251
|
Gan HK, Parakh S, Lassman AB, Seow A, Lau E, Lee ST, Ameratunga M, Perchyonok Y, Cao D, Burvenich IJG, O'Keefe GJ, Rigopoulos A, Gomez E, Maag D, Scott AM. Tumor volumes as a predictor of response to the anti-EGFR antibody drug conjugate depatuxizumab mafadotin. Neurooncol Adv 2021; 3:vdab102. [PMID: 34549181 PMCID: PMC8446913 DOI: 10.1093/noajnl/vdab102] [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] [Indexed: 11/13/2022] Open
Abstract
Background The adverse impact of increasing brain tumor size on the efficacy of antibody-drug conjugates (ADCs) was investigated preclinically then validated with clinical data. Methods—Preclinical study The impact of tumor size on ADC tumor delivery and treatment response was evaluated in an EGFR-amplified patient-derived glioblastoma (GBM) model following treatment with Depatuxizumab mafadotin (Depatux-M). Biodistribution and imaging studies correlated drug distribution with starting treatment volume and anti-tumor activity. Methods—Clinical study M12-356 was a Phase I study of Depatux-M in patients with GBM. Blinded volumetric analysis of baseline tumor volumes of M12-356 patients was undertaken by two reviewers and results correlated with response and survival. Results Preclinically, imaging and biodistribution studies showed specific and significantly higher tumor uptake of zirconium-89 labeled Depatux-M (89Zr-Depatux-M) in mice with smaller tumor volume (~98 mm3) versus those with larger volumes (~365 mm3); concordantly, mice with tumor volumes ≤100 mm3 at treatment commencement had significantly better growth inhibition by Depatux-M (93% vs 27%, P < .001) and significantly longer overall survival (P < .0001) compared to tumors ≥400 mm3. Clinically, patients with tumor volumes <25 cm3 had significantly higher response rates (17% vs. 0%, P = .009) and longer overall survival (0.5 vs 0.89 years, P = .001) than tumors above 25 cm3. Conclusion Both preclinical and clinical data showed intra-tumoral concentration and efficacy of Depatux-m inversely correlated with tumor size. This finding merit further investigation with pretreatment tumor volume as a predictor for response to ADCs, in both gliomas and other solid tumors.
Collapse
Affiliation(s)
- Hui K Gan
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia.,La Trobe University School of Cancer Medicine, Heidelberg, Melbourne, Australia.,Department of Medical Oncology, Austin Health, Heidelberg, Melbourne, Australia.,Department of Medicine, University of Melbourne, Parkville, Australia
| | - Sagun Parakh
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia.,La Trobe University School of Cancer Medicine, Heidelberg, Melbourne, Australia.,Department of Medical Oncology, Monash Health, Clayton, Melbourne, Australia
| | - Andrew B Lassman
- Division of Neuro-Oncology, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, NewYork-Presbyterian Hospital, New York, NY, USA
| | - Aidan Seow
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Melbourne, Australia
| | - Eddie Lau
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Melbourne, Australia.,Department of Radiology, Austin Health, Heidelberg, Melbourne, Australia
| | - Sze Ting Lee
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia.,La Trobe University School of Cancer Medicine, Heidelberg, Melbourne, Australia.,Department of Medicine, University of Melbourne, Parkville, Australia.,Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Melbourne, Australia
| | - Malaka Ameratunga
- Department of Medical Oncology, Austin Health, Heidelberg, Melbourne, Australia
| | - Yuliya Perchyonok
- Department of Radiology, Austin Health, Heidelberg, Melbourne, Australia
| | - Diana Cao
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia
| | - Ingrid J G Burvenich
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia.,La Trobe University School of Cancer Medicine, Heidelberg, Melbourne, Australia
| | - Graeme J O'Keefe
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Melbourne, Australia
| | - Angela Rigopoulos
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia
| | - Erica Gomez
- Research and Development Department, AbbVie Inc., North Chicago, Illinois, USA
| | - David Maag
- Research and Development Department, AbbVie Inc., North Chicago, Illinois, USA
| | - Andrew M Scott
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia.,La Trobe University School of Cancer Medicine, Heidelberg, Melbourne, Australia.,Department of Medicine, University of Melbourne, Parkville, Australia.,Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Melbourne, Australia
| |
Collapse
|
252
|
Localized blood-brain barrier opening in infiltrating gliomas with MRI-guided acoustic emissions-controlled focused ultrasound. Proc Natl Acad Sci U S A 2021; 118:2103280118. [PMID: 34504017 DOI: 10.1073/pnas.2103280118] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Pharmacological treatment of gliomas and other brain-infiltrating tumors remains challenging due to limited delivery of most therapeutics across the blood-brain barrier (BBB). Transcranial MRI-guided focused ultrasound (FUS), an emerging technology for noninvasive brain treatments, enables transient opening of the BBB through acoustic activation of circulating microbubbles. Here, we evaluate the safety and utility of transcranial microbubble-enhanced FUS (MB-FUS) for spatially targeted BBB opening in patients with infiltrating gliomas. In this Phase 0 clinical trial (NCT03322813), we conducted comparative and quantitative analyses of FUS exposures (sonications) and their effects on gliomas using MRI, histopathology, microbubble acoustic emissions (harmonic dose [HD]), and fluorescence-guided surgery metrics. Contrast-enhanced MRI and histopathology indicated safe and reproducible BBB opening in all patients. These observations occurred using a power cycling closed feedback loop controller, with the power varying by nearly an order of magnitude on average. This range underscores the need for monitoring and titrating the exposure on a patient-by-patient basis. We found a positive correlation between microbubble acoustic emissions (HD) and MR-evident BBB opening (P = 0.07) and associated interstitial changes (P < 0.01), demonstrating the unique capability to titrate the MB-FUS effects in gliomas. Importantly, we identified a 2.2-fold increase of fluorescein accumulation in MB-FUS-treated compared to untreated nonenhancing tumor tissues (P < 0.01) while accounting for vascular density. Collectively, this study demonstrates the capabilities of MB-FUS for safe, localized, controlled BBB opening and highlights the potential of this technology to improve the surgical and pharmacologic treatment of brain tumors.
Collapse
|
253
|
Pang Y, Yu G, Butler M, Sindiri S, Song YK, Wei JS, Wen X, Chou HC, Quezado M, Pack S, Xi L, Abdullaev Z, Kim O, Ranjan A, Merchant M, Antony R, Boris L, Aboud O, Kamson D, Kaplan R, Mackey M, Camphausen K, Zaghloul K, Armstrong TS, Gilbert MR, Aldape K, Holdhoff M, Khan J, Wu J. Report of Canonical BCR- ABL1 Fusion in Glioblastoma. JCO Precis Oncol 2021; 5:PO.20.00519. [PMID: 34485806 DOI: 10.1200/po.20.00519] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/25/2021] [Accepted: 07/27/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- Ying Pang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Guangyang Yu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Madison Butler
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Sivasish Sindiri
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Young K Song
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Jun S Wei
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Xinyu Wen
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Hisen-Chao Chou
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Martha Quezado
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Svetlana Pack
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Liqiang Xi
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Zied Abdullaev
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Olga Kim
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Alice Ranjan
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Mythili Merchant
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Ramya Antony
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Lisa Boris
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Orwa Aboud
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - David Kamson
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rosandra Kaplan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Megan Mackey
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD
| | - Kareem Zaghloul
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | - Terri S Armstrong
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Matthias Holdhoff
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Jing Wu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
254
|
Wu W, Klockow JL, Zhang M, Lafortune F, Chang E, Jin L, Wu Y, Daldrup-Link HE. Glioblastoma multiforme (GBM): An overview of current therapies and mechanisms of resistance. Pharmacol Res 2021; 171:105780. [PMID: 34302977 PMCID: PMC8384724 DOI: 10.1016/j.phrs.2021.105780] [Citation(s) in RCA: 237] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme (GBM) is a WHO grade IV glioma and the most common malignant, primary brain tumor with a 5-year survival of 7.2%. Its highly infiltrative nature, genetic heterogeneity, and protection by the blood brain barrier (BBB) have posed great treatment challenges. The standard treatment for GBMs is surgical resection followed by chemoradiotherapy. The robust DNA repair and self-renewing capabilities of glioblastoma cells and glioma initiating cells (GICs), respectively, promote resistance against all current treatment modalities. Thus, durable GBM management will require the invention of innovative treatment strategies. In this review, we will describe biological and molecular targets for GBM therapy, the current status of pharmacologic therapy, prominent mechanisms of resistance, and new treatment approaches. To date, medical imaging is primarily used to determine the location, size and macroscopic morphology of GBM before, during, and after therapy. In the future, molecular and cellular imaging approaches will more dynamically monitor the expression of molecular targets and/or immune responses in the tumor, thereby enabling more immediate adaptation of tumor-tailored, targeted therapies.
Collapse
Affiliation(s)
- Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Jessica L Klockow
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Michael Zhang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Famyrah Lafortune
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Edwin Chang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Linchun Jin
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| | - Yang Wu
- Department of Neuropathology, Institute of Pathology, Technical University of Munich, Munich, Bayern 81675, Germany
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
255
|
Andersen RS, Anand A, Harwood DSL, Kristensen BW. Tumor-Associated Microglia and Macrophages in the Glioblastoma Microenvironment and Their Implications for Therapy. Cancers (Basel) 2021; 13:cancers13174255. [PMID: 34503065 PMCID: PMC8428223 DOI: 10.3390/cancers13174255] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma is the most frequent and malignant primary brain tumor. Standard of care includes surgery followed by radiation and temozolomide chemotherapy. Despite treatment, patients have a poor prognosis with a median survival of less than 15 months. The poor prognosis is associated with an increased abundance of tumor-associated microglia and macrophages (TAMs), which are known to play a role in creating a pro-tumorigenic environment and aiding tumor progression. Most treatment strategies are directed against glioblastoma cells; however, accumulating evidence suggests targeting of TAMs as a promising therapeutic strategy. While TAMs are typically dichotomously classified as M1 and M2 phenotypes, recent studies utilizing single cell technologies have identified expression pattern differences, which is beginning to give a deeper understanding of the heterogeneous subpopulations of TAMs in glioblastomas. In this review, we evaluate the role of TAMs in the glioblastoma microenvironment and discuss how their interactions with cancer cells have an extensive impact on glioblastoma progression and treatment resistance. Finally, we summarize the effects and challenges of therapeutic strategies, which specifically aim to target TAMs.
Collapse
Affiliation(s)
- Rikke Sick Andersen
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark; (R.S.A.); (A.A.)
| | - Atul Anand
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark; (R.S.A.); (A.A.)
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Dylan Scott Lykke Harwood
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark;
- Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Bjarne Winther Kristensen
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark; (R.S.A.); (A.A.)
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark;
- Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
- Correspondence:
| |
Collapse
|
256
|
Wei X, Meel MH, Breur M, Bugiani M, Hulleman E, Phoenix TN. Defining tumor-associated vascular heterogeneity in pediatric high-grade and diffuse midline gliomas. Acta Neuropathol Commun 2021; 9:142. [PMID: 34425907 PMCID: PMC8381557 DOI: 10.1186/s40478-021-01243-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/10/2021] [Indexed: 12/23/2022] Open
Abstract
The blood–brain barrier (BBB) plays important roles in brain tumor pathogenesis and treatment response, yet our understanding of its function and heterogeneity within or across brain tumor types remains poorly characterized. Here we analyze the neurovascular unit (NVU) of pediatric high-grade glioma (pHGG) and diffuse midline glioma (DMG) using patient derived xenografts and natively forming glioma mouse models. We show tumor-associated vascular differences between these glioma subtypes, and parallels between PDX and mouse model systems, with DMG models maintaining a more normal vascular architecture, BBB function and endothelial transcriptional program relative to pHGG models. Unlike prior work in angiogenic brain tumors, we find that expression of secreted Wnt antagonists do not alter the tumor-associated vascular phenotype in DMG tumor models. Together, these findings highlight vascular heterogeneity between pHGG and DMG and differences in their response to alterations in developmental BBB signals that may participate in driving these pathological differences.
Collapse
|
257
|
Xavier MA, Rezende F, Titze-de-Almeida R, Cornelissen B. BRCAness as a Biomarker of Susceptibility to PARP Inhibitors in Glioblastoma Multiforme. Biomolecules 2021; 11:1188. [PMID: 34439854 PMCID: PMC8394995 DOI: 10.3390/biom11081188] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain cancer. GBMs commonly acquire resistance to standard-of-care therapies. Among the novel means to sensitize GBM to DNA-damaging therapies, a promising strategy is to combine them with inhibitors of the DNA damage repair (DDR) machinery, such as inhibitors for poly(ADP-ribose) polymerase (PARP). PARP inhibitors (PARPis) have already shown efficacy and have received regulatory approval for breast, ovarian, prostate, and pancreatic cancer treatment. In these cancer types, after PARPi administration, patients carrying specific mutations in the breast cancer 1 (BRCA1) and 2 (BRCA2) suppressor genes have shown better response when compared to wild-type carriers. Mutated BRCA genes are infrequent in GBM tumors, but their cells can carry other genetic alterations that lead to the same phenotype collectively referred to as 'BRCAness'. The most promising biomarkers of BRCAness in GBM are related to isocitrate dehydrogenases 1 and 2 (IDH1/2), epidermal growth factor receptor (EGFR), phosphatase and tensin homolog (PTEN), MYC proto-oncogene, and estrogen receptors beta (ERβ). BRCAness status identified by accurate biomarkers can ultimately predict responsiveness to PARPi therapy, thereby allowing patient selection for personalized treatment. This review discusses potential biomarkers of BRCAness for a 'precision medicine' of GBM patients.
Collapse
Affiliation(s)
- Mary-Ann Xavier
- Central Institute of Sciences, Technology for Gene Therapy Laboratory, University of Brasília—UnB/FAV, Brasília 70910-900, Brazil; (F.R.); (R.T.-d.-A.)
| | - Fernando Rezende
- Central Institute of Sciences, Technology for Gene Therapy Laboratory, University of Brasília—UnB/FAV, Brasília 70910-900, Brazil; (F.R.); (R.T.-d.-A.)
| | - Ricardo Titze-de-Almeida
- Central Institute of Sciences, Technology for Gene Therapy Laboratory, University of Brasília—UnB/FAV, Brasília 70910-900, Brazil; (F.R.); (R.T.-d.-A.)
| | - Bart Cornelissen
- Department of Oncology, Radiobiology Research Institute, University of Oxford, Oxford OX3 7LJ, UK;
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, 9700 RB Groningen, The Netherlands
| |
Collapse
|
258
|
Biomimetic and cell-based nanocarriers - New strategies for brain tumor targeting. J Control Release 2021; 337:482-493. [PMID: 34352316 DOI: 10.1016/j.jconrel.2021.07.047] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/29/2021] [Accepted: 07/31/2021] [Indexed: 12/16/2022]
Abstract
In the last two decades no significant advances were achieved in the treatment of the most frequent and malignant types of brain tumors. The main difficulties in achieving progress are related to the incapacity to deliver drugs in therapeutic amounts into the central nervous system and the associated severe side effects. Indeed, to obtain effective treatments, the drugs should be able to cross the intended biological barriers and not being inactivated before reaching the specific therapeutic target. To overcome these challenges the development of synthetic nanocarriers has been widely explored for brain tumor treatment but unfortunately with no clinical translation until date. The use of cell-derived nanocarriers or biomimetic nanocarriers has been studied in the last few years, considering their innate bio-interfacing properties. The ability to carry therapeutic agents and a higher selectivity towards brain tumors would bring new hope for the development of safe and effective treatments. In this review, we explore the biological barriers that need to be crossed for effective delivery in brain tumors, and the types and properties of cell-based nanocarriers (extracellular vesicles and cell-membrane coated nanocarriers) currently under investigation.
Collapse
|
259
|
Li J, Zheng M, Shimoni O, Banks WA, Bush AI, Gamble JR, Shi B. Development of Novel Therapeutics Targeting the Blood-Brain Barrier: From Barrier to Carrier. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101090. [PMID: 34085418 PMCID: PMC8373165 DOI: 10.1002/advs.202101090] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/11/2021] [Indexed: 05/05/2023]
Abstract
The blood-brain barrier (BBB) is a highly specialized neurovascular unit, initially described as an intact barrier to prevent toxins, pathogens, and potentially harmful substances from entering the brain. An intact BBB is also critical for the maintenance of normal neuronal function. In cerebral vascular diseases and neurological disorders, the BBB can be disrupted, contributing to disease progression. While restoration of BBB integrity serves as a robust biomarker of better clinical outcomes, the restrictive nature of the intact BBB presents a major hurdle for delivery of therapeutics into the brain. Recent studies show that the BBB is actively engaged in crosstalk between neuronal and the circulatory systems, which defines another important role of the BBB: as an interfacing conduit that mediates communication between two sides of the BBB. This role has been subject to extensive investigation for brain-targeted drug delivery and shows promising results. The dual roles of the BBB make it a unique target for drug development. Here, recent developments and novel strategies to target the BBB for therapeutic purposes are reviewed, from both barrier and carrier perspectives.
Collapse
Affiliation(s)
- Jia Li
- School of PharmacyHenan UniversityKaifeng475001China
- Centre for Motor Neuron DiseaseDepartment of Biomedical SciencesFaculty of Medicine & Health SciencesMacquarie UniversitySydneyNew South Wales2109Australia
| | - Meng Zheng
- Henan‐Macquarie University Joint Center for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifengHenan475004China
| | - Olga Shimoni
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesFaculty of ScienceUniversity of Technology SydneySydneyNew South Wales2007Australia
| | - William A. Banks
- Geriatric Research Education and Clinical CenterVeterans Affairs Puget Sound Health Care System and Division of Gerontology and Geriatric MedicineDepartment of MedicineUniversity of Washington School of MedicineSeattleWA98108USA
| | - Ashley I. Bush
- Melbourne Dementia Research CenterThe Florey Institute for Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoria3052Australia
| | - Jennifer R. Gamble
- Center for the EndotheliumVascular Biology ProgramCentenary InstituteThe University of SydneySydneyNew South Wales2042Australia
| | - Bingyang Shi
- School of PharmacyHenan UniversityKaifeng475001China
- Centre for Motor Neuron DiseaseDepartment of Biomedical SciencesFaculty of Medicine & Health SciencesMacquarie UniversitySydneyNew South Wales2109Australia
- Henan‐Macquarie University Joint Center for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifengHenan475004China
| |
Collapse
|
260
|
Rattray Z, Deng G, Zhang S, Shirali A, May CK, Chen X, Cuffari BJ, Liu J, Zou P, Rattray NJ, Johnson CH, Dubljevic V, Campbell JA, Huttner A, Baehring JM, Zhou J, Hansen JE. ENT2 facilitates brain endothelial cell penetration and blood-brain barrier transport by a tumor-targeting anti-DNA autoantibody. JCI Insight 2021; 6:e145875. [PMID: 34128837 PMCID: PMC8410084 DOI: 10.1172/jci.insight.145875] [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: 11/11/2020] [Accepted: 06/10/2021] [Indexed: 11/18/2022] Open
Abstract
The blood-brain barrier (BBB) prevents antibodies from penetrating the CNS and limits conventional antibody-based approaches to brain tumors. We now show that ENT2, a transporter that regulates nucleoside flux at the BBB, may offer an unexpected path to circumventing this barrier to allow targeting of brain tumors with an anti-DNA autoantibody. Deoxymab-1 (DX1) is a DNA-damaging autoantibody that localizes to tumors and is synthetically lethal to cancer cells with defects in the DNA damage response. We found that DX1 penetrated brain endothelial cells and crossed the BBB, and mechanistic studies identify ENT2 as the key transporter. In efficacy studies, DX1 crosses the BBB to suppress orthotopic glioblastoma and breast cancer brain metastases. ENT2-linked transport of autoantibodies across the BBB has potential to be exploited in brain tumor immunotherapy, and its discovery raises hypotheses on actionable mechanisms of CNS penetration by neurotoxic autoantibodies in CNS lupus.
Collapse
Affiliation(s)
| | - Gang Deng
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Shenqi Zhang
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | - Jun Liu
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Pan Zou
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Caroline H Johnson
- Yale School of Public Health, New Haven, Connecticut, USA.,Yale Cancer Center, New Haven, Connecticut, USA
| | | | | | - Anita Huttner
- Yale Cancer Center, New Haven, Connecticut, USA.,Department of Pathology and
| | - Joachim M Baehring
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA.,Yale Cancer Center, New Haven, Connecticut, USA.,Department of Neurology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA.,Yale Cancer Center, New Haven, Connecticut, USA
| | - James E Hansen
- Department of Therapeutic Radiology and.,Yale Cancer Center, New Haven, Connecticut, USA
| |
Collapse
|
261
|
Antibody-drug conjugates for H3K27M-mutant diffuse midline gliomas: prospects and challenges. Ther Deliv 2021; 12:553-557. [PMID: 34286602 DOI: 10.4155/tde-2021-0045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
262
|
Chen KT, Wei KC, Liu HL. Focused Ultrasound Combined with Microbubbles in Central Nervous System Applications. Pharmaceutics 2021; 13:pharmaceutics13071084. [PMID: 34371774 PMCID: PMC8308978 DOI: 10.3390/pharmaceutics13071084] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/20/2022] Open
Abstract
The blood–brain barrier (BBB) protects the central nervous system (CNS) from invasive pathogens and maintains the homeostasis of the brain. Penetrating the BBB has been a major challenge in the delivery of therapeutic agents for treating CNS diseases. Through a physical acoustic cavitation effect, focused ultrasound (FUS) combined with microbubbles achieves the local detachment of tight junctions of capillary endothelial cells without inducing neuronal damage. The bioavailability of therapeutic agents is increased only in the area targeted by FUS energy. FUS with circulating microbubbles is currently the only method for inducing precise, transient, reversible, and noninvasive BBB opening (BBBO). Over the past decade, FUS-induced BBBO (FUS-BBBO) has been preclinically confirmed to not only enhance the penetration of therapeutic agents in the CNS, but also modulate focal immunity and neuronal activity. Several recent clinical human trials have demonstrated both the feasibility and potential advantages of using FUS-BBBO in diseased patients. The promising results support adding FUS-BBBO as a multimodal therapeutic strategy in modern CNS disease management. This review article explores this technology by describing its physical mechanisms and the preclinical findings, including biological effects, therapeutic concepts, and translational design of human medical devices, and summarizes completed and ongoing clinical trials.
Collapse
Affiliation(s)
- Ko-Ting Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan;
- Ph.D. Program in Biomedical Engineering, Chang Gung University, Guishan, Taoyuan 333, Taiwan
- Neuroscience Research Center, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan;
- Neuroscience Research Center, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, Chang Gung Medical Foundation, TuCheng, New Taipei 236, Taiwan
- School of Medicine, Chang Gung University, Guishan, Taoyuan 333, Taiwan
- Correspondence: (K.-C.W.); (H.-L.L.)
| | - Hao-Li Liu
- Department of Electrical Engineering, National Taiwan University, Da’an, Taipei 106, Taiwan
- Department of Biomedical Engineering, National Taiwan University, Da’an, Taipei 106, Taiwan
- Correspondence: (K.-C.W.); (H.-L.L.)
| |
Collapse
|
263
|
MRI and PET of Brain Tumor Neuroinflammation in the Era of Immunotherapy, From the AJR Special Series on Inflammation. AJR Am J Roentgenol 2021; 218:582-596. [PMID: 34259035 DOI: 10.2214/ajr.21.26159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
With the emergence of immune-modulating therapies, brain tumors present significant diagnostic imaging challenges. These challenges include planning personalized treatment and adjudicating accurate monitoring approaches and therapeutically specific response criteria. This has been due, in part, to the reliance on nonspecific imaging metrics, such as gadolinium-contrast-enhanced MRI or FDG PET, and rapidly evolving biologic understanding of neuroinflammation. The importance of the tumor-immune interaction and ability to therapeutically augment inflammation to improve clinical outcomes necessitates that the radiologist develop a working knowledge of the immune system and its role in clinical neuroimaging. In this article, we review relevant biologic concepts of the tumor microenvironment of primary and metastatic brain tumors, these tumors' interactions with the immune system, and MRI and PET methods for imaging inflammatory elements associated with these malignancies. Recognizing the growing fields of immunotherapeutics and precision oncology, we highlight clinically translatable imaging metrics for the diagnosis and monitoring of brain tumor neuroinflammation. Practical guidance is provided for implementing iron nanoparticle imaging, including imaging indications, protocol, interpretation, and pitfalls. A comprehensive understanding of the inflammatory mechanisms within brain tumors and their imaging features will facilitate the development of innovative non-invasive prognostic and predictive imaging strategies for precision oncology.
Collapse
|
264
|
Burgenske DM, Talele S, Pokorny JL, Mladek AC, Bakken KK, Carlson BL, Schroeder MA, He L, Hu Z, Gampa G, Kosel ML, Decker PA, Kitange GJ, Schmitt-Hoffmann A, Bachmann F, Vaubel RA, Eckel-Passow JE, Giannini C, McSheehy P, Lane HA, Elmquist WF, Sarkaria JN. Preclinical modeling in glioblastoma patient-derived xenograft (GBM PDX) xenografts to guide clinical development of lisavanbulin-a novel tumor checkpoint controller targeting microtubules. Neuro Oncol 2021; 24:384-395. [PMID: 34232318 PMCID: PMC8917401 DOI: 10.1093/neuonc/noab162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is an incurable disease with few approved therapeutic interventions. Radiation therapy (RT) and temozolomide (TMZ) remain the standards of care. The efficacy and optimal deployment schedule of the orally bioavailable small-molecule tumor checkpoint controller lisavanbulin alone, and in combination with, standards of care were assessed using a panel of IDH-wildtype GBM patient-derived xenografts. METHODS Mice bearing intracranial tumors received lisavanbulin +/-RT +/-TMZ and followed for survival. Lisavanbulin concentrations in plasma and brain were determined by liquid chromatography with tandem mass spectrometry, while flow cytometry was used for cell cycle analysis. RESULTS Lisavanbulin monotherapy showed significant benefit (P < .01) in 9 of 14 PDXs tested (median survival extension 9%-84%) and brain-to-plasma ratios of 1.3 and 1.6 at 2- and 6-hours postdose, respectively, validating previous data suggesting significant exposure in the brain. Prolonged lisavanbulin dosing from RT start until moribund was required for maximal benefit (GBM6: median survival lisavanbulin/RT 90 vs. RT alone 69 days, P = .0001; GBM150: lisavanbulin/RT 143 days vs. RT alone 73 days, P = .06). Similar observations were seen with RT/TMZ combinations (GBM39: RT/TMZ/lisavanbulin 502 days vs. RT/TMZ 249 days, P = .0001; GBM26: RT/TMZ/lisavanbulin 172 days vs. RT/TMZ 121 days, P = .04). Immunohistochemical analyses showed a significant increase in phospho-histone H3 with lisavanbulin treatment (P = .01). CONCLUSIONS Lisavanbulin demonstrated excellent brain penetration, significant extension of survival alone or in RT or RT/TMZ combinations, and was associated with mitotic arrest. These data provide a strong clinical rationale for testing lisavanbulin in combination with RT or RT/TMZ in GBM patients.
Collapse
Affiliation(s)
| | - Surabhi Talele
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jenny L Pokorny
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA,Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ann C Mladek
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Katrina K Bakken
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Brett L Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark A Schroeder
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Lihong He
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Zeng Hu
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gautham Gampa
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Matthew L Kosel
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Paul A Decker
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Gaspar J Kitange
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Felix Bachmann
- Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | - Rachael A Vaubel
- Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Caterina Giannini
- Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Paul McSheehy
- Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | - Heidi A Lane
- Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | - William F Elmquist
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA,Corresponding Author: Jann N. Sarkaria, MD, Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA ()
| |
Collapse
|
265
|
Chokshi CR, Brakel BA, Tatari N, Savage N, Salim SK, Venugopal C, Singh SK. Advances in Immunotherapy for Adult Glioblastoma. Cancers (Basel) 2021; 13:cancers13143400. [PMID: 34298615 PMCID: PMC8305609 DOI: 10.3390/cancers13143400] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Therapy failure and disease recurrence are hallmarks of glioblastoma (GBM), the most common and lethal tumor in adults that originates in the brain. Despite aggressive standards of care, tumor recurrence is inevitable with no standardized second-line therapy. Recent clinical studies evaluating therapies that augment the anti-tumor immune response (i.e., immunotherapies) have yielded promising results in subsets of GBM patients. Here, we summarize clinical studies in the past decade that evaluate vaccines, immune checkpoint inhibitors and chimeric antigen receptor (CAR) T cells for treatment of GBM. Although immunotherapies have yet to return widespread efficacy for the majority of GBM patients, critical insights from completed and ongoing clinical trials are informing development of the next generation of therapies, with the goal to alleviate disease burden and extend patient survival. Abstract Despite aggressive multimodal therapy, glioblastoma (GBM) remains the most common malignant primary brain tumor in adults. With the advent of therapies that revitalize the anti-tumor immune response, several immunotherapeutic modalities have been developed for treatment of GBM. In this review, we summarize recent clinical and preclinical efforts to evaluate vaccination strategies, immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T cells. Although these modalities have shown long-term tumor regression in subsets of treated patients, the underlying biology that may predict efficacy and inform therapy development is being actively investigated. Common to all therapeutic modalities are fundamental mechanisms of therapy evasion by tumor cells, including immense intratumoral heterogeneity, suppression of the tumor immune microenvironment and low mutational burden. These insights have led efforts to design rational combinatorial therapies that can reignite the anti-tumor immune response, effectively and specifically target tumor cells and reliably decrease tumor burden for GBM patients.
Collapse
Affiliation(s)
- Chirayu R. Chokshi
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
| | - Benjamin A. Brakel
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
| | - Nazanin Tatari
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
| | - Neil Savage
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
| | - Sabra K. Salim
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
| | - Chitra Venugopal
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada;
| | - Sheila K. Singh
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.R.C.); (B.A.B.); (N.T.); (N.S.); (S.K.S.)
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada;
- Correspondence:
| |
Collapse
|
266
|
Li QY, Lee JH, Kim HW, Jin GZ. Research Models of the Nanoparticle-Mediated Drug Delivery across the Blood-Brain Barrier. Tissue Eng Regen Med 2021; 18:917-930. [PMID: 34181202 DOI: 10.1007/s13770-021-00356-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/11/2021] [Accepted: 05/16/2021] [Indexed: 12/17/2022] Open
Abstract
Brain diseases and damages come in many forms such as neurodegenerative diseases, tumors, and stroke. Millions of people currently suffer from neurological diseases worldwide. While Challenges of current diagnosis and treatment for neurological diseases are the drug delivery to the central nervous system. The Blood-Brain Barrier (BBB) limits the drug from reaching the targeted site thus showing poor effects. Nanoparticles that have advantage of the assembly at the nanoscale of available biomaterials can provide a delivery platform with potential to raising brain levels of either imaging therapeutic drugs or imaging. Therefore, successful modeling of the BBB is another crucial factor for the development of nanodrugs. In this review, we analyze the in vitro and in vivo findings achieved in various models, and outlook future development of nanodrugs for the successful treatment of brain diseases and damages.
Collapse
Affiliation(s)
- Quan-You Li
- Department of Orthopedics, Yanbian University Hospital , Yanji , China
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea.,Department of Nanobiomedical Science & BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, 256 Grays Inn Road, London, WC1X 8LD, UK
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea. .,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea. .,Department of Nanobiomedical Science & BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea. .,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, 256 Grays Inn Road, London, WC1X 8LD, UK.
| | - Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea. .,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea. .,Department of Nanobiomedical Science & BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
267
|
Mockenhaupt K, Gonsiewski A, Kordula T. RelB and Neuroinflammation. Cells 2021; 10:1609. [PMID: 34198987 PMCID: PMC8307460 DOI: 10.3390/cells10071609] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation within the central nervous system involves multiple cell types that coordinate their responses by secreting and responding to a plethora of inflammatory mediators. These factors activate multiple signaling cascades to orchestrate initial inflammatory response and subsequent resolution. Activation of NF-κB pathways in several cell types is critical during neuroinflammation. In contrast to the well-studied role of p65 NF-κB during neuroinflammation, the mechanisms of RelB activation in specific cell types and its roles during neuroinflammatory response are less understood. In this review, we summarize the mechanisms of RelB activation in specific cell types of the CNS and the specialized effects this transcription factor exerts during neuroinflammation.
Collapse
Affiliation(s)
| | | | - Tomasz Kordula
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VI 23298, USA; (K.M.); (A.G.)
| |
Collapse
|
268
|
Park JH, de Lomana ALG, Marzese DM, Juarez T, Feroze A, Hothi P, Cobbs C, Patel AP, Kesari S, Huang S, Baliga NS. A Systems Approach to Brain Tumor Treatment. Cancers (Basel) 2021; 13:3152. [PMID: 34202449 PMCID: PMC8269017 DOI: 10.3390/cancers13133152] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Brain tumors are among the most lethal tumors. Glioblastoma, the most frequent primary brain tumor in adults, has a median survival time of approximately 15 months after diagnosis or a five-year survival rate of 10%; the recurrence rate is nearly 90%. Unfortunately, this prognosis has not improved for several decades. The lack of progress in the treatment of brain tumors has been attributed to their high rate of primary therapy resistance. Challenges such as pronounced inter-patient variability, intratumoral heterogeneity, and drug delivery across the blood-brain barrier hinder progress. A comprehensive, multiscale understanding of the disease, from the molecular to the whole tumor level, is needed to address the intratumor heterogeneity resulting from the coexistence of a diversity of neoplastic and non-neoplastic cell types in the tumor tissue. By contrast, inter-patient variability must be addressed by subtyping brain tumors to stratify patients and identify the best-matched drug(s) and therapies for a particular patient or cohort of patients. Accomplishing these diverse tasks will require a new framework, one involving a systems perspective in assessing the immense complexity of brain tumors. This would in turn entail a shift in how clinical medicine interfaces with the rapidly advancing high-throughput (HTP) technologies that have enabled the omics-scale profiling of molecular features of brain tumors from the single-cell to the tissue level. However, several gaps must be closed before such a framework can fulfill the promise of precision and personalized medicine for brain tumors. Ultimately, the goal is to integrate seamlessly multiscale systems analyses of patient tumors and clinical medicine. Accomplishing this goal would facilitate the rational design of therapeutic strategies matched to the characteristics of patients and their tumors. Here, we discuss some of the technologies, methodologies, and computational tools that will facilitate the realization of this vision to practice.
Collapse
Affiliation(s)
- James H. Park
- Institute for Systems Biology, Seattle, WA 98109, USA; (J.H.P.); (S.H.)
| | | | - Diego M. Marzese
- Balearic Islands Health Research Institute (IdISBa), 07010 Palma, Spain;
| | - Tiffany Juarez
- St. John’s Cancer Institute, Santa Monica, CA 90401, USA; (T.J.); (S.K.)
| | - Abdullah Feroze
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA; (A.F.); (A.P.P.)
| | - Parvinder Hothi
- Swedish Neuroscience Institute, Seattle, WA 98122, USA; (P.H.); (C.C.)
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Seattle, WA 98122, USA
| | - Charles Cobbs
- Swedish Neuroscience Institute, Seattle, WA 98122, USA; (P.H.); (C.C.)
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Seattle, WA 98122, USA
| | - Anoop P. Patel
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA; (A.F.); (A.P.P.)
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Brotman-Baty Institute for Precision Medicine, University of Washington, Seattle, WA 98195, USA
| | - Santosh Kesari
- St. John’s Cancer Institute, Santa Monica, CA 90401, USA; (T.J.); (S.K.)
| | - Sui Huang
- Institute for Systems Biology, Seattle, WA 98109, USA; (J.H.P.); (S.H.)
| | - Nitin S. Baliga
- Institute for Systems Biology, Seattle, WA 98109, USA; (J.H.P.); (S.H.)
- Departments of Microbiology, Biology, and Molecular Engineering Sciences, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
269
|
Burns TC, Quinones-Hinojosa A. Regenerative medicine for neurological diseases-will regenerative neurosurgery deliver? BMJ 2021; 373:n955. [PMID: 34162530 DOI: 10.1136/bmj.n955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Regenerative medicine aspires to transform the future practice of medicine by providing curative, rather than palliative, treatments. Healing the central nervous system (CNS) remains among regenerative medicine's most highly prized but formidable challenges. "Regenerative neurosurgery" provides access to the CNS or its surrounding structures to preserve or restore neurological function. Pioneering efforts over the past three decades have introduced cells, neurotrophins, and genes with putative regenerative capacity into the CNS to combat neurodegenerative, ischemic, and traumatic diseases. In this review we critically evaluate the rationale, paradigms, and translational progress of regenerative neurosurgery, harnessing access to the CNS to protect, rejuvenate, or replace cell types otherwise irreversibly compromised by neurological disease. We discuss the evidence surrounding fetal, somatic, and pluripotent stem cell derived implants to replace endogenous neuronal and glial cell types and provide trophic support. Neurotrophin based strategies via infusions and gene therapy highlight the motivation to preserve neuronal circuits, the complex fidelity of which cannot be readily recreated. We specifically highlight ongoing translational efforts in Parkinson's disease, amyotrophic lateral sclerosis, stroke, and spinal cord injury, using these to illustrate the principles, challenges, and opportunities of regenerative neurosurgery. Risks of associated procedures and novel neurosurgical trials are discussed, together with the ethical challenges they pose. After decades of efforts to develop and refine necessary tools and methodologies, regenerative neurosurgery is well positioned to advance treatments for refractory neurological diseases. Strategic multidisciplinary efforts will be critical to harness complementary technologies and maximize mechanistic feedback, accelerating iterative progress toward cures for neurological diseases.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
270
|
Bunevicius A, McDannold NJ, Golby AJ. Focused Ultrasound Strategies for Brain Tumor Therapy. Oper Neurosurg (Hagerstown) 2021; 19:9-18. [PMID: 31853548 DOI: 10.1093/ons/opz374] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/20/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND A key challenge in the medical treatment of brain tumors is the limited penetration of most chemotherapeutic agents across the blood-brain barrier (BBB) into the tumor and the infiltrative margin around the tumor. Magnetic resonance-guided focused ultrasound (MRgFUS) is a promising tool to enhance the delivery of chemotherapeutic agents into brain tumors. OBJECTIVE To review the mechanism of FUS, preclinical evidence, and clinical studies that used low-frequency FUS for a BBB opening in gliomas. METHODS Literature review. RESULTS The potential of externally delivered low-intensity ultrasound for a temporally and spatially precise and predictable disruption of the BBB has been investigated for over a decade, yielding extensive preclinical literature demonstrating that FUS can disrupt the BBB in a spatially targeted and temporally reversible manner. Studies in animal models documented that FUS enhanced the delivery of numerous chemotherapeutic and investigational agents across the BBB and into brain tumors, including temozolomide, bevacizumab, 1,3-bis (2-chloroethyl)-1-nitrosourea, doxorubicin, viral vectors, and cells. Chemotherapeutic interventions combined with FUS slowed tumor progression and improved animal survival. Recent advances of MRgFUS systems allow precise, temporally and spatially controllable, and safe transcranial delivery of ultrasound energy. Initial clinical evidence in glioma patients has shown the efficacy of MRgFUS in disrupting the BBB, as demonstrated by an enhanced gadolinium penetration. CONCLUSION Thus far, a temporary disruption of the BBB followed by the administration of chemotherapy has been both feasible and safe. Further studies are needed to determine the actual drug delivery, including the drug distribution at a tissue-level scale, as well as effects on tumor growth and patient prognosis.
Collapse
Affiliation(s)
- Adomas Bunevicius
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Nathan Judson McDannold
- Harvard Medical School, Harvard University, Boston, Massachusetts.,Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Alexandra J Golby
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Harvard University, Boston, Massachusetts.,Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
271
|
Chan MH, Chen W, Li CH, Fang CY, Chang YC, Wei DH, Liu RS, Hsiao M. An Advanced In Situ Magnetic Resonance Imaging and Ultrasonic Theranostics Nanocomposite Platform: Crossing the Blood-Brain Barrier and Improving the Suppression of Glioblastoma Using Iron-Platinum Nanoparticles in Nanobubbles. ACS APPLIED MATERIALS & INTERFACES 2021; 13:26759-26769. [PMID: 34076419 DOI: 10.1021/acsami.1c04990] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Glioblastoma (GBM) is one of the deadliest and most invasive brain cancers/gliomas, and there is currently no established way to treat this disease. The treatment of GBM typically involves intracranial surgery followed by chemotherapy. However, the blood-brain barrier (BBB) impedes the delivery of the chemotherapeutic drug, making the treatment challenging. In this study, we embedded a chemotherapeutic drug and other nanomaterials into a nanobubble (NB), utilized active tracking and other guidance mechanisms to guide the nanocomposite to the tumor site, and then used high-intensity focused ultrasound oscillation to burst the nanobubbles, generating a transient cavitation impact on the BBB and allowing the drug to bypass it and reach the brain. FePt enhances the resolution of T2-weighted magnetic resonance imaging images and has magnetic properties that help guide the nanocomposite to the tumor location. FePt nanoparticles were loaded into the hydrophobic core of the NBs along with doxorubicin to form a bubble-based drug delivery system (Dox-FePt@NB). The surface of the NBs is modified with a targeting ligand, transferrin (Dox-FePt@NB-Tf), giving the nanocomposite active tracking abilities. The Dox-FePt@NB-Tf developed in the present study represents a potential breakthrough in GBM treatment through improved drug delivery and biological imaging.
Collapse
Affiliation(s)
- Ming-Hsien Chan
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - William Chen
- Upper School, Taipei American School, Taipei 11152, Taiwan
| | - Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Chih-Yeu Fang
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Da-Hua Wei
- Graduate Institute of Manufacturing Technology and Department of Mechanical Engineering, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Ru-Shi Liu
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
- Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
272
|
Arsiwala TA, Sprowls SA, Blethen KE, Adkins CE, Saralkar PA, Fladeland RA, Pentz W, Gabriele A, Kielkowski B, Mehta RI, Wang P, Carpenter JS, Ranjan M, Najib U, Rezai AR, Lockman PR. Ultrasound-mediated disruption of the blood tumor barrier for improved therapeutic delivery. Neoplasia 2021; 23:676-691. [PMID: 34139452 PMCID: PMC8208897 DOI: 10.1016/j.neo.2021.04.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022] Open
Abstract
The blood-brain barrier (BBB) is a major anatomical and physiological barrier limiting the passage of drugs into brain. Central nervous system tumors can impair the BBB by changing the tumor microenvironment leading to the formation of a leaky barrier, known as the blood-tumor barrier (BTB). Despite the change in integrity, the BTB remains effective in preventing delivery of chemotherapy into brain tumors. Focused ultrasound is a unique noninvasive technique that can transiently disrupt the BBB and increase accumulation of drugs within targeted areas of the brain. Herein, we summarize the current understanding of different types of targeted ultrasound mediated BBB/BTB disruption techniques. We also discuss influence of the tumor microenvironment on BBB opening, as well as the role of immunological response following disruption. Lastly, we highlight the gaps between evaluation of the parameters governing opening of the BBB/BTB. A deeper understanding of physical opening of the BBB/BTB and the biological effects following disruption can potentially enhance treatment strategies for patients with brain tumors.
Collapse
Affiliation(s)
- T A Arsiwala
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, WV
| | - S A Sprowls
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, WV
| | - K E Blethen
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, WV
| | - C E Adkins
- School of Pharmacy, South University, Savannah, GA
| | - P A Saralkar
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, WV
| | - R A Fladeland
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, WV
| | - W Pentz
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, WV
| | - A Gabriele
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, WV
| | - B Kielkowski
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, WV
| | - R I Mehta
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV; Department of Neuroradiology, West Virginia University, Morgantown, WV; Department of Neuroscience, West Virginia University, Morgantown, WV
| | - P Wang
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV; Department of Neuroradiology, West Virginia University, Morgantown, WV
| | - J S Carpenter
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV; Department of Neuroradiology, West Virginia University, Morgantown, WV
| | - M Ranjan
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV; Departments of Neuroscience and Neurosurgery, West Virginia University, Morgantown, WV
| | - U Najib
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV; Department of Neurology, West Virginia University, Morgantown, WV
| | - A R Rezai
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV; Departments of Neuroscience and Neurosurgery, West Virginia University, Morgantown, WV
| | - P R Lockman
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, WV.
| |
Collapse
|
273
|
Sarkar S, Yang R, Mirzaei R, Rawji K, Poon C, Mishra MK, Zemp FJ, Bose P, Kelly J, Dunn JF, Yong VW. Control of brain tumor growth by reactivating myeloid cells with niacin. Sci Transl Med 2021; 12:12/537/eaay9924. [PMID: 32238578 DOI: 10.1126/scitranslmed.aay9924] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 03/09/2020] [Indexed: 12/11/2022]
Abstract
Glioblastomas are generally incurable partly because monocytes, macrophages, and microglia in afflicted patients do not function in an antitumor capacity. Medications that reactivate these macrophages/microglia, as well as circulating monocytes that become macrophages, could thus be useful to treat glioblastoma. We have discovered that niacin (vitamin B3) is a potential stimulator of these inefficient myeloid cells. Niacin-exposed monocytes attenuated the growth of brain tumor-initiating cells (BTICs) derived from glioblastoma patients by producing anti-proliferative interferon-α14. Niacin treatment of mice bearing intracranial BTICs increased macrophage/microglia representation within the tumor, reduced tumor size, and prolonged survival. These therapeutic outcomes were negated in mice depleted of circulating monocytes or harboring interferon-α receptor-deleted BTICs. Combination treatment with temozolomide enhanced niacin-promoted survival. Monocytes from glioblastoma patients had increased interferon-α14 upon niacin exposure and were reactivated to reduce BTIC growth in culture. We highlight niacin, a common vitamin that can be quickly translated into clinical application, as an immune stimulator against glioblastomas.
Collapse
Affiliation(s)
- Susobhan Sarkar
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Runze Yang
- Department of Radiology and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Reza Mirzaei
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Khalil Rawji
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Candice Poon
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Manoj K Mishra
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Franz J Zemp
- Department of Oncology and the Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Pinaki Bose
- Department of Oncology and the Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Surgery, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - John Kelly
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Oncology and the Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Jeff F Dunn
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Radiology and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada. .,Department of Oncology and the Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
274
|
Marin BM, Porath KA, Jain S, Kim M, Conage-Pough JE, Oh JH, Miller CL, Talele S, Kitange GJ, Tian S, Burgenske DM, Mladek AC, Gupta SK, Decker PA, McMinn MH, Stopka SA, Regan MS, He L, Carlson BL, Bakken K, Burns TC, Parney IF, Giannini C, Agar NYR, Eckel-Passow JE, Cochran JR, Elmquist WF, Vaubel RA, White FM, Sarkaria JN. Heterogeneous delivery across the blood-brain barrier limits the efficacy of an EGFR-targeting antibody drug conjugate in glioblastoma. Neuro Oncol 2021; 23:2042-2053. [PMID: 34050676 PMCID: PMC8643472 DOI: 10.1093/neuonc/noab133] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Antibody drug conjugates (ADCs) targeting the epidermal growth factor receptor (EGFR), such as depatuxizumab mafodotin (Depatux-M), is a promising therapeutic strategy for glioblastoma (GBM) but recent clinical trials did not demonstrate a survival benefit. Understanding the mechanisms of failure for this promising strategy is critically important. METHODS PDX models were employed to study efficacy of systemic vs intracranial delivery of Depatux-M. Immunofluorescence and MALDI-MSI were performed to detect drug levels in the brain. EGFR levels and compensatory pathways were studied using quantitative flow cytometry, Western blots, RNAseq, FISH, and phosphoproteomics. RESULTS Systemic delivery of Depatux-M was highly effective in nine of 10 EGFR-amplified heterotopic PDXs with survival extending beyond one year in eight PDXs. Acquired resistance in two PDXs (GBM12 and GBM46) was driven by suppression of EGFR expression or emergence of a novel short-variant of EGFR lacking the epitope for the Depatux-M antibody. In contrast to the profound benefit observed in heterotopic tumors, only two of seven intrinsically sensitive PDXs were responsive to Depatux-M as intracranial tumors. Poor efficacy in orthotopic PDXs was associated with limited and heterogeneous distribution of Depatux-M into tumor tissues, and artificial disruption of the BBB or bypass of the BBB by direct intracranial injection of Depatux-M into orthotopic tumors markedly enhanced the efficacy of drug treatment. CONCLUSIONS Despite profound intrinsic sensitivity to Depatux-M, limited drug delivery into brain tumor may have been a key contributor to lack of efficacy in recently failed clinical trials.
Collapse
Affiliation(s)
- Bianca-Maria Marin
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kendra A Porath
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sonia Jain
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Minjee Kim
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jason E Conage-Pough
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ju-Hee Oh
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Caitlyn L Miller
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Surabhi Talele
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Gaspar J Kitange
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Shulan Tian
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ann C Mladek
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Shiv K Gupta
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Paul A Decker
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Madison H McMinn
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA,Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, USA
| | - Sylwia A Stopka
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA,Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael S Regan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lihong He
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Brett L Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Katrina Bakken
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Terence C Burns
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ian F Parney
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Caterina Giannini
- Department of Laboratory Medicine and Pathology; Mayo Clinic, Rochester, Minnesota, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA,Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - William F Elmquist
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rachael A Vaubel
- Department of Laboratory Medicine and Pathology; Mayo Clinic, Rochester, Minnesota, USA
| | - Forest M White
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA,Corresponding Author: Jann N. Sarkaria, MD, Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Mayo Clinic, Rochester, MN 55902, USA ()
| |
Collapse
|
275
|
Wang Y, Zhang F, Xiong N, Xu H, Chai S, Wang H, Wang J, Zhao H, Jiang X, Fu P, Xiang W. Remodelling and Treatment of the Blood-Brain Barrier in Glioma. Cancer Manag Res 2021; 13:4217-4232. [PMID: 34079374 PMCID: PMC8166259 DOI: 10.2147/cmar.s288720] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 03/30/2021] [Indexed: 11/23/2022] Open
Abstract
The blood-brain barrier (BBB) is an essential structure of the central nervous system (CNS), and its existence makes the local internal environment of the CNS a relatively independent structure distinct from other internal environments of the human body to ensure normal physiological and high stability of activities of the CNS. Changes in BBB structure and function are fundamental to the pathophysiology of many diseases. The occurrence and development of glioma are often accompanied by a series of changes in the structure and function of the internal environment, the most significant of which is remodelling of the BBB. The remodelling of the BBB usually leads to changes in the permeability of local microvessels, which provide certain favourable conditions for the occurrence and development of glioma. Meanwhile, the newly generated abnormal blood vessels and the remaining intact regions of the BBB also hinder the effects of drug treatments. Changes in permeability and structural function often lead to the creation of abnormally functioning vascular regions, which pose further treatment challenges. At present, therapeutic methods for glioma have not achieved satisfactory effects in clinical practice, and emerging therapeutic methods have not yet been widely used in clinical practice. In this review, we summarize the knowledge of remodelling of the BBB in the glioma environment, the type of changes that occur, and current BBB treatment methods and prospects for the treatment of glioma.
Collapse
Affiliation(s)
- Yihao Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Fangcheng Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Nanxiang Xiong
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Hao Xu
- Department of Neurosurgery, General Hospital of the Yangtze River Shipping, Wuhan, 430022, People's Republic of China
| | - Songshan Chai
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Haofei Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Jiajing Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Hongyang Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Wei Xiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| |
Collapse
|
276
|
Patel B, Yang PH, Kim AH. The effect of thermal therapy on the blood-brain barrier and blood-tumor barrier. Int J Hyperthermia 2021; 37:35-43. [PMID: 32672118 DOI: 10.1080/02656736.2020.1783461] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The blood-brain and blood-tumor barriers represent highly specialized structures responsible for tight regulation of molecular transit into the central nervous system. Under normal circumstances, the relative impermeability of the blood-brain barrier (BBB) protects the brain from circulating toxins and contributes to a brain microenvironment necessary for optimal neuronal function. However, in the context of tumors and other diseases of central nervous system, the BBB and the more recently appreciated blood-tumor barrier (BTB) represent barriers that prevent effective drug delivery. Overcoming both barriers to optimize treatment of central nervous system diseases remains the subject of intense scientific investigation. Although many newer technologies have been developed to overcome these barriers, thermal therapy, which dates back to the 1890 s, has been known to disrupt the BBB since at least the early 1980s. Recently, as a result of several technological advances, laser interstitial thermal therapy (LITT), a method of delivering targeted thermal therapy, has gained widespread use as a surgical technique to ablate brain tumors. In addition, accumulating evidence indicates that laser ablation may also increase local BBB/BTB permeability after treatment. We herein review the structure and function of the BBB and BTB and the impact of thermal injury, including LITT, on barrier function.
Collapse
Affiliation(s)
- Bhuvic Patel
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter H Yang
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Albert H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
277
|
Maggs L, Cattaneo G, Dal AE, Moghaddam AS, Ferrone S. CAR T Cell-Based Immunotherapy for the Treatment of Glioblastoma. Front Neurosci 2021; 15:662064. [PMID: 34113233 PMCID: PMC8185049 DOI: 10.3389/fnins.2021.662064] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/14/2021] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive malignant primary brain tumor in adults. Current treatment options typically consist of surgery followed by chemotherapy or more frequently radiotherapy, however, median patient survival remains at just over 1 year. Therefore, the need for novel curative therapies for GBM is vital. Characterization of GBM cells has contributed to identify several molecules as targets for immunotherapy-based treatments such as EGFR/EGFRvIII, IL13Rα2, B7-H3, and CSPG4. Cytotoxic T lymphocytes collected from a patient can be genetically modified to express a chimeric antigen receptor (CAR) specific for an identified tumor antigen (TA). These CAR T cells can then be re-administered to the patient to identify and eliminate cancer cells. The impressive clinical responses to TA-specific CAR T cell-based therapies in patients with hematological malignancies have generated a lot of interest in the application of this strategy with solid tumors including GBM. Several clinical trials are evaluating TA-specific CAR T cells to treat GBM. Unfortunately, the efficacy of CAR T cells against solid tumors has been limited due to several factors. These include the immunosuppressive tumor microenvironment, inadequate trafficking and infiltration of CAR T cells and their lack of persistence and activity. In particular, GBM has specific limitations to overcome including acquired resistance to therapy, limited diffusion across the blood brain barrier and risks of central nervous system toxicity. Here we review current CAR T cell-based approaches for the treatment of GBM and summarize the mechanisms being explored in pre-clinical, as well as clinical studies to improve their anti-tumor activity.
Collapse
Affiliation(s)
- Luke Maggs
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | | | | | | | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
278
|
Kim C, Guo Y, Velalopoulou A, Leisen J, Motamarry A, Ramajayam K, Aryal M, Haemmerich D, Arvanitis CD. Closed-loop trans-skull ultrasound hyperthermia leads to improved drug delivery from thermosensitive drugs and promotes changes in vascular transport dynamics in brain tumors. Am J Cancer Res 2021; 11:7276-7293. [PMID: 34158850 PMCID: PMC8210606 DOI: 10.7150/thno.54630] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 04/25/2021] [Indexed: 12/11/2022] Open
Abstract
Effective drug delivery in brain tumors remains a major challenge in oncology. Although local hyperthermia and stimuli-responsive delivery systems, such as thermosensitive liposomes, represent promising strategies to locally enhance drug delivery in solid tumors and improve outcomes, their application in intracranial malignancies remains unexplored. We hypothesized that the combined abilities of closed-loop trans-skull Magnetic Resonance Imaging guided Focused Ultrasound (MRgFUS) hyperthermia with those of thermosensitive drugs can alleviate challenges in drug delivery and improve survival in gliomas. Methods: To conduct our investigations, we first designed a closed loop MR-guided Focused Ultrasound (MRgFUS) system for localized trans-skull hyperthermia (ΔT < 0.5 °C) in rodents and established safety thresholds in healthy mice. To assess the abilities of the developed system and proposed therapeutic strategy for FUS-triggered chemotherapy release we employed thermosensitive liposomal Dox (TSL-Dox) and tested it in two different glioma tumor models (F98 in rats and GL261 in mice). To quantify Dox delivery and changes in the transvascular transport dynamics in the tumor microenvironment we combined fluorescent microscopy, dynamic contrast enhanced MRI (DCE-MRI), and physiologically based pharmacokinetic (PBPK) modeling. Lastly, to assess the therapeutic efficacy of the system and of the proposed therapeutic strategy we performed a survival study in the GL261 glioma bearing mice. Results: The developed closed-loop trans-skull MRgFUS-hyperthermia system that operated at 1.7 MHz, a frequency that maximized the brain (FUS-focus) to skull temperature ratio in mice, was able to attain and maintain the desired focal temperature within a narrow range. Histological evidence (H&E and Nissl) suggests that focal temperature at 41.5 ± 0.5 °C for 10 min is below the threshold for tissue damage. Quantitative analysis of doxorubicin delivery from TSLs with MRgFUS-hyperthermia demonstrated 3.5-fold improvement in cellular uptake in GL261 glioma mouse tumors (p < 0.001) and 5-fold increase in delivery in F98 glioma rat tumors (p < 0.05), as compared to controls (TSL-Dox-only). Moreover, PBPK modeling of drug transport that was calibrated using the experimental data indicated that thermal stress could lead to significant improvement in the transvascular transport (2.3-fold increase in the vessel diffusion coefficient; P < 0.001), in addition to promoting targeted Dox release. Prospective experimental investigations with DCE-MRI during FUS-hyperthermia, supported these findings and provided evidence that moderate thermal stress (≈41 °C for up to 10 min) can promote acute changes in the vascular transport dynamics in the brain tumor microenvironment (Ktrans value for control vs. FUS was 0.0097 and 0.0148 min-1, respectively; p = 0.026). Crucially, survival analysis demonstrated significant improvement in the survival in the TSL-Dox-FUS group as compared to TSL-Dox-only group (p < 0.05), providing supporting evidence on the therapeutic potential of the proposed strategy. Conclusions: Our investigations demonstrated that spatially controlled thermal stress can be attained and sustained in the mouse brain, using a trans-skull closed-loop MRgFUS system, and used to promote the effective delivery of chemotherapy in gliomas from thermosensitive drugs. This system also allowed us to conduct mechanistic investigations that resulted in the refinement of our understanding on the role of thermal stress in augmenting mass and drug transport in brain tumors. Overall, our study established a new paradigm for effective drug delivery in brain tumors based on closed-loop ultrasound-mediated thermal stress and thermosensitive drugs.
Collapse
|
279
|
Kim H, Jin S, Choi H, Kang M, Park SG, Jun H, Cho H, Kang S. Target-switchable Gd(III)-DOTA/protein cage nanoparticle conjugates with multiple targeting affibody molecules as target selective T 1 contrast agents for high-field MRI. J Control Release 2021; 335:269-280. [PMID: 34044091 DOI: 10.1016/j.jconrel.2021.05.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022]
Abstract
Magnetic resonance imaging (MRI) is a non-invasive in vivo imaging tool, providing high enough spatial resolution to obtain both the anatomical and the physiological information of patients. However, MRI generally suffers from relatively low sensitivity often requiring the aid of contrast agents (CA) to enhance the contrast of vessels and/or the tissues of interest from the background. The targeted delivery of diagnostic probes to the specific lesion is a powerful approach for early diagnosis and signal enhancement leading to the effective treatment of various diseases. Here, we established targeting ligand switchable nanoplatforms using lumazine synthase protein cage nanoparticles derived from Aquifex aeolicus (AaLS) by genetically introducing the SpyTag peptide (ST) to the C-terminus of the AaLS subunits to form an ST-displaying AaLS (AaLS-ST). Conversely, multiple targeting ligands were constructed by genetically fusing SpyCatcher protein (SC) to either HER2 or EGFR targeting affibody molecules (SC-HER2Afb or SC-EGFRAfb). Gd(III)-DOTA complexes were chemically attached to the AaLS-ST and the external surface of the Gd(III)-DOTA conjugated AaLS-ST (Gd(III)-DOTA-AaLS-ST) were successfully decorated with either the HER2Afb or the EGFRAfb. The resulting Gd(III)-DOTA-AaLS/HER2Afb and Gd(III)-DOTA-AaLS/EGFR2Afb exhibited high r1 relaxivity values of 57 and 25 mM-1 s-1 at 1.4 and 7 T, respectively, which were 10-fold or higher than those of the clinically used Dotarem. Their target-selective contrast enhancements were confirmed with in vitro cell-based MRI scans and the in vivo MR imaging of tumor-bearing mouse models at 7 T. A target-switchable AaLS-based nanoplatform that was developed in this study might serve as a promising T1 CA developing platform at a high magnetic field to detect various tumor sites in a target-specific manner in future clinical applications.
Collapse
Affiliation(s)
- Hansol Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seokha Jin
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hyukjun Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - MungSoo Kang
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seong Guk Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Heejin Jun
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - HyungJoon Cho
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| | - Sebyung Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| |
Collapse
|
280
|
Wen PY. Positron emission tomography imaging of drug concentrations in the brain. Neuro Oncol 2021; 23:537-538. [PMID: 33704490 DOI: 10.1093/neuonc/noab025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Patrick Y Wen
- Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
281
|
Jucaite A, Stenkrona P, Cselényi Z, De Vita S, Buil-Bruna N, Varnäs K, Savage A, Varrone A, Johnström P, Schou M, Davison C, Sykes A, Pilla Reddy V, Hoch M, Vazquez-Romero A, Moein MM, Halldin C, Merchant MS, Pass M, Farde L. Brain exposure of the ATM inhibitor AZD1390 in humans-a positron emission tomography study. Neuro Oncol 2021; 23:687-696. [PMID: 33123736 DOI: 10.1093/neuonc/noaa238] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The protein kinase ataxia telangiectasia mutated (ATM) mediates cellular response to DNA damage induced by radiation. ATM inhibition decreases DNA damage repair in tumor cells and affects tumor growth. AZD1390 is a novel, highly potent, selective ATM inhibitor designed to cross the blood-brain barrier (BBB) and currently evaluated with radiotherapy in a phase I study in patients with brain malignancies. In the present study, PET was used to measure brain exposure of 11C-labeled AZD1390 after intravenous (i.v.) bolus administration in healthy subjects with an intact BBB. METHODS AZD1390 was radiolabeled with carbon-11 and a microdose (mean injected mass 1.21 µg) was injected in 8 male subjects (21-65 y). The radioactivity concentration of [11C]AZD1390 in brain was measured using a high-resolution PET system. Radioactivity in arterial blood was measured to obtain a metabolite corrected arterial input function for quantitative image analysis. Participants were monitored by laboratory examinations, vital signs, electrocardiogram, adverse events. RESULTS The brain radioactivity concentration of [11C]AZD1390 was 0.64 SUV (standard uptake value) and reached maximum 1.00% of injected dose at Tmax[brain] of 21 min (time of maximum brain radioactivity concentration) after i.v. injection. The whole brain total distribution volume was 5.20 mL*cm-3. No adverse events related to [11C]AZD1390 were reported. CONCLUSIONS This study demonstrates that [11C]AZD1390 crosses the intact BBB and supports development of AZD1390 for the treatment of glioblastoma multiforme or other brain malignancies. Moreover, it illustrates the potential of PET microdosing in predicting and guiding dose range and schedule for subsequent clinical studies.
Collapse
Affiliation(s)
- Aurelija Jucaite
- PET Science Centre, Precision Medicine and Biosamples, R&D, AstraZeneca, Stockholm, Sweden.,Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Per Stenkrona
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Zsolt Cselényi
- PET Science Centre, Precision Medicine and Biosamples, R&D, AstraZeneca, Stockholm, Sweden.,Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | | | - Nuria Buil-Bruna
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Katarina Varnäs
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | | | - Andrea Varrone
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Peter Johnström
- PET Science Centre, Precision Medicine and Biosamples, R&D, AstraZeneca, Stockholm, Sweden.,Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Magnus Schou
- PET Science Centre, Precision Medicine and Biosamples, R&D, AstraZeneca, Stockholm, Sweden.,Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | | | - Andy Sykes
- Oncology R&D, AstraZeneca, Cambridge, UK
| | | | - Matthias Hoch
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Ana Vazquez-Romero
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Mohammad Mahdi Moein
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | | | | | - Lars Farde
- PET Science Centre, Precision Medicine and Biosamples, R&D, AstraZeneca, Stockholm, Sweden.,Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
282
|
Genovesi LA, Puttick S, Millar A, Kojic M, Ji P, Lagendijk AK, Brighi C, Bonder CS, Adolphe C, Wainwright BJ. Patient-derived orthotopic xenograft models of medulloblastoma lack a functional blood-brain barrier. Neuro Oncol 2021; 23:732-742. [PMID: 33258962 DOI: 10.1093/neuonc/noaa266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Novel targeted therapies for children diagnosed with medulloblastoma (MB), the most common malignant pediatric brain tumor, are urgently required. A major hurdle in the development of effective therapies is the impaired delivery of systemic therapies to tumor cells due to a specialized endothelial blood-brain barrier (BBB). Accordingly, the integrity of the BBB is an essential consideration in any preclinical model used for assessing novel therapeutics. This study sought to assess the functional integrity of the BBB in several preclinical mouse models of MB. METHODS Dynamic contrast enhancement magnetic resonance imaging (MRI) was used to evaluate blood-brain-tumor barrier (BBTB) permeability in a murine genetically engineered mouse model (GEMM) of Sonic Hedgehog (SHH) MB, patient-derived orthotopic xenograft models of MB (SHH and Gp3), and orthotopic transplantation of GEMM tumor cells, enabling a comparison of the direct effects of transplantation on the integrity of the BBTB. Immunofluorescence analysis was performed to compare the structural and subcellular features of tumor-associated vasculature in all models. RESULTS Contrast enhancement was observed in all transplantation models of MB. No contrast enhancement was observed in the GEMM despite significant tumor burden. Cellular analysis of BBTB integrity revealed aberrancies in all transplantation models, correlating to the varying levels of BBTB permeability observed by MRI in these models. CONCLUSIONS These results highlight functional differences in the integrity of the BBTB and tumor vessel phenotype between commonly utilized preclinical models of MB, with important implications for the preclinical evaluation of novel therapeutic agents for MB.
Collapse
Affiliation(s)
- Laura A Genovesi
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Simon Puttick
- Probing Biosystems Future Science Platform, Commonwealth Scientific and Industrial Research Organization, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Amanda Millar
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Marija Kojic
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Pengxiang Ji
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Anne K Lagendijk
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Caterina Brighi
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland, Australia
| | - Claudine S Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Christelle Adolphe
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Brandon J Wainwright
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
283
|
Yu MW, Quail DF. Immunotherapy for Glioblastoma: Current Progress and Challenge. Front Immunol 2021; 12:676301. [PMID: 34054867 PMCID: PMC8158294 DOI: 10.3389/fimmu.2021.676301] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is a highly lethal brain cancer with a median survival rate of less than 15 months when treated with the current standard of care, which consists of surgery, radiotherapy and chemotherapy. With the recent success of immunotherapy in other aggressive cancers such as advanced melanoma and advanced non-small cell lung cancer, glioblastoma has been brought to the forefront of immunotherapy research. Resistance to therapy has been a major challenge across a multitude of experimental candidates and no immunotherapies have been approved for glioblastoma to-date. Intra- and inter-tumoral heterogeneity, an inherently immunosuppressive environment and tumor plasticity remain barriers to be overcome. Moreover, the unique tissue-specific interactions between the central nervous system and the peripheral immune system present an additional challenge for immune-based therapies. Nevertheless, there is sufficient evidence that these challenges may be overcome, and immunotherapy continues to be actively pursued in glioblastoma. Herein, we review the primary ongoing immunotherapy candidates for glioblastoma with a focus on immune checkpoint inhibitors, myeloid-targeted therapies, vaccines and chimeric antigen receptor (CAR) immunotherapies. We further provide insight on mechanisms of resistance and how our understanding of these mechanisms may pave the way for more effective immunotherapeutics against glioblastoma.
Collapse
Affiliation(s)
- Miranda W Yu
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Physiology, McGill University, Montreal, QC, Canada
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Physiology, McGill University, Montreal, QC, Canada.,Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
284
|
Radiomics-based neural network predicts recurrence patterns in glioblastoma using dynamic susceptibility contrast-enhanced MRI. Sci Rep 2021; 11:9974. [PMID: 33976264 PMCID: PMC8113258 DOI: 10.1038/s41598-021-89218-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/22/2021] [Indexed: 02/03/2023] Open
Abstract
Glioblastoma remains the most devastating brain tumor despite optimal treatment, because of the high rate of recurrence. Distant recurrence has distinct genomic alterations compared to local recurrence, which requires different treatment planning both in clinical practice and trials. To date, perfusion-weighted MRI has revealed that perfusional characteristics of tumor are associated with prognosis. However, not much research has focused on recurrence patterns in glioblastoma: namely, local and distant recurrence. Here, we propose two different neural network models to predict the recurrence patterns in glioblastoma that utilizes high-dimensional radiomic profiles based on perfusion MRI: area under the curve (AUC) (95% confidence interval), 0.969 (0.903-1.000) for local recurrence; 0.864 (0.726-0.976) for distant recurrence for each patient in the validation set. This creates an opportunity to provide personalized medicine in contrast to studies investigating only group differences. Moreover, interpretable deep learning identified that salient radiomic features for each recurrence pattern are related to perfusional intratumoral heterogeneity. We also demonstrated that the combined salient radiomic features, or "radiomic risk score", increased risk of recurrence/progression (hazard ratio, 1.61; p = 0.03) in multivariate Cox regression on progression-free survival.
Collapse
|
285
|
Blood-brain barrier opening by intracarotid artery hyperosmolar mannitol induces sterile inflammatory and innate immune responses. Proc Natl Acad Sci U S A 2021; 118:2021915118. [PMID: 33906946 DOI: 10.1073/pnas.2021915118] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intracarotid arterial hyperosmolar mannitol (ICAHM) blood-brain barrier disruption (BBBD) is effective and safe for delivery of therapeutics for central nervous system malignancies. ICAHM osmotically alters endothelial cells and tight junction integrity to achieve BBBD. However, occurrence of neuroinflammation following hemispheric BBBD by ICAHM remains unknown. Temporal proteomic changes in rat brains following ICAHM included increased damage-associated molecular patterns, cytokines, chemokines, trophic factors, and cell adhesion molecules, indicative of a sterile inflammatory response (SIR). Proteomic changes occurred within 5 min of ICAHM infusion and returned to baseline by 96 h. Transcriptomic analyses following ICAHM BBBD further supported an SIR. Immunohistochemistry revealed activated astrocytes, microglia, and macrophages. Moreover, proinflammatory proteins were elevated in serum, and proteomic and histological findings from the contralateral hemisphere demonstrated a less pronounced SIR, suggesting neuroinflammation beyond regions of ICAHM infusion. Collectively, these results demonstrate ICAHM induces a transient SIR that could potentially be harnessed for neuroimmunomodulation.
Collapse
|
286
|
Wala K, Szlasa W, Saczko J, Rudno-Rudzińska J, Kulbacka J. Modulation of Blood-Brain Barrier Permeability by Activating Adenosine A2 Receptors in Oncological Treatment. Biomolecules 2021; 11:biom11050633. [PMID: 33923147 PMCID: PMC8146369 DOI: 10.3390/biom11050633] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022] Open
Abstract
The blood–brain barrier (BBB) plays an important protective role in the central nervous system and maintains its homeostasis. It regulates transport into brain tissue and protects neurons against the toxic effects of substances circulating in the blood. However, in the case of neurological diseases or primary brain tumors, i.e., gliomas, the higher permeability of the blood-derived substances in the brain tissue is necessary. Currently applied methods of treatment for the primary brain neoplasms include surgical removal of the tumor, radiation therapy, and chemotherapy. Despite the abovementioned treatment methods, the prognosis of primary brain tumors remains bad. Moreover, chemotherapy options seem to be limited due to low drug penetration into the cancerous tissue. Modulation of the blood–brain barrier permeability may contribute to an increase in the concentration of the drug in the CNS and thus increase the effectiveness of therapy. Interestingly, endothelial cells in cerebral vessels are characterized by the presence of adenosine 2A receptors (A2AR). It has been shown that substances affecting these receptors regulate the permeability of the BBB. The mechanism of increasing the BBB permeability by A2AR agonists is the actin-cytoskeletal reorganization and acting on the tight junctions. In this case, the A2AR seems to be a promising therapy target. This article aims to assess the possibility of increasing the BBB permeability through A2AR agonists to increase the effectiveness of chemotherapy and to improve the results of cancer therapy.
Collapse
Affiliation(s)
- Kamila Wala
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.W.); (W.S.)
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.W.); (W.S.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Julia Rudno-Rudzińska
- Department of General and Oncological Surgery, Medical University Hospital, Borowska 213, 50-556 Wrocław, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
- Correspondence: ; Tel.: +48-784-06-92
| |
Collapse
|
287
|
Hay BA, Godugu K, Darwish NHE, Fujioka K, Sudha T, Karakus OO, Mousa SA. New Thyrointegrin α vβ 3 Antagonist with a Scalable Synthesis, Brain Penetration, and Potent Activity against Glioblastoma Multiforme. J Med Chem 2021; 64:6300-6309. [PMID: 33886292 DOI: 10.1021/acs.jmedchem.1c00350] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We have previously reported that the αvβ3 inhibitor P-bi-TAT, a bifunctional version of the thyroid hormone metabolite tetraiodothyroacetic acid (tetrac) conjugated to polyethylene glycol (PEG) MW 4000, has excellent efficacy in a glioblastoma multiforme (GBM) mouse model. However, bioanalysis problems due to PEG polydispersity and large-scale synthesis issues led to a search for new molecules, culminating in the discovery of fb-PMT, a conjugate of tetrac and monodisperse PEG36, with a lipophilic 4-fluorobenzyl group at the opposite end of the PEG chain. fb-PMT reduces GBM tumor growth and viability by up to 98%, is suitable for large-scale synthesis, and is amenable to bioanalysis using mass spectrometry-based detection. We also showed that changes in lipophilicity at the opposite end of the PEG chain from the active tetrac component affected the proton NMR chemical shift of the tetrac moiety in D20 and brain levels of the compound after subcutaneous dosing.
Collapse
Affiliation(s)
- Bruce A Hay
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive (Room 238), Rensselaer 12144, New York, United States
| | - Kavitha Godugu
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive (Room 238), Rensselaer 12144, New York, United States
| | - Noureldien H E Darwish
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive (Room 238), Rensselaer 12144, New York, United States
| | - Kazutoshi Fujioka
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive (Room 238), Rensselaer 12144, New York, United States
| | - Thangirala Sudha
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive (Room 238), Rensselaer 12144, New York, United States
| | - Ozlem Ozen Karakus
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive (Room 238), Rensselaer 12144, New York, United States
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive (Room 238), Rensselaer 12144, New York, United States
| |
Collapse
|
288
|
Reddy S, Tatiparti K, Sau S, Iyer AK. Recent advances in nano delivery systems for blood-brain barrier (BBB) penetration and targeting of brain tumors. Drug Discov Today 2021; 26:1944-1952. [PMID: 33865978 DOI: 10.1016/j.drudis.2021.04.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/24/2021] [Accepted: 04/08/2021] [Indexed: 02/09/2023]
Abstract
Gliomas constitute about 80% of brain tumors and have a meager two-year survival rate. The treatment options available are very few because of poor prognosis and a lack of targeted nanodelivery systems that can cross the blood-brain barrier (BBB) and the blood-tumor barrier. This short review attempts to clarify the challenges for delivery systems designed to cross the BBB, and provides a brief description of the different types of targeted nanodelivery system that have shown potential for success in delivering drugs to the brain. Further, this review describes the most recent studies that have developed nanoparticles for brain delivery in the past five years. We also provide an insight into the most recent clinical trials designed to assess the efficacy of these nanodelivery systems for glioma.
Collapse
Affiliation(s)
- Shriya Reddy
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; Northville High School, Northville, MI 48168, USA
| | - Katyayani Tatiparti
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Samaresh Sau
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; Molecular Imaging Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
289
|
Guo Y, Lee H, Fang Z, Velalopoulou A, Kim J, Thomas MB, Liu J, Abramowitz RG, Kim Y, Coskun AF, Krummel DP, Sengupta S, MacDonald TJ, Arvanitis C. Single-cell analysis reveals effective siRNA delivery in brain tumors with microbubble-enhanced ultrasound and cationic nanoparticles. SCIENCE ADVANCES 2021; 7:7/18/eabf7390. [PMID: 33931452 PMCID: PMC8087400 DOI: 10.1126/sciadv.abf7390] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/12/2021] [Indexed: 05/08/2023]
Abstract
RNA-based therapies offer unique advantages for treating brain tumors. However, tumor penetrance and uptake are hampered by RNA therapeutic size, charge, and need to be "packaged" in large carriers to improve bioavailability. Here, we have examined delivery of siRNA, packaged in 50-nm cationic lipid-polymer hybrid nanoparticles (LPHs:siRNA), combined with microbubble-enhanced focused ultrasound (MB-FUS) in pediatric and adult preclinical brain tumor models. Using single-cell image analysis, we show that MB-FUS in combination with LPHs:siRNA leads to more than 10-fold improvement in siRNA delivery into brain tumor microenvironments of the two models. MB-FUS delivery of Smoothened (SMO) targeting siRNAs reduces SMO protein production and markedly increases tumor cell death in the SMO-activated medulloblastoma model. Moreover, our analysis reveals that MB-FUS and nanoparticle properties can be optimized to maximize delivery in the brain tumor microenvironment, thereby serving as a platform for developing next-generation tunable delivery systems for RNA-based therapy in brain tumors.
Collapse
Affiliation(s)
- Yutong Guo
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hohyun Lee
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhou Fang
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Anastasia Velalopoulou
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jinhwan Kim
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Midhun Ben Thomas
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jingbo Liu
- Department of Pediatrics, Aflac Cancer and Blood, Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan G Abramowitz
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - YongTae Kim
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Daniel Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tobey J MacDonald
- Department of Pediatrics, Aflac Cancer and Blood, Emory University School of Medicine, Atlanta, GA, USA
| | - Costas Arvanitis
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| |
Collapse
|
290
|
Kizilbash SH, Gupta SK, Parrish KE, Laramy JK, Kim M, Gampa G, Carlson BL, Bakken KK, Mladek AC, Schroeder MA, Decker PA, Elmquist WF, Sarkaria JN. In Vivo Efficacy of Tesevatinib in EGFR-Amplified Patient-Derived Xenograft Glioblastoma Models May Be Limited by Tissue Binding and Compensatory Signaling. Mol Cancer Ther 2021; 20:1009-1018. [PMID: 33785646 DOI: 10.1158/1535-7163.mct-20-0640] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 02/02/2021] [Accepted: 03/23/2021] [Indexed: 11/16/2022]
Abstract
Tesevatinib is a potent oral brain penetrant EGFR inhibitor currently being evaluated for glioblastoma therapy. Tesevatinib distribution was assessed in wild-type (WT) and Mdr1a/b(-/-)Bcrp(-/-) triple knockout (TKO) FVB mice after dosing orally or via osmotic minipump; drug-tissue binding was assessed by rapid equilibrium dialysis. Two hours after tesevatinib dosing, brain concentrations in WT and TKO mice were 0.72 and 10.03 μg/g, respectively. Brain-to-plasma ratios (Kp) were 0.53 and 5.73, respectively. With intraperitoneal infusion, brain concentrations were 1.46 and 30.6 μg/g (Kp 1.16 and 25.10), respectively. The brain-to-plasma unbound drug concentration ratios were substantially lower (WT mice, 0.03-0.08; TKO mice, 0.40-1.75). Unbound drug concentrations in brains of WT mice were 0.78 to 1.59 ng/g. In vitro cytotoxicity and EGFR pathway signaling were evaluated using EGFR-amplified patient-derived glioblastoma xenograft models (GBM12, GBM6). In vivo pharmacodynamics and efficacy were assessed using athymic nude mice bearing either intracranial or flank tumors treated by oral gavage. Tesevatinib potently reduced cell viability [IC50 GBM12 = 11 nmol/L (5.5 ng/mL), GBM6 = 102 nmol/L] and suppressed EGFR signaling in vitro However, tesevatinib efficacy compared with vehicle in intracranial (GBM12, median survival: 23 vs. 18 days, P = 0.003) and flank models (GBM12, median time to outcome: 41 vs. 33 days, P = 0.007; GBM6, 44 vs. 33 days, P = 0.007) was modest and associated with partial inhibition of EGFR signaling. Overall, tesevatinib efficacy in EGFR-amplified PDX GBM models is robust in vitro but relatively modest in vivo, despite a high brain-to-plasma ratio. This discrepancy may be explained by drug-tissue binding and compensatory signaling.
Collapse
Affiliation(s)
| | - Shiv K Gupta
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Karen E Parrish
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Janice K Laramy
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Minjee Kim
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Gautham Gampa
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Brett L Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Katrina K Bakken
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Ann C Mladek
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Mark A Schroeder
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Paul A Decker
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - William F Elmquist
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
291
|
Griffith JI, Sarkaria JN, Elmquist WF. Efflux Limits Tumor Drug Delivery Despite Disrupted BBB. Trends Pharmacol Sci 2021; 42:426-428. [PMID: 33736874 DOI: 10.1016/j.tips.2021.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Apparent blood-brain barrier (BBB) disruption is common in glioblastoma (GBM), but has not translated to improved drug delivery efficacy. Recently, de Gooijer et al. demonstrated that efflux transporters can have a prominent role in limiting drug delivery. These transport systems contribute to ineffective drug delivery to tumor cells in the brain.
Collapse
Affiliation(s)
- Jessica I Griffith
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
292
|
Baranyai Z, Biri-Kovács B, Krátký M, Szeder B, Debreczeni ML, Budai J, Kovács B, Horváth L, Pári E, Németh Z, Cervenak L, Zsila F, Méhes E, Kiss É, Vinšová J, Bősze S. Cellular Internalization and Inhibition Capacity of New Anti-Glioma Peptide Conjugates: Physicochemical Characterization and Evaluation on Various Monolayer- and 3D-Spheroid-Based in Vitro Platforms. J Med Chem 2021; 64:2982-3005. [PMID: 33719423 DOI: 10.1021/acs.jmedchem.0c01399] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Most therapeutic agents used for treating brain malignancies face hindered transport through the blood-brain barrier (BBB) and poor tissue penetration. To overcome these problems, we developed peptide conjugates of conventional and experimental anticancer agents. SynB3 cell-penetrating peptide derivatives were applied that can cross the BBB. Tuftsin derivatives were used to target the neuropilin-1 transport system for selectivity and better tumor penetration. Moreover, SynB3-tuftsin tandem compounds were synthesized to combine the beneficial properties of these peptides. Most of the conjugates showed high and selective efficacy against glioblastoma cells. SynB3 and tandem derivatives demonstrated superior cellular internalization. The penetration profile of the conjugates was determined on a lipid monolayer and Transwell co-culture system with noncontact HUVEC-U87 monolayers as simple ex vivo and in vitro BBB models. Importantly, in 3D spheroids, daunomycin-peptide conjugates possessed a better tumor penetration ability than daunomycin. These conjugates are promising tools for the delivery systems with tunable features.
Collapse
Affiliation(s)
- Zsuzsa Baranyai
- Eötvös Loránd Research Network, Research Group of Peptide Chemistry, Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| | - Beáta Biri-Kovács
- Eötvös Loránd Research Network, Research Group of Peptide Chemistry, Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary.,Institute of Chemistry, Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| | - Martin Krátký
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Bálint Szeder
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary
| | - Márta L Debreczeni
- 3rd Department of Medicine Research Laboratory, Semmelweis University, Kútvölgyi út 4, H-1125 Budapest, Hungary
| | - Johanna Budai
- Eötvös Loránd Research Network, Research Group of Peptide Chemistry, Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| | - Bence Kovács
- Centre for Ecological Research, Institute of Ecology and Botany, Alkotmány u. 2-4, H-2163 Vácrátót, Hungary
| | - Lilla Horváth
- Eötvös Loránd Research Network, Research Group of Peptide Chemistry, Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| | - Edit Pári
- Laboratory of Interfaces and Nanostructures, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| | - Zsuzsanna Németh
- 3rd Department of Medicine Research Laboratory, Semmelweis University, Kútvölgyi út 4, H-1125 Budapest, Hungary
| | - László Cervenak
- 3rd Department of Medicine Research Laboratory, Semmelweis University, Kútvölgyi út 4, H-1125 Budapest, Hungary
| | - Ferenc Zsila
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary
| | - Előd Méhes
- Department of Biological Physics, Institute of Physics, Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| | - Éva Kiss
- Laboratory of Interfaces and Nanostructures, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| | - Jarmila Vinšová
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Szilvia Bősze
- Eötvös Loránd Research Network, Research Group of Peptide Chemistry, Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| |
Collapse
|
293
|
Banerjee K, Núñez FJ, Haase S, McClellan BL, Faisal SM, Carney SV, Yu J, Alghamri MS, Asad AS, Candia AJN, Varela ML, Candolfi M, Lowenstein PR, Castro MG. Current Approaches for Glioma Gene Therapy and Virotherapy. Front Mol Neurosci 2021; 14:621831. [PMID: 33790740 PMCID: PMC8006286 DOI: 10.3389/fnmol.2021.621831] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor in the adult population and it carries a dismal prognosis. Inefficient drug delivery across the blood brain barrier (BBB), an immunosuppressive tumor microenvironment (TME) and development of drug resistance are key barriers to successful glioma treatment. Since gliomas occur through sequential acquisition of genetic alterations, gene therapy, which enables to modification of the genetic make-up of target cells, appears to be a promising approach to overcome the obstacles encountered by current therapeutic strategies. Gene therapy is a rapidly evolving field with the ultimate goal of achieving specific delivery of therapeutic molecules using either viral or non-viral delivery vehicles. Gene therapy can also be used to enhance immune responses to tumor antigens, reprogram the TME aiming at blocking glioma-mediated immunosuppression and normalize angiogenesis. Nano-particles-mediated gene therapy is currently being developed to overcome the BBB for glioma treatment. Another approach to enhance the anti-glioma efficacy is the implementation of viro-immunotherapy using oncolytic viruses, which are immunogenic. Oncolytic viruses kill tumor cells due to cancer cell-specific viral replication, and can also initiate an anti-tumor immunity. However, concerns still remain related to off target effects, and therapeutic and transduction efficiency. In this review, we describe the rationale and strategies as well as advantages and disadvantages of current gene therapy approaches against gliomas in clinical and preclinical studies. This includes different delivery systems comprising of viral, and non-viral delivery platforms along with suicide/prodrug, oncolytic, cytokine, and tumor suppressor-mediated gene therapy approaches. In addition, advances in glioma treatment through BBB-disruptive gene therapy and anti-EGFRvIII/VEGFR gene therapy are also discussed. Finally, we discuss the results of gene therapy-mediated human clinical trials for gliomas. In summary, we highlight the progress, prospects and remaining challenges of gene therapies aiming at broadening our understanding and highlighting the therapeutic arsenal for GBM.
Collapse
Affiliation(s)
- Kaushik Banerjee
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Felipe J. Núñez
- Laboratory of Molecular and Cellular Therapy, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Brandon L. McClellan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Immunology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Syed M. Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Stephen V. Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Cancer Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jin Yu
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Antonela S. Asad
- Departamento de Biología e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro J. Nicola Candia
- Departamento de Biología e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria Luisa Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Marianela Candolfi
- Departamento de Biología e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
294
|
Chien CH, Hsueh WT, Chuang JY, Chang KY. Dissecting the mechanism of temozolomide resistance and its association with the regulatory roles of intracellular reactive oxygen species in glioblastoma. J Biomed Sci 2021; 28:18. [PMID: 33685470 PMCID: PMC7938520 DOI: 10.1186/s12929-021-00717-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is the most common primary malignant brain tumor that is usually considered fatal even with treatment. This is often a result for tumor to develop resistance. Regarding the standard chemotherapy, the alkylating agent temozolomide is effective in disease control but the recurrence will still occur eventually. The mechanism of the resistance is various, and differs in terms of innate or acquired. To date, aberrations in O6-methylguanine-DNA methyltransferase are the clear factor that determines drug susceptibility. Alterations of the other DNA damage repair genes such as DNA mismatch repair genes are also known to affect the drug effect. Together these genes have roles in the innate resistance, but are not sufficient for explaining the mechanism leading to acquired resistance. Recent identification of specific cellular subsets with features of stem-like cells may have role in this process. The glioma stem-like cells are known for its superior ability in withstanding the drug-induced cytotoxicity, and giving the chance to repopulate the tumor. The mechanism is complicated to administrate cellular protection, such as the enhancing ability against reactive oxygen species and altering energy metabolism, the important steps to survive. In this review, we discuss the possible mechanism for these specific cellular subsets to evade cancer treatment, and the possible impact to the following treatment courses. In addition, we also discuss the possibility that can overcome this obstacle.
Collapse
Affiliation(s)
- Chia-Hung Chien
- National Institute of Cancer Research, National Health Research Institutes, 367 Sheng-Li Road, Tainan, 70456, Taiwan
| | - Wei-Ting Hsueh
- Department of Oncology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Jian-Ying Chuang
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kwang-Yu Chang
- National Institute of Cancer Research, National Health Research Institutes, 367 Sheng-Li Road, Tainan, 70456, Taiwan. .,Department of Oncology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
295
|
Weber CM, Clyne AM. Sex differences in the blood-brain barrier and neurodegenerative diseases. APL Bioeng 2021; 5:011509. [PMID: 33758788 PMCID: PMC7968933 DOI: 10.1063/5.0035610] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
The number of people diagnosed with neurodegenerative diseases is on the rise. Many of these diseases, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, and motor neuron disease, demonstrate clear sexual dimorphisms. While sex as a biological variable must now be included in animal studies, sex is rarely included in in vitro models of human neurodegenerative disease. In this Review, we describe these sex-related differences in neurodegenerative diseases and the blood-brain barrier (BBB), whose dysfunction is linked to neurodegenerative disease development and progression. We explain potential mechanisms by which sex and sex hormones affect BBB integrity. Finally, we summarize current in vitro BBB bioengineered models and highlight their potential to study sex differences in BBB integrity and neurodegenerative disease.
Collapse
Affiliation(s)
- Callie M. Weber
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA
| |
Collapse
|
296
|
Tripathi SK, Kean R, Bongiorno E, Hooper DC, Jin YY, Wickstrom E, McCue PA, Thakur ML. Targeting VPAC1 Receptors for Imaging Glioblastoma. Mol Imaging Biol 2021; 22:293-302. [PMID: 31292914 DOI: 10.1007/s11307-019-01388-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Scintigraphic imaging of malignant glioblastoma (MG) continues to be challenging. We hypothesized that VPAC1 cell surface receptors can be targeted for positron emission tomography (PET) imaging of orthotopically implanted MG in a mouse model, using a VPAC1-specific peptide [64Cu]TP3805. PROCEDURES The expression of VPAC1 in mouse GL261 and human U87 glioma cell lines was determined by western blot. The ability of [64Cu]TP3805 to bind to GL261 and U87 cells was studied by cell-binding. Receptor-blocking studies were performed to validate receptor specificity. GL261 tumors were implanted orthotopically in syngeneic T-bet knockout C57BL/6 mouse brain (N = 15) and allowed to grow for 2-3 weeks. Mice were injected i.v., first with ~ 150 μCi of 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) then 24 h later with ~ 200 μCi of [64Cu]TP3805. In another set of tumor-bearing mice, (N = 5), ionic [64Cu]Cl2 was injected as a control. Mice were imaged at a 2-h post-injection using an Inveon micro-PET/CT, sacrificed and % ID/g of [64Cu]TP3805 and [64Cu]Cl2 were calculated in a tumor, normal brain, and other tissues. For histologic tissue examination, 3-μm thick sections of the tumors and normal brain were prepared, digital autoradiography (DAR) was performed, and then the sections were H&E stained for histologic examination. RESULTS Western blots showed a strong signal for VPAC1 on both cell lines. [64Cu]TP3805 cell-binding was 87 ± 1.5 %. Receptor-blocking reduced cell-binding to 24.3 ± 1.5 % (P < 0.01). PET imaging revealed remarkable accumulation of [64Cu]TP3805 in GL261 MG with a negligible background in the normal brain, as compared to [18F]FDG. Micro-PET/CT image analyses and tissue distribution showed that the brain tumor uptake for [64Cu]TP3805 was 8.2 ± 1.7 % ID/g and for [64Cu]Cl2 2.1 ± 0.5 % ID/g as compared to 1.0 ± 0.3 % ID/g and 1.4 ± 0.3 % ID/g for normal mouse brains, respectively. The high tumor/normal brain ratio for [64Cu]TP3805 (8.1 ± 1.1) allowed tumors to be visualized unequivocally. Histology and [64Cu]TP3805 DAR differentiated malignant tumors from healthy brain and confirmed PET findings. CONCLUSION Targeting VPAC1 receptors using [64Cu]TP3805 for PET imaging of MG is a promising novel approach and calls for further investigation.
Collapse
Affiliation(s)
- Sushil K Tripathi
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rhonda Kean
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Emily Bongiorno
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Douglas C Hooper
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Eric Wickstrom
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peter A McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mathew L Thakur
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Radiology, Laboratories of Radiopharmaceutical Research and Molecular Imaging, 1020 Locust Street, 359-JAH, Philadelphia, PA, 19107, USA.
| |
Collapse
|
297
|
D'Amico RS, Aghi MK, Vogelbaum MA, Bruce JN. Convection-enhanced drug delivery for glioblastoma: a review. J Neurooncol 2021; 151:415-427. [PMID: 33611708 DOI: 10.1007/s11060-020-03408-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/18/2020] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Convection-enhanced delivery (CED) is a method of targeted, local drug delivery to the central nervous system (CNS) that bypasses the blood-brain barrier (BBB) and permits the delivery of high-dose therapeutics to large volumes of interest while limiting associated systemic toxicities. Since its inception, CED has undergone considerable preclinical and clinical study as a safe method for treating glioblastoma (GBM). However, the heterogeneity of both, the surgical procedure and the mechanisms of action of the agents studied-combined with the additional costs of performing a trial evaluating CED-has limited the field's ability to adequately assess the durability of any potential anti-tumor responses. As a result, the long-term efficacy of the agents studied to date remains difficult to assess. MATERIALS AND METHODS We searched PubMed using the phrase "convection-enhanced delivery and glioblastoma". The references of significant systematic reviews were also reviewed for additional sources. Articles focusing on physiological and physical mechanisms of CED were included as well as technological CED advances. RESULTS We review the history and principles of CED, procedural advancements and characteristics, and outcomes from key clinical trials, as well as discuss the potential future of this promising technique for the treatment of GBM. CONCLUSION While the long-term efficacy of the agents studied to date remains difficult to assess, CED remains a promising technique for the treatment of GBM.
Collapse
Affiliation(s)
- Randy S D'Amico
- Department of Neurological Surgery, Lenox Hill Hospital/Northwell Health, New York, NY, USA.
| | - Manish K Aghi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Jeffrey N Bruce
- Department of Neurological Surgery, New York Presbyterian/Columbia University Irving Medical Center, Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| |
Collapse
|
298
|
Abstract
Chimeric antigen receptor T (CAR-T) cells, an immunotherapy that demonstrates marked success in treatment of hematologic malignancies, are an emergent therapeutic for patients with glioblastoma (GBM). GBM CAR-T trials have focused on targeting well-characterized antigens in the pathogenesis of GBM. Early stage trials demonstrate initial success in terms of safety and tolerability. There is preliminary evidence of antitumor activity and localization of the CAR-T product to tumoral sites. There are mixed results regarding patient outcomes. Ongoing GBM CAR-T trials will target novel antigens, explore CAR-T combination therapy, design multivalent CAR constructs, and assess the impact of lymphodepletion before CAR-T delivery.
Collapse
Affiliation(s)
- Thilan Tudor
- University of Pennsylvania, 3600 Hamilton Walk, Stemmler Hall, Room 176, Philadelphia, PA 19104
| | - Zev A Binder
- University of Pennsylvania, 3600 Hamilton Walk, Stemmler Hall, Room 176, Philadelphia, PA 19104.
| | - Donald M O'Rourke
- John Templeton, Jr. M.D. Professor in Neurosurgery, Hospital of the University of Pennsylvania, 3400 Spruce St. Philadelphia, PA 19104, USA. https://twitter.com/DrORourke2
| |
Collapse
|
299
|
Nguyen HM, Guz-Montgomery K, Lowe DB, Saha D. Pathogenetic Features and Current Management of Glioblastoma. Cancers (Basel) 2021; 13:cancers13040856. [PMID: 33670551 PMCID: PMC7922739 DOI: 10.3390/cancers13040856] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is the most common form of primary malignant brain tumor with a devastatingly poor prognosis. The disease does not discriminate, affecting adults and children of both sexes, and has an average overall survival of 12-15 months, despite advances in diagnosis and rigorous treatment with chemotherapy, radiation therapy, and surgical resection. In addition, most survivors will eventually experience tumor recurrence that only imparts survival of a few months. GBM is highly heterogenous, invasive, vascularized, and almost always inaccessible for treatment. Based on all these outstanding obstacles, there have been tremendous efforts to develop alternative treatment options that allow for more efficient targeting of the tumor including small molecule drugs and immunotherapies. A number of other strategies in development include therapies based on nanoparticles, light, extracellular vesicles, and micro-RNA, and vessel co-option. Advances in these potential approaches shed a promising outlook on the future of GBM treatment. In this review, we briefly discuss the current understanding of adult GBM's pathogenetic features that promote treatment resistance. We also outline novel and promising targeted agents currently under development for GBM patients during the last few years with their current clinical status.
Collapse
|
300
|
Schaffenrath J, Wyss T, He L, Rushing EJ, Delorenzi M, Vasella F, Regli L, Neidert MC, Keller A. Blood-brain barrier alterations in human brain tumors revealed by genome-wide transcriptomic profiling. Neuro Oncol 2021; 23:2095-2106. [PMID: 33560373 DOI: 10.1093/neuonc/noab022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Brain tumors, whether primary or secondary, have limited therapeutic options despite advances in understanding driver gene mutations and heterogeneity within tumor cells. The cellular and molecular composition of brain tumor stroma, an important modifier of tumor growth, has been less investigated to date. Only few studies have focused on the vasculature of human brain tumors despite the fact that the blood-brain barrier (BBB) represents the major obstacle for efficient drug delivery. METHODS In this study, we employed RNA sequencing to characterize transcriptional alterations of endothelial cells isolated from primary and secondary human brain tumors. We used an immunoprecipitation approach to enrich for endothelial cells from normal brain, glioblastoma (GBM) and lung cancer brain metastasis (BM). RESULTS Analysis of the endothelial transcriptome showed deregulation of genes implicated in cell proliferation, angiogenesis and deposition of extracellular matrix (ECM) in the vasculature of GBM and BM. Deregulation of genes defining the BBB dysfunction module were found in both tumor types. We identified deregulated expression of genes in vessel-associated fibroblasts in GBM. CONCLUSION We characterize alterations in BBB genes in GBM and BM vasculature and identify proteins that might be exploited for developing drug delivery platforms. In addition, our analysis on vessel-associated fibroblasts in GBM shows that the cellular composition of brain tumor stroma merits further investigation.
Collapse
Affiliation(s)
- Johanna Schaffenrath
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zürich, Zürich University, Zürich, Switzerland.,Neuroscience Center Zürich, University of Zürich and ETH Zürich, Zürich, Switzerland
| | - Tania Wyss
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland.,Department of Oncology, University Lausanne, Lausanne, Switzerland
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Mauro Delorenzi
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland.,Department of Oncology, University Lausanne, Lausanne, Switzerland
| | - Flavio Vasella
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zürich, Zürich University, Zürich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zürich, Zürich University, Zürich, Switzerland
| | - Marian Christoph Neidert
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zürich, Zürich University, Zürich, Switzerland
| | - Annika Keller
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zürich, Zürich University, Zürich, Switzerland.,Neuroscience Center Zürich, University of Zürich and ETH Zürich, Zürich, Switzerland
| |
Collapse
|