1
|
Dhiman A, Rana D, Benival D, Garkhal K. Comprehensive insights into glioblastoma multiforme: drug delivery challenges and multimodal treatment strategies. Ther Deliv 2024:1-29. [PMID: 39445563 DOI: 10.1080/20415990.2024.2415281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and malignant brain tumors, with a high prevalence in elderly population. Most chemotherapeutic agents fail to reach the tumor site due to various challenges. However, smart nanocarriers have demonstrated excellent drug-loading capabilities, enabling them to cross the blood brain tumor barrier for the GBM treatment. Surface modification of nanocarriers has significantly enhanced their potential for targeting therapeutics. Moreover, recent innovations in drug therapies, such as the incorporation of theranostic agents in nanocarriers and antibody-drug conjugates, have offered newer insights for both diagnosis and treatment. This review focuses on recent advances in new therapeutic interventions for GBM, with an emphasis on the nanotheranostics systems to maximize therapeutic and diagnostic outcomes.
Collapse
Affiliation(s)
- Ashish Dhiman
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Dhwani Rana
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Derajram Benival
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Kalpna Garkhal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| |
Collapse
|
2
|
Bae WH, Maraka S, Daher A. Challenges and advances in glioblastoma targeted therapy: the promise of drug repurposing and biomarker exploration. Front Oncol 2024; 14:1441460. [PMID: 39439947 PMCID: PMC11493774 DOI: 10.3389/fonc.2024.1441460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Glioblastoma remains the most prevalent and aggressive primary malignant brain tumor in adults, characterized by limited treatment options and a poor prognosis. Previous drug repurposing efforts have yielded only marginal survival benefits, particularly those involving inhibitors targeting receptor tyrosine kinase and cyclin-dependent kinase-retinoblastoma pathways. This limited efficacy is likely due to several critical challenges, including the tumor's molecular heterogeneity, the dynamic evolution of its genetic profile, and the restrictive nature of the blood-brain barrier that impedes effective drug delivery. Emerging diagnostic tools, such as circulating tumor DNA and extracellular vesicles, offer promising non-invasive methods for real-time tumor monitoring, potentially enabling the application of targeted therapies to more selected patient populations. Moreover, innovative drug delivery strategies, including focused ultrasound, implantable drug-delivery systems, and engineered nanoparticles, hold potential for enhancing the bioavailability and therapeutic efficacy of treatments.
Collapse
Affiliation(s)
- William Han Bae
- Division of Hematology/Oncology, Department of Internal Medicine, University of Illinois Chicago, Chicago, IL, United States
| | - Stefania Maraka
- Department of Neurology and Rehabilitation, University of Illinois Chicago, Chicago, IL, United States
| | - Ahmad Daher
- Department of Neurology and Rehabilitation, University of Illinois Chicago, Chicago, IL, United States
| |
Collapse
|
3
|
Andrew Awuah W, Shah MH, Tan JK, Ranganathan S, Sanker V, Darko K, Tenkorang PO, Adageba BB, Ahluwalia A, Shet V, Aderinto N, Kundu M, Abdul‐Rahman T, Atallah O. Immunotherapeutic advances in glioma management: The rise of vaccine-based approaches. CNS Neurosci Ther 2024; 30:e70013. [PMID: 39215399 PMCID: PMC11364516 DOI: 10.1111/cns.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/23/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Gliomas, particularly glioblastoma multiforme (GBM), are highly aggressive brain tumors that present significant challenges in oncology due to their rapid progression and resistance to conventional therapies. Despite advancements in treatment, the prognosis for patients with GBM remains poor, necessitating the exploration of novel therapeutic approaches. One such emerging strategy is the development of glioma vaccines, which aim to stimulate the immune system to target and destroy tumor cells. AIMS This review aims to provide a comprehensive evaluation of the current landscape of glioma vaccine development, analyzing the types of vaccines under investigation, the outcomes of clinical trials, and the challenges and opportunities associated with their implementation. The goal is to highlight the potential of glioma vaccines in advancing more effective and personalized treatments for glioma patients. MATERIALS AND METHODS This narrative review systematically assessed the role of glioma vaccines by including full-text articles published between 2000 and 2024 in English. Databases such as PubMed/MEDLINE, EMBASE, the Cochrane Library, and Scopus were searched using key terms like "glioma," "brain tumor," "glioblastoma," "vaccine," and "immunotherapy." The review incorporated both pre-clinical and clinical studies, including descriptive studies, animal-model studies, cohort studies, and observational studies. Exclusion criteria were applied to omit abstracts, case reports, posters, and non-peer-reviewed studies, ensuring the inclusion of high-quality evidence. RESULTS Clinical trials investigating various glioma vaccines, including peptide-based, DNA/RNA-based, whole-cell, and dendritic-cell vaccines, have shown promising results. These vaccines demonstrated potential in extending survival rates and managing adverse events in glioma patients. However, significant challenges remain, such as therapeutic resistance due to tumor heterogeneity and immune evasion mechanisms. Moreover, the lack of standardized guidelines for evaluating vaccine responses and issues related to ethical considerations, regulatory hurdles, and vaccine acceptance among patients further complicate the implementation of glioma vaccines. DISCUSSION Addressing the challenges associated with glioma vaccines involves exploring combination therapies, targeted approaches, and personalized medicine. Combining vaccines with traditional therapies like radiotherapy or chemotherapy may enhance efficacy by boosting the immune system's ability to fight tumor cells. Personalized vaccines tailored to individual patient profiles present an opportunity for improved outcomes. Furthermore, global collaboration and equitable distribution are critical for ensuring access to glioma vaccines, especially in low- and middle-income countries with limited healthcare resources CONCLUSION: Glioma vaccines represent a promising avenue in the fight against gliomas, offering hope for improving patient outcomes in a disease that is notoriously difficult to treat. Despite the challenges, continued research and the development of innovative strategies, including combination therapies and personalized approaches, are essential for overcoming current barriers and transforming the treatment landscape for glioma patients.
Collapse
Affiliation(s)
| | | | | | | | - Vivek Sanker
- Department of NeurosurgeryTrivandrum Medical CollegeTrivandrumKeralaIndia
| | - Kwadwo Darko
- Department of NeurosurgeryKorle Bu Teaching HospitalAccraGhana
| | | | - Bryan Badayelba Adageba
- Kwame Nkrumah University of Science and Technology School of Medicine and DentistryKumasiGhana
| | | | - Vallabh Shet
- Faculty of MedicineBangalore Medical College and Research InstituteBangaloreKarnatakaIndia
| | - Nicholas Aderinto
- Department of Internal MedicineLAUTECH Teaching HospitalOgbomosoNigeria
| | - Mrinmoy Kundu
- Institute of Medical Sciences and SUM HospitalBhubaneswarOdishaIndia
| | | | - Oday Atallah
- Department of Neurosurgery, Hannover Medical SchoolHannoverGermany
| |
Collapse
|
4
|
Nagayach A, Bhaskar R, Ghosh S, Singh KK, Han SS, Sinha JK. Advancing the understanding of diabetic encephalopathy through unravelling pathogenesis and exploring future treatment perspectives. Ageing Res Rev 2024; 100:102450. [PMID: 39134179 DOI: 10.1016/j.arr.2024.102450] [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: 06/03/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/25/2024]
Abstract
Diabetic encephalopathy (DE), a significant micro-complication of diabetes, manifests as neurochemical, structural, behavioral, and cognitive alterations. This condition is especially dangerous for the elderly because aging raises the risk of neurodegenerative disorders and cognitive impairment, both of which can be made worse by diabetes. Despite its severity, diagnosis of this disease is challenging, and there is a paucity of information on its pathogenesis. The pivotal roles of various cellular pathways, activated or influenced by hyperglycemia, insulin sensitivity, amyloid accumulation, tau hyperphosphorylation, brain vasculopathy, neuroinflammation, and oxidative stress, are widely recognized for contributing to the potential causes of diabetic encephalopathy. We also reviewed current pharmacological strategies for DE encompassing a comprehensive approach targeting metabolic dysregulations and neurological manifestations. Antioxidant-based therapies hold promise in mitigating oxidative stress-induced neuronal damage, while anti-diabetic drugs offer neuroprotective effects through diverse mechanisms, including modulation of insulin signaling pathways and neuroinflammation. Additionally, tissue engineering and nanomedicine-based approaches present innovative strategies for targeted drug delivery and regenerative therapies for DE. Despite significant progress, challenges remain in translating these therapeutic interventions into clinical practice, including long-term safety, scalability, and regulatory approval. Further research is warranted to optimize these approaches and address remaining gaps in the management of DE and associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Aarti Nagayach
- Department of Cancer Biology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301 India
| | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology, Symbiosis International (Deemed University), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune, Maharashtra 411057, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea.
| | | |
Collapse
|
5
|
Ding S, Kim YJ, Huang KY, Um D, Jung Y, Kong H. Delivery-mediated exosomal therapeutics in ischemia-reperfusion injury: advances, mechanisms, and future directions. NANO CONVERGENCE 2024; 11:18. [PMID: 38689075 PMCID: PMC11061094 DOI: 10.1186/s40580-024-00423-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024]
Abstract
Ischemia-reperfusion injury (IRI) poses significant challenges across various organ systems, including the heart, brain, and kidneys. Exosomes have shown great potentials and applications in mitigating IRI-induced cell and tissue damage through modulating inflammatory responses, enhancing angiogenesis, and promoting tissue repair. Despite these advances, a more systematic understanding of exosomes from different sources and their biotransport is critical for optimizing therapeutic efficacy and accelerating the clinical adoption of exosomes for IRI therapies. Therefore, this review article overviews the administration routes of exosomes from different sources, such as mesenchymal stem cells and other somatic cells, in the context of IRI treatment. Furthermore, this article covers how the delivered exosomes modulate molecular pathways of recipient cells, aiding in the prevention of cell death and the promotions of regeneration in IRI models. In the end, this article discusses the ongoing research efforts and propose future research directions of exosome-based therapies.
Collapse
Affiliation(s)
- Shengzhe Ding
- Chemical & Biomolecular Engineering, University of Illinois, Urbana, IL, 61801, USA
| | - Yu-Jin Kim
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Kai-Yu Huang
- Chemical & Biomolecular Engineering, University of Illinois, Urbana, IL, 61801, USA
| | - Daniel Um
- Bioengineering, University of Illinois, Urbana, IL, 61801, USA
| | - Youngmee Jung
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Department of Electrical and Electronic Engineering, YU-KIST Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyunjoon Kong
- Chemical & Biomolecular Engineering, University of Illinois, Urbana, IL, 61801, USA.
- Bioengineering, University of Illinois, Urbana, IL, 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, IL, 61801, USA.
- Chan Zuckerberg Biohub-Chicago, Chicago, USA.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
6
|
Sharma S, Wang SA, Yang WB, Lin HY, Lai MJ, Chen HC, Kao TY, Hsu FL, Nepali K, Hsu TI, Liou JP. First-in-Class Dual EZH2-HSP90 Inhibitor Eliciting Striking Antiglioblastoma Activity In Vitro and In Vivo. J Med Chem 2024; 67:2963-2985. [PMID: 38285511 PMCID: PMC10895674 DOI: 10.1021/acs.jmedchem.3c02053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 01/31/2024]
Abstract
Structural analysis of tazemetostat, an FDA-approved EZH2 inhibitor, led us to pinpoint a suitable site for appendage with a pharmacophoric fragment of second-generation HSP90 inhibitors. Resultantly, a magnificent dual EZH2/HSP90 inhibitor was pinpointed that exerted striking cell growth inhibitory efficacy against TMZ-resistant Glioblastoma (GBM) cell lines. Exhaustive explorations of chemical probe 7 led to several revelations such as (i) compound 7 increased apoptosis/necrosis-related gene expression, whereas decreased M phase/kinetochore/spindle-related gene expression as well as CENPs protein expression in Pt3R cells; (ii) dual inhibitor 7 induced cell cycle arrest at the M phase; (iii) compound 7 suppressed reactive oxygen species (ROS) catabolism pathway, causing the death of TMZ-resistant GBM cells; and (iv) compound 7 elicited substantial in vivo anti-GBM efficacy in experimental mice xenografted with TMZ-resistant Pt3R cells. Collectively, the study results confirm the potential of dual EZH2-HSP90 inhibitor 7 as a tractable anti-GBM agent.
Collapse
Affiliation(s)
- Sachin Sharma
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
| | - Shao-An Wang
- School
of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Bin Yang
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
| | - Hong-Yi Lin
- Graduate
Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Mei-Jung Lai
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
| | - Hsien-Chung Chen
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
- Department
of Neurosurgery, Shuang Ho Hospital, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
| | - Tzu-Yuan Kao
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
| | - Feng-Lin Hsu
- School
of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Kunal Nepali
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Tsung-I Hsu
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
- International
Master Program in Medical Neuroscience, College of Medical Science
and Technology, Taipei Medical University, Taipei 110, Taiwan
- TMU
Research Center of Cancer Translational Medicine, Taipei 110 Taiwan
| | - Jing-Ping Liou
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center of Cancer Translational Medicine, Taipei 110 Taiwan
- Ph.D.
Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
7
|
Behrends A, Wei H, Neumann A, Friedrich T, Bakenecker AC, Franke J, Sajjamark K, Buchholz O, Bär S, Hofmann UG, Graeser M, Buzug TM. Integrable Magnetic Fluid Hyperthermia Systems for 3D Magnetic Particle Imaging. Nanotheranostics 2024; 8:163-178. [PMID: 38444740 PMCID: PMC10911971 DOI: 10.7150/ntno.90360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/20/2024] [Indexed: 03/07/2024] Open
Abstract
Background: Combining magnetic particle imaging (MPI) and magnetic fluid hyperthermia (MFH) offers the ability to perform localized hyperthermia and magnetic particle imaging-assisted thermometry of hyperthermia treatment. This allows precise regional selective heating inside the body without invasive interventions. In current MPI-MFH platforms, separate systems are used, which require object transfer from one system to another. Here, we present the design, development and evaluation process for integrable MFH platforms, which extends a commercial MPI scanner with the functionality of MFH. Methods: The biggest issue of integrating magnetic fluid hyperthermia platforms into a magnetic particle imaging system is the magnetic coupling of the devices, which induces high voltage in the imaging system, and is harming its components. In this paper, we use a self-compensation approach derived from heuristic algorithms to protect the magnetic particle imaging scanner. The integrable platforms are evaluated regarding electrical and magnetic characteristics, cooling capability, field strength, the magnetic coupling to a replica of the magnetic particle imaging system's main solenoid and particle heating. Results: The MFH platforms generate suitable magnetic fields for the magnetic heating of particles and are compatible with a commercial magnetic particle imaging scanner. In combination with the imaging system, selective heating with a gradient field and steerable heating positioning using the MPI focus fields are possible. Conclusion: The proposed MFH platforms serve as a therapeutic tool to unlock the MFH functionality of a commercial magnetic particle imaging scanner, enabling its use in future preclinical trials of MPI-guided, spatially selective magnetic hyperthermia therapy.
Collapse
Affiliation(s)
- André Behrends
- Fraunhofer Research Institution for Individualized and Cell-Based Medical Engineering (IMTE), Lübeck, Germany
| | - Huimin Wei
- Fraunhofer Research Institution for Individualized and Cell-Based Medical Engineering (IMTE), Lübeck, Germany
| | - Alexander Neumann
- Institute of Medical Engineering (IMT), University of Lübeck, Lübeck, Germany
| | - Thomas Friedrich
- Fraunhofer Research Institution for Individualized and Cell-Based Medical Engineering (IMTE), Lübeck, Germany
| | - Anna C. Bakenecker
- Fraunhofer Research Institution for Individualized and Cell-Based Medical Engineering (IMTE), Lübeck, Germany
| | - Jochen Franke
- Bruker BioSpin MRI GmbH, Preclinical Imaging Division, Ettlingen, Germany
| | - Kulthisa Sajjamark
- Bruker BioSpin MRI GmbH, Preclinical Imaging Division, Ettlingen, Germany
| | - Oliver Buchholz
- Section for Neuroelectronic Systems, Department of Neurosurgery, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sébastien Bär
- Section for Neuroelectronic Systems, Department of Neurosurgery, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ulrich G. Hofmann
- Section for Neuroelectronic Systems, Department of Neurosurgery, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matthias Graeser
- Fraunhofer Research Institution for Individualized and Cell-Based Medical Engineering (IMTE), Lübeck, Germany
- Institute of Medical Engineering (IMT), University of Lübeck, Lübeck, Germany
| | - Thorsten M. Buzug
- Fraunhofer Research Institution for Individualized and Cell-Based Medical Engineering (IMTE), Lübeck, Germany
- Institute of Medical Engineering (IMT), University of Lübeck, Lübeck, Germany
| |
Collapse
|
8
|
Khan IN, Navaid S, Waqar W, Hussein D, Ullah N, Khan MUA, Hussain Z, Javed A. Chitosan-Based Polymeric Nanoparticles as an Efficient Gene Delivery System to Cross Blood Brain Barrier: In Vitro and In Vivo Evaluations. Pharmaceuticals (Basel) 2024; 17:169. [PMID: 38399386 PMCID: PMC10893193 DOI: 10.3390/ph17020169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 02/25/2024] Open
Abstract
Significant progress has been made in the field of gene therapy, but effective treatments for brain tumors remain challenging due to their complex nature. Current treatment options have limitations, especially due to their inability to cross the blood-brain barrier (BBB) and precisely target cancer cells. Therefore options that are safer, more effective, and capable of specifically targeting cancer cells are urgently required as alternatives. This current study aimed to develop highly biocompatible natural biopolymeric chitosan nanoparticles (CNPs) as potential gene delivery vehicles that can cross the BBB and serve as gene or drug delivery vehicles for brain disease therapeutics. The efficiency of the CNPs was evaluated via in vitro transfection of Green Fluorescent Protein (GFP)-tagged plasmid in HEK293-293 and brain cancer MG-U87 cell lines, as well as within in vivo mouse models. The CNPs were prepared via a complex coacervation method, resulting in nanoparticles of approximately 260 nm in size. In vitro cytotoxicity analysis revealed that the CNPs had better cell viability (85%) in U87 cells compared to the chemical transfection reagent (CTR) (72%). Moreover, the transfection efficiency of the CNPs was also higher, as indicated by fluorescent emission microscopy (20.56% vs. 17.79%) and fluorescent-activated cell sorting (53% vs. 27%). In vivo assays using Balb/c mice revealed that the CNPs could efficiently cross the BBB, suggesting their potential as efficient gene delivery vehicles for targeted therapies against brain cancers as well as other brain diseases for which the efficient targeting of a therapeutic load to the brain cells has proven to be a real challenge.
Collapse
Affiliation(s)
- Ishaq N. Khan
- MIT Media Lab, Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
- Cancer Cell Culture & Precision Oncomedicine Lab, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar 25100, Pakistan;
| | - Shiza Navaid
- School of Chemical and Materials Engineering, National University of Sciences and Technology, Islamabad 44000, Pakistan;
| | - Walifa Waqar
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad 44000, Pakistan;
| | - Deema Hussein
- Neurooncology Translational Group, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia;
| | - Najeeb Ullah
- Cancer Cell Culture & Precision Oncomedicine Lab, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar 25100, Pakistan;
| | - Muhammad Umar Aslam Khan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar;
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Zakir Hussain
- School of Chemical and Materials Engineering, National University of Sciences and Technology, Islamabad 44000, Pakistan;
| | - Aneela Javed
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad 44000, Pakistan;
| |
Collapse
|
9
|
Bataille Backer P, Adekiya TA, Kim Y, Reid TER, Thomas M, Adesina SK. Development of a Targeted SN-38-Conjugate for the Treatment of Glioblastoma. ACS OMEGA 2024; 9:2615-2628. [PMID: 38250376 PMCID: PMC10795035 DOI: 10.1021/acsomega.3c07486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024]
Abstract
Glioblastoma (GBM) is the most aggressive and fatal brain tumor, with approximately 10,000 people diagnosed every year in the United States alone. The typical survival period for individuals with glioblastoma ranges from 12 to 18 months, with significant recurrence rates. Common therapeutic modalities for brain tumors are chemotherapy and radiotherapy. The main challenges with chemotherapy for the treatment of glioblastoma are high toxicity, poor selectivity, and limited accumulation of therapeutic anticancer agents in brain tumors as a result of the presence of the blood-brain barrier. To overcome these challenges, researchers have explored strategies involving the combination of targeting peptides possessing a specific affinity for overexpressed cell-surface receptors with conventional chemotherapy agents via the prodrug approach. This approach results in the creation of peptide drug conjugates (PDCs), which facilitate traversal across the blood-brain barrier (BBB), enable preferential accumulation of chemotherapy within the neoplastic microenvironment, and selectively target cancerous cells. This approach increases accumulation in tumors, thereby improving therapeutic efficiency and minimizing toxicity. Leveraging the affinity of the HAIYPRH (T7) peptide for the transferrin receptor (TfR) overexpressed on the blood-brain barrier and glioma cells, a novel T7-SN-38 peptide drug conjugate was developed. The T7-SN-38 peptide drug conjugate demonstrates about a 2-fold reduction in glide score (binding affinity) compared to T7 while maintaining a comparable orientation within the TfR target site using Schrödinger-2022-3 Maestro 13.3 for ligand preparation and Glide SP-Peptide docking. Additionally, SN-38 extends into a solvent-accessible region, enhancing its susceptibility to protease hydrolysis at the cathepsin B (Cat B) cleavable site. The SN-38-ether-peptide drug conjugate displayed high stability in buffer at physiological pH, and cleavage of the conjugate to release free cytotoxic SN-38 was observed in the presence of exogenous cathepsin B. The synthesized peptide drug conjugate exhibited potent cytotoxic activities in cellular models of glioblastoma in vitro. In addition, blocking transferrin receptors using the free T7 peptide resulted in a notable inhibition of cytotoxicity of the conjugate, which was reversed when exogenous cathepsin B was added to cells. This work demonstrates the potential for targeted drug delivery to the brain in the treatment of glioblastoma using the transferrin receptor-targeted T7-SN-38 conjugate.
Collapse
Affiliation(s)
| | - Tayo Alex Adekiya
- Department
of Pharmaceutical Sciences, Howard University, Washington D.C. 20059, United States
| | - Yushin Kim
- Department
of Pharmaceutical Sciences, Concordia University
of Wisconsin, Mequon, Wisconsin 53097-2402, United States
| | - Terry-Elinor R. Reid
- Department
of Pharmaceutical Sciences, Concordia University
of Wisconsin, Mequon, Wisconsin 53097-2402, United States
| | - Michael Thomas
- Department
of Biology, Howard University, Washington D.C. 20059, United States
| | - Simeon K. Adesina
- Department
of Pharmaceutical Sciences, Howard University, Washington D.C. 20059, United States
| |
Collapse
|
10
|
Chaves JCS, Wasielewska JM, Cuní-López C, Rantanen LM, Lee S, Koistinaho J, White AR, Oikari LE. Alzheimer's disease brain endothelial-like cells reveal differential drug transporter expression and modulation by potentially therapeutic focused ultrasound. Neurotherapeutics 2024; 21:e00299. [PMID: 38241156 PMCID: PMC10903103 DOI: 10.1016/j.neurot.2023.10.009] [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: 10/20/2023] [Accepted: 10/28/2023] [Indexed: 01/21/2024] Open
Abstract
The blood-brain barrier (BBB) has a key function in maintaining homeostasis in the brain, partly modulated by transporters, which are highly expressed in brain endothelial cells (BECs). Transporters mediate the uptake or efflux of compounds to and from the brain and they can also challenge the delivery of drugs for the treatment of Alzheimer's disease (AD). Currently there is a limited understanding of changes in BBB transporters in AD. To investigate this, we generated brain endothelial-like cells (iBECs) from induced pluripotent stem cells (iPSCs) with familial AD (FAD) Presenilin 1 (PSEN1) mutation and identified AD-specific differences in transporter expression compared to control (ctrl) iBECs. We first characterized the expression levels of 12 BBB transporters in AD-, Ctrl-, and isogenic (PSEN1 corrected) iBECs to identify any AD specific differences. We then exposed the cells to focused ultrasound (FUS) in the absence (FUSonly) or presence of microbubbles (MB) (FUS+MB), which is a novel therapeutic method that can be used to transiently open the BBB to increase drug delivery into the brain, however its effects on BBB transporter expression are largely unknown. Following FUSonly and FUS+MB, we investigated whether the expression or activity of key transporters could be modulated. Our findings demonstrate that PSEN1 mutant FAD (PSEN1AD) possess phenotypical differences compared to control iBECs in BBB transporter expression and function. Additionally, we show that FUSonly and FUS+MB can modulate BBB transporter expression and functional activity in iBECs, having potential implications on drug penetration and amyloid clearance. These findings highlight the differential responses of patient cells to FUS treatment, with patient-derived models likely providing an important tool for modelling therapeutic effects of FUS.
Collapse
Affiliation(s)
- Juliana C S Chaves
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QUT, Brisbane, QLD, Australia
| | - Joanna M Wasielewska
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Carla Cuní-López
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Laura M Rantanen
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QUT, Brisbane, QLD, Australia
| | - Serine Lee
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neuroscience Center, Kuopio, Finland; Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Anthony R White
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QUT, Brisbane, QLD, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Lotta E Oikari
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
| |
Collapse
|
11
|
Sahoo L, Tripathy NS, Dilnawaz F. Naringenin Nanoformulations for Neurodegenerative Diseases. Curr Pharm Biotechnol 2024; 25:2108-2124. [PMID: 38347794 DOI: 10.2174/0113892010281459240118091137] [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: 10/12/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 09/10/2024]
Abstract
Glioblastoma (GBM) is a grade-IV astrocytoma, which is the most common and aggressive type of brain tumor, spreads rapidly and has a life-threatening catastrophic effect. GBM mostly occurs in adults with an average survival time of 15 to 18 months, and the overall mortality rate is 5%. Significant invasion and drug resistance activity cause the poor diagnosis of GBM. Naringenin (NRG) is a plant secondary metabolite byproduct of the flavanone subgroup. NRG can cross the blood-brain barrier and deliver drugs into the central nervous system when conjugated with appropriate nanocarriers to overcome the challenges associated with gliomas through naringenin-loaded nanoformulations. Here, we discuss several nanocarriers employed that are as delivery systems, such as polymeric nanoparticles, micelles, liposomes, solid lipid nanoparticles (SLNs), nanosuspensions, and nanoemulsions. These naringenin-loaded nanoformulations have been tested in various in vitro and in vivo models as a potential treatment for brain disorders. This review nanoformulations of NRG can a possible therapeutic alternative for the treatment of neurological diseases are discussed.
Collapse
Affiliation(s)
- Liza Sahoo
- Department of Biotechnology, School of Engineering and Technology, Centurion University of Technology and Management, Jatni, 752050, Bhubaneswar, Odisha, India
| | - Nigam Sekhar Tripathy
- Department of Biotechnology, School of Engineering and Technology, Centurion University of Technology and Management, Jatni, 752050, Bhubaneswar, Odisha, India
| | - Fahima Dilnawaz
- Department of Biotechnology, School of Engineering and Technology, Centurion University of Technology and Management, Jatni, 752050, Bhubaneswar, Odisha, India
| |
Collapse
|
12
|
Koňáriková K, Girašková GM, Žitňanová I, Dvořáková M, Rollerová E, Scsuková S, Bizik J, Janubová M, Muchová J. Biological analyses of the effects of TiO2 and PEG-b-PLA nanoparticles on three-dimensional spheroid-based tumor. Physiol Res 2023; 72:S257-S266. [PMID: 37888969 PMCID: PMC10669953 DOI: 10.33549/physiolres.935152] [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: 06/07/2023] [Accepted: 06/27/2023] [Indexed: 12/01/2023] Open
Abstract
The aim of our study was to monitor the antiproliferative/ cytotoxic and genotoxic effects of both, poly(ethylene glycol)-block-poly(lactic acid) (PEG-b-PLA) and titanium dioxide (TiO2) nanoparticles on the tumor (HT-29, MCF-7, U118MG) and healthy (HEK-293T) cell lines during 2D cultivation and during cultivation in the spheroid form (3D cultivation). Cells or spheroids were cultivated with nanoparticles (0.01, 0.1, 1, 10, 50, and 100 ?g/ml) for 72 hours. The cytotoxic effect was determined by the MTT test and the genotoxic effect by the comet assay. We found that 2D cultivation of tumor cell lines with PEG-b-PLA and TiO2 nanoparticles had an anti-proliferative effect on human colon cancer cell line HT-29, human breast cancer cell line MCF-7, human glioma cell line U-118MG during 72h cultivation, but not on control/healthy HEK-293T cells. At the concentrations used, the tested nanoparticles caused no cytotoxic effect on tumor cell lines. Nanoparticles PEG-b-PLA induced significant damage to DNA in HT-29 and MCF-7 cells, while TiO2 nanoparticles in MCF-7 and U-118MG cells. Only PEG-b-PLA nanoparticles caused cytotoxic (IC50 = 7 mikrog/ml) and genotoxic effects on the healthy cell line HEK-293T after 72h cultivation. The cells which were cultivated in spheroid forms were more sensitive to both types of nanoparticles. After 72h cultivation, we observed the cytotoxic effect on both, the tumor and healthy cell lines.
Collapse
Affiliation(s)
- K Koňáriková
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Pires PC, Paiva-Santos AC, Veiga F. Liposome-Derived Nanosystems for the Treatment of Behavioral and Neurodegenerative Diseases: The Promise of Niosomes, Transfersomes, and Ethosomes for Increased Brain Drug Bioavailability. Pharmaceuticals (Basel) 2023; 16:1424. [PMID: 37895895 PMCID: PMC10610493 DOI: 10.3390/ph16101424] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Psychiatric and neurodegenerative disorders are amongst the most prevalent and debilitating diseases, but current treatments either have low success rates, greatly due to the low permeability of the blood-brain barrier, and/or are connected to severe side effects. Hence, new strategies are extremely important, and here is where liposome-derived nanosystems come in. Niosomes, transfersomes, and ethosomes are nanometric vesicular structures that allow drug encapsulation, protecting them from degradation, and increasing their solubility, permeability, brain targeting, and bioavailability. This review highlighted the great potential of these nanosystems for the treatment of Alzheimer's disease, Parkinson's disease, schizophrenia, bipolar disorder, anxiety, and depression. Studies regarding the encapsulation of synthetic and natural-derived molecules in these systems, for intravenous, oral, transdermal, or intranasal administration, have led to an increased brain bioavailability when compared to conventional pharmaceutical forms. Moreover, the developed formulations proved to have neuroprotective, anti-inflammatory, and antioxidant effects, including brain neurotransmitter level restoration and brain oxidative status improvement, and improved locomotor activity or enhancement of recognition and working memories in animal models. Hence, albeit being relatively new technologies, niosomes, transfersomes, and ethosomes have already proven to increase the brain bioavailability of psychoactive drugs, leading to increased effectiveness and decreased side effects, showing promise as future therapeutics.
Collapse
Affiliation(s)
- Patrícia C. Pires
- Faculty of Pharmacy, Faculty of Pharmacy of the University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana Cláudia Paiva-Santos
- Faculty of Pharmacy, Faculty of Pharmacy of the University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Faculty of Pharmacy, Faculty of Pharmacy of the University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
14
|
Gu Y, Wu L, Hameed Y, Nabi-Afjadi M. Overcoming the challenge: cell-penetrating peptides and membrane permeability. BIOMATERIALS AND BIOSENSORS 2023; 2. [DOI: 10.58567/bab02010002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
<p>Cell-penetrating peptides (CPPs) have emerged as a promising strategy for enhancing the membrane permeability of bioactive molecules, particularly in the treatment of central nervous system diseases. CPPs possess the ability to deliver a diverse array of bioactive molecules into cells using either covalent or non-covalent approaches, with a preference for non-covalent methods to preserve the biological activity of the transported molecules. By effectively traversing various physiological barriers, CPPs have exhibited significant potential in preclinical and clinical drug development. The discovery of CPPs represents a valuable solution to the challenge of limited membrane permeability of bioactive molecules and will continue to exert a crucial influence on the field of biomedical science.</p>
Collapse
Affiliation(s)
- Yuan Gu
- The Statistics Department, The George Washington University, Washington, United States
| | - Long Wu
- Department of Surgery, University of Maryland, Baltimore, United States
| | - Yasir Hameed
- Department of Applied Biological Sciences, Tokyo University of Science, Tokyo, Japan
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
15
|
Sahoo RK, Kumar H, Jain V, Sinha S, Gupta U. Angiopep-2 Grafted PAMAM Dendrimers for the Targeted Delivery of Temozolomide: In Vitro and In Vivo Effects of PEGylation in the Management of Glioblastoma Multiforme. ACS Biomater Sci Eng 2023. [PMID: 37307155 DOI: 10.1021/acsbiomaterials.3c00263] [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: 06/14/2023]
Abstract
The present study was aimed to synthesize, characterize, and evaluate the angiopep-2 grafted PAMAM dendrimers (Den, G 3.0 NH2) with and without PEGylation for the targeted and better delivery approach of temozolomide (TMZ) for the management of glioblastoma multiforme (GBM). Den-ANG and Den-PEG2-ANG conjugates were synthesized and characterized by 1H NMR spectroscopy. The PEGylated (TMZ@Den-PEG2-ANG) and non-PEGylated (TMZ@Den-ANG) drug loaded formulations were prepared and characterized for particle size, zeta potential, entrapment efficiency, and drug loading. An in vitro release study at physiological (pH 7.4) and acidic pH (pH 5.0) was performed. Preliminary toxicity studies were performed through hemolytic assay in human RBCs. MTT assay, cell uptake, and cell cycle analysis were performed to evaluate the in vitro efficacy against GBM cell lines (U87MG). Finally, the formulations were evaluated in vivo in a Sprague-Dawley rat model for pharmacokinetics and organ distribution analysis. The 1H NMR spectra confirmed the conjugation of angiopep-2 to both PAMAM and PEGylated PAMAM dendrimers, as the characteristic chemical shifts were observed in the range of 2.1 to 3.9 ppm. AFM results revealed that the surface of Den-ANG and Den-PEG2-ANG conjugates were rough. The particle size and zeta potential of TMZ@Den-ANG were observed to be 229.0 ± 17.8 nm and 9.06 ± 0.4 mV, respectively, whereas the same for TMZ@Den-PEG2-ANG were found to be 249.6 ± 12.9 nm and 10.9 ± 0.6 mV, respectively. The entrapment efficiency of TMZ@Den-ANG and TMZ@Den-PEG2-ANG were calculated to be 63.27 ± 5.1% and 71.48 ± 4.3%, respectively. Moreover, TMZ@Den-PEG2-ANG showed a better drug release profile with a controlled and sustained pattern at PBS pH 5.0 than at pH 7.4. The ex vivo hemolytic study revealed that TMZ@Den-PEG2-ANG was biocompatible in nature as it showed 2.78 ± 0.1% hemolysis compared to 4.12 ± 0.2% hemolysis displayed by TMZ@Den-ANG. The outcomes of the MTT assay inferred that TMZ@Den-PEG2-ANG possessed maximum cytotoxic effects against U87MG cells with IC50 values of 106.62 ± 11.43 μM (24 h) and 85.90 ± 9.12 μM (48 h). In the case of TMZ@Den-PEG2-ANG, the IC50 values were reduced by 2.23-fold (24 h) and 1.36-fold (48 h) in comparison to pure TMZ. The cytotoxicity findings were further confirmed by significantly higher cellular uptake of TMZ@Den-PEG2-ANG. Cell cycle analysis of the formulations suggested that the PEGylated formulation halts the cell cycle at G2/M phase with S-phase inhibition. In the in vivo studies, the half-life (t1/2) values of TMZ@Den-ANG and TMZ@Den-PEG2-ANG were enhanced by 2.22 and 2.76 times, respectively, than the pure TMZ. After 4 h of administration, the brain uptake values of TMZ@Den-ANG and TMZ@Den-PEG2-ANG were found to be 2.55 and 3.35 times, respectively, higher than that of pure TMZ. The outcomes of various in vitro and ex vivo experiments promoted the use of PEGylated nanocarriers for the management of GBM. Angiopep-2 grafted PEGylated PAMAM dendrimers can be potential and promising drug carriers for the targeted delivery of antiglioma drugs directly to the brain.
Collapse
Affiliation(s)
- Rakesh Kumar Sahoo
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka 570015, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka 570015, India
| | - Sonal Sinha
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh 490024, India
| | - Umesh Gupta
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| |
Collapse
|
16
|
Khatami SH, Karami N, Taheri-Anganeh M, Taghvimi S, Tondro G, Khorsand M, Soltani Fard E, Sedighimehr N, Kazemi M, Rahimi Jaberi K, Moradi M, Nafisi Fard P, Darvishi MH, Movahedpour A. Exosomes: Promising Delivery Tools for Overcoming Blood-Brain Barrier and Glioblastoma Therapy. Mol Neurobiol 2023:10.1007/s12035-023-03365-0. [PMID: 37138197 PMCID: PMC10155653 DOI: 10.1007/s12035-023-03365-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/19/2023] [Indexed: 05/05/2023]
Abstract
Gliomas make up virtually 80% of all lethal primary brain tumors and are categorized based on their cell of origin. Glioblastoma is an astrocytic tumor that has an inferior prognosis despite the ongoing advances in treatment modalities. One of the main reasons for this shortcoming is the presence of the blood-brain barrier and blood-brain tumor barrier. Novel invasive and non-invasive drug delivery strategies for glioblastoma have been developed to overcome both the intact blood-brain barrier and leverage the disrupted nature of the blood-brain tumor barrier to target cancer cells after resection-the first treatment stage of glioblastoma. Exosomes are among non-invasive drug delivery methods and have emerged as a natural drug delivery vehicle with high biological barrier penetrability. There are various exosome isolation methods from different origins, and the intended use of the exosomes and starting materials defines the choice of isolation technique. In the present review, we have given an overview of the structure of the blood-brain barrier and its disruption in glioblastoma. This review provided a comprehensive insight into novel passive and active drug delivery techniques to overcome the blood-brain barrier, emphasizing exosomes as an excellent emerging drug, gene, and effective molecule delivery vehicle used in glioblastoma therapy.
Collapse
Affiliation(s)
- Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Karami
- TU Wien, Institute of Solid State Electronics, A-1040, Vienna, Austria
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Sina Taghvimi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Gholamhossein Tondro
- Microbiology Department, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Marjan Khorsand
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elahe Soltani Fard
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Najmeh Sedighimehr
- Department of Physical Therapy, School of Rehabilitation, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Kazemi
- Department of Radio-oncology, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Khojaste Rahimi Jaberi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Melika Moradi
- Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Parvaneh Nafisi Fard
- Department of Veterinary Clinical Science, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mohammad Hasan Darvishi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
17
|
Tran TH, Tran PTT, Truong DH. Lactoferrin and Nanotechnology: The Potential for Cancer Treatment. Pharmaceutics 2023; 15:pharmaceutics15051362. [PMID: 37242604 DOI: 10.3390/pharmaceutics15051362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Lactoferrin (Lf)-a glycoprotein of the transferrin family-has been investigated as a promising molecule with diverse applications, including infection inhibition, anti-inflammation, antioxidant properties and immune modulation. Along with that, Lf was found to inhibit the growth of cancerous tumors. Owing to unique properties such as iron-binding and positive charge, Lf could interrupt the cancer cell membrane or influence the apoptosis pathway. In addition, being a common mammalian excretion, Lf offers is promising in terms of targeting delivery or the diagnosis of cancer. Recently, nanotechnology significantly enhanced the therapeutic index of natural glycoproteins such as Lf. Therefore, in the context of this review, the understanding of Lf is summarized and followed by different strategies of nano-preparation, including inorganic nanoparticles, lipid-based nanoparticles and polymer-based nanoparticles in cancer management. At the end of the study, the potential future applications are discussed to pave the way for translating Lf into actual usage.
Collapse
Affiliation(s)
- Tuan Hiep Tran
- Faculty of Pharmacy, Phenikaa University, Yen Nghia, Ha Dong, Hanoi 12116, Vietnam
| | - Phuong Thi Thu Tran
- Department of Life Sciences, University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi 10000, Vietnam
| | | |
Collapse
|
18
|
Yalamarty SSK, Filipczak N, Li X, Subhan MA, Parveen F, Ataide JA, Rajmalani BA, Torchilin VP. Mechanisms of Resistance and Current Treatment Options for Glioblastoma Multiforme (GBM). Cancers (Basel) 2023; 15:cancers15072116. [PMID: 37046777 PMCID: PMC10093719 DOI: 10.3390/cancers15072116] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive form of brain cancer that is difficult to treat due to its resistance to both radiation and chemotherapy. This resistance is largely due to the unique biology of GBM cells, which can evade the effects of conventional treatments through mechanisms such as increased resistance to cell death and rapid regeneration of cancerous cells. Additionally, the blood–brain barrier makes it difficult for chemotherapy drugs to reach GBM cells, leading to reduced effectiveness. Despite these challenges, there are several treatment options available for GBM. The standard of care for newly diagnosed GBM patients involves surgical resection followed by concurrent chemoradiotherapy and adjuvant chemotherapy. Emerging treatments include immunotherapy, such as checkpoint inhibitors, and targeted therapies, such as bevacizumab, that attempt to attack specific vulnerabilities in GBM cells. Another promising approach is the use of tumor-treating fields, a type of electric field therapy that has been shown to slow the growth of GBM cells. Clinical trials are ongoing to evaluate the safety and efficacy of these and other innovative treatments for GBM, intending to improve with outcomes for patients.
Collapse
Affiliation(s)
- Satya Siva Kishan Yalamarty
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Xiang Li
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Farzana Parveen
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- Department of Pharmacy Services, DHQ Hospital, Jhang 35200, Pakistan
| | - Janaína Artem Ataide
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas 13083-871, Brazil
| | - Bharat Ashok Rajmalani
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Vladimir P. Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
19
|
Rationally designed donepezil-based hydroxamates modulate Sig-1R and HDAC isoforms to exert anti-glioblastoma effects. Eur J Med Chem 2023; 248:115054. [PMID: 36630883 DOI: 10.1016/j.ejmech.2022.115054] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023]
Abstract
The pursuit of activating the HDAC inhibitory template towards additional mechanisms spurred us to design dual modulators (Sig-1R agonist - HDAC inhibitor) via utilization of the core structural unit of donepezil (an FDA-approved anti-Alzheimer's agent) as a surface recognition part. Literature precedents coupled with our experience rendered us with several insights that led to the inclusion of chemically diverse linkers and hydroxamic acid (zinc-binding motif) as the other components of HDAC inhibitory pharmacophore. With this envisionment and clarity, donepezil-based HDAC inhibitory adducts were furnished and exhaustively explored for their anti-GBM efficacy. Resultantly, a magnificently potent HDAC inhibitor 10 [IC50 (HDAC6) = 2.7 nM, IC50 (HDAC2) = 0.71 μM] was pinpointed that was endowed with the ability to: i) exert cell growth inhibitory effects against Human U87MG GBM cells ii) cause death in TMZ-resistant GBM cells iii) induce subG1 arrest in GBM cells iv) prolong the survival of TMZ-resistant U87MG inoculated orthotopic mice (in-vivo studies) v) induce GBM cell apoptosis via binding to Sig-1R. Collectively, the results led to the identification of compound 10 as a tractable anti-GBM agent that deserves detailed investigation for the accomplishment of its candidature as a GBM therapeutic.
Collapse
|
20
|
Ghosh A, Chakraborty D, Mukerjee N, Baishya D, Chigurupati S, Felemban SG, Almahmoud SA, Almikhlafi MA, Sehgal A, Singh S, Sharma N, Aleya L, Behl T. Target-based virtual screening and molecular interaction studies for lead identification of natural olive compounds against glioblastoma multiforme. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:6170-6191. [PMID: 35994146 DOI: 10.1007/s11356-022-22401-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
Glioblastoma multiforme, a rare traumatic brain disorder, is at the research climax for its uncontrolled growth leading to a catastrophic outcome. Throwing light on the target-based virtual screening of drugs using natural phytocompounds is a striking cornerstone in glioblastoma-based drug discovery, accelerating with leaps and bounds. This project aims to develop promising lead compounds against glioblastoma brain cancer using OliveNet™, an open-source database. In this pursuit, our rationale for selecting molecules was based on their capability to pass through the blood-brain barrier. Out of 51 derivative molecules from flavonoids and polyphenols, 17 molecules were screened out bearing the best ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties, alongside fulfilling our rationale of lead selection. Two polyphenols, 3,4,5-trimethoxybenzoic acid and 4-ethyl guaiacol, have binding affinity for the antioxidant flavonoid luteolin of -5.1 and -4.3 kcal/mol, respectively. According to docking studies, the residues ASN1960, ASN1966, ASN1960, PHE1984, TYR1896, VAL1911, and LYS1966 make both polar and nonpolar interactions with 3,4,5-trimethoxybenzoic acid and 4-ethylguanidine, respectively. LD50 values of toxicity screening using TOX Pro brought to limelight the excellent safety profile of polyphenols and flavonoids. Furthermore, studies using in silico cytotoxicity prediction and molecular modelling have decisively shown that these polyphenols are likely to be effective brain cancer inhibitors and promising future lead candidates against glioblastoma multiforme.
Collapse
Affiliation(s)
- Arabinda Ghosh
- Microbiology Division, Department of Botany, Gauhati University, Guwahati, Assam, India
| | - Dipanwita Chakraborty
- Department of Molecular Biology and Biotechnology, Cotton University, Guwahati, Assam, India
| | - Nobendu Mukerjee
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Kolkata, West Bengal, India
| | - Debabrat Baishya
- Department of Bioengineering and Technology, GUIST, Gauhati University, Guwahati, Assam, India
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Shatha Ghazi Felemban
- Department of Medical Laboratory Science, Fakeeh College for Medical Sciences, Jeddah, Kingdom of Saudi Arabia
| | - Suliman A Almahmoud
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Mohannad A Almikhlafi
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madina, Kingdom of Saudi Arabia
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sukhbir Singh
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133207, Haryana, India
| | - Neelam Sharma
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133207, Haryana, India
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besancon, France
| | - Tapan Behl
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India.
| |
Collapse
|
21
|
Rhaman MM, Islam MR, Akash S, Mim M, Noor alam M, Nepovimova E, Valis M, Kuca K, Sharma R. Exploring the role of nanomedicines for the therapeutic approach of central nervous system dysfunction: At a glance. Front Cell Dev Biol 2022; 10:989471. [PMID: 36120565 PMCID: PMC9478743 DOI: 10.3389/fcell.2022.989471] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/08/2022] [Indexed: 12/12/2022] Open
Abstract
In recent decades, research scientists, molecular biologists, and pharmacologists have placed a strong emphasis on cutting-edge nanostructured materials technologies to increase medicine delivery to the central nervous system (CNS). The application of nanoscience for the treatment of neurodegenerative diseases (NDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), Huntington’s disease (HD), brain cancer, and hemorrhage has the potential to transform care. Multiple studies have indicated that nanomaterials can be used to successfully treat CNS disorders in the case of neurodegeneration. Nanomedicine development for the cure of degenerative and inflammatory diseases of the nervous system is critical. Nanoparticles may act as a drug transporter that can precisely target sick brain sub-regions, boosting therapy success. It is important to develop strategies that can penetrate the blood–brain barrier (BBB) and improve the effectiveness of medications. One of the probable tactics is the use of different nanoscale materials. These nano-based pharmaceuticals offer low toxicity, tailored delivery, high stability, and drug loading capacity. They may also increase therapeutic effectiveness. A few examples of the many different kinds and forms of nanomaterials that have been widely employed to treat neurological diseases include quantum dots, dendrimers, metallic nanoparticles, polymeric nanoparticles, carbon nanotubes, liposomes, and micelles. These unique qualities, including sensitivity, selectivity, and ability to traverse the BBB when employed in nano-sized particles, make these nanoparticles useful for imaging studies and treatment of NDs. Multifunctional nanoparticles carrying pharmacological medications serve two purposes: they improve medication distribution while also enabling cell dynamics imaging and pharmacokinetic study. However, because of the potential for wide-ranging clinical implications, safety concerns persist, limiting any potential for translation. The evidence for using nanotechnology to create drug delivery systems that could pass across the BBB and deliver therapeutic chemicals to CNS was examined in this study.
Collapse
Affiliation(s)
- Md. Mominur Rhaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mobasharah Mim
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Noor alam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
| | - Martin Valis
- Department of Neurology, Charles University in Prague, Faculty of Medicine in Hradec Králové and University Hospital, Hradec Králové, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| |
Collapse
|
22
|
Israel LL, Galstyan A, Cox A, Shatalova ES, Sun T, Rashid MH, Grodzinski Z, Chiechi A, Fuchs DT, Patil R, Koronyo-Hamaoui M, Black KL, Ljubimova JY, Holler E. Signature Effects of Vector-Guided Systemic Nano Bioconjugate Delivery Across Blood-Brain Barrier of Normal, Alzheimer's, and Tumor Mouse Models. ACS NANO 2022; 16:11815-11832. [PMID: 35961653 PMCID: PMC9413444 DOI: 10.1021/acsnano.1c10034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The ability to cross the blood-brain barrier (BBB) is critical for targeted therapy of the central nerve system (CNS). Six peptide vectors were covalently attached to a 50 kDa poly(β-l-malic acid)-trileucine polymer forming P/LLL(40%)/vector conjugates. The vectors were Angiopep-2 (AP2), B6, Miniap-4 (M4), and d-configurated peptides D1, D3, and ACI-89, with specificity for transcytosis receptors low-density lipoprotein receptor-related protein-1 (LRP-1), transferrin receptor (TfR), bee venom-derived ion channel, and Aβ/LRP-1 related transcytosis complex, respectively. The BBB-permeation efficacies were substantially increased ("boosted") in vector conjugates of P/LLL(40%). We have found that the copolymer group binds at the endothelial membrane and, by an allosterically membrane rearrangement, exposes the sites for vector-receptor complex formation. The specificity of vectors is indicated by competition experiments with nonconjugated vectors. P/LLL(40%) does not function as an inhibitor, suggesting that the copolymer binding site is eliminated after binding of the vector-nanoconjugate. The two-step mechanism, binding to endothelial membrane and allosteric exposure of transcytosis receptors, is supposed to be an integral feature of nanoconjugate-transcytosis pathways. In vivo brain delivery signatures of the nanoconjugates were recapitulated in mouse brains of normal, tumor (glioblastoma), and Alzheimer's disease (AD) models. BBB permeation of the tumor was most efficient, followed by normal and then AD-like brain. In tumor-bearing and normal brains, AP2 was the top performing vector; however, in AD models, D3 and D1 peptides were superior ones. The TfR vector B6 was equally efficient in normal and AD-model brains. Cross-permeation efficacies are manifested through modulated vector coligation and dosage escalation such as supra-linear dose dependence and crossover transcytosis activities.
Collapse
Affiliation(s)
- Liron L. Israel
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Anna Galstyan
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Alysia Cox
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Ekaterina S. Shatalova
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Tao Sun
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Mohammad-Harun Rashid
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Zachary Grodzinski
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Antonella Chiechi
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Rameshwar Patil
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery and Department of Biomedical Sciences,
Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los
Angeles, California 90048, United States
| | - Keith L. Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Julia Y. Ljubimova
- Terasaki Institute for Biomedical Innovation
(TIBI), 1018 Westwood
Boulevard, Los Angeles, California 90024, United States
| | - Eggehard Holler
- Terasaki Institute for Biomedical Innovation
(TIBI), 1018 Westwood
Boulevard, Los Angeles, California 90024, United States
| |
Collapse
|
23
|
Asija S, Chatterjee A, Yadav S, Chekuri G, Karulkar A, Jaiswal AK, Goda JS, Purwar R. Combinatorial approaches to effective therapy in glioblastoma (GBM): Current status and what the future holds. Int Rev Immunol 2022; 41:582-605. [PMID: 35938932 DOI: 10.1080/08830185.2022.2101647] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
The aggressive and recurrent nature of glioblastoma is multifactorial and has been attributed to its biological heterogeneity, dysfunctional metabolic signaling pathways, rigid blood-brain barrier, inherent resistance to standard therapy due to the stemness property of the gliomas cells, immunosuppressive tumor microenvironment, hypoxia and neoangiogenesis which are very well orchestrated and create the tumor's own highly pro-tumorigenic milieu. Once the relay of events starts amongst these components, eventually it becomes difficult to control the cascade using only the balanced contemporary care of treatment consisting of maximal resection, radiotherapy and chemotherapy with temozolamide. Over the past few decades, implementation of contemporary treatment modalities has shown benefit to some extent, but no significant overall survival benefit is achieved. Therefore, there is an unmet need for advanced multifaceted combinatorial strategies. Recent advances in molecular biology, development of innovative therapeutics and novel delivery platforms over the years has resulted in a paradigm shift in gliomas therapeutics. Decades of research has led to emergence of several treatment molecules, including immunotherapies such as immune checkpoint blockade, oncolytic virotherapy, adoptive cell therapy, nanoparticles, CED and BNCT, each with the unique proficiency to overcome the mentioned challenges, present research. Recent years are seeing innovative combinatorial strategies to overcome the multifactorial resistance put forth by the GBM cell and its TME. This review discusses the contemporary and the investigational combinatorial strategies being employed to treat GBM and summarizes the evidence accumulated till date.
Collapse
Affiliation(s)
- Sweety Asija
- Department of Biosciences & Bioengineering, Indian Institute of Technology, Mumbai, India
| | - Abhishek Chatterjee
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sandhya Yadav
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Godhanjali Chekuri
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Atharva Karulkar
- Department of Biosciences & Bioengineering, Indian Institute of Technology, Mumbai, India
| | - Ankesh Kumar Jaiswal
- Department of Biosciences & Bioengineering, Indian Institute of Technology, Mumbai, India
| | - Jayant S Goda
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Rahul Purwar
- Department of Biosciences & Bioengineering, Indian Institute of Technology, Mumbai, India
| |
Collapse
|
24
|
Exosomes and Other Extracellular Vesicles with High Therapeutic Potential: Their Applications in Oncology, Neurology, and Dermatology. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041303. [PMID: 35209095 PMCID: PMC8879284 DOI: 10.3390/molecules27041303] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023]
Abstract
Until thirty years ago, it was believed that extracellular vesicles (EVs) were used to remove unnecessary compounds from the cell. Today, we know about their enormous potential in diagnosing and treating various diseases. EVs are essential mediators of intercellular communication, enabling the functional transfer of bioactive molecules from one cell to another. Compared to laboratory-created drug nanocarriers, they are stable in physiological conditions. Furthermore, they are less immunogenic and cytotoxic compared to polymerized vectors. Finally, EVs can transfer cargo to particular cells due to their membrane proteins and lipids, which can implement them to specific receptors in the target cells. Recently, new strategies to produce ad hoc exosomes have been devised. Cells delivering exosomes have been genetically engineered to overexpress particular macromolecules, or transformed to release exosomes with appropriate targeting molecules. In this way, we can say tailor-made therapeutic EVs are created. Nevertheless, there are significant difficulties to solve during the application of EVs as drug-delivery agents in the clinic. This review explores the diversity of EVs and the potential therapeutic options for exosomes as natural drug-delivery vehicles in oncology, neurology, and dermatology. It also reflects future challenges in clinical translation.
Collapse
|
25
|
Exploring the Antiglioma Mechanisms of Luteolin Based on Network Pharmacology and Experimental Verification. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:7765658. [PMID: 34873410 PMCID: PMC8643232 DOI: 10.1155/2021/7765658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/27/2021] [Accepted: 10/30/2021] [Indexed: 11/17/2022]
Abstract
Luteolin, a natural flavone compound, exists in a variety of fruits and vegetables, and its anticancer effect has been shown in many studies. However, its use in glioma treatment is hampered due to the fact that the underlying mechanism of action has not been fully explored. Therefore, we elucidated the potential antiglioma targets and pathways of luteolin systematically with the help of network pharmacology and molecular docking technology. The druggability of luteolin, including absorption, excretion, distribution, and metabolism, was assessed via the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). The potential targets of luteolin and glioma were extracted from public databases, and the intersecting targets between luteolin and glioma were integrated and visualized by a Venn diagram. In addition, GO and KEGG pathway analysis was engaged in Metascape. The network of the luteolin-target-pathway was visualized by Cytoscape. Ultimately, the interactions between luteolin and predicted key targets were confirmed by Discovery studio software. According to the ADME results, luteolin shows great potential for development into a drug. 4860 glioma-associated targets and 280 targets of luteolin were identified, of which 205 were intersection targets. 6 core targets of luteolin against glioma, including AKT1, JUN, ALB, MAPK3, MAPK1, and TNF, were identified via PPI network analysis of which AKT1, JUN, ALB, MAPK1, and TNF harbor diagnostic value. The biological processes of luteolin are mainly involved in the response to inorganic substances, response to oxidative stress, and apoptotic signaling pathway. The essential pathways of luteolin against glioma involve pathways in cancer, the PI3K-Akt signaling pathway, the TNF signaling pathway, and more. Meanwhile, luteolin's interaction with six core targets was verified by molecular docking simulation and its antiglioma effect was verified by in vitro experiments. This study suggests that luteolin has a promising potential for development into a drug and, moreover, it displays preventive effects against glioma by targeting various genes and pathways.
Collapse
|
26
|
Calabrese G, De Luca G, Nocito G, Rizzo MG, Lombardo SP, Chisari G, Forte S, Sciuto EL, Conoci S. Carbon Dots: An Innovative Tool for Drug Delivery in Brain Tumors. Int J Mol Sci 2021; 22:11783. [PMID: 34769212 PMCID: PMC8583729 DOI: 10.3390/ijms222111783] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 01/16/2023] Open
Abstract
Brain tumors are particularly aggressive and represent a significant cause of morbidity and mortality in adults and children, affecting the global population and being responsible for 2.6% of all cancer deaths (as well as 30% of those in children and 20% in young adults). The blood-brain barrier (BBB) excludes almost 100% of the drugs targeting brain neoplasms, representing one of the most significant challenges to current brain cancer therapy. In the last decades, carbon dots have increasingly played the role of drug delivery systems with theranostic applications against cancer, thanks to their bright photoluminescence, solubility in bodily fluids, chemical stability, and biocompatibility. After a summary outlining brain tumors and the current drug delivery strategies devised in their therapeutic management, this review explores the most recent literature about the advances and open challenges in the employment of carbon dots as both diagnostic and therapeutic agents in the treatment of brain cancers, together with the strategies devised to allow them to cross the BBB effectively.
Collapse
Affiliation(s)
- Giovanna Calabrese
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali—Università degli Studi di Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (G.N.); (M.G.R.); (S.C.)
| | - Giovanna De Luca
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali—Università degli Studi di Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (G.N.); (M.G.R.); (S.C.)
| | - Giuseppe Nocito
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali—Università degli Studi di Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (G.N.); (M.G.R.); (S.C.)
| | - Maria Giovanna Rizzo
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali—Università degli Studi di Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (G.N.); (M.G.R.); (S.C.)
| | - Sofia Paola Lombardo
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (S.P.L.); (G.C.)
| | - Giulia Chisari
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (S.P.L.); (G.C.)
| | - Stefano Forte
- IOM Ricerca, Via Penninazzo 11, 95029 Viagrande, Italy;
| | - Emanuele Luigi Sciuto
- A.O.-Universitaria Policlinico “G. Rodolico–San Marco”, Via Santa Sofia 78, 95123 Catania, Italy;
| | - Sabrina Conoci
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali—Università degli Studi di Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (G.N.); (M.G.R.); (S.C.)
| |
Collapse
|
27
|
Schiera G, Di Liegro CM, Di Liegro I. Involvement of Thyroid Hormones in Brain Development and Cancer. Cancers (Basel) 2021; 13:2693. [PMID: 34070729 PMCID: PMC8197921 DOI: 10.3390/cancers13112693] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/21/2022] Open
Abstract
The development and maturation of the mammalian brain are regulated by thyroid hormones (THs). Both hypothyroidism and hyperthyroidism cause serious anomalies in the organization and function of the nervous system. Most importantly, brain development is sensitive to TH supply well before the onset of the fetal thyroid function, and thus depends on the trans-placental transfer of maternal THs during pregnancy. Although the mechanism of action of THs mainly involves direct regulation of gene expression (genomic effects), mediated by nuclear receptors (THRs), it is now clear that THs can elicit cell responses also by binding to plasma membrane sites (non-genomic effects). Genomic and non-genomic effects of THs cooperate in modeling chromatin organization and function, thus controlling proliferation, maturation, and metabolism of the nervous system. However, the complex interplay of THs with their targets has also been suggested to impact cancer proliferation as well as metastatic processes. Herein, after discussing the general mechanisms of action of THs and their physiological effects on the nervous system, we will summarize a collection of data showing that thyroid hormone levels might influence cancer proliferation and invasion.
Collapse
Affiliation(s)
- Gabriella Schiera
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche) (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.S.); (C.M.D.L.)
| | - Carlo Maria Di Liegro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche) (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.S.); (C.M.D.L.)
| | - Italia Di Liegro
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Dipartimento di Biomedicina, Neuroscienze e Diagnostica avanzata) (Bi.N.D.), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
28
|
Oberacker E, Diesch C, Nadobny J, Kuehne A, Wust P, Ghadjar P, Niendorf T. Patient-Specific Planning for Thermal Magnetic Resonance of Glioblastoma Multiforme. Cancers (Basel) 2021; 13:cancers13081867. [PMID: 33919701 PMCID: PMC8070230 DOI: 10.3390/cancers13081867] [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: 01/31/2021] [Revised: 03/26/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Hyperthermia was proven to enhance the efficacy of chemo- and radiation therapy treatment of glioblastoma multiforme, an aggressive brain tumor of poor prognosis. Despite good clinical results in other tumor types and locations, hyperthermia induced by electromagnetic waves in the radiofrequency range is not available so far for the treatment of brain tumors due to the highly sensitive surrounding tissue and lack of non-invasive therapy monitoring. ThermalMR integrates non-invasive diagnosis, therapy, and therapy monitoring in a single RF applicator device by employing radiowaves for magnetic resonance imaging, radiofrequency heating, as well as magnetic resonance thermometry. This work examines three optimization algorithms for hyperthermia treatment planning and up to ten RF applicator configurations for a cohort of nine patient models with glioblastoma multiforme. Clinical diversity is represented in target size and location and the inclusion of post-operative models. Our findings indicate the need and potential for patient-specific treatment planning and RF applicator design when targeting brain tumors. Abstract Thermal intervention is a potent sensitizer of cells to chemo- and radiotherapy in cancer treatment. Glioblastoma multiforme (GBM) is a potential clinical target, given the cancer’s aggressive nature and resistance to current treatment options. This drives research into optimization algorithms for treatment planning as well as radiofrequency (RF) applicator design for treatment delivery. In this work, nine clinically realistic GBM target volumes (TVs) for thermal intervention are compared using three optimization algorithms and up to ten RF applicator designs for thermal magnetic resonance. Hyperthermia treatment planning (HTP) was successfully performed for all cases, including very small, large, and even split target volumes. Minimum requirements formulated for the metrics assessing HTP outcome were met and exceeded for all patient specific cases. Results indicate a 16 channel two row arrangement to be most promising. HTP of TVs with a small extent in the cranial–caudal direction in conjunction with a large radial extent remains challenging despite the advanced optimization algorithms used. In general, deep seated targets are favorable. Overall, our findings indicate that a one-size-fits-all RF applicator might not be the ultimate approach in hyperthermia of brain tumors. It stands to reason that modular and reconfigurable RF applicator configurations might best suit the needs of targeting individual GBM geometry.
Collapse
Affiliation(s)
- Eva Oberacker
- Berlin Ultrahigh Field Facility, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (C.D.); (T.N.)
- Department Radiation Oncology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (J.N.); (P.W.); (P.G.)
- Department of Physics, Faculty of Mathematics and Natural Sciences, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Correspondence: ; Tel.: +49-(0)30-450-557188
| | - Cecilia Diesch
- Berlin Ultrahigh Field Facility, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (C.D.); (T.N.)
| | - Jacek Nadobny
- Department Radiation Oncology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (J.N.); (P.W.); (P.G.)
| | | | - Peter Wust
- Department Radiation Oncology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (J.N.); (P.W.); (P.G.)
| | - Pirus Ghadjar
- Department Radiation Oncology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (J.N.); (P.W.); (P.G.)
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (C.D.); (T.N.)
- MRI.TOOLS GmbH, 13125 Berlin, Germany;
- Experimental and Clinical Research Center, Joint Cooperation between Charité Unversitätsmedizin and the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| |
Collapse
|
29
|
Nepali K, Hsu TI, Hsieh CM, Lo WL, Lai MJ, Hsu KC, Lin TE, Chuang JY, Liou JP. Pragmatic recruitment of memantine as the capping group for the design of HDAC inhibitors: A preliminary attempt to unravel the enigma of glioblastoma. Eur J Med Chem 2021; 217:113338. [PMID: 33744690 DOI: 10.1016/j.ejmech.2021.113338] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/01/2021] [Accepted: 02/19/2021] [Indexed: 02/04/2023]
Abstract
Hurdled and marred by the notorious nature of glioblastomas (GBM) in terms of resistance to therapy and limited drug delivery into the brain, the anti-GBM drug pipeline is required to be loaded with mechanistically diverse agents. The consideration of HDAC inhibition as a prudent approach to circumvent the resistance issue in GBM spurred us to pragmatically design and synthesizes hydroxamic acids endowed with CNS penetrating ability. By virtue of the blood brain barrier permeability (BBB), memantine was envisioned as an appropriate CAP component for the construction of the HDAC inhibitors. Diverse linkers were stapled for the tetheration of the zinc binding motif with the CAP group to pinpoint an appropriate combination (CAP and linker) that could confer inhibitory preference to HDAC6 isoform (overexpressed in GBM). Resultantly, hydroxamic acid 16 was identified as a promising compound that elicited striking antiproliferative effects against Human U87MG GBM cells as well as TMZ-resistant GBM cells and P1S cells, a concurrent chemo radiotherapy (CCRT)-resistant/patient-derived glioma cell line mediated through preferential HDAC6 inhibition (IC50 = 5.42 nM). Furthermore, 16 exerted cell cycle arrest at G2 phase, induced apoptosis in GBM cells at high concentration and exhibited high BBB permeability. To add on, in-vivo study revealed that the administration of compound 16 prolonged the survival of TMZ-resistant U87MG inoculated orthotopic mice. Overall, the cumulative results indicate that 16 is a tractable CNS penetrant preferential HDAC6 inhibitor that might emerge as a potent weapon against GBM.
Collapse
Affiliation(s)
- Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Tsung-I Hsu
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taiwan
| | - Chien-Ming Hsieh
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Wei-Lun Lo
- Division of Neurosurgery, Taipei Medical University-Shuang-Ho Hospital, Taiwan
| | - Mei-Jung Lai
- Biomedical Commercialization Center, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kai-Cheng Hsu
- Biomedical Commercialization Center, Taipei Medical University, Taipei, 11031, Taiwan; Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan; Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taiwan
| | - Tony Eight Lin
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taiwan
| | - Jian-Ying Chuang
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taiwan.
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan; Biomedical Commercialization Center, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
30
|
Integrin α vβ 3-targeted liposomal drug delivery system for enhanced lung cancer therapy. Colloids Surf B Biointerfaces 2021; 201:111623. [PMID: 33636597 DOI: 10.1016/j.colsurfb.2021.111623] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/25/2022]
Abstract
Conventional chemotherapy for tumor treatment remains flawed because it fails to limit cytotoxicity to a small set of selectable tissues. Active targeting techniques for the delivery of drugs to specific sites are increasingly used to enhance drug accumulation at tumor sites with the aim of reducing side effects in vivo. Liposomes, modified with different targeting ligands, are considered to be one of the most promising targeted drug carriers. Herein, novel linear and cyclic arginine-glycine-aspartate (RGD) peptide-based lipids were synthesized to develop modified liposomal drug delivery systems with active targeting and pH-sensitivity. The RGD-modified liposomes showed excellent active targeting ability for integrin αvβ3 receptors, resulting in improved cellular uptake. The modified liposomes also enhanced intracellular doxorubicin (DOX) release because of their degradation in an acidic environment. Consequently, the RGD-modified, DOX-loaded liposomes exhibited significant antitumor efficacy and low toxicity in vitro and in vivo. In particular, 5% cRGD-lipid modified DOX-loaded liposome showed the greatest inhibition of tumor growth in mice among the tested formulations, and much less toxicity than free DOX. In conclusion, the DOX-loaded pH-sensitive liposome modified with 5% cRGD-lipid developed in the current study provides a potential approach for improved tumor therapy.
Collapse
|
31
|
Zhou Y, Wang Y, Chen H, Xu Y, Luo Y, Deng Y, Zhang J, Shao A. Immuno-oncology: are TAM receptors in glioblastoma friends or foes? Cell Commun Signal 2021; 19:11. [PMID: 33509214 PMCID: PMC7841914 DOI: 10.1186/s12964-020-00694-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022] Open
Abstract
Tyro3, Axl, and Mertk (TAM) receptors are a subfamily of receptor tyrosine kinases. TAM receptors have been implicated in mediating efferocytosis, regulation of immune cells, secretion of inflammatory factors, and epithelial-to-mesenchymal transition in the tumor microenvironment, thereby serving as a critical player in tumor development and progression. The pro-carcinogenic role of TAM receptors has been widely confirmed, overexpression of TAM receptors is tied to tumor cells growth, metastasis, invasion and treatment resistance. Nonetheless, it is surprising to detect that inhibiting TAM signaling is not all beneficial in the tumor immune microenvironment. The absence of TAM receptors also affects anti-tumor immunity under certain conditions by modulating different immune cells, as the functional diversification of TAM signaling is closely related to tumor immunotherapy. Glioblastoma is the most prevalent and lethal primary brain tumor in adults. Although research regarding the crosstalk between TAM receptors and glioblastoma remains scarce, it appears likely that TAM receptors possess potential anti-tumor effects rather than portraying a total cancer-driving role in the context of glioblastoma. Accordingly, we doubt whether TAM receptors play a double-sided role in glioblastoma, and propose the Janus-faced TAM Hypothesis as a conceptual framework for comprehending the precise underlying mechanisms of TAMs. In this study, we aim to cast a spotlight on the potential multidirectional effects of TAM receptors in glioblastoma and provide a better understanding for TAM receptor-related targeted intervention. Video Abstract
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yali Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hailong Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yanyan Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, 211126, Jiangsu, China
| | - Yi Luo
- The Second Affiliated Hospital of Zhejiang University School of Medicine (Changxing Branch), Changxing, Huzhou, 313100, Zhejiang, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
32
|
Patel D, Wairkar S, Yergeri MC. Current Developments in Targeted Drug Delivery Systems for Glioma. Curr Pharm Des 2021; 26:3973-3984. [PMID: 32329681 DOI: 10.2174/1381612826666200424161929] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/01/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Glioma is one of the most commonly observed tumours, representing about 75% of brain tumours in the adult population. Generally, glioma treatment includes surgical resection followed by radiotherapy and chemotherapy. The current chemotherapy for glioma involves the use of temozolomide, doxorubicin, monoclonal antibodies, etc. however, the clinical outcomes in patients are not satisfactory. Primarily, the blood-brain barrier hinders these drugs from reaching the target leading to the recurrence of glioma post-surgery. In addition, these drugs are not target-specific and affect the healthy cells of the body. Therefore, glioma-targeted drug delivery is essential to reduce the rate of recurrence and treat the condition with more reliable alternatives. METHODS A literature search was conducted to understand glioma pathophysiology, its current therapeutic approaches for targeted delivery using databases like Pub Med, Web of Science, Scopus, and Google Scholar, etc. Results: This review gives an insight to challenges associated with current treatments, factors influencing drug delivery in glioma, and recent advancements in targeted drug delivery. CONCLUSION The promising results could be seen with nanotechnology-based approaches, like polymeric, lipidbased, and hybrid nanoparticles in the treatment of glioma. Biotechnological developments, such as carrier peptides and gene therapy, are future prospects in glioma therapy. Therefore, these targeted delivery systems will be beneficial in clinical practices for glioma treatment.
Collapse
Affiliation(s)
- Dhrumi Patel
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra - 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra - 400056, India
| | - Mayur C Yergeri
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra - 400056, India
| |
Collapse
|
33
|
Fluorescent bovine serum albumin-silver nanoclusters loaded with paclitaxel can traverse the blood-brain barrier to inhibit the migration of glioma. JOURNAL OF BIO-X RESEARCH 2020. [DOI: 10.1097/jbr.0000000000000082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
34
|
Wang T, Meng Z, Kang Z, Ding G, Zhao B, Han Z, Zheng Z, Wang C, Meng Q. Peptide Gene Delivery Vectors for Specific Transfection of Glioma Cells. ACS Biomater Sci Eng 2020; 6:6778-6789. [DOI: 10.1021/acsbiomaterials.0c01336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Taoran Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Zhao Meng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Ziyao Kang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Guihua Ding
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Baoquan Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Zhenbin Han
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Zhibing Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Chenhong Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Qingbin Meng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin 133002, China
| |
Collapse
|
35
|
Caprifico AE, Foot PJS, Polycarpou E, Calabrese G. Overcoming the Blood-Brain Barrier: Functionalised Chitosan Nanocarriers. Pharmaceutics 2020; 12:pharmaceutics12111013. [PMID: 33114020 PMCID: PMC7690755 DOI: 10.3390/pharmaceutics12111013] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/17/2020] [Accepted: 10/22/2020] [Indexed: 12/15/2022] Open
Abstract
The major impediment to the delivery of therapeutics to the brain is the presence of the blood-brain barrier (BBB). The BBB allows for the entrance of essential nutrients while excluding harmful substances, including most therapeutic agents; hence, brain disorders, especially tumours, are very difficult to treat. Chitosan is a well-researched polymer that offers advantageous biological and chemical properties, such as mucoadhesion and the ease of functionalisation. Chitosan-based nanocarriers (CsNCs) establish ionic interactions with the endothelial cells, facilitating the crossing of drugs through the BBB by adsorptive mediated transcytosis. This process is further enhanced by modifications of the structure of chitosan, owing to the presence of reactive amino and hydroxyl groups. Finally, by permanently binding ligands or molecules, such as antibodies or lipids, CsNCs have showed a boosted passage through the BBB, in both in vivo and in vitro studies which will be discussed in this review.
Collapse
|
36
|
Zhao L, Zhu J, Gong J, Song N, Wu S, Qiao W, Yang J, Zhu M, Zhao J. Polyethylenimine-based theranostic nanoplatform for glioma-targeting single-photon emission computed tomography imaging and anticancer drug delivery. J Nanobiotechnology 2020; 18:143. [PMID: 33054757 PMCID: PMC7557081 DOI: 10.1186/s12951-020-00705-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/07/2020] [Indexed: 12/22/2022] Open
Abstract
Background Glioma is the deadliest brain cancer in adults because the blood–brain-barrier (BBB) prevents the vast majority of therapeutic drugs from entering into the central nervous system. The development of BBB-penetrating drug delivery systems for glioma therapy still remains a great challenge. In this study, we aimed to design and develop a theranostic nanocomplex with enhanced BBB penetrability and tumor-targeting efficiency for glioma single-photon emission computed tomography (SPECT) imaging and anticancer drug delivery. Results This multifunctional nanocomplex was manufactured using branched polyethylenimine (PEI) as a template to sequentially conjugate with methoxypolyethylene glycol (mPEG), glioma-targeting peptide chlorotoxin (CTX), and diethylenetriaminepentaacetic acid (DTPA) for 99mTc radiolabeling on the surface of PEI. After the acetylation of the remaining PEI surface amines using acetic anhydride (Ac2O), the CTX-modified PEI (mPEI-CTX) was utilized as a carrier to load chemotherapeutic drug doxorubicin (DOX) in its interior cavity. The formed mPEI-CTX/DOX complex had excellent water dispersibility and released DOX in a sustainable and pH-dependent manner; furthermore, it showed targeting specificity and therapeutic effect of DOX toward glioma cells in vitro and in vivo (a subcutaneous tumor mouse model). Owing to the unique biological properties of CTX, the mPEI-CTX/DOX complex was able to cross the BBB and accumulate at the tumor site in an orthotopic rat glioma model. In addition, after efficient radiolabeling of PEI with 99mTc via DTPA, the 99mTc-labeled complex could help to visualize the drug accumulation in tumors of glioma-bearing mice and the drug delivery into the brains of rats through SPECT imaging. Conclusions These results indicate the potential of the developed PEI-based nanocomplex in facilitating glioma-targeting SPECT imaging and chemotherapy. ![]()
Collapse
Affiliation(s)
- Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Jingyi Zhu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, People's Republic of China
| | - Jiali Gong
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Ningning Song
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Shan Wu
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Wenli Qiao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Jiqin Yang
- Department of Nuclear Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China.
| | - Meilin Zhu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China.
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
37
|
Jnaidi R, Almeida AJ, Gonçalves LM. Solid Lipid Nanoparticles and Nanostructured Lipid Carriers as Smart Drug Delivery Systems in the Treatment of Glioblastoma Multiforme. Pharmaceutics 2020; 12:E860. [PMID: 32927610 PMCID: PMC7558650 DOI: 10.3390/pharmaceutics12090860] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant type of brain tumor. In fact, tumor recurrence usually appears a few months after surgical resection and chemotherapy, mainly due to many factors that make GBM treatment a real challenge, such as tumor location, heterogeneity, presence of the blood-brain barrier (BBB), and others. Solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) represent the most promising carriers for therapeutics delivery into the central nervous system (CNS) owing to their inherent ability to cross the BBB. In this review, we present the main challenges in GBM treatment, a description of SLNs and NLCs and their valuable role as drug carriers in GBM treatment, and finally, a detailed description of all modification strategies that aim to change composition of SLNs and NLCs to enhance treatment outcomes. This includes modification of SLNs and NLCs to improve crossing the BBB, reduced GBM cell resistance, target GBM cells selectively minimizing side effects, and modification strategies to enhance SLNs and NLCs nose-to-brain delivery. Finally, future perspectives on their use are also be discussed, to provide insight about all strategies with SLNs and NLCs formulation that could result in drug delivery systems for GBM treatment with highly effective theraputic and minimum undesirable effects.
Collapse
Affiliation(s)
| | | | - Lídia M. Gonçalves
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (R.J.); (A.J.A.)
| |
Collapse
|
38
|
Ebrahimi Z, Talaei S, Aghamiri S, Goradel NH, Jafarpour A, Negahdari B. Overcoming the blood-brain barrier in neurodegenerative disorders and brain tumours. IET Nanobiotechnol 2020; 14:441-448. [PMID: 32755952 PMCID: PMC8676526 DOI: 10.1049/iet-nbt.2019.0351] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/30/2020] [Accepted: 04/24/2020] [Indexed: 07/31/2023] Open
Abstract
Drug delivery is one of the major challenges in the treatment of central nervous system disorders. The brain needs to be protected from harmful agents, which are done by the capillary network, the so-called blood-brain barrier (BBB). This protective guard also prevents the delivery of therapeutic agents to the brain and limits the effectiveness of treatment. For this reason, various strategies have been explored by scientists for overcoming the BBB from disruption of the BBB to targeted delivery of nanoparticles (NPs) and cells and immunotherapy. In this review, different promising brain drug delivery strategies including disruption of tight junctions in the BBB, enhanced transcellular transport by peptide-based delivery, local delivery strategies, NP delivery, and cell-based delivery have been fully discussed.
Collapse
Affiliation(s)
- Zahra Ebrahimi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sam Talaei
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahin Aghamiri
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Jafarpour
- Students' Scientific Research Center, Virology Division, Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Wu Y, Lu Z, Li Y, Yang J, Zhang X. Surface Modification of Iron Oxide-Based Magnetic Nanoparticles for Cerebral Theranostics: Application and Prospection. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1441. [PMID: 32722002 PMCID: PMC7466388 DOI: 10.3390/nano10081441] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 12/22/2022]
Abstract
Combining diagnosis with therapy, magnetic iron oxide nanoparticles (INOPs) act as an important vehicle for drug delivery. However, poor biocompatibility of INOPs limits their application. To improve the shortcomings, various surface modifications have been developed, including small molecules coatings, polymers coatings, lipid coatings and lipopolymer coatings. These surface modifications facilitate iron nanoparticles to cross the blood-brain-barrier, which is essential for diagnosis and treatments of brain diseases. Here we focus on the characteristics of different coated INOPs and their application in brain disease, particularly gliomas, Alzheimer's disease (AD) and Parkinson's disease (PD). Moreover, we summarize the current progress and expect to provide help for future researches.
Collapse
Affiliation(s)
- Yanyue Wu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiguo Lu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Li
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Jun Yang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
40
|
Garcia-Fabiani MB, Ventosa M, Comba A, Candolfi M, Nicola Candia AJ, Alghamri MS, Kadiyala P, Carney S, Faisal SM, Schwendeman A, Moon JJ, Scheetz L, Lahann J, Mauser A, Lowenstein PR, Castro MG. Immunotherapy for gliomas: shedding light on progress in preclinical and clinical development. Expert Opin Investig Drugs 2020; 29:659-684. [PMID: 32400216 DOI: 10.1080/13543784.2020.1768528] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Gliomas are infiltrating brain tumors associated with high morbidity and mortality. Current standard of care includes radiation, chemotherapy, and surgical resection. Today, survival rates for malignant glioma patients remain dismal and unchanged for decades. The glioma microenvironment is highly immunosuppressive and consequently this has motivated the development of immunotherapies for counteracting this condition, enabling the immune cells within the tumor microenvironment to react against this tumor. AREAS COVERED The authors discuss immunotherapeutic strategies for glioma in phase-I/II clinical trials and illuminate their mechanisms of action, limitations, and key challenges. They also examine promising approaches under preclinical development. EXPERT OPINION In the last decade there has been an expansion in immune-mediated anti-cancer therapies. In the glioma field, sophisticated strategies have been successfully implemented in preclinical models. Unfortunately, clinical trials have not yet yielded consistent results for glioma patients. This could be attributed to our limited understanding of the complex immune cell infiltration and its interaction with the tumor cells, the selected time for treatment, the combination with other therapies and the route of administration of the agent. Applying these modalities to treat malignant glioma is challenging, but many new alternatives are emerging to by-pass these hurdles.
Collapse
Affiliation(s)
- Maria B Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Maria Ventosa
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires , Buenos Aires, Argentina
| | - Alejandro J Nicola Candia
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires , Buenos Aires, Argentina
| | - Mahmoud S Alghamri
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Stephen Carney
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Cancer Biology Graduate Program, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Syed M Faisal
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA.,Department of Biomedical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Lindsay Scheetz
- Department of Pharmaceutical Sciences, University of Michigan , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| | - Joerg Lahann
- Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA.,Department of Chemical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Ava Mauser
- Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA.,Department of Chemical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| |
Collapse
|
41
|
Lübtow MM, Oerter S, Quader S, Jeanclos E, Cubukova A, Krafft M, Haider MS, Schulte C, Meier L, Rist M, Sampetrean O, Kinoh H, Gohla A, Kataoka K, Appelt-Menzel A, Luxenhofer R. In Vitro Blood–Brain Barrier Permeability and Cytotoxicity of an Atorvastatin-Loaded Nanoformulation Against Glioblastoma in 2D and 3D Models. Mol Pharm 2020; 17:1835-1847. [DOI: 10.1021/acs.molpharmaceut.9b01117] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Michael M. Lübtow
- Functional Polymer Materials, Chair for Advanced Materials Synthesis, Department of Chemistry and Pharmacy and Bavarian Polymer Institute, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Sabrina Oerter
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Sabina Quader
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-Ku, Kawasaki-Shi 210-0821, Japan
| | - Elisabeth Jeanclos
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Straße 9, 97078 Würzburg, Germany
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
| | - Alevtina Cubukova
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies TLC-RT, Röntgenring 11, 97070 Würzburg, Germany
| | - Marion Krafft
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Malik Salman Haider
- Functional Polymer Materials, Chair for Advanced Materials Synthesis, Department of Chemistry and Pharmacy and Bavarian Polymer Institute, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Clemens Schulte
- Functional Polymer Materials, Chair for Advanced Materials Synthesis, Department of Chemistry and Pharmacy and Bavarian Polymer Institute, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Laura Meier
- Functional Polymer Materials, Chair for Advanced Materials Synthesis, Department of Chemistry and Pharmacy and Bavarian Polymer Institute, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Maximilian Rist
- Functional Polymer Materials, Chair for Advanced Materials Synthesis, Department of Chemistry and Pharmacy and Bavarian Polymer Institute, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Oltea Sampetrean
- Institute for Advanced Medical Research (IAMR), Division of Gene Regulation, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiroaki Kinoh
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-Ku, Kawasaki-Shi 210-0821, Japan
| | - Antje Gohla
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Straße 9, 97078 Würzburg, Germany
| | - Kazunori Kataoka
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-Ku, Kawasaki-Shi 210-0821, Japan
- Policy Alternatives Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Antje Appelt-Menzel
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies TLC-RT, Röntgenring 11, 97070 Würzburg, Germany
| | - Robert Luxenhofer
- Functional Polymer Materials, Chair for Advanced Materials Synthesis, Department of Chemistry and Pharmacy and Bavarian Polymer Institute, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
- Soft Matter Chemistry, Department of Chemistry, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
42
|
Wang D, Wang C, Wang L, Chen Y. A comprehensive review in improving delivery of small-molecule chemotherapeutic agents overcoming the blood-brain/brain tumor barriers for glioblastoma treatment. Drug Deliv 2020; 26:551-565. [PMID: 31928355 PMCID: PMC6534214 DOI: 10.1080/10717544.2019.1616235] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and lethal primary brain tumor which is highly resistant to conventional radiotherapy and chemotherapy, and cannot be effectively controlled by surgical resection. Due to inevitable recurrence of GBM, it remains essentially incurable with a median overall survival of less than 18 months after diagnosis. A great challenge in current therapies lies in the abrogated delivery of most of the chemotherapeutic agents to the tumor location in the presence of blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB). These protective barriers serve as a selectively permeable hurdle reducing the efficacy of anti-tumor drugs in GBM therapy. This work systematically gives a comprehensive review on: (i) the characteristics of the BBB and the BBTB, (ii) the influence of BBB/BBTB on drug delivery and the screening strategy of small-molecule chemotherapeutic agents with promising BBB/BBTB-permeable potential, (iii) the strategies to overcome the BBB/BBTB as well as the techniques which can lead to transient BBB/BBTB opening or disruption allowing for improving BBB/BBTB-penetration of drugs. It is hoped that this review provide practical guidance for the future development of small BBB/BBTB-permeable agents against GBM as well as approaches enhancing drug delivery across the BBB/BBTB to GBM.
Collapse
Affiliation(s)
- Da Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Chao Wang
- Department of Chemistry, Yale University, New Haven, CT, USA
| | - Liang Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yue Chen
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
43
|
Ghali GZ, Ghali MGZ. β adrenergic receptor modulated signaling in glioma models: promoting β adrenergic receptor-β arrestin scaffold-mediated activation of extracellular-regulated kinase 1/2 may prove to be a panacea in the treatment of intracranial and spinal malignancy and extra-neuraxial carcinoma. Mol Biol Rep 2020; 47:4631-4650. [PMID: 32303958 PMCID: PMC7165076 DOI: 10.1007/s11033-020-05427-1] [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: 02/25/2020] [Accepted: 04/03/2020] [Indexed: 12/03/2022]
Abstract
Neoplastically transformed astrocytes express functionally active cell surface β adrenergic receptors (βARs). Treatment of glioma models in vitro and in vivo with β adrenergic agonists variably amplifies or attenuates cellular proliferation. In the majority of in vivo models, β adrenergic agonists generally reduce cellular proliferation. However, treatment with β adrenergic agonists consistently reduces tumor cell invasive potential, angiogenesis, and metastasis. β adrenergic agonists induced decreases of invasive potential are chiefly mediated through reductions in the expression of matrix metalloproteinases types 2 and 9. Treatment with β adrenergic agonists also clearly reduce tumoral neoangiogenesis, which may represent a putatively useful mechanism to adjuvantly amplify the effects of bevacizumab. Bevacizumab is a monoclonal antibody targeting the vascular endothelial growth factor receptor. We may accordingly designate βagonists to represent an enhancer of bevacizumab. The antiangiogenic effects of β adrenergic agonists may thus effectively render an otherwise borderline effective therapy to generate significant enhancement in clinical outcomes. β adrenergic agonists upregulate expression of the major histocompatibility class II DR alpha gene, effectively potentiating the immunogenicity of tumor cells to tumor surveillance mechanisms. Authors have also demonstrated crossmodal modulation of signaling events downstream from the β adrenergic cell surface receptor and microtubular polymerization and depolymerization. Complex effects and desensitization mechanisms of the β adrenergic signaling may putatively represent promising therapeutic targets. Constant stimulation of the β adrenergic receptor induces its phosphorylation by β adrenergic receptor kinase (βARK), rendering it a suitable substrate for alternate binding by β arrestins 1 or 2. The binding of a β arrestin to βARK phosphorylated βAR promotes receptor mediated internalization and downregulation of cell surface receptor and contemporaneously generates a cell surface scaffold at the βAR. The scaffold mediated activation of extracellular regulated kinase 1/2, compared with protein kinase A mediated activation, preferentially favors cytosolic retention of ERK1/2 and blunting of nuclear translocation and ensuant pro-transcriptional activity. Thus, βAR desensitization and consequent scaffold assembly effectively retains the cytosolic homeostatic functions of ERK1/2 while inhibiting its pro-proliferative effects. We suggest these mechanisms specifically will prove quite promising in developing primary and adjuvant therapies mitigating glioma growth, angiogenesis, invasive potential, and angiogenesis. We suggest generating compounds and targeted mutations of the β adrenergic receptor favoring β arrestin binding and scaffold facilitated activation of ERK1/2 may hold potential promise and therapeutic benefit in adjuvantly treating most or all cancers. We hope our discussion will generate fruitful research endeavors seeking to exploit these mechanisms.
Collapse
Affiliation(s)
- George Zaki Ghali
- United States Environmental Protection Agency, Arlington, VA, USA.,Emeritus Professor, Department of Toxicology, Purdue University, West Lafayette, IN, USA
| | - Michael George Zaki Ghali
- Department of Neurological Surgery, University of California, San Francisco, 505 Parnassus Avenue, Box-0112, San Francisco, CA, 94143, USA. .,Department of Neurological Surgery, Karolinska Institutet, Nobels väg 6, Solna and Alfred Nobels Allé 8, Huddinge, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
44
|
LRP-1 Mediated Endocytosis of EFE Across the Blood–Brain Barrier; Protein–Protein Interaction and Molecular Dynamics Analysis. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-020-10065-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
45
|
Li J, Zhao J, Tan T, Liu M, Zeng Z, Zeng Y, Zhang L, Fu C, Chen D, Xie T. Nanoparticle Drug Delivery System for Glioma and Its Efficacy Improvement Strategies: A Comprehensive Review. Int J Nanomedicine 2020; 15:2563-2582. [PMID: 32368041 PMCID: PMC7173867 DOI: 10.2147/ijn.s243223] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/21/2020] [Indexed: 12/22/2022] Open
Abstract
Gliomas are the most common tumor of the central nervous system. However, the presence of the brain barrier blocks the effective delivery of drugs and leads to the treatment failure of various drugs. The development of a nanoparticle drug delivery system (NDDS) can solve this problem. In this review, we summarized the brain barrier (including blood-brain barrier (BBB), blood-brain tumor barriers (BBTB), brain-cerebrospinal fluid barrier (BCB), and nose-to-brain barrier), NDDS of glioma (such as passive targeting systems, active targeting systems, and environmental responsive targeting systems), and NDDS efficacy improvement strategies and deficiencies. The research prospect of drug-targeted delivery systems for glioma is also discussed.
Collapse
Affiliation(s)
- Jie Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Jiaqian Zhao
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
- College of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Tiantian Tan
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Mengmeng Liu
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Zhaowu Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Yiying Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Lele Zhang
- School of Medicine, Chengdu University, Chengdu, People’s Republic of China
| | - Chaomei Fu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Dajing Chen
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Tian Xie
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
46
|
Altshuler DB, Kadiyala P, Núñez FJ, Núñez FM, Carney S, Alghamri MS, Garcia-Fabiani MB, Asad AS, Nicola Candia AJ, Candolfi M, Lahann J, Moon JJ, Schwendeman A, Lowenstein PR, Castro MG. Prospects of biological and synthetic pharmacotherapies for glioblastoma. Expert Opin Biol Ther 2020; 20:305-317. [PMID: 31959027 PMCID: PMC7059118 DOI: 10.1080/14712598.2020.1713085] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 01/06/2020] [Indexed: 01/05/2023]
Abstract
Introduction: The field of neuro-oncology has experienced significant advances in recent years. More is known now about the molecular and genetic characteristics of glioma than ever before. This knowledge leads to the understanding of glioma biology and pathogenesis, guiding the development of targeted therapeutics and clinical trials. The goal of this review is to describe the state of basic, translational, and clinical research as it pertains to biological and synthetic pharmacotherapy for gliomas.Areas covered: Challenges remain in designing accurate preclinical models and identifying patients that are likely to respond to a particular targeted therapy. Preclinical models for therapeutic assessment are critical to identify the most promising treatment approaches.Expert opinion: Despite promising new therapeutics, there have been no significant breakthroughs in glioma treatment and patient outcomes. Thus, there is an urgent need to better understand the mechanisms of treatment resistance and to design effective clinical trials.
Collapse
Affiliation(s)
- David B. Altshuler
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Felipe J. Núñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Fernando M. Núñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Stephen Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria B. Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Antonela S. Asad
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires. Argentina
| | - Alejandro J. Nicola Candia
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires. Argentina
| | - Marianela Candolfi
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires. Argentina
| | - Joerg Lahann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
47
|
Oberacker E, Kuehne A, Oezerdem C, Nadobny J, Weihrauch M, Beck M, Zschaeck S, Diesch C, Eigentler TW, Waiczies H, Ghadjar P, Wust P, Winter L, Niendorf T. Radiofrequency applicator concepts for thermal magnetic resonance of brain tumors at 297 MHz (7.0 Tesla). Int J Hyperthermia 2020; 37:549-563. [PMID: 32484019 PMCID: PMC8352381 DOI: 10.1080/02656736.2020.1761462] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 04/02/2020] [Accepted: 04/17/2020] [Indexed: 12/28/2022] Open
Abstract
Purpose: Thermal intervention is a potent sensitizer of cells to chemo- and radiotherapy in cancer treatment. Glioblastoma multiforme (GBM) is a potential clinical target, given the cancer's aggressive nature and resistance to current treatment options. The annular phased array (APA) technique employing electromagnetic waves in the radiofrequency (RF) range allows for localized temperature increase in deep seated target volumes (TVs). Reports on clinical applications of the APA technique in the brain are still missing. Ultrahigh field magnetic resonance (MR) employs higher frequencies than conventional MR and has potential to provide focal temperature manipulation, high resolution imaging and noninvasive temperature monitoring using an integrated RF applicator (ThermalMR). This work examines the applicability of RF applicator concepts for ThermalMR of brain tumors at 297 MHz (7.0 Tesla).Methods: Electromagnetic field (EMF) simulations are performed for clinically realistic data based on GBM patients. Two algorithms are used for specific RF energy absorption rate based thermal intervention planning for small and large TVs in the brain, aiming at maximum RF power deposition or RF power uniformity in the TV for 10 RF applicator designs.Results: For both TVs , the power optimization outperformed the uniformity optimization. The best results for the small TV are obtained for the 16 element interleaved RF applicator using an elliptical antenna arrangement with water bolus. The two row elliptical RF applicator yielded the best result for the large TV.Discussion: This work investigates the capacity of ThermalMR to achieve targeted thermal interventions in model systems resembling human brain tissue and brain tumors.
Collapse
Affiliation(s)
- Eva Oberacker
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Physics, Faculty of Mathematics and Natural Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Celal Oezerdem
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Joint Cooperation Between the Charité Medical Faculty and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jacek Nadobny
- Clinic for Radiation Oncology, Charité Universitätsmedizin, Berlin, Germany
| | - Mirko Weihrauch
- Clinic for Radiation Oncology, Charité Universitätsmedizin, Berlin, Germany
| | - Marcus Beck
- Clinic for Radiation Oncology, Charité Universitätsmedizin, Berlin, Germany
| | - Sebastian Zschaeck
- Clinic for Radiation Oncology, Charité Universitätsmedizin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Cecilia Diesch
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Thomas Wilhelm Eigentler
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Chair of Medical Engineering, Technische Universität Berlin, Berlin, Germany
| | | | - Pirus Ghadjar
- Clinic for Radiation Oncology, Charité Universitätsmedizin, Berlin, Germany
| | - Peter Wust
- Clinic for Radiation Oncology, Charité Universitätsmedizin, Berlin, Germany
| | - Lukas Winter
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Physikalisch Technische Bundesanstalt, Braunschweig, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- MRI.TOOLS GmbH, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Joint Cooperation Between the Charité Medical Faculty and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
48
|
Ghosh S, Lalani R, Patel V, Bhowmick S, Misra A. Surface engineered liposomal delivery of therapeutics across the blood brain barrier: recent advances, challenges and opportunities. Expert Opin Drug Deliv 2019; 16:1287-1311. [DOI: 10.1080/17425247.2019.1676721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Saikat Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Rohan Lalani
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Vivek Patel
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Subhas Bhowmick
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Ambikanandan Misra
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
49
|
Challenges on the processing of plant-based neuronutraceuticals and functional foods with emerging technologies: Extraction, encapsulation and therapeutic applications. Trends Food Sci Technol 2019. [DOI: 10.1016/j.tifs.2019.07.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
50
|
Kobrlova T, Korabecny J, Soukup O. Current approaches to enhancing oxime reactivator delivery into the brain. Toxicology 2019; 423:75-83. [PMID: 31112674 DOI: 10.1016/j.tox.2019.05.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/17/2019] [Accepted: 05/13/2019] [Indexed: 01/21/2023]
Abstract
The misuse of organophosphate compounds still represents a current threat worldwide. Treatment of poisoning with organophosphates (OPs) remains unsatisfactorily resolved despite the extensive investment in research in academia. There are no universal, effective and centrally-active acetylcholinesterase (AChE) reactivators to countermeasure OP intoxication. One major obstacle is to overcome the blood-brain barrier (BBB). The central compartment is readily accessible by the OPs which are lipophilic bullets that can easily cross the BBB, whereas first-line therapeutics, namely oxime-based AChE reactivators and atropine, do not cross or do so rather slowly. The limitation of oxime-based AChE reactivators can be ascribed to their chemical nature, bearing a positive charge which is essential either for their AChE affinity or their reactivating potency. The aim of this article is to review the methods for targeting the brain by oxime reactivators that have been developed so far. Approaches using prodrugs, lipophilicity enhancement, or sugar-based oximes have been rather unsuccessful. However, other strategies have been more promising, such as the use of nanoparticles or co-administration of the reactivator with efflux transporter inhibitors. Encouraging results have also been associated with intranasal delivery, but research in this field is still at the beginning. Further research of auspicious approaches is inevitable.
Collapse
Affiliation(s)
- Tereza Kobrlova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic; Biomedical Research Centre, University Hospital, Sokolska 581, 500 05, Hradec Kralove, Czech Republic.
| | - Jan Korabecny
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic; Biomedical Research Centre, University Hospital, Sokolska 581, 500 05, Hradec Kralove, Czech Republic.
| | - Ondrej Soukup
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic; Biomedical Research Centre, University Hospital, Sokolska 581, 500 05, Hradec Kralove, Czech Republic.
| |
Collapse
|