1
|
Fu W, Liang Q, Ma Y, Lei S, Li R, Zheng X, Chen L, Chen J, Cai X, Dai X, Duan H, He W, Ren J. Fn14-targeting, NIR-II responsive nanomaterials for enhanced radiotherapy against glioblastomas. NANOSCALE ADVANCES 2025; 7:2634-2647. [PMID: 40109505 PMCID: PMC11915457 DOI: 10.1039/d4na00788c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/01/2025] [Indexed: 03/22/2025]
Abstract
Radiotherapy is a common treatment option for patients with glioblastoma multiforme. However, tumor heterogeneity causes varying responses to radiation among different tumor subpopulations. Cancer cells that endure radiotherapy exhibit radioresistance, resulting in the ineffectiveness of radiation therapy and eventual tumor relapse. In this study, we discovered that the fibroblast growth factor-inducible 14 (Fn14)-positive tumor cells were enriched in tumor residual foci after radiation, ultimately leading to treatment failure. Fn14-expressing glioma cells survived ionizing radiation through preferential activation of DNA damage checkpoint response. We have thus engineered an Fn14-targeting and NIR-II responsive plasmonic gold nanosystem named Fn14-AuNPs, which can precisely internalize into Fn14-overexpressed glioma cells and have an excellent BBB-crossing capability. As gold nanoparticles, by inhibition of DNA repair processes and induction of G2/M cells cycle arrest, Fn14-AuNPs nanoparticles improved the radiosensitivity of tumor cells. Meanwhile, Fn14-AuNPs induced localized heat under NIR-II photoirradiation, thus impeding RT-induced DNA damage checkpoint response. This versatile nanosensitizer, combined with NIR-II laser photoirradiation, can eradicate radioresistant subpopulations of glioblastoma and improve the therapeutic effect of radiotherapy. This finding presents an effective radiosensitization strategy by targeting radioresistant subpopulations, which can efficiently overcome the constraints imposed in clinical radiotherapy and offer a hopeful avenue to enhance the treatment effectivity of radiotherapy in glioblastoma.
Collapse
Affiliation(s)
- Wei Fu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Qing Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Yuxi Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Shiqiong Lei
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
| | - Ruiqi Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Xin Zheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Lian Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Jiayuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Xing Cai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Xiaofang Dai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Hongwei Duan
- School of Chemical and Biomedical Engineering, Nanyang Technological University Singapore 637457 Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore 636921 Singapore
| | - Wenshan He
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
- Shenzhen Huazhong University of Science and Technology Research Institute Shenzhen 518057 China
| |
Collapse
|
2
|
Rivera D, Bouras A, Mattioli M, Anastasiadou M, Pacentra AC, Pelcher O, Koziel C, Schupper AJ, Chanenchuk T, Carlton H, Ivkov R, Hadjipanayis CG. Magnetic hyperthermia therapy enhances the chemoradiosensitivity of glioblastoma. Sci Rep 2025; 15:10532. [PMID: 40148452 PMCID: PMC11950323 DOI: 10.1038/s41598-025-95544-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/21/2025] [Indexed: 03/29/2025] Open
Abstract
Glioblastoma (GBM) is the most common primary brain cancer and is resistant to standard-of-care chemoradiation therapy (CRT). Magnetic hyperthermia therapy (MHT) exposes magnetic iron oxide nanoparticles (MIONPs) to an alternating magnetic field (AMF) to generate local hyperthermia. This study evaluated MHT-mediated enhancement of CRT in preclinical GBM models. Cell viability and apoptosis were assessed in GBM cell lines after water bath heating with radiation and/or temozolomide. Heating efficiency of MIONPs after intracranial delivery was measured in healthy mice. MHT with CRT was performed in syngeneic and patient-derived xenograft (PDX) GBM tumors. Tissue sections were analyzed for γ-H2AX, HSP90, CD4 + T cells, and microglial cells. Tumor burden and survival were assessed. Hyperthermia with radiation and temozolomide significantly reduced cell viability and increased apoptosis. Hyperthermia predominantly exhibited additive to synergistic interactions with both treatment modalities and reduced doses needed for tumor cell growth inhibition. In vivo, MHT with CRT decreased tumor burden and increased survival in PDX and syngeneic models. Immunohistochemistry showed increased γ-H2AX, HSP90, microglial activation, and CD4 + T cells after MHT in combination with CRT. Overall, adjuvant hyperthermia enhances CRT efficacy in GBM cells, with MHT improving survival outcomes in rodents. Sufficient intracranial heating and MIONP retention for repeated treatments was achieved, supporting further clinical translation.
Collapse
Affiliation(s)
- Daniel Rivera
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Milena Mattioli
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Maria Anastasiadou
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Anna Chiara Pacentra
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Olivia Pelcher
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Corrine Koziel
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Alexander J Schupper
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Tori Chanenchuk
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Hayden Carlton
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Constantinos G Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA.
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| |
Collapse
|
3
|
Zhao X, Li C, Wang K, Zhang Y, Wan H, Yang P, Zhou D. MR‑guided laser interstitial thermal therapy followed by early application of temozolomide for recurrent IDH-wildtype glioblastomas: preliminary results from a prospective study. Neurosurg Rev 2025; 48:254. [PMID: 39971799 DOI: 10.1007/s10143-025-03402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/19/2025] [Accepted: 02/09/2025] [Indexed: 02/21/2025]
Abstract
This study aims to evaluate the safety, tolerability, and preliminary efficacy of combining laser interstitial thermal therapy (LITT) with early administration of temozolomide (TMZ) in patients with recurrent glioblastoma (rGBM). Ten patients with rGBM were enrolled. Following the LITT procedure, TMZ was administered at a dose of 75 mg/m2/day during the early-TMZ phase for three weeks. After a 7-day interval, TMZ was given according to the standard dosage scheme for 6 cycles. Adverse events and complications encountered were documented. Regular follow-up assessments were conducted to evaluate both patient performance status and tumor progression. All patients demonstrated good tolerance to LITT, with six out of ten achieving an ablation rate above 90%, and only one patient had an ablation rate below 70%. Oral administration of TMZ was well-tolerated by all patients during the early-TMZ phase. Mild headache was the most common adverse event (3/10), and only one severe adverse event occurred. At a 6-month follow-up post-LITT, tumor progression was observed in five patients; noneof the patients reached survival endpoints. This preliminary report substantiates the favorable tolerability of early application of TMZ in combination with LITT. The safety profile was found to be acceptable, and the initial efficacy results were promising. Future studies should explore the potential of LITT combination therapy in greater detail and with larger patient samples.
Collapse
Affiliation(s)
- Xuzhe Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Chao Li
- Department of Neurosurgery, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Kai Wang
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yutao Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Haibin Wan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Pei Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Dabiao Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
4
|
Yu JS, Meade SM, Zhao R, Wei W, Dashora H, Prayson R, Grabowski MM, Stevens G, Lobbous M, Murphy ES, Suh JH, Chao ST, Barnett GH, Peereboom D, Ahluwalia MS, Mohammadi AM. Expedited chemoradiation after laser interstitial thermal therapy (LITT) is feasible and safe in patients with newly diagnosed glioblastoma. Neurooncol Adv 2025; 7:vdaf038. [PMID: 40276375 PMCID: PMC12019959 DOI: 10.1093/noajnl/vdaf038] [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: 04/26/2025] Open
Abstract
Background High-grade gliomas (HGG) are incurable primary brain tumors. Laser interstitial thermal therapy (LITT) has emerged as an alternative to surgery for select patients. Hyperthermia can improve the efficacy of radiation and chemotherapy. Shortening the time between LITT and chemoradiation may maximize their biological and clinical benefits. This trial evaluated the safety and feasibility of expediting chemoradiation after biopsy and LITT in patients with newly diagnosed HGG. Methods Patients with suspected HGG were enrolled. Those with pathologic confirmation of HGG and deemed appropriate candidates for LITT and chemoradiation were considered evaluable. Participants underwent 6 weeks of adjuvant chemoradiation initiated within 7 days of LITT. Endpoints were assessed until the completion of radiation and included the occurrence of wound dehiscence; new, treatment-refractory seizures; cerebral edema; and completion of planned radiotherapy. Results Thirteen patients with suspected HGG were enrolled, and ten were considered evaluable. All 10 patients were diagnosed with glioblastoma (GBM, IDHwt). Three patients were deemed unevaluable: 2 patients with other CNS tumors and one GBM patient who developed grade 4 postoperative edema. Of 10 evaluable patients, the median age was 60.2 years (IQR: 51.0, 69.4), and median preoperative KPS was 90 (IQR: 90, 80). The median time between LITT and the initiation of chemoradiation was 7 days. There were no occurrences of significant protocol-related adverse events. Conclusions Accelerated initiation of chemoradiation after biopsy and LITT is safe and feasible for patients with newly diagnosed GBM. A larger study is needed to assess potential synergy of hyperthermia and chemoradiation to improve survival.
Collapse
Affiliation(s)
- Jennifer S Yu
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Seth M Meade
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ran Zhao
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Wei Wei
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Himanshu Dashora
- Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Richard Prayson
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Matthew M Grabowski
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Glen Stevens
- Department of Neurology, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mina Lobbous
- Department of Neurology, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Erin S Murphy
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - John H Suh
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Samuel T Chao
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Gene H Barnett
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - David Peereboom
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Manmeet S Ahluwalia
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alireza M Mohammadi
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
5
|
Righini MF, Durham A, Tsoutsou PG. Hyperthermia and radiotherapy: physiological basis for a synergistic effect. Front Oncol 2024; 14:1428065. [PMID: 39165690 PMCID: PMC11333208 DOI: 10.3389/fonc.2024.1428065] [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: 05/05/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024] Open
Abstract
In cancer treatment, mild hyperthermia (HT) represents an old, but recently revived opportunity to increase the efficacy of radiotherapy (RT) without increasing side effects, thereby widening the therapeutic window. HT disrupts cellular homeostasis by acting on multiple targets, and its combination with RT produces synergistic antitumoral effects on specific pathophysiological mechanisms, associated to DNA damage and repair, hypoxia, stemness and immunostimulation. HT is furthermore associated to direct tumor cell kill, particularly in higher temperature levels. A phenomenon of temporary resistance to heat, known as thermotolerance, follows each HT session. Cancer treatment requires innovative concepts and combinations to be tested but, for a meaningful development of clinical trials, the understanding of the underlying mechanisms of the tested modalities is essential. In this mini-review, we aimed to describe the synergistic effects of the combination of HT with RT as well as the phenomena of thermal shock and thermotolerance, in order to stimulate clinicians in new, clinically relevant concepts and combinations, which become particularly relevant in the era of technological advents in both modalities but also cancer immunotherapy.
Collapse
Affiliation(s)
| | - André Durham
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
- Department of Radiation Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Pelagia G. Tsoutsou
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
- Department of Radiation Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| |
Collapse
|
6
|
Smadja DM. Hyperthermia for Targeting Cancer and Cancer Stem Cells: Insights from Novel Cellular and Clinical Approaches. Stem Cell Rev Rep 2024; 20:1532-1539. [PMID: 38795304 DOI: 10.1007/s12015-024-10736-0] [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] [Accepted: 05/14/2024] [Indexed: 05/27/2024]
Abstract
The Cellular Heat Shock Response and in particular heat shock protein activation are vital stress reactions observed in both healthy and cancer cells. Hyperthermia (HT) has been proposed for several years as an advancing non-invasive cancer therapy. It selectively targets cancer cells through mechanisms influenced by temperature and temperature variations. This article delves into the impact of HT on cancer cells, especially cancer stem cells (CSCs), essential contributors to cancer recurrence and metastasis. HT has shown promise in eliminating CSCs, sensitizing them to conventional treatments and modulating the tumor microenvironment. The exploration extends to mesenchymal stem cells (MSCs), which exhibit both pro-tumorigenic and anti-tumorigenic effects. HT's potential in recruiting therapeutic MSCs for targeted delivery of antitumoral agents is also discussed. Furthermore, the article introduces Brain Thermodynamics-guided Hyperthermia (BTGH) technology, a breakthrough in temperature control and modulation of heat transfer under different conditions. This non-invasive method leverages the brain-eyelid thermal tunnel (BTT) to monitor and regulate internal brain temperature. BTGH technology, with its precision and noninvasive continuous monitoring capabilities, is under clinical investigation for applications in neurological disorders and cancer. The innovative three-phase approach involves whole-body HT, targeted brain HT, and organ-specific HT. In conclusion, the exploration of localized or whole-body HT offers promising avenues for cancer, psychiatric and neurological diseases. The ongoing clinical investigations and potential applications underscore the significance of understanding and harnessing heat's responses to enhance human health.
Collapse
Affiliation(s)
- David M Smadja
- Paris Cité University, INSERM, Innovative Therapies in Hemostasis, Paris, F-75006, France.
- Hematology Department, AP-HP, Georges Pompidou European Hospital, 20 rue Leblanc, Paris, F-75015, France.
| |
Collapse
|
7
|
Ali BH, Khoee S, Mafakheri F, Sadri E, Mahabadi VP, Karimi MR, Shirvalilou S, Khoei S. Active targeted delivery of theranostic thermo/pH dual-responsive magnetic Janus nanoparticles functionalized with folic acid/fluorescein ligands for enhanced DOX combination therapy of rat glioblastoma. J Mater Chem B 2024; 12:5957-5973. [PMID: 38808630 DOI: 10.1039/d3tb02429f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Doxorubicin (DOX), a chemotherapy drug, has demonstrated limited efficacy against glioblastoma, an aggressive brain tumor with resistance attributed to the blood-brain barrier (BBB). This study aims to overcome this challenge by proposing the targeted delivery of magnetic Janus nanoparticles (MJNPs) functionalized with folic acid ligands, fluorescent dye, and doxorubicin (DOX/MJNPs-FLA). The properties of these nanoparticles were comprehensively evaluated using bio-physiochemical techniques such as Fourier transform infrared (FTIR) spectroscopy, dynamic light scattering (DLS), zeta potential analysis, high-resolution transmission electron microscopy (HR-TEM), vibrating sample magnetometry (VSM), fluorescence microscopy, MTT assay, hemolysis assay, and liver enzyme level evaluation. Dual-controlled DOX release was investigated under different pH and temperature conditions. Additionally, the impact of DOX/MJNPs-FLA on apoptosis induction in tumor cells, body weight, and survival time of cancerous animals was assessed. The targeted delivery system was assessed using C6 and OLN-93 cell lines as representatives of cancerous and healthy cell lines, respectively, alongside Wistar rat tumor-bearing models. Results from Prussian blue staining and confocal microscopy tests demonstrated the effective targeted internalization of MJNPs-FLA by glioblastoma cells. Additionally, we investigated the biodistribution of the nanoparticles utilizing fluorescence imaging techniques. This enabled us to track the distribution pattern of MJNPs-FLA in vivo, shedding light on their movement and accumulation within the biological system. Furthermore, the combination of chemotherapy and magnetic hyperthermia exhibited enhanced efficacy in inducing apoptosis, as evidenced by the increase of the pro-apoptotic Bax gene and a decrease in the anti-apoptotic Bcl-2 gene. Remarkably, this combination treatment did not cause any hepatotoxicity. This study highlights the potential of DOX/MJNPs-FLA as carriers for therapeutic and diagnostic agents in the context of theranostic applications for the treatment of brain malignancies. Additionally, it demonstrates the promising performance of DOX/MJNPs-FLA in combination treatment through passive and active targeting.
Collapse
Affiliation(s)
- Bahareh Haji Ali
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Sepideh Khoee
- Department of Polymer Chemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Fariba Mafakheri
- Department of Polymer Chemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Elahe Sadri
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Canada
| | | | - Mohammad Reza Karimi
- Department of Polymer Chemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Sakine Shirvalilou
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Samideh Khoei
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Chang C, Chavarro VS, Gerstl JVE, Blitz SE, Spanehl L, Dubinski D, Valdes PA, Tran LN, Gupta S, Esposito L, Mazzetti D, Gessler FA, Arnaout O, Smith TR, Friedman GK, Peruzzi P, Bernstock JD. Recurrent Glioblastoma-Molecular Underpinnings and Evolving Treatment Paradigms. Int J Mol Sci 2024; 25:6733. [PMID: 38928445 PMCID: PMC11203521 DOI: 10.3390/ijms25126733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Glioblastoma is the most common and lethal central nervous system malignancy with a median survival after progression of only 6-9 months. Major biochemical mechanisms implicated in glioblastoma recurrence include aberrant molecular pathways, a recurrence-inducing tumor microenvironment, and epigenetic modifications. Contemporary standard-of-care (surgery, radiation, chemotherapy, and tumor treating fields) helps to control the primary tumor but rarely prevents relapse. Cytoreductive treatment such as surgery has shown benefits in recurrent glioblastoma; however, its use remains controversial. Several innovative treatments are emerging for recurrent glioblastoma, including checkpoint inhibitors, chimeric antigen receptor T cell therapy, oncolytic virotherapy, nanoparticle delivery, laser interstitial thermal therapy, and photodynamic therapy. This review seeks to provide readers with an overview of (1) recent discoveries in the molecular basis of recurrence; (2) the role of surgery in treating recurrence; and (3) novel treatment paradigms emerging for recurrent glioblastoma.
Collapse
Affiliation(s)
- Christopher Chang
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA;
| | - Velina S. Chavarro
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
| | - Jakob V. E. Gerstl
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
| | - Sarah E. Blitz
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Lennard Spanehl
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Department of Neurosurgery, University of Rostock, 18055 Rostock, Germany; (D.D.); (F.A.G.)
| | - Daniel Dubinski
- Department of Neurosurgery, University of Rostock, 18055 Rostock, Germany; (D.D.); (F.A.G.)
| | - Pablo A. Valdes
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Lily N. Tran
- Division of Biology and Medicine, Brown University, Providence, RI 02912, USA;
| | - Saksham Gupta
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Luisa Esposito
- Department of Medicine and Surgery, Unicamillus University, 00131 Rome, Italy;
| | - Debora Mazzetti
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
| | - Florian A. Gessler
- Department of Neurosurgery, University of Rostock, 18055 Rostock, Germany; (D.D.); (F.A.G.)
| | - Omar Arnaout
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Timothy R. Smith
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Gregory K. Friedman
- Division of Pediatrics, Neuro-Oncology Section, MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Pierpaolo Peruzzi
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Joshua D. Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
9
|
Nwafor DC, Obiri-Yeboah D, Fazad F, Blanks W, Mut M. Focused ultrasound as a treatment modality for gliomas. Front Neurol 2024; 15:1387986. [PMID: 38813245 PMCID: PMC11135048 DOI: 10.3389/fneur.2024.1387986] [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: 02/19/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
Ultrasound waves were initially used as a diagnostic tool that provided critical insights into several pathological conditions (e.g., gallstones, ascites, pneumothorax, etc.) at the bedside. Over the past decade, advancements in technology have led to the use of ultrasound waves in treating many neurological conditions, such as essential tremor and Parkinson's disease, with high specificity. The convergence of ultrasound waves at a specific region of interest/target while avoiding surrounding tissue has led to the coined term "focused ultrasound (FUS)." In tumor research, ultrasound technology was initially used as an intraoperative guidance tool for tumor resection. However, in recent years, there has been growing interest in utilizing FUS as a therapeutic tool in the management of brain tumors such as gliomas. This mini-review highlights the current knowledge surrounding using FUS as a treatment modality for gliomas. Furthermore, we discuss the utility of FUS in enhanced drug delivery to the central nervous system (CNS) and highlight promising clinical trials that utilize FUS as a treatment modality for gliomas.
Collapse
Affiliation(s)
- Divine C. Nwafor
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, United States
| | - Derrick Obiri-Yeboah
- Department of Neurological Surgery, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
| | - Faraz Fazad
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, United States
| | - William Blanks
- Department of Neurosurgery, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Melike Mut
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
10
|
Rentzeperis F, Rivera D, Zhang JY, Brown C, Young T, Rodriguez B, Schupper A, Price G, Gomberg J, Williams T, Bouras A, Hadjipanayis C. Recent Developments in Magnetic Hyperthermia Therapy (MHT) and Magnetic Particle Imaging (MPI) in the Brain Tumor Field: A Scoping Review and Meta-Analysis. MICROMACHINES 2024; 15:559. [PMID: 38793132 PMCID: PMC11123314 DOI: 10.3390/mi15050559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/28/2024] [Accepted: 04/17/2024] [Indexed: 05/26/2024]
Abstract
Magnetic hyperthermia therapy (MHT) is a promising treatment modality for brain tumors using magnetic nanoparticles (MNPs) locally delivered to the tumor and activated with an external alternating magnetic field (AMF) to generate antitumor effects through localized heating. Magnetic particle imaging (MPI) is an emerging technology offering strong signal-to-noise for nanoparticle localization. A scoping review was performed by systematically querying Pubmed, Scopus, and Embase. In total, 251 articles were returned, 12 included. Articles were analyzed for nanoparticle type used, MHT parameters, and MPI applications. Preliminary results show that MHT is an exciting treatment modality with unique advantages over current heat-based therapies for brain cancer. Effective application relies on the further development of unique magnetic nanoparticle constructs and imaging modalities, such as MPI, that can enable real-time MNP imaging for improved therapeutic outcomes.
Collapse
Affiliation(s)
- Frederika Rentzeperis
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
- Sinai BioDesign, Department of Neurosurgery, Mount Sinai, New York, NY 10029, USA;
| | - Daniel Rivera
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
| | - Jack Y. Zhang
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
| | - Cole Brown
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
| | - Tirone Young
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
- Sinai BioDesign, Department of Neurosurgery, Mount Sinai, New York, NY 10029, USA;
| | - Benjamin Rodriguez
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
- Sinai BioDesign, Department of Neurosurgery, Mount Sinai, New York, NY 10029, USA;
| | - Alexander Schupper
- Department of Neurological Surgery, Mount Sinai Hospital, New York, NY 10029, USA;
| | - Gabrielle Price
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
| | - Jack Gomberg
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
| | - Tyree Williams
- Sinai BioDesign, Department of Neurosurgery, Mount Sinai, New York, NY 10029, USA;
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA;
| | - Constantinos Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA;
- Center for Image-Guided Neurosurgery, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
11
|
Zhao X, Li R, Guo Y, Wan H, Zhou D. Laser interstitial thermal therapy for recurrent glioblastomas: a systematic review and meta-analysis. Neurosurg Rev 2024; 47:159. [PMID: 38625588 DOI: 10.1007/s10143-024-02409-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/29/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
We aim to investigate the efficacy and safety of laser interstitial thermal therapy (LITT) in treating recurrent glioblastomas (rGBMs). A comprehensive search was conducted in four databases to identify studies published between January 2001 and June 2022 that reported prognosis information of rGBM patients treated with LITT as the primary therapy. The primary outcomes of interest were progression-free survival (PFS) and overall survival (OS) at 6 and 12 months after LITT intervention. Adverse events and complications were also evaluated. Eight eligible non-comparative studies comprising 128 patients were included in the analysis. Seven studies involving 120 patients provided data for the analysis of PFS. The pooled PFS rate at 6 months after LITT was 25% (95% CI 15-37%, I2 = 53%), and at 12 months, it was 9% (95% CI 4-15%, I2 = 24%). OS analysis was performed on 54 patients from six studies, with an OS rate of 92% (95% CI 84-100%, I2 = 0%) at 6 months and 42% (95% CI 13-73%, I2 = 67%) at 12 months after LITT. LITT demonstrates a favorable safety profile with low complication rates and promising tumor control and overall survival rates in patients with rGBMs. Tumor volume and performance status are important factors that may influence the effectiveness of LITT in selected patients. Additionally, the combination of LITT with immune-based therapy holds promise. Further well-designed clinical trials are needed to expand the application of LITT in glioma treatment.
Collapse
Affiliation(s)
- Xuzhe Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, #119 Fanyang Road, Fengtai District, Beijing, 100070, China
| | - Runting Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, #119 Fanyang Road, Fengtai District, Beijing, 100070, China
| | - Yiding Guo
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, #119 Fanyang Road, Fengtai District, Beijing, 100070, China
| | - Haibin Wan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, #119 Fanyang Road, Fengtai District, Beijing, 100070, China
| | - Dabiao Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, #119 Fanyang Road, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
12
|
Huang S, Zhang P, Yin N, Xu Z, Liu X, Wu A, Zhang X, Li Z, Zhang Z, Zhong T, Liu L, Shi Y, Dong J. Glioblastoma stem cell-derived exosomal miR-374b-3p promotes tumor angiogenesis and progression through inducing M2 macrophages polarization. iScience 2024; 27:109270. [PMID: 38487014 PMCID: PMC10937837 DOI: 10.1016/j.isci.2024.109270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/30/2024] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Glioblastoma stem cells (GSCs) reside in hypoxic periarteriolar niches of glioblastoma micro-environment, however, the crosstalk of GSCs with macrophages on regulating tumor angiogenesis and progression are not fully elucidated. GSCs-derived exosomes (GSCs-exos) are essential mediators during tumor immune-microenvironment remodeling initiated by GSCs, resulting in M2 polarization of tumor-associated macrophages (TAMs) as we reported previously. Our data disclosed aberrant upregulation of miR-374b-3p in both clinical glioblastoma specimens and human cell lines of GSCs. MiR-374b-3p level was high in GSCs-exos and can be internalized by macrophages. Mechanistically, GSCs exosomal miR-374b-3p induced M2 polarization of macrophages by downregulating phosphatase and tensin expression, thereby promoting migration and tube formation of vascular endothelial cells after coculture with M2 macrophages. Cumulatively, these data indicated that GSCs exosomal miR-374b-3p can enhance tumor angiogenesis by inducing M2 polarization of macrophages, as well as promote malignant progression of glioblastoma. Targeting exosomal miR-374b-3p may serve as a potential target against glioblastoma.
Collapse
Affiliation(s)
- Shilu Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Peng Zhang
- Department of Neurosurgery, Rugao People’s Hospital, RuGao 226500, China
| | - Nanheng Yin
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Zhipeng Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Xinglei Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Anyi Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Xiaopei Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Zengyang Li
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Zhicheng Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Tao Zhong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Yan Shi
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Jun Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| |
Collapse
|
13
|
Lin TY, Jia JS, Luo WR, Lin XL, Xiao SJ, Yang J, Xia JW, Zhou C, Zhou ZH, Lin SJ, Li QW, Yang ZZ, Lei Y, Yang WQ, Shen HF, Huang SH, Wang SC, Chen LB, Yang YL, Xue SW, Li YL, Dai GQ, Zhou Y, Li YC, Wei F, Rong XX, Luo XJ, Zhao BX, Huang WH, Xiao D, Sun Y. ThermomiR-377-3p-induced suppression of Cirbp expression is required for effective elimination of cancer cells and cancer stem-like cells by hyperthermia. J Exp Clin Cancer Res 2024; 43:62. [PMID: 38419081 PMCID: PMC10903011 DOI: 10.1186/s13046-024-02983-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND In recent years, the development of adjunctive therapeutic hyperthermia for cancer therapy has received considerable attention. However, the mechanisms underlying hyperthermia resistance are still poorly understood. In this study, we investigated the roles of cold‑inducible RNA binding protein (Cirbp) in regulating hyperthermia resistance and underlying mechanisms in nasopharyngeal carcinoma (NPC). METHODS CCK-8 assay, colony formation assay, tumor sphere formation assay, qRT-PCR, Western blot were employed to examine the effects of hyperthermia (HT), HT + oridonin(Ori) or HT + radiotherapy (RT) on the proliferation and stemness of NPC cells. RNA sequencing was applied to gain differentially expressed genes upon hyperthermia. Gain-of-function and loss-of-function experiments were used to evaluate the effects of RNAi-mediated Cirbp silencing or Cirbp overexpression on the sensitivity or resistance of NPC cells and cancer stem-like cells to hyperthermia by CCK-8 assay, colony formation assay, tumorsphere formation assay and apoptosis assay, and in subcutaneous xenograft animal model. miRNA transient transfection and luciferase reporter assay were used to demonstrate that Cirbp is a direct target of miR-377-3p. The phosphorylation levels of key members in ATM-Chk2 and ATR-Chk1 pathways were detected by Western blot. RESULTS Our results firstly revealed that hyperthermia significantly attenuated the stemness of NPC cells, while combination treatment of hyperthermia and oridonin dramatically increased the killing effect on NPC cells and cancer stem cell (CSC)‑like population. Moreover, hyperthermia substantially improved the sensitivity of radiation‑resistant NPC cells and CSC‑like cells to radiotherapy. Hyperthermia noticeably suppressed Cirbp expression in NPC cells and xenograft tumor tissues. Furthermore, Cirbp inhibition remarkably boosted anti‑tumor‑killing activity of hyperthermia against NPC cells and CSC‑like cells, whereas ectopic expression of Cirbp compromised tumor‑killing effect of hyperthermia on these cells, indicating that Cirbp overexpression induces hyperthermia resistance. ThermomiR-377-3p improved the sensitivity of NPC cells and CSC‑like cells to hyperthermia in vitro by directly suppressing Cirbp expression. More importantly, our results displayed the significantly boosted sensitization of tumor xenografts to hyperthermia by Cirbp silencing in vivo, but ectopic expression of Cirbp almost completely counteracted hyperthermia-mediated tumor cell-killing effect against tumor xenografts in vivo. Mechanistically, Cirbp silencing-induced inhibition of DNA damage repair by inactivating ATM-Chk2 and ATR-Chk1 pathways, decrease in stemness and increase in cell death contributed to hyperthermic sensitization; conversely, Cirbp overexpression-induced promotion of DNA damage repair, increase in stemness and decrease in cell apoptosis contributed to hyperthermia resistance. CONCLUSION Taken together, these findings reveal a previously unrecognized role for Cirbp in positively regulating hyperthermia resistance and suggest that thermomiR-377-3p and its target gene Cirbp represent promising targets for therapeutic hyperthermia.
Collapse
Affiliation(s)
- Tao-Yan Lin
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun-Shuang Jia
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Wei-Ren Luo
- Cancer Research Institute, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | - Xiao-Lin Lin
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Sheng-Jun Xiao
- Department of Pathology, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Jie Yang
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Imaging, Central Hospital of Shaoyang, Shaoyang, 422000, China
| | - Jia-Wei Xia
- The Third People's Hospital of Kunming (The Sixth Affiliated Hospital of Dali University), Kunming, 650041, China
| | - Chen Zhou
- Department of Pathology, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Zhi-Hao Zhou
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shu-Jun Lin
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qi-Wen Li
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhi-Zhi Yang
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ye Lei
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wen-Qing Yang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Hong-Fen Shen
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shi-Hao Huang
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Sheng-Chun Wang
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Lin-Bei Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Yu-Lin Yang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Shu-Wen Xue
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yong-Long Li
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guan-Qi Dai
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ying Zhou
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ying-Chun Li
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fang Wei
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiao-Xiang Rong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guang‑zhou, 510515, China
| | - Xiao-Jun Luo
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Bing-Xia Zhao
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Wen-Hua Huang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510000, China.
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524001, China.
| | - Dong Xiao
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Guangzhou Southern Medical Laboratory Animal Sci.&Tech. Co.,Ltd, Guangzhou, 510515, China.
- National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Department of Stomatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Yan Sun
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
14
|
Hasan U, Rajakumara E, Giri J. Reversal of Multidrug Resistance by the Synergistic Effect of Reversan and Hyperthermia to Potentiate the Chemotherapeutic Response of Doxorubicin in Glioblastoma and Glioblastoma Stem Cells. ACS APPLIED BIO MATERIALS 2023; 6:5399-5413. [PMID: 37975516 DOI: 10.1021/acsabm.3c00644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
The glioblastoma stem cell (GSC) population in glioblastoma multiforme (GBM) poses major complication in clinical oncology owing to increased resistance to chemotherapeutic drugs, thereby limiting treatment in patients with recurring glioblastoma. To completely eradicate glioblastoma, a single therapy module is not enough; therefore, there is a need to develop a multimodal approach to eliminate bulk tumors along with the CSC population. With an aim to target transporters associated with multidrug resistance (MDR), such as P-glycoprotein (P-gp), a small-molecule inhibitor, reversan (RV) was used along with multifunctional magnetic nanoparticles (MNPs) for hyperthermia (HT) therapy and targeted drug delivery. Higher efflux of free doxorubicin (Dox) from the cells was stabilized by encapsulation in PPS-MnFe nanoparticles, whose physicochemical properties were determined by dynamic light scattering (DLS) and transmission electron microscopy (TEM). Treatment with RV also enhanced the cellular uptake of PPS-MnFe-Dox, whereas RV and magnetic hyperthermia (MHT) together showed prolonged retention of fluorescence dye, Rhodamine123 (R123), in glioblastoma cells compared with individual treatment. Overall, in this work, we demonstrated the synergistic action of RV and HT to combat MDR in GBM and GSCs, and chemo-hyperthermia therapy enhanced the cytotoxic effect of the chemotherapeutic drug Dox (with lower effective concentration) and induced a higher degree of apoptosis compared to single-drug dosage.
Collapse
Affiliation(s)
- Uzma Hasan
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Hyderabad, Telangana 502284, India
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Hyderabad, Telangana 502284, India
| | - Eerappa Rajakumara
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Hyderabad, Telangana 502284, India
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Hyderabad, Telangana 502284, India
| |
Collapse
|
15
|
Tewari AB, Saini A, Sharma D. Extirpating the cancer stem cell hydra: Differentiation therapy and Hyperthermia therapy for targeting the cancer stem cell hierarchy. Clin Exp Med 2023; 23:3125-3145. [PMID: 37093450 DOI: 10.1007/s10238-023-01066-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 04/02/2023] [Indexed: 04/25/2023]
Abstract
Ever since the discovery of cancer stem cells (CSCs), they have progressively attracted more attention as a therapeutic target. Like the mythical hydra, this subpopulation of cells seems to contribute to cancer immortality, spawning more cells each time that some components of the cancer cell hierarchy are destroyed. Traditional modalities focusing on cancer treatment have emphasized apoptosis as a route to eliminate the tumor burden. A major problem is that cancer cells are often in varying degrees of dedifferentiation contributing to what is known as the CSCs hierarchy and cells which are known to be resistant to conventional therapy. Differentiation therapy is an experimental therapeutic modality aimed at the conversion of malignant phenotype to a more benign one. Hyperthermia therapy (HT) is a modality exploiting the changes induced in cells by the application of heat produced to aid in cancer therapy. While differentiation therapy has been successfully employed in the treatment of acute myeloid leukemia, it has not been hugely successful for other cancer types. Mounting evidence suggests that hyperthermia therapy may greatly augment the effects of differentiation therapy while simultaneously overcoming many of the hard-to-treat facets of recurrent tumors. This review summarizes the progress made so far in integrating hyperthermia therapy with existing modules of differentiation therapy. The focus is on studies related to the successful application of both hyperthermia and differentiation therapy when used alone or in conjunction for hard-to-treat cancer cell niche with emphasis on combined approaches to target the CSCs hierarchy.
Collapse
Affiliation(s)
- Amit B Tewari
- Institute of Nano Science and Technology (INST), Knowledge City, Sector 81, Mohali, Punjab, 140306, India
| | - Anamika Saini
- Institute of Nano Science and Technology (INST), Knowledge City, Sector 81, Mohali, Punjab, 140306, India
| | - Deepika Sharma
- Institute of Nano Science and Technology (INST), Knowledge City, Sector 81, Mohali, Punjab, 140306, India.
| |
Collapse
|
16
|
Wang X, Zhang H, Chen X, Wu C, Ding K, Sun G, Luo Y, Xiang D. Overcoming tumor microenvironment obstacles: Current approaches for boosting nanodrug delivery. Acta Biomater 2023; 166:42-68. [PMID: 37257574 DOI: 10.1016/j.actbio.2023.05.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
In order to achieve targeted delivery of anticancer drugs, efficacy improvement, and side effect reduction, various types of nanoparticles are employed. However, their therapeutic effects are not ideal. This phenomenon is caused by tumor microenvironment abnormalities such as abnormal blood vessels, elevated interstitial fluid pressure, and dense extracellular matrix that affect nanoparticle penetration into the tumor's interstitium. Furthermore, nanoparticle properties including size, charge, and shape affect nanoparticle transport into tumors. This review comprehensively goes over the factors hindering nanoparticle penetration into tumors and describes methods for improving nanoparticle distribution by remodeling the tumor microenvironment and optimizing nanoparticle physicochemical properties. Finally, a critical analysis of future development of nanodrug delivery in oncology is further discussed. STATEMENT OF SIGNIFICANCE: This article reviews the factors that hinder the distribution of nanoparticles in tumors, and describes existing methods and approaches for improving the tumor accumulation from the aspects of remodeling the tumor microenvironment and optimizing the properties of nanoparticles. The description of the existing methods and approaches is followed by highlighting their advantages and disadvantages and put forward possible directions for the future researches. At last, the challenges of improving tumor accumulation in nanomedicines design were also discussed. This review will be of great interest to the broad readers who are committed to delivering nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Xiaohui Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Hong Zhang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Xiaohui Chen
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Chunrong Wu
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Ke Ding
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Guiyin Sun
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Debing Xiang
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| |
Collapse
|
17
|
Chen C, Ren A, Yi Q, Cai J, Khan M, Lin Y, Huang Z, Lin J, Zhang J, Liu W, Xu A, Tian Y, Yuan Y, Zheng R. Therapeutic hyperthermia regulates complement C3 activation and suppresses tumor development through HSPA5/NFκB/CD55 pathway in nasopharyngeal carcinoma. Clin Exp Immunol 2023; 213:221-234. [PMID: 37249005 PMCID: PMC10361742 DOI: 10.1093/cei/uxad060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/09/2023] [Accepted: 05/29/2023] [Indexed: 05/31/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is endemic in Southern China and Southeast Asia. Hyperthermia is widely used in combination with chemotherapy and radiotherapy to enhance therapeutic efficacy in NPC treatment, but the underlying anti-tumor mechanisms of hyperthermia remain unclear. Complement C3 has been reported to participate in the activation of immune system in the tumor microenvironment, leading to tumor growth inhibition. In this study, we aimed to explore the effect and mechanisms of hyperthermia and investigate the functional role of complement C3 in NPC hyperthermia therapy (HT). The serum levels of complement C3 before and after hyperthermia therapy in patients with NPC were analyzed. NPC cell lines SUNE1 and HONE1 were used for in vitro experiment to evaluate the function of complement C3 and HT on cell proliferation and apoptosis. SUNE1 xenograft mouse model was established and tumor-bearing mice were treated in water bath at a constant temperature of 43°C. Tumor samples were collected at different time points to verify the expression of complement C3 by immunohistochemical staining and western blot. The differential expressed genes after hyperthermia were analyzed by using RNA sequencing. We found that complement could enhance hyperthermia effect on suppressing proliferation and promoting apoptosis of tumor cells in NPC. Hyperthermia decreased the mRNA expression of complement C3 in tumor cells, but promoted the aggregation and activation circulating C3 in NPC tumor tissue. By using in vitro hyperthermia-treated NPC cell lines and SUNE1 xenograft tumor-bearing mice, we found that the expression of heat shock protein 5 (HSPA5) was significantly upregulated. Knockdown of HSPA5 abrogated the anti-tumor effect of hyperthermia. Moreover, we demonstrated that hyperthermia downregulated CD55 expression via HSPA5/NFκB (P65) signaling and activated complement cascade. Our findings suggest that therapeutic hyperthermia regulates complement C3 activation and suppresses tumor development via HSPA5/NFκB/CD55 pathway in NPC.
Collapse
Affiliation(s)
- Chengcong Chen
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Anbang Ren
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Qi Yi
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Jiazuo Cai
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Muhammad Khan
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Yunen Lin
- Department of Pathology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhong Huang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Jie Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Wei Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Anan Xu
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Yunhong Tian
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - YaWei Yuan
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Ronghui Zheng
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
18
|
Kwon S, Jung S, Baek SH. Combination Therapy of Radiation and Hyperthermia, Focusing on the Synergistic Anti-Cancer Effects and Research Trends. Antioxidants (Basel) 2023; 12:antiox12040924. [PMID: 37107299 PMCID: PMC10136118 DOI: 10.3390/antiox12040924] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/03/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Despite significant therapeutic advances, the toxicity of conventional therapies remains a major obstacle to their application. Radiation therapy (RT) is an important component of cancer treatment. Therapeutic hyperthermia (HT) can be defined as the local heating of a tumor to 40-44 °C. Both RT and HT have the advantage of being able to induce and regulate oxidative stress. Here, we discuss the effects and mechanisms of RT and HT based on experimental research investigations and summarize the results by separating them into three phases. Phase (1): RT + HT is effective and does not provide clear mechanisms; phase (2): RT + HT induces apoptosis via oxygenation, DNA damage, and cell cycle arrest; phase (3): RT + HT improves immunological responses and activates immune cells. Overall, RT + HT is an effective cancer modality complementary to conventional therapy and stimulates the immune response, which has the potential to improve cancer treatments, including immunotherapy, in the future.
Collapse
Affiliation(s)
- Seeun Kwon
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea
| | - Sumin Jung
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea
| | - Seung Ho Baek
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea
| |
Collapse
|
19
|
Rivera D, Schupper AJ, Bouras A, Anastasiadou M, Kleinberg L, Kraitchman DL, Attaluri A, Ivkov R, Hadjipanayis CG. Neurosurgical Applications of Magnetic Hyperthermia Therapy. Neurosurg Clin N Am 2023; 34:269-283. [PMID: 36906333 PMCID: PMC10726205 DOI: 10.1016/j.nec.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Magnetic hyperthermia therapy (MHT) is a highly localized form of hyperthermia therapy (HT) that has been effective in treating various forms of cancer. Many clinical and preclinical studies have applied MHT to treat aggressive forms of brain cancer and assessed its role as a potential adjuvant to current therapies. Initial results show that MHT has a strong antitumor effect in animal studies and a positive association with overall survival in human glioma patients. Although MHT is a promising therapy with the potential to be incorporated into the future treatment of brain cancer, significant advancement of current MHT technology is required.
Collapse
Affiliation(s)
- Daniel Rivera
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA 15213, USA; Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15232, USA
| | - Alexander J Schupper
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Alexandros Bouras
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA 15213, USA; Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15232, USA
| | - Maria Anastasiadou
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Lawrence Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, 1550 Orleans Street, Baltimore, MD 21231-5678, USA
| | - Dara L Kraitchman
- Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | - Anilchandra Attaluri
- Department of Mechanical Engineering, The Pennsylvania State University, 777 West Harrisburg Pike Middletown, PA 17057, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, 1550 Orleans Street, Baltimore, MD 21231-5678, USA; Department of Oncology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21231-5678, USA; Department of Mechanical Engineering, Johns Hopkins University, Whiting School of Engineering, 3400 North Charles Street, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Whiting School of Engineering, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Constantinos G Hadjipanayis
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA 15213, USA; Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15232, USA.
| |
Collapse
|
20
|
Dhawan U, Tseng CL, Wu PH, Liao MY, Wang HY, Wu KCW, Chung RJ. Theranostic doxorubicin encapsulated FeAu alloy@metal-organic framework nanostructures enable magnetic hyperthermia and medical imaging in oral carcinoma. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 48:102652. [PMID: 36623714 DOI: 10.1016/j.nano.2023.102652] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/08/2023]
Abstract
Metal-organic frameworks (MOFs) have emerged as attractive candidates in cancer theranostics due to their ability to envelop magnetic nanoparticles, resulting in reduced cytotoxicity and high porosity, enabling chemodrug encapsulation. Here, FeAu alloy nanoparticles (FeAu NPs) are synthesized and coated with MIL-100(Fe) MOFs to fabricate FeAu@MOF nanostructures. We encapsulated Doxorubicin within the nanostructures and evaluated the suitability of this platform for medical imaging and cancer theranostics. FeAu@MOF nanostructures (FeAu@MIL-100(Fe)) exhibited superparamagnetism, magnetic hyperthermia behavior and displayed DOX encapsulation and release efficiency of 69.95 % and 97.19 %, respectively, when stimulated with alternating magnetic field (AMF). In-vitro experiments showed that AMF-induced hyperthermia resulted in 90 % HSC-3 oral squamous carcinoma cell death, indicating application in cancer theranostics. Finally, in an in-vivo mouse model, FeAu@MOF nanostructures improved image contrast, reduced tumor volume by 30-fold and tumor weight by 10-fold, which translated to enhancement in cumulative survival, highlighting the prospect of this platform for oral cancer treatment.
Collapse
Affiliation(s)
- Udesh Dhawan
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8LT, UK
| | - Ching-Li Tseng
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City 110, Taiwan; International Ph. D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City 110, Taiwan; Research Center of Biomedical Device, College of Biomedical Engineering, Taipei Medical University, Taipei City 110, Taiwan; International Ph. D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan
| | - Ping-Hsuan Wu
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), Taipei 106344, Taiwan
| | - Mei-Yi Liao
- Department of Applied Chemistry, National Pingtung University, Pingtung 90003, Taiwan
| | - Huey-Yuan Wang
- Department of Stomatology, MacKay Memorial Hospital, Taipei 104217, Taiwan.
| | - Kevin C-W Wu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan; Institute of Biomedical Engineering & Nanomedicine, National Health Research Institute, Keyan Road, Zhunan, Miaoli City 350, Taiwan; Center of Atomic Initiative for New Materials (Al-MAT), National Taiwan University, Taipei 106216, Taiwan.
| | - Ren-Jei Chung
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), Taipei 106344, Taiwan.
| |
Collapse
|
21
|
Sharma A, Jangam A, Shen JLY, Ahmad A, Arepally N, Rodriguez B, Borrello J, Bouras A, Kleinberg L, Ding K, Hadjipanayis C, Kraitchman DL, Ivkov R, Attaluri A. Validation of a Temperature-Feedback Controlled Automated Magnetic Hyperthermia Therapy Device. Cancers (Basel) 2023; 15:327. [PMID: 36672278 PMCID: PMC9856953 DOI: 10.3390/cancers15020327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023] Open
Abstract
We present in vivo validation of an automated magnetic hyperthermia therapy (MHT) device that uses real-time temperature input measured at the target to control tissue heating. MHT is a thermal therapy that uses heat generated by magnetic materials exposed to an alternating magnetic field. For temperature monitoring, we integrated a commercial fiber optic temperature probe containing four gallium arsenide (GaAs) temperature sensors. The controller device used temperature from the sensors as input to manage power to the magnetic field applicator. We developed a robust, multi-objective, proportional-integral-derivative (PID) algorithm to control the target thermal dose by modulating power delivered to the magnetic field applicator. The magnetic field applicator was a 20 cm diameter Maxwell-type induction coil powered by a 120 kW induction heating power supply operating at 160 kHz. Finite element (FE) simulations were performed to determine values of the PID gain factors prior to verification and validation trials. Ex vivo verification and validation were conducted in gel phantoms and sectioned bovine liver, respectively. In vivo validation of the controller was achieved in a canine research subject following infusion of magnetic nanoparticles (MNPs) into the brain. In all cases, performance matched controller design criteria, while also achieving a thermal dose measured as cumulative equivalent minutes at 43 °C (CEM43) 60 ± 5 min within 30 min.
Collapse
Affiliation(s)
- Anirudh Sharma
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Avesh Jangam
- Department of Mechanical Engineering, School of Science, Engineering, and Technology, The Pennsylvania State University—Harrisburg, Harrisburg, PA 17057, USA
| | - Julian Low Yung Shen
- Department of Mechanical Engineering, School of Science, Engineering, and Technology, The Pennsylvania State University—Harrisburg, Harrisburg, PA 17057, USA
| | - Aiman Ahmad
- Department of Mechanical Engineering, School of Science, Engineering, and Technology, The Pennsylvania State University—Harrisburg, Harrisburg, PA 17057, USA
| | - Nageshwar Arepally
- Department of Mechanical Engineering, School of Science, Engineering, and Technology, The Pennsylvania State University—Harrisburg, Harrisburg, PA 17057, USA
| | - Benjamin Rodriguez
- Sinai BioDesign, Mount Sinai Hospital, New York, NY 10029, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joseph Borrello
- Sinai BioDesign, Mount Sinai Hospital, New York, NY 10029, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexandros Bouras
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Lawrence Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kai Ding
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Constantinos Hadjipanayis
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Dara L. Kraitchman
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anilchandra Attaluri
- Department of Mechanical Engineering, School of Science, Engineering, and Technology, The Pennsylvania State University—Harrisburg, Harrisburg, PA 17057, USA
| |
Collapse
|
22
|
Wang K, Jalil AT, Saleh MM, Talaei S, Wang L. Glutathione (GSH) conjugated Bi2S3 NPs as a promising radiosensitizer against glioblastoma cancer cells. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-022-02592-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
23
|
Hersh AM, Bhimreddy M, Weber-Levine C, Jiang K, Alomari S, Theodore N, Manbachi A, Tyler BM. Applications of Focused Ultrasound for the Treatment of Glioblastoma: A New Frontier. Cancers (Basel) 2022; 14:4920. [PMID: 36230843 PMCID: PMC9563027 DOI: 10.3390/cancers14194920] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/21/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive primary astrocytoma associated with short overall survival. Treatment for GBM primarily consists of maximal safe surgical resection, radiation therapy, and chemotherapy using temozolomide. Nonetheless, recurrence and tumor progression is the norm, driven by tumor stem cell activity and a high mutational burden. Focused ultrasound (FUS) has shown promising results in preclinical and clinical trials for treatment of GBM and has received regulatory approval for the treatment of other neoplasms. Here, we review the range of applications for FUS in the treatment of GBM, which depend on parameters, including frequency, power, pulse duration, and duty cycle. Low-intensity FUS can be used to transiently open the blood-brain barrier (BBB), which restricts diffusion of most macromolecules and therapeutic agents into the brain. Under guidance from magnetic resonance imaging, the BBB can be targeted in a precise location to permit diffusion of molecules only at the vicinity of the tumor, preventing side effects to healthy tissue. BBB opening can also be used to improve detection of cell-free tumor DNA with liquid biopsies, allowing non-invasive diagnosis and identification of molecular mutations. High-intensity FUS can cause tumor ablation via a hyperthermic effect. Additionally, FUS can stimulate immunological attack of tumor cells, can activate sonosensitizers to exert cytotoxic effects on tumor tissue, and can sensitize tumors to radiation therapy. Finally, another mechanism under investigation, known as histotripsy, produces tumor ablation via acoustic cavitation rather than thermal effects.
Collapse
Affiliation(s)
- Andrew M. Hersh
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Meghana Bhimreddy
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Carly Weber-Levine
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kelly Jiang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Safwan Alomari
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Nicholas Theodore
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Amir Manbachi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Mechanical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Electrical and Computer Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Betty M. Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
24
|
Redr J, Pokorny T, Drizdal T, Fiser O, Brunat M, Vrba J, Vrba D. Microwave Hyperthermia of Brain Tumors: A 2D Assessment Parametric Numerical Study. SENSORS (BASEL, SWITZERLAND) 2022; 22:6115. [PMID: 36015874 PMCID: PMC9416291 DOI: 10.3390/s22166115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 06/15/2023]
Abstract
Due to the clinically proven benefit of hyperthermia treatments if added to standard cancer therapies for various tumor sites and the recent development of non-invasive temperature measurements using magnetic resonance systems, the hyperthermia community is convinced that it is a time when even patients with brain tumors could benefit from regional microwave hyperthermia, even if they are the subject of a treatment to a vital organ. The purpose of this study was to numerically analyze the ability to achieve a therapeutically relevant constructive superposition of electromagnetic (EM) waves in the treatment of hyperthermia targets within the brain. We evaluated the effect of the target size and position, operating frequency, and the number of antenna elements forming the phased array applicator on the treatment quality. In total, 10 anatomically realistic 2D human head models were considered, in which 10 circular hyperthermia targets with diameters of 20, 25, and 30 mm were examined. Additionally, applicators with 8, 12, 16, and 24 antenna elements and operating frequencies of 434, 650, 915, and 1150 MHz, respectively, were analyzed. For all scenarios considered (4800 combinations), the EM field distributions of individual antenna elements were calculated and treatment planning was performed. Their quality was evaluated using parameters applied in clinical practice, i.e., target coverage (TC) and the target to hot-spot quotient (THQ). The 12-antenna phased array system operating at 434 MHz was the best candidate among all tested systems for HT treatments of glioblastoma tumors. The 12 antenna elements met all the requirements to cover the entire target area; an additional increase in the number of antenna elements did not have a significant effect on the treatment quality.
Collapse
|
25
|
Hersh AM, Gaitsch H, Alomari S, Lubelski D, Tyler BM. Molecular Pathways and Genomic Landscape of Glioblastoma Stem Cells: Opportunities for Targeted Therapy. Cancers (Basel) 2022; 14:3743. [PMID: 35954407 PMCID: PMC9367289 DOI: 10.3390/cancers14153743] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive tumor of the central nervous system categorized by the World Health Organization as a Grade 4 astrocytoma. Despite treatment with surgical resection, adjuvant chemotherapy, and radiation therapy, outcomes remain poor, with a median survival of only 14-16 months. Although tumor regression is often observed initially after treatment, long-term recurrence or progression invariably occurs. Tumor growth, invasion, and recurrence is mediated by a unique population of glioblastoma stem cells (GSCs). Their high mutation rate and dysregulated transcriptional landscape augment their resistance to conventional chemotherapy and radiation therapy, explaining the poor outcomes observed in patients. Consequently, GSCs have emerged as targets of interest in new treatment paradigms. Here, we review the unique properties of GSCs, including their interactions with the hypoxic microenvironment that drives their proliferation. We discuss vital signaling pathways in GSCs that mediate stemness, self-renewal, proliferation, and invasion, including the Notch, epidermal growth factor receptor, phosphatidylinositol 3-kinase/Akt, sonic hedgehog, transforming growth factor beta, Wnt, signal transducer and activator of transcription 3, and inhibitors of differentiation pathways. We also review epigenomic changes in GSCs that influence their transcriptional state, including DNA methylation, histone methylation and acetylation, and miRNA expression. The constituent molecular components of the signaling pathways and epigenomic regulators represent potential sites for targeted therapy, and representative examples of inhibitory molecules and pharmaceuticals are discussed. Continued investigation into the molecular pathways of GSCs and candidate therapeutics is needed to discover new effective treatments for GBM and improve survival.
Collapse
Affiliation(s)
- Andrew M. Hersh
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (H.G.); (S.A.); (D.L.)
| | - Hallie Gaitsch
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (H.G.); (S.A.); (D.L.)
- NIH Oxford-Cambridge Scholars Program, Wellcome—MRC Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK
| | - Safwan Alomari
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (H.G.); (S.A.); (D.L.)
| | - Daniel Lubelski
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (H.G.); (S.A.); (D.L.)
| | - Betty M. Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (H.G.); (S.A.); (D.L.)
| |
Collapse
|
26
|
Interaction of Radiotherapy and Hyperthermia with the Immune System: a Brief Current Overview. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-022-00215-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Abstract
Purpose of Review
This review focuses on the opposing effects on the immune system of radiotherapy (RT) and the consequences for combined cancer treatment strategies of RT with immunotherapies, including hyperthermia (HT). How RT and HT might affect cancer stem cell populations is also briefly outlined in this context.
Recent Findings
RT is one of the crucial standard cancer therapies. Most patients with solid tumors receive RT for curative and palliative purposes in the course of their disease. RT achieves a local tumor control by inducing DNA damage which can lead to tumor cell death. In recent years, it has become evident that RT does not only have local effects, but also systemic effects which involves induction of anti-tumor immunity and possible alteration of the immunosuppressive properties of the tumor microenvironment. Though, often RT alone is not able to induce potent anti-tumor immune responses since the effects of RT on the immune system can be both immunostimulatory and immunosuppressive.
Summary
RT with additional therapies such as HT and immune checkpoint inhibitors (ICI) are promising approaches to induce anti-tumor immunity effectively. HT is not only a potent sensitizer for RT, but it might also improve the efficacy of RT and certain chemotherapeutic agents (CT) by additionally sensitizing resistant cancer stem cells (CSCs).
Graphical abstract
Collapse
|
27
|
Munir MU. Nanomedicine Penetration to Tumor: Challenges, and Advanced Strategies to Tackle This Issue. Cancers (Basel) 2022; 14:cancers14122904. [PMID: 35740570 PMCID: PMC9221319 DOI: 10.3390/cancers14122904] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/01/2023] Open
Abstract
Nanomedicine has been under investigation for several years to improve the efficiency of chemotherapeutics, having minimal pharmacological effects clinically. Ineffective tumor penetration is mediated by tumor environments, including limited vascular system, rising cancer cells, higher interstitial pressure, and extra-cellular matrix, among other things. Thus far, numerous methods to increase nanomedicine access to tumors have been described, including the manipulation of tumor micro-environments and the improvement of nanomedicine characteristics; however, such outdated approaches still have shortcomings. Multi-functional convertible nanocarriers have recently been developed as an innovative nanomedicine generation with excellent tumor infiltration abilities, such as tumor-penetrating peptide-mediated transcellular transport. The developments and limitations of nanomedicines, as well as expectations for better outcomes of tumor penetration, are discussed in this review.
Collapse
Affiliation(s)
- Muhammad Usman Munir
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia
| |
Collapse
|
28
|
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
|
29
|
Li X, Duan S, Zheng Y, Yang Y, Wang L, Li X, Zhang Q, Thorne RF, Li W, Yang D. Hyperthermia inhibits growth of nasopharyngeal carcinoma through degradation of c-Myc. Int J Hyperthermia 2022; 39:358-371. [PMID: 35184661 DOI: 10.1080/02656736.2022.2038282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Xiaole Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Department of Radiotherapy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shichao Duan
- Department of Pathology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingjuan Zheng
- Department of Radiotherapy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongqiang Yang
- Department of Radiotherapy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Wang
- Department of Pathology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinqiang Li
- Department of Pathology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qing Zhang
- Translational Research Institute, Henan Provincial People’s Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Rick F. Thorne
- Translational Research Institute, Henan Provincial People’s Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Wencai Li
- Department of Pathology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Daoke Yang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Department of Radiotherapy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
30
|
Tani T, Tojo N, Ohnishi K. Preferential radiosensitization to glioblastoma cancer stem cell‑like cells by a Hsp90 inhibitor, N‑vinylpyrrolidone‑AUY922. Oncol Lett 2022; 23:102. [PMID: 35154433 PMCID: PMC8822487 DOI: 10.3892/ol.2022.13222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/22/2021] [Indexed: 11/06/2022] Open
Abstract
The present study examined the radiosensitization induced by a heat shock protein 90 inhibitor, N-vinylpyrrolidone (NVP)-AUY922, in CD133-positive cells in a hypoxic area of T98G spheroids. CD133-positive cells that are induced in the hypoxic microenvironment of spheroids have previously been reported to exhibit cancer stem cell-like properties. The present study used CD133-positive cells from a glioblastoma cell line (T98G) as cancer stem cell-like cells. CD133-positive and negative cells were sorted from T98G spheroids using fluorescence-activated cell sorting and used for colony formation assay. Colony formation assay results indicated that NVP-AUY922 enhanced radiosensitivity more strongly in CD133-positive cells compared with CD133-negative cells. This result showed that NVP-AUY922 was a preferential radiosensitization candidate targeting glioblastoma cancer stem cells. The mechanisms underlying radiosensitization by NVP-AUY922 are discussed in relation to the properties of cancer stem cells. Overall, HIF-1α inhibition by NVP-AUY922 may induce higher sensitization of cancer stem cells to radiation.
Collapse
Affiliation(s)
- Toshiaki Tani
- Radiological Technology Section, QST Hospital, National Institutes for Quantum and Radiological Science and Technology, Chiba 263‑8555, Japan
| | - Naomi Tojo
- Department of Biology, Ibaraki Prefectural University of Health Sciences, Inashiki, Ibaraki 300‑0394, Japan
| | - Ken Ohnishi
- Department of Biology, Ibaraki Prefectural University of Health Sciences, Inashiki, Ibaraki 300‑0394, Japan
| |
Collapse
|
31
|
Zhang M, Rodrigues A, Zhou Q, Li G. Focused ultrasound: growth potential and future directions in neurosurgery. J Neurooncol 2022; 156:23-32. [PMID: 34410576 DOI: 10.1007/s11060-021-03820-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/31/2021] [Indexed: 12/18/2022]
Abstract
Over the past two decades, vast improvements in focused ultrasound (FUS) technology have made the therapy an exciting addition to the neurosurgical armamentarium. In this time period, FUS has gained US Food and Drug Administration (FDA) approval for the treatment of two neurological disorders, and ongoing efforts seek to expand the lesion profile that is amenable to ultrasonic intervention. In the following review, we highlight future applications for FUS therapy and compare its potential role against established technologies, including deep brain stimulation and stereotactic radiosurgery. Particular attention is paid to tissue ablation, blood-brain-barrier opening, and gene therapy. We also address technical and infrastructural challenges involved with FUS use and summarize the hurdles that must be overcome before FUS becomes widely accepted in the neurosurgical community.
Collapse
Affiliation(s)
- Michael Zhang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.
- Center for Academic Medicine, Neurosurgery, Stanford University School of Medicine, MC 5327, 453 Quarry Road, Palo Alto, CA, 94304, USA.
| | - Adrian Rodrigues
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Quan Zhou
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Gordon Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
32
|
Abstract
BACKGROUND Glioblastoma stem cells (GSCs) and their interplay with tumor-associated macrophages (TAMs) are responsible for malignant growth and tumor recurrence of glioblastoma multiforme (GBM), but the underlying mechanisms are largely unknown. METHODS Cell viability, stemness, migration, and invasion were measured in GSCs after the knockdown of upstream stimulating factor 1 (USF1). Luciferase assay and chromatin immunoprecipitation qPCR were performed to determine the regulation of CD90 by USF1. Immunohistochemistry and immunofluorescent staining were used to examine the expression of USF1 and GSC markers, as well as the crosstalk between GSCs and TAMs. In addition, the interaction between GSCs and TAMs was confirmed using in vivo GBM models. RESULTS We show that USF1 promotes malignant glioblastoma phenotypes and GSCs-TAMs physical interaction by inducing CD90 expression. USF1 predicts a poor prognosis for glioma patients and is upregulated in patient-derived GSCs and glioblastoma cell lines. USF1 overexpression increases the proliferation, invasion, and neurosphere formation of GSCs and glioblastoma cell lines, while USF1 knockdown exerts an opposite effect. Further mechanistic studies reveal that USF1 promotes GSC stemness by directly regulating CD90 expression. Importantly, CD90 of GSCs functions as an anchor for physical interaction with macrophages. Additionally, the USF1/CD90 signaling axis supports the GSCs and TAMs adhesion and immunosuppressive feature of TAMs, which in turn enhance the stemness of GSCs. Moreover, the overexpression of CD90 restores the stemness property in USF1 knockdown GSCs and its immunosuppressive microenvironment. CONCLUSIONS Our findings indicate that the USF1/CD90 axis might be a potential therapeutic target for the treatment of glioblastoma.
Collapse
|
33
|
Pang S, Kapur A, Zhou K, Anastasiadis P, Ballirano N, Kim AJ, Winkles JA, Woodworth GF, Huang H. Nanoparticle-assisted, image-guided laser interstitial thermal therapy for cancer treatment. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1826. [PMID: 35735205 PMCID: PMC9540339 DOI: 10.1002/wnan.1826] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 11/18/2022]
Abstract
Laser interstitial thermal therapy (LITT) guided by magnetic resonance imaging (MRI) is a new treatment option for patients with brain and non-central nervous system (non-CNS) tumors. MRI guidance allows for precise placement of optical fiber in the tumor, while MR thermometry provides real-time monitoring and assessment of thermal doses during the procedure. Despite promising clinical results, LITT complications relating to brain tumor procedures, such as hemorrhage, edema, seizures, and thermal injury to nearby healthy tissues, remain a significant concern. To address these complications, nanoparticles offer unique prospects for precise interstitial hyperthermia applications that increase heat transport within the tumor while reducing thermal impacts on neighboring healthy tissues. Furthermore, nanoparticles permit the co-delivery of therapeutic compounds that not only synergize with LITT, but can also improve overall effectiveness and safety. In addition, efficient heat-generating nanoparticles with unique optical properties can enhance LITT treatments through improved real-time imaging and thermal sensing. This review will focus on (1) types of inorganic and organic nanoparticles for LITT; (2) in vitro, in silico, and ex vivo studies that investigate nanoparticles' effect on light-tissue interactions; and (3) the role of nanoparticle formulations in advancing clinically relevant image-guided technologies for LITT. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Implantable Materials and Surgical Technologies > Nanoscale Tools and Techniques in Surgery.
Collapse
Affiliation(s)
- Sumiao Pang
- Fischell Department of Bioengineering, University of Maryland at College ParkCollege ParkMarylandUSA
| | - Anshika Kapur
- Department of NeurosurgeryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Keri Zhou
- Fischell Department of Bioengineering, University of Maryland at College ParkCollege ParkMarylandUSA
| | - Pavlos Anastasiadis
- Department of NeurosurgeryUniversity of Maryland School of MedicineBaltimoreMarylandUSA,University of Maryland Marlene and Stewart Greenebaum Cancer CenterBaltimoreMarylandUSA
| | - Nicholas Ballirano
- Fischell Department of Bioengineering, University of Maryland at College ParkCollege ParkMarylandUSA
| | - Anthony J. Kim
- Department of NeurosurgeryUniversity of Maryland School of MedicineBaltimoreMarylandUSA,University of Maryland Marlene and Stewart Greenebaum Cancer CenterBaltimoreMarylandUSA
| | - Jeffrey A. Winkles
- Department of NeurosurgeryUniversity of Maryland School of MedicineBaltimoreMarylandUSA,University of Maryland Marlene and Stewart Greenebaum Cancer CenterBaltimoreMarylandUSA
| | - Graeme F. Woodworth
- Department of NeurosurgeryUniversity of Maryland School of MedicineBaltimoreMarylandUSA,University of Maryland Marlene and Stewart Greenebaum Cancer CenterBaltimoreMarylandUSA
| | - Huang‐Chiao Huang
- Fischell Department of Bioengineering, University of Maryland at College ParkCollege ParkMarylandUSA,University of Maryland Marlene and Stewart Greenebaum Cancer CenterBaltimoreMarylandUSA
| |
Collapse
|
34
|
Cheng B, Bing C, Chu TH, Alzahrani S, Pichardo S, Pike GB. Simultaneous Localized Brain Mild Hyperthermia and Blood-Brain Barrier Opening via Feedback-Controlled Transcranial MR-guided Focused Ultrasound and Microbubbles. IEEE Trans Biomed Eng 2021; 69:1880-1888. [PMID: 34813464 DOI: 10.1109/tbme.2021.3130164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Non-invasive methods to enhance drug delivery and efficacy in the brain have been pursued for decades. Focused ultrasound hyperthermia (HT) combined with thermosensitive therapeutics have been demonstrated promising in enhancing local drug delivery to solid tumors. We hypothesized that the presence of microbubbles (MBs) combined with transcranial MR-guided focused ultrasound (MRgFUS) could be used to reduce the ultrasound power required for HT while simultaneously increasing drug delivery by locally opening the blood-brain barrier (BBB). METHODS Transcranial HT (42 C, 10 min) was performed in wild-type mice using a small animal MRgFUS system incorporated into a 9.4T Bruker MR scanner, with infusions of saline or Definity MBs with doses of 20 or 100 l/kg/min (denoted as MB-20 and MB-100). MR thermometry data was continuously acquired as feedback for the ultrasound controller during the procedure. RESULTS Spatiotemporally precise transcranial HT was achieved in both saline and MB groups. A significant ultrasound power reduction (-45.7%, p = 0.006) was observed in the MB-20 group compared to saline. Localized BBB opening was achieved in MB groups confirmed by CE-T1w MR images. There were no structural abnormalities, edema, hemorrhage, or acute microglial activation in all groups, confirmed by T2w MR imaging and histology. CONCLUSION Our investigations showed that it is feasible and safe to achieve spatiotemporally precise brain HT at significantly reduced power and simultaneous localized BBB opening via transcranial MRgFUS and MBs. SIGNIFICANCE This study provides a new synergistic brain drug delivery method with clinical translation potential.
Collapse
|
35
|
Role of Laser Interstitial Thermal Therapy in the Management of Primary and Metastatic Brain Tumors. Curr Treat Options Oncol 2021; 22:108. [PMID: 34687357 DOI: 10.1007/s11864-021-00912-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2021] [Indexed: 10/20/2022]
Abstract
OPINION STATEMENT Laser interstitial thermal therapy (LITT) is a minimally invasive treatment option for brain tumors including glioblastoma, other primary central nervous system (CNS) neoplasms, metastases, and radiation necrosis. LITT employs a fiber optic coupled laser delivery probe stabilized via stereotaxis to deliver thermal energy that induces coagulative necrosis in tumors to achieve effective cytoreduction. LITT complements surgical resection, radiation treatment, tumor treating fields, and systemic therapy, especially in patients who are high risk for surgical resection due to tumor location in eloquent regions or poor functional status. These factors must be balanced with the increased rate of cerebral edema post LITT compared to surgical resection. LITT has also been shown to induce transient disruption of the blood-brain barrier (BBB), especially in the peritumoral region, which allows for enhanced CNS delivery of anti-neoplastic agents, thus greatly expanding the armamentarium against brain tumors to include highly effective anti-neoplastic agents that have poor BBB penetration. In addition, hyperthermia-induced immunogenic cell death is another secondary side effect of LITT that opens up immunotherapy as an attractive adjuvant treatment for brain tumors. Numerous large studies have demonstrated the safety and efficacy of LITT against various CNS tumors and as the literature continues to grow on this novel technique so will its indications.
Collapse
|
36
|
Combinatorial Effect of PLK1 Inhibition with Temozolomide and Radiation in Glioblastoma. Cancers (Basel) 2021; 13:cancers13205114. [PMID: 34680264 PMCID: PMC8533781 DOI: 10.3390/cancers13205114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary There is a critical need to identify readily translatable adjuncts to potentiate the dismal median survivals of only 15–20 months in glioblastoma (GBM) patients after standard of care, i.e., concurrent Temozolomide (TMZ) and radiation (XRT) therapy. Here we demonstrated that the Polo-like kinase 1 (PLK1) inhibitor volasertib, which has been employed in cancer clinical trials, has activity against GBM in the contexts of both as monotherapy and as an adjunct to standard of care (SOC). In addition to corroborating the known effects of volasertib, we found novel impacts of volasertib on mitochondrial membrane potential, ROS generation, persistent DNA damage and signaling pathways such as ERK/MAPK, AMPK and glucocorticoid receptor. Together these studies support the potential importance of PLK1 inhibitors as an adjunct to GBM SOC therapy that warrants further preclinical investigation. Abstract New strategies that improve median survivals of only ~15–20 months for glioblastoma (GBM) with the current standard of care (SOC) which is concurrent temozolomide (TMZ) and radiation (XRT) treatment are urgently needed. Inhibition of polo-like kinase 1 (PLK1), a multifunctional cell cycle regulator, overexpressed in GBM has shown therapeutic promise but has never been tested in the context of SOC. Therefore, we examined the mechanistic and therapeutic impact of PLK1 specific inhibitor (volasertib) alone and in combination with TMZ and/or XRT on GBM cells. We quantified the effects of volasertib alone and in combination with TMZ and/or XRT on GBM cell cytotoxicity/apoptosis, mitochondrial membrane potential (MtMP), reactive oxygen species (ROS), cell cycle, stemness, DNA damage, DNA repair genes, cellular signaling and in-vivo tumor growth. Volasertib alone and in combination with TMZ and/or XRT promoted apoptotic cell death, altered MtMP, increased ROS and G2/M cell cycle arrest. Combined volasertib and TMZ treatment reduced side population (SP) indicating activity against GBM stem-like cells. Volasertib combinatorial treatment also significantly increased DNA damage and reduced cell survival by inhibition of DNA repair gene expression and modulation of ERK/MAPK, AMPK and glucocorticoid receptor signaling. Finally, as observed in-vitro, combined volasertib and TMZ treatment resulted in synergistic inhibition of tumor growth in-vivo. Together these results identify new mechanisms of action for volasertib that provide a strong rationale for further investigation of PLK1 inhibition as an adjunct to current GBM SOC therapy.
Collapse
|
37
|
Knackstedt R, Smile T, Yu J, Gastman BR. Non-Operative Options for Loco-regional Melanoma. Clin Plast Surg 2021; 48:631-642. [PMID: 34503723 DOI: 10.1016/j.cps.2021.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Malignant melanoma is the 5th most common cancer and stage IV melanoma accounts for approximately 4% of new melanoma diagnoses in the United States. The prognosis for regionally advanced disease is poor, but there have been numerous recent advances in the medical management of melanoma in-transit metastases. The goal of this paper is to review currently accepted treatment options for in-transit metastases and introduce emerging therapies. Therapies to be discussed include limb perfusion and infusion, immunotherapy, checkpoint inhibitors, and radiation therapy.
Collapse
Affiliation(s)
- Rebecca Knackstedt
- Department of Plastic Surgery, Cleveland Clinic, 2049 East 100th Street, Desk A60, Cleveland, OH 44195, USA
| | - Timothy Smile
- Department of Radiation Oncology, Cleveland Clinic, Taussig Cancer Center, 10201 Carnegie Avenue, Cleveland, OH 44195, USA
| | - Jennifer Yu
- Department of Radiation Oncology, Cleveland Clinic, Taussig Cancer Center, 10201 Carnegie Avenue, Cleveland, OH 44195, USA
| | - Brian R Gastman
- Department of Plastic Surgery, Cleveland Clinic, Cleveland Clinic Lerner College of Medicine, 2049 East 100th Street, Desk A60, Cleveland, OH 44195, USA.
| |
Collapse
|
38
|
Khan AB, Matsuoka CK, Lee S, Rahman M, Rao G. Prolonged survival after laser interstitial thermal therapy in glioblastoma. Surg Neurol Int 2021; 12:228. [PMID: 34221559 PMCID: PMC8248111 DOI: 10.25259/sni_174_2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023] Open
Abstract
Background: Glioblastoma (GBM) is the most common primary malignant brain tumor in adults. Management includes surgical resection followed by chemoradiation, and prognosis remains poor. Surgical resection is not possible for some deep-seated or eloquent tumors. Laser interstitial thermal therapy (LITT) has emerged as a new, minimally invasive surgical option for deep-seated GBM. Case Description: We report a case of newly diagnosed thalamic GBM managed with LITT followed by radiation and chemotherapy. Conclusion: The patient remains well at 50-month post-LITT, indicating a potentially unique durability of LITT treatment in GBM.
Collapse
Affiliation(s)
- A Basit Khan
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, United States
| | | | - Sungho Lee
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, United States
| | - Maryam Rahman
- Department of Neurosurgery, University of Florida, Gainesville, Florida, United States
| | - Ganesh Rao
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
39
|
Enhancement of Radio-Thermo-Sensitivity of 5-Iodo-2-Deoxyuridine-Loaded Polymeric-Coated Magnetic Nanoparticles Triggers Apoptosis in U87MG Human Glioblastoma Cancer Cell Line. Cell Mol Bioeng 2021; 14:365-377. [PMID: 34295445 DOI: 10.1007/s12195-021-00675-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 05/05/2021] [Indexed: 01/22/2023] Open
Abstract
Introduction With an emphasis on the radioresistant nature of glioblastoma cells, the aim of the present study was to evaluate the radio-thermo-sensitizing effects of PCL-PEG-coated Superparamagnetic iron oxide nanoparticles (SPIONs) as a carrier of 5-iodo-2-deoxyuridine (IUdR) in monolayer culture of U87MG human glioma cell line. Methods Following monolayer culture of U87MG cells, nanoparticle uptake was assessed using Prussian blue staining and ICP-OES method. The U87MG cells were treated with an appropriate concentration of free IUdR and PCL-PEG-coated SPIONs (MNPs) loaded with IUdR (IUdR/MNPs) for 24 h, subjected to hyperthermia (water bath and alternating magnetic field (AMF)) at 43 °C, and exposed to X-ray (2 Gy, 6 MV). The combined effects of hyperthermia with or without magnetic nanoparticles on radiosensitivity of the U87MG cells were evaluated using colony formation assay (CFA) and Flowcytometry. Results Prussian blue staining and ICP-OES showed that the nanoparticles were able to enter the cells. The results also indicated that IUdR/MNPs combined with X-ray radiation and hyperthermia significantly decreased the colony formation ability of monolayer cells (1.11, 1.41 fold) and increased the percentage of apoptotic (2.47, 4.1 fold) and necrotic cells (12.28, 29.34 fold), when compared to IUdR combined with X-ray and hyperthermia or IUdR/MNPs + X-ray. MTT results revealed that the presence of IUdR/MNPs significantly increased the toxicity of AMF hyperthermia compared to the water bath method. Conclusions Our study showed that SPIONs/PCL-PEG, as a carrier of IUdR, can enhance the cytotoxic effects of radiotherapy and hyperthermia and act as a radio-thermo-sensitizing agent. Graphic Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s12195-021-00675-y.
Collapse
|
40
|
Yu JS, Mohammadi AM. Introduction to laser thermal therapy for brain disorders. Int J Hyperthermia 2021; 37:1-2. [PMID: 32672120 PMCID: PMC7673657 DOI: 10.1080/02656736.2020.1789764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Jennifer S Yu
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alireza M Mohammadi
- Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
41
|
Avecillas-Chasin JM, Atik A, Mohammadi AM, Barnett GH. Laser thermal therapy in the management of high-grade gliomas. Int J Hyperthermia 2021; 37:44-52. [PMID: 32672121 DOI: 10.1080/02656736.2020.1767807] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Laser interstitial thermal therapy (LITT) is a minimally invasive therapy that have been used for brain tumors, epilepsy, chronic pain, and other spine pathologies. This therapy is performed under imaging and stereotactic guidance to precisely direct the probe and ablate the area of interest using real-time magnetic resonance (MR) thermography. LITT has gained popularity as a treatment for glioma because of its minimally invasive nature, small skin incision, repeatability, shorter hospital stay, and the possibility of receiving adjuvant therapy shortly after surgery instead of several weeks as required after open surgical resection. Several reports have demonstrated the usefulness of LITT in the treatment of newly-diagnosed and recurrent gliomas. In this review, we will summarize the recent evidence of this therapy in the field of glioma surgery and the future perspectives of the use of LITT combined with other treatment strategies for this devastating disease.
Collapse
Affiliation(s)
- Josue M Avecillas-Chasin
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA.,Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ahmet Atik
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA.,Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alireza M Mohammadi
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA.,Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Gene H Barnett
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA.,Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
42
|
Kavya S, Reghu R. An Overview of High-grade Glioma: Current and Emerging Treatment Approaches. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394716666200721155514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
High grade glioma is one of the severe form of tumour that progresses in the glial cells
of the brain and spinal cord. Age, gender, exposure to infections, race, ethnicity, viruses and allergens,
environmental carcinogens, diet, head injury or trauma and ionizing radiation may report
with increased glioma risk. Headache, seizure mainly generalized tonic-clonic seizure, memory
loss and altered sensorium are considered as common symptoms of glioma. Magnetic Resonance
Imaging (MRI), CT scans, neurological examinations and biopsy are considered as the diagnostic
option for glioma. Treatment for glioma mainly depended upon the tumour progression, malignancy,
cell type, age, location of tumour growth and anatomic structure. The standard treatment includes
surgery, radiation therapy and chemotherapy. Temozolomide is usually prescribed at a
dosage of 75 mg/m2 and began in combination with radiation therapy and continued daily. The primary
indicator of hepatotoxicity is the elevation of the liver profiles, i.e. the changes in any of the
liver panels may be considered to be hepatotoxic. Serum glutamic oxaloacetic transaminase (SGOT),
Serum Glutamic Pyruvic Transaminase (SGPT), Alkaline phosphatase (ALP) are rising panels
of the liver, which are elevated during toxicity. In some patients, albumin and globulin levels
may show variations. Treatment for glioma associated symptoms like seizures, depression anxiety
etc. are also mentioned along with supportive care for glioma. New trends in the treatment for glioma
are RINTEGA, an experimental immunotherapeutic agent and bevazizumab, a recombinant
monoclonal, a humanized antibody against the VEGF ligand [VEGF-A (vascular endothelial
growth factor)] in tumor cells.
Collapse
Affiliation(s)
- S.G. Kavya
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - R. Reghu
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| |
Collapse
|
43
|
Nanotechnology and Nanocarrier-Based Drug Delivery as the Potential Therapeutic Strategy for Glioblastoma Multiforme: An Update. Cancers (Basel) 2021; 13:cancers13020195. [PMID: 33430494 PMCID: PMC7827410 DOI: 10.3390/cancers13020195] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/20/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Glioblastoma multiforme (GBM) are among the most lethal tumors. The highly invasive nature and presence of GBM stem cells, as well as the blood brain barrier (BBB) which limits chemotherapeutic drugs from entering the tumor mass, account for the high chance of treatment failure. Recent developments have found that nanoparticles can be conjugated to liposomes, dendrimers, metal irons, or polymeric micelles, which enhance the drug-loaded compounds to efficiently penetrate the BBB, thus offering new possibilities for overcoming GBM stem cell-mediated resistance to chemotherapy and radiation therapy. In addition, there have been new emerging strategies that use nanocarriers for successful GBM treatment in animal models. This review highlights the recent development of nanotechnology and nanocarrier-based drug delivery for treatment of GBMs, which may be a promising therapeutic strategy for this tumor entity. Abstract Glioblastoma multiforme (GBM) is the most common and malignant brain tumor with poor prognosis. The heterogeneous and aggressive nature of GBMs increases the difficulty of current standard treatment. The presence of GBM stem cells and the blood brain barrier (BBB) further contribute to the most important compromise of chemotherapy and radiation therapy. Current suggestions to optimize GBM patients’ outcomes favor controlled targeted delivery of chemotherapeutic agents to GBM cells through the BBB using nanoparticles and monoclonal antibodies. Nanotechnology and nanocarrier-based drug delivery have recently gained attention due to the characteristics of biosafety, sustained drug release, increased solubility, and enhanced drug bioactivity and BBB penetrability. In this review, we focused on recently developed nanoparticles and emerging strategies using nanocarriers for the treatment of GBMs. Current studies using nanoparticles or nanocarrier-based drug delivery system for treatment of GBMs in clinical trials, as well as the advantages and limitations, were also reviewed.
Collapse
|
44
|
Abstract
The previous decade has seen an expansion in the use of laser interstitial thermal therapy (LITT) for a variety of pathologies. LITT has been used to treat both newly diagnosed and recurrent glioblastoma (GBM), especially in deep-seated, difficult-to-access lesions where open resection is otherwise infeasible or in patients who would not tolerate craniotomy. This review aims to describe the current state of the technology and operative technique, as well as summarize the outcomes data and future research regarding LITT as a treatment of GBM.
Collapse
Affiliation(s)
- Matthew M Grabowski
- Department of Neurosurgery, Rose Ella Burkhardt Brain Tumor & Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA
| | - Balint Otvos
- Department of Neurosurgery, Rose Ella Burkhardt Brain Tumor & Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA
| | - Alireza M Mohammadi
- Department of Neurological Surgery, Cleveland Clinic Lerner College of Medicine at CWRU, Rose Ella Burkhardt Brain Tumor & Neuro-Oncology Center, Cleveland Clinic, CA-51, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| |
Collapse
|
45
|
Lou M, Li R, Lang TY, Zhang LY, Zhou Q, Li L. Aberrant methylation of GADD45A is associated with decreased radiosensitivity in cervical cancer through the PI3K/AKT signaling pathway. Oncol Lett 2020; 21:8. [PMID: 33240414 PMCID: PMC7681222 DOI: 10.3892/ol.2020.12269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 10/08/2020] [Indexed: 12/12/2022] Open
Abstract
Epigenetic inactivation of GADD45A is a common occurrence in different types of cancer. However, little is known regarding its association with radiosensitivity in cervical cancer (CC). Thus, the present study aimed to investigate the association between aberrant GADD45A methylation and radiosensitivity in CC. SiHa, HeLa and CaSki CC cells were treated with 5-azacytidine (5-azaC), with or without irradiation. The expression levels of GADD45A and AKT related molecules were detected via reverse transcription-quantitative PCR and western blot analyses. The methylation status of GADD45A was assessed via methylation-specific PCR and cell proliferation assays, while clonogenic assays and flow cytometric analysis were performed to assess the function of the genes (GADD45A and AKT) in the CC cell lines. The results demonstrated that methylation of GADD45A was significantly higher in the radioresistant tissues (63.16%) compared with the radiosensitive samples (33.33%). In addition, the surviving fraction of SiHa cells following irradiation with 2 Gy was demonstrated to be highest amongst the three CC cells (CaSki, 57±9.5%; HeLa, 70±4% and SiHa, 75±10%). The survival rate of SiHa cells following treatment with 5-azaC and ionizing radiation (IR) significantly decreased as the radiation dose increased, compared with treatment with IR alone. Following overexpression of GADD45A or treatment with 5-azaC, the radiosensitivity of SiHa cells significantly increased compared with both the control vector and PBS treated groups. In addition, the AKT inhibitor, MK-2206, increased the radiosensitivity of SiHa cells. Notably, aberrant methylation of GADD45A was associated with decreased radiosensitivity in CC, and the PI3K/AKT signaling pathway was essential for radioresistance, which was mediated through downregulation of GADD45A.
Collapse
Affiliation(s)
- Meng Lou
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Rong Li
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing 400030, P.R. China
| | - Ting-Yuan Lang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing 400030, P.R. China
| | - Li-Ying Zhang
- Department of Gynecology, The Second Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Qi Zhou
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Department of Gynecologic Oncology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing 400030, P.R. China.,Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing 400030, P.R. China.,Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing 400030, P.R. China
| | - Li Li
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
46
|
Sawai H, Kurimoto M, Suzuki Y, Yamaguchi Y, Murata A, Suganuma E, Yamamoto K, Kuzuya H, Ueno S, Koide S, Koide H, Kamiya A. Efficacy of Hyperthermia in Treatment of Recurrent Metastatic Breast Cancer After Long-Term Chemotherapy: A Report of 2 Cases. AMERICAN JOURNAL OF CASE REPORTS 2020; 21:e926647. [PMID: 33141812 PMCID: PMC7649113 DOI: 10.12659/ajcr.926647] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/24/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Breast cancer has a long-term prognosis with various multimodality treatments. This report introduces the effectiveness of radiofrequency (RF) hyperthermia in the long-term treatment for recurrent/metastatic breast cancer. CASE REPORT In the first case, the patient had bone and liver metastases during the course of chemotherapy, hormone therapy, and radiotherapy for 27 years after curative resection of breast cancer. Finally, she received RF hyperthermia alone for liver metastasis and showed a decrease in tumor markers and reduction in liver metastasis on computed tomography (CT). In the second case, the patient underwent curative resection for multiple occurrences on the left side of the breast. She received postoperative chemotherapy combined with hormone therapy but had metachronous local recurrences. She continued hormone therapy after 2 local recurrence resections; unfortunately, she had bone, liver, and lung metastases and pleural dissemination. Eventually, the patient received RF hyperthermia combined with oral chemotherapy. Her tumor markers decreased, and CT showed disappearance of lung metastasis and improved pleural dissemination. Furthermore, the reduction of chemotherapy adverse events due to hyperthermia allowed the patient to continue chemotherapy and improved her quality of life. CONCLUSIONS We present 2 cases in which RF hyperthermia had a positive effect despite the presence of a recurrent tumor after various types of surgery, chemotherapy, and radiotherapy. This report suggests that the addition of RF hyperthermia to conventional multidisciplinary therapies may enhance the therapeutic effect of these treatments and improve the quality of life in patients with recurrent breast cancer.
Collapse
Affiliation(s)
- Hirozumi Sawai
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Masaaki Kurimoto
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Yuka Suzuki
- Department of Medical Technique (Radiological Technology), Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Yoshimi Yamaguchi
- Department of Medical Technique (Radiological Technology), Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Akemi Murata
- Department of Medical Technique (Radiological Technology), Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Enami Suganuma
- Department of Medical Technique (Radiological Technology), Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Kazuya Yamamoto
- Department of Medical Technique (Radiological Technology), Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Hiromasa Kuzuya
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Shuhei Ueno
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Shuji Koide
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Hajime Koide
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Atsushi Kamiya
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| |
Collapse
|
47
|
Meng Y, Hynynen K, Lipsman N. Applications of focused ultrasound in the brain: from thermoablation to drug delivery. Nat Rev Neurol 2020; 17:7-22. [PMID: 33106619 DOI: 10.1038/s41582-020-00418-z] [Citation(s) in RCA: 247] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
Focused ultrasound (FUS) is a disruptive medical technology, and its implementation in the clinic represents the culmination of decades of research. Lying at the convergence of physics, engineering, imaging, biology and neuroscience, FUS offers the ability to non-invasively and precisely intervene in key circuits that drive common and challenging brain conditions. The actions of FUS in the brain take many forms, ranging from transient blood-brain barrier opening and neuromodulation to permanent thermoablation. Over the past 5 years, we have seen a dramatic expansion of indications for and experience with FUS in humans, with a resultant exponential increase in academic and public interest in the technology. Applications now span the clinical spectrum in neurological and psychiatric diseases, with insights still emerging from preclinical models and human trials. In this Review, we provide a comprehensive overview of therapeutic ultrasound and its current and emerging indications in the brain. We examine the potential impact of FUS on the landscape of brain therapies as well as the challenges facing further advancement and broader adoption of this promising minimally invasive therapeutic alternative.
Collapse
Affiliation(s)
- Ying Meng
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Sunnybrook Research Institute, Hurvitz Brain Sciences Program, Harquail Centre for Neuromodulation, Toronto, ON, Canada.,Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Kullervo Hynynen
- Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Medical Biophysics and Institute of Biomaterials & Biomedical Engineering (IBBME), University of Toronto, Toronto, ON, Canada
| | - Nir Lipsman
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. .,Sunnybrook Research Institute, Hurvitz Brain Sciences Program, Harquail Centre for Neuromodulation, Toronto, ON, Canada. .,Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
48
|
Frenster JD, Desai S, Placantonakis DG. In vitro evidence for glioblastoma cell death in temperatures found in the penumbra of laser-ablated tumors. Int J Hyperthermia 2020; 37:20-26. [PMID: 32672127 PMCID: PMC7725000 DOI: 10.1080/02656736.2020.1774082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/11/2020] [Accepted: 05/20/2020] [Indexed: 12/30/2022] Open
Abstract
The concept of thermal therapy toward the treatment of brain tumors has gained traction in recent years. Traditionally, thermal therapy has been subdivided into hyperthermia, with mild elevation of temperature in treated tissue above the physiologic baseline; and thermal ablation, where even higher temperatures are achieved. The recent surge in interest has been driven by the use of novel thermal ablation technologies, including laser interstitial thermal therapy (LITT), that are implemented in brain tumor treatment. Here, we review previous scientific literature on the biologic effects of thermal therapy on brain tumors, with an emphasis on glioblastoma (GBM), an aggressive brain malignancy. In addition, we present in vitro evidence from our laboratory that even moderate elevations in temperature achieved in the penumbra around laser-ablated coagulum may also produce GBM cell death. While much remains to be elucidated in terms of the biology of thermal therapy, we propose that it is a welcome addition to the neuro-oncology armamentarium, in particular with regard to GBM, which is generally resistant to current chemoradiotherapeutic regimens.
Collapse
Affiliation(s)
- Joshua D. Frenster
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Shivang Desai
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY, USA
- Emory School of Medicine, Atlanta, GA, USA
| | - Dimitris G. Placantonakis
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA
- Kimmel Center for Stem Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Brain and Spine Tumor Center, NYU Grossman School of Medicine, New York, NY 10016, USA
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
49
|
Skandalakis GP, Rivera DR, Rizea CD, Bouras A, Raj JGJ, Bozec D, Hadjipanayis CG. Hyperthermia treatment advances for brain tumors. Int J Hyperthermia 2020; 37:3-19. [PMID: 32672123 PMCID: PMC7756245 DOI: 10.1080/02656736.2020.1772512] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/15/2020] [Accepted: 05/16/2020] [Indexed: 02/06/2023] Open
Abstract
Hyperthermia therapy (HT) of cancer is a well-known treatment approach. With the advent of new technologies, HT approaches are now important for the treatment of brain tumors. We review current clinical applications of HT in neuro-oncology and ongoing preclinical research aiming to advance HT approaches to clinical practice. Laser interstitial thermal therapy (LITT) is currently the most widely utilized thermal ablation approach in clinical practice mainly for the treatment of recurrent or deep-seated tumors in the brain. Magnetic hyperthermia therapy (MHT), which relies on the use of magnetic nanoparticles (MNPs) and alternating magnetic fields (AMFs), is a new quite promising HT treatment approach for brain tumors. Initial MHT clinical studies in combination with fractionated radiation therapy (RT) in patients have been completed in Europe with encouraging results. Another combination treatment with HT that warrants further investigation is immunotherapy. HT approaches for brain tumors will continue to a play an important role in neuro-oncology.
Collapse
Affiliation(s)
- Georgios P. Skandalakis
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Daniel R. Rivera
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Caroline D. Rizea
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Joe Gerald Jesu Raj
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Dominique Bozec
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Constantinos G. Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
50
|
Mohammadi AM, Sharma M, Beaumont TL, Juarez KO, Kemeny H, Dechant C, Seas A, Sarmey N, Lee BS, Jia X, Fecci PE, Baehring J, Moliterno J, Chiang VL, Ahluwalia MS, Kim AH, Barnett GH, Leuthardt EC. Upfront Magnetic Resonance Imaging-Guided Stereotactic Laser-Ablation in Newly Diagnosed Glioblastoma: A Multicenter Review of Survival Outcomes Compared to a Matched Cohort of Biopsy-Only Patients. Neurosurgery 2020; 85:762-772. [PMID: 30476325 DOI: 10.1093/neuros/nyy449] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/21/2018] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Laser ablation (LA) is used as an upfront treatment in patients with deep seated newly diagnosed Glioblastoma (nGBM). OBJECTIVE To evaluate the outcomes of LA in patients with nGBM and compare them with a matched biopsy-only cohort. METHODS Twenty-four nGBM patients underwent upfront LA at Cleveland clinic, Washington University in St. Louis, and Yale University (6/2011-12/2014) followed by chemo/radiotherapy. Also, 24 out of 171 nGBM patients with biopsy followed by chemo/radiotherapy were matched based on age (< 70 vs ≥ 70), gender, tumor location (deep vs lobar), and volume (<11 cc vs ≥11 cc). Progression-free survival (PFS), overall survival (OS), and disease-specific PFS and OS were outcome measures. Three prognostic groups were identified based on extent of tumor ablation by thermal-damage-threshold (TDT)-lines. RESULTS The median tumor volume in LA (n = 24) and biopsy only (n = 24) groups was 9.3 cm3 and 8.2 cm3 respectively. Overall, median estimate of OS and PFS in LA cohort was 14.4 and 4.3 mo compared to 15.8 mo and 5.9 mo for biopsy only cohort. On multivariate analysis, favorable TDT-line prognostic groups were associated with lower incidence of disease specific death (P = .03) and progression (P = .05) compared to other groups including biopsy only cohort. Only age (<70 yr, P = .02) and tumor volume (<11 cc, P = .03) were favorable prognostic factors for OS. CONCLUSION The maximum tumor coverage by LA followed by radiation/chemotherapy is an effective treatment modality in patients with nGBM, compared to biopsy only cohort. The TDT-line prognostic groups were independent predictor of disease specific death and progression after LA.
Collapse
Affiliation(s)
- Alireza M Mohammadi
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Mayur Sharma
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Thomas L Beaumont
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Kevin O Juarez
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Hanna Kemeny
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Cosette Dechant
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Andreas Seas
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Nehaw Sarmey
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Bryan S Lee
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Xuefei Jia
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio
| | - Peter E Fecci
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Joachim Baehring
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Jennifer Moliterno
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Veronica L Chiang
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Manmeet S Ahluwalia
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Albert H Kim
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Gene H Barnett
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Eric C Leuthardt
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
- Department of Biomedical Engineering, Center for Innovation in Neuroscience and Technology, Washington University School of Medicine, St. Louis, Missouri
- Department of Mechanical Engineering and Material Science, Center for Innovation in Neuroscience and Technology, Washington University, School of Medicine, St. Louis, Missouri
| |
Collapse
|