1
|
Stepanenko AA, Sosnovtseva AO, Valikhov MP, Vasiukova AA, Abramova OV, Lipatova AV, Yusubalieva GM, Chekhonin VP. Comparison of the L3-23K and L5-Fiber Regions for Arming the Oncolytic Adenovirus Ad5-Delta-24-RGD with Reporter and Therapeutic Transgenes. Int J Mol Sci 2025; 26:3700. [PMID: 40332328 PMCID: PMC12027834 DOI: 10.3390/ijms26083700] [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: 03/10/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
The insertion of a transgene downstream of the L3-23K or L5-Fiber region was reported as a vital strategy for arming E3 non-deleted oncolytic adenoviruses. However, depending on the percentage of codons with G/C at the third base position (GC3%) and the type of splicing acceptor, an insert downstream of the L5-Fiber region may substantially affect virus fitness. Since the insertion of transgenes downstream of the L3-23K and L5-Fiber regions has never been compared in terms of their expression levels and impact on virus fitness, we assessed the total virus yield, cytolytic efficacy, and plaque size of Ad5-delta-24-RGD (Ad5Δ24RGD) armed with EGFP, FLuc, the suppressor of RNA silencing p19, soluble wild-type human/mouse and high-affinity human programmed cell death receptor-1 (PD-1/PDCD1) ectodomains, and soluble human hyaluronidase PH20/SPAM1. The insertion of transgenes downstream of the L3-23K region ensures their production at considerably higher levels. However, the insertion of transgenes downstream of either region differentially and unpredictably affects the oncolytic potency of Ad5Δ24RGD, which cannot be explained by GC3% or expression level alone. Surprisingly, the expression of the human and mouse PD-1 ectodomains with 83.1% and 70.1% GC3%, respectively, does not affect cytolytic efficacy but increases the plaque size in a cell line-dependent manner.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, Ministry of Health of the Russian Federation, 119034 Moscow, Russia; (A.O.S.); (M.P.V.); (A.A.V.); (O.V.A.); (V.P.C.)
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| | - Anastasiia O. Sosnovtseva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, Ministry of Health of the Russian Federation, 119034 Moscow, Russia; (A.O.S.); (M.P.V.); (A.A.V.); (O.V.A.); (V.P.C.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
- Laboratory of Regulation of Intracellular Proteolysis, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Marat P. Valikhov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, Ministry of Health of the Russian Federation, 119034 Moscow, Russia; (A.O.S.); (M.P.V.); (A.A.V.); (O.V.A.); (V.P.C.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Anastasiia A. Vasiukova
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, Ministry of Health of the Russian Federation, 119034 Moscow, Russia; (A.O.S.); (M.P.V.); (A.A.V.); (O.V.A.); (V.P.C.)
| | - Olga V. Abramova
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, Ministry of Health of the Russian Federation, 119034 Moscow, Russia; (A.O.S.); (M.P.V.); (A.A.V.); (O.V.A.); (V.P.C.)
| | - Anastasiia V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Gaukhar M. Yusubalieva
- Laboratory of Cell Technologies, Federal Scientific and Clinical Center for Specialized Types of Medical Care and Medical Technologies, Federal Medical and Biological Agency of the Russian Federation, 115682 Moscow, Russia;
- Laboratory of Immunotherapy of Solid Tumors, Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of the Russian Federation, 117513 Moscow, Russia
- Laboratory of Molecular Mechanisms of Regeneration, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, Ministry of Health of the Russian Federation, 119034 Moscow, Russia; (A.O.S.); (M.P.V.); (A.A.V.); (O.V.A.); (V.P.C.)
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| |
Collapse
|
2
|
Qiu X, Wang F, Gao X, Zhang H, Wu H, Gong X, Lin J. Detection of hyaluronidase in urine using hyaluronic acid-coated silver nanoparticles-based surface-enhanced Raman spectroscopy for the diagnosis of bladder cancer. Talanta 2025; 294:128139. [PMID: 40262342 DOI: 10.1016/j.talanta.2025.128139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025]
Abstract
Bladder cancer, a malignancy of the urinary tract, has shown a rising incidence rate in recent years. Current diagnostic methods often suffer from issues such as invasiveness, high costs, or insufficient sensitivity, creating an urgent need for a fast, simple, and non-invasive diagnostic approach. In this study, a novel diagnostic method for bladder cancer is proposed, using hyaluronic acid-coated silver nanoparticles (HA-AgNPs) as the substrate for surface-enhanced Raman scattering (SERS) to detect hyaluronidase (HAase), a biomarker for bladder cancer, in urine. This method is based on the hydrolysis of hyaluronic acid on HA-AgNPs by HAase, which generates oligomers and causes the breakdown of HA-AgNPs into smaller nanoparticles. The formation of oligomers enhances the surface shielding effect of the silver nanoparticles, promoting aggregation between particles and significantly weakening the SERS signal. By detecting changes in the SERS signal using Rhodamine (R6G), HAase was quantified. The experimental results show that there is a good linear relationship (R2 = 0.9991) between the SERS signal and the HAase concentration in the range of 3 × 10-4 U/mL to 3 × 101 U/mL. The method demonstrates high sensitivity and can effectively detect HAase concentration in urine. Combining principal component analysis (PCA) with linear discriminant analysis (LDA), effective classification of SERS spectra from 42 normal and 40 bladder cancer urine samples was achieved. Experimental results show that the sensitivity and specificity of this algorithm in distinguishing between the normal and bladder cancer groups reached 95.2 % and 95 %, respectively.
Collapse
Affiliation(s)
- Xinhao Qiu
- School of Opto-electronic and Communication Engineering, Xiamen University of Technology, Xiamen, Fujian, China
| | - Fuqiang Wang
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Xiamen, 361004, Fujian, China
| | - Xingen Gao
- School of Opto-electronic and Communication Engineering, Xiamen University of Technology, Xiamen, Fujian, China
| | - Hongyi Zhang
- School of Opto-electronic and Communication Engineering, Xiamen University of Technology, Xiamen, Fujian, China
| | - Haochen Wu
- Department of Hepatobiliary Surgery, Xiamen Hospital, Beijing University of Chinese Medicine, Xiamen, Fujian, China; Department of Hepatobiliary Surgery, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian, China
| | - Xianqiong Gong
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Xiamen, 361004, Fujian, China
| | - Juqiang Lin
- School of Opto-electronic and Communication Engineering, Xiamen University of Technology, Xiamen, Fujian, China.
| |
Collapse
|
3
|
Jang HJ, Park JW. Microenvironmental Drivers of Glioma Progression. Int J Mol Sci 2025; 26:2108. [PMID: 40076738 PMCID: PMC11900340 DOI: 10.3390/ijms26052108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Gliomas, particularly glioblastoma (GBM), are among the most challenging brain tumors due to their complex and dynamic tumor microenvironment (TME). The TME plays a pivotal role in tumor progression, immune evasion, and resistance to therapy through intricate interactions among glioma cells, immune components, neurons, astrocytes, the extracellular matrix, and the blood-brain barrier. Targeting the TME has demonstrated potential, with immunotherapies such as checkpoint inhibitors and neoadjuvant therapies enhancing immune responses. Nonetheless, overcoming the immunosuppressive landscape and metabolic adaptations continues to pose significant challenges. This review explores the diverse cellular and molecular mechanisms that shape the glioma TME. A deeper understanding of these mechanisms holds promise for providing novel therapeutic opportunities to improve glioma treatment outcomes.
Collapse
Affiliation(s)
- Hyun Ji Jang
- Department of Life Sciences, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
| | - Jong-Whi Park
- Department of Life Sciences, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
4
|
Sukegawa M, Miyagawa Y, Kuroda S, Yamazaki Y, Yamamoto M, Adachi K, Sato H, Sato Y, Taniai N, Yoshida H, Umezawa A, Sakai M, Okada T. Mesenchymal stem cell origin contributes to the antitumor effect of oncolytic virus carriers. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200896. [PMID: 39554905 PMCID: PMC11568361 DOI: 10.1016/j.omton.2024.200896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/18/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Oncolytic virotherapy shows promise as a cancer treatment approach; however, its systemic application is hindered by antibody neutralization. This issue can be overcome by using mesenchymal stem cells (MSCs) as carrier cells for oncolytic viruses (OVs). However, it remains elusive whether MSC source influences the antitumor effect. Here, we demonstrate that their source affects the migration ability and oncolytic activity of OV-loaded MSCs. Among human MSCs derived from different tissues, bone marrow-derived MSCs (BMMSCs) showed a high migration ability toward cancer cells in two- and three-dimensional MSC-cancer cell co-culture models. Comprehensive gene expression and Gene Ontology-based functional analyses suggested that genes involved in cell migration and cytokine response influence the cancer-specific tropism of BMMSCs. Furthermore, MSC origin affected the susceptibility to OVs, including cytotoxicity resistance and OV release from MSCs. MSC-mediated OV delivery significantly increased the viral spread and antitumor activity compared with delivery by OVs alone, and OV-loaded BMMSCs demonstrated the most potent antitumor effect among OV-loaded MSCs. Our results offer promising insights into cancer gene therapy with carrier cells and can help with the selection of an appropriate MSC source for MSC-based OV therapy.
Collapse
Affiliation(s)
- Makoto Sukegawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Department of Gastrointestinal Surgery, Graduate School of Medicine, Nippon Medical School Musashikosugi Hospital, Kawasaki, Japan
- Department of Surgery, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yoshitaka Miyagawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Seiji Kuroda
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yoshiyuki Yamazaki
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Motoko Yamamoto
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Kumi Adachi
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Hirofumi Sato
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yuriko Sato
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Nobuhiko Taniai
- Department of Gastrointestinal Surgery, Graduate School of Medicine, Nippon Medical School Musashikosugi Hospital, Kawasaki, Japan
| | - Hiroshi Yoshida
- Department of Surgery, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Akihiro Umezawa
- Center for Regenerative Medicine, National Center for Child Health and Development Research Institute, Tokyo, Japan
| | - Mashito Sakai
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Omole AO, Zhao Z, Chang-Liao S, de Oliveira JFA, Boone CE, Sutorus L, Sack M, Varner J, Fiering SN, Steinmetz NF. Virus nanotechnology for intratumoural immunotherapy. NATURE REVIEWS BIOENGINEERING 2024; 2:916-929. [PMID: 39698315 PMCID: PMC11655125 DOI: 10.1038/s44222-024-00231-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 12/20/2024]
Abstract
Viruses can be designed to be tools and carrier vehicles for intratumoural immunotherapy. Their nanometre-scale size and shape allow for functionalization with or encapsulation of medical cargoes and tissue-specific ligands. Importantly, immunotherapies may particularly benefit from the inherent immunomodulatory properties of viruses. For example, mammalian viruses have already been tested for oncolytic virotherapy, and bacteriophages and plant viruses can be engineered for immunotherapeutic treatment approaches. In this Review, we discuss how viruses - including oncolytic viruses, immunomodulatory plant viruses and bacteriophages - and virus-like particles can be designed for intratumoural immunotherapy to elicit anti-tumour immunity and induce systemic anti-tumour responses at distant non-injected sites. We further highlight the engineering of viruses and virus-like particles as drug-delivery systems, and outline key translational challenges and clinical opportunities.
Collapse
Affiliation(s)
- Anthony O. Omole
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Zhongchao Zhao
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Sabrina Chang-Liao
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Jessica Fernanda Affonso de Oliveira
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Christine E. Boone
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Lucas Sutorus
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | | | - Judith Varner
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
- Center for Engineering in Cancer, Institute of Engineering Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Steven N. Fiering
- Department of Microbiology and Immunology, Dartmouth Cancer Center, Dartmouth Geisel School of Medicine and Dartmouth-Hitchock Health, Lebanon, NH, USA
| | - Nicole F. Steinmetz
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Center for Engineering in Cancer, Institute of Engineering Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
6
|
Alsharabasy AM, Pandit A. Hyaluronan-Based Hydrogels for 3D Modeling of Tumor Tissues. Tissue Eng Part C Methods 2024; 30:452-499. [PMID: 39345138 DOI: 10.1089/ten.tec.2024.0271] [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] [Indexed: 10/01/2024] Open
Abstract
Although routine two-dimensional (2D) cell culture techniques have advanced basic cancer research owing to their simplicity, cost-effectiveness, and reproducibility, they have limitations that necessitate the development of advanced three-dimensional (3D) tumor models that better recapitulate the tumor microenvironment. Various biomaterials have been used to establish these 3D models, enabling the study of cancer cell behavior within different matrices. Hyaluronic acid (HA), a key component of the extracellular matrix (ECM) in tumor tissues, has been widely studied and employed in the development of multiple cancer models. This review first examines the role of HA in tumors, including its function as an ECM component and regulator of signaling pathways that affect tumor progression. It then explores HA-based models for various cancers, focusing on HA as a central component of the 3D matrix and its mobilization within the matrix for targeted studies of cell behavior and drug testing. The tumor models discussed included those for breast cancer, glioblastoma, fibrosarcoma, gastric cancer, hepatocellular carcinoma, and melanoma. The review concludes with a discussion of future prospects for developing more robust and high-throughput HA-based models to more accurately mimic the tumor microenvironment and improve drug testing. Impact Statement This review underscores the transformative potential of hyaluronic acid (HA)-based hydrogels in developing advanced tumor models. By exploring HA's dual role as a critical extracellular matrix component and a regulator of cancer cell dynamics, we highlight its unique contributions to replicating the tumor microenvironment. The recent advancements in HA-based models provide new opportunities for more accurate studies of cancer cell behavior and drug responses. Looking ahead, these innovations pave the way for high-throughput, biomimetic platforms that could revolutionize drug testing and accelerate the discovery of effective cancer therapies.
Collapse
Affiliation(s)
- Amir M Alsharabasy
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
7
|
Neves ER, Anand A, Mueller J, Remy RA, Xu H, Selting KA, Sarkaria JN, Harley BAC, Pedron-Haba S. Targeting glioblastoma tumor hyaluronan to enhance therapeutic interventions that regulate metabolic cell properties. ADVANCED THERAPEUTICS 2024; 7:2400041. [PMID: 40248278 PMCID: PMC12002556 DOI: 10.1002/adtp.202400041] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Indexed: 04/19/2025]
Abstract
Despite extensive advances in cancer research, glioblastoma (GBM) still remains a very locally invasive and thus challenging tumor to treat, with a poor median survival. Tumor cells remodel their microenvironment and utilize extracellular matrix to promote invasion and therapeutic resistance. We aim here to determine how GBM cells exploit hyaluronan (HA) to maintain proliferation using ligand-receptor dependent and ligand-receptor independent signaling. We use tissue engineering approaches to recreate the three-dimensional tumor microenvironment in vitro, then analyze shifts in metabolism, hyaluronan secretion, HA molecular weight distribution, as well as hyaluronan synthetic enzymes (HAS) and hyaluronidases (HYAL) activity in an array of patient derived xenograft GBM cells. We reveal that endogenous HA plays a role in mitochondrial respiration and cell proliferation in a tumor subtype dependent manner. We propose a tumor specific combination treatment of HYAL and HAS inhibitors to disrupt the HA stabilizing role in GBM cells. Taken together, these data shed light on the dual metabolic and ligand - dependent signaling roles of hyaluronan in glioblastoma.
Collapse
Affiliation(s)
- Edward R Neves
- Department of Chemical and Biomolecular Engineering, Carl R Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, 1206 W Gregory Dr., Urbana IL 61801, USA
| | - Achal Anand
- Department of Chemical and Biomolecular Engineering, Carl R Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, 1206 W Gregory Dr., Urbana IL 61801, USA
| | - Joseph Mueller
- Department of Chemical and Biomolecular Engineering, Carl R Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, 1206 W Gregory Dr., Urbana IL 61801, USA
| | - Roddel A Remy
- Materials Research Laboratory, University of Illinois Urbana-Champaign, 104 S Goodwin Ave MC-230, Urbana, IL 61801, USA
| | - Hui Xu
- Tumor Engineering and Phenotyping, Cancer Center at Illinois, University of Illinois Urbana Champaign, Beckman Institute, 405 N. Mathews, Urbana, IL 61801, USA
| | - Kim A Selting
- Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois Urbana-Champaign, 2001 S Lincoln Ave. Urbana, IL 61802, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Brendan AC Harley
- Dept. of Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, Cancer Center at Illinois, University of Illinois Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801
| | - Sara Pedron-Haba
- Dept. of Chemical and Biomolecular Engineering, Carle Illinois College of Medicine, Carle Illinois College of Medicine, Carl R. Woese Institute for Genomic Biology, Cancer Center at Illinois, University of Illinois Urbana-Champaign, 1206 W Gregory Dr. Urbana, IL 61801, USA
| |
Collapse
|
8
|
Mongeon B, Hébert-Doutreloux J, Surendran A, Karimi E, Fiset B, Quail DF, Walsh LA, Jenner AL, Craig M. Spatial computational modelling illuminates the role of the tumour microenvironment for treating glioblastoma with immunotherapies. NPJ Syst Biol Appl 2024; 10:91. [PMID: 39155294 PMCID: PMC11330976 DOI: 10.1038/s41540-024-00419-4] [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: 11/23/2023] [Accepted: 08/07/2024] [Indexed: 08/20/2024] Open
Abstract
Glioblastoma is the most common and deadliest brain tumour in adults, with a median survival of 15 months under the current standard of care. Immunotherapies like immune checkpoint inhibitors and oncolytic viruses have been extensively studied to improve this endpoint. However, most thus far have failed. To improve the efficacy of immunotherapies to treat glioblastoma, new single-cell imaging modalities like imaging mass cytometry can be leveraged and integrated with computational models. This enables a better understanding of the tumour microenvironment and its role in treatment success or failure in this hard-to-treat tumour. Here, we implemented an agent-based model that allows for spatial predictions of combination chemotherapy, oncolytic virus, and immune checkpoint inhibitors against glioblastoma. We initialised our model with patient imaging mass cytometry data to predict patient-specific responses and found that oncolytic viruses drive combination treatment responses determined by intratumoral cell density. We found that tumours with higher tumour cell density responded better to treatment. When fixing the number of cancer cells, treatment efficacy was shown to be a function of CD4 + T cell and, to a lesser extent, of macrophage counts. Critically, our simulations show that care must be put into the integration of spatial data and agent-based models to effectively capture intratumoral dynamics. Together, this study emphasizes the use of predictive spatial modelling to better understand cancer immunotherapy treatment dynamics, while highlighting key factors to consider during model design and implementation.
Collapse
Affiliation(s)
- Blanche Mongeon
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, QC, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada
| | | | - Anudeep Surendran
- Center for Advanced Systems Understanding, Görlitz, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Elham Karimi
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Benoit Fiset
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montréal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, QC, Canada
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Adrianne L Jenner
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Morgan Craig
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, QC, Canada.
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
9
|
Di Vito A, Donato A, Bria J, Conforti F, La Torre D, Malara N, Donato G. Extracellular Matrix Structure and Interaction with Immune Cells in Adult Astrocytic Tumors. Cell Mol Neurobiol 2024; 44:54. [PMID: 38969910 PMCID: PMC11226480 DOI: 10.1007/s10571-024-01488-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/21/2024] [Indexed: 07/07/2024]
Abstract
The extracellular matrix (ECM) is a dynamic set of molecules produced by the cellular component of normal and pathological tissues of the embryo and adult. ECM acts as critical regulator in various biological processes such as differentiation, cell proliferation, angiogenesis, and immune control. The most frequent primary brain tumors are gliomas and by far the majority are adult astrocytic tumors (AATs). The prognosis for patients with these neoplasms is poor and the treatments modestly improves survival. In the literature, there is a fair number of studies concerning the composition of the ECM in AATs, while the number of studies relating the composition of the ECM with the immune regulation is smaller. Circulating ECM proteins have emerged as a promising biomarker that reflect the general immune landscape of tumor microenvironment and may represent a useful tool in assessing disease activity. Given the importance it can have for therapeutic and prognostic purposes, the aim of our study is to summarize the biological properties of ECM components and their effects on the tumor microenvironment and to provide an overview of the interactions between major ECM proteins and immune cells in AATs. As the field of immunotherapy in glioma is quickly expanding, we retain that current data together with future studies on ECM organization and functions in glioma will provide important insights into the tuning of immunotherapeutic approaches.
Collapse
Affiliation(s)
- Anna Di Vito
- Department of Clinical and Experimental Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy.
| | - Annalidia Donato
- Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Jessica Bria
- Department of Clinical and Experimental Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | | | - Domenico La Torre
- Unit of Neurosurgery, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Natalia Malara
- Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Giuseppe Donato
- Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
10
|
Taheri M, Tehrani HA, Dehghani S, Alibolandi M, Arefian E, Ramezani M. Nanotechnology and bioengineering approaches to improve the potency of mesenchymal stem cell as an off-the-shelf versatile tumor delivery vehicle. Med Res Rev 2024; 44:1596-1661. [PMID: 38299924 DOI: 10.1002/med.22023] [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: 12/08/2022] [Revised: 11/28/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
Targeting actionable mutations in oncogene-driven cancers and the evolution of immuno-oncology are the two prominent revolutions that have influenced cancer treatment paradigms and caused the emergence of precision oncology. However, intertumoral and intratumoral heterogeneity are the main challenges in both fields of precision cancer treatment. In other words, finding a universal marker or pathway in patients suffering from a particular type of cancer is challenging. Therefore, targeting a single hallmark or pathway with a single targeted therapeutic will not be efficient for fighting against tumor heterogeneity. Mesenchymal stem cells (MSCs) possess favorable characteristics for cellular therapy, including their hypoimmune nature, inherent tumor-tropism property, straightforward isolation, and multilineage differentiation potential. MSCs can be loaded with various chemotherapeutics and oncolytic viruses. The combination of these intrinsic features with the possibility of genetic manipulation makes them a versatile tumor delivery vehicle that can be used for in vivo selective tumor delivery of various chemotherapeutic and biological therapeutics. MSCs can be used as biofactory for the local production of chemical or biological anticancer agents at the tumor site. MSC-mediated immunotherapy could facilitate the sustained release of immunotherapeutic agents specifically at the tumor site, and allow for the achievement of therapeutic concentrations without the need for repetitive systemic administration of high therapeutic doses. Despite the enthusiasm evoked by preclinical studies that used MSC in various cancer therapy approaches, the translation of MSCs into clinical applications has faced serious challenges. This manuscript, with a critical viewpoint, reviewed the preclinical and clinical studies that have evaluated MSCs as a selective tumor delivery tool in various cancer therapy approaches, including gene therapy, immunotherapy, and chemotherapy. Then, the novel nanotechnology and bioengineering approaches that can improve the potency of MSC for tumor targeting and overcoming challenges related to their low localization at the tumor sites are discussed.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Gil-Chinchilla JI, Zapata AG, Moraleda JM, García-Bernal D. Bioengineered Mesenchymal Stem/Stromal Cells in Anti-Cancer Therapy: Current Trends and Future Prospects. Biomolecules 2024; 14:734. [PMID: 39062449 PMCID: PMC11275142 DOI: 10.3390/biom14070734] [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: 05/08/2024] [Revised: 06/11/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are one of the most widely used cell types in advanced therapies due to their therapeutic potential in the regulation of tissue repair and homeostasis, and immune modulation. However, their use in cancer therapy is controversial: they can inhibit cancer cell proliferation, but also potentially promote tumour growth by supporting angiogenesis, modulation of the immune milieu and increasing cancer stem cell invasiveness. This opposite behaviour highlights the need for careful and nuanced use of MSCs in cancer treatment. To optimize their anti-cancer effects, diverse strategies have bioengineered MSCs to enhance their tumour targeting and therapeutic properties or to deliver anti-cancer drugs. In this review, we highlight the advanced uses of MSCs in cancer therapy, particularly as carriers of targeted treatments due to their natural tumour-homing capabilities. We also discuss the potential of MSC-derived extracellular vesicles to improve the efficiency of drug or molecule delivery to cancer cells. Ongoing clinical trials are evaluating the therapeutic potential of these cells and setting the stage for future advances in MSC-based cancer treatment. It is critical to identify the broad and potent applications of bioengineered MSCs in solid tumour targeting and anti-cancer agent delivery to position them as effective therapeutics in the evolving field of cancer therapy.
Collapse
Affiliation(s)
- Jesús I. Gil-Chinchilla
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Virgen de la Arrixaca University Hospital, University of Murcia, 30120 Murcia, Spain;
| | - Agustín G. Zapata
- Department of Cell Biology, Complutense University, 28040 Madrid, Spain;
| | - Jose M. Moraleda
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Virgen de la Arrixaca University Hospital, University of Murcia, 30120 Murcia, Spain;
- Department of Medicine, University of Murcia, 30120 Murcia, Spain
| | - David García-Bernal
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Virgen de la Arrixaca University Hospital, University of Murcia, 30120 Murcia, Spain;
- Department of Biochemistry, Molecular Biology and Immunology, University of Murcia, 30120 Murcia, Spain
| |
Collapse
|
12
|
Shirazi MMA, Saedi TA, Moghaddam ZS, Nemati M, Shiri R, Negahdari B, Goradel NH. Nanotechnology and nano-sized tools: Newer approaches to circumvent oncolytic adenovirus limitations. Pharmacol Ther 2024; 256:108611. [PMID: 38387653 DOI: 10.1016/j.pharmthera.2024.108611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/03/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Oncolytic adenoviruses (OAds), engineered Ads preferentially infect and lyse tumor cells, have attracted remarkable attention as immunotherapy weapons for the treatment of various malignancies. Despite hopeful successes in preclinical investigations and translation into clinical phases, they face some challenges that thwart their therapeutic effectiveness, including low infectivity of cancer cells, liver sequestration, pre-existing neutralizing antibodies, physical barriers to the spread of Ads, and immunosuppressive TME. Nanotechnology and nano-sized tools provide several advantages to overcome these limitations of OAds. Nano-sized tools could improve the therapeutic efficacy of OAds by enhancing infectivity and cellular uptake, targeting and protecting from pre-existing immune responses, masking and preventing liver tropism, and co-delivery with other therapeutic agents. Herein, we reviewed the constructs of various OAds and their application in clinical trials, as well as the limitations they have faced. Furthermore, we emphasized the potential applications of nanotechnology to solve the constraints of OAds to improve their anti-tumor activities.
Collapse
Affiliation(s)
| | - Tayebeh Azam Saedi
- Department of Genetics, Faculty of Science, Islamic Azad University, Tonekabon Branch, Tonekabon, Iran
| | - Zahra Samadi Moghaddam
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Shiri
- Department of Basic Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasser Hashemi Goradel
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran; Arthropod-Borne Diseases Research Centre, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
13
|
Zeng M, Zhang W, Li Y, Yu L. Harnessing adenovirus in cancer immunotherapy: evoking cellular immunity and targeting delivery in cell-specific manner. Biomark Res 2024; 12:36. [PMID: 38528632 DOI: 10.1186/s40364-024-00581-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/09/2024] [Indexed: 03/27/2024] Open
Abstract
Recombinant adenovirus (rAd) regimens, including replication-competent oncolytic adenovirus (OAV) and replication-deficient adenovirus, have been identified as potential cancer therapeutics. OAV presents advantages such as selective replication, oncolytic efficacy, and tumor microenvironment (TME) remodeling. In this perspective, the principles and advancements in developing OAV toolkits are reviewed. The burgeoning rAd may dictate efficacy of conventional cancer therapies as well as cancer immunotherapies, including cancer vaccines, synergy with adoptive cell therapy (ACT), and TME reshaping. Concurrently, we explored the potential of rAd hitchhiking to adoptive immune cells or stem cells, highlighting how this approach facilitates synergistic interactions between rAd and cellular therapeutics at tumor sites. Results from preclinical and clinical trials in which immune and stem cells were infected with rAd have been used to address significant oncological challenges, such as postsurgical residual tumor tissue and metastatic tissue. Briefly, rAd can eradicate tumors through various mechanisms, resulting from tumor immunogenicity, reprogramming of the TME, enhancement of cellular immunity, and effective tumor targeting. In this context, we argue that rAd holds immense potential for enhancing cellular immunity and synergistically improving antitumor effects in combination with novel cancer immunotherapies.
Collapse
Affiliation(s)
- Miao Zeng
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Wei Zhang
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Yisheng Li
- Shenzhen Haoshi Biotechnology Co., Ltd. No, 155 Hongtian Road, Xinqiao Street, Bao'an District, Shenzhen, Guangdong, 518125, China.
| | - Li Yu
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| |
Collapse
|
14
|
Neves ER, Anand A, Mueller J, Remy RA, Xu H, Selting KA, Sarkaria JN, Harley BA, Pedron-Haba S. Targeting glioblastoma tumor hyaluronan to enhance therapeutic interventions that regulate metabolic cell properties. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574065. [PMID: 38260497 PMCID: PMC10802468 DOI: 10.1101/2024.01.05.574065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Despite extensive advances in cancer research, glioblastoma (GBM) still remains a very locally invasive and thus challenging tumor to treat, with a poor median survival. Tumor cells remodel their microenvironment and utilize extracellular matrix to promote invasion and therapeutic resistance. We aim here to determine how GBM cells exploit hyaluronan (HA) to maintain proliferation using ligand-receptor dependent and ligand-receptor independent signaling. We use tissue engineering approaches to recreate the three-dimensional tumor microenvironment in vitro, then analyze shifts in metabolism, hyaluronan secretion, HA molecular weight distribution, as well as hyaluronan synthetic enzymes (HAS) and hyaluronidases (HYAL) activity in an array of patient derived xenograft GBM cells. We reveal that endogenous HA plays a role in mitochondrial respiration and cell proliferation in a tumor subtype dependent manner. We propose a tumor specific combination treatment of HYAL and HAS inhibitors to disrupt the HA stabilizing role in GBM cells. Taken together, these data shed light on the dual metabolic and ligand - dependent signaling roles of hyaluronan in glioblastoma. Significance The control of aberrant hyaluronan metabolism in the tumor microenvironment can improve the efficacy of current treatments. Bioengineered preclinical models demonstrate potential to predict, stratify and accelerate the development of cancer treatments.
Collapse
|
15
|
Zhang L, Guo S, Chang S, Jiang G. Revolutionizing Cancer Treatment: Unleashing the Power of Combining Oncolytic Viruses with CAR-T Cells. Anticancer Agents Med Chem 2024; 24:1407-1418. [PMID: 39051583 DOI: 10.2174/0118715206308253240723055019] [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: 03/02/2024] [Revised: 06/07/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024]
Abstract
Oncolytic Viruses (OVs) have emerged as a promising treatment option for cancer thanks to their significant research potential and encouraging results. These viruses exert a profound impact on the tumor microenvironment, making them effective against various types of cancer. In contrast, the efficacy of Chimeric antigen receptor (CAR)-T cell therapy in treating solid tumors is relatively low. The combination of OVs and CAR-T cell therapy, however, is a promising area of research. OVs play a crucial role in enhancing the tumor-suppressive microenvironment, which in turn enables CAR-T cells to function efficiently in the context of solid malignancies. This review aims to provide a comprehensive analysis of the benefits and drawbacks of OV therapy and CAR-T cell therapy, with a focus on the potential of combining these two treatment approaches.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - ShuXian Guo
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - ShuYing Chang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Guan Jiang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| |
Collapse
|
16
|
Chen L, Zuo M, Zhou Q, Wang Y. Oncolytic virotherapy in cancer treatment: challenges and optimization prospects. Front Immunol 2023; 14:1308890. [PMID: 38169820 PMCID: PMC10758479 DOI: 10.3389/fimmu.2023.1308890] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
Oncolytic viruses (OVs) are emerging cancer therapeutics that offer a multifaceted therapeutic platform for the benefits of replicating and lysing tumor cells, being engineered to express transgenes, modulating the tumor microenvironment (TME), and having a tolerable safety profile that does not overlap with other cancer therapeutics. The mechanism of OVs combined with other antitumor agents is based on immune-mediated attack resistance and might benefit patients who fail to achieve durable responses after immune checkpoint inhibitor (ICI) treatment. In this Review, we summarize data on the OV mechanism and limitations of monotherapy, which are currently in the process of combination partner development, especially with ICIs. We discuss some of the hurdles that have limited the preclinical and clinical development of OVs. We also describe the available data and provide guidance for optimizing OVs in clinical practice, as well as a summary of approved and promising novel OVs with clinical indications.
Collapse
Affiliation(s)
- Lingjuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Mengsi Zuo
- Department of Oncology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Qin Zhou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Yang Wang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), College of Bioengineering, Hubei University of Technology, Wuhan, China
| |
Collapse
|
17
|
Zhang L, Pakmehr SA, Shahhosseini R, Hariri M, Fakhrioliaei A, Karkon Shayan F, Xiang W, Karkon Shayan S. Oncolytic viruses improve cancer immunotherapy by reprogramming solid tumor microenvironment. Med Oncol 2023; 41:8. [PMID: 38062315 DOI: 10.1007/s12032-023-02233-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/21/2023] [Indexed: 12/18/2023]
Abstract
Immunotherapies using immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T-cell therapy have achieved successful results against several types of human tumors, particularly hematological malignancies. However, their clinical results for the treatment of solid tumors remain poor and unsatisfactory. The immunosuppressive tumor microenvironment (TME) plays an important role by interfering with intratumoral T-cell infiltration, promoting effector T-cell exhaustion, upregulating inhibitory molecules, inducing hypoxia, and so on. Oncolytic viruses are an encouraging biocarrier that could be used in both natural and genetically engineered platforms to induce oncolysis in a targeted manner. Oncolytic virotherapy (OV) contributes to the reprogramming of the TME, thus synergizing the functional effects of current ICIs and CAR T-cell therapy to overcome resistant barriers in solid tumors. Here, we summarize the TME-related inhibitory factors affecting the therapeutic outcomes of ICIs and CAR T cells and discuss the potential of OV-based approaches to alleviate these barriers and improve future therapies for advanced solid tumors.
Collapse
Affiliation(s)
- Ling Zhang
- The Second People's Hospital of Lianyungang, Jiangsu, 222000, China
| | | | | | - Maryam Hariri
- Department of Pathobiology, Auburn University, Auburn, AL, 36832, USA
| | | | - Farid Karkon Shayan
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Wenxue Xiang
- The Second People's Hospital of Lianyungang, Jiangsu, 222000, China.
| | - Sepideh Karkon Shayan
- Student Research Committee, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
- Clinical Research Development Unit, Bohlool Hospital, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
18
|
Hu D, Tian Y, Xu J, Xie D, Wang Y, Liu M, Wang Y, Yang L. Oncolytic viral therapy as promising immunotherapy against glioma. MEDCOMM – FUTURE MEDICINE 2023; 2. [DOI: 10.1002/mef2.61] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/22/2023] [Indexed: 03/19/2025]
Abstract
AbstractGlioma is a common primary central nervous system malignant tumor in clinical, traditional methods such as surgery and chemoradiotherapy are not effective in treatment. Therefore, more effective treatments need to be found. Oncolytic viruses (OVs) are a new type of immunotherapy that selectively infects and kills tumor cells instead of normal cells. OVs can mediate antitumor immune responses through a variety of mechanisms, and have the ability to activate antitumor immune responses, transform the tumor microenvironment from “cold” to “hot,” and enhance the efficacy of immune checkpoint inhibitors. Recently, a large number of preclinical and clinical studies have shown that OVs show great prospects in the treatment of gliomas. In this review, we summarize the current status of glioma therapies with a focus on OVs. First, this article introduces the current status of treatment of glioma and their respective shortcomings. Then, the important progress of OVs of in clinical trials of glioma is summarized. Finally, the urgent challenges of oncolytic virus treatment for glioma are sorted out, and related solutions are proposed. This review will help to further promote the use of OVs in the treatment of glioma.
Collapse
Affiliation(s)
- Die Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Yaomei Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
- College of Bioengineering Sichuan University of Science & Engineering Zigong China
| | - Jie Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Daoyuan Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Mohan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Yuanda Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| |
Collapse
|
19
|
Li Z, Wu F, Zeng Y, Xu Y, Liu H, Wang X, Luo F, Lin Z. Design of target response wettability switchable core-shell-shell electrochemiluminescence nanoprobes for sensitive hyaluronidase detection. Chem Commun (Camb) 2023; 59:12621-12624. [PMID: 37791621 DOI: 10.1039/d3cc04408d] [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: 10/05/2023]
Abstract
Electrochemiluminescence nanoprobes with a core-shell-shell structure have been designed and applied for hyaluronidase detection. The nanoprobes can precipitate efficiently through target-regulation wettability for collection, and enrich near to the hydrophobic electrode surface through hydrophobic interaction to enhance the performance of the biosensor.
Collapse
Affiliation(s)
- Zhixin Li
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China.
- Institute for Advanced Study, Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Fangcai Wu
- Department of Radiation Oncology, Esophageal Cancer Prevention and Control Research Center, the Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Yulan Zeng
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China.
| | - Yiwei Xu
- Department of Clinical Laboratory Medicine, Esophageal Cancer Prevention and Control Research Center, the Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Hongning Liu
- Institute for Advanced Study, Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Xinjia Wang
- Department of Orthopedics, Esophageal Cancer Prevention and Control Research Center, the Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China.
| | - Fang Luo
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China.
| | - Zhenyu Lin
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China.
| |
Collapse
|
20
|
Wang M, Wang X, Jin X, Zhou J, Zhang Y, Yang Y, Liu Y, Zhang J. Cell-based and cell-free immunotherapies for glioblastoma: current status and future directions. Front Immunol 2023; 14:1175118. [PMID: 37304305 PMCID: PMC10248152 DOI: 10.3389/fimmu.2023.1175118] [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: 02/27/2023] [Accepted: 05/08/2023] [Indexed: 06/13/2023] Open
Abstract
Glioblastoma (GBM) is among the most fatal and recurring malignant solid tumors. It arises from the GBM stem cell population. Conventional neurosurgical resection, temozolomide (TMZ)-dependent chemotherapy and radiotherapy have rendered the prognosis of patients unsatisfactory. Radiotherapy and chemotherapy can frequently induce non-specific damage to healthy brain and other tissues, which can be extremely hazardous. There is therefore a pressing need for a more effective treatment strategy for GBM to complement or replace existing treatment options. Cell-based and cell-free immunotherapies are currently being investigated to develop new treatment modalities against cancer. These treatments have the potential to be both selective and successful in minimizing off-target collateral harm in the normal brain. In this review, several aspects of cell-based and cell-free immunotherapies related to GBM will be discussed.
Collapse
Affiliation(s)
- Mingming Wang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Xiaojie Wang
- Basic Medical School, Shenyang Medical College, Shenyang, Liaoning, China
| | - Xiaoyan Jin
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Jingjing Zhou
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yufu Zhang
- Department of Hepatobiliary Surgery, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Yiyuan Yang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yusi Liu
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Jing Zhang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| |
Collapse
|
21
|
Lim XY, Capinpin SM, Bolem N, Foo ASC, Yip WG, Kumar AP, Teh DBL. Biomimetic nanotherapeutics for targeted drug delivery to glioblastoma multiforme. Bioeng Transl Med 2023; 8:e10483. [PMID: 37206213 PMCID: PMC10189489 DOI: 10.1002/btm2.10483] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 02/17/2023] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain tumor with poor prognosis and high mortality, with no curative treatment to date as limited trafficking across the blood-brain barrier (BBB) combined with tumor heterogeneity often leads to therapeutic failure. Although modern medicine poses a wide range of drugs that are otherwise efficacious in treating other tumors, they often do not achieve therapeutic concentrations in the brain, hence driving the need for more effective drug delivery strategies. Nanotechnology, an interdisciplinary field, has been gaining immense popularity in recent years for remarkable advancements such as nanoparticle (NP) drug carriers, which possess extraordinary versatility in modifying surface coatings to home in on target cells, including those beyond the BBB. In this review, we will be highlighting recent developments in biomimetic NPs in GBM therapy and how these allowed us to overcome the physiological and anatomical challenges that have long plagued GBM treatment.
Collapse
Affiliation(s)
- Xin Yuan Lim
- MBBS ProgrammeYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Sharah Mae Capinpin
- Department of PharmacologyYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- NUS Centre for Cancer ResearchYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Nagarjun Bolem
- Department of Surgery, Division of NeurosurgeryNational University HospitalSingaporeSingapore
| | - Aaron Song Chuan Foo
- MBBS ProgrammeYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Department of Surgery, Division of NeurosurgeryNational University HospitalSingaporeSingapore
| | - Wai‐Cheong George Yip
- Department of AnatomyYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Alan Prem Kumar
- Department of PharmacologyYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- NUS Centre for Cancer ResearchYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Daniel Boon Loong Teh
- Department of AnatomyYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Department of BiochemistryYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Department of OphthalmologyYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- NeurobiologyLife Science Institute, National University of SingaporeSingaporeSingapore
| |
Collapse
|
22
|
Chartouni A, Mouawad A, Boutros M, Attieh F, Medawar N, Kourie HR. Mesenchymal stem cells: a trojan horse to treat glioblastoma. Invest New Drugs 2023; 41:240-250. [PMID: 37017885 DOI: 10.1007/s10637-023-01352-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/23/2023] [Indexed: 04/06/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common and lethal primary tumor of the central nervous system. What makes it so dreadful is the very low survival rate, despite the existence of a standard treatment plan. An innovative and more effective way to treat glioblastoma based on Mesenchymal Stem Cells (MSCs) has been explored recently. MSCs are a group of endogenous multipotent stem cells that could mainly be harvested from adipose tissue, bone marrow, and umbilical cord. Having the ability to migrate toward the tumor using multiple types of binding receptors, they could be used either as a direct treatment (whether they are enhanced or not) or as a delivery vehicle carrying various anti-tumoral agents. Some of these agents are: chemotherapy drugs, prodrug activating therapy, oncolytic viruses, nanoparticles, human artificial chromosome… Promising results have started to surface; however, more evidence is needed to perfect their use as a glioblastoma multiforme treatment option. Alternative treatment, using unloaded or loaded MSCs, leading to a better outcome.
Collapse
Affiliation(s)
- Antoine Chartouni
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon.
| | - Antoine Mouawad
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon
| | - Marc Boutros
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon
| | - Fouad Attieh
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon
| | - Nicolas Medawar
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon
| | - Hampig Raphaël Kourie
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon
- Department of hematology-oncology, Faculty of medicine, Saint-joseph university of beirut, Beirut, Lebanon
| |
Collapse
|
23
|
Karami Fath M, Moayedi Banan Z, Barati R, Mohammadrezakhani O, Ghaderi A, Hatami A, Ghiabi S, Zeidi N, Asgari K, Payandeh Z, Barati G. Recent advancements to engineer mesenchymal stem cells and their extracellular vesicles for targeting and destroying tumors. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 178:1-16. [PMID: 36781149 DOI: 10.1016/j.pbiomolbio.2023.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/24/2023] [Accepted: 02/10/2023] [Indexed: 02/13/2023]
Abstract
Mesenchymal stem cells (MSCs) have the ability to migrate into tumor sites and release growth factors to modulate the tumor microenvironment. MSC therapy have shown a dual role in cancers, promoting or inhibiting. However, MSCs could be used as a carrier of anticancer agents for targeted tumor therapy. Recent technical improvements also allow engineering MSCs to improve tumor-targeting properties, protect anticancer agents, and decrease the cytotoxicity of drugs. While some of MSC functions are mediated through their secretome, MSCs-derived extracellular vesicles (EVs) are also proposed as a possible viechle for cancer therapy. EVs allow efficient loading of anticancer agents and have an intrinsic ability to target tumor cells, making them suitable for targeted therapy of tumors. In addition, the specificity and selectivity of EVs to the tumor sites could be enhanced by surface modification. In this review, we addressed the current approaches used for engineering MSCs and EVs to effectively target tumor sites and deliver anticancer agents.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Zahra Moayedi Banan
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Barati
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Omid Mohammadrezakhani
- Faculty of Pharmacy, Ramsar Campus, Mazandaran University of Medical Sciences, Sari, Iran
| | - Aliasghar Ghaderi
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hatami
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shamim Ghiabi
- Department of Medical Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nazanin Zeidi
- Division of Pharmaceutical Science, Long Island University, Brooklyn, NY, USA
| | - Katayoon Asgari
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Payandeh
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|
24
|
Abstract
Oncolytic viruses (OVs) are an emerging class of cancer therapeutics that offer the benefits of selective replication in tumour cells, delivery of multiple eukaryotic transgene payloads, induction of immunogenic cell death and promotion of antitumour immunity, and a tolerable safety profile that largely does not overlap with that of other cancer therapeutics. To date, four OVs and one non-oncolytic virus have been approved for the treatment of cancer globally although talimogene laherparepvec (T-VEC) remains the only widely approved therapy. T-VEC is indicated for the treatment of patients with recurrent melanoma after initial surgery and was initially approved in 2015. An expanding body of data on the clinical experience of patients receiving T-VEC is now becoming available as are data from clinical trials of various other OVs in a range of other cancers. Despite increasing research interest, a better understanding of the underlying biology and pharmacology of OVs is needed to enable the full therapeutic potential of these agents in patients with cancer. In this Review, we summarize the available data and provide guidance on optimizing the use of OVs in clinical practice, with a focus on the clinical experience with T-VEC. We describe data on selected novel OVs that are currently in clinical development, either as monotherapies or as part of combination regimens. We also discuss some of the preclinical, clinical and regulatory hurdles that have thus far limited the development of OVs.
Collapse
|
25
|
Zamboni F, Wong CK, Collins MN. Hyaluronic acid association with bacterial, fungal and viral infections: Can hyaluronic acid be used as an antimicrobial polymer for biomedical and pharmaceutical applications? Bioact Mater 2023; 19:458-473. [PMID: 35574061 PMCID: PMC9079116 DOI: 10.1016/j.bioactmat.2022.04.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/05/2022] [Accepted: 04/23/2022] [Indexed: 12/21/2022] Open
Abstract
The relationships between hyaluronic acid (HA) and pathological microorganisms incite new understandings on microbial infection, tissue penetration, disease progression and lastly, potential treatments. These understandings are important for the advancement of next generation antimicrobial therapeutical strategies for the control of healthcare-associated infections. Herein, this review will focus on the interplay between HA, bacteria, fungi, and viruses. This review will also comprehensively detail and discuss the antimicrobial activity displayed by various HA molecular weights for a variety of biomedical and pharmaceutical applications, including microbiology, pharmaceutics, and tissue engineering.
Collapse
Affiliation(s)
- Fernanda Zamboni
- Bernal Institute, School of Engineering, University of Limerick, Ireland
- Health Research Institute, University of Limerick, Ireland
| | - Chun Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Maurice N. Collins
- Bernal Institute, School of Engineering, University of Limerick, Ireland
- Health Research Institute, University of Limerick, Ireland
| |
Collapse
|
26
|
Bikfalvi A, da Costa CA, Avril T, Barnier JV, Bauchet L, Brisson L, Cartron PF, Castel H, Chevet E, Chneiweiss H, Clavreul A, Constantin B, Coronas V, Daubon T, Dontenwill M, Ducray F, Enz-Werle N, Figarella-Branger D, Fournier I, Frenel JS, Gabut M, Galli T, Gavard J, Huberfeld G, Hugnot JP, Idbaih A, Junier MP, Mathivet T, Menei P, Meyronet D, Mirjolet C, Morin F, Mosser J, Moyal ECJ, Rousseau V, Salzet M, Sanson M, Seano G, Tabouret E, Tchoghandjian A, Turchi L, Vallette FM, Vats S, Verreault M, Virolle T. Challenges in glioblastoma research: focus on the tumor microenvironment. Trends Cancer 2023; 9:9-27. [PMID: 36400694 DOI: 10.1016/j.trecan.2022.09.005] [Citation(s) in RCA: 134] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/20/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022]
Abstract
Glioblastoma (GBM) is the most deadly type of malignant brain tumor, despite extensive molecular analyses of GBM cells. In recent years, the tumor microenvironment (TME) has been recognized as an important player and therapeutic target in GBM. However, there is a need for a full and integrated understanding of the different cellular and molecular components involved in the GBM TME and their interactions for the development of more efficient therapies. In this review, we provide a comprehensive report of the GBM TME, which assembles the contributions of physicians and translational researchers working on brain tumor pathology and therapy in France. We propose a holistic view of the subject by delineating the specific features of the GBM TME at the cellular, molecular, and therapeutic levels.
Collapse
Affiliation(s)
- Andreas Bikfalvi
- Bordeaux University, INSERM, U1312 BRIC, Tumor and Vascular Biology Laboratory, F-33600, Pessac, France.
| | - Cristine Alves da Costa
- Côte d'Azur University, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Team "Laboratory of Excellence (LABEX) Distalz", F-06560 Nice, France
| | - Tony Avril
- Rennes University, Inserm U1242, Centre de Lutte contre le Cancer Eugène Marquis, F- 35000 Rennes, France
| | - Jean-Vianney Barnier
- Institute of Neuroscience Paris-Saclay, UMR9197, CNRS, Univ. Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Luc Bauchet
- Montpellier University Medical Center, Department of Neurosurgery, INSERM U1191, F-34090 Montpellier, France
| | - Lucie Brisson
- Bordeaux University, INSERM, U1312 BRIC, Tumor and Vascular Biology Laboratory, F-33600, Pessac, France
| | | | - Hélène Castel
- Normandie University, INSERM U1239, DC2N, Institute for Research and Innovation in Biomedicine (IRIB), F-76000 Rouen, France
| | - Eric Chevet
- Rennes University, Inserm U1242, Centre de Lutte contre le Cancer Eugène Marquis, F- 35000 Rennes, France
| | - Hervé Chneiweiss
- Sorbonne University, CNRS UMR8246, Inserm U1130, IBPS-Neuroscience Paris Seine, F- 75005 Paris, France
| | - Anne Clavreul
- Angers University, CHU d'Angers, CRCINA, F-49000 Angers, France
| | - Bruno Constantin
- Poitiers University, CNRS UMR 6041, Laboratory Channels & Connexins in Cancers and Cell Stemness, F-86000 Poitiers, France
| | - Valérie Coronas
- Poitiers University, CNRS UMR 6041, Laboratory Channels & Connexins in Cancers and Cell Stemness, F-86000 Poitiers, France
| | - Thomas Daubon
- Bordeaux University, CNRS, IBGC, UMR 5095, F-33 077 Bordeaux, France
| | - Monique Dontenwill
- Strasbourg University, Laboratoire de Bioimagerie et Pathologie, UMR7021 CNRS, F-67401 Illkirch-Graffenstaden, France
| | - Francois Ducray
- Lyon I University, Cancer Research Centre of Lyon (CRCL) INSERM 1052&CNRS UMR5286, Centre Léon Bérard, Lyon 69008, France., F-69622 Villeurbanne, France
| | - Natacha Enz-Werle
- Strasbourg University, Laboratoire de Bioimagerie et Pathologie, UMR7021 CNRS, F-67401 Illkirch-Graffenstaden, France
| | - Dominique Figarella-Branger
- Aix-Marseille University, Service d'Anatomie Pathologique et de Neuropathologie, Hôpital de la Timone, F-13385 Marseille, France
| | - Isabelle Fournier
- Lille University, Inserm, CHU Lille, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Jean-Sébastien Frenel
- Normandie University, INSERM U1239, DC2N, Institute for Research and Innovation in Biomedicine (IRIB), F-76000 Rouen, France
| | - Mathieu Gabut
- Lyon I University, Cancer Research Centre of Lyon (CRCL) INSERM 1052&CNRS UMR5286, Centre Léon Bérard, Lyon 69008, France., F-69622 Villeurbanne, France
| | - Thierry Galli
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, GHU PARIS Psychiatrie & Neurosciences, F-75014 Paris, France
| | - Julie Gavard
- CRCI2NA, INSERM U1307, CNRS UMR6075, Nantes Universite, 44007 Nantes, France
| | - Gilles Huberfeld
- College de France, Center for Interdisciplinary Research in Biology (CIRB), CNRS, INSERM, Université PSL, Paris 75005, France
| | - Jean-Philippe Hugnot
- Montpellier University, Institut de Génomique Fonctionnelle, CNRS, INSERM, F-34094 Montpellier, France
| | - Ahmed Idbaih
- Sorbonne University, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, F-75013, Paris, France
| | - Marie-Pierre Junier
- Sorbonne University, CNRS UMR8246, Inserm U1130, IBPS-Neuroscience Paris Seine, F- 75005 Paris, France
| | - Thomas Mathivet
- Bordeaux University, INSERM, U1312 BRIC, Tumor and Vascular Biology Laboratory, F-33600, Pessac, France
| | - Philippe Menei
- Angers University, CHU d'Angers, CRCINA, F-49000 Angers, France
| | - David Meyronet
- Institute of Neuropathology, Hospices Civils de Lyon, F-69008, Lyon, France
| | - Céline Mirjolet
- Centre Georges-François Leclerc, UNICANCER, Dijon, France. Inserm U1231, Equipe Cadir, F-21000 Dijon, France
| | - Fabrice Morin
- Normandie University, INSERM U1239, DC2N, Institute for Research and Innovation in Biomedicine (IRIB), F-76000 Rouen, France
| | - Jean Mosser
- Rennes University, Inserm U1242, Centre de Lutte contre le Cancer Eugène Marquis, F- 35000 Rennes, France
| | - Elisabeth Cohen-Jonathan Moyal
- Institut Claudius Regaud, NSERM 1037, CRCT Team RADOPT, Département de Radiothérapie, IUCT-Oncopole, F-31100 Toulouse, France
| | - Véronique Rousseau
- Institute of Neuroscience Paris-Saclay, UMR9197, CNRS, Univ. Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Michel Salzet
- Lille University, Inserm, CHU Lille, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Marc Sanson
- Sorbonne University, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, F-75013, Paris, France
| | - Giorgio Seano
- Curie Institute Research Center, Tumor Microenvironment Laboratory, PSL Research University, Inserm U1021, CNRS UMR3347, F-91898 Orsay, France
| | - Emeline Tabouret
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, F-13005 Marseille, France
| | - Aurélie Tchoghandjian
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, F-13005 Marseille, France
| | - Laurent Turchi
- Côte D'Azur University, CNRS, INSERM, Institut de Biologie Valrose, Team INSERM "Cancer Stem Cell Plasticity and Functional Intra-tumor Heterogeneity", F-06108 Nice, France
| | - Francois M Vallette
- CRCI2NA, INSERM U1307, CNRS UMR6075, Nantes Universite, 44007 Nantes, France
| | - Somya Vats
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, GHU PARIS Psychiatrie & Neurosciences, F-75014 Paris, France
| | - Maité Verreault
- Sorbonne University, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, F-75013, Paris, France
| | - Thierry Virolle
- Côte D'Azur University, CNRS, INSERM, Institut de Biologie Valrose, Team INSERM "Cancer Stem Cell Plasticity and Functional Intra-tumor Heterogeneity", F-06108 Nice, France
| |
Collapse
|
27
|
Wang L, Shi Y, Jiang J, Li C, Zhang H, Zhang X, Jiang T, Wang L, Wang Y, Feng L. Micro-Nanocarriers Based Drug Delivery Technology for Blood-Brain Barrier Crossing and Brain Tumor Targeting Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203678. [PMID: 36103614 DOI: 10.1002/smll.202203678] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/04/2022] [Indexed: 06/15/2023]
Abstract
The greatest obstacle to using drugs to treat brain tumors is the blood-brain barrier (BBB), making it difficult for conventional drug molecules to enter the brain. Therefore, how to safely and effectively penetrate the BBB to achieve targeted drug delivery to brain tumors has been a challenging research problem. With the intensive research in micro- and nanotechnology in recent years, nano drug-targeted delivery technologies have shown great potential to overcome this challenge, such as inorganic nanocarriers, organic polymer-carriers, liposomes, and biobased carriers, which can be designed in different sizes, shapes, and surface functional groups to enhance their ability to penetrate the BBB and targeted drug delivery for brain tumors. In this review, the composition and overcoming patterns of the BBB are detailed, and then the hot research topics of drug delivery carriers for brain tumors in recent years are summarized, and their mechanisms of action on the BBB and the factors affecting drug delivery are described in detail, and the effectiveness of targeted therapy for brain tumors is evaluated. Finally, the challenges and dilemmas in developing brain tumor drug delivery systems are discussed, which will be promising in the future for targeted drug delivery to brain tumors based on micro-nanocarriers technology.
Collapse
Affiliation(s)
- Luyao Wang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Youyuan Shi
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Jingzhen Jiang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Chan Li
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Hengrui Zhang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Xinhui Zhang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Tao Jiang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yinyan Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Lin Feng
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| |
Collapse
|
28
|
Yan G, Kong B, Zhao J, Ni H, Zhan L, Huang C, Zou H. Fluorescence turn-on Cu 2-xSe@HA-rhodamine 6G FRET nanoprobe for hyaluronidase detection and imaging. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2022; 233:112496. [PMID: 35689932 DOI: 10.1016/j.jphotobiol.2022.112496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/06/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
The application of nanostructures to design fluorescence resonance energy transfer (FRET) based sensing platforms has been greatly concerned with the demand for sensitive and selective detection of biomolecules. Here, a novel sensitive turn-on fluorescence strategy based on the FRET mechanism has been designed for hyaluronidase (HAase) detection through the modulation of Cu2-xSe@HA-Rh6G nanoprobe fabricated by self-assembly of rhodamine 6G (Rh6G) together with Cu2-xSe@HA nanoparticles through electrostatic adsorption. The Cu2-xSe@HA had extensive localized surface plasma resonance (LSPR) absorption in the wide range of ultraviolet (UV) to near-infrared (NIR) wavelengths and showed good light capture characteristics, which can be acted as good acceptors in the FRET interactions with Rh6G, inducing its efficient fluorescence quenching. In the presence of HAase, the FRET process was disrupted and the fluorescence signal was recovered. In the range of 0.1-10.0 U/mL, the fluorescence recovery of Rh6G showed a good linear relationship with the concentration of HAase, and the detection limit was 0.06 U/mL. The sensing platform has been used for HAase detection in real urine samples and cancer cells imaging.
Collapse
Affiliation(s)
- Guojuan Yan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Bo Kong
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jiaqiang Zhao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Huanhuan Ni
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Lei Zhan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Chengzhi Huang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Luminescent and Real-Time Analysis System, Chongqing Science and Technology Commission, College of Chemistry and Chemical Engineering, Southwest University, Beibei, Chongqing 400715, China
| | - Hongyan Zou
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
29
|
Heris RM, Shirvaliloo M, Abbaspour-Aghdam S, Hazrati A, Shariati A, Youshanlouei HR, Niaragh FJ, Valizadeh H, Ahmadi M. The potential use of mesenchymal stem cells and their exosomes in Parkinson's disease treatment. Stem Cell Res Ther 2022; 13:371. [PMID: 35902981 PMCID: PMC9331055 DOI: 10.1186/s13287-022-03050-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 07/17/2022] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is the second most predominant neurodegenerative disease worldwide. It is recognized clinically by severe complications in motor function caused by progressive degeneration of dopaminergic neurons (DAn) and dopamine depletion. As the current standard of treatment is focused on alleviating symptoms through Levodopa, developing neuroprotective techniques is critical for adopting a more pathology-oriented therapeutic approach. Regenerative cell therapy has provided us with an unrivalled platform for evaluating potentially effective novel methods for treating neurodegenerative illnesses over the last two decades. Mesenchymal stem cells (MSCs) are most promising, as they can differentiate into dopaminergic neurons and produce neurotrophic substances. The precise process by which stem cells repair neuronal injury is unknown, and MSC-derived exosomes are suggested to be responsible for a significant portion of such effects. The present review discusses the application of mesenchymal stem cells and MSC-derived exosomes in PD treatment.
Collapse
Affiliation(s)
| | - Milad Shirvaliloo
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ali Hazrati
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Shariati
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farhad Jadidi Niaragh
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Valizadeh
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
30
|
Scanlan H, Coffman Z, Bettencourt J, Shipley T, Bramblett DE. Herpes simplex virus 1 as an oncolytic viral therapy for refractory cancers. Front Oncol 2022; 12:940019. [PMID: 35965554 PMCID: PMC9364694 DOI: 10.3389/fonc.2022.940019] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022] Open
Abstract
The need for efficacious and non-toxic cancer therapies is paramount. Oncolytic viruses (OVs) are showing great promise and are introducing new possibilities in cancer treatment with their ability to selectively infect tumor cells and trigger antitumor immune responses. Herpes Simplex Virus 1 (HSV-1) is a commonly selected OV candidate due to its large genome, relative safety profile, and ability to infect a variety of cell types. Talimogene laherparevec (T-VEC) is an HSV-1-derived OV variant and the first and only OV therapy currently approved for clinical use by the United States Food and Drug Administration (FDA). This review provides a concise description of HSV-1 as an OV candidate and the genomic organization of T-VEC. Furthermore, this review focuses on the advantages and limitations in the use of T-VEC compared to other HSV-1 OV variants currently in clinical trials. In addition, approaches for future directions of HSV-1 OVs as cancer therapy is discussed.
Collapse
Affiliation(s)
- Hayle Scanlan
- Rowan School of Medicine, RowanSOM-Jefferson Health-Virtua Our Lady of Lourdes Hospital, Stratford, NJ, United States
| | - Zachary Coffman
- Monroe Clinic Rural Family Medicine Program, The University of Illinois College of Medicine Rockford, Monroe, WI, United States
| | - Jeffrey Bettencourt
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Timothy Shipley
- Department of Biomedical Sciences, A.T. Still University School of Osteopathic Medicine in Arizona, Mesa, AZ, United States
| | - Debra E. Bramblett
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
- *Correspondence: Debra E. Bramblett,
| |
Collapse
|
31
|
Jenner AL, Smalley M, Goldman D, Goins WF, Cobbs CS, Puchalski RB, Chiocca EA, Lawler S, Macklin P, Goldman A, Craig M. Agent-based computational modeling of glioblastoma predicts that stromal density is central to oncolytic virus efficacy. iScience 2022; 25:104395. [PMID: 35637733 PMCID: PMC9142563 DOI: 10.1016/j.isci.2022.104395] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/18/2022] [Accepted: 04/08/2022] [Indexed: 11/26/2022] Open
Abstract
Oncolytic viruses (OVs) are emerging cancer immunotherapy. Despite notable successes in the treatment of some tumors, OV therapy for central nervous system cancers has failed to show efficacy. We used an ex vivo tumor model developed from human glioblastoma tissue to evaluate the infiltration of herpes simplex OV rQNestin (oHSV-1) into glioblastoma tumors. We next leveraged our data to develop a computational, model of glioblastoma dynamics that accounts for cellular interactions within the tumor. Using our computational model, we found that low stromal density was highly predictive of oHSV-1 therapeutic success, suggesting that the efficacy of oHSV-1 in glioblastoma may be determined by stromal-to-tumor cell regional density. We validated these findings in heterogenous patient samples from brain metastatic adenocarcinoma. Our integrated modeling strategy can be applied to suggest mechanisms of therapeutic responses for central nervous system cancers and to facilitate the successful translation of OVs into the clinic.
Collapse
Affiliation(s)
- Adrianne L. Jenner
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada
- Sainte-Justine University Hospital Research Centre, Montréal, QC, Canada
| | - Munisha Smalley
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | | | - William F. Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles S. Cobbs
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Ralph B. Puchalski
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
| | - E. Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sean Lawler
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul Macklin
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Aaron Goldman
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Morgan Craig
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada
- Sainte-Justine University Hospital Research Centre, Montréal, QC, Canada
| |
Collapse
|
32
|
Immunotherapy by mesenchymal stromal cell delivery of oncolytic viruses for treating metastatic tumors. Mol Ther Oncolytics 2022; 25:78-97. [PMID: 35434272 PMCID: PMC8989711 DOI: 10.1016/j.omto.2022.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Oncolytic viruses (OVs) have emerged as a very promising anti-cancer therapeutic strategy in the past decades. However, despite their pre-clinical promise, many OV clinical evaluations for cancer therapy have highlighted the continued need for their improved delivery and targeting. Mesenchymal stromal cells (MSCs) have emerged as excellent candidate vehicles for the delivery of OVs due to their tumor-homing properties and low immunogenicity. MSCs can enhance OV delivery by protecting viruses from rapid clearance following administration and also by more efficiently targeting tumor sites, consequently augmenting the therapeutic potential of OVs. MSCs can function as “biological factories,” enabling OV amplification within these cells to promote tumor lysis following MSC-OV arrival at the tumor site. MSC-OVs can promote enhanced safety profiles and therapeutic effects relative to OVs alone. In this review we explore the general characteristics of MSCs as delivery tools for cancer therapeutic agents. Furthermore, we discuss the potential of OVs as immune therapeutics and highlight some of the promising applications stemming from combining MSCs to achieve enhanced delivery and anti-tumor effectiveness of OVs at different pre-clinical and clinical stages. We further provide potential pitfalls of the MSC-OV platform and the strategies under development for enhancing the efficacy of these emerging therapeutics.
Collapse
|
33
|
Yang J, Zhang L. The roles and therapeutic approaches of MSC-derived exosomes in colorectal cancer. Clin Transl Oncol 2022; 24:959-967. [PMID: 35037237 DOI: 10.1007/s12094-021-02750-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer in both men and women, accounting for 8% of all new cancer cases in both. CRC is typically diagnosed at advanced stages, which leads to a higher mortality rate. The 5-year survival rate for CRC is 64% in all cases and just 12% in metastatic cases. Mesenchymal stem cells (MSCs) are one of the most recent approaches for therapeutic interventions in cancer. MSCs have multiple properties, including paracrine signaling, immunologic functions, and the ability to migrate to the targeted tissue. MSCs can produce and secrete exosomes in tumor microenvironments. These exosomes can transfer compounds across tumor cells, stromal cells, fibroblasts, endothelial cells, and immune cells. Studies showed that modified MCS-derived exosomes have enhanced specificity, reduced immunogenicity, and better targeting capabilities in comparison to other frequently used delivery systems such as liposomes. Therefore, this study aimed to provide a comprehensive view of the role of natural MSC-derived exosomes in CRC, as well as the most current and prospective advancements in MSC-derived exosome therapeutic modifications.
Collapse
Affiliation(s)
- Jie Yang
- Anorectal, Shijiazhuang Hospital of Traditional Chinese Medicine, Shijiazhuang, 050051, China.
| | - Liman Zhang
- Anorectal, Shijiazhuang Hospital of Traditional Chinese Medicine, Shijiazhuang, 050051, China
| |
Collapse
|
34
|
Icardi A, Lompardia SL, Papademetrio DL, Rosales P, Díaz M, Pibuel MA, Alaniz L, Alvarez E. Hyaluronan in the Extracellular Matrix of Hematological and Solid Tumors. Its Biological Effects. BIOLOGY OF EXTRACELLULAR MATRIX 2022:161-196. [DOI: 10.1007/978-3-030-99708-3_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
35
|
Niknam MR, Attari F. The Potential Applications of Stem Cells for Cancer Treatment. Curr Stem Cell Res Ther 2022; 17:26-42. [PMID: 35048802 DOI: 10.2174/1574888x16666210810100858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 01/10/2023]
Abstract
:
Scientists encounter many obstacles in traditional cancer therapies, including the side effects
on the healthy cells, drug resistance, tumor relapse, the short half-life of employed drugs in
the blood circulation, and the improper delivery of drugs toward the tumor site. The unique traits of
stem cells (SCs) such as self-renewal, differentiation, tumor tropism, the release of bioactive
molecules, and immunosuppression have opened a new window for utilizing SCs as a novel tool in
cancer treatment. In this regard, engineered SCs can secrete anti-cancer proteins or express enzymes
used in suicide gene therapy which locally induce apoptosis in neoplastic cells via the bystander
effect. These cells also stand as proper candidates to serve as careers for drug-loaded nanoparticles
or to play suitable hosts for oncolytic viruses. Moreover, they harbor great potential to be
employed in immunotherapy and combination therapy. However, tactful strategies should be devised
to allow easier transplantation and protection of SCs from in vivo immune responses. In spite
of the great hope concerning SCs application in cancer therapy, there are shortcomings and challenges
to be addressed. This review tends to elaborate on recent advances on the various applications
of SCs in cancer therapy and existing challenges in this regard.
Collapse
Affiliation(s)
- Malikeh Rad Niknam
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Farnoosh Attari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
36
|
Attia N, Mashal M, Pemminati S, Omole A, Edmondson C, Jones W, Priyadarshini P, Mughal T, Aziz P, Zenick B, Perez A, Lacken M. Cell-Based Therapy for the Treatment of Glioblastoma: An Update from Preclinical to Clinical Studies. Cells 2021; 11:116. [PMID: 35011678 PMCID: PMC8750228 DOI: 10.3390/cells11010116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 01/12/2023] Open
Abstract
Glioblastoma (GB), an aggressive primary tumor of the central nervous system, represents about 60% of all adult primary brain tumors. It is notorious for its extremely low (~5%) 5-year survival rate which signals the unsatisfactory results of the standard protocol for GB therapy. This issue has become, over time, the impetus for the discipline of bringing novel therapeutics to the surface and challenging them so they can be improved. The cell-based approach in treating GB found its way to clinical trials thanks to a marvelous number of preclinical studies that probed various types of cells aiming to combat GB and increase the survival rate. In this review, we aimed to summarize and discuss the up-to-date preclinical studies that utilized stem cells or immune cells to treat GB. Likewise, we tried to summarize the most recent clinical trials using both cell categories to treat or prevent recurrence of GB in patients. As with any other therapeutics, cell-based therapy in GB is still hampered by many drawbacks. Therefore, we highlighted several novel techniques, such as the use of biomaterials, scaffolds, nanoparticles, or cells in the 3D context that may depict a promising future when combined with the cell-based approach.
Collapse
Affiliation(s)
- Noha Attia
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
- Laboratory of Pharmaceutics, NanoBioCel Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
- Histology and Cell Biology Department, Faculty of Medicine, University of Alexandria, Alexandria 21561, Egypt
| | - Mohamed Mashal
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
- Laboratory of Pharmaceutics, NanoBioCel Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
| | - Sudhakar Pemminati
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Adekunle Omole
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Carolyn Edmondson
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Will Jones
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Priyanka Priyadarshini
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Temoria Mughal
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Pauline Aziz
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Blesing Zenick
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Ambar Perez
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Morgan Lacken
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| |
Collapse
|
37
|
Kułach N, Pilny E, Cichoń T, Czapla J, Jarosz-Biej M, Rusin M, Drzyzga A, Matuszczak S, Szala S, Smolarczyk R. Mesenchymal stromal cells as carriers of IL-12 reduce primary and metastatic tumors of murine melanoma. Sci Rep 2021; 11:18335. [PMID: 34526531 PMCID: PMC8443548 DOI: 10.1038/s41598-021-97435-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 08/20/2021] [Indexed: 01/14/2023] Open
Abstract
Due to immunosuppressive properties and confirmed tropism towards cancer cells mesenchymal stromal cells (MSC) have been used in many trials. In our study we used these cells as carriers of IL-12 in the treatment of mice with primary and metastatic B16-F10 melanomas. IL-12 has confirmed anti-cancer activity, induces a strong immune response against cancer cells and acts as an anti-angiogenic agent. A major limitation of the use of IL-12 in therapy is its systemic toxicity. The aim of the work was to develop a system in which cytokine may be administered intravenously without toxic side effects. In this study MSC were used as carriers of the IL-12. We confirmed antitumor effectiveness of the cells secreting IL-12 (MSC/IL-12) in primary and metastatic murine melanoma models. We observed inhibition of tumor growth and a significant reduction in the number of metastases in mice after MSC/IL-12 administration. MSC/IL-12 decreased vascular density and increased the number of anticancer M1 macrophages and CD8+ cytotoxic T lymphocytes in tumors of treated mice. To summarize, we showed that MSC are an effective, safe carrier of IL-12 cytokine. Administered systemically they exert therapeutic properties of IL-12 cytokine without toxicity. Therapeutic effect may be a result of pleiotropic (proinflammatory and anti-angiogenic) properties of IL-12 released by modified MSC.
Collapse
Affiliation(s)
- Natalia Kułach
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Ewelina Pilny
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Magdalena Jarosz-Biej
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Marek Rusin
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Alina Drzyzga
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Sybilla Matuszczak
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Stanisław Szala
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| |
Collapse
|
38
|
Immunomodulatory Arming Factors-The Current Paradigm for Oncolytic Vectors Relies on Immune Stimulating Molecules. Int J Mol Sci 2021; 22:ijms22169051. [PMID: 34445759 DOI: 10.3390/ijms22169051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/15/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
The dogma of engineering oncolytic viral vectors has shifted from emphasizing the viral lysis of individual cancer cells to the recruitment and coordination of the adaptive immune system to clear the tumor. To accomplish this, researchers have been adding several classes of transgenes to their preferred viral platforms. The most prevalent of these include antibodies and targeting moieties, interleukins and cytokines, and genes which rely on small molecule co-administration for tumor killing. Most current vectors rely exclusively on one of these types of transgenes to elicit the desired immune response to clear tumors, but are not mutually exclusive, with several larger OVs armed with several of these factors. The common theme of emerging armed vectors is to simply initiate or enhance infiltration of effector CD8+ T cells to clear the tumor locally at OV infection sites, and systemically throughout the body where the OV has not infected tumor cells. The precision of oncolytic vectors to target a cell type or tissue remains its key advantage over small-molecule drugs. Unlike chemo- and other drug therapies, viral vectors can be made to specifically infect and grow within tumor cells. This ensures localized expression of the therapeutic transgene to the diseased tissue, thereby limiting systemic toxicity. This review will examine the immunomodulating transgenes of current OVs, describe their general effect on the immune system, and provide the rationale for each vector's use in clearing its targeted tumor.
Collapse
|
39
|
Mohiuddin E, Wakimoto H. Extracellular matrix in glioblastoma: opportunities for emerging therapeutic approaches. Am J Cancer Res 2021; 11:3742-3754. [PMID: 34522446 PMCID: PMC8414390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 06/13/2023] Open
Abstract
Extracellular matrix is a complex network of macromolecules that constitute a microenvironment of normal tissues and malignancies such as the primary brain tumor glioblastoma (GBM). The unique composition of the GBM ECM, compared with the brain, contributes to angiogenesis, invasion, and therapeutic resistance of GBM. On the other hand, components of tumor ECM and related aberrant signaling pathways offer opportunities for various therapeutic strategies that are under active investigations. Here we provide a comprehensive overview of emerging therapeutic approaches for GBM that target or utilize its unique ECM via antibodies or ligands, RNA interference, pharmacological agents and modification of ECM molecules. Furthermore, drug-loaded nanoparticles displaying ECM-directed antibodies or peptides enable tumor selective delivery of the payload. As an in vitro research platform, 3D tumor cell culture incorporating ECM can advance our understanding of tumor-ECM interactions.
Collapse
Affiliation(s)
- Enaya Mohiuddin
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School Boston, MA 02114, USA
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School Boston, MA 02114, USA
| |
Collapse
|
40
|
Glioblastoma Therapy: Rationale for a Mesenchymal Stem Cell-based Vehicle to Carry Recombinant Viruses. Stem Cell Rev Rep 2021; 18:523-543. [PMID: 34319509 DOI: 10.1007/s12015-021-10207-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2021] [Indexed: 12/12/2022]
Abstract
Evasion of growth suppression is among the prominent hallmarks of cancer. Phosphatase and tensin homolog (PTEN) and p53 tumor-suppressive pathways are compromised in most human cancers, including glioblastoma (GB). Hence, these signaling pathways are an ideal point of focus for novel cancer therapeutics. Recombinant viruses can selectivity kill cancer cells and carry therapeutic genes to tumors. Specifically, oncolytic viruses (OV) have been successfully employed for gene delivery in GB animal models and showed potential to neutralize immunosuppression at the tumor site. However, the associated systemic immunogenicity, inefficient transduction of GB cells, and inadequate distribution to metastatic tumors have been the major bottlenecks in clinical studies. Mesenchymal stem cells (MSCs), with tumor-tropic properties and immune privilege, can improve OVs targeting. Remarkably, combining the two approaches can address their individual issues. Herein, we summarize findings to advocate the reactivation of tumor suppressors p53 and PTEN in GB treatment and use MSCs as a "Trojan horse" to carry oncolytic viral cargo to disseminated tumor beds. The integration of MSCs and OVs can emerge as the new paradigm in cancer treatment.
Collapse
|
41
|
Wang Y, Gao W. A label-free and sensitive fluorescence assay for hyaluronidase activity through electrostatic-controlled quantum dots self-assembly. JOURNAL OF CHEMICAL RESEARCH 2021. [DOI: 10.1177/17475198211018973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A label-free fluorescence assay for hyaluronidase (HAase) activity based on self-assembly of quantum dots is developed. A cationic polymer (polycation) can induce aggregation of the negatively charged quantum dots through electrostatic interactions and the fluorescence of the quantum dots is quenched. When the polycation is mixed with hyaluronic acid (HA), intense binding of HA to the polycation makes the quantum dots free and recovery of the fluorescence of the quantum dots is observed. However, in the presence of HAase, HA is hydrolyzed into small fragments and the polycation induces reaggregation of the quantum dots. A simple and rapid fluorescence sensor with high sensitivity and selectivity for HAase activity detection is therefore successfully established with a detection limit of 0.01 U/mL. Moreover, we have demonstrated an assay that can be applied to detect HAase activity in a complex mixture sample including 1% human serum.
Collapse
Affiliation(s)
- Yan Wang
- School of Chemistry and Chemical Engineering, Yulin University, Yulin, People’s Republic of China
| | - Wenwen Gao
- School of Chemistry and Chemical Engineering, Yulin University, Yulin, People’s Republic of China
| |
Collapse
|
42
|
Nowak B, Rogujski P, Janowski M, Lukomska B, Andrzejewska A. Mesenchymal stem cells in glioblastoma therapy and progression: How one cell does it all. Biochim Biophys Acta Rev Cancer 2021; 1876:188582. [PMID: 34144129 DOI: 10.1016/j.bbcan.2021.188582] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) are among the most investigated and applied somatic stem cells in experimental therapies for the regeneration of damaged tissues. Moreover, as it was recently postulated, MSCs may demonstrate anti-tumor properties. Glioblastoma (GBM) is a grade IV central nervous system tumor with no available effective therapy and an inevitably fatal prognosis. Experimental studies utilizing MSCs in GBM treatment resulted in numerous controversies. Native MSCs were shown to exert anti-GBM activity by controlling angiogenesis, regulating cell cycle, and inducing apoptosis. They also were used as sensitizing factors and vehicles delivering various anti-cancer compounds. On the other hand, some experiments revealed significant risks related to MSC-based therapies for GBM, such as enhancement of tumor cell proliferation, invasion, and aggressiveness. The following review elaborates on all mentioned contradictory data and provides a realistic, current clinical perspective on MSCs' potential in GBM treatment.
Collapse
Affiliation(s)
- Blazej Nowak
- Department of Neurosurgery, Central Clinical Hospital of Ministry of the Interior and Administration, Warsaw, Poland; Neurosurgery Department, John Paul II Western Hospital, Grodzisk Mazowiecki, Poland
| | - Piotr Rogujski
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Miroslaw Janowski
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA; Tumor Immunology and Immunotherapy Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Andrzejewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
43
|
Banijamali RS, Soleimanjahi H, Soudi S, Karimi H. Mesenchymal stem cells support delivery and boost the efficacy of oncolytic reoviruses in TC-1 tumor cells. J Cell Biochem 2021; 122:1360-1375. [PMID: 34056765 DOI: 10.1002/jcb.29955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 11/08/2022]
Abstract
Cancer has remained a major health problem around the world. Mesenchymal stem cells (MSCs)-based therapy exhibits a therapeutic effect via different mechanisms. By using MSCs as carrier cells, the major problem of clearance of oncolytic viruses is resolved by neutralizing antibodies before they react with cancer cells. The aim of this study was to characterize the effect of infected MSCs by reovirus type-3 Dearing (T3D) for in vitro cancer therapy. Adipose-derived MSCs (AD-MSCs) were infected with reovirus T3D and its biological properties were evaluated. Then, the effects of reovirus-infected AD-MSCs on cytokine profile, nitric oxide (NO) production, and apoptosis induction in TC-1 cells were assessed. Our results indicated that the differentiation potential of AD-MSCs was affected by reovirus. However, phenotypes were not affected after infection. Then, the effects of reovirus-infected AD-MSCs in TC-1 cells showed an increased amount of tumor necrosis factor-alpha (TNF-α) and NO production and a decreased amount of transforming growth factor-beta 1 (TGF-β1) and interleukin-10 (IL-10). Moreover, apoptosis significantly increased via coculturing of TC-1 cells with infected AD-MSCs, compared with control, and both internal and external apoptosis pathways are activated in experimental groups. In conclusion, the data showed that with increasing TNF-α and NO production and reducing IL-10 and TGF-β production, AD-MSCs can enhance the oncolytic effect of reovirus in cancer cells. Furthermore, the results suggested that AD-MSCs can be used as effective carrier cells candidate for reovirus T3D to maximize their anticancer cell activity.
Collapse
Affiliation(s)
- Razieh S Banijamali
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hesam Karimi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
44
|
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: 13] [Impact Index Per Article: 3.3] [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
|
45
|
Zhang Y, Li Y, Chen K, Qian L, Wang P. Oncolytic virotherapy reverses the immunosuppressive tumor microenvironment and its potential in combination with immunotherapy. Cancer Cell Int 2021; 21:262. [PMID: 33985527 PMCID: PMC8120729 DOI: 10.1186/s12935-021-01972-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
It has been intensively reported that the immunosuppressive tumor microenvironment (TME) results in tumor resistance to immunotherapy, especially immune checkpoint blockade and chimeric T cell antigen therapy. As an emerging therapeutic agent, oncolytic viruses (OVs) can specifically kill malignant cells and modify immune and non-immune TME components through their intrinsic properties or genetically incorporated with TME regulators. Strategies of manipulating OVs against the immunosuppressive TME include serving as a cancer vaccine, expressing proinflammatory factors and immune checkpoint inhibitors, and regulating nonimmune stromal constituents. In this review, we summarized the mechanisms and applications of OVs against the immunosuppressive TME, and strategies of OVs in combination with immunotherapy. We also introduced future directions to achieve efficient clinical translation including optimization of preclinical models that simulate the human TME and achieving systemic delivery of OVs.
Collapse
Affiliation(s)
- Yalei Zhang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ye Li
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
| | - Kun Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ling Qian
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Peng Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
46
|
Seyed-Khorrami SM, Soleimanjahi H, Soudi S, Habibian A. MSCs loaded with oncolytic reovirus: migration and in vivo virus delivery potential for evaluating anti-cancer effect in tumor-bearing C57BL/6 mice. Cancer Cell Int 2021; 21:244. [PMID: 33933086 PMCID: PMC8088007 DOI: 10.1186/s12935-021-01848-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/20/2021] [Indexed: 02/07/2023] Open
Abstract
Background and aims Several oncolytic viruses applications have been approved in the clinic or in different phases of clinical trials. However, these methods have some rudimentary problems. Therefore, to enhance the delivery and quality of treatment, considering the advantage of cell carrier-based methods such as Mesenchymal Stem Cells (MSC) have been proposed. This study was designed to evaluate the performance and quality of cancer treatment based on MSCs loaded by oncolytic reovirus in the cancerous C57BL/6 mouse model. Also, we evaluated MSCs migration potency in vitro and in vivo following the oncolytic reovirus infection. Methods C57BL/6 mice were inoculated with TC-1 cell lines and tumors were established in the right flank. Mice were systemically treated with reovirus, MSCs-loaded with reovirus, MSCs, and PBS as a control in separated groups. Effects of infected AD-MSCs with reovirus on tumor growth and penetration in the tumor site were monitored. All groups of mice were monitored for two months in order to therapeutic and anticancer potential. After treatments, tumor size alteration and apoptosis rate, as well as cytokine release pattern was assessed. Results The results of the current study indicated that the effect of reovirus infection on AD-MSCs is not devastating the migration capacity especially in MOI 1 and 5 while intact cells remain. On the other hand, MSCs play an efficient role as a carrier to deliver oncolytic virus into the tumor site in comparison with systemic administration of reovirus alone. Apoptosis intensity relies on viral titration and passing time. Followed by systemic administration, treatment with oncolytic reovirus-infected AD-MSCs and MSCs alone had shown significant inhibition in tumor growth. Also, treatment by reovirus causes an increase in IFN-γ secretion. Conclusion The results of in vitro and in vivo study confirmed the tumor-homing properties of infected AD-MSCs and the significant antitumor activity of this platform. Hence, our results showed that the cell carrier strategy using oncolytic reovirus-loaded AD-MSCs enhanced virus delivery, infiltration, and antitumor activity can be effectively applied in most cancers.
Collapse
Affiliation(s)
| | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ala Habibian
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
47
|
Santos J, Heiniö C, Quixabeira D, Zafar S, Clubb J, Pakola S, Cervera-Carrascon V, Havunen R, Kanerva A, Hemminki A. Systemic Delivery of Oncolytic Adenovirus to Tumors Using Tumor-Infiltrating Lymphocytes as Carriers. Cells 2021; 10:cells10050978. [PMID: 33922052 PMCID: PMC8143525 DOI: 10.3390/cells10050978] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/10/2021] [Accepted: 04/19/2021] [Indexed: 01/29/2023] Open
Abstract
Immunotherapy with tumor-infiltrating lymphocytes (TIL) or oncolytic adenoviruses, have shown promising results in cancer treatment, when used as separate therapies. When used in combination, the antitumor effect is synergistically potentiated due oncolytic adenovirus infection and its immune stimulating effects on T cells. Indeed, studies in hamsters have shown a 100% complete response rate when animals were treated with oncolytic adenovirus coding for TNFa and IL-2 (Ad5/3-E2F-D24-hTNFa-IRES-hIL2; TILT-123) and TIL therapy. In humans, one caveat with oncolytic virus therapy is that intratumoral injection has been traditionally preferred over systemic administration, for achieving sufficient virus concentrations in tumors, especially when neutralizing antibodies emerge. We have previously shown that 5/3 chimeric oncolytic adenovirus can bind to human lymphocytes for avoidance of neutralization. In this study, we hypothesized that incubation of oncolytic adenovirus (TILT-123) with TILs prior to systemic injection would allow delivery of virus to tumors. This approach would deliver both components in one self-amplifying product. TILs would help deliver TILT-123, whose replication will recruit more TILs and increase their cytotoxicity. In vitro, TILT-123 was seen binding efficiently to lymphocytes, supporting the idea of dual administration. We show in vivo in different models that virus could be delivered to tumors with TILs as carriers.
Collapse
Affiliation(s)
- Joao Santos
- Cancer Gene Therapy Group, Faculty of Medicine, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland; (J.S.); (C.H.); (D.Q.); (S.Z.); (J.C.); (S.P.); (V.C.-C.); (R.H.); (A.K.)
- TILT Biotherapeutics Ltd., 00290 Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Faculty of Medicine, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland; (J.S.); (C.H.); (D.Q.); (S.Z.); (J.C.); (S.P.); (V.C.-C.); (R.H.); (A.K.)
| | - Dafne Quixabeira
- Cancer Gene Therapy Group, Faculty of Medicine, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland; (J.S.); (C.H.); (D.Q.); (S.Z.); (J.C.); (S.P.); (V.C.-C.); (R.H.); (A.K.)
| | - Sadia Zafar
- Cancer Gene Therapy Group, Faculty of Medicine, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland; (J.S.); (C.H.); (D.Q.); (S.Z.); (J.C.); (S.P.); (V.C.-C.); (R.H.); (A.K.)
| | - James Clubb
- Cancer Gene Therapy Group, Faculty of Medicine, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland; (J.S.); (C.H.); (D.Q.); (S.Z.); (J.C.); (S.P.); (V.C.-C.); (R.H.); (A.K.)
- TILT Biotherapeutics Ltd., 00290 Helsinki, Finland
| | - Santeri Pakola
- Cancer Gene Therapy Group, Faculty of Medicine, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland; (J.S.); (C.H.); (D.Q.); (S.Z.); (J.C.); (S.P.); (V.C.-C.); (R.H.); (A.K.)
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Faculty of Medicine, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland; (J.S.); (C.H.); (D.Q.); (S.Z.); (J.C.); (S.P.); (V.C.-C.); (R.H.); (A.K.)
- TILT Biotherapeutics Ltd., 00290 Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Faculty of Medicine, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland; (J.S.); (C.H.); (D.Q.); (S.Z.); (J.C.); (S.P.); (V.C.-C.); (R.H.); (A.K.)
- TILT Biotherapeutics Ltd., 00290 Helsinki, Finland
| | - Anna Kanerva
- Cancer Gene Therapy Group, Faculty of Medicine, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland; (J.S.); (C.H.); (D.Q.); (S.Z.); (J.C.); (S.P.); (V.C.-C.); (R.H.); (A.K.)
- Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, 00290 Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Faculty of Medicine, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland; (J.S.); (C.H.); (D.Q.); (S.Z.); (J.C.); (S.P.); (V.C.-C.); (R.H.); (A.K.)
- TILT Biotherapeutics Ltd., 00290 Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland
- Correspondence:
| |
Collapse
|
48
|
Shin DH, Nguyen T, Ozpolat B, Lang F, Alonso M, Gomez-Manzano C, Fueyo J. Current strategies to circumvent the antiviral immunity to optimize cancer virotherapy. J Immunother Cancer 2021; 9:jitc-2020-002086. [PMID: 33795384 PMCID: PMC8021759 DOI: 10.1136/jitc-2020-002086] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer virotherapy is a paradigm-shifting treatment modality based on virus-mediated oncolysis and subsequent antitumor immune responses. Clinical trials of currently available virotherapies showed that robust antitumor immunity characterizes the remarkable and long-term responses observed in a subset of patients. These data suggest that future therapies should incorporate strategies to maximize the immunotherapeutic potential of oncolytic viruses. In this review, we highlight the recent evidence that the antiviral immunity of the patients may limit the immunotherapeutic potential of oncolytic viruses and summarize the most relevant approaches to strategically redirect the immune response away from the viruses and toward tumors to heighten the clinical impact of viro-immunotherapy platforms.
Collapse
Affiliation(s)
- Dong Ho Shin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Teresa Nguyen
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Frederick Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marta Alonso
- Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
49
|
Li ZX, Zhang JL, Wang J, Luo F, Qiu B, Guo LH, Lin ZY. A Novel Enzyme-Responded Controlled Release Electrochemical Biosensor for Hyaluronidase Activity Detection. JOURNAL OF ANALYSIS AND TESTING 2021. [DOI: 10.1007/s41664-021-00158-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
50
|
Raj P, Lee SY, Lee TY. Carbon Dot/Naphthalimide Based Ratiometric Fluorescence Biosensor for Hyaluronidase Detection. MATERIALS 2021; 14:ma14051313. [PMID: 33803381 PMCID: PMC7967242 DOI: 10.3390/ma14051313] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/27/2021] [Accepted: 03/07/2021] [Indexed: 12/14/2022]
Abstract
Bladder cancer is the leading cause of death in patients with genitourinary cancer. An elevated level of hyaluronidase (HAase) was found in bladder cancer, which acts as an important biomarker for the early diagnosis of bladder cancer. Hence, there is a need to develop a simple enzymatic assay for the early recognition of HAase. Herein, we report a simple, sensitive, and ratiometric fluorescence assay for HAase detection under physiological conditions. The fluorescence assay was constructed by the adsorption of cationic carbon dots and positively charged naphthalimide on negatively charged hyaluronic acid and the development of a Förster resonance energy transfer (FRET) mechanism from carbon dots to a naphthalimide fluorophores. The hyaluronidase enzyme cleaves the hyaluronic acid in this assay, and breaking down the FRET mechanism induces ratiometric changes. A detection limit of 0.09 U/mL was achieved, which is less than the HAase level found in normal human body fluids. Moreover, this assay may be used for diagnosing HAase-related diseases.
Collapse
Affiliation(s)
- Pushap Raj
- Department of Convergence System Engineering and Department of Biomedical Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea;
| | - Seon-yeong Lee
- Department of Technology Education, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea;
| | - Tae Yoon Lee
- Department of Convergence System Engineering and Department of Biomedical Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea;
- Department of Technology Education, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea;
- Correspondence:
| |
Collapse
|