1
|
Valerius AR, Webb LM, Thomsen A, Lehrer EJ, Breen WG, Campian JL, Riviere-Cazaux C, Burns TC, Sener U. Review of Novel Surgical, Radiation, and Systemic Therapies and Clinical Trials in Glioblastoma. Int J Mol Sci 2024; 25:10570. [PMID: 39408897 PMCID: PMC11477105 DOI: 10.3390/ijms251910570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor in adults. Despite an established standard of care including surgical resection, radiation therapy, and chemotherapy, GBM unfortunately is associated with a dismal prognosis. Therefore, researchers are extensively evaluating avenues to expand GBM therapy and improve outcomes in patients with GBM. In this review, we provide a broad overview of novel GBM therapies that have recently completed or are actively undergoing study in clinical trials. These therapies expand across medical, surgical, and radiation clinical trials. We additionally review methods for improving clinical trial design in GBM.
Collapse
Affiliation(s)
| | - Lauren M. Webb
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA (U.S.)
| | - Anna Thomsen
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA (U.S.)
| | - Eric J. Lehrer
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - William G. Breen
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jian L. Campian
- Department of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Terry C. Burns
- Department of Neurosurgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Ugur Sener
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA (U.S.)
- Department of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
2
|
Watson SS, Duc B, Kang Z, de Tonnac A, Eling N, Font L, Whitmarsh T, Massara M, Bodenmiller B, Hausser J, Joyce JA. Microenvironmental reorganization in brain tumors following radiotherapy and recurrence revealed by hyperplexed immunofluorescence imaging. Nat Commun 2024; 15:3226. [PMID: 38622132 PMCID: PMC11018859 DOI: 10.1038/s41467-024-47185-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
The tumor microenvironment plays a crucial role in determining response to treatment. This involves a series of interconnected changes in the cellular landscape, spatial organization, and extracellular matrix composition. However, assessing these alterations simultaneously is challenging from a spatial perspective, due to the limitations of current high-dimensional imaging techniques and the extent of intratumoral heterogeneity over large lesion areas. In this study, we introduce a spatial proteomic workflow termed Hyperplexed Immunofluorescence Imaging (HIFI) that overcomes these limitations. HIFI allows for the simultaneous analysis of > 45 markers in fragile tissue sections at high magnification, using a cost-effective high-throughput workflow. We integrate HIFI with machine learning feature detection, graph-based network analysis, and cluster-based neighborhood analysis to analyze the microenvironment response to radiation therapy in a preclinical model of glioblastoma, and compare this response to a mouse model of breast-to-brain metastasis. Here we show that glioblastomas undergo extensive spatial reorganization of immune cell populations and structural architecture in response to treatment, while brain metastases show no comparable reorganization. Our integrated spatial analyses reveal highly divergent responses to radiation therapy between brain tumor models, despite equivalent radiotherapy benefit.
Collapse
Affiliation(s)
- Spencer S Watson
- Department of Oncology, University of Lausanne, Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, 1011, Switzerland.
- L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, 1011, Switzerland.
| | - Benoit Duc
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, 1011, Switzerland
- L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, 1011, Switzerland
| | - Ziqi Kang
- Department of Cellular and Molecular Biology, Karolinska Institutet and SciLifeLab, Stockholm, Sweden
| | - Axel de Tonnac
- Department of Cellular and Molecular Biology, Karolinska Institutet and SciLifeLab, Stockholm, Sweden
| | - Nils Eling
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute for Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Laure Font
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- École Polytechnique Fédérale Lausanne, Lausanne, Switzerland
| | - Tristan Whitmarsh
- Machine Intelligence Laboratory, Department of Engineering, University of Cambridge, Cambridge, UK
| | - Matteo Massara
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, 1011, Switzerland
- L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, 1011, Switzerland
| | - Bernd Bodenmiller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute for Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Jean Hausser
- Department of Cellular and Molecular Biology, Karolinska Institutet and SciLifeLab, Stockholm, Sweden
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, 1011, Switzerland.
- L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, 1011, Switzerland.
- Cancer Research UK, Cancer Grand Challenges iMAXT Consortium, University of Cambridge, Cambridge, UK.
| |
Collapse
|
3
|
Karabuga M, Erdogan S, Filikci K, Hazıroglu R, Tuncel M, Cengiz M. Evaluation of efficacy of tumor-specific nanoliposomal radiosensitizer in radiotherapy. J Drug Deliv Sci Technol 2023; 86:104586. [DOI: 10.1016/j.jddst.2023.104586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
|
4
|
Liang X, Yang Y, Huang C, Ye Z, Lai W, Luo J, Li X, Yi X, Fan JB, Wang Y, Wang Y. cRGD-targeted heparin nanoparticles for effective dual drug treatment of cisplatin-resistant ovarian cancer. J Control Release 2023; 356:691-701. [PMID: 36933699 DOI: 10.1016/j.jconrel.2023.03.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/23/2023] [Accepted: 03/10/2023] [Indexed: 03/20/2023]
Abstract
Resistance to the chemotherapeutic agent cisplatin (DDP) is the primary reason for invalid chemotherapy of ovarian cancer. Given the complex mechanisms underlying chemo-resistance, the design of combination therapies based on blocking multiple mechanisms is a rationale to synergistically elevate therapeutic effect for effectively overcoming cancer chemo-resistance. Herein, we demonstrated a multifunctional nanoparticle (DDP-Ola@HR), which could simultaneously co-deliver DDP and Olaparib (Ola, DNA damage repair inhibitor) using targeted ligand cRGD peptide modified with heparin (HR) as nanocarrier, enabling the concurrent tackling of multiple resistance mechanisms to effectively inhibit the growth and metastasis of DDP-resistant ovarian cancer. In combination strategy, heparin could suppress the function of multidrug resistance-associated protein 2 (MRP2) and P-glycoprotein (P-gp) to promote the intracellular accumulation of DDP and Ola by specifically binding with heparanase (HPSE) to down-regulate PI3K/AKT/mTOR signaling pathway, and simultaneously served as a carrier combined with Ola to synergistically enhance the anti-proliferation ability of DDP for resistant ovarian cancer, thus achieving great therapeutic efficacy. Our DDP-Ola@HR could provide a simple and multifunctional combination strategy to trigger an anticipated cascading effect, thus effectively overcoming the chemo-resistance of ovarian cancer.
Collapse
Affiliation(s)
- Xiaomei Liang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Yulu Yang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Chuanqing Huang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Zhibin Ye
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Wujiang Lai
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Jiamao Luo
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Xiaoxuan Li
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Xiao Yi
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Jun-Bing Fan
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China.
| | - Ying Wang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China.
| | - Yifeng Wang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China.
| |
Collapse
|
5
|
Szklener K, Mazurek M, Wieteska M, Wacławska M, Bilski M, Mańdziuk S. New Directions in the Therapy of Glioblastoma. Cancers (Basel) 2022; 14:5377. [PMID: 36358795 PMCID: PMC9655599 DOI: 10.3390/cancers14215377] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma is the most common histologic type of all gliomas and contributes to 57.3% of all cases. Despite the standard management based on surgical resection and radiotherapy, it is related to poor outcome, with a 5-year relative survival rate below 6.9%. In order to improve the overall outcome for patients, the new therapeutic strategies are needed. Herein, we describe the current state of knowledge on novel targeted therapies in glioblastoma. Based on recent studies, we compared treatment efficacy measured by overall survival and progression-free survival in patients treated with selected potential antitumor drugs. The results of the application of the analyzed inhibitors are highly variable despite the encouraging conclusions of previous preclinical studies. This paper focused on drugs that target major glioblastoma kinases. As far, the results of some BRAF inhibitors are favorable. Vemurafenib demonstrated a long-term efficacy in clinical trials while the combination of dabrafenib and trametinib improves PFS compared with both vemurafenib and dabrafenib alone. There is no evidence that any MEK inhibitor is effective in monotherapy. According to the current state of knowledge, BRAF and MEK inhibition are more advantageous than BRAF inhibitor monotherapy. Moreover, mTOR inhibitors (especially paxalisib) may be considered a particularly important group. Everolimus demonstrated a partial response in a significant proportion of patients when combined with bevacizumab, however its actual role in the treatment is unclear. Neither nintedanib nor pemigatinib were efficient in treatment of GBM. Among the anti-VEGF drugs, bevacizumab monotherapy was a well-tolerated option, significantly associated with anti-GBM activity in patients with recurrent GBM. The efficacy of aflibercept and pazopanib in monotherapy has not been demonstrated. Apatinib has been proven to be effective and tolerable by a single clinical trial, but more research is needed. Lenvatinib is under trial. Finally, promising results from a study with regorafenib may be confirmed by the ongoing randomized AGILE trial. The studies conducted so far have provided a relatively wide range of drugs, which are at least well tolerated and demonstrated some efficacy in the randomized clinical trials. The comprehensive understanding of the molecular biology of gliomas promises to further improve the treatment outcomes of patients.
Collapse
Affiliation(s)
- Katarzyna Szklener
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 8 Jaczewski Street, 20-090 Lublin, Poland
| | - Marek Mazurek
- Department of Neurosurgery, Medical University of Lublin, 20-090 Lublin, Poland
| | - Małgorzata Wieteska
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 8 Jaczewski Street, 20-090 Lublin, Poland
| | - Monika Wacławska
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 8 Jaczewski Street, 20-090 Lublin, Poland
| | - Mateusz Bilski
- Department of Radiotherapy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Sławomir Mańdziuk
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 8 Jaczewski Street, 20-090 Lublin, Poland
| |
Collapse
|
6
|
Ruiz-Molina D, Mao X, Alfonso-Triguero P, Lorenzo J, Bruna J, Yuste VJ, Candiota AP, Novio F. Advances in Preclinical/Clinical Glioblastoma Treatment: Can Nanoparticles Be of Help? Cancers (Basel) 2022; 14:4960. [PMID: 36230883 PMCID: PMC9563739 DOI: 10.3390/cancers14194960] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma multiforme (GB) is the most aggressive and frequent primary malignant tumor in the central nervous system (CNS), with unsatisfactory and challenging treatment nowadays. Current standard of care includes surgical resection followed by chemotherapy and radiotherapy. However, these treatments do not much improve the overall survival of GB patients, which is still below two years (the 5-year survival rate is below 7%). Despite various approaches having been followed to increase the release of anticancer drugs into the brain, few of them demonstrated a significant success, as the blood brain barrier (BBB) still restricts its uptake, thus limiting the therapeutic options. Therefore, enormous efforts are being devoted to the development of novel nanomedicines with the ability to cross the BBB and specifically target the cancer cells. In this context, the use of nanoparticles represents a promising non-invasive route, allowing to evade BBB and reducing systemic concentration of drugs and, hence, side effects. In this review, we revise with a critical view the different families of nanoparticles and approaches followed so far with this aim.
Collapse
Affiliation(s)
- Daniel Ruiz-Molina
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Xiaoman Mao
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Paula Alfonso-Triguero
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Julia Lorenzo
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Jordi Bruna
- Neuro-Oncology Unit, Bellvitge University Hospital-ICO (IDIBELL), Avinguda de la Gran Via de l’Hospitalet, 199-203, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Victor J. Yuste
- Instituto de Neurociencias. Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Campus UAB, 08193 Cerdanyola del Vallès, Spain
| | - Ana Paula Candiota
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193 Cerdanyola del Vallès, Spain
| | - Fernando Novio
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Departament de Química, Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193 Cerdanyola del Vallès, Spain
| |
Collapse
|
7
|
Zhang J, Zhang T, Gao J. Biocompatible Iron Oxide Nanoparticles for Targeted Cancer Gene Therapy: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12193323. [PMID: 36234452 PMCID: PMC9565336 DOI: 10.3390/nano12193323] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 05/14/2023]
Abstract
In recent years, gene therapy has made remarkable achievements in tumor treatment. In a successfully cancer gene therapy, a smart gene delivery system is necessary for both protecting the therapeutic genes in circulation and enabling high gene expression in tumor sites. Magnetic iron oxide nanoparticles (IONPs) have demonstrated their bright promise for highly efficient gene delivery target to tumor tissues, partly due to their good biocompatibility, magnetic responsiveness, and extensive functional surface modification. In this review, the latest progress in targeting cancer gene therapy is introduced, and the unique properties of IONPs contributing to the efficient delivery of therapeutic genes are summarized with detailed examples. Furthermore, the diagnosis potentials and synergistic tumor treatment capacity of IONPs are highlighted. In addition, aiming at potential risks during the gene delivery process, several strategies to improve the efficiency or reduce the potential risks of using IONPs for cancer gene therapy are introduced and addressed. The strategies and applications summarized in this review provide a general understanding for the potential applications of IONPs in cancer gene therapy.
Collapse
Affiliation(s)
- Jinsong Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Correspondence: (T.Z.); (J.G.)
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Correspondence: (T.Z.); (J.G.)
| |
Collapse
|
8
|
Teng XQ, Qu J, Li GH, Zhuang HH, Qu Q. Small Interfering RNA for Gliomas Treatment: Overcoming Hurdles in Delivery. Front Cell Dev Biol 2022; 10:824299. [PMID: 35874843 PMCID: PMC9304887 DOI: 10.3389/fcell.2022.824299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Gliomas are central nervous system tumors originating from glial cells, whose incidence and mortality rise in coming years. The current treatment of gliomas is surgery combined with chemotherapy or radiotherapy. However, developing therapeutic resistance is one of the significant challenges. Recent research suggested that small interfering RNA (siRNA) has excellent potential as a therapeutic to silence genes that are significantly involved in the manipulation of gliomas’ malignant phenotypes, including proliferation, invasion, metastasis, therapy resistance, and immune escape. However, it is challenging to deliver the naked siRNA to the action site in the cells of target tissues. Therefore, it is urgent to develop delivery strategies to transport siRNA to achieve the optimal silencing effect of the target gene. However, there is no systematic discussion about siRNAs’ clinical potential and delivery strategies in gliomas. This review mainly discusses siRNAs’ delivery strategies, especially nanotechnology-based delivery systems, as a potential glioma therapy. Moreover, we envisage the future orientation and challenges in translating these findings into clinical applications.
Collapse
Affiliation(s)
- Xin-Qi Teng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jian Qu
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Guo-Hua Li
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Hai-Hui Zhuang
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Qiang Qu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Qiang Qu,
| |
Collapse
|
9
|
Włodarczyk A, Gorgoń S, Radoń A, Bajdak-Rusinek K. Magnetite Nanoparticles in Magnetic Hyperthermia and Cancer Therapies: Challenges and Perspectives. NANOMATERIALS 2022; 12:nano12111807. [PMID: 35683663 PMCID: PMC9182445 DOI: 10.3390/nano12111807] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 12/12/2022]
Abstract
Until now, strategies used to treat cancer are imperfect, and this generates the need to search for better and safer solutions. The biggest issue is the lack of selective interaction with neoplastic cells, which is associated with occurrence of side effects and significantly reduces the effectiveness of therapies. The use of nanoparticles in cancer can counteract these problems. One of the most promising nanoparticles is magnetite. Implementation of this nanoparticle can improve various treatment methods such as hyperthermia, targeted drug delivery, cancer genotherapy, and protein therapy. In the first case, its feature makes magnetite useful in magnetic hyperthermia. Interaction of magnetite with the altered magnetic field generates heat. This process results in raised temperature only in a desired part of a patient body. In other therapies, magnetite-based nanoparticles could serve as a carrier for various types of therapeutic load. The magnetic field would direct the drug-related magnetite nanoparticles to the pathological site. Therefore, this material can be used in protein and gene therapy or drug delivery. Since the magnetite nanoparticle can be used in various types of cancer treatment, they are extensively studied. Herein, we summarize the latest finding on the applicability of the magnetite nanoparticles, also addressing the most critical problems faced by smart nanomedicine in oncological therapies.
Collapse
Affiliation(s)
- Agnieszka Włodarczyk
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland;
| | - Szymon Gorgoń
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, 901 87 Umeå, Sweden;
| | - Adrian Radoń
- Łukasiewicz Research Network—Institute of Non-Ferrous Metals, Sowinskiego 5 St., 44-100 Gliwice, Poland;
| | - Karolina Bajdak-Rusinek
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland;
- Correspondence: ; Tel.: +48-32-208-8382
| |
Collapse
|
10
|
Lin G, Huang J, Zhang M, Chen S, Zhang M. Chitosan-Crosslinked Low Molecular Weight PEI-Conjugated Iron Oxide Nanoparticle for Safe and Effective DNA Delivery to Breast Cancer Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:584. [PMID: 35214917 PMCID: PMC8876741 DOI: 10.3390/nano12040584] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/30/2022] [Accepted: 02/04/2022] [Indexed: 02/06/2023]
Abstract
Breast cancer has attracted tremendous research interest in treatment development as one of the major threats to public health. The use of non-viral carriers for therapeutic DNA delivery has shown promise in treating various cancer types, including breast cancer, due to their high DNA loading capacity, high cell transfection efficiency, and design versatility. However, cytotoxicity and large sizes of non-viral DNA carriers often raise safety concerns and hinder their applications in the clinic. Here we report the development of a novel nanoparticle formulation (termed NP-Chi-xPEI) that can safely and effectively deliver DNA into breast cancer cells for successful transfection. The nanoparticle is composed of an iron oxide core coated with low molecular weight (800 Da) polyethyleneimine crosslinked with chitosan via biodegradable disulfide bonds. The NP-Chi-xPEI can condense DNA into a small nanoparticle with the overall size of less than 100 nm and offer full DNA protection. Its biodegradable coating of small-molecular weight xPEI and mildly positive surface charge confer extra biocompatibility. NP-Chi-xPEI-mediated DNA delivery was shown to achieve high transfection efficiency across multiple breast cancer cell lines with significantly lower cytotoxicity as compared to the commercial transfection agent Lipofectamine 3000. With demonstrated favorable physicochemical properties and functionality, NP-Chi-xPEI may serve as a reliable vehicle to deliver DNA to breast cancer cells.
Collapse
Affiliation(s)
| | | | | | | | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA; (G.L.); (J.H.); (M.Z.); (S.C.)
| |
Collapse
|
11
|
Li J, Yu X, Shi X, Shen M. Cancer nanomedicine based on polyethylenimine-mediated multifunctional nanosystems. PROGRESS IN MATERIALS SCIENCE 2022; 124:100871. [DOI: 10.1016/j.pmatsci.2021.100871] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
Lehrer EJ, Ruiz-Garcia H, Nehlsen AD, Sindhu KK, Estrada RS, Borst GR, Sheehan JP, Quinones-Hinojosa A, Trifiletti DM. Preoperative Stereotactic Radiosurgery for Glioblastoma. BIOLOGY 2022; 11:194. [PMID: 35205059 PMCID: PMC8869151 DOI: 10.3390/biology11020194] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/16/2022]
Abstract
Glioblastoma is a devastating primary brain tumor with a median overall survival of approximately 15 months despite the use of optimal modern therapy. While GBM has been studied for decades, modern therapies have allowed for a reduction in treatment-related toxicities, while the prognosis has largely been unchanged. Adjuvant stereotactic radiosurgery (SRS) was previously studied in GBM; however, the results were disappointing. SRS is a highly conformal radiation technique that permits the delivery of high doses of ionizing radiation in 1-5 sessions while largely sparing surrounding healthy tissues. Furthermore, studies have shown that the delivery of ablative doses of ionizing radiation within the central nervous system is associated with enhanced anti-tumor immunity. While SRS is commonly used in the definitive and adjuvant settings for other CNS malignancies, its role in the preoperative setting has become a topic of great interest due to the potential for reduced treatment volumes due to the treatment of an intact tumor, and a lower risk of nodular leptomeningeal disease and radiation necrosis. While early reports of SRS in the adjuvant setting for glioblastoma were disappointing, its role in the preoperative setting and its impact on the anti-tumor adaptive immune response is largely unknown. In this review, we provide an overview of GBM, discuss the potential role of preoperative SRS, and discuss the possible immunogenic effects of this therapy.
Collapse
Affiliation(s)
- Eric J. Lehrer
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (E.J.L.); (A.D.N.); (K.K.S.)
| | - Henry Ruiz-Garcia
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (R.S.E.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Anthony D. Nehlsen
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (E.J.L.); (A.D.N.); (K.K.S.)
| | - Kunal K. Sindhu
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (E.J.L.); (A.D.N.); (K.K.S.)
| | - Rachel Sarabia Estrada
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (R.S.E.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Gerben R. Borst
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK;
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, UK
| | - Jason P. Sheehan
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA 22908, USA;
| | | | - Daniel M. Trifiletti
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (R.S.E.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| |
Collapse
|
13
|
Li R, Wang H, Liang Q, Chen L, Ren J. Radiotherapy for glioblastoma: clinical issues and nanotechnology strategies. Biomater Sci 2022; 10:892-908. [PMID: 34989724 DOI: 10.1039/d1bm01401c] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain cancer in adults with poor prognosis. Despite the current state of knowledge on its genetic characteristics, relatively little progress has been made in improving the treatment of patients with this fatal disease. Radiotherapy (RT) has been identified as a crucial treatment for GBM following surgical resection to improve both local control and survival. Unfortunately, radiotherapy resistance is frequently observed in GBM patients, which is the major reason for the high mortality rate of cancer patients. Radioresistance of GBM is often multifactorial and heterogeneous, and associated with the recurrence of GBM after surgery. Nanotechnology has gained increasing attention and has already been investigated for optimization of radiosensitization due to the unique properties of nanobiomaterials, such as photoelectric decay characteristics or potential as carriers for drug delivery to the central nervous system. A large body of preclinical data has accumulated over the past several years, in which nanotechnology-based strategies exhibit promising potential to enhance the radiosensitivity of GBM, both in cellular and animal models. In this review, we summarize the mechanisms of GBM radioresistance, including tumor cell-intrinsic factors as well as tumor microenvironment (TME). We further discuss current nano-biotechnology-based radiosensitizer in the treatment of GBM, summarize the latest findings, highlight challenges, and put forward prospects for the future of nano-radiosensitizers. These data suggest that nanotechnology has the potential to address many of the clinical challenges and nanobiomaterials would become promising next-generation radiotherapy sensitizers for GBM treatment.
Collapse
Affiliation(s)
- Ruiqi Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Haihong Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Qing Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Lian Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| |
Collapse
|
14
|
Bilynsky C, Millot N, Papa A. Radiation nanosensitizers in cancer therapy-From preclinical discoveries to the outcomes of early clinical trials. Bioeng Transl Med 2022; 7:e10256. [PMID: 35079631 PMCID: PMC8780058 DOI: 10.1002/btm2.10256] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/05/2021] [Accepted: 08/12/2021] [Indexed: 12/31/2022] Open
Abstract
Improving the efficacy and spatial targeting of radiation therapy while sparing surrounding normal tissues has been a guiding principle for its use in cancer therapy. Nanotechnologies have shown considerable growth in terms of innovation and the development of new therapeutic approaches, particularly as radiosensitizers. The aim of this study was to systematically review how nanoparticles (NPs) are used to enhance the radiotherapeutic effect, including preclinical and clinical studies. Clinicaltrials.gov was used to perform the search using the following terms: radiation, cancer, and NPs. In this review, we describe the various designs of nano-radioenhancers, the rationale for using such technology, as well as their chemical and biological effects. Human trials are then discussed with an emphasis on their design and detailed clinical outcomes.
Collapse
Affiliation(s)
- Colette Bilynsky
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
- Present address:
Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Nadine Millot
- Laboratoire Interdisciplinaire Carnot de BourgogneUMR 6303, CNRS, Université Bourgogne Franche‐ComtéDijon CedexFrance
| | - Anne‐Laure Papa
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| |
Collapse
|
15
|
Singh M, Jindal D, Agarwal V, Pathak D, Sharma M, Pancham P, Mani S, Rachana. New phase therapeutic pursuits for targeted drug delivery in glioblastoma multiforme. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:866-888. [PMID: 36654821 PMCID: PMC9834280 DOI: 10.37349/etat.2022.00118] [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: 06/11/2022] [Accepted: 08/19/2022] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme (GBM) is known as the most aggressive and prevalent brain tumor with a high mortality rate. It is reported in people who are as young as 10 years old to as old as over 70 years old, exhibiting inter and intra tumor heterogeneity. There are several genomic and proteomic investigations that have been performed to find the unexplored potential targets of the drug against GBM. Therefore, certain effective targets have been taken to further validate the studies embarking on the robustness in the field of medicinal chemistry followed by testing in clinical trials. Also, The Cancer Genome Atlas (TCGA) project has identified certain overexpressed targets involved in the pathogenesis of GBM in three major pathways, i.e., tumor protein 53 (p53), retinoblastoma (RB), and receptor tyrosine kinase (RTK)/rat sarcoma virus (Ras)/phosphoinositide 3-kinase (PI3K) pathways. This review focuses on the compilation of recent developments in the fight against GBM thus, directing future research into the elucidation of pathogenesis and potential cure for GBM. Also, it highlights the potential biomarkers that have undergone extensive research and have promising prognostic and predictive values. Additionally, this manuscript analyses the advent of gene therapy and immunotherapy, unlocking the way to consider treatment approaches other than, or in addition to, conventional chemo-radiation therapies. This review study encompasses all the relevant research studies associated with the pathophysiology, occurrence, diagnostic tools, and therapeutic intervention for GBM. It highlights the evolution of various therapeutic perspectives against GBM from the most conventional form of radiotherapy to the recent advancement of gene/cell/immune therapy. Further, the review focuses on various targeted therapies for GBM including chemotherapy sensitization, radiotherapy, nanoparticles based, immunotherapy, cell therapy, and gene therapy which would offer a comprehensive account for exploring several facets related to GBM prognostics.
Collapse
Affiliation(s)
- Manisha Singh
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India,Correspondence: Manisha Singh, Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India.
| | - Divya Jindal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Vinayak Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Deepanshi Pathak
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Mansi Sharma
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Pranav Pancham
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Shalini Mani
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| | - Rachana
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201301, India
| |
Collapse
|
16
|
Morás AM, Henn JG, Steffens Reinhardt L, Lenz G, Moura DJ. Recent developments in drug delivery strategies for targeting DNA damage response in glioblastoma. Life Sci 2021; 287:120128. [PMID: 34774874 DOI: 10.1016/j.lfs.2021.120128] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 12/19/2022]
Abstract
Glioblastoma is the most frequent and malignant brain tumor. The median survival for this disease is approximately 15 months, and despite all the available treatment strategies employed, it remains an incurable disease. Preclinical and clinical research have shown that the resistance process related to DNA damage repair pathways, glioma stem cells, blood-brain barrier selectivity, and dose-limiting toxicity of systemic treatment leads to poor clinical outcomes. In this context, the advent of drug delivery systems associated with localized treatment seems to be a promising and versatile alternative to overcome the failure of the current treatment approaches. In order to bypass therapeutic tumor resistance mechanisms, more effective combinatorial therapies should be identified, such as the use of cytotoxic drugs combined with the inhibition of DNA damage response (DDR)-related targets. Additionally, critical reasoning about the delivery approach and administration route in brain tumors treatment innovation is essential. The outcomes of future experimental studies regarding the association of delivery systems, alternative treatment routes, and DDR targets are expected to lead to the development of refined therapeutic interventions. Novel therapeutic approaches could improve the life's quality of glioblastoma patients and increase their survival rate.
Collapse
Affiliation(s)
- A M Morás
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre, (UFCSPA), Porto Alegre, Brazil.
| | - J G Henn
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre, (UFCSPA), Porto Alegre, Brazil.
| | - L Steffens Reinhardt
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre, (UFCSPA), Porto Alegre, Brazil.
| | - G Lenz
- Department of Biophysics and Center of Biotechnology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
| | - D J Moura
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre, (UFCSPA), Porto Alegre, Brazil.
| |
Collapse
|
17
|
Elbanna M, Chowdhury NN, Rhome R, Fishel ML. Clinical and Preclinical Outcomes of Combining Targeted Therapy With Radiotherapy. Front Oncol 2021; 11:749496. [PMID: 34733787 PMCID: PMC8558533 DOI: 10.3389/fonc.2021.749496] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022] Open
Abstract
In the era of precision medicine, radiation medicine is currently focused on the precise delivery of highly conformal radiation treatments. However, the tremendous developments in targeted therapy are yet to fulfill their full promise and arguably have the potential to dramatically enhance the radiation therapeutic ratio. The increased ability to molecularly profile tumors both at diagnosis and at relapse and the co-incident progress in the field of radiogenomics could potentially pave the way for a more personalized approach to radiation treatment in contrast to the current ‘‘one size fits all’’ paradigm. Few clinical trials to date have shown an improved clinical outcome when combining targeted agents with radiation therapy, however, most have failed to show benefit, which is arguably due to limited preclinical data. Several key molecular pathways could theoretically enhance therapeutic effect of radiation when rationally targeted either by directly enhancing tumor cell kill or indirectly through the abscopal effect of radiation when combined with novel immunotherapies. The timing of combining molecular targeted therapy with radiation is also important to determine and could greatly affect the outcome depending on which pathway is being inhibited.
Collapse
Affiliation(s)
- May Elbanna
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, United States.,Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Nayela N Chowdhury
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ryan Rhome
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, United States.,Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Melissa L Fishel
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
18
|
Liu W, Chen B, Zheng H, Xing Y, Chen G, Zhou P, Qian L, Min Y. Advances of Nanomedicine in Radiotherapy. Pharmaceutics 2021; 13:pharmaceutics13111757. [PMID: 34834172 PMCID: PMC8622383 DOI: 10.3390/pharmaceutics13111757] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy (RT) remains one of the current main treatment strategies for many types of cancer. However, how to improve RT efficiency while reducing its side effects is still a large challenge to be overcome. Advancements in nanomedicine have provided many effective approaches for radiosensitization. Metal nanoparticles (NPs) such as platinum-based or hafnium-based NPs are proved to be ideal radiosensitizers because of their unique physicochemical properties and high X-ray absorption efficiency. With nanoparticles, such as liposomes, bovine serum albumin, and polymers, the radiosensitizing drugs can be promoted to reach the tumor sites, thereby enhancing anti-tumor responses. Nowadays, the combination of some NPs and RT have been applied to clinical treatment for many types of cancer, including breast cancer. Here, as well as reviewing recent studies on radiotherapy combined with inorganic, organic, and biomimetic nanomaterials for oncology, we analyzed the underlying mechanisms of NPs radiosensitization, which may contribute to exploring new directions for the clinical translation of nanoparticle-based radiosensitizers.
Collapse
Affiliation(s)
- Wei Liu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (W.L.); (P.Z.)
| | - Bo Chen
- Department of Bio-X Interdisciplinary Science at Hefei National Laboratory (HFNL) for Physical Science at the Microscale, University of Science and Technology of China, Hefei 230026, China; (B.C.); (Y.M.)
| | - Haocheng Zheng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Yun Xing
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Guiyuan Chen
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Peijie Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (W.L.); (P.Z.)
| | - Liting Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (W.L.); (P.Z.)
- Correspondence:
| | - Yuanzeng Min
- Department of Bio-X Interdisciplinary Science at Hefei National Laboratory (HFNL) for Physical Science at the Microscale, University of Science and Technology of China, Hefei 230026, China; (B.C.); (Y.M.)
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.Z.); (Y.X.); (G.C.)
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
19
|
Ruiz-Garcia H, Ramirez-Loera C, Malouff TD, Seneviratne DS, Palmer JD, Trifiletti DM. Novel Strategies for Nanoparticle-Based Radiosensitization in Glioblastoma. Int J Mol Sci 2021; 22:9673. [PMID: 34575840 PMCID: PMC8465220 DOI: 10.3390/ijms22189673] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 01/09/2023] Open
Abstract
Radiotherapy (RT) is one of the cornerstones in the current treatment paradigm for glioblastoma (GBM). However, little has changed in the management of GBM since the establishment of the current protocol in 2005, and the prognosis remains grim. Radioresistance is one of the hallmarks for treatment failure, and different therapeutic strategies are aimed at overcoming it. Among these strategies, nanomedicine has advantages over conventional tumor therapeutics, including improvements in drug delivery and enhanced antitumor properties. Radiosensitizing strategies using nanoparticles (NP) are actively under study and hold promise to improve the treatment response. We aim to describe the basis of nanomedicine for GBM treatment, current evidence in radiosensitization efforts using nanoparticles, and novel strategies, such as preoperative radiation, that could be synergized with nanoradiosensitizers.
Collapse
Affiliation(s)
- Henry Ruiz-Garcia
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| | | | - Timothy D. Malouff
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
| | - Danushka S. Seneviratne
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
| | - Joshua D. Palmer
- Department of Radiation Oncology, Ohio State University, Columbus, OH 43210, USA;
| | - Daniel M. Trifiletti
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| |
Collapse
|
20
|
Alavian F, Ghasemi S. The Effectiveness of Nanoparticles on Gene Therapy for Glioblastoma Cells Apoptosis: A Systematic Review. Curr Gene Ther 2021; 21:230-245. [PMID: 33655831 DOI: 10.2174/1566523221666210224110454] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most common and fatal type of glioma. Nanoparticles (NPs) are used in new approaches for the delivery of gene therapy in the treatment of GBM. INTRODUCTION The purpose of this article was to review the efficacy of NPs as the targeted carriers in the gene therapy aimed at apoptosis in GBM. METHODS The appropriate keywords such as nanoparticle, glioblastoma, gene therapy, apoptosis, and related words were used to search from PubMed, ISI Web of Science, and Scopus for relevant publications up to September 4, 2020, with no language restrictions. The present systematic review was performed based on PRISMA protocol and reviewed the articles evaluating the effects of nanoparticles, carriers of various gene therapies essentials, on GBM cells apoptosis in vitro and in vivo. The selected articles were considered using specific scores on the quality of the articles. Data extraction and quality evaluation were performed by two reviewers. RESULTS Of 101 articles retrieved, forty-two met the inclusion criteria and were, therefore, subjected to the final deduction. The most widely used NP in GBM gene therapy studies is polyamidoamine (PAMAM). The most common gene therapy approach for apoptosis in GBM is using siRNAs. CONCLUSION In conclusion, these studies validated that NPs could be a practical choice to enhance the efficiency and specific delivery in gene therapies for GBM cell apoptosis. However, the choice of NP type and gene therapy mechanism affect the GBM cell apoptotic efficiency.
Collapse
Affiliation(s)
- Firoozeh Alavian
- Department of Biology, School of Basic Sciences, Farhangian University, Tehran, Iran
| | - Sorayya Ghasemi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
21
|
Kanvinde PP, Malla AP, Connolly NP, Szulzewsky F, Anastasiadis P, Ames HM, Kim AJ, Winkles JA, Holland EC, Woodworth GF. Leveraging the replication-competent avian-like sarcoma virus/tumor virus receptor-A system for modeling human gliomas. Glia 2021; 69:2059-2076. [PMID: 33638562 PMCID: PMC8591561 DOI: 10.1002/glia.23984] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/20/2022]
Abstract
Gliomas are the most common primary intrinsic brain tumors occurring in adults. Of all malignant gliomas, glioblastoma (GBM) is considered the deadliest tumor type due to diffuse brain invasion, immune evasion, cellular, and molecular heterogeneity, and resistance to treatments resulting in high rates of recurrence. An extensive understanding of the genomic and microenvironmental landscape of gliomas gathered over the past decade has renewed interest in pursuing novel therapeutics, including immune checkpoint inhibitors, glioma-associated macrophage/microglia (GAMs) modulators, and others. In light of this, predictive animal models that closely recreate the conditions and findings found in human gliomas will serve an increasingly important role in identifying new, effective therapeutic strategies. Although numerous syngeneic, xenograft, and transgenic rodent models have been developed, few include the full complement of pathobiological features found in human tumors, and therefore few accurately predict bench-to-bedside success. This review provides an update on how genetically engineered rodent models based on the replication-competent avian-like sarcoma (RCAS) virus/tumor virus receptor-A (tv-a) system have been used to recapitulate key elements of human gliomas in an immunologically intact host microenvironment and highlights new approaches using this model system as a predictive tool for advancing translational glioma research.
Collapse
Affiliation(s)
- Pranjali P Kanvinde
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Adarsha P Malla
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nina P Connolly
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Frank Szulzewsky
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Pavlos Anastasiadis
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Heather M Ames
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jeffrey A Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Eric C Holland
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Seattle Tumor Translational Research Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Wiwatchaitawee K, Quarterman JC, Geary SM, Salem AK. Enhancement of Therapies for Glioblastoma (GBM) Using Nanoparticle-based Delivery Systems. AAPS PharmSciTech 2021; 22:71. [PMID: 33575970 DOI: 10.1208/s12249-021-01928-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/10/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of malignant brain tumor. Current FDA-approved treatments include surgical resection, radiation, and chemotherapy, while hyperthermia, immunotherapy, and most relevantly, nanoparticle (NP)-mediated delivery systems or combinations thereof have shown promise in preclinical studies. Drug-carrying NPs are a promising approach to brain delivery as a result of their potential to facilitate the crossing of the blood-brain barrier (BBB) via two main types of transcytosis mechanisms: adsorptive-mediated transcytosis (AMT) and receptor-mediated transcytosis (RMT). Their ability to accumulate in the brain can thus provide local sustained release of tumoricidal drugs at or near the site of GBM tumors. NP-based drug delivery has the potential to significantly reduce drug-related toxicity, increase specificity, and consequently improve the lifespan and quality of life of patients with GBM. Due to significant advances in the understanding of the molecular etiology and pathology of GBM, the efficacy of drugs loaded into vectors targeting this disease has increased in both preclinical and clinical settings. Multitargeting NPs, such as those incorporating multiple specific targeting ligands, are an innovative technology that can lead to decreased off-target effects while simultaneously having increased accumulation and action specifically at the tumor site. Targeting ligands can include antibodies, or fragments thereof, and peptides or small molecules, which can result in a more controlled drug delivery system compared to conventional drug treatments. This review focuses on GBM treatment strategies, summarizing current options and providing a detailed account of preclinical findings with prospective NP-based approaches aimed at improving tumor targeting and enhancing therapeutic outcomes for GBM patients.
Collapse
|
23
|
Hwang D, Dismuke T, Tikunov A, Rosen EP, Kagel JR, Ramsey JD, Lim C, Zamboni W, Kabanov AV, Gershon TR, Sokolsky-Papkov PhD M. Poly(2-oxazoline) nanoparticle delivery enhances the therapeutic potential of vismodegib for medulloblastoma by improving CNS pharmacokinetics and reducing systemic toxicity. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 32:102345. [PMID: 33259959 PMCID: PMC8160025 DOI: 10.1016/j.nano.2020.102345] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/16/2020] [Accepted: 11/20/2020] [Indexed: 10/22/2022]
Abstract
We report a nanoparticle formulation of the SHH-pathway inhibitor vismodegib that improves efficacy for medulloblastoma, while reducing toxicity. Limited blood-brain barrier (BBB) penetration and dose-limiting extitle/citraneural toxicities complicate systemic therapies for brain tumors. Vismodegib is FDA-approved for SHH-driven basal cell carcinoma, but implementation for medulloblastoma has been limited by inadequate efficacy and excessive bone toxicity. To address these issues through optimized drug delivery, we formulated vismodegib in polyoxazoline block copolymer micelles (POx-vismo). We then evaluated POx-vismo in transgenic mice that develop SHH-driven medulloblastomas with native vasculature and tumor microenvironment. POx-vismo improved CNS pharmacokinetics and reduced bone toxicity. Mechanistically, the nanoparticle carrier did not enter the CNS, and acted within the vascular compartment to improve drug delivery. Unlike conventional vismodegib, POx-vismo extended survival in medulloblastoma-bearing mice. Our results show the broad potential for non-targeted nanoparticle formulation to improve systemic brain tumor therapy, and specifically to improve vismodegib therapy for SHH-driven cancers.
Collapse
Affiliation(s)
- Duhyeong Hwang
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
| | - Taylor Dismuke
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Andrey Tikunov
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Elias P Rosen
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States
| | - John R Kagel
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Jacob D Ramsey
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
| | - Chaemin Lim
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
| | - William Zamboni
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Timothy R Gershon
- Department of Neurology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA; Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA.
| | - Marina Sokolsky-Papkov PhD
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA.
| |
Collapse
|
24
|
Lin G, Revia RA, Zhang M. Inorganic Nanomaterial-Mediated Gene Therapy in Combination with Other Antitumor Treatment Modalities. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007096. [PMID: 34366761 PMCID: PMC8336227 DOI: 10.1002/adfm.202007096] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Indexed: 05/05/2023]
Abstract
Cancer is a genetic disease originating from the accumulation of gene mutations in a cellular subpopulation. Although many therapeutic approaches have been developed to treat cancer, recent studies have revealed an irrefutable challenge that tumors evolve defenses against some therapies. Gene therapy may prove to be the ultimate panacea for cancer by correcting the fundamental genetic errors in tumors. The engineering of nanoscale inorganic carriers of cancer therapeutics has shown promising results in the efficacious and safe delivery of nucleic acids to treat oncological diseases in small-animal models. When these nanocarriers are used for co-delivery of gene therapeutics along with auxiliary treatments, the synergistic combination of therapies often leads to an amplified health benefit. In this review, an overview of the inorganic nanomaterials developed for combinatorial therapies of gene and other treatment modalities is presented. First, the main principles of using nucleic acids as therapeutics, inorganic nanocarriers for medical applications and delivery of gene/drug payloads are introduced. Next, the utility of recently developed inorganic nanomaterials in different combinations of gene therapy with each of chemo, immune, hyperthermal, and radio therapy is examined. Finally, current challenges in the clinical translation of inorganic nanomaterial-mediated therapies are presented and outlooks for the field are provided.
Collapse
Affiliation(s)
- Guanyou Lin
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Richard A Revia
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
25
|
Conniot J, Talebian S, Simões S, Ferreira L, Conde J. Revisiting gene delivery to the brain: silencing and editing. Biomater Sci 2020; 9:1065-1087. [PMID: 33315025 DOI: 10.1039/d0bm01278e] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative disorders, ischemic brain diseases, and brain tumors are debilitating diseases that severely impact a person's life and could possibly lead to their demise if left untreated. Many of these diseases do not respond to small molecule therapeutics and have no effective long-term therapy. Gene therapy offers the promise of treatment or even a cure for both genetic and acquired brain diseases, mediated by either silencing or editing disease-specific genes. Indeed, in the last 5 years, significant progress has been made in the delivery of non-coding RNAs as well as gene-editing formulations to the brain. Unfortunately, the delivery is a major limiting factor for the success of gene therapies. Both viral and non-viral vectors have been used to deliver genetic information into a target cell, but they have limitations. Viral vectors provide excellent transduction efficiency but are associated with toxic effects and have limited packaging capacity; however, non-viral vectors are less toxic and show a high packaging capacity at the price of low transfection efficiency. Herein, we review the progress made in the field of brain gene therapy, particularly in the design of non-toxic and trackable non-viral vectors, capable of controlled release of genes in response to internal/external triggers, and in the delivery of formulations for gene editing. The application of these systems in the context of various brain diseases in pre-clinical and clinical tests will be discussed. Such promising approaches could potentially pave the way for clinical realization of brain gene therapies.
Collapse
Affiliation(s)
- João Conniot
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal.
| | | | | | | | | |
Collapse
|
26
|
Clement S, Campbell JM, Deng W, Guller A, Nisar S, Liu G, Wilson BC, Goldys EM. Mechanisms for Tuning Engineered Nanomaterials to Enhance Radiation Therapy of Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2003584. [PMID: 33344143 PMCID: PMC7740107 DOI: 10.1002/advs.202003584] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Indexed: 05/12/2023]
Abstract
Engineered nanomaterials that produce reactive oxygen species on exposure to X- and gamma-rays used in radiation therapy offer promise of novel cancer treatment strategies. Similar to photodynamic therapy but suitable for large and deep tumors, this new approach where nanomaterials acting as sensitizing agents are combined with clinical radiation can be effective at well-tolerated low radiation doses. Suitably engineered nanomaterials can enhance cancer radiotherapy by increasing the tumor selectivity and decreasing side effects. Additionally, the nanomaterial platform offers therapeutically valuable functionalities, including molecular targeting, drug/gene delivery, and adaptive responses to trigger drug release. The potential of such nanomaterials to be combined with radiotherapy is widely recognized. In order for further breakthroughs to be made, and to facilitate clinical translation, the applicable principles and fundamentals should be articulated. This review focuses on mechanisms underpinning rational nanomaterial design to enhance radiation therapy, the understanding of which will enable novel ways to optimize its therapeutic efficacy. A roadmap for designing nanomaterials with optimized anticancer performance is also shown and the potential clinical significance and future translation are discussed.
Collapse
Affiliation(s)
- Sandhya Clement
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Jared M. Campbell
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Wei Deng
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Anna Guller
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
- Institute for Regenerative MedicineSechenov First Moscow State Medical University (Sechenov University)Trubetskaya StreetMoscow119991Russia
| | - Saadia Nisar
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Guozhen Liu
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| | - Brian C. Wilson
- Department of Medical BiophysicsUniversity of Toronto/Princess Margaret Cancer CentreUniversity Health NetworkColledge StreetTorontoOntarioON M5G 2C1Canada
| | - Ewa M. Goldys
- ARC Centre of Excellence for Nanoscale BiophotonicsThe Graduate School of Biomedical EngineeringUniversity of New South WalesHigh StreetKensingtonNew South Wales2052Australia
| |
Collapse
|
27
|
Lee JA, Ayat N, Sun Z, Tofilon PJ, Lu ZR, Camphausen K. Improving Radiation Response in Glioblastoma Using ECO/siRNA Nanoparticles Targeting DNA Damage Repair. Cancers (Basel) 2020; 12:cancers12113260. [PMID: 33158243 PMCID: PMC7694254 DOI: 10.3390/cancers12113260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/30/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Glioblastoma (GBM) is the most common form of brain cancer and among the most lethal of human cancers. Radiation therapy is a mainstay in the standard of care for GBM, killing tumor cells by creating DNA damage. Inhibiting DNA damage repair (DDR) proteins enhances radiation therapy by not allowing tumor cells to repair the DNA damage caused by radiation. The aim of our study was to investigate whether the novel nanoparticle material, ECO, could be used to deliver small interfering RNA (siRNA) to GBM tumor cells and temporarily reduce the production of DDR proteins to improve radiation therapy outcomes. SiRNAs can be designed to target an innumerable number of genes and with the right delivery vehicle can be used in a variety of disease settings. Our work provides support for the use of the novel ECO material for delivery of siRNA in GBM. Abstract Radiation therapy is a mainstay in the standard of care for glioblastoma (GBM), thus inhibiting the DNA damage response (DDR) is a major strategy to improve radiation response and therapeutic outcomes. Small interfering RNA (siRNA) therapy holds immeasurable potential for the treatment of GBM, however delivery of the siRNA payload remains the largest obstacle for clinical implementation. Here we demonstrate the effectiveness of the novel nanomaterial, ECO (1-aminoethylimino[bis(N-oleoylcysteinylaminoethyl) propionamide]), to deliver siRNA targeting DDR proteins ataxia telangiectasia mutated and DNA-dependent protein kinase (DNApk-cs) for the radiosensitzation of GBM in vitro and in vivo. ECO nanoparticles (NPs) were shown to efficiently deliver siRNA and silence target protein expression in glioma (U251) and glioma stem cell lines (NSC11, GBMJ1). Importantly, ECO NPs displayed no cytotoxicity and minimal silencing of genes in normal astrocytes. Treatment with ECO/siRNA NPs and radiation resulted in the prolonged presence of γH2AX foci, indicators of DNA damage, and increased radiosensitivity in all tumor cell lines. In vivo, intratumoral injection of ECO/siDNApk-cs NPs with radiation resulted in a significant increase in survival compared with injection of NPs alone. These data suggest the ECO nanomaterial can effectively deliver siRNA to more selectively target and radiosensitize tumor cells to improve therapeutic outcomes in GBM.
Collapse
Affiliation(s)
- Jennifer A. Lee
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (P.J.T.); (K.C.)
- Correspondence:
| | - Nadia Ayat
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44140, USA; (N.A.); (Z.S.); (Z.-R.L.)
| | - Zhanhu Sun
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44140, USA; (N.A.); (Z.S.); (Z.-R.L.)
| | - Philip J. Tofilon
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (P.J.T.); (K.C.)
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44140, USA; (N.A.); (Z.S.); (Z.-R.L.)
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (P.J.T.); (K.C.)
| |
Collapse
|
28
|
Reda M, Bagley AF, Zaidan HY, Yantasee W. Augmenting the therapeutic window of radiotherapy: A perspective on molecularly targeted therapies and nanomaterials. Radiother Oncol 2020; 150:225-235. [PMID: 32598976 DOI: 10.1016/j.radonc.2020.06.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/25/2022]
Abstract
Radiation therapy is a cornerstone of modern cancer therapy alongside surgery, chemotherapy, and immunotherapy, with over half of all cancer patients receiving radiation therapy as part of their treatment regimen. Development of novel radiation sensitizers that can improve the therapeutic window of radiation therapy are sought after, particularly for tumors at an elevated risk of local and regional recurrence such as locally-advanced lung, head and neck, and gastrointestinal tumors. This review discusses clinical strategies to enhance radiotherapy efficacy and decrease toxicity, hence, increasing the overall therapeutic window. A focus is given to the molecular targets that have been identified and their associated mechanisms of action in enhancing radiotherapy. Examples include cell survival and proliferation signaling such as the EGFR and PI3K/AKT/mTOR pathways, DNA repair genes including PARP and ATM/ATR, angiogenic growth factors, epigenetic regulators, and immune checkpoint proteins. By manipulating various mechanisms of tumor resistance to ionizing radiation (IR), targeted therapies hold significant value to increase the therapeutic window of radiotherapy. Further, the use of novel nanoparticles to enhance radiotherapy is also reviewed, including nanoparticle delivery of chemotherapies, metallic (high-Z) nanoparticles, and nanoparticle delivery of targeted therapies - all of which may improve the therapeutic window of radiotherapy by enhancing the tumor response to IR or reducing normal tissue toxicity.
Collapse
Affiliation(s)
- Moataz Reda
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, United States; PDX Pharmaceuticals, Portland, OR 97239, United States
| | - Alexander F Bagley
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | | | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, United States; PDX Pharmaceuticals, Portland, OR 97239, United States.
| |
Collapse
|
29
|
Kumar A, Chaudhary RK, Singh R, Singh SP, Wang SY, Hoe ZY, Pan CT, Shiue YL, Wei DQ, Kaushik AC, Dai X. Nanotheranostic Applications for Detection and Targeting Neurodegenerative Diseases. Front Neurosci 2020; 14:305. [PMID: 32425743 PMCID: PMC7203731 DOI: 10.3389/fnins.2020.00305] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 03/16/2020] [Indexed: 12/13/2022] Open
Abstract
Nanotechnology utilizes engineered materials and devices which function with biological systems at the molecular level and could transform the management of neurodegenerative diseases (NDs) by provoking, reacting to, and intermingling with target sites to stimulate physiological responses while minimizing side effects. Blood-brain barrier (BBB) protects the brain from harmful agents, and transporting drugs across the BBB is a major challenge for diagnosis, targeting, and treatment of NDs. The BBB provides severe limitations for diagnosis and treatment of Alzheimer's disease (AD), Parkinson's disease (PD), and various other neurological diseases. Conventional drug delivery systems generally fail to cross the BBB, thus are inefficient in treatment. Although gradual development through research is ensuring the progress of nanotheranostic approaches from animal to human modeling, aspects of translational applicability and safety are a key concern. This demands a deep understanding of the interaction of body systems with nanomaterials. There are various plant-based nanobioactive compounds which are reported to have applicability in the diagnosis and treatment of these NDs. This review article provides an overview of applications of nanotheranostics in AD and PD. The review also discusses nano-enabled drug delivery systems and their current and potential applications for the treatment of various NDs.
Collapse
Affiliation(s)
- Ajay Kumar
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ravi Kumar Chaudhary
- Department of Biotechnology, Institute of Applied Medicines & Research, Ghaziabad, India
| | - Rachita Singh
- Department of Electrical and Electronics Engineering, IIMT Engineering College, Uttar Pradesh Technical University, Meerut, India
| | - Satya P. Singh
- School of Computer Science & Engineering, Nanyang Technological University, Singapore, Singapore
| | - Shao-Yu Wang
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Zheng-Yu Hoe
- Department of Physical Medicine and Rehabilitation, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Cheng-Tang Pan
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Dong-Qing Wei
- Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Aman Chandra Kaushik
- Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
30
|
Sun H, Wang X, Zhai S. The Rational Design and Biological Mechanisms of Nanoradiosensitizers. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E504. [PMID: 32168899 PMCID: PMC7153263 DOI: 10.3390/nano10030504] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 01/01/2023]
Abstract
Radiotherapy (RT) has been widely used for cancer treatment. However, the intrinsic drawbacks of RT, such as radiotoxicity in normal tissues and tumor radioresistance, promoted the development of radiosensitizers. To date, various kinds of nanoparticles have been found to act as radiosensitizers in cancer radiotherapy. This review focuses on the current state of nanoradiosensitizers, especially the related biological mechanisms, and the key design strategies for generating nanoradiosensitizers. The regulation of oxidative stress, DNA damage, the cell cycle, autophagy and apoptosis by nanoradiosensitizers in vitro and in vivo is highlighted, which may guide the rational design of therapeutics for tumor radiosensitization.
Collapse
Affiliation(s)
- Hainan Sun
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
- Shandong Vocational College of Light Industry, Zibo 255300, Shandong, China
| | - Xiaoling Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
| | - Shumei Zhai
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
| |
Collapse
|
31
|
Nuzhina JV, Shtil AA, Prilepskii AY, Vinogradov VV. Preclinical Evaluation and Clinical Translation of Magnetite-Based Nanomedicines. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101282] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
32
|
Zhang C, Yan L, Gu Z, Zhao Y. Strategies based on metal-based nanoparticles for hypoxic-tumor radiotherapy. Chem Sci 2019; 10:6932-6943. [PMID: 31588260 PMCID: PMC6676466 DOI: 10.1039/c9sc02107h] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/10/2019] [Indexed: 01/02/2023] Open
Abstract
Radiotherapy (RT) is one of the most effective and frequent clinical cancer treatments. Nevertheless, RT can cause damage to normal tissues around tumors under high-dose ionizing radiation. Inspired by versatile metal-based nanomaterials, great efforts have been devoted to developing nanomaterials with high-Z metal elements as radiosensitizers by depositing more energy into tumors for RT enhancement. However, these metal-based nanomaterial-mediated RTs are highly O2-dependent. Unfortunately, O2 concentrations within the majority of solid tumors exhibit low levels, which seriously hampers the antitumor efficacy of these nanomaterials during RT. Therefore, the development of novel metal-based nanomaterials as radiosensitizers capable of avoiding the radioresistance induced by tumor hypoxia is highly desirable and important. Currently, the most effective approaches to reverse the radioresistance of hypoxic tumors are to introduce nanomaterials with O2-elevating ability by delivering exogenous O2, generating O2 in situ, increasing intratumoral blood flow, or reducing HIF-1 expression to harness the O2 level in solid tumors. Besides these, recently, some innovative and simple strategies by employing nanoradiosensitizers with diminished oxygen dependence have also been applied to combat unmet hypoxic challenges, in which nanoradiosensitizers can target tumor hypoxia for selective RT, enhance oxygen-independent ROS generation, or combine with non-oxygen dependent cancer therapies for synergistic treatments. These approaches and strategies provide new avenues for enhanced hypoxic-tumor RT. Nevertheless, an overall review aiming specifically at these strategies is still rare. Herein, we present an overview about recent advances in metal-based nanomaterials for hypoxic-tumor RT, and give a detailed discussion about the design and working mechanisms of these strategies in their application of RT. Finally, current challenges and future perspectives are also pointed out in this field.
Collapse
Affiliation(s)
- Chenyang Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics , Chinese Academy of Sciences , Beijing 100049 , China .
- College of Materials Science and Optoelectronic Technology , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Liang Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics , Chinese Academy of Sciences , Beijing 100049 , China .
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics , Chinese Academy of Sciences , Beijing 100049 , China .
- College of Materials Science and Optoelectronic Technology , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Yuliang Zhao
- CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Chinese Academy of Sciences , Beijing 100190 , China .
- College of Materials Science and Optoelectronic Technology , University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
33
|
Abstract
Certain genetic mutations lead to the development of cancer through unchecked cell growth and division. Cancer is typically treated through surgical resection, radiotherapy, and small-molecule chemotherapy. A relatively recent approach to cancer therapy involves the use of a natural process wherein small RNA molecules regulate gene expression in a pathway known as RNA interference (RNAi). RNA oligomers pair with a network of proteins to form an RNA-induced silencing complex, which inhibits the translation of mRNA into proteins, thereby controlling the expression of gene products. Synthetically produced RNA oligomers may be designed to target and silence specific oncogenes to provide cancer therapy. The primary challenges facing the use of the RNAi pathway for cancer therapy are the safe and efficacious delivery of RNA payloads and their release at pertinent sites within disease-causing cells. Nucleases are abundant in the bloodstream and intracellular environment, and therapeutic RNA sequences often require a suitable carrier to provide protection from degradation prior to reaching their site of action in the body. The use of metal core nanoparticles (NPs) serving as targeted delivery vehicles able to shield and direct RNA payloads to their intended destinations have recently gained favor. Biological barriers present in the body establish a size prerequisite for drug delivery vehicles; to overcome recognition by the body's immune system and to gain access to intracellular environments, drug carriers must be small (< 100 nm). Iron oxide and gold core NPs can be synthesized with a high degree of control to create uniform ultrasmall drug delivery vehicles capable of bypassing key biological barriers. While progress is being made in size control of liposomal and polymer NPs, such advances still lag in comparison to the exquisite tunability and time stability of size engineering achievable with metal core NPs at bulk scales. Further, unlike lipid- and viral-based transfection agents, the biodistribution of metal core NPs can be traced using noninvasive imaging techniques that capitalize on the interaction of electromagnetic radiation and the inorganic atoms at the core of the NPs. Finally, metal core NPs have been shown to match the transfection efficiency of conventional RNA-delivery vehicles while also providing less immunogenicity and minimal side effects through the addition of tumor-targeting ligands on their surface. This Account reviews recent advances in the use of iron oxide and gold NPs for RNAi therapy. An overview of the different types of RNA-based therapies is provided along with a discussion of the advantages and current limitations of the technique. We highlight design considerations for the use of iron oxide and gold NP carriers in RNAi, including a discussion of the importance of size and its role in traversing biological barriers, NP surface modifications required for targeted delivery and RNA payload release, and auxiliary properties supporting imaging functionality for treatment monitoring. Applications of NPs for combination therapies including the pairing of RNAi with chemotherapy, photothermal therapy, immunotherapy, and radiotherapy are explored through examples. Finally, future perspectives are provided with a focus on the current limitations and the potential for clinical translation of iron oxide and gold NPs in RNAi therapy.
Collapse
Affiliation(s)
- Richard A. Revia
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Zachary R. Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
34
|
Jain KK. A Critical Overview of Targeted Therapies for Glioblastoma. Front Oncol 2018; 8:419. [PMID: 30374421 PMCID: PMC6196260 DOI: 10.3389/fonc.2018.00419] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/10/2018] [Indexed: 01/07/2023] Open
Abstract
Over the past century, treatment of malignant tumors of the brain has remained a challenge. Refinements in neurosurgical techniques, discovery of powerful chemotherapeutic agents, advances in radiotherapy, applications of biotechnology, and improvements in methods of targeted delivery have led to some extension of length of survival of glioblastoma patients. Refinements in surgery are mentioned because most of the patients with glioblastoma undergo surgery and many of the other innovative therapies are combined with surgery. However, cure of glioblastoma has remained elusive because it requires complete destruction of the tumor. Radical surgical ablation is not possible in the brain and even a small residual tumor leads to rapid recurrence that eventually kills the patient. Blood-brain barrier (BBB) comprising brain endothelial cells lining the cerebral microvasculature, limits delivery of drugs to the brain. Even though opening of the BBB in tumor core occurs locally, BBB limits systemic chemotherapy especially at the tumor periphery, where tumor cells invade normal brain structure comprising intact BBB. Comprehensive approaches are necessary to gain maximally from promising targeted therapies. Common methods used for critical evaluation of targeted therapies for glioblastoma include: (1) novel methods for targeted delivery of chemotherapy; (2) strategies for delivery through BBB and blood-tumor barriers; (3) innovations in radiotherapy for selective destruction of tumor; (4) techniques for local destruction of tumor; (5) tumor growth inhibitors; (6) immunotherapy; and (7) cell/gene therapies. Suggestions for improvements in glioblastoma therapy include: (1) controlled targeted delivery of anticancer therapy to glioblastoma through the BBB using nanoparticles and monoclonal antibodies; (2) direct introduction of genetically modified bacteria that selectively destroy cancer cells but spare the normal brain into the remaining tumor after resection; (3) use of better animal models for preclinical testing; and (4) personalized/precision medicine approaches to therapy in clinical trials and translation into practice of neurosurgery and neurooncology. Advances in these techniques suggest optimism for the future management of glioblastoma.
Collapse
|
35
|
Abstract
Glioblastoma multiforme (GBM), a grade IV astrocytoma as defined by the World Health Organization (WHO) criteria, is the most common primary central nervous system tumor in adults. After treatment with the current standard of care consisting of surgical resection, concurrent temozolomide (TMZ), and radiation, the median survival is only 15 months. The limited and less-effective treatment options for these highly aggressive GBMs call for the development of new techniques and the improvement of existing technologies. Nanotechnology has shown promise in treating this disease, and some nanomaterials have demonstrated the ability to cross the blood–brain barrier (BBB) and remain in GBM tissues. Although the retention of nanoparticles (NPs) in GBM tissue is necessary to elicit an antitumor response, the delivery of the NP needs to be enhanced. Current research in nanotechnology is directed at increasing the active targeting of GBM tissue not only for the aid of chemotherapeutic drug delivery but also for imaging studies. This review is aimed at describing advancements in increasing nanotechnology specificity to GBM tissue.
Collapse
|
36
|
Wang Y, Cuzzucoli F, Escobar A, Lu S, Liang L, Wang S. Tumor-on-a-chip platforms for assessing nanoparticle-based cancer therapy. NANOTECHNOLOGY 2018; 29:332001. [PMID: 29794338 DOI: 10.1088/1361-6528/aac7a4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Cancer has become the most prevalent cause of deaths, placing a huge economic and healthcare burden worldwide. Nanoparticles (NPs), as a key component of nanomedicine, provide alternative options for promoting the efficacy of cancer therapy. Current conventional cancer models have limitations in predicting the effects of various cancer treatments. To overcome these limitations, biomimetic and novel 'tumor-on-a-chip' platforms have emerged with other innovative biomedical engineering methods that enable the evaluation of NP-based cancer therapy. In this review, we first describe cancer models for evaluation of NP-based cancer therapy techniques, and then present the latest advances in 'tumor-on-a-chip' platforms that can potentially facilitate clinical translation of NP-based cancer therapies.
Collapse
Affiliation(s)
- Yimin Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, People's Republic of China. Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang Province, 310003, People's Republic of China. Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, People's Republic of China
| | | | | | | | | | | |
Collapse
|
37
|
Mochizuki AY, Frost IM, Mastrodimos MB, Plant AS, Wang AC, Moore TB, Prins RM, Weiss PS, Jonas SJ. Precision Medicine in Pediatric Neurooncology: A Review. ACS Chem Neurosci 2018; 9:11-28. [PMID: 29199818 PMCID: PMC6656379 DOI: 10.1021/acschemneuro.7b00388] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Central nervous system tumors are the leading cause of cancer related death in children. Despite much progress in the field of pediatric neurooncology, modern combination treatment regimens often result in significant late effects, such as neurocognitive deficits, endocrine dysfunction, secondary malignancies, and a host of other chronic health problems. Precision medicine strategies applied to pediatric neurooncology target specific characteristics of individual patients' tumors to achieve maximal killing of neoplastic cells while minimizing unwanted adverse effects. Here, we review emerging trends and the current literature that have guided the development of new molecularly based classification schemas, promising diagnostic techniques, targeted therapies, and delivery platforms for the treatment of pediatric central nervous system tumors.
Collapse
Affiliation(s)
- Aaron Y. Mochizuki
- Department
of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Isaura M. Frost
- Department
of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Melina B. Mastrodimos
- Department
of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Ashley S. Plant
- Division
of Pediatric Oncology, Children’s Hospital of Orange County, Orange, California 92868, United States
| | - Anthony C. Wang
- Department
of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Theodore B. Moore
- Department
of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Robert M. Prins
- Department
of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
- Jonsson
Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, United States
| | - Paul S. Weiss
- California
NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Materials Science and Engineering, University of California, Los Angeles, Los
Angeles, California 90095, United States
- Jonsson
Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Steven J. Jonas
- California
NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, United States
- Children’s
Discovery and Innovation Institute, University of California, Los Angeles, Los
Angeles, California 90095, United States
| |
Collapse
|