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Hyo Sook Han
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Daniel K Jeong
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Stephan G Klinz
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Jasgit C Sachdev
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Ronald L Korn
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Natarajan Raghunand
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| |
Collapse
|
6
|
Li Y, Xie M, Jones JB, Zhang Z, Wang Z, Dang T, Wang X, Lipowska M, Mao H. Targeted Delivery of DNA Topoisomerase Inhibitor SN38 to Intracranial Tumors of Glioblastoma Using Sub-5 Ultrafine Iron Oxide Nanoparticles. Adv Healthc Mater 2022; 11:e2102816. [PMID: 35481625 DOI: 10.1002/adhm.202102816] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 04/07/2022] [Indexed: 11/09/2022]
Abstract
Effectively delivering therapeutics for treating brain tumors is hindered by the physical and biological barriers in the brain. Even with the compromised blood-brain barrier and highly angiogenic blood-tumor barrier seen in glioblastoma (GBM), most drugs, including nanomaterial-based formulations, hardly reach intracranial tumors. This work investigates sub-5 nm ultrafine iron oxide nanoparticles (uIONP) with 3.5 nm core diameter as a carrier for delivering DNA topoisomerase inhibitor 7-ethyl-10-hydroxyl camptothecin (SN38) to treat GBM. Given a higher surface-to-volume ratio, uIONP shows one- or three-folds higher SN38 loading efficiency (48.3 ± 6.1%, mg/mg Fe) than those with core sizes of 10 or 20 nm. SN38 encapsulated in the coating polymer exhibits pH sensitive release with <10% over 48 h at pH 7.4, but 86% at pH 5, thus being protected from converting to inactive glucuronide by UDP-glucuronosyltransferase 1A1. Conjugating αv β3 -integrin-targeted cyclo(Arg-Gly-Asp-D-Phe-Cys) (RGD) as ligands, RGD-uIONP/SN38 demonstrates targeted cytotoxicity to αv β3 -integrin-overexpressed U87MG GBM cells with a half-maximal inhibitory concentration (IC50 ) of 30.9 ± 2.2 nm. The efficacy study using an orthotopic mouse model of GBM reveals tumor-specific delivery of 11.5% injected RGD-uIONP/SN38 (10 mg Fe kg-1 ), significantly prolonging the survival in mice by 41%, comparing to those treated with SN38 alone (p < 0.001).
Collapse
Affiliation(s)
- Yuancheng Li
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
- 5M Biomed, LLC Atlanta GA 30303 USA
| | - Manman Xie
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Joshua B. Jones
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Zhaobin Zhang
- Department of Neurosurgery Emory University Atlanta GA 30329 USA
| | - Zi Wang
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Tu Dang
- Division of Research Philadelphia College of Osteopathic Medicine – Georgia Campus Suwanee GA 30024 USA
| | - Xinyu Wang
- Department of Pharmaceutical Sciences Philadelphia College of Osteopathic Medicine – Georgia Campus Suwanee GA 30024 USA
| | - Malgorzata Lipowska
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| |
Collapse
|
7
|
Ng TSC, Allen HH, Rashidian M, Miller MA. Probing immune infiltration dynamics in cancer by in vivo imaging. Curr Opin Chem Biol 2022; 67:102117. [PMID: 35219177 PMCID: PMC9118268 DOI: 10.1016/j.cbpa.2022.102117] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/11/2022]
Abstract
Cancer immunotherapies typically aim to stimulate the accumulation and activity of cytotoxic T-cells or pro-inflammatory antigen-presenting cells, reduce immunosuppressive myeloid cells or regulatory T-cells, or elicit some combination of effects thereof. Notwithstanding the encouraging results, immunotherapies such as PD-1/PD-L1-targeted immune checkpoint blockade act heterogeneously across individual patients. It remains challenging to predict and monitor individual responses, especially across multiple sites of metastasis or sites of potential toxicity. To address this need, in vivo imaging of both adaptive and innate immune cell populations has emerged as a tool to quantify spatial leukocyte accumulation in tumors non-invasively. Here we review recent progress in the translational development of probes for in vivo leukocyte imaging, focusing on complementary perspectives provided by imaging of T-cells, phagocytic macrophages, and their responses to therapy.
Collapse
Affiliation(s)
- Thomas S C Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Boston, MA 02114, United States; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Boston, MA 02114, United States
| | - Harris H Allen
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA 02115, United States
| | - Mohammad Rashidian
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA 02115, United States; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, United States
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Boston, MA 02114, United States; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Boston, MA 02114, United States.
| |
Collapse
|
8
|
Huang Y, Hsu JC, Koo H, Cormode DP. Repurposing ferumoxytol: Diagnostic and therapeutic applications of an FDA-approved nanoparticle. Am J Cancer Res 2022; 12:796-816. [PMID: 34976214 PMCID: PMC8692919 DOI: 10.7150/thno.67375] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/12/2021] [Indexed: 02/07/2023] Open
Abstract
Ferumoxytol is an intravenous iron oxide nanoparticle formulation that has been approved by the U.S. Food and Drug Administration (FDA) for treating anemia in patients with chronic kidney disease. In recent years, ferumoxytol has also been demonstrated to have potential for many additional biomedical applications due to its excellent inherent physical properties, such as superparamagnetism, biocatalytic activity, and immunomodulatory behavior. With good safety and clearance profiles, ferumoxytol has been extensively utilized in both preclinical and clinical studies. Here, we first introduce the medical needs and the value of current iron oxide nanoparticle formulations in the market. We then focus on ferumoxytol nanoparticles and their physicochemical, diagnostic, and therapeutic properties. We include examples describing their use in various biomedical applications, including magnetic resonance imaging (MRI), multimodality imaging, iron deficiency treatment, immunotherapy, microbial biofilm treatment and drug delivery. Finally, we provide a brief conclusion and offer our perspectives on the current limitations and emerging applications of ferumoxytol in biomedicine. Overall, this review provides a comprehensive summary of the developments of ferumoxytol as an agent with diagnostic, therapeutic, and theranostic functionalities.
Collapse
|
9
|
MRI and PET of Brain Tumor Neuroinflammation in the Era of Immunotherapy, From the AJR Special Series on Inflammation. AJR Am J Roentgenol 2021; 218:582-596. [PMID: 34259035 DOI: 10.2214/ajr.21.26159] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
With the emergence of immune-modulating therapies, brain tumors present significant diagnostic imaging challenges. These challenges include planning personalized treatment and adjudicating accurate monitoring approaches and therapeutically specific response criteria. This has been due, in part, to the reliance on nonspecific imaging metrics, such as gadolinium-contrast-enhanced MRI or FDG PET, and rapidly evolving biologic understanding of neuroinflammation. The importance of the tumor-immune interaction and ability to therapeutically augment inflammation to improve clinical outcomes necessitates that the radiologist develop a working knowledge of the immune system and its role in clinical neuroimaging. In this article, we review relevant biologic concepts of the tumor microenvironment of primary and metastatic brain tumors, these tumors' interactions with the immune system, and MRI and PET methods for imaging inflammatory elements associated with these malignancies. Recognizing the growing fields of immunotherapeutics and precision oncology, we highlight clinically translatable imaging metrics for the diagnosis and monitoring of brain tumor neuroinflammation. Practical guidance is provided for implementing iron nanoparticle imaging, including imaging indications, protocol, interpretation, and pitfalls. A comprehensive understanding of the inflammatory mechanisms within brain tumors and their imaging features will facilitate the development of innovative non-invasive prognostic and predictive imaging strategies for precision oncology.
Collapse
|
10
|
Programmed cell death, redox imbalance, and cancer therapeutics. Apoptosis 2021; 26:385-414. [PMID: 34236569 DOI: 10.1007/s10495-021-01682-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 02/06/2023]
Abstract
Cancer cells are disordered by nature and thus featured by higher internal redox level than healthy cells. Redox imbalance could trigger programmed cell death if exceeded a certain threshold, rendering therapeutic strategies relying on redox control a possible cancer management solution. Yet, various programmed cell death events have been consecutively discovered, complicating our understandings on their associations with redox imbalance and clinical implications especially therapeutic design. Thus, it is imperative to understand differences and similarities among programmed cell death events regarding their associations with redox imbalance for improved control over these events in malignant cells as well as appropriate design on therapeutic approaches relying on redox control. This review addresses these issues and concludes by bringing affront cold atmospheric plasma as an emerging redox controller with translational potential in clinics.
Collapse
|
11
|
Abstract
Magnetic resonance (MR) imaging is a crucial tool for evaluation of the skull base, enabling characterization of complex anatomy by utilizing multiple image contrasts. Recent technical MR advances have greatly enhanced radiologists' capability to diagnose skull base pathology and help direct management. In this paper, we will summarize cutting-edge clinical and emerging research MR techniques for the skull base, including high-resolution, phase-contrast, diffusion, perfusion, vascular, zero echo-time, elastography, spectroscopy, chemical exchange saturation transfer, PET/MR, ultra-high-field, and 3D visualization. For each imaging technique, we provide a high-level summary of underlying technical principles accompanied by relevant literature review and clinical imaging examples.
Collapse
Affiliation(s)
- Claudia F Kirsch
- Division Chief, Neuroradiology, Professor of Neuroradiology and Otolaryngology, Department of Radiology, Northwell Health, Zucker Hofstra School of Medicine at Northwell, North Shore University Hospital, Manhasset, NY
| | - Mai-Lan Ho
- Associate Professor of Radiology, Director of Research, Department of Radiology, Director, Advanced Neuroimaging Core, Chair, Asian Pacific American Network, Secretary, Association for Staff and Faculty Women, Nationwide Children's Hospital and The Ohio State University, Columbus, OH; Division Chief, Neuroradiology, Professor of Neuroradiology and Otolaryngology, Department of Radiology, Northwell Health, Zucker Hofstra School of Medicine at Northwell, North Shore University Hospital, Manhasset, NY.
| |
Collapse
|