1
|
Saha S, Zhang Y, Gilbert MK, Dube C, Hanif F, Mulcahy EQX, Bednarek S, Marcinkiewicz P, Wang X, Kwak G, Hudson K, Sun Y, Dinda M, Saha T, Guessous F, Cruickshanks N, Colon RR, Dell'Olio L, Anbu R, Kefas B, Kumar P, Klibanov AL, Schiff D, Suk JS, Hanes J, Mata J, Hafner M, Abounader R. Discovery and therapeutic exploitation of Master Regulatory miRNAs in Glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646663. [PMID: 40236125 PMCID: PMC11996502 DOI: 10.1101/2025.04.01.646663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Glioblastoma is a fatal primary malignant brain tumor. Despite therapies involving surgical resection, chemotherapy, and radiation therapy, the average survival for glioblastoma patients remains at approximately 15 months. MicroRNAs (miRNAs) are short noncoding RNA molecules that regulate the expression of the majority of human genes. Numerous genes are concurrently deregulated in glioblastoma. Consequently, molecular monotherapies have failed to achieve improvements in clinical outcomes. Several lines of evidence suggest that simultaneous targeting of several deregulated molecules is required to achieve better therapies. However, the simultaneous targeting of several deregulated oncogenic drivers is severely limited by the fact that the drugs needed to target many deregulated molecules do not currently exist, and because combining several drugs in a clinical setting leads to an exponential increase in toxicity. We hypothesized that we can develop and use miRNA to simultaneously inhibit multiple deregulated genes for more efficacious glioblastoma therapies. The goal of this study was therefore to identify master regulatory microRNAs (miRNAs) and use them to simultaneously target multiple deregulated molecules for GBM therapy. We defined master regulatory miRNAs as those that target several deregulated genes in glioblastoma. To find master regulatory miRNAs, we first used PAR-CLIP screenings to identify all targets of all miRNAs in glioblastoma cells. We then analyzed TCGA tumor data to determine which of these targets are deregulated in human tumors. We developed and used an algorithm to rank these targets for significance in glioblastoma malignancy based on their magnitude of deregulation, frequency of deregulation, and correlation with patient survival. We then ranked the miRNAs for their capacity of targeting multiple glioblastoma-deregulated genes and therefore the potential to exhibit strong anti-tumor effects when delivered as therapy. Using this strategy, we selected two tumor suppressor master regulatory miRNAs, miR-340, miR-382 and an oncogenic master regulatory miRNA, miR-17. We validated the target genes of the miRNAs and showed that they form part of important glioblastoma regulatory pathways. We then showed that the miRNAs (miR-340 and miR-582) or the miR-17 inhibitor have strong inhibitory effects on glioblastoma cell growth, survival, invasion, stemness and in vivo tumor growth. Ultimately, we developed and successfully tested a new therapeutic approach to delivery miR-340 using MRI guided focused ultrasound and microbubbles (FUS-MB) and special brain penetrating nanoparticles (BPN). This approach resulted in a substantial reduction in tumor volume and prolongation of the survival of glioblastoma-bearing mice and can be translated into clinical trials. We therefore developed and successfully tested a novel strategy to discover and deliver miRNAs for glioblastoma and cancer therapy. One Sentence Summary We developed and used new computational, experimental, and therapeutic approaches to identify and therapeutically deliver master regulatory miRNAs to inhibit the growth of glioblastoma, the most common and deadly primary brain tumor.
Collapse
|
2
|
Lefaucheur JP, Moro E, Shirota Y, Ugawa Y, Grippe T, Chen R, Benninger DH, Jabbari B, Attaripour S, Hallett M, Paulus W. Clinical neurophysiology in the treatment of movement disorders: IFCN handbook chapter. Clin Neurophysiol 2024; 164:57-99. [PMID: 38852434 PMCID: PMC11418354 DOI: 10.1016/j.clinph.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/02/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024]
Abstract
In this review, different aspects of the use of clinical neurophysiology techniques for the treatment of movement disorders are addressed. First of all, these techniques can be used to guide neuromodulation techniques or to perform therapeutic neuromodulation as such. Neuromodulation includes invasive techniques based on the surgical implantation of electrodes and a pulse generator, such as deep brain stimulation (DBS) or spinal cord stimulation (SCS) on the one hand, and non-invasive techniques aimed at modulating or even lesioning neural structures by transcranial application. Movement disorders are one of the main areas of indication for the various neuromodulation techniques. This review focuses on the following techniques: DBS, repetitive transcranial magnetic stimulation (rTMS), low-intensity transcranial electrical stimulation, including transcranial direct current stimulation (tDCS) and transcranial alternating current stimulation (tACS), and focused ultrasound (FUS), including high-intensity magnetic resonance-guided FUS (MRgFUS), and pulsed mode low-intensity transcranial FUS stimulation (TUS). The main clinical conditions in which neuromodulation has proven its efficacy are Parkinson's disease, dystonia, and essential tremor, mainly using DBS or MRgFUS. There is also some evidence for Tourette syndrome (DBS), Huntington's disease (DBS), cerebellar ataxia (tDCS), and axial signs (SCS) and depression (rTMS) in PD. The development of non-invasive transcranial neuromodulation techniques is limited by the short-term clinical impact of these techniques, especially rTMS, in the context of very chronic diseases. However, at-home use (tDCS) or current advances in the design of closed-loop stimulation (tACS) may open new perspectives for the application of these techniques in patients, favored by their easier use and lower rate of adverse effects compared to invasive or lesioning methods. Finally, this review summarizes the evidence for keeping the use of electromyography to optimize the identification of muscles to be treated with botulinum toxin injection, which is indicated and widely performed for the treatment of various movement disorders.
Collapse
Affiliation(s)
- Jean-Pascal Lefaucheur
- Clinical Neurophysiology Unit, Henri Mondor University Hospital, AP-HP, Créteil, France; EA 4391, ENT Team, Paris-Est Créteil University, Créteil, France.
| | - Elena Moro
- Grenoble Alpes University, Division of Neurology, CHU of Grenoble, Grenoble Institute of Neuroscience, Grenoble, France
| | - Yuichiro Shirota
- Department of Neurology, Division of Neuroscience, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshikazu Ugawa
- Department of Human Neurophysiology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Talyta Grippe
- Division of Neurology, University of Toronto, Toronto, Ontario, Canada; Neuroscience Graduate Program, Federal University of Minas Gerais, Belo Horizonte, Brazil; Krembil Brain Institute, Toronto, Ontario, Canada
| | - Robert Chen
- Division of Neurology, University of Toronto, Toronto, Ontario, Canada; Krembil Brain Institute, Toronto, Ontario, Canada
| | - David H Benninger
- Service of Neurology, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Bahman Jabbari
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Sanaz Attaripour
- Department of Neurology, University of California, Irvine, CA, USA
| | - Mark Hallett
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Walter Paulus
- Department of Neurology, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
3
|
Chen KS, Koubek EJ, Sakowski SA, Feldman EL. Stem cell therapeutics and gene therapy for neurologic disorders. Neurotherapeutics 2024; 21:e00427. [PMID: 39096590 PMCID: PMC11345629 DOI: 10.1016/j.neurot.2024.e00427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/05/2024] Open
Abstract
Rapid advances in biological knowledge and technological innovation have greatly advanced the fields of stem cell and gene therapies to combat a broad spectrum of neurologic disorders. Researchers are currently exploring a variety of stem cell types (e.g., embryonic, progenitor, induced pluripotent) and various transplantation strategies, each with its own advantages and drawbacks. Similarly, various gene modification techniques (zinc finger, TALENs, CRISPR-Cas9) are employed with various delivery vectors to modify underlying genetic contributors to neurologic disorders. While these two individual fields continue to blaze new trails, it is the combination of these technologies which enables genetically engineered stem cells and vastly increases investigational and therapeutic opportunities. The capability to culture and expand stem cells outside the body, along with their potential to correct genetic abnormalities in patient-derived cells or enhance cells with extra gene products, unleashes the full biological potential for innovative, multifaceted approaches to treat complex neurological disorders. In this review, we provide an overview of stem cell and gene therapies in the context of neurologic disorders, highlighting recent advances and current shortcomings, and discuss prospects for future therapies in clinical settings.
Collapse
Affiliation(s)
- Kevin S Chen
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA; Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily J Koubek
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stacey A Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
4
|
Farokhi E, Alaofi AL, Prasasty VD, Stephanie F, Laksitorini MD, Kuczera K, Siahaan TJ. Mechanism of the blood-brain barrier modulation by cadherin peptides. EXPLORATION OF DRUG SCIENCE 2024; 2:322-338. [PMID: 39118806 PMCID: PMC11309765 DOI: 10.37349/eds.2024.00049] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/21/2024] [Indexed: 08/10/2024]
Abstract
Aim This study was aimed at finding the binding site on the human E-cadherin for Ala-Asp-Thr Cyclic 5 (ADTC5), ADTC7, and ADTC9 peptides as blood-brain barrier modulator (BBBM) for determining their mechanism of action in modulating the blood-brain barrier (BBB). Methods ADTC7 and ADTC9 were derivatives of ADTC5 where the Val6 residue in ADTC5 was replaced by Glu6 and Tyr6 residues, respectively. The binding properties of ADTC5, ADTC7, and ADTC9 to the extracellular-1 (EC1) domain of E-cadherin were evaluated using chemical shift perturbation (CSP) method in the two dimensional (2D) 1H-15N-heteronuclear single quantum coherence (HSQC) nuclear magnetic resonance (NMR) spectroscopy. Molecular docking experiments were used to determine the binding sites of these peptides to the EC1 domain of E-cadherin. Results This study indicates that ADTC5 has the highest binding affinity to the EC1 domain of E-cadherin compared to ADTC7 and ADTC9, suggesting the importance of the Val6 residue as shown in our previous in vitro study. All three peptides have a similar binding site at the hydrophobic binding pocket where the domain swapping occurs. ADTC5 has a higher overlapping binding site with ADTC7 than that of ADTC9. Binding of ADTC5 on the EC1 domain influences the conformation of the EC1 C-terminal tail. Conclusions These peptides bind the domain swapping region of the EC1 domain to inhibit the trans-cadherin interaction that creates intercellular junction modulation to increase the BBB paracellular porosity.
Collapse
Affiliation(s)
- Elinaz Farokhi
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: Analytical Department, Johnson & Johnson, San Diego, CA 92123, USA
| | - Ahmed L. Alaofi
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Vivitri D. Prasasty
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Filia Stephanie
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
| | - Marlyn D. Laksitorini
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: School of Pharmacy, University of Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Krzysztof Kuczera
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66047, USA
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
| |
Collapse
|
5
|
Antoniou A, Stavrou M, Evripidou N, Georgiou E, Kousiappa I, Koupparis A, Papacostas SS, Kleopa KA, Damianou C. FUS-mediated blood-brain barrier disruption for delivering anti-Aβ antibodies in 5XFAD Alzheimer's disease mice. J Ultrasound 2024; 27:251-262. [PMID: 37516718 PMCID: PMC11178731 DOI: 10.1007/s40477-023-00805-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 06/28/2023] [Indexed: 07/31/2023] Open
Abstract
PURPOSE Amyloid-β (Aβ) peptides, the main component of amyloid plaques found in the Alzheimer's disease (AD) brain, are implicated in its pathogenesis, and are considered a key target in AD therapeutics. We herein propose a reliable strategy for non-invasively delivering a specific anti-Aβ antibody in a mouse model of AD by microbubbles-enhanced Focused Ultrasound (FUS)-mediated Blood-brain barrier disruption (BBBD), using a simple single stage MR-compatible positioning device. METHODS The initial experimental work involved wild-type mice and was devoted to selecting the sonication protocol for efficient and safe BBBD. Pulsed FUS was applied using a single-element FUS transducer of 1 MHz (80 mm radius of curvature and 50 mm diameter). The success and extent of BBBD were assessed by Evans Blue extravasation and brain damage by hematoxylin and eosin staining. 5XFAD mice were divided into different subgroups; control (n = 1), FUS + MBs alone (n = 5), antibody alone (n = 5), and FUS + antibody combined (n = 10). The changes in antibody deposition among groups were determined by immunohistochemistry. RESULTS It was confirmed that the antibody could not normally enter the brain parenchyma. A single treatment with MBs-enhanced pulsed FUS using the optimized protocol (1 MHz, 0.5 MPa in-situ pressure, 10 ms bursts, 1% duty factor, 100 s duration) transiently disrupted the BBB allowing for non-invasive antibody delivery to amyloid plaques within the sonicated brain regions. This was consistently reproduced in ten mice. CONCLUSION These preliminary findings should be confirmed by longer-term studies examining the antibody effects on plaque clearance and cognitive benefit to hold promise for developing disease-modifying anti-Aβ therapeutics for clinical use.
Collapse
Affiliation(s)
- Anastasia Antoniou
- Department of Electrical Engineering, Computer Engineering, and Informatics, Cyprus University of Technology, Limassol, Cyprus
| | - Marios Stavrou
- Department of Neurobiology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Nikolas Evripidou
- Department of Electrical Engineering, Computer Engineering, and Informatics, Cyprus University of Technology, Limassol, Cyprus
| | - Elena Georgiou
- Department of Neuroscience, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Ioanna Kousiappa
- Department of Neurobiology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Andreas Koupparis
- Department of Neurobiology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Savvas S Papacostas
- Department of Neurobiology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Kleopas A Kleopa
- Department of Neuroscience, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Christakis Damianou
- Department of Electrical Engineering, Computer Engineering, and Informatics, Cyprus University of Technology, Limassol, Cyprus.
| |
Collapse
|
6
|
Schwinghamer K, Line S, Tesar DB, Miller DW, Sreedhara A, Siahaan TJ. Selective Uptake of Macromolecules to the Brain in Microfluidics and Animal Models Using the HAVN1 Peptide as a Blood-Brain Barrier Modulator. Mol Pharm 2024; 21:1639-1652. [PMID: 38395041 PMCID: PMC10984760 DOI: 10.1021/acs.molpharmaceut.3c00775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Monoclonal antibodies (mAbs) possess favorable pharmacokinetic properties, high binding specificity and affinity, and minimal off-target effects, making them promising therapeutic agents for central nervous system (CNS) disorders. However, their development as effective therapeutic and diagnostic agents for brain disorders is hindered by their limited ability to efficiently penetrate the blood-brain barrier (BBB). Therefore, it is crucial to develop efficient delivery methods that enhance the penetration of antibodies into the brain. Previous studies have demonstrated the potential of cadherin-derived peptides (i.e., ADTC5, HAVN1 peptides) as BBB modulators (BBBMs) to increase paracellular porosities for penetration of molecules across the BBB. Here, we test the effectiveness of the leading BBBM peptide, HAVN1 (Cyclo(1,6)SHAVSS), in enhancing the permeation of various monoclonal antibodies through the BBB using both in vitro and in vivo systems. In vitro, HAVN1 has been shown to increase the permeability of fluorescently labeled macromolecules, such as a 70 kDa dextran, 50 kDa Fab1, and 150 kDa mAb1, by 4- to 9-fold in a three-dimensional blood-brain barrier (3D-BBB) microfluidics model using a human BBB endothelial cell line (i.e., hCMEC/D3). HAVN1 was selective in modulating the BBB endothelial cell, compared to the pulmonary vascular endothelial (PVE) cell barrier. Co-administration of HAVN1 significantly improved brain depositions of mAb1, mAb2, and Fab1 in C57BL/6 mice after 15 min in the systemic circulation. Furthermore, HAVN1 still significantly enhanced brain deposition of mAb2 when it was administered 24 h after the administration of the mAb. Lastly, we observed that multiple doses of HAVN1 may have a cumulative effect on the brain deposition of mAb2 within a 24-h period. These findings offer promising insights into optimizing HAVN1 and mAb dosing regimens to control or modulate mAb brain deposition for achieving desired mAb dose in the brain to provide its therapeutic effects.
Collapse
Affiliation(s)
- Kelly Schwinghamer
- Department of Pharmaceutical Chemistry, The University of Kansas, 2093 Constant Ave., Lawrence, KS 66047, USA
| | - Stacey Line
- Department of Pharmacology and Therapeutics, University of Manitoba, 753 McDermot Avenue Winnipeg, MB, R3E 0T6, Canada
| | - Devin B. Tesar
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Donald W. Miller
- Department of Pharmacology and Therapeutics, University of Manitoba, 753 McDermot Avenue Winnipeg, MB, R3E 0T6, Canada
| | - Alavattam Sreedhara
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, The University of Kansas, 2093 Constant Ave., Lawrence, KS 66047, USA
| |
Collapse
|
7
|
AIUM Official Statement for the Statement on Biological Effects of Therapeutic Ultrasound. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2023; 42:E68-E73. [PMID: 37584480 DOI: 10.1002/jum.16315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/17/2023]
|
8
|
Sharma A, Fernandes DC, Reis RL, Gołubczyk D, Neumann S, Lukomska B, Janowski M, Kortylewski M, Walczak P, Oliveira JM, Maciaczyk J. Cutting-edge advances in modeling the blood-brain barrier and tools for its reversible permeabilization for enhanced drug delivery into the brain. Cell Biosci 2023; 13:137. [PMID: 37501215 PMCID: PMC10373415 DOI: 10.1186/s13578-023-01079-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023] Open
Abstract
The blood-brain barrier (BBB) is a sophisticated structure whose full functionality is required for maintaining the executive functions of the central nervous system (CNS). Tight control of transport across the barrier means that most drugs, particularly large size, which includes powerful biologicals, cannot reach their targets in the brain. Notwithstanding the remarkable advances in characterizing the cellular nature of the BBB and consequences of BBB dysfunction in pathology (brain metastasis, neurological diseases), it remains challenging to deliver drugs to the CNS. Herein, we outline the basic architecture and key molecular constituents of the BBB. In addition, we review the current status of approaches that are being explored to temporarily open the BBB in order to allow accumulation of therapeutics in the CNS. Undoubtedly, the major concern in field is whether it is possible to open the BBB in a meaningful way without causing negative consequences. In this context, we have also listed few other important key considerations that can improve our understanding about the dynamics of the BBB.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Stereotacitc and Functional Neurosurgery, University Hospital Bonn, 53127, Bonn, Germany
| | - Diogo C Fernandes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga, Portugal
| | - Dominika Gołubczyk
- Ti-Com, Polish Limited Liability Company, 10-683, Olsztyn, Poland
- Center for Translational Medicine, Warsaw University of Life Sciences, 02-797, Warsaw, Poland
| | - Silke Neumann
- Department of Pathology, University of Otago, Dunedin, 9054, New Zealand
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcin Kortylewski
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J Miguel Oliveira
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga, Portugal.
| | - Jarek Maciaczyk
- Department of Stereotacitc and Functional Neurosurgery, University Hospital Bonn, 53127, Bonn, Germany.
- Department of Surgical Sciences, University of Otago, Dunedin, 9054, New Zealand.
| |
Collapse
|
9
|
Bao Y, Lu W. Targeting cerebral diseases with enhanced delivery of therapeutic proteins across the blood-brain barrier. Expert Opin Drug Deliv 2023; 20:1681-1698. [PMID: 36945117 DOI: 10.1080/17425247.2023.2193390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Cerebral diseases have been threatening public physical and psychological health in the recent years. With the existence of the blood-brain barrier (BBB), it is particularly hard for therapeutic proteins like peptides, enzymes, antibodies, etc. to enter the central nervous system (CNS) and function in diagnosis and treatment in cerebral diseases. Fortunately, the past decade has witnessed some emerging strategies of delivering macromolecular therapeutic proteins across the BBB. AREAS COVERED Based on the structure, functions, and substances transport mechanisms, various enhanced delivery strategies of therapeutic proteins were reviewed, categorized by molecule-mediated delivery strategies, carrier-mediated delivery strategies, and other delivery strategies. EXPERT OPINION As for molecule-mediated delivery strategies, development of genetic engineering technology, optimization of protein expression and purification techniques, and mature of quality control systems all help to realize large-scale production of recombinant antibodies, making it possible to apply to the clinical practice. In terms of carrier-mediated delivery strategies and others, although nano-carriers/adeno-associated virus (AAV) are also promising candidates for delivering therapeutic proteins or genes across the BBB, some issues still remain to be further investigated, including safety concerns related to applied materials, large-scale production costs, quality control standards, combination therapies with auxiliary delivery strategies like focused ultrasound, etc.
Collapse
Affiliation(s)
- Yanning Bao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, China
- Department of Research and Development, Shanghai Tayzen PharmLab Co., Ltd. Lingang of Shanghai, China
| |
Collapse
|
10
|
Park EY, Cai X, Foiret J, Bendjador H, Hyun D, Fite BZ, Wodnicki R, Dahl JJ, Boutin RD, Ferrara KW. Fast volumetric ultrasound facilitates high-resolution 3D mapping of tissue compartments. SCIENCE ADVANCES 2023; 9:eadg8176. [PMID: 37256942 PMCID: PMC10413648 DOI: 10.1126/sciadv.adg8176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/28/2023] [Indexed: 06/02/2023]
Abstract
Volumetric ultrasound imaging has the potential for operator-independent acquisition and enhanced field of view. Panoramic acquisition has many applications across ultrasound; spanning musculoskeletal, liver, breast, and pediatric imaging; and image-guided therapy. Challenges in high-resolution human imaging, such as subtle motion and the presence of bone or gas, have limited such acquisition. These issues can be addressed with a large transducer aperture and fast acquisition and processing. Programmable, ultrafast ultrasound scanners with a high channel count provide an unprecedented opportunity to optimize volumetric acquisition. In this work, we implement nonlinear processing and develop distributed beamformation to achieve fast acquisition over a 47-centimeter aperture. As a result, we achieve a 50-micrometer -6-decibel point spread function at 5 megahertz and resolve in-plane targets. A large volume scan of a human limb is completed in a few seconds, and in a 2-millimeter dorsal vein, the image intensity difference between the vessel center and surrounding tissue was ~50 decibels, facilitating three-dimensional reconstruction of the vasculature.
Collapse
Affiliation(s)
- Eun-Yeong Park
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Xiran Cai
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Josquin Foiret
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Hanna Bendjador
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Dongwoon Hyun
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Brett Z. Fite
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Robert Wodnicki
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Jeremy J. Dahl
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Robert D. Boutin
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
11
|
Petrovskaya A, Tverskoi A, Medvedeva A, Nazarova M. Is blood-brain barrier a probable mediator of non-invasive brain stimulation effects on Alzheimer's disease? Commun Biol 2023; 6:416. [PMID: 37059824 PMCID: PMC10104838 DOI: 10.1038/s42003-023-04717-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/16/2023] [Indexed: 04/16/2023] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease with no existing treatment leading to full recovery. The blood-brain barrier (BBB) breakdown usually precedes the advent of first symptoms in AD and accompanies the progression of the disease. At the same time deliberate BBB opening may be beneficial for drug delivery in AD. Non-invasive brain stimulation (NIBS) techniques, primarily transcranial magnetic stimulation (TMS) and transcranial direct current stimulation (tDCS), have shown multiple evidence of being able to alleviate symptoms of AD. Currently, TMS/tDCS mechanisms are mostly investigated in terms of their neuronal effects, while their possible non-neuronal effects, including mitigation of the BBB disruption, are less studied. We argue that studies of TMS/tDCS effects on the BBB in AD are necessary to boost the effectiveness of neuromodulation in AD. Moreover, such studies are important considering the safety issues of TMS/tDCS use in the advanced AD stages when the BBB is usually dramatically deteriorated. Here, we elucidate the evidence of NIBS-induced BBB opening and closing in various models from in vitro to humans, and highlight its importance in AD.
Collapse
Affiliation(s)
- Aleksandra Petrovskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Artem Tverskoi
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Angela Medvedeva
- Department of Chemistry, Rice University, Houston, TX, 77005, US
| | - Maria Nazarova
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
- Center for Cognition and Decision Making, Institute for Cognitive Neuroscience, National Research University Higher School of Economics, Moscow, 101000, Russian Federation
| |
Collapse
|
12
|
Critchley BJ, Gaspar HB, Benedetti S. Targeting the central nervous system in lysosomal storage diseases: Strategies to deliver therapeutics across the blood-brain barrier. Mol Ther 2023; 31:657-675. [PMID: 36457248 PMCID: PMC10014236 DOI: 10.1016/j.ymthe.2022.11.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/18/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are multisystem inherited metabolic disorders caused by dysfunctional lysosomal activity, resulting in the accumulation of undegraded macromolecules in a variety of organs/tissues, including the central nervous system (CNS). Treatments include enzyme replacement therapy, stem/progenitor cell transplantation, and in vivo gene therapy. However, these treatments are not fully effective in treating the CNS as neither enzymes, stem cells, nor viral vectors efficiently cross the blood-brain barrier. Here, we review the latest advancements in improving delivery of different therapeutic agents to the CNS and comment upon outstanding questions in the field of neurological LSDs.
Collapse
Affiliation(s)
- Bethan J Critchley
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK
| | - H Bobby Gaspar
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK; Orchard Therapeutics Ltd., London EC4N 6EU, UK
| | - Sara Benedetti
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
13
|
Sánchez-Dengra B, González-Álvarez I, Bermejo M, González-Álvarez M. Access to the CNS: Strategies to overcome the BBB. Int J Pharm 2023; 636:122759. [PMID: 36801479 DOI: 10.1016/j.ijpharm.2023.122759] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/31/2023] [Accepted: 02/17/2023] [Indexed: 02/21/2023]
Abstract
The blood-brain barrier (BBB) limits the access of substances to the central nervous system (CNS) which hinders the treatment of pathologies affecting the brain and the spinal cord. Nowadays, research is focus on new strategies to overcome the BBB and can treat the pathologies affecting the CNS are needed. In this review, the different strategies that allow and increase the access of substances to the CNS are analysed and extended commented, not only invasive strategies but also non-invasive ones. The invasive techniques include the direct injection into the brain parenchyma or the CSF and the therapeutic opening of the BBB, while the non-invasive techniques include the use of alternative routes of administration (nose-to-brain route), the inhibition of efflux transporters (as it is important to prevent the drug efflux from the brain and enhance the therapeutic efficiency), the chemical modification of the molecules (prodrugs and chemical drug delivery systems (CDDS)) and the use of nanocarriers. In the future, knowledge about nanocarriers to treat CNS diseases will continue to increase, but the use of other strategies such as drug repurposing or drug reprofiling, which are cheaper and less time consuming, may limit its transfer to society. The main conclusion is that the combination of different strategies may be the most interesting approach to increase the access of substances to the CNS.
Collapse
Affiliation(s)
- Bárbara Sánchez-Dengra
- Pharmacokinetics and Pharmaceutical Technology Area, Department of Engineering, Miguel Hernandez University, 03550 Alicante, Spain
| | - Isabel González-Álvarez
- Pharmacokinetics and Pharmaceutical Technology Area, Department of Engineering, Miguel Hernandez University, 03550 Alicante, Spain.
| | - Marival Bermejo
- Pharmacokinetics and Pharmaceutical Technology Area, Department of Engineering, Miguel Hernandez University, 03550 Alicante, Spain
| | - Marta González-Álvarez
- Pharmacokinetics and Pharmaceutical Technology Area, Department of Engineering, Miguel Hernandez University, 03550 Alicante, Spain
| |
Collapse
|
14
|
Neurotrophic Factors as Regenerative Therapy for Neurodegenerative Diseases: Current Status, Challenges and Future Perspectives. Int J Mol Sci 2023; 24:ijms24043866. [PMID: 36835277 PMCID: PMC9968045 DOI: 10.3390/ijms24043866] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), spinal cord injury (SCI), and amyotrophic lateral sclerosis (ALS), are characterized by acute or chronic progressive loss of one or several neuronal subtypes. However, despite their increasing prevalence, little progress has been made in successfully treating these diseases. Research has recently focused on neurotrophic factors (NTFs) as potential regenerative therapy for neurodegenerative diseases. Here, we discuss the current state of knowledge, challenges, and future perspectives of NTFs with a direct regenerative effect in chronic inflammatory and degenerative disorders. Various systems for delivery of NTFs, such as stem and immune cells, viral vectors, and biomaterials, have been applied to deliver exogenous NTFs to the central nervous system, with promising results. The challenges that currently need to be overcome include the amount of NTFs delivered, the invasiveness of the delivery route, the blood-brain barrier permeability, and the occurrence of side effects. Nevertheless, it is important to continue research and develop standards for clinical applications. In addition to the use of single NTFs, the complexity of chronic inflammatory and degenerative diseases may require combination therapies targeting multiple pathways or other possibilities using smaller molecules, such as NTF mimetics, for effective treatment.
Collapse
|
15
|
Lenka A, Jankovic J. How should future clinical trials be designed in the search for disease-modifying therapies for Parkinson's disease? Expert Rev Neurother 2023; 23:107-122. [PMID: 36803618 DOI: 10.1080/14737175.2023.2177535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
INTRODUCTION Although there has been substantial progress in research and innovations in symptomatic treatments, similar success has not been achieved in disease-modifying therapy (DMT) for Parkinson's disease (PD). Considering the enormous motor, psychosocial and financial burden associated with PD, safe and effective DMT is of paramount importance. AREAS COVERED One of the reasons for the lack of progress in DMT for PD is poor or inappropriate design of clinical trials. In the first part of the article, the authors focus on the plausible reasons why the previous trials have failed and in the latter part, they provide their perspectives on future DMT trials. EXPERT OPINION There are several potential reasons why previous trials have failed, including broad clinical and etiopathogenic heterogeneity of PD, poor definition and documentation of target engagement, lack of appropriate biomarkers and outcome measures, and short duration of follow-up. To address these deficiencies, future trials may consider- (i) a more customized approach to select the most suitable participants and therapeutic approaches, (ii) explore combination therapies that would target multiple pathogenetic mechanisms, and (iii) moving beyond targeting only motor symptoms to also assessing non-motor features of PD in well-designed longitudinal studies.
Collapse
Affiliation(s)
- Abhishek Lenka
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
16
|
Stefani A, Pierantozzi M, Cardarelli S, Stefani L, Cerroni R, Conti M, Garasto E, Mercuri NB, Marini C, Sucapane P. Neurotrophins as Therapeutic Agents for Parkinson’s Disease; New Chances From Focused Ultrasound? Front Neurosci 2022; 16:846681. [PMID: 35401084 PMCID: PMC8990810 DOI: 10.3389/fnins.2022.846681] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/31/2022] [Indexed: 01/02/2023] Open
Abstract
Magnetic Resonance–guided Focused Ultrasound (MRgFUS) represents an effective micro-lesioning approach to target pharmaco-resistant tremor, mostly in patients afflicted by essential tremor (ET) and/or Parkinson’s disease (PD). So far, experimental protocols are verifying the clinical extension to other facets of the movement disorder galaxy (i.e., internal pallidus for disabling dyskinesias). Aside from those neurosurgical options, one of the most intriguing opportunities of this technique relies on its capability to remedy the impermeability of blood–brain barrier (BBB). Temporary BBB opening through low-intensity focused ultrasound turned out to be safe and feasible in patients with PD, Alzheimer’s disease, and amyotrophic lateral sclerosis. As a mere consequence of the procedures, some groups described even reversible but significant mild cognitive amelioration, up to hippocampal neurogenesis partially associated to the increased of endogenous brain-derived neurotrophic factor (BDNF). A further development elevates MRgFUS to the status of therapeutic tool for drug delivery of putative neurorestorative therapies. Since 2012, FUS-assisted intravenous administration of BDNF or neurturin allowed hippocampal or striatal delivery. Experimental studies emphasized synergistic modalities. In a rodent model for Huntington’s disease, engineered liposomes can carry glial cell line–derived neurotrophic factor (GDNF) plasmid DNA (GDNFp) to form a GDNFp-liposome (GDNFp-LPs) complex through pulsed FUS exposures with microbubbles; in a subacute MPTP-PD model, the combination of intravenous administration of neurotrophic factors (either through protein or gene delivery) plus FUS did curb nigrostriatal degeneration. Here, we explore these arguments, focusing on the current, translational application of neurotrophins in neurodegenerative diseases.
Collapse
Affiliation(s)
- Alessandro Stefani
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
- *Correspondence: Alessandro Stefani,
| | | | - Silvia Cardarelli
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Lucrezia Stefani
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Rocco Cerroni
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Matteo Conti
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Elena Garasto
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Nicola B. Mercuri
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Carmine Marini
- UOC Neurology and Stroke Unit, University of L’Aquila, L’Aquila, Italy
| | | |
Collapse
|
17
|
Poustforoosh A, Nematollahi MH, Hashemipour H, Pardakhty A. Recent advances in Bio-conjugated nanocarriers for crossing the Blood-Brain Barrier in (pre-)clinical studies with an emphasis on vesicles. J Control Release 2022; 343:777-797. [DOI: 10.1016/j.jconrel.2022.02.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 12/12/2022]
|
18
|
Thombre R, Mess G, Kempski Leadingham KM, Kapoor S, Hersh A, Acord M, Kaovasia T, Theodore N, Tyler B, Manbachi A. Towards standardization of the parameters for opening the blood-brain barrier with focused ultrasound to treat glioblastoma multiforme: A systematic review of the devices, animal models, and therapeutic compounds used in rodent tumor models. Front Oncol 2022; 12:1072780. [PMID: 36873300 PMCID: PMC9978816 DOI: 10.3389/fonc.2022.1072780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/20/2022] [Indexed: 02/18/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a deadly and aggressive malignant brain cancer that is highly resistant to treatments. A particular challenge of treatment is caused by the blood-brain barrier (BBB), the relatively impermeable vasculature of the brain. The BBB prevents large molecules from entering the brain parenchyma. This protective characteristic of the BBB, however, also limits the delivery of therapeutic drugs for the treatment of brain tumors. To address this limitation, focused ultrasound (FUS) has been safely utilized to create transient openings in the BBB, allowing various high molecular weight drugs access to the brain. We performed a systematic review summarizing current research on treatment of GBMs using FUS-mediated BBB openings in in vivo mouse and rat models. The studies gathered here highlight how the treatment paradigm can allow for increased brain and tumor perfusion of drugs including chemotherapeutics, immunotherapeutics, gene therapeutics, nanoparticles, and more. Given the promising results detailed here, the aim of this review is to detail the commonly used parameters for FUS to open the BBB in rodent GBM models.
Collapse
Affiliation(s)
- Rasika Thombre
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Griffin Mess
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Kelley M Kempski Leadingham
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Shivani Kapoor
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Andrew Hersh
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Molly Acord
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Tarana Kaovasia
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Nicholas Theodore
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amir Manbachi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States.,Department of Electrical Engineering and Computer Science, Johns Hopkins University, Baltimore, MD, United States.,Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, United States.,Department of Anesthesiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|