1
|
Roncali L, Hindré F, Samarut E, Lacoeuille F, Rousseau A, Lemée JM, Garcion E, Chérel M. Current landscape and future directions of targeted-alpha-therapy for glioblastoma treatment. Theranostics 2025; 15:4861-4889. [PMID: 40303349 PMCID: PMC12036880 DOI: 10.7150/thno.106081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/02/2025] [Indexed: 05/02/2025] Open
Abstract
Glioblastoma (GB) is the most aggressive malignancy of the central nervous system. Despite two decades of intensive research since the establishment of the standard of care, emerging strategies have yet to produce consistent satisfactory outcomes. Because of its specific localisation and intricate characteristics, GB is a uniquely regulated solid tumour with a strong resistance to therapy. Advances in targeted radionuclide therapy (TRT), particularly with the introduction of a-emitting radionuclides, have unveiled potential avenues for the management of GB. Recent preclinical and clinical studies underscored promising advancements for targeted-α-therapy (TAT), but these therapeutic approaches exhibit a vast design heterogeneity, encompassing diverse radionuclides, vectors, target molecules, and administration modalities. This review seeks to critically assess the therapeutic landscape of GB through the perspective of TAT. Here, the focus is made on the advancements and limitations of in vivo explorations, pilot studies, and clinical trials, to determine the best directions for future investigations. In doing so, we hope to identify existing challenges and draw insights that might pave the way towards a more effective therapeutic approach.
Collapse
Affiliation(s)
- Loris Roncali
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela; E-15782 Santiago de Compostela, Spain
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
| | - François Hindré
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- PRIMEX (Experimental Imagery and Radiobiology Platform), University of Angers, SFR 4208; F-49000 Angers, France
| | - Edouard Samarut
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
- Department of Neurosurgery & Neurotraumatology, University Hospital of Nantes; F-44093 Nantes, France
| | - Franck Lacoeuille
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Nuclear Medicine, University Hospital of Angers; F-49000 Angers, France
| | - Audrey Rousseau
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Pathology, University Hospital of Angers; F-49000 Angers, France
| | - Jean-Michel Lemée
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Neurosurgery, University Hospital of Angers; F-49000 Angers, France
| | - Emmanuel Garcion
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- PACEM (Platform of Cellular and Molecular Analysis), University of Angers, SFR 4208; F-49000 Angers, France
| | - Michel Chérel
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
- Institut de Cancérologie de l'Ouest, Department of Nuclear Medicine; F-44160 Saint-Herblain, France
| |
Collapse
|
2
|
Brenner AJ, Patel T, Bao A, Phillips WT, Michalek JE, Youssef M, Weinberg JS, Kamiya Matsuoka C, Hedrick MH, LaFrance N, Moore M, Floyd JR. Convection enhanced delivery of Rhenium ( 186Re) Obisbemeda ( 186RNL) in recurrent glioma: a multicenter, single arm, phase 1 clinical trial. Nat Commun 2025; 16:2079. [PMID: 40055350 PMCID: PMC11889265 DOI: 10.1038/s41467-025-57263-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Rhenium (186Re) Obisbemeda (186RNL), chelated-186Re encapsulated in nanoliposomes and delivered to brain tumors via convection enhanced delivery (CED), was evaluated in a Phase 1 dose escalation trial (NCT01906385). The primary objective was to determine the maximum tolerated dose (MTD). Secondary objectives included safety and tolerability, dose distribution, the overall response rate (ORR), disease-specific progression-free survival (PFS), and overall survival (OS). 21 patients received up to 22.3 mCi 186RNL over 6 dosing cohorts. Most adverse events (AEs) were unrelated to 186RNL and the MTD was not reached. Although not predefined outcomes, the mOS and mPFS were 11 and 4 months, respectively, and found to correlate with radiation absorbed dose to the tumor and percent tumor treated. When dichotomized by absorbed dose of 100 Gy, the mOS and mPFS were 17 months and 6 months, respectively, for >100 Gy, compared to 6 (mOS) and 2 (mPFS) months, respectively, for <100 Gy. For ORR, 57.1% exhibited stable disease (SD), 4.8% partial response, and 38.1% progressive disease. Overall, patients received radiation absorbed doses without significant toxicity higher than possible with external beam radiation therapy (EBRT) and demonstrated mOS beyond standard of care for recurrent glioblastoma (~8 months).
Collapse
Affiliation(s)
- Andrew J Brenner
- Mays Cancer Center at UT Health San Antonio, San Antonio, TX, USA.
| | - Toral Patel
- UT Southwestern Medical Center of Dallas, Dallas, TX, USA
| | - Ande Bao
- Case Western Reserve University, Cleveland, OH, USA
| | | | - Joel E Michalek
- Mays Cancer Center at UT Health San Antonio, San Antonio, TX, USA
| | | | | | | | | | | | | | - John R Floyd
- Mays Cancer Center at UT Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
3
|
Kozak A, Lavrih E, Mikhaylov G, Turk B, Vasiljeva O. Navigating the Clinical Landscape of Liposomal Therapeutics in Cancer Treatment. Pharmaceutics 2025; 17:276. [PMID: 40006643 PMCID: PMC11859495 DOI: 10.3390/pharmaceutics17020276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Liposome-based targeted drug delivery systems represent a significant advancement in pharmaceutical science, offering distinct advantages that enhance the efficacy and safety of various therapies. These versatile carriers can encapsulate both hydrophilic and hydrophobic drugs, making them particularly valuable in clinical settings. This review explores the critical role of liposomal formulations in improving drug pharmacokinetics and minimizing side effects, especially in oncology, where targeted delivery to tumor cells is essential. Outlining the properties of different types of liposomes, we focus on the effects of these properties on the liposomes' targeting and drug release capabilities through innovative surface modifications and describe the most common methods of liposome preparation and characterization. Furthermore, this review provides an in-depth analysis of the properties and composition of liposomal-based nanocarriers, with a unique focus on ongoing clinical trials and recently approved therapies. It offers a comprehensive overview of the latest advancements in pre-clinical research and highlights the critical progress in clinical development, offering insights into the clinical impact and regulatory approvals. Ultimately, this review underscores the transformative potential of liposomal nanocarriers in modern therapeutics, suggesting avenues for future innovations and clinical breakthroughs.
Collapse
Affiliation(s)
- Andreja Kozak
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (A.K.); (E.L.); (G.M.); (B.T.)
| | - Ernestina Lavrih
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (A.K.); (E.L.); (G.M.); (B.T.)
- Jožef Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
| | - Georgy Mikhaylov
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (A.K.); (E.L.); (G.M.); (B.T.)
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (A.K.); (E.L.); (G.M.); (B.T.)
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Olga Vasiljeva
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (A.K.); (E.L.); (G.M.); (B.T.)
| |
Collapse
|
4
|
Razavi R, Khajouei G, Divsalar F, Dawi E, Amiri M. Recent advances on brain drug delivery via nanoparticles: alternative future materials for neuroscience applications; a review. Rev Neurosci 2025:revneuro-2024-0086. [PMID: 39829237 DOI: 10.1515/revneuro-2024-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/29/2024] [Indexed: 01/22/2025]
Abstract
Essentially, the blood-brain barrier (BBB) serves as a line of demarcation between neural tissues and the bloodstream. A unique and protective characteristic of the blood-brain barrier is its ability to maintain cerebral homeostasis by regulating the flux of molecules and ions. The inability to uphold proper functioning in any of these constituents leads to the disruption of this specialized multicellular arrangement, consequently fostering neuroinflammation and neurodegeneration. Recent advancements in nanomedicine have been regarded as a promising avenue for improving the delivery of drugs to the central nervous system in the modern era. A major benefit of this innovation is that it allows drugs to accumulate selectively within the cerebral area by circumventing the blood-brain barrier. Although brain-targeted nanomedicines have demonstrated impressive achievements, certain limitations in targeting specificity still exist. In this examination, we scrutinize the distinctive physical and chemical attributes of nanoparticles (NPs) contributing to their facilitation in BBB traversal. We explore the various mechanisms governing NP passage over the BBB, encompassing paracellular conveyance, mediated transport, as well as adsorptive- and receptor-mediated transcytosis. The therapeutic success of NPs for the treatment of brain tumors has been extensively investigated through the use of various categories of NPs. Among these are polymeric nanoparticles, liposomes, solid lipid nanoparticles, dendrimers, metallic nanoparticles, quantum dots, and nanogels. The potential utility of nanoparticles goes beyond their ability to transport pharmaceuticals. They can serve as adept imaging contrast agents, capable of being linked with imaging probes. This will facilitate tumor visualization, delineate lesion boundaries and margins, and monitor drug delivery and treatment response. Versatile nanoparticles can be engineered to effectively target neoplastic lesions, serving dual roles in diagnostic imaging and therapeutic interventions. Subsequently, this discourse explores the constraints associated with nanoparticles in the context of treating brain tumors.
Collapse
Affiliation(s)
- Razieh Razavi
- Department of Chemistry, Faculty of Science, University of Jiroft, Jiroft, Iran
| | - Ghazal Khajouei
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| | - Fatemeh Divsalar
- Sina Hospital, Zarand Network and Health Center, 48463 Kerman University of Medical Sciences , Kerman, Iran
| | - Elmuez Dawi
- College of Humanities and Sciences, College of Humanities and Sciences, Department of Mathematics and Sciences, Ajman University, P.O. Box 346, Ajman, United Arab Emirates
| | - Mahnaz Amiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| |
Collapse
|
5
|
Wu C, Hormuth DA, Christenson CD, Woodall RT, Abdelmalik MRA, Phillips WT, Hughes TJR, Brenner AJ, Yankeelov TE. Image-guided patient-specific optimization of catheter placement for convection-enhanced nanoparticle delivery in recurrent glioblastoma. Comput Biol Med 2024; 179:108889. [PMID: 39032243 DOI: 10.1016/j.compbiomed.2024.108889] [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: 01/11/2024] [Revised: 06/15/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Proper catheter placement for convection-enhanced delivery (CED) is required to maximize tumor coverage and minimize exposure to healthy tissue. We developed an image-based model to patient-specifically optimize the catheter placement for rhenium-186 (186Re)-nanoliposomes (RNL) delivery to treat recurrent glioblastoma (rGBM). METHODS The model consists of the 1) fluid fields generated via catheter infusion, 2) dynamic transport of RNL, and 3) transforming RNL concentration to the SPECT signal. Patient-specific tissue geometries were assigned from pre-delivery MRIs. Model parameters were personalized with either 1) individual-based calibration with longitudinal SPECT images, or 2) population-based assignment via leave-one-out cross-validation. The concordance correlation coefficient (CCC) was used to quantify the agreement between the predicted and measured SPECT signals. The model was then used to simulate RNL distributions from a range of catheter placements, resulting in a ratio of the cumulative RNL dose outside versus inside the tumor, the "off-target ratio" (OTR). Optimal catheter placement) was identified by minimizing OTR. RESULTS Fifteen patients with rGBM from a Phase I/II clinical trial (NCT01906385) were recruited to the study. Our model, with either individual-calibrated or population-assigned parameters, achieved high accuracy (CCC > 0.80) for predicting RNL distributions up to 24 h after delivery. The optimal catheter placements identified using this model achieved a median (range) of 34.56 % (14.70 %-61.12 %) reduction on OTR at the 24 h post-delivery in comparison to the original placements. CONCLUSIONS Our image-guided model achieved high accuracy for predicting patient-specific RNL distributions and indicates value for optimizing catheter placement for CED of radiolabeled liposomes.
Collapse
Affiliation(s)
- Chengyue Wu
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA; Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - David A Hormuth
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA; Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Chase D Christenson
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Ryan T Woodall
- Division of Mathematical Oncology, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd, Duarte, CA, 91010, USA
| | - Michael R A Abdelmalik
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA; Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - William T Phillips
- Department of Radiology, UT Health San Antonio, San Antonio, TX, 78229, USA
| | - Thomas J R Hughes
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA; Department of Aerospace Engineering and Engineering Mechanics, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew J Brenner
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX, 78229, USA
| | - Thomas E Yankeelov
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA; Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA; Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX, 78712, USA; Department of Oncology, The University of Texas at Austin, Austin, TX, 78712, USA; Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, 78712, USA; Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
6
|
Zhao X, Jakobsson V, Tao Y, Zhao T, Wang J, Khong PL, Chen X, Zhang J. Targeted Radionuclide Therapy in Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39042829 DOI: 10.1021/acsami.4c07850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Despite the development of various novel therapies, glioblastoma (GBM) remains a devastating disease, with a median survival of less than 15 months. Recently, targeted radionuclide therapy has shown significant progress in treating solid tumors, with the approval of Lutathera for neuroendocrine tumors and Pluvicto for prostate cancer by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA). This achievement has shed light on the potential of targeted radionuclide therapy for other solid tumors, including GBM. This review presents the current status of targeted radionuclide therapy in GBM, highlighting the commonly used therapeutic radionuclides emitting alpha, beta particles, and Auger electrons that could induce potent molecular and cellular damage to treat GBM. We then explore a range of targeting vectors, including small molecules, peptides, and antibodies, which selectively target antigen-expressing tumor cells with minimal or no binding to healthy tissues. Considering that radiopharmaceuticals for GBM are often administered locoregionally to bypass the blood-brain barrier (BBB), we review prominent delivery methods such as convection-enhanced delivery, local implantation, and stereotactic injections. Finally, we address the challenges of this therapeutic approach for GBM and propose potential solutions.
Collapse
Affiliation(s)
- Xiaobin Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Vivianne Jakobsson
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yucen Tao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Tianzhi Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jingyan Wang
- Xiamen University, School of Public Health, Xiang'an South Road, Xiamen 361102, China
| | - Pek-Lan Khong
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Departments of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
7
|
Lei J, Huang Y, Zhao Y, Zhou Z, Mao L, Liu Y. Nanotechnology as a new strategy for the diagnosis and treatment of gliomas. J Cancer 2024; 15:4643-4655. [PMID: 39006067 PMCID: PMC11242339 DOI: 10.7150/jca.96859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
Glioma is the most common malignant tumor of the central nervous system (CNS), and is characterized by high aggressiveness and a high recurrence rate. Currently, the main treatments for gliomas include surgical resection, temozolomide chemotherapy and radiotherapy. However, the prognosis of glioma patients after active standardized treatment is still poor, especially for glioblastoma (GBM); the median survival is still only 14.6 months, and the 5-year survival rate is only 4-5%. The current challenges in glioma treatment include difficulty in complete surgical resection, poor blood‒brain barrier (BBB) drug permeability, therapeutic resistance, and difficulty in tumor-specific targeting. In recent years, the rapid development of nanotechnology has provided new directions for diagnosing and treating gliomas. Nanoparticles (NPs) are characterized by excellent surface tunability, precise targeting, excellent biocompatibility, and high safety. In addition, NPs can be used for gene therapy, photodynamic therapy, and antiangiogenic therapy and can be combined with biomaterials for thermotherapy. In recent decades, breakthroughs in diagnosing and treating gliomas have been made with various functional NPs, and NPs are expected to become a new strategy for glioma diagnosis and treatment. In this paper, we review the main obstacles in the treatment of glioma and discuss the potential and challenges of the latest nanotechnology in the diagnosis and treatment of glioma.
Collapse
Affiliation(s)
- Jun Lei
- Department of Neurosurgery, The First People's Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yiyang Huang
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yichuan Zhao
- Southwest Medical University, Luzhou 646000, China
| | - Zhi Zhou
- Department of Neurosurgery, The First People's Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China
| | - Lei Mao
- Department of Neurosurgery, The First People's Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China
| | - Yanhui Liu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
Reilly RM, Georgiou CJ, Brown MK, Cai Z. Radiation nanomedicines for cancer treatment: a scientific journey and view of the landscape. EJNMMI Radiopharm Chem 2024; 9:37. [PMID: 38703297 PMCID: PMC11069497 DOI: 10.1186/s41181-024-00266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Radiation nanomedicines are nanoparticles labeled with radionuclides that emit α- or β-particles or Auger electrons for cancer treatment. We describe here our 15 years scientific journey studying locally-administered radiation nanomedicines for cancer treatment. We further present a view of the radiation nanomedicine landscape by reviewing research reported by other groups. MAIN BODY Gold nanoparticles were studied initially for radiosensitization of breast cancer to X-radiation therapy. These nanoparticles were labeled with 111In to assess their biodistribution after intratumoural vs. intravenous injection. Intravenous injection was limited by high liver and spleen uptake and low tumour uptake, while intratumoural injection provided high tumour uptake but low normal tissue uptake. Further, [111In]In-labeled gold nanoparticles modified with trastuzumab and injected iintratumourally exhibited strong tumour growth inhibition in mice with subcutaneous HER2-positive human breast cancer xenografts. In subsequent studies, strong tumour growth inhibition in mice was achieved without normal tissue toxicity in mice with human breast cancer xenografts injected intratumourally with gold nanoparticles labeled with β-particle emitting 177Lu and modified with panitumumab or trastuzumab to specifically bind EGFR or HER2, respectively. A nanoparticle depot (nanodepot) was designed to incorporate and deliver radiolabeled gold nanoparticles to tumours using brachytherapy needle insertion techniques. Treatment of mice with s.c. 4T1 murine mammary carcinoma tumours with a nanodepot incorporating [90Y]Y-labeled gold nanoparticles inserted into one tumour arrested tumour growth and caused an abscopal growth-inhibitory effect on a distant second tumour. Convection-enhanced delivery of [177Lu]Lu-AuNPs to orthotopic human glioblastoma multiforme (GBM) tumours in mice arrested tumour growth without normal tissue toxicity. Other groups have explored radiation nanomedicines for cancer treatment in preclinical animal tumour xenograft models using gold nanoparticles, liposomes, block copolymer micelles, dendrimers, carbon nanotubes, cellulose nanocrystals or iron oxide nanoparticles. These nanoparticles were labeled with radionuclides emitting Auger electrons (111In, 99mTc, 125I, 103Pd, 193mPt, 195mPt), β-particles (177Lu, 186Re, 188Re, 90Y, 198Au, 131I) or α-particles (225Ac, 213Bi, 212Pb, 211At, 223Ra). These studies employed intravenous or intratumoural injection or convection enhanced delivery. Local administration of these radiation nanomedicines was most effective and minimized normal tissue toxicity. CONCLUSIONS Radiation nanomedicines have shown great promise for treating cancer in preclinical studies. Local intratumoural administration avoids sequestration by the liver and spleen and is most effective for treating tumours, while minimizing normal tissue toxicity.
Collapse
Affiliation(s)
- Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.
- Princess Margaret Cancer Centre, Toronto, ON, Canada.
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.
- Joint Department of Medical Imaging, University Health Network, Toronto, ON, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| | | | - Madeline K Brown
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Iqbal J, Courville E, Kazim SF, Kogan M, Schmidt MH, Bowers CA. Role of nanotechnology in neurosurgery: A review of recent advances and their applications. World Neurosurg X 2024; 22:100298. [PMID: 38455250 PMCID: PMC10918265 DOI: 10.1016/j.wnsx.2024.100298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Affiliation(s)
- Javed Iqbal
- School of Medicine, King Edward Medical University, Lahore, Pakistan
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Evan Courville
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Syed Faraz Kazim
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Michael Kogan
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Meic H. Schmidt
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Christian A. Bowers
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| |
Collapse
|
10
|
Christenson C, Wu C, Hormuth DA, Huang S, Bao A, Brenner A, Yankeelov TE. Predicting the spatio-temporal response of recurrent glioblastoma treated with rhenium-186 labelled nanoliposomes. BRAIN MULTIPHYSICS 2023; 5:100084. [PMID: 38187909 PMCID: PMC10768931 DOI: 10.1016/j.brain.2023.100084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024] Open
Abstract
Rhenium-186 (186Re) labeled nanoliposome (RNL) therapy for recurrent glioblastoma patients has shown promise to improve outcomes by locally delivering radiation to affected areas. To optimize the delivery of RNL, we have developed a framework to predict patient-specific response to RNL using image-guided mathematical models. Methods We calibrated a family of reaction-diffusion type models with multi-modality imaging data from ten patients (NCR01906385) to predict the spatio-temporal dynamics of each patient's tumor. The data consisted of longitudinal magnetic resonance imaging (MRI) and single photon emission computed tomography (SPECT) to estimate tumor burden and local RNL activity, respectively. The optimal model from the family was selected and used to predict future growth. A simplified version of the model was used in a leave-one-out analysis to predict the development of an individual patient's tumor, based on cohort parameters. Results Across the cohort, predictions using patient-specific parameters with the selected model were able to achieve Spearman correlation coefficients (SCC) of 0.98 and 0.93 for tumor volume and total cell number, respectively, when compared to the measured data. Predictions utilizing the leave-one-out method achieved SCCs of 0.89 and 0.88 for volume and total cell number across the population, respectively. Conclusion We have shown that patient-specific calibrations of a biology-based mathematical model can be used to make early predictions of response to RNL therapy. Furthermore, the leave-one-out framework indicates that radiation doses determined by SPECT can be used to assign model parameters to make predictions directly following the conclusion of RNL treatment. Statement of Significance This manuscript explores the application of computational models to predict response to radionuclide therapy in glioblastoma. There are few, to our knowledge, examples of mathematical models used in clinical radionuclide therapy. We have tested a family of models to determine the applicability of different radiation coupling terms for response to the localized radiation delivery. We show that with patient-specific parameter estimation, we can make accurate predictions of future glioblastoma response to the treatment. As a comparison, we have shown that population trends in response can be used to forecast growth from the moment the treatment has been delivered.In addition to the high simulation and prediction accuracy our modeling methods have achieved, the evaluation of a family of models has given insight into the response dynamics of radionuclide therapy. These dynamics, while different than we had initially hypothesized, should encourage future imaging studies involving high dosage radiation treatments, with specific emphasis on the local immune and vascular response.
Collapse
Affiliation(s)
| | - Chengyue Wu
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - David A. Hormuth
- Livestrong Cancer Institutes, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Shiliang Huang
- Department of Oncology, The University of Texas Health Sciences Center at San Antonio, San Antonio, TX 78229, USA
| | - Ande Bao
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Andrew Brenner
- Department of Oncology, The University of Texas Health Sciences Center at San Antonio, San Antonio, TX 78229, USA
| | - Thomas E. Yankeelov
- Departments of Biomedical Engineering, USA
- Departments of Diagnostic Medicine, USA
- Departments of Oncology, USA
- Livestrong Cancer Institutes, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA
- The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
11
|
Krasnovskaya OO, Abramchuck D, Erofeev A, Gorelkin P, Kuznetsov A, Shemukhin A, Beloglazkina EK. Recent Advances in 64Cu/ 67Cu-Based Radiopharmaceuticals. Int J Mol Sci 2023; 24:9154. [PMID: 37298101 PMCID: PMC10288943 DOI: 10.3390/ijms24119154] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/19/2023] [Accepted: 05/21/2023] [Indexed: 06/12/2023] Open
Abstract
Copper-64 (T1/2 = 12.7 h) is a positron and beta-emitting isotope, with decay characteristics suitable for both positron emission tomography (PET) imaging and radiotherapy of cancer. Copper-67 (T1/2 = 61.8 h) is a beta and gamma emitter, appropriate for radiotherapy β-energy and with a half-life suitable for single-photon emission computed tomography (SPECT) imaging. The chemical identities of 64Cu and 67Cu isotopes allow for convenient use of the same chelating molecules for sequential PET imaging and radiotherapy. A recent breakthrough in 67Cu production opened previously unavailable opportunities for a reliable source of 67Cu with high specific activity and purity. These new opportunities have reignited interest in the use of copper-containing radiopharmaceuticals for the therapy, diagnosis, and theranostics of various diseases. Herein, we summarize recent (2018-2023) advances in the use of copper-based radiopharmaceuticals for PET, SPECT imaging, radiotherapy, and radioimmunotherapy.
Collapse
Affiliation(s)
- Olga O. Krasnovskaya
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia; (D.A.)
| | - Daniil Abramchuck
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia; (D.A.)
| | - Alexander Erofeev
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia; (D.A.)
- Research Laboratory of Biophysics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, 119049 Moscow, Russia
| | - Peter Gorelkin
- Research Laboratory of Biophysics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, 119049 Moscow, Russia
| | - Alexander Kuznetsov
- Skobeltsyn Institute of Nuclear Physics, Lomonosov Moscow State University, Leninskie Gory, 1, Bld. 2, 119991 Moscow, Russia
- Department of Physics, Lomonosov Moscow State University, Leninskie Gory, 1/2, 119991 Moscow, Russia
| | - Andrey Shemukhin
- Skobeltsyn Institute of Nuclear Physics, Lomonosov Moscow State University, Leninskie Gory, 1, Bld. 2, 119991 Moscow, Russia
| | - Elena K. Beloglazkina
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia; (D.A.)
| |
Collapse
|
12
|
Szychot E, Bhagawati D, Sokolska MJ, Walker D, Gill S, Hyare H. Evaluating drug distribution in children and young adults with diffuse midline glioma of the pons (DIPG) treated with convection-enhanced drug delivery. FRONTIERS IN NEUROIMAGING 2023; 2:1062493. [PMID: 37554653 PMCID: PMC10406269 DOI: 10.3389/fnimg.2023.1062493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/08/2023] [Indexed: 08/10/2023]
Abstract
AIMS To determine an imaging protocol that can be used to assess the distribution of infusate in children with DIPG treated with CED. METHODS 13 children diagnosed with DIPG received between 3.8 and 5.7 ml of infusate, through two pairs of catheters to encompass tumor volume on day 1 of cycle one of treatment. Volumetric T2-weighted (T2W) and diffusion-weighted MRI imaging (DWI) were performed before and after day 1 of CED. Apparent diffusion coefficient (ADC) maps were calculated. The tumor volume pre and post CED was automatically segmented on T2W and ADC on the basis of signal intensity. The ADC maps pre and post infusion were aligned and subtracted to visualize the infusate distribution. RESULTS There was a significant increase (p < 0.001) in mean ADC and T2W signal intensity (SI) ratio and a significant (p < 0.001) increase in mean tumor volume defined by ADC and T2W SI post infusion (mean ADC volume pre: 19.8 ml, post: 24.4 ml; mean T2W volume pre: 19.4 ml, post: 23.4 ml). A significant correlation (p < 0.001) between infusate volume and difference in ADC/T2W SI defined tumor volume was observed (ADC, r = 0.76; T2W, r = 0.70). Finally, pixel-by-pixel subtraction of the ADC maps pre and post infusion demonstrated a volume of high signal intensity, presumed infusate distribution. CONCLUSIONS ADC and T2W MRI are proposed as a combined parameter method for evaluation of CED infusate distribution in brainstem tumors in future clinical trials.
Collapse
Affiliation(s)
- Elwira Szychot
- Department of Paediatric Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
- Department of Paediatric Oncology, Harley Street Children's Hospital, London, United Kingdom
- Department of Paediatrics, Paediatric Oncology and Immunology, Pomeranian Medical University, Szczecin, Poland
| | - Dolin Bhagawati
- Department of Paediatric Oncology, Harley Street Children's Hospital, London, United Kingdom
- Department of Neurosurgery, Charing Cross Hospital, Imperial College, London, United Kingdom
| | - Magdalena Joanna Sokolska
- Department of Medical Physics and Biomedical Engineering, Faculty of Engineering Sciences, University College London, London, United Kingdom
| | - David Walker
- Department of Paediatric Oncology, Harley Street Children's Hospital, London, United Kingdom
- Division of Child Health, School of Human Development, University of Nottingham, Nottingham, United Kingdom
| | - Steven Gill
- Department of Paediatric Oncology, Harley Street Children's Hospital, London, United Kingdom
- Department of Translational Health Sciences, Institute of Clinical Neurosciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Harpreet Hyare
- Department of Paediatric Oncology, Harley Street Children's Hospital, London, United Kingdom
- Department of Neuroradiology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
13
|
Georgiou C, Cai Z, Alsaden N, Cho H, Behboudi M, Winnik MA, Rutka JT, Reilly RM. Treatment of Orthotopic U251 Human Glioblastoma Multiforme Tumors in NRG Mice by Convection-Enhanced Delivery of Gold Nanoparticles Labeled with the β-Particle-Emitting Radionuclide, 177Lu. Mol Pharm 2023; 20:582-592. [PMID: 36516432 PMCID: PMC9812026 DOI: 10.1021/acs.molpharmaceut.2c00815] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this study, we investigated convection-enhanced delivery (CED) of 23 ± 3 nm gold nanoparticles (AuNPs) labeled with the β-particle-emitting radionuclide 177Lu (177Lu-AuNPs) for treatment of orthotopic U251-Luc human glioblastoma multiforme (GBM) tumors in NRG mice. The cytotoxicity in vitro of 177Lu-AuNPs (0.0-2.0 MBq, 4 × 1011 AuNPs) on U251-Luc cells was also studied by a clonogenic survival assay, and DNA double-strand breaks (DSBs) caused by β-particle emissions of 177Lu were measured by confocal immunofluorescence microscopy for γH2AX. NRG mice with U251-Luc tumors in the right cerebral hemisphere of the brain were treated by CED of 1.1 ± 0.2 MBq of 177Lu-AuNPs (4 × 1011 AuNPs). Control mice received unlabeled AuNPs or normal saline. Tumor retention of 177Lu-AuNPs was assessed by single-photon emission computed tomography/computed tomography (SPECT/CT) imaging and biodistribution studies. Radiation doses were estimated for the tumor, brain, and other organs. The effectiveness for treating GBM tumors was determined by bioluminescence imaging (BLI) and T2-weighted magnetic resonance imaging (MRI) and by Kaplan-Meier median survival. Normal tissue toxicity was assessed by monitoring body weight and hematology and blood biochemistry analyses at 14 d post-treatment. 177Lu-AuNPs (2.0 MBq, 4 × 1011 AuNPs) decreased the clonogenic survival of U251-Luc cells to 0.005 ± 0.002 and increased DNA DSBs by 14.3-fold compared to cells treated with unlabeled AuNPs or normal saline. A high proportion of 177Lu-AuNPs was retained in the U251-Luc tumor in NRG mice up to 21 d with minimal re-distribution to the brain or other organs. The radiation dose in the tumor was high (599 Gy). The dose in the normal right cerebral hemisphere of the brain excluding the tumor was 93-fold lower (6.4 Gy), and 2000-3000-fold lower doses were calculated for the contralateral left cerebral hemisphere (0.3 Gy) or cerebellum (0.2 Gy). The doses in peripheral organs were <0.1 Gy. BLI revealed almost complete tumor growth arrest in mice treated with 177Lu-AuNPs, while tumors grew rapidly in control mice. MRI at 28 d post-treatment and histological staining showed no visible tumor in mice treated with 177Lu-AuNPs but large GBM tumors in control mice. All control mice reached a humane endpoint requiring sacrifice within 39 d (normal saline) or 45 d post-treatment (unlabeled AuNPs), while 5/8 mice treated with 177Lu-AuNPs survived up to 150 d. No normal tissue toxicity was observed in mice treated with 177Lu-AuNPs. We conclude that CED of 177Lu-AuNPs was highly effective for treating U251-Luc human GBM tumors in the brain in NRG mice at amounts that were non-toxic to normal tissues. These 177Lu-AuNPs administered by CED hold promise for treating patients with GBM to prevent recurrence and improve long-term outcome.
Collapse
Affiliation(s)
- Constantine
J. Georgiou
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, OntarioM5S 3M2, Canada
| | - Zhongli Cai
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, OntarioM5S 3M2, Canada
| | - Noor Alsaden
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, OntarioM5S 3M2, Canada
| | - Hyungjun Cho
- Department
of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
| | - Minou Behboudi
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, OntarioM5S 3M2, Canada
| | - Mitchell A. Winnik
- Department
of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
| | - James T. Rutka
- Division
of Neurosurgery, The Hospital for Sick Children, 555 University Avenue, Toronto, OntarioM5G 1X8, Canada,Division
of Neurosurgery, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 149 College Street, Toronto, OntarioM5T 1P5, Canada
| | - Raymond M. Reilly
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, OntarioM5S 3M2, Canada,Department
of Medical Imaging, Temerty Faculty of Medicine, University of Toronto, Toronto, OntarioM5S 1A8, Canada,Joint Department
of Medical Imaging and Princess Margaret Cancer Centre, University Health Network, Toronto, OntarioM5G 2C1, Canada,
| |
Collapse
|
14
|
Wilcox JA, Li MJ, Boire AA. Leptomeningeal Metastases: New Opportunities in the Modern Era. Neurotherapeutics 2022; 19:1782-1798. [PMID: 35790709 PMCID: PMC9723010 DOI: 10.1007/s13311-022-01261-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2022] [Indexed: 02/07/2023] Open
Abstract
Leptomeningeal metastases arise from cancer cell entry into the subarachnoid space, inflicting significant neurologic morbidity and mortality across a wide range of malignancies. The modern era of cancer therapeutics has seen an explosion of molecular-targeting agents and immune-mediated strategies for patients with breast, lung, and melanoma malignancies, with meaningful extracranial disease control and improvement in patient survival. However, the clinical efficacy of these agents in those with leptomeningeal metastases remains understudied, due to the relative rarity of this patient population, the investigational challenges associated with studying this dynamic disease state, and brisk disease pace. Nevertheless, retrospective studies, post hoc analyses, and small prospective trials in the last two decades provide a glimmer of hope for patients with leptomeningeal metastases, suggesting that several cancer-directed strategies are not only active in the intrathecal space but also improve survival against historical odds. The continued development of clinical trials devoted to patients with leptomeningeal metastases is critical to establish robust efficacy outcomes in this patient population, define drug pharmacokinetics in the intrathecal space, and uncover new avenues for treatment in the face of leptomeningeal therapeutic resistance.
Collapse
Affiliation(s)
- Jessica A Wilcox
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Min Jun Li
- Brain Tumor Center, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adrienne A Boire
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Brain Tumor Center, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
15
|
Nishri Y, Vatarescu M, Luz I, Epstein L, Dumančić M, Del Mare S, Shai A, Schmidt M, Deutsch L, Den RB, Kelson I, Keisari Y, Arazi L, Cooks T, Domankevich V. Diffusing alpha-emitters radiation therapy in combination with temozolomide or bevacizumab in human glioblastoma multiforme xenografts. Front Oncol 2022; 12:888100. [PMID: 36237307 PMCID: PMC9552201 DOI: 10.3389/fonc.2022.888100] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is at present an incurable disease with a 5-year survival rate of 5.5%, despite improvements in treatment modalities such as surgery, radiation therapy, chemotherapy [e.g., temozolomide (TMZ)], and targeted therapy [e.g., the antiangiogenic agent bevacizumab (BEV)]. Diffusing alpha-emitters radiation therapy (DaRT) is a new modality that employs radium-224-loaded seeds that disperse alpha-emitting atoms inside the tumor. This treatment was shown to be effective in mice bearing human-derived GBM tumors. Here, the effect of DaRT in combination with standard-of-care therapies such as TMZ or BEV was investigated. In a viability assay, the combination of alpha radiation with TMZ doubled the cytotoxic effect of each of the treatments alone in U87 cultured cells. A colony formation assay demonstrated that the surviving fraction of U87 cells treated by TMZ in combination with alpha irradiation was lower than was achieved by alpha- or x-ray irradiation as monotherapies, or by x-ray combined with TMZ. The treatment of U87-bearing mice with DaRT and TMZ delayed tumor development more than the monotherapies. Unlike other radiation types, alpha radiation did not increase VEGF secretion from U87 cells in culture. BEV treatment introduced several days after DaRT implantation improved tumor control, compared to BEV or DaRT as monotherapies. The combination was also shown to be superior when starting BEV administration prior to DaRT implantation in large tumors relative to the seed size. BEV induced a decrease in CD31 staining under DaRT treatment, increased the diffusive spread of 224Ra progeny atoms in the tumor tissue, and decreased their clearance from the tumor through the blood. Taken together, the combinations of DaRT with standard-of-care chemotherapy or antiangiogenic therapy are promising approaches, which may improve the treatment of GBM patients.
Collapse
Affiliation(s)
- Yossi Nishri
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
| | - Maayan Vatarescu
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Ishai Luz
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Lior Epstein
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Radiation Protection Department, Soreq Nuclear Research Center, Yavne, Israel
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Mirta Dumančić
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sara Del Mare
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
| | - Amit Shai
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
| | | | - Lisa Deutsch
- Biostatistics Department, BioStats Statistical Consulting Ltd., Maccabim, Israel
| | - Robert B. Den
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
- Department of Radiation Oncology, Urology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Itzhak Kelson
- School of Physics and Astronomy, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yona Keisari
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lior Arazi
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- *Correspondence: Lior Arazi, ; Tomer Cooks, ; Vered Domankevich,
| | - Tomer Cooks
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
- *Correspondence: Lior Arazi, ; Tomer Cooks, ; Vered Domankevich,
| | - Vered Domankevich
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
- *Correspondence: Lior Arazi, ; Tomer Cooks, ; Vered Domankevich,
| |
Collapse
|
16
|
Uccelli L, Martini P, Urso L, Ghirardi T, Marvelli L, Cittanti C, Carnevale A, Giganti M, Bartolomei M, Boschi A. Rhenium Radioisotopes for Medicine, a Focus on Production and Applications. Molecules 2022; 27:5283. [PMID: 36014521 PMCID: PMC9412410 DOI: 10.3390/molecules27165283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/08/2022] [Accepted: 08/17/2022] [Indexed: 11/28/2022] Open
Abstract
In recent decades, the use of alpha; pure beta; or beta/gamma emitters in oncology, endocrinology, and interventional cardiology rheumatology, has proved to be an important alternative to the most common therapeutic regimens. Among radionuclides used for therapy in nuclear medicine, two rhenium radioisotopes are of particular relevance: rhenium-186 and rhenium-188. The first is routinely produced in nuclear reactors by direct neutron activation of rhenium-186 via 185Re(n,γ)186Re nuclear reaction. Rhenium-188 is produced by the decay of the parent tungsten-188. Separation of rhenium-188 is mainly performed using a chromatographic 188W/188Re generator in which tungsten-188 is adsorbed on the alumina column, similar to the 99Mo/99mTc generator system, and the radionuclide eluted in saline solution. The application of rhenium-186 and rhenium-188 depends on their specific activity. Rhenium-186 is produced in low specific activity and is mainly used for labeling particles or diphosphonates for bone pain palliation. Whereas, rhenium-188 of high specific activity can be used for labeling peptides or bioactive molecules. One of the advantages of rhenium is its chemical similarity with technetium. So, diagnostic technetium analogs labeled with radiorhenium can be developed for therapeutic applications. Clinical trials promoting the use of 186/188Re-radiopharmaceuticals is, in particular, are discussed.
Collapse
Affiliation(s)
- Licia Uccelli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Petra Martini
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Luca Urso
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Teresa Ghirardi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Lorenza Marvelli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Aldo Carnevale
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Melchiore Giganti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Alessandra Boschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
17
|
Schulz JA, Rodgers LT, Kryscio RJ, Hartz AMS, Bauer B. Characterization and comparison of human glioblastoma models. BMC Cancer 2022; 22:844. [PMID: 35922758 PMCID: PMC9347152 DOI: 10.1186/s12885-022-09910-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
Glioblastoma (GBM) is one of the deadliest cancers. Treatment options are limited, and median patient survival is only several months. Translation of new therapies is hindered by a lack of GBM models that fully recapitulate disease heterogeneity. Here, we characterize two human GBM models (U87-luc2, U251-RedFLuc). In vitro, both cell lines express similar levels of luciferase and show comparable sensitivity to temozolomide and lapatinib exposure. In vivo, however, the two GBM models recapitulate different aspects of the disease. U87-luc2 cells quickly grow into large, well-demarcated tumors; U251-RedFLuc cells form small, highly invasive tumors. Using a new method to assess GBM invasiveness based on detecting tumor-specific anti-luciferase staining in brain slices, we found that U251-RedFLuc cells are more invasive than U87-luc2 cells. Lastly, we determined expression levels of ABC transporters in both models. Our findings indicate that U87-luc2 and U251-RedFLuc GBM models recapitulate different aspects of GBM heterogeneity that need to be considered in preclinical research.
Collapse
Affiliation(s)
- Julia A Schulz
- Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington, KY, USA
| | - Louis T Rodgers
- Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington, KY, USA
| | - Richard J Kryscio
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, USA
- Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, USA
- Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, USA
| | - Björn Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington, KY, USA.
- Drug Discovery, Delivery and Translational Therapeutics Track, Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, USA.
| |
Collapse
|
18
|
Hersh AM, Alomari S, Tyler BM. Crossing the Blood-Brain Barrier: Advances in Nanoparticle Technology for Drug Delivery in Neuro-Oncology. Int J Mol Sci 2022; 23:4153. [PMID: 35456971 PMCID: PMC9032478 DOI: 10.3390/ijms23084153] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 12/10/2022] Open
Abstract
The blood-brain barrier (BBB) constitutes a microvascular network responsible for excluding most drugs from the brain. Treatment of brain tumors is limited by the impermeability of the BBB and, consequently, survival outcomes for malignant brain tumors remain poor. Nanoparticles (NPs) represent a potential solution to improve drug transport to brain tumors, given their small size and capacity to target tumor cells. Here, we review the unique physical and chemical properties of NPs that aid in BBB transport and discuss mechanisms of NP transport across the BBB, including paracellular transport, carrier-mediated transport, and adsorptive- and receptor-mediated transcytosis. The major types of NPs investigated for treatment of brain tumors are detailed, including polymeric NPs, liposomes, solid lipid NPs, dendrimers, metals, quantum dots, and nanogels. In addition to their role in drug delivery, NPs can be used as imaging contrast agents and can be conjugated with imaging probes to assist in visualizing tumors, demarcating lesion boundaries and margins, and monitoring drug delivery and treatment response. Multifunctional NPs can be designed that are capable of targeting tumors for both imaging and therapeutic purposes. Finally, limitations of NPs for brain tumor treatment are discussed.
Collapse
Affiliation(s)
| | | | - Betty M. Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (S.A.)
| |
Collapse
|
19
|
Advances in local therapy for glioblastoma - taking the fight to the tumour. Nat Rev Neurol 2022; 18:221-236. [PMID: 35277681 PMCID: PMC10359969 DOI: 10.1038/s41582-022-00621-0] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2022] [Indexed: 12/21/2022]
Abstract
Despite advances in neurosurgery, chemotherapy and radiotherapy, glioblastoma remains one of the most treatment-resistant CNS malignancies, and the tumour inevitably recurs. The majority of recurrences appear in or near the resection cavity, usually within the area that received the highest dose of radiation. Many new therapies focus on combatting these local recurrences by implementing treatments directly in or near the tumour bed. In this Review, we discuss the latest developments in local therapy for glioblastoma, focusing on recent preclinical and clinical trials. The approaches that we discuss include novel intraoperative techniques, various treatments of the surgical cavity, stereotactic injections directly into the tumour, and new developments in convection-enhanced delivery and intra-arterial treatments.
Collapse
|
20
|
Melis DR, Burgoyne AR, Ooms M, Gasser G. Bifunctional chelators for radiorhenium: past, present and future outlook. RSC Med Chem 2022; 13:217-245. [PMID: 35434629 PMCID: PMC8942221 DOI: 10.1039/d1md00364j] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/14/2022] [Indexed: 01/16/2023] Open
Abstract
Targeted radionuclide therapy (TRNT) is an ever-expanding field of nuclear medicine that provides a personalised approach to cancer treatment while limiting toxicity to normal tissues. It involves the radiolabelling of a biological targeting vector with an appropriate therapeutic radionuclide, often facilitated by the use of a bifunctional chelator (BFC) to stably link the two entities. The radioisotopes of rhenium, 186Re (t 1/2 = 90 h, 1.07 MeV β-, 137 keV γ (9%)) and 188Re (t 1/2 = 16.9 h, 2.12 MeV β-, 155 keV γ (15%)), are particularly attractive for radiotherapy because of their convenient and high-abundance β--particle emissions as well as their imageable γ-emissions and chemical similarity to technetium. As a transition metal element with multiple oxidation states and coordination numbers accessible for complexation, there is great opportunity available when it comes to developing novel BFCs for rhenium. The purpose of this review is to provide a recap on some of the past successes and failings, as well as show some more current efforts in the design of BFCs for 186/188Re. Future use of these radionuclides for radiotherapy depends on their cost-effective availability and this will also be discussed. Finally, bioconjugation strategies for radiolabelling biomolecules with 186/188Re will be touched upon.
Collapse
Affiliation(s)
- Diana R Melis
- SCK CEN, Belgian Nuclear Research Centre Boeretang 200 BE-2400 Mol Belgium +1 865 341 1413 +32 14 33 32 83
- Chimie ParisTech, Laboratory for Inorganic Chemical Biology, PSL University F-75005 Paris France www.gassergroup.com +33 1 44 27 56 02
| | - Andrew R Burgoyne
- SCK CEN, Belgian Nuclear Research Centre Boeretang 200 BE-2400 Mol Belgium +1 865 341 1413 +32 14 33 32 83
| | - Maarten Ooms
- SCK CEN, Belgian Nuclear Research Centre Boeretang 200 BE-2400 Mol Belgium +1 865 341 1413 +32 14 33 32 83
| | - Gilles Gasser
- Chimie ParisTech, Laboratory for Inorganic Chemical Biology, PSL University F-75005 Paris France www.gassergroup.com +33 1 44 27 56 02
| |
Collapse
|
21
|
Kaynak A, Davis HW, Vallabhapurapu SD, Pak KY, Gray BD, Qi X. SapC-DOPS as a Novel Therapeutic and Diagnostic Agent for Glioblastoma Therapy and Detection: Alternative to Old Drugs and Agents. Pharmaceuticals (Basel) 2021; 14:1193. [PMID: 34832975 PMCID: PMC8619974 DOI: 10.3390/ph14111193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma multiforme (GBM), the most common type of brain cancer, is extremely aggressive and has a dreadful prognosis. GBM comprises 60% of adult brain tumors and the 5 year survival rate of GBM patients is only 4.3%. Standard-of-care treatment includes maximal surgical removal of the tumor in combination with radiation and temozolomide (TMZ) chemotherapy. TMZ is the "gold-standard" chemotherapy for patients suffering from GBM. However, the median survival is only about 12 to 18 months with this protocol. Consequently, there is a critical need to develop new therapeutic options for treatment of GBM. Nanomaterials have unique properties as multifunctional platforms for brain tumor therapy and diagnosis. As one of the nanomaterials, lipid-based nanocarriers are capable of delivering chemotherapeutics and imaging agents to tumor sites by enhancing the permeability of the compound through the blood-brain barrier, which makes them ideal for GBM therapy and imaging. Nanocarriers also can be used for delivery of radiosensitizers to the tumor to enhance the efficacy of the radiation therapy. Previously, high-atomic-number element-containing particles such as gold nanoparticles and liposomes have been used as radiosensitizers. SapC-DOPS, a protein-based liposomal drug comprising the lipid, dioleoylphosphatidylserine (DOPS), and the protein, saposin C (SapC), has been shown to be effective for treatment of a variety of cancers in small animals, including GBM. SapC-DOPS also has the unique ability to be used as a carrier for delivery of radiotheranostic agents for nuclear imaging and radiotherapeutic purposes. These unique properties make tumor-targeting proteo-liposome nanocarriers novel therapeutic and diagnostic alternatives to traditional chemotherapeutics and imaging agents. This article reviews various treatment modalities including nanolipid-based delivery and therapeutic systems used in preclinical and clinical trial settings for GBM treatment and detection.
Collapse
Affiliation(s)
- Ahmet Kaynak
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, and Brain Tumor Center at UC Neuroscience Institute, 3512 Eden Avenue, Cincinnati, OH 45267, USA; (A.K.); (H.W.D.); (S.D.V.)
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Harold W. Davis
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, and Brain Tumor Center at UC Neuroscience Institute, 3512 Eden Avenue, Cincinnati, OH 45267, USA; (A.K.); (H.W.D.); (S.D.V.)
| | - Subrahmanya D. Vallabhapurapu
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, and Brain Tumor Center at UC Neuroscience Institute, 3512 Eden Avenue, Cincinnati, OH 45267, USA; (A.K.); (H.W.D.); (S.D.V.)
| | - Koon Y. Pak
- Molecular Targeting Technologies, Inc., West Chester, PA 19380, USA; (K.Y.P.); (B.D.G.)
| | - Brian D. Gray
- Molecular Targeting Technologies, Inc., West Chester, PA 19380, USA; (K.Y.P.); (B.D.G.)
| | - Xiaoyang Qi
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, and Brain Tumor Center at UC Neuroscience Institute, 3512 Eden Avenue, Cincinnati, OH 45267, USA; (A.K.); (H.W.D.); (S.D.V.)
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
22
|
Shalgunov V, Engudar G, Bohrmann L, Wharton L, Maskell K, Johann K, Barz M, Schaffer P, Herth MM, Radchenko V. Radiolabeling of a polypeptide polymer for intratumoral delivery of alpha-particle emitter, 225Ac, and beta-particle emitter, 177Lu. Nucl Med Biol 2021; 104-105:11-21. [PMID: 34839209 DOI: 10.1016/j.nucmedbio.2021.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/22/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Radiotherapy of cancer requires both alpha- and beta-particle emitting radionuclides, as these radionuclide types are efficient at destroying different types of tumors. Both classes of radionuclides require a vehicle, such as an antibody or a polymer, to be delivered and retained within the tumor. Polyglutamic acid (pGlu) is a polymer that has proven itself effective as a basis of drug-polymer conjugates in the clinic, while its derivatives have been used for pretargeted tumor imaging in a research setup. trans-Cyclooctene (TCO) modified pGlu is suitable for pretargeted imaging or therapy, as well as for intratumoral radionuclide therapy. In all cases, it becomes indirectly radiolabeled via the bioorthogonal click reaction with the tetrazine (Tz) molecule carrying the radionuclide. In this study, we report the radiolabeling of TCO-modified pGlu with either lutetium-177 (177Lu), a beta-particle emitter, or actinium-225 (225Ac), an alpha-particle emitter, using the click reaction between TCO and Tz. METHODS A panel of Tz derivatives containing a metal ion binding chelator (DOTA or macropa) connected to the Tz moiety directly or through a polyethylene glycol (PEG) linker was synthesized and tested for their ability to chelate 177Lu and 225Ac, and click to pGlu-TCO. Radiolabeled 177Lu-pGlu and 225Ac-pGlu were isolated by size exclusion chromatography. The retention of 177Lu or 225Ac by the obtained conjugates was investigated in vitro in human serum. RESULTS All DOTA-modified Tzs efficiently chelated 177Lu resulting in average radiochemical conversions (RCC) of >75%. Isolated radiochemical yields (RCY) for 177Lu-pGlu prepared from 177Lu-Tzs ranged from 31% to 55%. TLC analyses detected <5% unchelated 177Lu for all 177Lu-pGlu preparations over six days in human serum. For 225Ac chelation, optimized RCCs ranged from 61 ± 34% to quantitative for DOTA-Tzs and were quantitative for the macropa-modified Tz (>98%). Isolated radiochemical yields (RCY) for 225Ac-pGlu prepared from 225Ac-Tzs ranged from 28% to 51%. For 3 out of 5 225Ac-pGlu conjugates prepared from DOTA-Tzs, the amount of unchelated 225Ac stayed below 10% over six days in human serum, while 225Ac-pGlu prepared from macropa-Tz showed a steady release of up to 37% 225Ac. CONCLUSION We labeled TCO-modified pGlu polymers with alpha- and beta-emitting radionuclides in acceptable RCYs. All 177Lu-pGlu preparations and some 225Ac-pGlu preparations showed excellent stability in human plasma. Our work shows the potential of pGlu as a vehicle for alpha- and beta-radiotherapy of tumors and demonstrated the usefulness of Tz ligation for indirect radiolabeling.
Collapse
Affiliation(s)
- Vladimir Shalgunov
- Department for Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Gokce Engudar
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | - Lennart Bohrmann
- Department for Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Luke Wharton
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada; Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z, Canada
| | - Keiran Maskell
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada; Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 0A7, Canada
| | - Kerstin Johann
- Department of Chemistry, Johannes Gutenberg University, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Matthias Barz
- Department of Chemistry, Johannes Gutenberg University, Duesbergweg 10-14, 55128 Mainz, Germany; Division of Biotherapeutics, Leiden Academic Center for Drug Research (LACDR), Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada; Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 0A7, Canada; Department of Radiology, University of British Columbia, 2775 Lauret St, Vancouver, BC V5Z 1M9, Canada
| | - Matthias M Herth
- Department for Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada; Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z, Canada.
| |
Collapse
|
23
|
Woodall RT, Hormuth Ii DA, Wu C, Abdelmalik MRA, Phillips WT, Bao A, Hughes TJR, Brenner AJ, Yankeelov TE. Patient specific, imaging-informed modeling of rhenium-186 nanoliposome delivery via convection-enhanced delivery in glioblastoma multiforme. Biomed Phys Eng Express 2021; 7. [PMID: 34050041 DOI: 10.1088/2057-1976/ac02a6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/18/2021] [Indexed: 12/25/2022]
Abstract
Convection-enhanced delivery of rhenium-186 (186Re)-nanoliposomes is a promising approach to provide precise delivery of large localized doses of radiation for patients with recurrent glioblastoma multiforme. Current approaches for treatment planning utilizing convection-enhanced delivery are designed for small molecule drugs and not for larger particles such as186Re-nanoliposomes. To enable the treatment planning for186Re-nanoliposomes delivery, we have developed a computational fluid dynamics approach to predict the distribution of nanoliposomes for individual patients. In this work, we construct, calibrate, and validate a family of computational fluid dynamics models to predict the spatio-temporal distribution of186Re-nanoliposomes within the brain, utilizing patient-specific pre-operative magnetic resonance imaging (MRI) to assign material properties for an advection-diffusion transport model. The model family is calibrated to single photon emission computed tomography (SPECT) images acquired during and after the infusion of186Re-nanoliposomes for five patients enrolled in a Phase I/II trial (NCT Number NCT01906385), and is validated using a leave-one-out bootstrapping methodology for predicting the final distribution of the particles. After calibration, our models are capable of predicting the mid-delivery and final spatial distribution of186Re-nanoliposomes with a Dice value of 0.69 ± 0.18 and a concordance correlation coefficient of 0.88 ± 0.12 (mean ± 95% confidence interval), using only the patient-specific, pre-operative MRI data, and calibrated model parameters from prior patients. These results demonstrate a proof-of-concept for a patient-specific modeling framework, which predicts the spatial distribution of nanoparticles. Further development of this approach could enable optimizing catheter placement for future studies employing convection-enhanced delivery.
Collapse
Affiliation(s)
- Ryan T Woodall
- Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - David A Hormuth Ii
- Oden Institute for Computational Engineering and Sciences,The University of Texas at Austin, Austin, Texas, United States of America.,Oncology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Chengyue Wu
- Oden Institute for Computational Engineering and Sciences,The University of Texas at Austin, Austin, Texas, United States of America
| | - Michael R A Abdelmalik
- Oden Institute for Computational Engineering and Sciences,The University of Texas at Austin, Austin, Texas, United States of America.,Mechanical Engineering, Eindhoven University of Technology, The Netherlands
| | - William T Phillips
- Departments of Radiology at UT Health San Antonio, San Antonio, Texas, United States of America
| | - Ande Bao
- Department of Radiation Oncology, Seidman Cancer Center, University Hospitals, Cleveland Medical Center, Cleveland, Ohio, United States of America.,School of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Thomas J R Hughes
- Oden Institute for Computational Engineering and Sciences,The University of Texas at Austin, Austin, Texas, United States of America.,Aerospace Engineering and Engineering Mechanics, The University of Texas at Austin, Austin, Texas, United States of America
| | - Andrew J Brenner
- Mays Cancer Center at UT Health San Antonio, San Antonio, Texas, United States of America
| | - Thomas E Yankeelov
- Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America.,Oden Institute for Computational Engineering and Sciences,The University of Texas at Austin, Austin, Texas, United States of America.,Diagnostic Medicine, The University of Texas at Austin, Austin, Texas, United States of America.,Oncology, The University of Texas at Austin, Austin, Texas, United States of America.,Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas, United States of America.,Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
24
|
Fach M, Fliedner FP, Kempen PJ, Melander F, Hansen AE, Bruun LM, Köster U, Sporer E, Kjær A, Andresen TL, Jensen AI, Henriksen JR. Effective Intratumoral Retention of [ 103 Pd]AuPd Alloy Nanoparticles Embedded in Gel-Forming Liquids Paves the Way for New Nanobrachytherapy. Adv Healthc Mater 2021; 10:e2002009. [PMID: 33763995 DOI: 10.1002/adhm.202002009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/10/2021] [Indexed: 12/31/2022]
Abstract
Local application of radioactive sources as brachytherapy is well established in oncology. This treatment is highly invasive however, due to the insertion of millimeter sized metal seeds. The authors report the development of a new concept for brachytherapy, based on gold-palladium (AuPd) alloy nanoparticles, intrinsically radiolabeled with 103 Pd. These are formulated in a carbohydrate-ester based liquid, capable of forming biodegradable gel-like implants upon injection. This allows for less invasive administration through small-gauge needles. [103 Pd]AuPd nanoparticles with sizes around 20 nm are prepared with radiolabeling efficiencies ranging from 79% to >99%. Coating with the hydrophobic polymer poly(N-isopropylacrylamide) leads to nanoparticle diameters below 40 nm. Dispersing the nanoparticles in ethanol with water insoluble carbohydrate esters gives "nanogels", a low viscosity liquid capable of solidifying upon injection into aqueous environments. Both nanoparticles and radioactivity are stably retained in the nanogel over 25 days (>99%) after formation in aqueous buffers. Animals bearing CT26 murine tumors are injected intratumorally with 25 MBq of the 103 Pd-nanogel, and display tumor growth delay and significantly increase median survival times compared with control groups. Excellent retention in the tumor of both the 103 Pd and the nanoparticle matrix itself is observed, demonstrating a potential for replacing currently used brachytherapy seeds.
Collapse
Affiliation(s)
- Matthias Fach
- DTU Health Technology Center for Nanomedicine and Theranostics Technical University of Denmark Ørsteds Plads 345C Lyngby 2800 Denmark
| | - Frederikke P. Fliedner
- Department of Clinical Physiology Nuclear Medicine & PET and Cluster for Molecular Imaging Department of Biomedical Sciences Rigshospitalet and University of Copenhagen Blegdamsvej 3B Copenhagen 2100 Denmark
| | - Paul J. Kempen
- DTU Health Technology Center for Nanomedicine and Theranostics Technical University of Denmark Ørsteds Plads 345C Lyngby 2800 Denmark
| | - Fredrik Melander
- DTU Health Technology Center for Nanomedicine and Theranostics Technical University of Denmark Ørsteds Plads 345C Lyngby 2800 Denmark
| | - Anders E. Hansen
- DTU Health Technology Center for Nanomedicine and Theranostics Technical University of Denmark Ørsteds Plads 345C Lyngby 2800 Denmark
- Department of Clinical Physiology Nuclear Medicine & PET and Cluster for Molecular Imaging Department of Biomedical Sciences Rigshospitalet and University of Copenhagen Blegdamsvej 3B Copenhagen 2100 Denmark
| | - Linda M. Bruun
- DTU Health Technology Center for Nanomedicine and Theranostics Technical University of Denmark Ørsteds Plads 345C Lyngby 2800 Denmark
| | - Ulli Köster
- Institut Laue‐Langevin 71 Avenue des Martyrs Grenoble 38042 France
| | - Emanuel Sporer
- The Hevesy Laboratory DTU Health Technology Center for Nanomedicine and Theranostics Technical University of Denmark (DTU) Frederiksborgvej 399 Roskilde 4000 Denmark
| | - Andreas Kjær
- Department of Clinical Physiology Nuclear Medicine & PET and Cluster for Molecular Imaging Department of Biomedical Sciences Rigshospitalet and University of Copenhagen Blegdamsvej 3B Copenhagen 2100 Denmark
| | - Thomas L. Andresen
- DTU Health Technology Center for Nanomedicine and Theranostics Technical University of Denmark Ørsteds Plads 345C Lyngby 2800 Denmark
| | - Andreas I. Jensen
- The Hevesy Laboratory DTU Health Technology Center for Nanomedicine and Theranostics Technical University of Denmark (DTU) Frederiksborgvej 399 Roskilde 4000 Denmark
| | - Jonas R. Henriksen
- DTU Health Technology Center for Nanomedicine and Theranostics Technical University of Denmark Ørsteds Plads 345C Lyngby 2800 Denmark
| |
Collapse
|
25
|
Yang S, Han G, Chen Q, Yu L, Wang P, Zhang Q, Dong J, Zhang W, Huang J. Au-Pt Nanoparticle Formulation as a Radiosensitizer for Radiotherapy with Dual Effects. Int J Nanomedicine 2021; 16:239-248. [PMID: 33469284 PMCID: PMC7811476 DOI: 10.2147/ijn.s287523] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Background Radiotherapy occupies an essential position as one of the most significant approaches for the clinical treatment of cancer. However, we cannot overcome the shortcoming of X-rays which is the high value of the oxygen enhancement ratio (OER). Radiosensitizers with the ability to enhance the radiosensitivity of tumor cells provide an alternative to changing X-rays to protons and heavy ion radiotherapy. Materials and Methods We prepared the Au-Pt nanoparticles (Au-Pt NPs) using a one-step method. The characteristics of the Au-Pt NPs were determined using TEM, HAADF-STEM, elemental mapping images, and DLS. The enhanced radiotherapy was demonstrated in vitro using MTT assays, colony formation assays, fluorescence imaging, and flow cytometric analyses of the apoptosis. The biodistribution of the Au-Pt NPs was analyzed using ICP-OES, and thermal images. The enhanced radiotherapy was demonstrated in vitro using immunofluorescence images, tumor volume and weigh, and hematoxylin & eosin (H&E) staining. Results Polyethylene glycol (PEG) functionalized nanoparticles composed of the metallic elements Au and Pt were designed to increase synergistic radiosensitivity. The mechanism demonstrated that heavy metal NPs possess a high X-ray photon capture cross-section and Compton scattering effect which increased DNA damage. Furthermore, the Au-Pt NPs exhibited enzyme-mimicking activities by catalyzing the decomposition of endogenous H2O2 to O2 in the solid tumor microenvironment (TME). Conclusion Our work provides a systematically administered radiosensitizer that can selectively reside in a tumor via the EPR effect and enhances the efficiency of treating cancer with radiotherapy.
Collapse
Affiliation(s)
- Song Yang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Gaohua Han
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Quan Chen
- Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China.,Department of Thoracic Surgery, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China
| | - Lei Yu
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Peng Wang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Qi Zhang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jiang Dong
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Wei Zhang
- Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China.,Department of Infectious Disease, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China
| | - Junxing Huang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
26
|
Anderson AR, Segura T. Injectable biomaterials for treatment of glioblastoma. ADVANCED MATERIALS INTERFACES 2020; 7:2001055. [PMID: 34660174 PMCID: PMC8513688 DOI: 10.1002/admi.202001055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Indexed: 06/13/2023]
Abstract
Despite ongoing advancements in the field of medicine, glioblastoma multiforme (GBM) is presently incurable, making this advanced brain tumor the deadliest tumor type in the central nervous system. The primary treatment strategies for GBM (i.e. surgical resection, radiation therapy, chemotherapy, and newly incorporated targeted therapies) fail to overcome the challenging characteristics of highly aggressive GBM tumors and are presently given with the goal of increasing the quality of life for patients. With the aim of creating effective treatment solutions, research has shifted toward utilizing injectable biomaterial adjuncts to minimize invasiveness of treatment, provide spatiotemporal control of therapeutic delivery, and engage with cells through material-cell interfaces. This review aims to summarize the limitations of the current standard of care for GBM, discuss how these limitations can be addressed by local employment of injectable biomaterial systems, and highlight developments in the field of biomaterials for these applications.
Collapse
Affiliation(s)
- Alexa R. Anderson
- Duke University Department of Biomedical Engineering, 101 Science Drive, Durham, NC 27708, U.S.A
| | - Tatiana Segura
- Duke University Department of Biomedical Engineering, 101 Science Drive, Durham, NC 27708, U.S.A
| |
Collapse
|
27
|
Liew HS, Mai CW, Zulkefeli M, Madheswaran T, Kiew LV, Delsuc N, Low ML. Recent Emergence of Rhenium(I) Tricarbonyl Complexes as Photosensitisers for Cancer Therapy. Molecules 2020; 25:4176. [PMID: 32932573 PMCID: PMC7571230 DOI: 10.3390/molecules25184176] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/05/2023] Open
Abstract
Photodynamic therapy (PDT) is emerging as a significant complementary or alternative approach for cancer treatment. PDT drugs act as photosensitisers, which upon using appropriate wavelength light and in the presence of molecular oxygen, can lead to cell death. Herein, we reviewed the general characteristics of the different generation of photosensitisers. We also outlined the emergence of rhenium (Re) and more specifically, Re(I) tricarbonyl complexes as a new generation of metal-based photosensitisers for photodynamic therapy that are of great interest in multidisciplinary research. The photophysical properties and structures of Re(I) complexes discussed in this review are summarised to determine basic features and similarities among the structures that are important for their phototoxic activity and future investigations. We further examined the in vitro and in vivo efficacies of the Re(I) complexes that have been synthesised for anticancer purposes. We also discussed Re(I) complexes in conjunction with the advancement of two-photon PDT, drug combination study, nanomedicine, and photothermal therapy to overcome the limitation of such complexes, which generally absorb short wavelengths.
Collapse
Affiliation(s)
- Hui Shan Liew
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
| | - Chun-Wai Mai
- Centre for Cancer and Stem Cell Research, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| | - Mohd Zulkefeli
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| | - Thiagarajan Madheswaran
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| | - Lik Voon Kiew
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Nicolas Delsuc
- Laboratoire des Biomolécules, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, 75005 Paris, France;
| | - May Lee Low
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| |
Collapse
|
28
|
Convection Enhanced Delivery for Diffuse Intrinsic Pontine Glioma: Review of a Single Institution Experience. Pharmaceutics 2020; 12:pharmaceutics12070660. [PMID: 32674336 PMCID: PMC7407112 DOI: 10.3390/pharmaceutics12070660] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 01/24/2023] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are a pontine subtype of diffuse midline gliomas (DMGs), primary central nervous system (CNS) tumors of childhood that carry a terrible prognosis. Because of the highly infiltrative growth pattern and the anatomical position, cytoreductive surgery is not an option. An initial response to radiation therapy is invariably followed by recurrence; mortality occurs approximately 11 months after diagnosis. The development of novel therapeutics with great preclinical promise has been hindered by the tightly regulated blood-brain barrier (BBB), which segregates the tumor comportment from the systemic circulation. One possible solution to this obstacle is the use of convection enhanced delivery (CED), a local delivery strategy that bypasses the BBB by direct infusion into the tumor through a small caliber cannula. We have recently shown CED to be safe in children with DIPG (NCT01502917). In this review, we discuss our experience with CED, its advantages, and technical advancements that are occurring in the field. We also highlight hurdles that will likely need to be overcome in demonstrating clinical benefit with this therapeutic strategy.
Collapse
|
29
|
Xu Y, Wei L, Wang H. Progress and perspectives on nanoplatforms for drug delivery to the brain. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101636] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
30
|
Rice KM, Ginjupalli GK, Manne NDPK, Jones CB, Blough ER. A review of the antimicrobial potential of precious metal derived nanoparticle constructs. NANOTECHNOLOGY 2019; 30:372001. [PMID: 30840941 DOI: 10.1088/1361-6528/ab0d38] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The field of nanotechnology is rapidly growing. The promise of pharmacotherapeutics emerging from this vast field has drawn the attention of many researchers. However, with the increase in the prevalence of antibiotic resistant microorganisms, the manifestations of these promises are needed now more than ever. Many have postulated the antimicrobial potential of nanoparticle constructs derived from precious metals/noble metals nanoparticles (NMNPs), such as silver nanoparticles that show activity against multidrug resistant bacteria. In this review we will evaluate the current studies and explore the data to obtain a clear picture of the potential of these particles and the validity of the claims of drug resistant treatments with NMNPs.
Collapse
Affiliation(s)
- Kevin M Rice
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, United States of America. Department of Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States of America. Biotechnology Graduate Program West Virginia State University, Institute, WV, United States of America. Department of Health and Human Service, School of Kinesiology, Marshall University, Huntington, WV, United States of America
| | | | | | | | | |
Collapse
|
31
|
Haase AA, Bauer EB, Kühn FE, Crans DC. Speciation and toxicity of rhenium salts, organometallics and coordination complexes. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
32
|
Bauer EB, Haase AA, Reich RM, Crans DC, Kühn FE. Organometallic and coordination rhenium compounds and their potential in cancer therapy. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.04.014] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
33
|
Bailly C, Vidal A, Bonnemaire C, Kraeber-Bodéré F, Chérel M, Pallardy A, Rousseau C, Garcion E, Lacoeuille F, Hindré F, Valable S, Bernaudin M, Bodet-Milin C, Bourgeois M. Potential for Nuclear Medicine Therapy for Glioblastoma Treatment. Front Pharmacol 2019; 10:772. [PMID: 31354487 PMCID: PMC6637301 DOI: 10.3389/fphar.2019.00772] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/14/2019] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma is the most common malignant adult brain tumor and has a very poor patient prognosis. The mean survival for highly proliferative glioblastoma is only 10 to 14 months despite an aggressive current therapeutic approach known as Stupp's protocol, which consists of debulking surgery followed by radiotherapy and chemotherapy. Despite several clinical trials using anti-angiogenic targeted therapies, glioblastoma medical care remains without major progress in the last decade. Recent progress in nuclear medicine, has been mainly driven by advances in biotechnologies such as radioimmunotherapy, radiopeptide therapy, and radionanoparticles, and these bring a new promising arsenal for glioblastoma therapy. For therapeutic purposes, nuclear medicine practitioners classically use β- particle emitters like 131I, 90Y, 186/188Re, or 177Lu. In the glioblastoma field, these radioisotopes are coupled with nanoparticles, monoclonal antibodies, or peptides. These radiopharmaceutical compounds have resulted in a stabilization and/or improvement of the neurological status with only transient side effects. In nuclear medicine, the glioblastoma-localized and targeted internal radiotherapy proof-of-concept stage has been successfully demonstrated using β- emitting isotopes. Similarly, α particle emitters like 213Bi, 211At, or 225Ac appear to be an innovative and interesting alternative. Indeed, α particles deliver a high proportion of their energy inside or at close proximity to the targeted cells (within a few micrometers from the emission point versus several millimeters for β- particles). This physical property is based on particle-matter interaction differences and results in α particles being highly efficient in killing tumor cells with minimal irradiation of healthy tissues and permits targeting of isolated tumor cells. The first clinical trials confirmed this idea and showed good therapeutic efficacy and less side effects, thus opening a new and promising era for glioblastoma medical care using α therapy. The objective of this literature review is focused on the developing field of nuclear medicine and aims to describe the various parameters such as targets, vectors, isotopes, or injection route (systemic and local) in relation to the clinical and preclinical results in glioblastoma pathology.
Collapse
Affiliation(s)
- Clément Bailly
- Nuclear Medicine, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | | | | | - Françoise Kraeber-Bodéré
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medecine, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France
| | - Michel Chérel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Institut de Cancérologie de l'Ouest (ICO), Angers, France
| | - Amandine Pallardy
- Nuclear Medicine, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France
| | | | - Emmanuel Garcion
- Team 17-Design and Application of Innovative Local Treatments in Glioblastoma, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France
| | - Franck Lacoeuille
- Team 17-Design and Application of Innovative Local Treatments in Glioblastoma, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France.,Nuclear Medicine, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - François Hindré
- Team 17-Design and Application of Innovative Local Treatments in Glioblastoma, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France
| | | | | | - Caroline Bodet-Milin
- Nuclear Medicine, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Mickaël Bourgeois
- Nuclear Medicine, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Arronax, Saint-Herblain, France
| |
Collapse
|
34
|
Man F, Gawne PJ, T M de Rosales R. Nuclear imaging of liposomal drug delivery systems: A critical review of radiolabelling methods and applications in nanomedicine. Adv Drug Deliv Rev 2019; 143:134-160. [PMID: 31170428 PMCID: PMC6866902 DOI: 10.1016/j.addr.2019.05.012] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/25/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022]
Abstract
The integration of nuclear imaging with nanomedicine is a powerful tool for efficient development and clinical translation of liposomal drug delivery systems. Furthermore, it may allow highly efficient imaging-guided personalised treatments. In this article, we critically review methods available for radiolabelling liposomes. We discuss the influence that the radiolabelling methods can have on their biodistribution and highlight the often-overlooked possibility of misinterpretation of results due to decomposition in vivo. We stress the need for knowing the biodistribution/pharmacokinetics of both the radiolabelled liposomal components and free radionuclides in order to confidently evaluate the images, as they often share excretion pathways with intact liposomes (e.g. phospholipids, metallic radionuclides) and even show significant tumour uptake by themselves (e.g. some radionuclides). Finally, we describe preclinical and clinical studies using radiolabelled liposomes and discuss their impact in supporting liposomal drug development and clinical translation in several diseases, including personalised nanomedicine approaches.
Collapse
Affiliation(s)
- Francis Man
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Peter J Gawne
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom; London Centre for Nanotechnology, King's College London, Strand Campus, London WC2R 2LS, United Kingdom.
| |
Collapse
|
35
|
Halle B, Mongelard K, Poulsen FR. Convection-enhanced Drug Delivery for Glioblastoma: A Systematic Review Focused on Methodological Differences in the Use of the Convection-enhanced Delivery Method. Asian J Neurosurg 2019; 14:5-14. [PMID: 30937002 PMCID: PMC6417332 DOI: 10.4103/ajns.ajns_302_17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Glioblastoma (GBM) is a leading cause of brain cancer-related death. The blood–brain barrier (BBB) prevents the transport of most systemic delivered molecules to the brain. This constitutes a major problem in the therapy of brain tumors. In the last decade, numerous different drug-delivery approaches have been developed to overcome the BBB. The objective of this study is to provide an overview of the methodological aspects used in all preclinical and clinical studies published from 2011 to 2016 where convection-enhanced delivery (CED) was used for drug delivery in the treatment of GBM. A systematic review of English articles published in the past 5 years was undertaken using PubMed and Embase. The search terms (brain tumor [MeSH Terms]) AND (CED OR convection enhanced delivery) were used in PubMed and a similar search was carried out in Embase using their “multi-field search.” All studies using CED on an intracranial GBM model were included. The search resulted in 151 hits after duplicates were removed. In total, 30 studies were included in the review. Of these, two publications studied the technical aspects of the CED method. Furthermore, only one study was a clinical study. The research field is focused on preclinical drug development trials and less emphasis is placed on the CED technique itself. However, it is important that future studies focus on establishing optimal protocols for the use of CED in rodents as well as for big brain models to be able to use the CED method in patients with GBM.
Collapse
Affiliation(s)
- Bo Halle
- Department of Neurosurgery, Odense University Hospital and BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kristian Mongelard
- Department of Neurosurgery, Odense University Hospital and BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Frantz Rom Poulsen
- Department of Neurosurgery, Odense University Hospital and BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
36
|
Engudar G, Schaarup-Jensen H, Fliedner FP, Hansen AE, Kempen P, Jølck RI, Kjæer A, Andresen TL, Clausen MH, Jensen AI, Henriksen JR. Remote loading of liposomes with a 124I-radioiodinated compound and their in vivo evaluation by PET/CT in a murine tumor model. Am J Cancer Res 2018; 8:5828-5841. [PMID: 30613265 PMCID: PMC6299439 DOI: 10.7150/thno.26706] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/21/2018] [Indexed: 11/09/2022] Open
Abstract
Long circulating liposomes entrapping iodinated and radioiodinated compounds offer a highly versatile theranostic platform. Here we report a new methodology for efficient and high-yield loading of such compounds into liposomes, enabling CT/SPECT/PET imaging and 131I-radiotherapy. Methods: The CT contrast agent diatrizoate was synthetically functionalized with a primary amine, which enabled its remote loading into PEGylated liposomes by either an ammonium sulfate- or a citrate-based pH transmembrane gradient. Further, the amino-diatrizoate was radiolabeled with either 124I (t1/2 = 4.18 days) for PET or 125I (t1/2 = 59.5 days) for SPECT, through an aromatic Finkelstein reaction. Results: Quantitative loading efficiencies (>99%) were achieved at optimized conditions. The 124I-labeled compound was remote-loaded into liposomes, with an overall radiolabeling efficiency of 77 ± 1%, and imaged in vivo in a CT26 murine colon cancer tumor model by PET/CT. A prolonged blood circulation half-life of 19.5 h was observed for the radiolabeled liposomes, whereas injections of the free compound were rapidly cleared. Lower accumulation was observed in the spleen, liver, kidney and tumor than what is usually seen for long-circulating liposomes. Conclusion: The lower accumulation was interpreted as release of the tracer from the liposomes within these organs after accumulation. These results may guide the design of systems for controlled release of remote loadable drugs from liposomes.
Collapse
|
37
|
Kleynhans J, Grobler AF, Ebenhan T, Sathekge MM, Zeevaart JR. Radiopharmaceutical enhancement by drug delivery systems: A review. J Control Release 2018; 287:177-193. [DOI: 10.1016/j.jconrel.2018.08.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022]
|
38
|
Lacoeuille F, Arlicot N, Faivre-Chauvet A. Targeted alpha and beta radiotherapy: An overview of radiopharmaceutical and clinical aspects. MEDECINE NUCLEAIRE-IMAGERIE FONCTIONNELLE ET METABOLIQUE 2018. [DOI: 10.1016/j.mednuc.2017.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Séhédic D, Chourpa I, Tétaud C, Griveau A, Loussouarn C, Avril S, Legendre C, Lepareur N, Wion D, Hindré F, Davodeau F, Garcion E. Locoregional Confinement and Major Clinical Benefit of 188Re-Loaded CXCR4-Targeted Nanocarriers in an Orthotopic Human to Mouse Model of Glioblastoma. Theranostics 2017; 7:4517-4536. [PMID: 29158842 PMCID: PMC5695146 DOI: 10.7150/thno.19403] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 09/11/2017] [Indexed: 02/02/2023] Open
Abstract
PURPOSE Gold standard beam radiation for glioblastoma (GBM) treatment is challenged by resistance phenomena occurring in cellular populations well prepared to survive or to repair damage caused by radiation. Among signals that have been linked with radio-resistance, the SDF1/CXCR4 axis, associated with cancer stem-like cell, may be an opportune target. To avoid the problem of systemic toxicity and blood-brain barrier crossing, the relevance and efficacy of an original system of local brain internal radiation therapy combining a radiopharmaceutical with an immuno-nanoparticle was investigated. EXPERIMENT DESIGN The nanocarrier combined lipophilic thiobenzoate complexes of rhenium-188 loaded in the core of a lipid nanocapsule (LNC188Re) with a function-blocking antibody, 12G5 directed at the CXCR4, on its surface. The efficiency of 12G5-LNC188Re was investigated in an orthotopic and xenogenic GBM model of CXCR4-positive U87MG cells implanted in the striatum of Scid mice. RESULTS We demonstrated that 12G5-LNC188Re single infusion treatment by convection-enhanced delivery resulted in a major clinical improvement in median survival that was accompanied by locoregional effects on tumor development including hypovascularization and stimulation of the recruitment of bone marrow derived CD11b- or CD68-positive cells as confirmed by immunohistochemistry analysis. Interestingly, thorough analysis by spectral imaging in a chimeric U87MG GBM model containing CXCR4-positive/red fluorescent protein (RFP)-positive- and CXCR4-negative/RFP-negative-GBM cells revealed greater confinement of DiD-labeled 12G5-LNCs than control IgG2a-LNCs in RFP compartments. Main conclusion: These findings on locoregional impact and targeting of disseminated cancer cells in tumor margins suggest that intracerebral active targeting of nanocarriers loaded with radiopharmaceuticals may have considerable benefits in clinical applications.
Collapse
Affiliation(s)
- Delphine Séhédic
- CRCINA, INSERM, Université de Nantes, Université d'Angers, 49933 Angers, France
| | - Igor Chourpa
- Université Tours, EA6295, “Nanomédicaments et Nanosondes”, 37020 Tours, France
| | - Clément Tétaud
- CRCINA, INSERM, Université de Nantes, Université d'Angers, 49933 Angers, France
- PRIMEX, “Plateforme de Radiobiologie et d'Imagerie EXperimentale”, Université d'Angers, 49933 Angers, France
| | - Audrey Griveau
- CRCINA, INSERM, Université de Nantes, Université d'Angers, 49933 Angers, France
| | - Claire Loussouarn
- CRCINA, INSERM, Université de Nantes, Université d'Angers, 49933 Angers, France
| | - Sylvie Avril
- CRCINA, INSERM, Université de Nantes, Université d'Angers, 49933 Angers, France
| | - Claire Legendre
- CRCINA, INSERM, Université de Nantes, Université d'Angers, 49933 Angers, France
| | - Nicolas Lepareur
- Centre Regional de Lutte Contre le Cancer Eugène Marquis, Department of Nuclear Medicine, 35042 Rennes, France
| | - Didier Wion
- INSERM U1205, bâtiment modulaire 40-23, CEA 17 rue des Martyrs, 38054 Grenoble cedex, France
| | - François Hindré
- CRCINA, INSERM, Université de Nantes, Université d'Angers, 49933 Angers, France
- PRIMEX, “Plateforme de Radiobiologie et d'Imagerie EXperimentale”, Université d'Angers, 49933 Angers, France
| | - François Davodeau
- CRCINA, INSERM, Université d'Angers, Université de Nantes, 44035 Nantes, France
| | - Emmanuel Garcion
- CRCINA, INSERM, Université de Nantes, Université d'Angers, 49933 Angers, France
- PACeM, “Plateforme d'Analyses Cellulaires et Moléculaires”, Université d'Angers, 49933 Angers, France
- PRIMEX, “Plateforme de Radiobiologie et d'Imagerie EXperimentale”, Université d'Angers, 49933 Angers, France
| |
Collapse
|
40
|
Seo YE, Bu T, Saltzman WM. Nanomaterials for convection-enhanced delivery of agents to treat brain tumors. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2017; 4:1-12. [PMID: 29333521 DOI: 10.1016/j.cobme.2017.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Nanomaterials represent a promising and versatile platform for the delivery of therapeutics to the brain. Treatment of brain tumors has been a long-standing challenge in the field of neuro-oncology. The current standard of care - a multimodal approach of surgery, radiation and chemotherapy - yields only a modest therapeutic benefit for patients with malignant gliomas. A major obstacle for treatment is the failure to achieve sufficient delivery of therapeutics at the tumor site. Recent advances in local drug delivery techniques, along with the development of highly effective brain-penetrating nanocarriers, have significantly improved treatment and imaging of brain tumors in preclinical studies. The major advantage of this combined strategy is the ability to optimize local therapy, by maintaining an effective and sustained concentration of therapeutics in the brain with minimal systemic toxicity. This review highlights some of the latest developments, significant advancements and current challenges in local delivery of nanomaterials for the treatment of brain tumors.
Collapse
Affiliation(s)
- Young-Eun Seo
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Tom Bu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
41
|
Surface chemistry governs cellular tropism of nanoparticles in the brain. Nat Commun 2017; 8:15322. [PMID: 28524852 PMCID: PMC5454541 DOI: 10.1038/ncomms15322] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 03/21/2017] [Indexed: 02/07/2023] Open
Abstract
Nanoparticles are of long-standing interest for the treatment of neurological diseases such as glioblastoma. Most past work focused on methods to introduce nanoparticles into the brain, suggesting that reaching the brain interstitium will be sufficient to ensure therapeutic efficacy. However, optimized nanoparticle design for drug delivery to the central nervous system is limited by our understanding of their cellular deposition in the brain. Here, we investigated the cellular fate of poly(lactic acid) nanoparticles presenting different surface chemistries, after administration by convection-enhanced delivery. We demonstrate that nanoparticles with ‘stealth' properties mostly avoid internalization by all cell types, but internalization can be enhanced by functionalization with bio-adhesive end-groups. We also show that association rates measured in cultured cells predict the extent of internalization of nanoparticles in cell populations. Finally, evaluating therapeutic efficacy in an orthotopic model of glioblastoma highlights the need to balance significant uptake without inducing adverse toxicity. There have been numerous attempts to develop nanomaterials to reach cells of the central nervous system for drug delivery. Here, the authors investigate the cellular fate of polymer-based nanoparticles with varying surface chemistries after administration directly into the brain.
Collapse
|
42
|
Tagami T, Ozeki T. Recent Trends in Clinical Trials Related to Carrier-Based Drugs. J Pharm Sci 2017; 106:2219-2226. [PMID: 28259767 DOI: 10.1016/j.xphs.2017.02.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 02/07/2017] [Accepted: 02/13/2017] [Indexed: 02/06/2023]
Abstract
Clinical trials related to carrier-based drugs have recently attracted attention because carrier-based drugs hold promise for high efficiency drug delivery and for reducing drug-related side effects. In this commentary, we introduce recent clinical trials involving the use of various carriers, including liposomes, nano and micro particles, micelles, emulsions, and polymeric carriers. Liposomal drug carriers are currently the most intensively tested carriers in clinical trials, but other carriers such as polymeric carriers, albumin-based carriers, and metal nanocarriers have also recently been studied in clinical trials. Each carrier has specific properties, advantages, and disadvantages. The recent clinical trials introduced herein provide information critical to understanding current trends in carrier-based drug research.
Collapse
Affiliation(s)
- Tatsuaki Tagami
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Tetsuya Ozeki
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan.
| |
Collapse
|
43
|
Goins B, Bao A, Phillips WT. Techniques for Loading Technetium-99m and Rhenium-186/188 Radionuclides into Preformed Liposomes for Diagnostic Imaging and Radionuclide Therapy. Methods Mol Biol 2017; 1522:155-178. [PMID: 27837538 DOI: 10.1007/978-1-4939-6591-5_13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liposomes can serve as carriers of radionuclides for diagnostic imaging and therapeutic applications. Herein, procedures are outlined for radiolabeling liposomes with the gamma-emitting radionuclide, technetium-99m (99mTc), for noninvasive detection of disease and for monitoring the pharmacokinetics and biodistribution of liposomal drugs, and/or with therapeutic beta-emitting radionuclides, rhenium-186/188 (186/188Re), for radionuclide therapy. These efficient and practical liposome radiolabeling methods use a post-labeling mechanism to load 99mTc or 186/188Re into preformed liposomes prepared in advance of the labeling procedure. For all liposome radiolabeling methods described, a lipophilic chelator is used to transport 99mTc or 186/188Re across the lipid bilayer of the preformed liposomes. Once within the liposome interior, the pre-encapsulated glutathione or ammonium sulfate (pH) gradient provides for stable entrapment of the 99mTc and 186/188Re within the liposomes. In the first method, 99mTc is transported across the lipid bilayer by the lipophilic chelator, hexamethylpropyleneamine oxime (HMPAO) and 99mTc-HMPAO becomes trapped by interaction with the pre-encapsulated glutathione within the liposomes. In the second method, 99mTc or 186/188Re is transported across the lipid bilayer by the lipophilic chelator, N,N-bis(2-mercaptoethyl)-N',N'-diethylethylenediamine (BMEDA), and 99mTc-BMEDA or 186/188Re-BMEDA becomes trapped by interaction with pre-encapsulated glutathione within the liposomes. In the third method, an ammonium sulfate (pH) gradient loading technique is employed using liposomes with an extraliposomal pH of 7.4 and an interior pH of 5.1. BMEDA, which is lipophilic at pH 7.4, serves as a lipophilic chelator for 99mTc or 186/188Re to transport the radionuclides across the lipid bilayer. Once within the more acidic liposome interior, 99mTc/186/188Re-BMEDA complex becomes protonated and more hydrophilic, which results in stable entrapment of the 99mTc/186/188Re-BMEDA complex within the liposomes. Since many commercially available liposomal drugs use an ammonium sulfate (pH) gradient for drug loading, these liposomal drugs can be directly radiolabeled with 99mTc-BMEDA for noninvasive monitoring of tissue distribution during treatment or with 186/188Re-BMEDA for combination chemo-radionuclide therapy.
Collapse
Affiliation(s)
- Beth Goins
- Department of Radiology, MSC 7800, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Ande Bao
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland, OH, 44106, USA
| | - William T Phillips
- Department of Radiology, MSC 7800, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
44
|
Vieira DB, Gamarra LF. Getting into the brain: liposome-based strategies for effective drug delivery across the blood-brain barrier. Int J Nanomedicine 2016; 11:5381-5414. [PMID: 27799765 PMCID: PMC5077137 DOI: 10.2147/ijn.s117210] [Citation(s) in RCA: 264] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review summarizes articles that have been reported in literature on liposome-based strategies for effective drug delivery across the blood–brain barrier. Due to their unique physicochemical characteristics, liposomes have been widely investigated for their application in drug delivery and in vivo bioimaging for the treatment and/or diagnosis of neurological diseases, such as Alzheimer’s, Parkinson’s, stroke, and glioma. Several strategies have been used to deliver drug and/or imaging agents to the brain. Covalent ligation of such macromolecules as peptides, antibodies, and RNA aptamers is an effective method for receptor-targeting liposomes, which allows their blood–brain barrier penetration and/or the delivery of their therapeutic molecule specifically to the disease site. Additionally, methods have been employed for the development of liposomes that can respond to external stimuli. It can be concluded that the development of liposomes for brain delivery is still in its infancy, although these systems have the potential to revolutionize the ways in which medicine is administered.
Collapse
Affiliation(s)
| | - Lionel F Gamarra
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| |
Collapse
|
45
|
You S, Luo J, Grossniklaus HE, Gou ML, Meng K, Zhang Q. Nanomedicine in the application of uveal melanoma. Int J Ophthalmol 2016; 9:1215-25. [PMID: 27588278 DOI: 10.18240/ijo.2016.08.20] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/16/2016] [Indexed: 12/28/2022] Open
Abstract
Rapid advances in nanomedicine have significantly changed many aspects of nanoparticle application to the eye including areas of diagnosis, imaging and more importantly drug delivery. The nanoparticle-based drug delivery systems has provided a solution to various drug solubility-related problems in ophthalmology treatment. Nanostructured compounds could be used to achieve local ocular delivery with minimal unwanted systematic side effects produced by taking advantage of the phagocyte system. In addition, the in vivo control release by nanomaterials encapsulated drugs provides prolong exposure of the compound in the body. Furthermore, certain nanoparticles can overcome important body barriers including the blood-retinal barrier as well as the corneal-retinal barrier of the eye for effective delivery of the drug. In summary, the nanotechnology based drug delivery system may serve as an important tool for uveal melanoma treatment.
Collapse
Affiliation(s)
- Shuo You
- Department of Endocrinology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China; Winship Cancer Institute, School of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Jing Luo
- Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Hans E Grossniklaus
- Winship Cancer Institute, School of Medicine, Emory University, Atlanta, Georgia 30322, USA; Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia 30322, USA; Department of Pathology, School of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Ma-Ling Gou
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ke Meng
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Qing Zhang
- Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China; Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia 30322, USA
| |
Collapse
|
46
|
van der Geest T, Laverman P, Metselaar JM, Storm G, Boerman OC. Radionuclide imaging of liposomal drug delivery. Expert Opin Drug Deliv 2016; 13:1231-42. [PMID: 27351233 DOI: 10.1080/17425247.2016.1205584] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Ever since their discovery, liposomes have been radiolabeled to monitor their fate in vivo. Despite extensive preclinical studies, only a limited number of radiolabeled liposomal formulations have been examined in patients. Since they can play a crucial role in patient management, it is of importance to enable translation of radiolabeled liposomes into the clinic. AREAS COVERED Liposomes have demonstrated substantial advantages as drug delivery systems and can be efficiently radiolabeled. Potentially, radiolabeled drug-loaded liposomes form an elegant theranostic system, which can be tracked in vivo using single-photon emission computed tomography (SPECT) or positron emission tomography (PET) imaging. In this review, we discuss important aspects of liposomal research with a focus on the use of radiolabeled liposomes and their potential role in drug delivery and monitoring therapeutic effects. EXPERT OPINION Radiolabeled drug-loaded liposomes have been poorly investigated in patients and no radiolabeled liposomes have been approved for use in clinical practice. Evaluation of the risks, pharmacokinetics, pharmacodynamics and toxicity is necessary to meet pharmaceutical and commercial requirements. It remains to be demonstrated whether the results found in animal studies translate to humans before radiolabeled liposomes can be implemented into clinical practice.
Collapse
Affiliation(s)
- Tessa van der Geest
- a Department of Radiology and Nuclear Medicine , Radboud University Medical Center , Nijmegen , The Netherlands
| | - Peter Laverman
- a Department of Radiology and Nuclear Medicine , Radboud University Medical Center , Nijmegen , The Netherlands
| | - Josbert M Metselaar
- b Department of Experimental Molecular Imaging , University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH - Aachen University , Aachen , Germany.,c Department of Targeted Therapeutics , MIRA Institute, University of Twente , Enschede , The Netherlands
| | - Gert Storm
- c Department of Targeted Therapeutics , MIRA Institute, University of Twente , Enschede , The Netherlands.,d Department of Pharmaceutics , Utrecht Institute for Pharmaceutical Sciences, Utrecht University , Utrecht , The Netherlands
| | - Otto C Boerman
- a Department of Radiology and Nuclear Medicine , Radboud University Medical Center , Nijmegen , The Netherlands
| |
Collapse
|
47
|
Goins B, Phillips WT, Bao A. Strategies for improving the intratumoral distribution of liposomal drugs in cancer therapy. Expert Opin Drug Deliv 2016; 13:873-89. [PMID: 26981891 DOI: 10.1517/17425247.2016.1167035] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION A major limitation of current liposomal cancer therapies is the inability of liposome therapeutics to penetrate throughout the entire tumor mass. This inhomogeneous distribution of liposome therapeutics within the tumor has been linked to treatment failure and drug resistance. Both liposome particle transport properties and tumor microenvironment characteristics contribute to this challenge in cancer therapy. This limitation is relevant to both intravenously and intratumorally administered liposome therapeutics. AREAS COVERED Strategies to improve the intratumoral distribution of liposome therapeutics are described. Combination therapies of intravenous liposome therapeutics with pharmacologic agents modulating abnormal tumor vasculature, interstitial fluid pressure, extracellular matrix components, and tumor associated macrophages are discussed. Combination therapies using external stimuli (hyperthermia, radiofrequency ablation, magnetic field, radiation, and ultrasound) with intravenous liposome therapeutics are discussed. Intratumoral convection-enhanced delivery (CED) of liposomal therapeutics is reviewed. EXPERT OPINION Optimization of the combination therapies and drug delivery protocols are necessary. Further research should be conducted in appropriate cancer types with consideration of physiochemical features of liposomes and their timing sequence. More investigation of the role of tumor associated macrophages in intratumoral distribution is warranted. Intratumoral infusion of liposomes using CED is a promising approach to improve their distribution within the tumor mass.
Collapse
Affiliation(s)
- Beth Goins
- a Department of Radiology , University of Texas Health Science Center San Antonio , San Antonio , TX , USA
| | - William T Phillips
- a Department of Radiology , University of Texas Health Science Center San Antonio , San Antonio , TX , USA
| | - Ande Bao
- b Department of Radiation Oncology, School of Medicine, Case Western Reserve University/University Hospitals Case Medical Center , Cleveland , OH , USA
| |
Collapse
|
48
|
Lai SF, Ko BH, Chien CC, Chang CJ, Yang SM, Chen HH, Petibois C, Hueng DY, Ka SM, Chen A, Margaritondo G, Hwu Y. Gold nanoparticles as multimodality imaging agents for brain gliomas. J Nanobiotechnology 2015; 13:85. [PMID: 26589283 PMCID: PMC4654925 DOI: 10.1186/s12951-015-0140-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/22/2015] [Indexed: 11/20/2022] Open
Abstract
Background Nanoparticles can be used for targeted drug delivery, in particular for brain cancer therapy. However, this requires a detailed analysis of nanoparticles from the associated microvasculature to the tumor, not easy because of the required high spatial resolution. The objective of this study is to demonstrate an experimental solution of this problem, based in vivo and post-mortem whole organ imaging plus nanoscale 3-dimensional (3D) X-ray microscopy. Results The use of gold nanoparticles (AuNPs) as contrast agents paved the way to a detailed high-resolution three dimensional (3D) X-ray and fluorescence imaging analysis of the relation between xenografted glioma cells and the tumor-induced angiogenic microvasculature. The images of the angiogenic microvessels revealed nanoparticle leakage. Complementary tests showed that after endocytotic internalization fluorescent AuNPs allow the visible-light detection of cells. Conclusions AuNP-loading of cells could be extended from the case presented here to other imaging techniques. In our study, they enabled us to (1) identify primary glioma cells at inoculation sites in mice brains; (2) follow the subsequent development of gliomas. (3) Detect the full details of the tumor-related microvasculature; (4) Finding leakage of AuNPs from the tumor-related vasculature, in contrast to no leakage from normal vasculature. Electronic supplementary material The online version of this article (doi:10.1186/s12951-015-0140-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sheng-Feng Lai
- Department of Engineering Science, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Bai-Hung Ko
- Department of Engineering Science, National Cheng Kung University, Tainan, 701, Taiwan. .,Institute of Physics, Academia Sinica, Nankang, Taipei, 115, Taiwan.
| | - Chia-Chi Chien
- Institute of Physics, Academia Sinica, Nankang, Taipei, 115, Taiwan.
| | - Chia-Ju Chang
- Institute of Physics, Academia Sinica, Nankang, Taipei, 115, Taiwan.
| | - Shun-Ming Yang
- Institute of Physics, Academia Sinica, Nankang, Taipei, 115, Taiwan.
| | - Hsiang-Hsin Chen
- Inserm U1029 LMMA, University of Bordeaux, 33600, Pessac Cedex, France.
| | - Cyril Petibois
- Inserm U1029 LMMA, University of Bordeaux, 33600, Pessac Cedex, France.
| | - Dueng-Yuan Hueng
- Department of Biochemistry, School of Medicine, National Defense Medical Center, Taipei, 114, Taiwan. .,Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan.
| | - Shuk-Man Ka
- Institute of Aerospace and Undersea Medicine, School of Medicine, National Defense Medical Center, Taipei, 114, Taiwan.
| | - Ann Chen
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan.
| | - G Margaritondo
- School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland.
| | - Y Hwu
- Department of Engineering Science, National Cheng Kung University, Tainan, 701, Taiwan. .,Institute of Physics, Academia Sinica, Nankang, Taipei, 115, Taiwan. .,Advanced Optoelectronic Technology Center, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
49
|
Gruslova A, Cavazos DA, Miller JR, Breitbart E, Cohen YC, Bangio L, Yakov N, Soundararajan A, Floyd JR, Brenner AJ. VB-111: a novel anti-vascular therapeutic for glioblastoma multiforme. J Neurooncol 2015; 124:365-72. [PMID: 26108658 PMCID: PMC4584173 DOI: 10.1007/s11060-015-1853-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/14/2015] [Indexed: 02/01/2023]
Abstract
Glioblastoma multiforme (GBM) is among the most highly vascularized of solid tumors, contributing to the infiltrative nature of the disease, and conferring poor outcome. Due to the critical dependency of GBM on growth of new endothelial vasculature, we evaluated the preclinical activity of a novel adenoviral gene therapy that targets the endothelium within newly formed blood vessels for apoptosis. VB-111, currently in phase II clinical trials, consists of a non-replicating Adenovirus 5 (El deleted) carrying a proapoptotic human Fas-chimera (transgene) under the control of a modified murine promoter (PPE-1-3×) which specifically targets endothelial cells within the tumor vasculature. Here we report that a single intravenous dose of 2.5 × 10(11) or 1 × 10(11) VPs was sufficient to extend survival in nude rats bearing U87MG-luc2 or nude mice bearing U251-luc, respectively. Bioluminescence imaging of nude rats showed that VB-111 effectively inhibited tumor growth within four weeks of treatment. This was confirmed in a select group of animals by MRI. In our mouse model we observed that 3 of 10 nude mice treated with VB-111 completely lost U251 luciferase signal and were considered long term survivors. To assess the antiangiogenic effects of VB-111, we evaluated the tumor-associated microvaculature by CD31, a common marker of neovascularization, and found a significant decrease in the microvessel density by IHC. We further assessed the neovasculature by confocal microscopy and found that VB-111 inhibits vascular density in two separate mouse models bearing U251-RFP xenografts. Collectively, this study supports the clinical development of VB-111 as a treatment for GBM.
Collapse
Affiliation(s)
- Aleksandra Gruslova
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - David A Cavazos
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Jessica R Miller
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Eyal Breitbart
- VBL Therapeutics, 6 Jonathan Netanyahu St., Or Yehuda, 60376, Israel
| | - Yael C Cohen
- VBL Therapeutics, 6 Jonathan Netanyahu St., Or Yehuda, 60376, Israel
| | - Livnat Bangio
- VBL Therapeutics, 6 Jonathan Netanyahu St., Or Yehuda, 60376, Israel
| | - Niva Yakov
- VBL Therapeutics, 6 Jonathan Netanyahu St., Or Yehuda, 60376, Israel
| | - Anu Soundararajan
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - John R Floyd
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Andrew J Brenner
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
50
|
Luwor RB, Stylli SS, Kaye AH. Using bioluminescence imaging in glioma research. J Clin Neurosci 2015; 22:779-84. [DOI: 10.1016/j.jocn.2014.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 11/03/2014] [Indexed: 01/02/2023]
|