1
|
Khan M, Nasim M, Feizy M, Parveen R, Gull A, Khan S, Ali J. Contemporary strategies in glioblastoma therapy: Recent developments and innovations. Neuroscience 2024; 560:211-237. [PMID: 39368608 DOI: 10.1016/j.neuroscience.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/30/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024]
Abstract
Glioblastoma multiforme (GBM) represents one of the most prevailing and aggressive primary brain tumors among adults. Despite advances in therapeutic approaches, the complex microenvironment of GBM poses significant challenges in its optimal therapy, which are attributed to immune evasion, tumor repopulation by stem cells, and limited drug penetration across the blood-brain barrier (BBB). Nanotechnology has emerged as a promising avenue for GBM treatment, offering biosafety, sustained drug release, enhanced solubility, and improved BBB penetrability. In this review, a comprehensive overview of recent advancements in nanocarrier-based drug delivery systems for GBM therapy is emphasized. The conventional and novel treatment modalities for GBM and the potential of nanocarriers to overcome existing limitations are comprehensively covered. Furthermore, the updates in the clinical landscape of GBM therapeutics are presented in addition to the current status of drugs and patents in the same context. Through a critical evaluation of existing literature, the therapeutic prospect and limitations of nanocarrier-based drug delivery strategies are highlighted offering insights into future research directions and clinical translation.
Collapse
Affiliation(s)
- Mariya Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Modassir Nasim
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Mohammadamin Feizy
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Rabea Parveen
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Azka Gull
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Saba Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India.
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India.
| |
Collapse
|
2
|
Oishi T, Koizumi S, Kurozumi K. Mesenchymal stem cells as therapeutic vehicles for glioma. Cancer Gene Ther 2024; 31:1306-1314. [PMID: 38654128 DOI: 10.1038/s41417-024-00775-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Glioma is a disease with a poor prognosis despite the availability of multimodality treatments, and the development of novel therapies is urgently needed. Challenges in glioma treatment include the difficulty for drugs to cross the blood-brain barrier when administered systemically and poor drug diffusion when administered locally. Mesenchymal stem cells exhibit advantages for glioma therapy because of their ability to pass through the blood-brain barrier and migrate to tumor cells and their tolerance to the immune system. Therefore, mesenchymal stem cells have been explored as vehicles for various therapeutic agents for glioma treatment. Mesenchymal stem cells loaded with chemotherapeutic drugs show improved penetration and tumor accumulation. For gene therapy, mesenchymal stem cells can be used as vehicles for suicide genes, the so-called gene-directed enzyme prodrug therapy. Mesenchymal stem cell-based oncolytic viral therapies have been attempted in recent years to enhance the efficacy of infection against the tumor, viral replication, and distribution of viral particles. Many uncertainties remain regarding the function and behavior of mesenchymal stem cells in gliomas. However, strategies to increase mesenchymal stem cell migration to gliomas may improve the delivery of therapeutic agents and enhance their anti-tumor effects, representing promising potential for patient treatment.
Collapse
Affiliation(s)
- Tomoya Oishi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shinichiro Koizumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuhiko Kurozumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| |
Collapse
|
3
|
Wang X, Deng Y, Xiao Y, Wang F, Tang Z, Qi X. A double inducible cell ablation system for eliminating senescent astrocytes via apoptosis. Mol Biol Rep 2024; 51:363. [PMID: 38403730 DOI: 10.1007/s11033-024-09297-9] [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: 10/08/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024]
Abstract
PURPOSE Cell senescence stands as a principal risk factor for various neurodegenerative diseases, with astrocytic senescence emerging as a potentially pivotal player in the pathogenesis of aging and neurodegenerative disorders. Clearing senescent astrocytes holds promise as a potential therapeutic approach for senescence-related diseases. METHODS In this study, we designed and constructed two plasmids aimed at inducing apoptosis in senescent astrocytes. This was achieved through the ligation of FKBP (FK506-binding protein) and FRB (FKBP and FKBP rapamycin binding domain) and the formation of caspase8 dimers, thereby achieving the purpose of eliminating senescent astrocytes. RESULTS The developed vector system demonstrates a specifically capability to induce apoptosis in aging astrocytes, offering a targeted approach to eliminate these cells. CONCLUSION The utilization of the double -inducible suicide gene system provides a versatile tool forstimulating cell apoptosis and inhibiting cellular senescence. This system proves valuable in exploring the intrinsic roles and molecular mechanisms of senescent cells in the occurrence and development of aging-related diseases. Ultimately, it offers a potential avenue for developing an efficient treatment system for such conditions.
Collapse
Affiliation(s)
- Xiaoling Wang
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yuxin Deng
- Key Laboratory of Endemic and Minority Disease, Ministry of Education/Key Laboratory of Molecular Biology/Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yan Xiao
- Key Laboratory of Endemic and Minority Disease, Ministry of Education/Key Laboratory of Molecular Biology/Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China
| | - Fan Wang
- Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhi Tang
- Key Laboratory of Endemic and Minority Disease, Ministry of Education/Key Laboratory of Molecular Biology/Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Minority Disease, Ministry of Education/Key Laboratory of Molecular Biology/Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China.
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-Constructed By the Province and Ministry, Guiyang, Guizhou, China.
| |
Collapse
|
4
|
Gysel E, Larijani L, Kallos MS, Krawetz RJ. Suicide gene-enabled cell therapy: A novel approach to scalable human pluripotent stem cell quality control. Bioessays 2023; 45:e2300037. [PMID: 37582645 DOI: 10.1002/bies.202300037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/28/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023]
Abstract
There are an increasing number of cell therapy approaches being studied and employed world-wide. An emerging area in this field is the use of human pluripotent stem cell (hPSC) products for the treatment of injuries/diseases that cannot be effectively managed through current approaches. However, as with any cell therapy, vast numbers of functional and safe cells are required. Bioreactors provide an attractive avenue to generate clinically relevant cell numbers with decreased labour and decreased batch to batch variation. Yet, current methods of performing quality control are not readily scalable to the cell densities produced during bioreactor scale-up. One potential solution is the application of inducible/controllable suicide genes that can trigger cell death in unwanted cell types. These types of approaches have been demonstrated to increase the quality and safety of the resultant cell products. In this review, we will provide background on these approaches and how they could be used together with bioreactor technology to create effective bioprocesses for the generation of high quality and safe hPSCs for use in regenerative medicine approaches.
Collapse
Affiliation(s)
- Emilie Gysel
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
| | - Leila Larijani
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
| | - Michael S Kallos
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Roman J Krawetz
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
5
|
Wang L, Han Y, Gu Z, Han M, Hu C, Li Z. Boosting the therapy of glutamine-addiction glioblastoma by combining glutamine metabolism therapy with photo-enhanced chemodynamic therapy. Biomater Sci 2023; 11:6252-6266. [PMID: 37534821 DOI: 10.1039/d3bm00897e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
The complete treatment of high grade invasive glioblastoma (GBM) remains to be a great challenge, and it is of great importance to develop innovative therapeutic approaches. Herein, we found that GBM derived from U87 MG cells is a glutamine-addiction tumor, and jointly using glutamine-starvation therapy and photo-enhanced chemodynamic therapy (CDT) can significantly boost its therapy. We rationally fabricated tumor cell membrane coated Cu2-xSe nanoparticles (CS NPs) and an inhibitor of glutamine metabolism (Purpurin) for combined therapy, because glutamine rather than glucose plays a crucial role in the proliferation and growth of GBM cells, and serves as a precursor for the synthesis of glutathione (GSH). The resultant CS-P@CM NPs can be specifically delivered to the tumor site to inhibit glutamine metabolism in tumor cells, suppress tumor intracellular GSH, and increase H2O2 content, which benefit the CDT catalyzed by CS NPs. The cascade reaction can be further enhanced by irradiation with the second near-infrared (NIR-II) light at the maximum concentration of H2O2, which can be monitored by photoacoustic imaging. The NIR-II light irradiation can generate a large amount of reactive oxygen species (ROS) within a short time to kill tumor cells and enhance the CDT efficacy. This is the first work on the treatment of orthotopic malignant GBM through combined glutamine metabolism therapy and photo-enhanced CDT, and provides insights into the treatment of other solid tumors by modulating the metabolism of tumor cells.
Collapse
Affiliation(s)
- Ling Wang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Zhengpeng Gu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Mengxiao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Chunhong Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| |
Collapse
|
6
|
Oraee-Yazdani S, Tavanaei R, Rostami F, Hajarizadeh A, Mehrabadi M, Akhlaghpasand M, Tamaddon M, Khannejad S, Yazdani KO, Zali A. Suicide gene therapy using allogeneic adipose tissue-derived mesenchymal stem cell gene delivery vehicles in recurrent glioblastoma multiforme: a first-in-human, dose-escalation, phase I clinical trial. J Transl Med 2023; 21:350. [PMID: 37245011 DOI: 10.1186/s12967-023-04213-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/19/2023] [Indexed: 05/29/2023] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is associated with remarkably poor prognosis, and its treatment is challenging. This investigation aimed to evaluate the safety of suicide gene therapy using allogeneic adipose tissue-derived mesenchymal stem cells (ADSCs) carrying herpes simplex virus-thymidine kinase (HSV-TK) gene for the first time in patients with recurrent GBM. METHODS This study was a first-in-human, open-label, single-arm, phase I clinical trial with a classic 3 + 3 dose escalation design. Patients who did not undergo surgery for their recurrence were included and received this gene therapy protocol. Patients received the intratumoral stereotactic injection of ADSCs according to the assigned dose followed by prodrug administration for 14 days. The first dosing cohort (n = 3) received 2.5 × 105 ADSCs; the second dosing cohort (n = 3) received 5 × 105 ADSCs; the third dosing cohort (n = 6) received 10 × 105 ADSCs. The primary outcome measure was the safety profile of the intervention. RESULTS A total of 12 patients with recurrent GBM were recruited. The median follow-up was 16 (IQR, 14-18.5) months. This gene therapy protocol was safe and well tolerated. During the study period, eleven (91.7%) patients showed tumor progression, and nine (75.0%) died. The median overall survival (OS) was 16.0 months (95% CI 14.3-17.7) and the median progression-free survival (PFS) was 11.0 months (95% CI 8.3-13.7). A total of 8 and 4 patients showed partial response and stable disease, respectively. Moreover, significant changes were observed in volumetric analysis, peripheral blood cell counts, and cytokine profile. CONCLUSIONS The present clinical trial, for the first time, showed that suicide gene therapy using allogeneic ADSCs carrying the HSV-TK gene is safe in patients with recurrent GBM. Future phase II/III clinical trials with multiple arms are warranted to validate our findings and further investigate the efficacy of this protocol compared with standard therapy alone. TRIAL REGISTRATION Iranian Registry of Clinical Trials (IRCT), IRCT20200502047277N2. Registered 8 October 2020, https://www.irct.ir/ .
Collapse
Affiliation(s)
- Saeed Oraee-Yazdani
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran.
| | - Roozbeh Tavanaei
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
| | - Fatemeh Rostami
- Stem Cell Technology Research Center (STRC), Iran university of medical science (IUMS), Tehran, Iran
| | - Atieh Hajarizadeh
- Stem Cell Technology Research Center (STRC), Iran university of medical science (IUMS), Tehran, Iran
| | - Marzieh Mehrabadi
- Stem Cell Technology Research Center (STRC), Iran university of medical science (IUMS), Tehran, Iran
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mohammadhosein Akhlaghpasand
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
| | - Mona Tamaddon
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Samin Khannejad
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
| | - Kaveh Oraii Yazdani
- Department of cardiovascular diseases, Zahedan university of medical science, Zahedan, Iran
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
| |
Collapse
|
7
|
Ju H, Yu C, Liu W, Li HH, Fu Z, Wu YC, Gong PX, Li HJ. Polysaccharides from marine resources exhibit great potential in the treatment of tumor: A review. CARBOHYDRATE POLYMER TECHNOLOGIES AND APPLICATIONS 2023. [DOI: 10.1016/j.carpta.2023.100308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
|
8
|
Allami P, Heidari A, Rezaei N. The role of cell membrane-coated nanoparticles as a novel treatment approach in glioblastoma. Front Mol Biosci 2023; 9:1083645. [PMID: 36660431 PMCID: PMC9846545 DOI: 10.3389/fmolb.2022.1083645] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023] Open
Abstract
Glioblastoma multiform (GBM) is the most prevalent and deadliest primary brain malignancy in adults, whose median survival rate does not exceed 15 months after diagnosis. The conventional treatment of GBM, including maximal safe surgery followed by chemotherapy and radiotherapy, usually cannot lead to notable improvements in the disease prognosis and the tumor always recurs. Many GBM characteristics make its treatment challenging. The most important ones are the impermeability of the blood-brain barrier (BBB), preventing chemotherapeutic drugs from reaching in adequate amounts to the tumor site, intratumoral heterogeneity, and roles of glioblastoma stem cells (GSCs). To overcome these barriers, the recently-developed drug-carrying approach using nanoparticles (NPs) may play a significant role. NPs are tiny particles, usually less than 100 nm showing various diagnostic and therapeutic medical applications. In this regard, cell membrane (CM)-coated NPs demonstrated several promising effects in GBM in pre-clinical studies. They benefit from fewer adverse effects due to their specific targeting of tumor cells, biocompatibility because of their CM surfaces, prolonged half-life, easy penetrating of the BBB, and escaping from the immune reaction, making them an attractive option for GBM treatment. To date, CM-coated NPs have been applied to enhance the effectiveness of major therapeutic approaches in GBM treatment, including chemotherapy, immunotherapy, gene therapy, and photo-based therapies. Despite the promising results in pre-clinical studies regarding the effectiveness of CM-coated NPs in GBM, significant barriers like high expenses, complex preparation processes, and unknown long-term effects still hinder its mass production for the clinic. In this regard, the current study aims to provide an overview of different characteristics of CM-coated NPs and comprehensively investigate their application as a novel treatment approach in GBM.
Collapse
Affiliation(s)
- Pantea Allami
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Heidari
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Achón Buil B, Tackenberg C, Rust R. Editing a gateway for cell therapy across the blood-brain barrier. Brain 2022; 146:823-841. [PMID: 36397727 PMCID: PMC9976985 DOI: 10.1093/brain/awac393] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 11/19/2022] Open
Abstract
Stem cell therapy has been shown to improve stroke outcomes in animal models and is currently advancing towards clinical practice. However, uncertainty remains regarding the optimal route for cell delivery to the injured brain. Local intracerebral injections are effective in precisely delivering cells into the stroke cavity but carry the risk of damaging adjacent healthy tissue. Systemic endovascular injections, meanwhile, are minimally invasive, but most injected cells do not cross CNS barriers and become mechanically trapped in peripheral organs. Although the blood-brain barrier and the blood-CSF barrier tightly limit the entrance of cells and molecules into the brain parenchyma, immune cells can cross these barriers especially under pathological conditions, such as stroke. Deciphering the cell surface signature and the molecular mechanisms underlying this pathophysiological process holds promise for improving the targeted delivery of systemic injected cells to the injured brain. In this review, we describe experimental approaches that have already been developed in which (i) cells are either engineered to express cell surface proteins mimicking infiltrating immune cells; or (ii) cell grafts are preconditioned with hypoxia or incubated with pharmacological agents or cytokines. Modified cell grafts can be complemented with strategies to temporarily increase the permeability of the blood-brain barrier. Although these approaches could significantly enhance homing of stem cells into the injured brain, cell entrapment in off-target organs remains a non-negligible risk. Recent developments in safety-switch systems, which enable the precise elimination of transplanted cells on the administration of a drug, represent a promising strategy for selectively removing stem cells stuck in untargeted organs. In sum, the techniques described in this review hold great potential to substantially improve efficacy and safety of future cell therapies in stroke and may be relevant to other brain diseases.
Collapse
Affiliation(s)
- Beatriz Achón Buil
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Christian Tackenberg
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ruslan Rust
- Correspondence to: Ruslan Rust Institute for Regenerative Medicine Wagistrasse 12, 8952 Schlieren Zurich, Switzerland E-mail:
| |
Collapse
|
10
|
From Immunotoxins to Suicide Toxin Delivery Approaches: Is There a Clinical Opportunity? Toxins (Basel) 2022; 14:toxins14090579. [PMID: 36136517 PMCID: PMC9506092 DOI: 10.3390/toxins14090579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Suicide gene therapy is a relatively novel form of cancer therapy in which a gene coding for enzymes or protein toxins is delivered through targeting systems such as vesicles, nanoparticles, peptide or lipidic co-adjuvants. The use of toxin genes is particularly interesting since their catalytic activity can induce cell death, damaging in most cases the translation machinery (ribosomes or protein factors involved in protein synthesis) of quiescent or proliferating cells. Thus, toxin gene delivery appears to be a promising tool in fighting cancer. In this review we will give an overview, describing some of the bacterial and plant enzymes studied so far for their delivery and controlled expression in tumor models.
Collapse
|
11
|
Wu J, Li XY, Liang J, Fang DL, Yang ZJ, Wei J, Chen ZJ. Network pharmacological analysis of active components of Xiaoliu decoction in the treatment of glioblastoma multiforme. Front Genet 2022; 13:940462. [PMID: 36046228 PMCID: PMC9420933 DOI: 10.3389/fgene.2022.940462] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/29/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Glioblastoma multiforme (GBM) is the most aggressive primary nervous system brain tumor. There is still a lack of effective methods to control its progression and recurrence in clinical treatment. It is clinically found that Xiaoliu Decoction (XLD) has the effect of treating brain tumors and preventing tumor recurrence. However, its mechanism is still unclear. Methods: Search the Traditional Chinese Medicine System Pharmacology Database (TCSMP) for efficient substances for the treatment of XLD in the treatment of GBM, and target the targeted genes of the effective ingredients to construct a network. At the same time, download GBM-related gene expression data from the TCGA and GTEX databases, screen differential expression bases, and establish a drug target disease network. Through bioinformatics analysis, the target genes and shared genes of the selected Chinese medicines are analyzed. Finally, molecular docking was performed to further clarify the possibility of XLD in multiple GBMs. Results: We screened 894 differentially expressed genes in GBM, 230 XLD active ingredients and 169 predicted targets of its active compounds, of which 19 target genes are related to the differential expression of GBM. Bioinformatics analysis shows that these targets are closely related to cell proliferation, cell cycle regulation, and DNA synthesis. Finally, through molecular docking, it was further confirmed that Tanshinone IIA, the active ingredient of XLD, was tightly bound to key proteins. Conclusion: To sum up, the results of this study suggest that the mechanism of XLD in the treatment of GBM involves multiple targets and signal pathways related to tumorigenesis and development. This study not only provides a new theoretical basis for the treatment of glioblastoma multiforme with traditional Chinese medicine, but also provides a new idea for the research and development of targeted drugs for the treatment of glioblastoma multiforme.
Collapse
Affiliation(s)
- Ji Wu
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xue-Yu Li
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jing Liang
- Department of Pediatrics, The Second Affiliated Hospital of Xinjiang Medical University, Urumchi, China
| | - Da-Lang Fang
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- *Correspondence: Da-Lang Fang, ; Zhao-Jian Yang, ; Jie Wei, ; Zhi-Jun Chen,
| | - Zhao-Jian Yang
- Department of Neurosurgery, Red Cross Hospital of Yulin City, Yulin, China
- *Correspondence: Da-Lang Fang, ; Zhao-Jian Yang, ; Jie Wei, ; Zhi-Jun Chen,
| | - Jie Wei
- Department of Hematology, People’s Hospital of Baise, Baise, China
- *Correspondence: Da-Lang Fang, ; Zhao-Jian Yang, ; Jie Wei, ; Zhi-Jun Chen,
| | - Zhi-Jun Chen
- Department of Neurosurgery, Red Cross Hospital of Yulin City, Yulin, China
- *Correspondence: Da-Lang Fang, ; Zhao-Jian Yang, ; Jie Wei, ; Zhi-Jun Chen,
| |
Collapse
|
12
|
Thomas S, Bikeyeva V, Abdullah A, Radivojevic A, Abu Jad AA, Ravanavena A, Ravindra C, Igweonu-Nwakile EO, Ali S, Paul S, Yakkali S, Teresa Selvin S, Hamid P. Systematic Review of Pediatric Brain Tumors in Neurofibromatosis Type 1: Status of Gene Therapy. Cureus 2022; 14:e27963. [PMID: 36120213 PMCID: PMC9467501 DOI: 10.7759/cureus.27963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/13/2022] [Indexed: 11/18/2022] Open
Abstract
As oncology practice is rapidly shifting away from toxic chemotherapy, gene therapy provides a highly specific therapeutic approach for brain tumors. In this systematic review, we investigate gene therapy's status in pediatric brain tumors and future recommendations. The search was conducted systematically using PubMed, Cochrane, Google Scholar, and ClinicalTrials.gov databases. The field search used in the process was selected based on the keywords and Medical Subject Headings (MeSH), depending on the database used. We included cases of neurofibromatosis type 1 (NF1) brain tumors in all age groups with the additional inclusion of English language, free full text, articles published within the last 20 years, randomized controlled trials (RCTs), observational studies, systematic reviews, and meta-analyses. We excluded case reports, case studies, and editorials. The search identified a total of 1,213 articles from the databases. We included 19 studies with 16 narrative reviews, one systematic review, and two randomized clinical trials with 43 patients. After reviewing all data in the articles, we found that gene therapy can improve standard treatment efficacy when used as adjuvant therapy. It can be used to overcome barriers such as chemotherapy resistance by downregulating resistance genes. It is associated with mild toxicity when compared with other available treatment options, but given the overall poor prognosis in pediatric brain tumors, further studies are warranted.
Collapse
Affiliation(s)
- Sonu Thomas
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Viktoriya Bikeyeva
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ahmed Abdullah
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Aleksandra Radivojevic
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Anas A Abu Jad
- Behavioral Neurosciences and Psychology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Anvesh Ravanavena
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Chetna Ravindra
- General Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | | | - Safina Ali
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Salomi Paul
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Shreyas Yakkali
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sneha Teresa Selvin
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Pousette Hamid
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
13
|
Singh R, Kumar S. Cancer Targeting and Diagnosis: Recent Trends with Carbon Nanotubes. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2283. [PMID: 35808119 PMCID: PMC9268713 DOI: 10.3390/nano12132283] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023]
Abstract
Cancer belongs to a category of disorders characterized by uncontrolled cell development with the potential to invade other bodily organs, resulting in an estimated 10 million deaths globally in 2020. With advancements in nanotechnology-based systems, biomedical applications of nanomaterials are attracting increasing interest as prospective vehicles for targeted cancer therapy and enhancing treatment results. In this context, carbon nanotubes (CNTs) have recently garnered a great deal of interest in the field of cancer diagnosis and treatment due to various factors such as biocompatibility, thermodynamic properties, and varied functionalization. In the present review, we will discuss recent advancements regarding CNT contributions to cancer diagnosis and therapy. Various sensing strategies like electrochemical, colorimetric, plasmonic, and immunosensing are discussed in detail. In the next section, therapy techniques like photothermal therapy, photodynamic therapy, drug targeting, gene therapy, and immunotherapy are also explained in-depth. The toxicological aspect of CNTs for biomedical application will also be discussed in order to ensure the safe real-life and clinical use of CNTs.
Collapse
Affiliation(s)
- Ragini Singh
- College of Agronomy, Liaocheng University, Liaocheng 252059, China;
| | - Santosh Kumar
- Shandong Key Laboratory of Optical Communication Science and Technology, School of Physics Science and Information Technology, Liaocheng University, Liaocheng 252059, China
| |
Collapse
|
14
|
Tumor-Associated Macrophages in Gliomas—Basic Insights and Treatment Opportunities. Cancers (Basel) 2022; 14:cancers14051319. [PMID: 35267626 PMCID: PMC8909866 DOI: 10.3390/cancers14051319] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Macrophages are a specialized immune cell type found in both invertebrates and vertebrates. Versatile in functionality, macrophages carry out important tasks such as cleaning cellular debris in healthy tissues and mounting immune responses during infection. In many cancer types, macrophages make up a significant portion of tumor tissue, and these are aptly called tumor-associated macrophages. In gliomas, a group of primary brain tumors, these macrophages are found in very high frequency. Tumor-associated macrophages can promote glioma development and influence the outcome of various therapeutic regimens. At the same time, these cells provide various potential points of intervention for therapeutic approaches in glioma patients. The significance of tumor-associated macrophages in the glioma microenvironment and potential therapeutic targets are the focus of this review. Abstract Glioma refers to a group of primary brain tumors which includes glioblastoma (GBM), astrocytoma and oligodendroglioma as major entities. Among these, GBM is the most frequent and most malignant one. The highly infiltrative nature of gliomas, and their intrinsic intra- and intertumoral heterogeneity, pose challenges towards developing effective treatments. The glioma microenvironment, in addition, is also thought to play a critical role during tumor development and treatment course. Unlike most other solid tumors, the glioma microenvironment is dominated by macrophages and microglia—collectively known as tumor-associated macrophages (TAMs). TAMs, like their homeostatic counterparts, are plastic in nature and can polarize to either pro-inflammatory or immunosuppressive states. Many lines of evidence suggest that immunosuppressive TAMs dominate the glioma microenvironment, which fosters tumor development, contributes to tumor aggressiveness and recurrence and, very importantly, impedes the therapeutic effect of various treatment regimens. However, through the development of new therapeutic strategies, TAMs can potentially be shifted towards a proinflammatory state which is of great therapeutic interest. In this review, we will discuss various aspects of TAMs in the context of glioma. The focus will be on the basic biology of TAMs in the central nervous system (CNS), potential biomarkers, critical evaluation of model systems for studying TAMs and finally, special attention will be given to the potential targeted therapeutic options that involve the TAM compartment in gliomas.
Collapse
|
15
|
Barca C, Griessinger CM, Faust A, Depke D, Essler M, Windhorst AD, Devoogdt N, Brindle KM, Schäfers M, Zinnhardt B, Jacobs AH. Expanding Theranostic Radiopharmaceuticals for Tumor Diagnosis and Therapy. Pharmaceuticals (Basel) 2021; 15:13. [PMID: 35056071 PMCID: PMC8780589 DOI: 10.3390/ph15010013] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023] Open
Abstract
Radioligand theranostics (RT) in oncology use cancer-type specific biomarkers and molecular imaging (MI), including positron emission tomography (PET), single-photon emission computed tomography (SPECT) and planar scintigraphy, for patient diagnosis, therapy, and personalized management. While the definition of theranostics was initially restricted to a single compound allowing visualization and therapy simultaneously, the concept has been widened with the development of theranostic pairs and the combination of nuclear medicine with different types of cancer therapies. Here, we review the clinical applications of different theranostic radiopharmaceuticals in managing different tumor types (differentiated thyroid, neuroendocrine prostate, and breast cancer) that support the combination of innovative oncological therapies such as gene and cell-based therapies with RT.
Collapse
Affiliation(s)
- Cristina Barca
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
| | - Christoph M. Griessinger
- Roche Innovation Center, Early Clinical Development Oncology, Roche Pharmaceutical Research and Early Development, CH-4070 Basel, Switzerland;
| | - Andreas Faust
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
- Department of Nuclear Medicine, University Hospital Münster, D-48149 Münster, Germany
| | - Dominic Depke
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
| | - Markus Essler
- Department of Nuclear Medicine, University Hospital Bonn, D-53127 Bonn, Germany;
| | - Albert D. Windhorst
- Department Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands;
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, B-1090 Brussel, Belgium;
| | - Kevin M. Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 ORE, UK;
| | - Michael Schäfers
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
- Department of Nuclear Medicine, University Hospital Münster, D-48149 Münster, Germany
| | - Bastian Zinnhardt
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
- Department of Nuclear Medicine, University Hospital Münster, D-48149 Münster, Germany
- Biomarkers and Translational Technologies, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Andreas H. Jacobs
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
- Department of Geriatrics and Neurology, Johanniter Hospital, D-53113 Bonn, Germany
- Centre of Integrated Oncology, University Hospital Bonn, D-53127 Bonn, Germany
| |
Collapse
|
16
|
Głów D, Maire CL, Schwarze LI, Lamszus K, Fehse B. CRISPR-to-Kill (C2K)-Employing the Bacterial Immune System to Kill Cancer Cells. Cancers (Basel) 2021; 13:cancers13246306. [PMID: 34944926 PMCID: PMC8699370 DOI: 10.3390/cancers13246306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/01/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Reasoning that multiple DNA breaks will trigger programmed cell death, we generated lentiviral CRISPR-to-kill (C2K) vectors targeting highly repetitive SINE sequences for cancer gene therapy. In proof-of-concept experiments, C2K-Alu-vectors selectively killed human, but not murine cell lines, and efficiently inhibited the growth of patient-derived glioblastoma cell lines resistant to high-dose irradiation. In combination with tumor-targeting approaches, the C2K system might represent a promising tool for cancer gene therapy. Abstract CRISPR/Cas9 was described as a bacterial immune system that uses targeted introduction of DNA double-strand breaks (DSBs) to destroy invaders. We hypothesized that we can analogously employ CRISPR/Cas9 nucleases to kill cancer cells by inducing maximal numbers of DSBs in their genome and thus triggering programmed cell death. To do so, we generated CRISPR-to-kill (C2K) lentiviral particles targeting highly repetitive Short Interspersed Nuclear Element-Alu sequences. Our Alu-specific sgRNA has more than 15,000 perfectly matched target sites within the human genome. C2K-Alu-vectors selectively killed human, but not murine cell lines. More importantly, they efficiently inhibited the growth of cancer cells including patient-derived glioblastoma cell lines resistant to high-dose irradiation. Our data provide proof-of-concept for the potential of C2K as a novel treatment strategy overcoming common resistance mechanisms. In combination with tumor-targeting approaches, the C2K system might therefore represent a promising tool for cancer gene therapy.
Collapse
Affiliation(s)
- Dawid Głów
- Research Department, Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.G.); (L.I.S.)
| | - Cecile L. Maire
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (C.L.M.); (K.L.)
| | - Lea Isabell Schwarze
- Research Department, Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.G.); (L.I.S.)
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (C.L.M.); (K.L.)
| | - Boris Fehse
- Research Department, Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.G.); (L.I.S.)
- Correspondence: ; Tel.: +49-40-7410-55518; Fax: +49-40-7410-55468
| |
Collapse
|
17
|
Garcia-Fabiani MB, Haase S, Comba A, Carney S, McClellan B, Banerjee K, Alghamri MS, Syed F, Kadiyala P, Nunez FJ, Candolfi M, Asad A, Gonzalez N, Aikins ME, Schwendeman A, Moon JJ, Lowenstein PR, Castro MG. Genetic Alterations in Gliomas Remodel the Tumor Immune Microenvironment and Impact Immune-Mediated Therapies. Front Oncol 2021; 11:631037. [PMID: 34168976 PMCID: PMC8217836 DOI: 10.3389/fonc.2021.631037] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
High grade gliomas are malignant brain tumors that arise in the central nervous system, in patients of all ages. Currently, the standard of care, entailing surgery and chemo radiation, exhibits a survival rate of 14-17 months. Thus, there is an urgent need to develop new therapeutic strategies for these malignant brain tumors. Currently, immunotherapies represent an appealing approach to treat malignant gliomas, as the pre-clinical data has been encouraging. However, the translation of the discoveries from the bench to the bedside has not been as successful as with other types of cancer, and no long-lasting clinical benefits have been observed for glioma patients treated with immune-mediated therapies so far. This review aims to discuss our current knowledge about gliomas, their molecular particularities and the impact on the tumor immune microenvironment. Also, we discuss several murine models used to study these therapies pre-clinically and how the model selection can impact the outcomes of the approaches to be tested. Finally, we present different immunotherapy strategies being employed in clinical trials for glioma and the newest developments intended to harness the immune system against these incurable brain tumors.
Collapse
Affiliation(s)
- Maria B. Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Stephen Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Brandon McClellan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Immunology graduate program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Kaushik Banerjee
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Faisal Syed
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Antonela Asad
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nazareno Gonzalez
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marisa E. Aikins
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
18
|
Calinescu AA, Kauss MC, Sultan Z, Al-Holou WN, O'Shea SK. Stem cells for the treatment of glioblastoma: a 20-year perspective. CNS Oncol 2021; 10:CNS73. [PMID: 34006134 PMCID: PMC8162173 DOI: 10.2217/cns-2020-0026] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma, the deadliest form of primary brain tumor, remains a disease without cure. Treatment resistance is in large part attributed to limitations in the delivery and distribution of therapeutic agents. Over the last 20 years, numerous preclinical studies have demonstrated the feasibility and efficacy of stem cells as antiglioma agents, leading to the development of trials to test these therapies in the clinic. In this review we present and analyze these studies, discuss mechanisms underlying their beneficial effect and highlight experimental progress, limitations and the emergence of promising new therapeutic avenues. We hope to increase awareness of the advantages brought by stem cells for the treatment of glioblastoma and inspire further studies that will lead to accelerated implementation of effective therapies. Glioblastoma is the deadliest and most common form of brain tumor, for which there is no cure. It is very difficult to deliver medicine to the tumor cells, because they spread out widely into the normal brain, and local blood vessels represent a barrier that most medicines cannot cross. It was shown, in many studies over the last 20 years, that stem cells are attracted toward the tumor and that they can deliver many kinds of therapeutic agents directly to brain cancer cells and shrink the tumor. In this review we analyze these studies and present new discoveries that can be used to make stem cell therapies for glioblastoma more effective to prolong the life of patients with brain tumors.
Collapse
Affiliation(s)
| | - McKenzie C Kauss
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,College of Literature Science & Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zain Sultan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,College of Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wajd N Al-Holou
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sue K O'Shea
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
19
|
Sawdon AJ, Zhang J, Peng S, Alyami EM, Peng CA. Polymeric Nanovectors Incorporated with Ganciclovir and HSV- tk Encoding Plasmid for Gene-Directed Enzyme Prodrug Therapy. Molecules 2021; 26:molecules26061759. [PMID: 33801024 PMCID: PMC8003905 DOI: 10.3390/molecules26061759] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/14/2021] [Accepted: 03/19/2021] [Indexed: 11/16/2022] Open
Abstract
In the area of gene-directed enzyme prodrug therapy (GDEPT), using herpes simplex virus thymidine kinase (HSV-tk) paired with prodrug ganciclovir (GCV) for cancer treatment has been extensively studied. It is a process involved with two steps whereby the gene (HSV-tk) is first delivered to malignant cells. Afterward, non-toxic GCV is administered to that site and activated to cytotoxic ganciclovir triphosphate by HSV-tk enzyme expressed exogenously. In this study, we presented a one-step approach that both gene and prodrug were delivered at the same time by incorporating them with polymeric micellar nanovectors. GCV was employed as an initiator in the ring-opening polymerization of ε-caprolactone (ε-CL) to synthesize hydrophobic GCV-poly(caprolactone) (GCV-PCL), which was furthered grafted with hydrophilic chitosan to obtain amphiphilic polymer (GCV-PCL-chitosan) for the fabrication of self-assembled micellar nanoparticles. The synthesized amphiphilic polymer was characterized using Fourier transform infrared spectroscopy and proton nuclear magnetic resonance. Micellar prodrug nanoparticles were analyzed by dynamic light scattering, zeta potential, critical micelle concentration, and transmission electron microscopy. Polymeric prodrug micelles with optimal features incorporated with HSV-tk encoding plasmids were cultivated with HT29 colorectal cancer cells and anticancer effectiveness was determined. Our results showed that prodrug GCV and HSV-tk cDNA encoded plasmid incorporated in GCV-PCL-chitosan polymeric nanocarriers could be delivered in a one-step manner to HT-29 cells and triggered high cytotoxicity.
Collapse
Affiliation(s)
- Alicia J. Sawdon
- Department of Chemical Engineering, Michigan Technological University, Houghton, MI 49931, USA;
| | - Jun Zhang
- Department of Chemical and Biological Engineering, University of Idaho, Moscow, ID 83844, USA; (J.Z.); (S.P.); (E.M.A.)
| | - Sarah Peng
- Department of Chemical and Biological Engineering, University of Idaho, Moscow, ID 83844, USA; (J.Z.); (S.P.); (E.M.A.)
| | - Esmael M. Alyami
- Department of Chemical and Biological Engineering, University of Idaho, Moscow, ID 83844, USA; (J.Z.); (S.P.); (E.M.A.)
| | - Ching-An Peng
- Department of Chemical and Biological Engineering, University of Idaho, Moscow, ID 83844, USA; (J.Z.); (S.P.); (E.M.A.)
- Correspondence: ; Tel.: +1-208-885-7461
| |
Collapse
|
20
|
Chelliah SS, Paul EAL, Kamarudin MNA, Parhar I. Challenges and Perspectives of Standard Therapy and Drug Development in High-Grade Gliomas. Molecules 2021; 26:1169. [PMID: 33671796 PMCID: PMC7927069 DOI: 10.3390/molecules26041169] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022] Open
Abstract
Despite their low incidence rate globally, high-grade gliomas (HGG) remain a fatal primary brain tumor. The recommended therapy often is incapable of resecting the tumor entirely and exclusively targeting the tumor leads to tumor recurrence and dismal prognosis. Additionally, many HGG patients are not well suited for standard therapy and instead, subjected to a palliative approach. HGG tumors are highly infiltrative and the complex tumor microenvironment as well as high tumor heterogeneity often poses the main challenges towards the standard treatment. Therefore, a one-fit-approach may not be suitable for HGG management. Thus, a multimodal approach of standard therapy with immunotherapy, nanomedicine, repurposing of older drugs, use of phytochemicals, and precision medicine may be more advantageous than a single treatment model. This multimodal approach considers the environmental and genetic factors which could affect the patient's response to therapy, thus improving their outcome. This review discusses the current views and advances in potential HGG therapeutic approaches and, aims to bridge the existing knowledge gap that will assist in overcoming challenges in HGG.
Collapse
Affiliation(s)
- Shalini Sundramurthi Chelliah
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Ervin Ashley Lourdes Paul
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| | - Muhamad Noor Alfarizal Kamarudin
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| | - Ishwar Parhar
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| |
Collapse
|
21
|
Xu X, Chen W, Zhu W, Chen J, Ma B, Ding J, Wang Z, Li Y, Wang Y, Zhang X. Adeno-associated virus (AAV)-based gene therapy for glioblastoma. Cancer Cell Int 2021; 21:76. [PMID: 33499886 PMCID: PMC7836184 DOI: 10.1186/s12935-021-01776-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/16/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM) is the most common and malignant Grade IV primary craniocerebral tumor caused by glial cell carcinogenesis with an extremely poor median survival of 12–18 months. The current standard treatments for GBM, including surgical resection followed by chemotherapy and radiotherapy, fail to substantially prolong survival outcomes. Adeno-associated virus (AAV)-mediated gene therapy has recently attracted considerable interest because of its relatively low cytotoxicity, poor immunogenicity, broad tissue tropism, and long-term stable transgene expression. Furthermore, a range of gene therapy trials using AAV as vehicles are being investigated to thwart deadly GBM in mice models. At present, AAV is delivered to the brain by local injection, intracerebroventricular (ICV) injection, or systematic injection to treat experimental GBM mice model. In this review, we summarized the experimental trials of AAV-based gene therapy as GBM treatment and compared the advantages and disadvantages of different AAV injection approaches. We systematically introduced the prospect of the systematic injection of AAV as an approach for AAV-based gene therapy for GBM.
Collapse
Affiliation(s)
- Xin Xu
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| | - Wenli Chen
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenjun Zhu
- Department of Laboratory Medicine, The Second People's Hospital of Lianyungang, Lianyungang, 222006, China
| | - Jing Chen
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Bin Ma
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Jianxia Ding
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Zaichuan Wang
- School of Medicine, Yangzhou University, Yangzhou, 225600, China
| | - Yifei Li
- School of Medicine, Yangzhou University, Yangzhou, 225600, China
| | - Yeming Wang
- Department of Hepatobiliary Surgery, The Second People's Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China.
| | - Xiaochun Zhang
- School of Medicine, Yangzhou University, Yangzhou, 225600, China. .,Department of Oncology, Yangzhou Traditional Chinese Medical Hospital, Yangzhou, 225600, Jiangsu, China.
| |
Collapse
|
22
|
Yahya EB, Alqadhi AM. Recent trends in cancer therapy: A review on the current state of gene delivery. Life Sci 2021; 269:119087. [PMID: 33476633 DOI: 10.1016/j.lfs.2021.119087] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/04/2021] [Accepted: 01/12/2021] [Indexed: 02/06/2023]
Abstract
Cancer treatment has been always considered one of the most critical and vital themes of clinical issues. Many approaches have been developed, depending on the type and the stage of tumor. Gene therapy has the potential to revolutionize different cancer therapy. With the advent of recent bioinformatics technologies and genetic science, it become possible to identify, diagnose and determine the potential treatment using the technology of gene delivery. Several approaches have been developed and experimented in vitro and vivo for cancer therapy including: naked nucleic acids based therapy, targeting micro RNAs, oncolytic virotherapy, suicide gene based therapy, targeting telomerase, cell mediated gene therapy, and CRISPR/Cas9 based therapy. In this review, we present a straightforward introduction to cancer biology and occurrence, highlighting different viral and non-viral gene delivery systems for gene therapy and critically discussed the current and various strategies for cancer gene therapy.
Collapse
Affiliation(s)
- Esam Bashir Yahya
- School of Industrial Technology, Universiti Sains Malaysia, Penang 11800, Malaysia.
| | | |
Collapse
|
23
|
Das MK, Lunavat TR, Miletic H, Hossain JA. The Potentials and Pitfalls of Using Adult Stem Cells in Cancer Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1326:139-157. [PMID: 33615422 DOI: 10.1007/5584_2021_619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Stem cells play a pivotal role in the developmental stages of an organism and in adulthood as well. Therefore, it is not surprising that stem cells constitute a focus of extensive research. Indeed, several decades of stem cell research have tremendously increased our knowledge on the mechanistic understandings of stem cell biology. Interestingly, revealing the fundamental principles of stem cell biology has also fostered its application for therapeutic purposes. Many of the attributes that the stem cells possess, some of which are unique, allow multifaceted exploitation of stem cells in the treatment of various diseases. Cancer, the leading cause of mortality worldwide, is one of the disease groups that has been benefited by the potentials of therapeutic applications of the stem cells. While the modi operandi of how stem cells contribute to cancer treatment are many-sided, two major principles can be conceived. One mode involves harnessing the regenerative power of the stem cells to promote the generation of blood-forming cells in cancer patients after cytotoxic regimens. A totally different kind of utility of stem cells has been exercised in another mode where the stem cells can potentially deliver a plethora of anti-cancer therapeutics in a tumor-specific manner. While both these approaches can improve the treatment of cancer patients, there exist several issues that warrant further research. This review summarizes the basic principles of the utility of the stem cells in cancer treatment along with the current trends and pinpoints the major obstacles to focus on in the future for further improvement.
Collapse
Affiliation(s)
- Mrinal K Das
- Department of Molecular Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Taral R Lunavat
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Hrvoje Miletic
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Jubayer A Hossain
- Department of Biomedicine, University of Bergen, Bergen, Norway. .,Department of Pathology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
24
|
Kenarkoohi A, Bamdad T, Soleimani M, Soleimanjahi H, Fallah A, Falahi S. HSV-TK Expressing Mesenchymal Stem Cells Exert Inhibitory Effect on Cervical Cancer Model. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2020; 9:146-154. [PMID: 32934952 PMCID: PMC7489112 DOI: 10.22088/ijmcm.bums.9.2.146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 06/23/2020] [Indexed: 02/06/2023]
Abstract
A growing area of research is focused on cancer therapy, and new therapeutic approaches are welcomed. Mesenchymal stem cell (MSC)-based gene therapy is a promising strategy in oncology. Intrinsic tropism and migration to tumor microenvironment with off lights are attractive features of this type of cell carrier. In this way, suicide genes have also found a good platform for better performance and have shown a stronger anti-tumor mechanism by riding on mesenchymal cells. In this study, we investigated the anti-tumor activity of intratumoral injected MSCs transduced with a lentivector expressing the HSV/TK in a mouse cervical cancer model. Following the injection of MSCs transduced with lentivector carrying TK, MSCs alone or PBS into the mice tumor, ganciclovir was administered intraperitoneally during 14 days, and tumor size, survival time, natural killer (NK) cells and cytotoxic T lymphocyte (CTL) activities were assessed. We demonstrated that combination of suicide therapy and cell therapy leading m,to successful tumor inhibition. Significant reduction in tumor size was detected in test group in comparison with controls. Also, potent antitumor NK and CTL activity was seen in treatment group in comparison with controls. Our data demonstrated that the mesenchymal cells expressing TK had inhibitory effect on cervical cancer model.
Collapse
Affiliation(s)
- Azra Kenarkoohi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Department of Microbiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Taravat Bamdad
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Shahab Falahi
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|