1
|
Kim JH, Yang HJ, Lee SH, Song YS. Cytosine Deaminase-Overexpressing hTERT-Immortalized Human Adipose Stem Cells Enhance the Inhibitory Effects of Fluorocytosine on Tumor Growth in Castration Resistant Prostate Cancer. Int J Mol Sci 2024; 25:5519. [PMID: 38791557 PMCID: PMC11121865 DOI: 10.3390/ijms25105519] [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: 02/12/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
A promising de novo approach for the treatment of Castration-resistant prostate cancer (CRPC) exploits cell-mediated enzyme prodrug therapy comprising cytosine deaminase (CD) and fluorouracil (5-FC). The aim of this study was to determine the potential of bacterial CD-overexpressing hTERT-immortalized human adipose stem cells (hTERT-ADSC.CD) to suppress CRPC. A lentiviral vector encoding a bacterial CD gene was used to transfect and to generate the hTERT-ADSC.CD line. The ability of the cells to migrate selectively towards malignant cells was investigated in vitro. PC3 and hTERT-ADSC.CD cells were co-cultured. hTERT-ADSC.CD and 1 × 106 PC3 cells were administered to nude mice via intracardiac and subcutaneous injections, respectively, and 5-FC was given for 14 days. hTERT-ADSC.CD were successfully engineered. Enhanced in vitro hTERT-ADSC.CD cytotoxicity and suicide effect were evident following administration of 5 μM 5-FC. hTERT-ADSC.CD, together with 5-FC, augmented the numbers of PC3 cells undergoing apoptosis. In comparison to controls administered hTERT-ADSC.CD monotherapy, hTERT-ADSC.CD in combination with 5-FC demonstrated a greater suppressive effect on tumor. In CPRC-bearing mice, tumor suppression was enhanced by the combination of CD-overexpressing ADSC and the prodrug 5-FC. Stem cells exhibiting CD gene expression are a potential novel approach to treatment for CRPC.
Collapse
Affiliation(s)
- Jae Heon Kim
- Department of Urology, Soonchunhyang University School of Medicine, Seoul 04401, Republic of Korea
| | - Hee Jo Yang
- Department of Urology, Soonchunhyang University School of Medicine, Cheonan 31151, Republic of Korea
| | - Sang Hun Lee
- Program in Biomedical Sciences & Engineering, Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Yun Seob Song
- Department of Urology, Soonchunhyang University School of Medicine, Seoul 04401, Republic of Korea
| |
Collapse
|
2
|
Merino JJ, Cabaña-Muñoz ME. Nanoparticles and Mesenchymal Stem Cell (MSC) Therapy for Cancer Treatment: Focus on Nanocarriers and a si-RNA CXCR4 Chemokine Blocker as Strategies for Tumor Eradication In Vitro and In Vivo. MICROMACHINES 2023; 14:2068. [PMID: 38004925 PMCID: PMC10673568 DOI: 10.3390/mi14112068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/07/2023] [Accepted: 10/13/2023] [Indexed: 11/26/2023]
Abstract
Mesenchymal stem cells (MSCs) have a high tropism for the hypoxic microenvironment of tumors. The combination of nanoparticles in MSCs decreases tumor growth in vitro as well as in rodent models of cancers in vivo. Covalent conjugation of nanoparticles with the surface of MSCs can significantly increase the drug load delivery in tumor sites. Nanoparticle-based anti-angiogenic systems (gold, silica and silicates, diamond, silver, and copper) prevented tumor growth in vitro. For example, glycolic acid polyconjugates enhance nanoparticle drug delivery and have been reported in human MSCs. Labeling with fluorescent particles (coumarin-6 dye) identified tumor cells using fluorescence emission in tissues; the conjugation of different types of nanoparticles in MSCs ensured success and feasibility by tracking the migration and its intratumor detection using non-invasive imaging techniques. However, the biosafety and efficacy; long-term stability of nanoparticles, and the capacity for drug release must be improved for clinical implementation. In fact, MSCs are vehicles for drug delivery with nanoparticles and also show low toxicity but inefficient accumulation in tumor sites by clearance of reticuloendothelial organs. To solve these problems, the internalization or conjugation of drug-loaded nanoparticles should be improved in MSCs. Finally, CXCR4 may prove to be a promising target for immunotherapy and cancer treatment since the delivery of siRNA to knock down this alpha chemokine receptor or CXCR4 antagonism has been shown to disrupt tumor-stromal interactions.
Collapse
Affiliation(s)
- José Joaquín Merino
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid (U.C.M.), 28040 Madrid, Spain
| | | |
Collapse
|
3
|
Askari N, Asadi F, Nazer A, Falahati-pour SK. Anti-aging effects of the pistachio Extract on Mesenchymal Stem Cells proliferation and telomerase activity. Arch Gerontol Geriatr 2023; 111:105016. [PMID: 37031654 DOI: 10.1016/j.archger.2023.105016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/16/2023] [Accepted: 04/02/2023] [Indexed: 04/05/2023]
Abstract
PURPOSE Using mesenchymal stem cells (MSCs) is a promising method in regenerative medicine. Limited proliferation and aging process of MSC are the most common problems in MSCs application. In the present study, we intend to investigate the anti-aging properties of pistachio pericarp in bone marrow-derived MSCs of old male rats. MATERIALS AND METHODS First, 1000, 2000, and 3000 µg/mL AEPP were used to treat MSCs derived from bone marrow for 24 h at 37 °C. Then, cell viability, population doubling time, the percentage of senescent cells, telomere length, telomerase activity, and the expression of TRF1 and RAP1 when bone marrow-derived MSCs treated with AEPP were investigated. RESULTS The results showed that cell viability increased when MSCs derived from bone marrow were treated with 2000 and 3000 µg/mL AEPP, indicating this extract may stimulate proliferation. The population doubling time was also enhanced with an increase in AEPP concentration. Importantly, an increase in AEPP concentration significantly reduced senescent cell percentage. Telomere length, telomerase activity, and the expression of anti-aging genes were significantly increased with the increase of AEPP dose. CONCLUSION Taken together, AEPP has been used as a natural compound with excellent proliferation and anti-aging ability in MSCs. As new therapeutic candidates with promising effects, it can be used with high safety by multiplying cells and delaying the aging process. However, more studies are needed and the anti-aging effects of this extract should be well confirmed in animal models and clinical trials.
Collapse
|
4
|
Kim JH, Bak SH, Yang HJ, Doo SW, Kim DK, Yang WJ, Kim SU, Lee HJ, Song YS. Improvement of erectile dysfunction using endothelial progenitor cells from fetal cerebral vasculature in the cavernous nerve injury of rats. Basic Clin Androl 2022; 32:21. [PMID: 36451096 PMCID: PMC9714194 DOI: 10.1186/s12610-022-00171-x] [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: 07/09/2022] [Accepted: 09/05/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Because of limited differentiation to endothelium from mesenchymal stem cells, it has been strongly recommended to use endothelial progenitor cells for the regeneration of the damaged endothelium of corpora cavernosa. This study was performed to investigate the immortalized human cerebral endothelial cells and their capability for repairing erectile dysfunction in a rat model of cavernous nerve injury. Circulating endothelial progenitor cells were isolated from human fetal brain vasculature at the periventricular region of telencephalic tissues. Over 95% of CD 31-positive cells were sorted and cultured for 10 days. Human cerebral endothelial progenitor cells were injected into the cavernosa of rats with cavernous nerve injury. Erectile response was then assessed. In in vivo assays, rats were divided into three groups: group 1, sham operation: group 2, bilateral cavernous nerve injury: and group 3, treatment with human cerebral endothelial cells after cavernous nerve injury. RESULTS Established immortalized circulating endothelial progenitor cells showed expression of human telomerase reverse transcriptase transcript by RT-PCR. They also showed the expression of vascular endothelial growth factor, von Willebrand factor, vascular endothelial growth factor receptor, and CD31, cell type-specific markers for endothelial cells by RT-PCR. In in vitro angiogenesis assays, they demonstrated tube formation that suggested morphological properties of endothelial progenitor cells. In in vivo assays, impaired erectile function of rat with cavernous nerve injury recovered at 2, 4, and 12 weeks after transplantation of human cerebral endothelial cells into the cavernosa. CONCLUSIONS Telomerase reverse transcriptase-circulating endothelial progenitor cells from fetal brain vasculature could repair erectile dysfunction of rats with cavernous nerve injury.
Collapse
Affiliation(s)
- Jae Heon Kim
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, 04401 Seoul, Republic of Korea
| | - Sang Hong Bak
- Research Institute, e-Biogen Inc., Seoul, Republic of Korea
| | - Hee Jo Yang
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, Cheonan, Republic of Korea
| | - Seung Whan Doo
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, 04401 Seoul, Republic of Korea
| | - Do Kyung Kim
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, 04401 Seoul, Republic of Korea
| | - Won Jae Yang
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, 04401 Seoul, Republic of Korea
| | - Seung U. Kim
- grid.416957.80000 0004 0633 8774Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, Canada
| | - Hong J. Lee
- Research Institute, e-Biogen Inc., Seoul, Republic of Korea ,grid.254229.a0000 0000 9611 0917Medical Research Institute, Chungbuk National University, Cheongju, Chungbuk Republic of Korea
| | - Yun Seob Song
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, 04401 Seoul, Republic of Korea
| |
Collapse
|
5
|
Keshavarz M, Mohammad Miri S, Behboudi E, Arjeini Y, Dianat-Moghadam H, Ghaemi A. Oncolytic virus delivery modulated immune responses toward cancer therapy: Challenges and perspectives. Int Immunopharmacol 2022; 108:108882. [PMID: 35623296 DOI: 10.1016/j.intimp.2022.108882] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/11/2022] [Accepted: 05/18/2022] [Indexed: 11/05/2022]
Abstract
Oncolytic viruses (OVs) harness the hallmarks of tumor cells and cancer-related immune responses for the lysis of malignant cells, modulation of the tumor microenvironment, and exertion of vaccine-like activities. However, efficient clinical exploitation of these potent therapeutic modules requires their systematic administration, especially against metastatic and solid tumors. Therefore, developing methods for shielding a virus from the neutralizing environment of the bloodstream while departing toward tumor sites is a must. This paper reports the latest advancements in the employment of chemical and biological compounds aimed at safe and efficient delivery of OVs to target tissues or tumor deposits within the host.
Collapse
Affiliation(s)
- Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Seyed Mohammad Miri
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran.
| | - Emad Behboudi
- Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Yaser Arjeini
- Department of Research and Development, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran.
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Amir Ghaemi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
6
|
Sarwar A, Hashim L, Faisal MS, Haider MZ, Ahmed Z, Ahmed TF, Shahzad M, Ansar I, Ali S, Aslam MM, Anwer F. Advances in viral oncolytics for treatment of multiple myeloma - a focused review. Expert Rev Hematol 2021; 14:1071-1083. [PMID: 34428997 DOI: 10.1080/17474086.2021.1972802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Oncolytic viruses are genetically engineered viruses that target myeloma-affected cells by detecting specific cell surface receptors (CD46, CD138), causing cell death by activating the signaling pathway to induce apoptosis or by immune-mediated cellular destruction. AREAS COVERED This article summarizes oncolytic virotherapy advancements such as the therapeutic use of viruses by targeting cell surface proteins of myeloma cells as well as the carriers to deliver viruses to the target tissues safely. The major classes of viruses that have been studied for this include measles, myxoma, adenovirus, reovirus, vaccinia, vesicular-stomatitis virus, coxsackie, and others. The measles virus acts as oncolytic viral therapy by binding to the CD46 receptors on the myeloma cells to utilize its surface H protein. These H-protein and CD46 interactions lead to cellular syncytia formation resulting in cellular apoptosis. Vesicular-stomatitis virus acts by downregulation of anti-apoptotic factors (Mcl-2, BCL-2). Based upon the published literature searches till December 2020, we have summarized the data supporting the advances in viral oncolytic for the treatment of MM. EXPERT OPINION Oncolytic virotherapy is an experimental approach in multiple myeloma (MM); many issues need to be addressed for safe viral delivery to the target tissue.
Collapse
Affiliation(s)
- Ayesha Sarwar
- Department of Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | | | - Muhammad Salman Faisal
- Department of Internal Medicine, Division of Hematology, The Ohio State University Columbus Oh, USA
| | | | - Zahoor Ahmed
- Department of Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | - Tehniat Faraz Ahmed
- Department of Biochemistry, Dow University of Health Sciences, Karachi, Pakistan
| | - Moazzam Shahzad
- Department of Internal Medicine, St Mary's Medical Center, Huntington, WV, USA
| | - Iqraa Ansar
- Department of Internal medicine, Riverside Methodist hospital, Columbus OH
| | - Sundas Ali
- Department of Internal medicine, Rawalpindi Medical University, Rawalpindi, Pakistan
| | | | - Faiz Anwer
- Department of Hematology and Oncology, Taussig Cancer Center, Cleveland Clinic, Ohio, USA
| |
Collapse
|
7
|
Su Y, Zhang T, Huang T, Gao J. Current advances and challenges of mesenchymal stem cells-based drug delivery system and their improvements. Int J Pharm 2021; 600:120477. [PMID: 33737099 DOI: 10.1016/j.ijpharm.2021.120477] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/03/2021] [Accepted: 03/07/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) have recently emerged as a promising living carrier for targeted drug delivery. A wealth of literature has shown evidence for great advances in MSCs-based drug delivery system (MSCs-DDS) in the treatment of various diseases. Nevertheless, as this field of study rapidly advances, several challenges associated with this delivery strategy have arisen, mainly due to the inherent limitations of MSCs. To this end, several novel technologies are being developed in parallel to improve the efficiency or safety of this system. In this review, we introduce recent advances and summarize the present challenges of MSCs-DDS. We also highlight some potential technologies to improve MSCs-DDS, including nanotechnology, genome engineering technology, and biomimetic technology. Finally, prospects for application of artificially improved MSCs-DDS are addressed. The technologies summarized in this review provide a general guideline for the improvement of MSCs-DDS.
Collapse
Affiliation(s)
- Yuanqin Su
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ting Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
8
|
Martini V, D'Avanzo F, Maggiora PM, Varughese FM, Sica A, Gennari A. Oncolytic virotherapy: new weapon for breast cancer treatment. Ecancermedicalscience 2020; 14:1149. [PMID: 33574894 PMCID: PMC7864690 DOI: 10.3332/ecancer.2020.1149] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
The recent introduction of viruses as a weapon against cancer can be regarded as one of the most intriguing approaches in the context of precision medicine. The role of immune checkpoint inhibitors has been extensively studied in early and advanced cancer stages, with extraordinary results. Although there is a good tolerability profile, especially when compared with conventional chemotherapy, severe immune-related adverse events have emerged as a potential limitation. Moreover, there are still treatment-resistant cases and thus further treatment options need to be implemented. Several in vitro and in vivo studies have been conducted and are ongoing to develop oncolytic viruses (OVs) as a tool to modulate the immune system response. OVs are attenuated viruses that can kill cancer cells after having infected them, producing microenvironment remodelling and antitumour immune response. The potential of oncolytic virotherapy is to contrast the absence of T cell infiltrates, converting ‘cold’ tumours into ‘hot’ ones, thus improving the performance of the immune system. Breast cancer, the second most common cause of cancer-related deaths among women, is considered a ‘cold’ tumour. In this context, oncolytic virotherapy might well be considered as a promising strategy. This review summarises the current status, clinical applications and future development of OVs, focusing on breast cancer treatment.
Collapse
Affiliation(s)
- Veronica Martini
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy.,Center for Translational Research on Autoimmune & Allergic Diseases - CAAD, Novara 28100, Italy.,https://orcid.org/0000-0002-0887-4082
| | - Francesca D'Avanzo
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy
| | - Paola Maria Maggiora
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy
| | - Feba Maria Varughese
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy.,Center for Translational Research on Autoimmune & Allergic Diseases - CAAD, Novara 28100, Italy
| | - Antonio Sica
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A Avogadro 28100, Italy.,Department of Inflammation and Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano (MI) 20089, Italy.,https://orcid.org/0000-0002-8342-7442
| | - Alessandra Gennari
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy.,Center for Translational Research on Autoimmune & Allergic Diseases - CAAD, Novara 28100, Italy.,https://orcid.org/0000-0002-0928-2281
| |
Collapse
|
9
|
Hung HS, Yu AYH, Hsieh SC, Kung ML, Huang HY, Fu RH, Yeh CA, Hsu SH. Enhanced Biocompatibility and Differentiation Capacity of Mesenchymal Stem Cells on Poly(dimethylsiloxane) by Topographically Patterned Dopamine. ACS APPLIED MATERIALS & INTERFACES 2020; 12:44393-44406. [PMID: 32697572 DOI: 10.1021/acsami.0c05747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Controlling the behavior of mesenchymal stem cells (MSCs) through topographic patterns is an effective approach for stem cell studies. We, herein, reported a facile method to create a dopamine (DA) pattern on poly(dimethylsiloxane) (PDMS). The topography of micropatterned DA was produced on PDMS after plasma treatment. The grid-topographic-patterned surface of PDMS-DA (PDMS-DA-P) was measured for adhesion force and Young's modulus by atomic force microscopy. The surface of PDMS-DA-P demonstrated less stiff and more elastic characteristics compared to either nonpatterned PDMS-DA or PDMS. The PDMS-DA-P evidently enhanced the differentiation of MSCs into various tissue cells, including nerve, vessel, bone, and fat. We further designed comprehensive experiments to investigate adhesion, proliferation, and differentiation of MSCs in response to PDMS-DA-P and showed that the DA-patterned surface had good biocompatibility and did not activate macrophages or platelets in vitro and had low foreign body reaction in vivo. Besides, it protected MSCs from apoptosis as well as excessive reactive oxygen species (ROS) generation. Particularly, the patterned surface enhanced the differentiation capacity of MSCs toward neural and endothelial cells. The stromal cell-derived factor-1α/CXantiCR4 pathway may be involved in mediating the self-recruitment and promoting the differentiation of MSCs. These findings support the potential application of PDMS-DA-P in either cell treatment or tissue repair.
Collapse
Affiliation(s)
- Huey-Shan Hung
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan, R.O.C
- Translational Medicine Research, China Medical University Hospital, Taichung 40402, Taiwan, R.O.C
| | - Alex Yang-Hao Yu
- Ministry of Health & Welfare, Changhua Hospital, Changhua 51341, Taiwan, R.O.C
| | - Shu-Chen Hsieh
- Department of Chemistry, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan, R.O.C
| | - Mei-Lang Kung
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan, R.O.C
| | - Hsiu-Yuan Huang
- Department of Cosmeceutics and Graduate Institute of Cosmeceutics, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Ru-Huei Fu
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan, R.O.C
- Translational Medicine Research, China Medical University Hospital, Taichung 40402, Taiwan, R.O.C
| | - Chun-An Yeh
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei 10617, Taiwan, R.O.C
| |
Collapse
|
10
|
Gharbavi M, Sharafi A, Ghanbarzadeh S. Mesenchymal Stem Cells: A New Generation of Therapeutic Agents as Vehicles in Gene Therapy. Curr Gene Ther 2020; 20:269-284. [PMID: 32515309 DOI: 10.2174/1566523220666200607190339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/26/2022]
Abstract
In recent years, mesenchymal stem cells (MSCs) as a new tool for therapeutic gene delivery in clinics have attracted much attention. Their advantages cover longer lifespan, better isolation, and higher transfection efficiency and proliferation rate. MSCs are the preferred approach for cell-based therapies because of their in vitro self-renewal capacity, migrating especially to tumor tissues, as well as anti-inflammatory and immunomodulatory properties. Therefore, they have considerable efficiency in genetic engineering for future clinical applications in cancer gene therapy and other diseases. For improving therapeutic efficiency, targeted therapy of cancers can be achieved through the sustained release of therapeutic agents and functional gene expression induction to the intended tissues. The development of a new vector in gene therapy can improve the durability of a transgene expression. Also, the safety of the vector, if administered systemically, may resolve several problems, such as durability of expression and the host immune response. Currently, MSCs are prominent candidates as cell vehicles for both preclinical and clinical trials due to the secretion of therapeutic agents in several cancers. In the present study, we discuss the status of gene therapy in both viral and non-viral vectors along with their limitations. Throughout this study, the use of several nano-carriers for gene therapy is also investigated. Finally, we critically discuss the promising advantages of MSCs in targeted gene delivery, tumor inhibition and their utilization as the gene carriers in clinical situations.
Collapse
Affiliation(s)
- Mahmoud Gharbavi
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan,
Iran,Cancer Gene Therapy Research Center, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan,
Iran,Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Sharafi
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran,Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Saeed Ghanbarzadeh
- Cancer Gene Therapy Research Center, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan,
Iran,Zanjan Pharmaceutical Nanotechnology Research Center and Department of Pharmaceutics, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
11
|
Jayawardena N, Poirier JT, Burga LN, Bostina M. Virus-Receptor Interactions and Virus Neutralization: Insights for Oncolytic Virus Development. Oncolytic Virother 2020; 9:1-15. [PMID: 32185149 PMCID: PMC7064293 DOI: 10.2147/ov.s186337] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 02/09/2020] [Indexed: 12/24/2022] Open
Abstract
Oncolytic viruses (OVs) are replication competent agents that selectively target cancer cells. After penetrating the tumor cell, viruses replicate and eventually trigger cell lysis, releasing the new viral progeny, which at their turn will attack and kill neighbouring cells. The ability of OVs to self-amplify within the tumor while sparing normal cells can provide several advantages including the capacity to encode and locally produce therapeutic protein payloads, and to prime the host immune system. OVs targeting of cancer cells is mediated by host factors that are differentially expressed between normal tissue and tumors, including viral receptors and internalization factors. In this review article, we will discuss the evolution of oncolytic viruses that have reached the stage of clinical trials, their mechanisms of oncolysis, cellular receptors, strategies for targeting cancers, viral neutralization and developments to bypass virus neutralization.
Collapse
Affiliation(s)
- Nadishka Jayawardena
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - John T Poirier
- Department of Medicine and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laura N Burga
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Mihnea Bostina
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Otago Micro and Nano Imaging, University of Otago, Dunedin, New Zealand
| |
Collapse
|
12
|
Galland S, Stamenkovic I. Mesenchymal stromal cells in cancer: a review of their immunomodulatory functions and dual effects on tumor progression. J Pathol 2019; 250:555-572. [PMID: 31608444 PMCID: PMC7217065 DOI: 10.1002/path.5357] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/03/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem or stromal cells (MSCs) are pluripotent cells implicated in a broad range of physiological events, including organogenesis and maintenance of tissue homeostasis as well as tissue regeneration and repair. Because their current definition is somewhat loose – based primarily on their ability to differentiate into a variety of mesenchymal tissues, adhere to plastic, and express, or lack, a handful of cell surface markers – MSCs likely encompass several subpopulations, which may have diverse properties. Their diversity may explain, at least in part, the pleiotropic functions that they display in different physiological and pathological settings. In the context of tissue injury, MSCs can respectively promote and attenuate inflammation during the early and late phases of tissue repair. They may thereby act as sensors of the inflammatory response and secrete mediators that boost or temper the response as required by the stage of the reparatory and regenerative process. MSCs are also implicated in regulating tumor development, in which they are increasingly recognized to play a complex role. Thus, MSCs can both promote and constrain tumor progression by directly affecting tumor cells via secreted mediators and cell–cell interactions and by modulating the innate and adaptive immune response. This review summarizes our current understanding of MSC involvement in tumor development and highlights the mechanistic underpinnings of their implication in tumor growth and progression. © 2020 Authors. Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Sabine Galland
- Laboratory of Experimental Pathology, Institute of Pathology, CHUV, Lausanne, Switzerland
| | - Ivan Stamenkovic
- Laboratory of Experimental Pathology, Institute of Pathology, CHUV, Lausanne, Switzerland
| |
Collapse
|
13
|
Heidari Barchi Nezhad R, Asadi F, Abtahi Froushani SM, Hassanshahi G, Kaeidi A, Khanamani Falahati-Pour S, Hashemi Z, Mirzaei MR. The effects of transplanted mesenchymal stem cells treated with 17-b estradiol on experimental autoimmune encephalomyelitis. Mol Biol Rep 2019; 46:6135-6146. [PMID: 31555971 DOI: 10.1007/s11033-019-05048-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 08/28/2019] [Indexed: 01/09/2023]
Abstract
The present study was conducted aimed at exploring the modulatory effects of 17-b estradiol (17-bED) on mesenchymal stem cells (MSCs) in the EAE (experimental autoimmune encephalomyelitis) animal model of multiple sclerosis (MS). Following the isolation of bone marrow-derived MSCs from the bilateral femurs and tibias of the male Wistar rats, the cells were harvested and cultured in the presence of 100 nM 17-bED for 24 h. EAE was induced in male Wistar rats (8-12 weeks old) using guinea pig spinal cord homogenate, in combination with the complete Freund's adjuvant. The MSC therapy was triggered when all of the animals obtained a disability score. The symptoms were monitored on a daily basis throughout the study until the rats were euthanized. The mRNA expression of cytokines, including IL-17, IFN-γ, TNF-α, IL-10, IL-4, and TGF-β together with MMP8 and MMP9 as the family members of matrix metalloproteinases (MMPs) in the brain and spinal cord tissues were examined using real-time PCR. The levels of splenocytes-originated IL-10 and IFN-γ cytokines were also measured by ELISA. The MTT-based research findings showed that the infiltration of lymphocytes into the spleen decreased considerably. It was also observed that the mRNA expression of proinflammatory cytokines decreased significantly, while the mRNA levels of anti-inflammatory cytokines increased remarkably. It was also found that the mRNA levels of the examined matrix metalloproteinases (MMP8 and MMP9) were downregulated significantly. The findings of the present study indicated that the administration of 17-bED enhanced the efficacy of MSCs transplantation and modulated immune responses relatively in the EAE model, via the regulation of either pro- or anti-inflammatory cytokines and matrix metalloproteinases.
Collapse
Affiliation(s)
- Rahim Heidari Barchi Nezhad
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, 7718175911, Iran
| | - Fateme Asadi
- Department of Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, 7718175911, Iran
| | - Ayat Kaeidi
- Physiology-Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Zahra Hashemi
- Department of General Subjects, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Reza Mirzaei
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, 7718175911, Iran.
| |
Collapse
|
14
|
Na Y, Nam JP, Hong J, Oh E, Shin HC, Kim HS, Kim SW, Yun CO. Systemic administration of human mesenchymal stromal cells infected with polymer-coated oncolytic adenovirus induces efficient pancreatic tumor homing and infiltration. J Control Release 2019; 305:75-88. [PMID: 31071373 DOI: 10.1016/j.jconrel.2019.04.040] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 04/22/2019] [Accepted: 04/28/2019] [Indexed: 12/19/2022]
Abstract
Oncolytic adenovirus (oAd)-mediated gene therapy is a promising approach for cancer treatment because of its cancer cell-restricted replication and therapeutic gene expression. However, systemic administration of oAd is severely restricted by their immunogenic nature and poor tumor homing ability, thus oAd cannot be utilized to treat disseminated metastases. In this study, human bone marrow-derived mesenchymal stromal cell (hMSCs) was used as a viral replication-permissive carrier for oAd with an aim to improve the systemic delivery of the virus to tumor tissues. To overcome the poor delivery of oAd into hMSCs, a relaxin (RLX)-expressing oncolytic Ad (oAd/RLX), which degrades dense tumor extracellular matrix of highly desmoplastic pancreatic cancer, was complexed with biodegradable polymer (poly (ethyleneimine)-conjugated poly(CBA-DAH); PCDP), generating oAd/RLX-PCDP complex. oAd/RLX-PCDP complex enhanced the internalization of oAd into hMSC, leading to superior viral production and release from hMSCs, along with high RLX expression. Furthermore, systemic administration of oAd/RLX-PCDP-treated hMSCs elicited more potent antitumor effect compared to naked oAd/RLX or oAd/RLX-treated hMSC in pancreatic tumor model. This potent antitumor effect of systemically administered oAd/RLX-PCDP-treated hMSCs was achieved by superior viral replication in tumor tissues than any other treatment group. In conclusion, these results demonstrate that hMSCs are effective carriers for the systemic delivery of oAd to tumor sites and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Youjin Na
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Joung-Pyo Nam
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - JinWoo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - Eonju Oh
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | | | | | - Sung Wan Kim
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Hernandez Y, González-Pastor R, Belmar-Lopez C, Mendoza G, de la Fuente JM, Martin-Duque P. Gold nanoparticle coatings as efficient adenovirus carriers to non-infectable stem cells. RSC Adv 2019; 9:1327-1334. [PMID: 35517997 PMCID: PMC9059632 DOI: 10.1039/c8ra09088b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/07/2019] [Accepted: 12/24/2018] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are adult pluripotent cells with the plasticity to be converted into different cell types. Their self-renewal capacity, relative ease of isolation, expansion and inherent migration to tumors, make them perfect candidates for cell therapy against cancer. However, MSCs are notoriously refractory to adenoviral infection, mainly because CAR (Coxsackie-Adenovirus Receptor) expression is absent or downregulated. Over the last years, nanoparticles have attracted a great deal of attention as potential vehicle candidates for gene delivery, but with limited effects on their own. Our data showed that the use of positively charged 14 nm gold nanoparticles either functionalized with arginine-glycine-aspartate (RGD) motif or not, increases the efficiency of adenovirus infection in comparison to commercial reagents without altering cell viability or cell phenotype. This system represents a simple, efficient and safe method for the transduction of MSCs, being attractive for cancer gene and cell therapies.
Collapse
Affiliation(s)
- Yulan Hernandez
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza 50018 Spain
| | - Rebeca González-Pastor
- Instituto Aragonés de Ciencias de la Salud 50009 Zaragoza Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón) 50009 Zaragoza Spain
| | - Carolina Belmar-Lopez
- Instituto Aragonés de Ciencias de la Salud 50009 Zaragoza Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón) 50009 Zaragoza Spain
| | - Gracia Mendoza
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza 50018 Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón) 50009 Zaragoza Spain
| | - Jesus M de la Fuente
- Instituto de Ciencias de Materiales (ICMA), CSIC 50009 Zaragoza Spain
- CIBER-BBN 28029 Madrid Spain
| | - Pilar Martin-Duque
- Instituto Aragonés de Ciencias de la Salud 50009 Zaragoza Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón) 50009 Zaragoza Spain
- Fundación Araid 50001 Zaragoza Spain
| |
Collapse
|
16
|
Alshareeda AT, Rakha E, Alghwainem A, Alrfaei B, Alsowayan B, Albugami A, Alsubayyil AM, Abomraee M, Mohd Zin NK. The effect of human placental chorionic villi derived mesenchymal stem cell on triple-negative breast cancer hallmarks. PLoS One 2018; 13:e0207593. [PMID: 30458011 PMCID: PMC6245746 DOI: 10.1371/journal.pone.0207593] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/03/2018] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can influence the tumour microenvironment (TEM) and play a major role in tumourigenesis. Triple-negative [Ostrogen receptor (ER-), Progesterone receptor (PgR-), and HER2/neu receptor (HER2-)] breast cancer (TNBC) is an aggressive class of BC characterized by poor prognosis and lacks the benefit of routinely available targeted therapies. This study aims to investigate the effect of human placental chorionic villi derived MSCs (CVMSCs) on the behavior of TNBC in vitro. This was done by assaying different cancer hallmarks including proliferation, migration and angiogenesis. Cell proliferation rate of TNBC cell line (MDA-MB231) was monitored in real time using the xCELLigence system. Whereas, Boyden chamber migration assay was used to measure MDA-MB231 motility and invasiveness toward CVMSCs. Finally, a three-dimensional (3D) model using a co-culture system of CVMSCs with MDA-MB231 with or without the addition of human umbilical vein endothelial cells (HUVECs) was created to assess tumour angiogenesis in vitro. CVMSCs were able to significantly reduce the proliferative and migratory capacity of MDA-MB231 cells. Co-culturing of MDA-MB231 with CVMSCs, not only inhibited the tube formation ability of HUVECs but also reduced the expression of the BC characteristic cytokines; IL-10, IL-12, CXCL9 and CXCL10 of CVMSCs. These results support the hypothesis that CVMSCs can influence the behavior of TNBC cells and provides a basic for a potential therapeutic approach in a pre-clinical settings. The data from this study also highlight the complexity of the in vitro cancer angiogenesis model settings and regulations.
Collapse
Affiliation(s)
- Alaa T Alshareeda
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Emad Rakha
- University of Nottingham and Nottingham University Hospitals NHS Trust, City Hospital, Department of Cellular Pathology, UK, Nottingham, United Kingdom
| | - Ayidah Alghwainem
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Bahauddeen Alrfaei
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Batla Alsowayan
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Abdullah Albugami
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Abdullah M Alsubayyil
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Mohmed Abomraee
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | | |
Collapse
|
17
|
Adenovirus platform enhances transduction efficiency of human mesenchymal stem cells: An opportunity for cellular carriers of targeted TRAIL-based TR3 biologics in ovarian cancer. PLoS One 2017; 12:e0190125. [PMID: 29267342 PMCID: PMC5739501 DOI: 10.1371/journal.pone.0190125] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 11/19/2017] [Indexed: 12/31/2022] Open
Abstract
Clinical application of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-based cancer therapeutics has not reached optimal potencies in part due to inadequate drug stability and inefficiencies in cancer-selective drug delivery. As such, innovative strategies regarding drug design and delivery are of utmost importance to achieve improved treatment results. With our current study, we aimed at exploring the groundwork for a two-stage targeting concept, which is based on the intrinsic tumor homing capacity of mesenchymal stem cells (MSCs) as cellular drug factories for the in situ production of our newly designed and biomarker-targeted TRAIL-based TR3 therapeutics. Since MSCs are primary cells, capable in vitro of only a limited number of cell divisions, identification of suitable strategies for their efficient genetic manipulation is of critical importance. We chose adenoviral (Ad) vectors as a transduction vehicle due to its ability to infect dividing and non-dividing cells and because of their limited restrictions regarding the packaging capacity of their genetic payload. In order to enhance the transduction efficacy of MSCs using Ad5 wild-type-based vectors, we tested a variety of fiber knob modifications on a panel of patient-derived MSC lines established from adipose tissue. We identified Ad5pK7, an Ad5 vector containing a polylysine fiber knob modification, exhibiting the highest transduction rates across a panel of 16 patient-derived MSC lines. We further demonstrated that MSCs could be efficiently transduced with an Ad5pK7 vector containing membrane-anchored and secreted TR3 expression units, including the MUC16 (CA125)-targeted variant Meso64-TR3. In both in vitro and in vivo experiments, MSC-derived Meso64-TR3 was far more potent on MUC16-expressing ovarian cancer compared to its non-targeted TR3 counterpart. Our findings thus provide the foundation to initiate further preclinical investigations on MSC-mediated treatment options in ovarian cancer using biomarker-targeted TR3-based biologics.
Collapse
|
18
|
Khan M, Adil SER, Olson AL. The role of mesenchymal stem cells in oncology and regenerative medicine. Future Oncol 2017; 13:821-831. [DOI: 10.2217/fon-2016-0264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have long been used as therapeutic agents in disease affecting various organ systems. However, MSCs are fast emerging as promising anticancer agents which have the potential to treat a number of different cancer types, including glioblastoma and metastatic breast, ovarian and hepatic carcinoma. The ability of MSC to migrate directly into the tumor microenvironment and to produce IFN-α and -β makes this possible. However, the possibility of MSC undergoing either malignant transformation or transformation into protumorigenic fibroblasts currently limits their role in clinical use. It is hoped that future research can overcome these limitations and facilitate the use of MSC clinically.
Collapse
Affiliation(s)
- Maliha Khan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Amanda L Olson
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
19
|
iPS-derived MSCs from an expandable bank to deliver a prodrug-converting enzyme that limits growth and metastases of human breast cancers. Cell Death Discov 2017; 3:16064. [PMID: 28179988 PMCID: PMC5292869 DOI: 10.1038/cddiscovery.2016.64] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 06/11/2016] [Indexed: 12/12/2022] Open
Abstract
One attractive strategy to treat cancers is to deliver an exogenous enzyme that will convert a non-toxic compound to a highly toxic derivative. The strategy was tested with viral vectors but was disappointing because the efficiency of transduction into tumor cells was too low. Recent reports demonstrated that the limitation can be addressed by using tissue-derived mesenchymal stromal cells (MSCs) to deliver enzyme/prodrug systems that kill adjacent cancer cells through bystander effects. Here we addressed the limitation that tissue-derived MSCs vary in their properties and are difficult to generate in the large numbers needed for clinical applications. We prepared a Feeder Stock of MSCs from induced pluripotent stem cells (iPSs) that provided an extensively expandable source of standardized cells. We then transduced the iPS-derived MSCs to express cytosine deaminase and injected them locally into a mouse xenogeneic model of human breast cancer. After administration of the prodrug (5-fluorocytosine), the transduced iPS-MSCs both limited growth of preformed tumors and decreased lung metastases.
Collapse
|
20
|
Torkaman M, Ghollasi M, Mohammadnia-Afrouzi M, Salimi A, Amari A. The effect of transplanted human Wharton's jelly mesenchymal stem cells treated with IFN-γ on experimental autoimmune encephalomyelitis mice. Cell Immunol 2016; 311:1-12. [PMID: 27697286 DOI: 10.1016/j.cellimm.2016.09.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/26/2016] [Accepted: 09/26/2016] [Indexed: 12/29/2022]
Abstract
Interferon gamma (IFN-γ) increases the immunosuppressive property of human Wharton's jelly mesenchymal stem cells (hWJ-MSCs). In this study, we evaluated the therapeutic effects of IFN-γ primed WJ-MSCs in EAE mice. IFN-γ primed WJ-MSCs were injected on days 3 and 11 after EAE induction. 21 days after EAE induction, splenocytes and cervical lymph node cells were isolated and cell proliferation, secretion of inflammatory cytokines and frequency of regulatory T-cells was measured. On day 50 of the study, cell infiltration and gene expression of inflammatory cytokines in brain of mice were studied. Leukocyte infiltration and symptoms were significantly reduced in IFN-γ primed WJ-MSCs treated group compared to other groups. These cells showed significantly reduced proliferation and increased Treg cells as well as decreased secretion and gene expression of inflammatory cytokines in EAE mice. Our data suggest that IFN-γ may be used to stimulate the immunomodulatory property of WJ-MSCs in clinical situations.
Collapse
Affiliation(s)
- Mohammad Torkaman
- Department of Pediatrics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Marzieh Ghollasi
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran.
| | | | - Ali Salimi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Afshin Amari
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
21
|
Su Y, Xie Z, Kim GB, Dong C, Yang J. Design strategies and applications of circulating cell-mediated drug delivery systems. ACS Biomater Sci Eng 2015; 1:201-217. [PMID: 25984572 PMCID: PMC4428174 DOI: 10.1021/ab500179h] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Drug delivery systems, particularly nanomaterial-based drug delivery systems, possess a tremendous amount of potential to improve diagnostic and therapeutic effects of drugs. Controlled drug delivery targeted to a specific disease is designed to significantly improve the pharmaceutical effects of drugs and reduce their side effects. Unfortunately, only a few targeted drug delivery systems can achieve high targeting efficiency after intravenous injection, even with the development of numerous surface markers and targeting modalities. Thus, alternative drug and nanomedicine targeting approaches are desired. Circulating cells, such as erythrocytes, leukocytes, and stem cells, present innate disease sensing and homing properties. Hence, using living cells as drug delivery carriers has gained increasing interest in recent years. This review highlights the recent advances in the design of cell-mediated drug delivery systems and targeting mechanisms. The approaches of drug encapsulation/conjugation to cell-carriers, cell-mediated targeting mechanisms, and the methods of controlled drug release are elaborated here. Cell-based "live" targeting and delivery could be used to facilitate a more specific, robust, and smart payload distribution for the next-generation drug delivery systems.
Collapse
Affiliation(s)
- Yixue Su
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Zhiwei Xie
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Gloria B. Kim
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Cheng Dong
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Jian Yang
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| |
Collapse
|
22
|
Hammer K, Kazcorowski A, Liu L, Behr M, Schemmer P, Herr I, Nettelbeck DM. Engineered adenoviruses combine enhanced oncolysis with improved virus production by mesenchymal stromal carrier cells. Int J Cancer 2015; 137:978-90. [PMID: 25604186 DOI: 10.1002/ijc.29442] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 12/03/2014] [Accepted: 12/17/2014] [Indexed: 12/12/2022]
Abstract
Oncolytic viruses have demonstrated in pre-clinical and clinical studies safety and a unique pleiotropic activity profile of tumor destruction. Yet, their delivery suffers from virus inactivation by blood components and sequestration to healthy tissues. Therefore, mesenchymal stromal cells (MSCs) have been applied as carrier cells for shielded virus delivery to tumors after ex vivo infection with oncolytic viruses. However, infection and particle production by MSCs have remained unsatisfying. Here, we report engineered oncolytic adenoviruses (OAds) for improved virus production and delivery by MSCs. OAds are uniquely amenable to molecular engineering, which has facilitated improved tumor cell destruction. But for MSC-mediated regimens, OAd engineering needs to achieve efficient infection and replication in both MSCs and tumor cells. We show that an Ad5/3 chimeric OAd capsid, containing the adenovirus serotype 3 cell-binding domain, strongly increases the entry into human bone marrow-derived MSCs and into established and primary pancreatic cancer cells. Further, we reveal that OAd with engineered post-entry functions-by deletion of the anti-apoptotic viral gene E1B19K or expression of the death ligand TRAIL--markedly increased virus titers released from MSCs, while MSC migration was not hampered. Finally, these virus modifications, or viral expression of FCU1 for local 5-FC prodrug activation, improved tumor cell killing implementing complementary cytotoxicity profiles in a panel of pancreatic cancer cell cultures. Together, our study establishes post-entry modification of OAd replication for improving virus delivery by carrier cells and suggests a panel of optimized OAds for future clinical development in personalized treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Katharina Hammer
- Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Adam Kazcorowski
- Molecular OncoSurgery, German Cancer Research Center (DKFZ) and Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Li Liu
- Molecular OncoSurgery, German Cancer Research Center (DKFZ) and Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Michael Behr
- Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Schemmer
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Ingrid Herr
- Molecular OncoSurgery, German Cancer Research Center (DKFZ) and Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Dirk M Nettelbeck
- Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
23
|
Abstract
Stem cell-based therapeutic strategies have emerged as very attractive treatment options over the past decade. Stem cells are now being utilized as delivery vehicles especially in cancer therapy to deliver a number of targeted proteins and viruses. This chapter aims to shed light on numerous studies that have successfully employed these strategies to target various cancer types with a special emphasis on numerous aspects that are critical to the success of future stem cell-based therapies for cancer.
Collapse
|
24
|
Amari A, Ebtekar M, Moazzeni SM, Soleimani M, Amirabad LM, Tahoori MT, Massumi M. Investigation of immunomodulatory properties of human Wharton's Jelly-derived mesenchymal stem cells after lentiviral transduction. Cell Immunol 2014; 293:59-66. [PMID: 25569483 DOI: 10.1016/j.cellimm.2014.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 11/26/2014] [Accepted: 12/12/2014] [Indexed: 12/29/2022]
Abstract
Human Wharton's Jelly-derived Mesenchymal Stem Cells (hWJ-MSCs) are considered as an alternative for bone-marrow-derived MSCs. These cells have immunosuppressive properties. It was unclear whether the WJ-MSCs would sustain their immunomodulatory characteristics after lentiviral transduction or not. In this study, we evaluated immunomodulatory properties of WJ-MSCs after lentiviral transduction. HWJ-MSCs were transduced with lentiviral particles. Expression of transduced and un-transduced hWJ-MSCs surface molecules and secretion of IL-10, HGF, VEGF and TGF-β was analyzed. Cell proliferation and frequency of CD4(+)CD25(+) CD127(low/neg) Foxp3(+) T regulatory cells was measured. There was no difference between the surface markers and secretion of IL-10, HGF, VEGF and TGF-β in transduced and un-transduced hWJ-MSCs. Both cells inhibited the proliferation of PHA stimulated PBMCs, and improved the frequency of T regulatory cells. These findings suggest that lentiviral transduction does not alter the immunomodulatory function of hWJ-MSCs. However, lentiviral transduction may have a wide range of applications in gene therapy.
Collapse
Affiliation(s)
- Afshin Amari
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Massoumeh Ebtekar
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Seyed Mohammad Moazzeni
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoud Soleimani
- Hematology Department, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran; Stem Cells Biology Department, Stem Cell Technology Research Center, Tehran, Iran
| | - Leila Mohammadi Amirabad
- Medical Genetics Department, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mohammad Taher Tahoori
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Massumi
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran; Stem Cells Biology Department, Stem Cell Technology Research Center, Tehran, Iran.
| |
Collapse
|
25
|
Duebgen M, Martinez-Quintanilla J, Tamura K, Hingtgen S, Redjal N, Wakimoto H, Shah K. Stem cells loaded with multimechanistic oncolytic herpes simplex virus variants for brain tumor therapy. J Natl Cancer Inst 2014; 106:dju090. [PMID: 24838834 DOI: 10.1093/jnci/dju090] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The current treatment regimen for malignant glioblastoma multiforme (GBM) is tumor resection followed by chemotherapy and radiation therapy. Despite the proven safety of oncolytic herpes simplex virus (oHSV) in clinical trials for GBMs, its efficacy is suboptimal mainly because of insufficient viral spread after tumor resection. METHODS Human mesenchymal stem cells (MSC) were loaded with oHSV (MSC-oHSV), and their fate was explored by real-time imaging in vitro and in vivo. Using novel diagnostic and armed oHSV mutants and real-time multimodality imaging, the efficacy of MSC-oHSV and its proapoptotic variant, oHSV-TRAIL encapsulated in biocompatible synthetic extracellular matrix (sECM), was tested in different mouse GBM models, which more accurately reflect the current clinical settings of malignant, resistant, and resected tumors. All statistical tests were two-sided. RESULTS MSC-oHSVs effectively produce oHSV progeny, which results in killing of GBMs in vitro and in vivo mediated by a dynamic process of oHSV infection and tumor destruction. sECM-encapsulated MSC-oHSVs result in statistically significant increased anti-GBM efficacy compared with direct injection of purified oHSV in a preclinical model of GBM resection, resulting in prolonged median survival in mice (P < .001 with Gehan-Breslow-Wilcoxin test). To supersede resistant tumors, MSC loaded with oHSV-TRAIL effectively induce apoptosis-mediated killing and prolonged median survival in mice bearing oHSV- and TRAIL-resistant GBM in vitro (P < .001 with χ(2) contingency test). CONCLUSIONS Human MSC loaded with different oHSV variants provide a platform to translate oncolytic virus therapies to clinics in a broad spectrum of GBMs after resection and could also have direct implications in different cancer types.
Collapse
Affiliation(s)
- Matthias Duebgen
- Affiliations of authors: Molecular Neurotherapy and Imaging Laboratory (MD, JM-Q, SH, NR, HW, KS), Department of Radiology (MD, JM-Q, KT, SH, NR, HW, KS), Department of Neurosurgery (NR, HW), and Department of Neurology (KS), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA (KS)
| | - Jordi Martinez-Quintanilla
- Affiliations of authors: Molecular Neurotherapy and Imaging Laboratory (MD, JM-Q, SH, NR, HW, KS), Department of Radiology (MD, JM-Q, KT, SH, NR, HW, KS), Department of Neurosurgery (NR, HW), and Department of Neurology (KS), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA (KS)
| | - Kaoru Tamura
- Affiliations of authors: Molecular Neurotherapy and Imaging Laboratory (MD, JM-Q, SH, NR, HW, KS), Department of Radiology (MD, JM-Q, KT, SH, NR, HW, KS), Department of Neurosurgery (NR, HW), and Department of Neurology (KS), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA (KS)
| | - Shawn Hingtgen
- Affiliations of authors: Molecular Neurotherapy and Imaging Laboratory (MD, JM-Q, SH, NR, HW, KS), Department of Radiology (MD, JM-Q, KT, SH, NR, HW, KS), Department of Neurosurgery (NR, HW), and Department of Neurology (KS), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA (KS)
| | - Navid Redjal
- Affiliations of authors: Molecular Neurotherapy and Imaging Laboratory (MD, JM-Q, SH, NR, HW, KS), Department of Radiology (MD, JM-Q, KT, SH, NR, HW, KS), Department of Neurosurgery (NR, HW), and Department of Neurology (KS), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA (KS)
| | - Hiroaki Wakimoto
- Affiliations of authors: Molecular Neurotherapy and Imaging Laboratory (MD, JM-Q, SH, NR, HW, KS), Department of Radiology (MD, JM-Q, KT, SH, NR, HW, KS), Department of Neurosurgery (NR, HW), and Department of Neurology (KS), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA (KS)
| | - Khalid Shah
- Affiliations of authors: Molecular Neurotherapy and Imaging Laboratory (MD, JM-Q, SH, NR, HW, KS), Department of Radiology (MD, JM-Q, KT, SH, NR, HW, KS), Department of Neurosurgery (NR, HW), and Department of Neurology (KS), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA (KS).
| |
Collapse
|
26
|
Sun YP, Zhang BL, Duan JW, Wu HH, Wang BQ, Yu ZP, Yang WJ, Shan YF, Zhou MT, Zhang QY. Effect of NK4 transduction in bone marrow-derived mesenchymal stem cells on biological characteristics of pancreatic cancer cells. Int J Mol Sci 2014; 15:3729-45. [PMID: 24595237 PMCID: PMC3975364 DOI: 10.3390/ijms15033729] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 01/20/2014] [Accepted: 02/10/2014] [Indexed: 12/25/2022] Open
Abstract
Pancreatic cancer usually has a poor prognosis, and no gene therapy has yet been developed that is effective to treat it. Since a unique characteristic of bone marrow-derived mesenchymal stem cells (MSCs) is that they migrate to tumor tissues, we wanted to determine whether MSCs could serve as a vehicle of gene therapy for targeting pancreatic cancer. First, we successfully extracted MSCs from SD rats. Next, MSCs were efficiently transduced with NK4, an antagonist of hepatocyte growth factor (HGF) which comprising the N-terminal and the subsequent four kringle domains of HGF, by an adenoviral vector. Then, we confirmed that rat MSCs preferentially migrate to pancreatic cancer cells. Last, MSCs expressing NK4 (NK4-MSCs) strongly inhibited proliferation and migration of the pancreatic cancer cell line SW1990 after co-culture. These results indicate that MSCs can serve as a vehicle of gene therapy for targeting pancreatic cancer.
Collapse
Affiliation(s)
- Yun-Peng Sun
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| | - Ben-Long Zhang
- Department of General Surgery, Yiwu Chouzhou Hospital, Yiwu 322000, Zhejiang, China.
| | - Jian-Wen Duan
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| | - Huan-Huan Wu
- Department of Infectious Disease, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| | - Ben-Quan Wang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| | - Zheng-Ping Yu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| | - Wen-Jun Yang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| | - Yun-Feng Shan
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| | - Meng-Tao Zhou
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| | - Qi-Yu Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| |
Collapse
|
27
|
Lee HJ, Doo SW, Kim DH, Cha YJ, Kim JH, Song YS, Kim SU. Cytosine deaminase-expressing human neural stem cells inhibit tumor growth in prostate cancer-bearing mice. Cancer Lett 2013; 335:58-65. [DOI: 10.1016/j.canlet.2013.01.048] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Revised: 01/29/2013] [Accepted: 01/29/2013] [Indexed: 01/14/2023]
|
28
|
Belmar-Lopez C, Mendoza G, Oberg D, Burnet J, Simon C, Cervello I, Iglesias M, Ramirez JC, Lopez-Larrubia P, Quintanilla M, Martin-Duque P. Tissue-derived mesenchymal stromal cells used as vehicles for anti-tumor therapy exert different in vivo effects on migration capacity and tumor growth. BMC Med 2013; 11:139. [PMID: 23710709 PMCID: PMC3670996 DOI: 10.1186/1741-7015-11-139] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 04/19/2013] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have been promoted as an attractive option to use as cellular delivery vehicles to carry anti-tumor agents, owing to their ability to home into tumor sites and secrete cytokines. Multiple isolated populations have been described as MSCs, but despite extensive in vitro characterization, little is known about their in vivo behavior.The aim of this study was to investigate the efficacy and efficiency of different MSC lineages derived from five different sources (bone marrow, adipose tissue, epithelial endometrium, stroma endometrium, and amniotic membrane), in order to assess their adequacy for cell-based anti-tumor therapies. Our study shows the crucial importance of understanding the interaction between MSCs and tumor cells, and provides both information and a methodological approach, which could be used to develop safer and more accurate targeted therapeutic applications. METHODS We first measured the in vivo migration capacity and effect on tumor growth of the different MSCs using two imaging techniques: (i) single-photon emission computed tomography combined with computed tomography (SPECT-CT), using the human sodium iodine symporter gene (hNIS) and (ii) magnetic resonance imaging using superparamagnetic iron oxide. We then sought correlations between these parameters and expression of pluripotency-related or migration-related genes. RESULTS Our results show that migration of human bone marrow-derived MSCs was significantly reduced and slower than that obtained with the other MSCs assayed and also with human induced pluripotent stem cells (hiPSCs). The qPCR data clearly show that MSCs and hiPSCs exert a very different pluripotency pattern, which correlates with the differences observed in their engraftment capacity and with their effects on tumor growth. CONCLUSION This study reveals differences in MSC recruitment/migration toward the tumor site and the corresponding effects on tumor growth. Three observations stand out: 1) tracking of the stem cell is essential to check the safety and efficacy of cell therapies; 2) the MSC lineage to be used in the cell therapy needs to be carefully chosen to balance efficacy and safety for a particular tumor type; and 3) different pluripotency and mobility patterns can be linked to the engraftment capacity of the MSCs, and should be checked as part of the clinical characterization of the lineage.
Collapse
|
29
|
Gene therapy of gastric cancer using LIGHT-secreting human umbilical cord blood-derived mesenchymal stem cells. Gastric Cancer 2013; 16:155-66. [PMID: 22850801 DOI: 10.1007/s10120-012-0166-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 05/11/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have the ability to migrate into tumors and therefore are potential vehicles for the therapy of malignant diseases. In this study, we investigated the use of umbilical cord blood mesenchymal stem cells (UCB-MSCs) as carriers for a constant source of transgenic LIGHT (TNFSF14) to target tumor cells in vivo. METHODS Lentiviral vectors carrying LIGHT genes were constructed, producing viral particles with a titer of 2 × 10(8) TU/L. Fourteen days after UCB-MSCs transfected by LIGHT gene packaged lentivirus had been injected into mouse gastric cancer models, the expression levels of LIGHT mRNA and protein were detected by reverse transcription polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). Then the tumors' approximate volumes were measured. RESULTS The treatment with MSC-LIGHT demonstrated a strong suppressive effect on tumor growth compared to treatment with MSC and NaCl (p < 0.001). Examination of pathological sections of the tumor tissues showed that the areas of tumor necrocis in the MSC-LIGHT group were larger than those in the MSC group. Moreover, we found that MSCs with LIGHT were able to significantly induce apoptosis of tumor cells. The expression levels of LIGHT mRNA and protein were significantly higher in the UCB-MSCs with the LIGHT gene than the levels in UCB-MSCs (p < 0.001). CONCLUSION These results suggest that UCB-MSCs carrying the LIGHT gene have the potential to be used as effective delivery vehicles in the treatment of gastric cancers.
Collapse
|
30
|
Collet G, Grillon C, Nadim M, Kieda C. Trojan horse at cellular level for tumor gene therapies. Gene 2013; 525:208-16. [PMID: 23542073 DOI: 10.1016/j.gene.2013.03.057] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 02/27/2013] [Accepted: 03/07/2013] [Indexed: 01/14/2023]
Abstract
Among innovative strategies developed for cancer treatments, gene therapies stand of great interest despite their well-known limitations in targeting, delivery, toxicity or stability. The success of any given gene-therapy is highly dependent on the carrier efficiency. New approaches are often revisiting the mythic trojan horse concept to carry therapeutic nucleic acid, i.e. DNAs, RNAs or small interfering RNAs, to pathologic tumor site. Recent investigations are focusing on engineering carrying modalities to overtake the above limitations bringing new promise to cancer patients. This review describes recent advances and perspectives for gene therapies devoted to tumor treatment, taking advantage of available knowledge in biotechnology and medicine.
Collapse
Affiliation(s)
- Guillaume Collet
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071, Orléans, cedex 2, France.
| | | | | | | |
Collapse
|
31
|
Antitumor activities of human placenta-derived mesenchymal stem cells expressing endostatin on ovarian cancer. PLoS One 2012; 7:e39119. [PMID: 22911684 PMCID: PMC3404061 DOI: 10.1371/journal.pone.0039119] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Accepted: 05/16/2012] [Indexed: 01/14/2023] Open
Abstract
Endostatin is an important endogenous inhibitor of neovascularization that has been widely used in anti-angiogenesis therapy for the treatment of cancer. However, its clinical application is largely hampered by its low efficacy. Human placenta-derived mesenchymal stem cells (hpMSCs) are particularly attractive cells for clinical use in cell-based therapies. In the present study, hpMSCs were isolated and characterized. We then evaluated the tumor targeting properties and antitumor effects of hpMSCs as gene delivery vehicles for ovarian cancer therapy. We efficiently engineered hpMSCs to deliver endostatin via adenoviral transduction mediated by Lipofectamine 2000. The tropism capacity of the engineered hpMSCs toward tumor cells was then confirmed by in vitro migration assays and in vivo by intraperitoneal injection of hpMSCs into nude mice. The hpMSCs expressing the human endostatin gene demonstrated preferential homing to the tumor site and significantly decreased the tumor volume without apparent systemic toxic effects. These observations were associated with significantly decreased blood sprouts and tumor cell proliferation as well as a dramatically increased tumor apoptosis index. These results suggested that hpMSCs are potentially an effective delivery vehicle for therapeutic genes for the treatment of ovarian cancer.
Collapse
|
32
|
Shah K. Mesenchymal stem cells engineered for cancer therapy. Adv Drug Deliv Rev 2012; 64:739-48. [PMID: 21740940 DOI: 10.1016/j.addr.2011.06.010] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 06/20/2011] [Accepted: 06/22/2011] [Indexed: 12/22/2022]
Abstract
Recent pre-clinical and clinical studies have shown that stem cell-based therapies hold tremendous promise for the treatment of human disease. Mesenchymal stem cells (MSC) are emerging as promising anti-cancer agents which have an enormous potential to be utilized to treat a number of different cancer types. MSC have inherent tumor-trophic migratory properties, which allows them to serve as vehicles for delivering effective, targeted therapy to isolated tumors and metastatic disease. MSC have been readily engineered to express anti-proliferative, pro-apoptotic, anti-angiogenic agents that specifically target different cancer types. Many of these strategies have been validated in a wide range of studies evaluating treatment feasibility or efficacy, as well as establishing methods for real-time monitoring of stem cell migration in vivo for optimal therapy surveillance and accelerated development. This review aims to provide an in depth status of current MSC-based cancer therapies, as well as the prospects for their clinical translation.
Collapse
|
33
|
Mao W, Zhu X, Tang D, Zhao Y, Zhao B, Ma G, Zhang X, An G, Li Y. TNF-α expression in the UCB-MSCs as stable source inhibits gastric cancers growth in nude mice. Cancer Invest 2012; 30:463-72. [PMID: 22536934 DOI: 10.3109/07357907.2012.675385] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mesenchymal stem cells (MSCs) are potentially vehicles for therapy of malignant diseases. In our study, we investigated whether UCB-MSCs are capable to carry TNF-α to target tumor cells in vivo. The human gastric cancer cells SGC-7901 were subcutaneously injected into the abdomen near groins of 15 nude mice to establish experiment tumor models. MSC-TNF-α demonstrated a strong suppressive effect on the tumor growth compared with MSC and NaCl. Thus, MSC-TNF-α can obviously inhibit Gastric cancers growth in nude mice, indicating that UCB-MSCs may have the potential to become a prevention approach against gastric cancer.
Collapse
Affiliation(s)
- Weizheng Mao
- Department of General Surgery, Qingdao Municipal Hospital, Qingdao, China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Zhang X, Xu W, Qian H, Zhu W, Zhang R. Mesenchymal stem cells modified to express lentivirus TNF-α Tumstatin(45-132) inhibit the growth of prostate cancer. J Cell Mol Med 2012; 15:433-44. [PMID: 19799647 PMCID: PMC3822807 DOI: 10.1111/j.1582-4934.2009.00920.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a potential novel delivery system for cell-based gene therapies. Although tumour necrosis factor (TNF)-α has been shown to have antitumour activity, its use in therapy is limited by its systemic toxicity. For the present study, we designed lentivirus-mediated signal peptide TNF-α-Tumstatin(45-132) -expressing mesenchymal stem cells (SPTT-MSCs) as a novel anti-cancer approach. We evaluated the effects of this approach on human prostate cancer cells (PC3 and LNCaP) by co-culturing them with either SPTT-MSCs or supernatants from their culture medium in vitro. The antitumour effects and possible mechanisms of action of SPTT-MSCs were then determined in PC3 cells in vivo. The results showed that efficient TNF-α-Tumstatin(45-132) -expressing MSCs had been established, and demonstrated that SPTT-MSCs inhibited the proliferation of and induced apoptosis in prostate cancer cells and xenograft tumours. As would be expected, given the properties of the individual proteins, the TNF-α-Tumstatin(45-132) fusion exerted potent cytotoxic effects on human prostate cancer cells and tumours via the death receptor-dependent apoptotic pathway and via antiangiogenic effects. Our findings suggest that SPTT-MSCs have significant activity against prostate cancer cells, and that they may represent a promising new therapy for prostate cancer.
Collapse
Affiliation(s)
- Xu Zhang
- School of Medical Science and Laboratory Medicine, Jiangsu University, Jiangsu, China
| | | | | | | | | |
Collapse
|
35
|
Duffy MR, Parker AL, Bradshaw AC, Baker AH. Manipulation of adenovirus interactions with host factors for gene therapy applications. Nanomedicine (Lond) 2012; 7:271-88. [DOI: 10.2217/nnm.11.186] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nanomedicine based on the use of adenovirus vectors for therapeutic gene delivery shows broad potential. Specific targeting for many gene therapy applications, such as metastatic cancers or cardiovascular diseases requires intravascular delivery of the vector. However, a major barrier to successful adenovirus vector targeting follows systemic delivery, as upon contact with the bloodstream the virus interacts with a variety of host proteins, in particular coagulation factor X, which mediates profound liver gene transfer. This inherent hepatic tropism combined with macrophage scavenging minimizes the efficacy of the virus at the desired sites and induces toxic side effects. Understanding the complex, multifaceted interactions of adenovirus with host factors is of vital importance to the design of safer vectors with improved efficacy and pharmacokinetic profiles. Increased knowledge of adenovirus biology provides the opportunity to develop innovative strategies to detarget the virus from the liver following intravascular delivery and redirect the vector to disease areas.
Collapse
Affiliation(s)
- Margaret R Duffy
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Alan L Parker
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Angela C Bradshaw
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Andrew H Baker
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
36
|
Iguchi K, Sakurai F, Tomita K, Katayama K, Yamaguchi T, Kawabata K, Tagawa M, Kawabata M, Shirakawa T, Mizuguchi H. Efficient antitumor effects of carrier cells loaded with a fiber-substituted conditionally replicating adenovirus on CAR-negative tumor cells. Cancer Gene Ther 2011; 19:118-25. [PMID: 22076042 DOI: 10.1038/cgt.2011.74] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Carrier cells delivering a conditionally replicating adenovirus (CRAd), which selectively replicates in tumor cells and induces tumor cell lysis, have promising potential for treatment of cancer because CRAd-loaded carrier cells evade inhibition by neutralizing anti-adenovirus (Ad) antibodies and because the carrier cells are locally retained at the injection point after local injection. A previous study by Hamada et al. demonstrated that carrier cells (CRAd-containing cell fragments derived from the carrier cells) are engulfed into the target cells, probably through a pathway independent of the primary receptor for Ad, the coxsackievirus and Ad receptor (CAR) (Mol Ther, 15: 1121-1128; 2007); however, it remains to be elucidated whether carrier cells infected with a conventional CRAd, which is composed of subgroup-C Ad serotype-5 (Ad5), mediate antitumor effects on CAR-negative cells. In order to examine whether carrier cells delivering a conventional CRAd (Carrier-F5) induce lysis of CAR-negative tumor cells, CAR-positive and CAR-negative tumor cells were incubated with Carrier-F5. Carrier-F5 mediated efficient killing of CAR-positive tumor cells; however, CAR-negative tumor cells were almost refractory to Carrier-F5. On the other hand, carrier cells loaded with a fiber-substituted CRAd containing fiber proteins of Ad serotype-35 (Ad35) (CRAd-F35), which binds to human CD46 for infection, showed efficient killing of both CAR-positive and CAR-negative tumor cells. Intra-tumoral injection of carrier cells loaded with CRAd-F35 (Carrier-F35) also resulted in efficient regression of both CAR-positive and CAR-negative tumors. These results demonstrated that the expression levels of receptors for Ad are an important factor for CRAd-loaded carrier cell-mediated cancer therapy, and that Carrier-F35 would have potential as a cancer treatment for not only CAR-positive tumors but also CAR-negative tumors.
Collapse
Affiliation(s)
- K Iguchi
- Laboratory of Gene Transfer and Regulation, National Institute of Biomedical Innovation, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Jiang J, Fan CY, Zeng BF. Experimental construction of BMP2 and VEGF gene modified tissue engineering bone in vitro. Int J Mol Sci 2011; 12:1744-55. [PMID: 21673920 PMCID: PMC3111631 DOI: 10.3390/ijms12031744] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 02/22/2011] [Accepted: 03/01/2011] [Indexed: 01/30/2023] Open
Abstract
The purpose of this study was to investigate the feasibility and advantages of constructing a novel tissue engineering bone, using β-tricalcium phosphate (β-TCP) and rat bone marrow mesenchymal stem cells (MSCs), modified with human bone morphogenetic protein 2 gene (hBMP2) and human vascular endothelial growth factor 165 gene (hVEGF165), through lentiviral transfection. Both genes were successfully co-expressed in the co-transfection group for up to eight weeks confirmed by enzyme-linked immunosorbent assay (ELISA). After seeding MSCs onto the scaffolds, scanning electron microscopy (SEM) observation showed that MSCs grew and proliferated well in co-transfection group at 7 and 14 days. There was no significant difference among all the groups in hoechst DNA assay for cell proliferation for 14 days after cell seeding (P > 0.05), but the highest alkaline phosphatase (ALP) activity was observed in the co-transfection group at 14 days after cell seeding (p < 0.01). These results demonstrated that it was advantageous to construct tissue engineering bone using β-TCP combined with MSCs lentivirally co-transfected with BMP2 and VEGF165, providing an innovative way for treating bone defects.
Collapse
Affiliation(s)
- Jia Jiang
- Department of Sports Medicine and Arthroscopic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mail:
| | - Cun-Yi Fan
- Department of Orthopedics, The Sixth Affiliated People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, China; E-Mail:
| | - Bing-Fang Zeng
- Department of Orthopedics, The Sixth Affiliated People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, China; E-Mail:
| |
Collapse
|
38
|
Tang C, Russell PJ, Martiniello-Wilks R, Rasko JEJ, Khatri A. Concise review: Nanoparticles and cellular carriers-allies in cancer imaging and cellular gene therapy? Stem Cells 2010; 28:1686-702. [PMID: 20629172 PMCID: PMC2996089 DOI: 10.1002/stem.473] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ineffective treatment and poor patient management continue to plague the arena of clinical oncology. The crucial issues include inadequate treatment efficacy due to ineffective targeting of cancer deposits, systemic toxicities, suboptimal cancer detection and disease monitoring. This has led to the quest for clinically relevant, innovative multifaceted solutions such as development of targeted and traceable therapies. Mesenchymal stem cells (MSCs) have the intrinsic ability to "home" to growing tumors and are hypoimmunogenic. Therefore, these can be used as (a) "Trojan Horses" to deliver gene therapy directly into the tumors and (b) carriers of nanoparticles to allow cell tracking and simultaneous cancer detection. The camouflage of MSC carriers can potentially tackle the issues of safety, vector, and/or transgene immunogenicity as well as nanoparticle clearance and toxicity. The versatility of the nanotechnology platform could allow cellular tracking using single or multimodal imaging modalities. Toward that end, noninvasive magnetic resonance imaging (MRI) is fast becoming a clinical favorite, though there is scope for improvement in its accuracy and sensitivity. In that, use of superparamagnetic iron-oxide nanoparticles (SPION) as MRI contrast enhancers may be the best option for tracking therapeutic MSC. The prospects and consequences of synergistic approaches using MSC carriers, gene therapy, and SPION in developing cancer diagnostics and therapeutics are discussed.
Collapse
Affiliation(s)
- Catherine Tang
- Oncology Research Centre, Prince of Wales Hospital, Randwick, Sydney, NSW, Australia
| | | | | | | | | |
Collapse
|
39
|
Dwyer RM, Khan S, Barry FP, O'Brien T, Kerin MJ. Advances in mesenchymal stem cell-mediated gene therapy for cancer. Stem Cell Res Ther 2010; 1:25. [PMID: 20699014 PMCID: PMC2941117 DOI: 10.1186/scrt25] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells have a natural tropism for tumours and their metastases, and are also considered immunoprivileged. This remarkable combination of properties has formed the basis for many studies investigating their potential as tumour-specific delivery vehicles for suicide genes, oncolytic viruses and secreted therapeutic proteins. The aim of the present review is to discuss the range of approaches that have been used to exploit the tumour-homing capacity of mesenchymal stem cells for gene delivery, and to highlight advances required to realize the full potential of this promising approach.
Collapse
Affiliation(s)
- Roisin M Dwyer
- Division of Surgery, School of Medicine, Clinical Science Institute, National University of Ireland Galway, Galway, Ireland.
| | | | | | | | | |
Collapse
|
40
|
Pulukuri SMK, Gorantla B, Dasari VR, Gondi CS, Rao JS. Epigenetic upregulation of urokinase plasminogen activator promotes the tropism of mesenchymal stem cells for tumor cells. Mol Cancer Res 2010; 8:1074-83. [PMID: 20663859 DOI: 10.1158/1541-7786.mcr-09-0495] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A major obstacle for the effective treatment of cancer is the invasive capacity of the tumor cells. Previous studies have shown the capability of mesenchymal stem cells (MSC) to target these disseminated tumor cells and to serve as therapeutic delivery vehicles. However, the molecular mechanisms that would enhance the migration of MSCs toward tumor areas are not well understood. In particular, very little is known about the role that epigenetic mechanisms play in cell migration and tropism of MSCs. In this study, we investigated whether histone deacetylation was involved in the repression of urokinase plasminogen activator (uPA) expression in MSCs derived from umbilical cord blood (CB) and bone marrow (BM). Induction of uPA expression by histone deacetylase inhibitors trichostatin A and sodium butyrate was observed in CB- and BM-derived MSCs examined. In vitro migration assays showed that induction of uPA expression by histone deacetylase inhibitors in CB- and BM-derived MSCs significantly enhanced tumor tropism of these cells. Furthermore, overexpression of uPA in CB-MSCs induced migration capacity toward human cancer cells in vitro. In addition, our results showed that uPA-uPAR knockdown in PC3 prostate cancer cells significantly inhibited tumor-specific migration of uPA-overexpressing MSCs. These results have significant implications for the development of MSC-mediated, tumor-selective gene therapies.
Collapse
Affiliation(s)
- Sai Murali Krishna Pulukuri
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | | | | | | | | |
Collapse
|
41
|
Grisendi G, Annerén C, Cafarelli L, Sternieri R, Veronesi E, Cervo GL, Luminari S, Maur M, Frassoldati A, Palazzi G, Otsuru S, Bambi F, Paolucci P, Pierfranco C, Horwitz E, Dominici M. GMP-manufactured density gradient media for optimized mesenchymal stromal/stem cell isolation and expansion. Cytotherapy 2010; 12:466-77. [DOI: 10.3109/14653241003649510] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
42
|
Bak XY, Yang J, Wang S. Baculovirus-transduced bone marrow mesenchymal stem cells for systemic cancer therapy. Cancer Gene Ther 2010; 17:721-9. [DOI: 10.1038/cgt.2010.32] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
43
|
Ghaedi M, Lotfi AS, Soleimani M. Establishment of lentiviral-vector-mediated model of human alpha-1 antitrypsin delivery into hepatocyte-like cells differentiated from mesenchymal stem cells. Tissue Cell 2010; 42:181-9. [DOI: 10.1016/j.tice.2010.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2010] [Revised: 03/25/2010] [Accepted: 03/27/2010] [Indexed: 02/03/2023]
|
44
|
Komarova S, Roth J, Alvarez R, Curiel DT, Pereboeva L. Targeting of mesenchymal stem cells to ovarian tumors via an artificial receptor. J Ovarian Res 2010; 3:12. [PMID: 20500878 PMCID: PMC2883983 DOI: 10.1186/1757-2215-3-12] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Accepted: 05/25/2010] [Indexed: 02/07/2023] Open
Abstract
Background Mesenchymal Progenitor/Stem Cells (MSC) respond to homing cues providing an important mechanism to deliver therapeutics to sites of injury and tumors. This property has been confirmed by many investigators, however, the efficiency of tumor homing needs to be improved for effective therapeutic delivery. We investigated the feasibility of enhancing MSC tumor targeting by expressing an artificial tumor-binding receptor on the MSC surface. Methods Human MSC expressing an artificial receptor that binds to erbB2, a tumor cell marker, were obtained by transduction with genetically modified adenoviral vectors encoding an artificial receptor (MSC-AR). MSC-AR properties were tested in vitro in cell binding assays and in vivo using two model systems: transient transgenic mice that express human erbB2 in the lungs and ovarian xenograft tumor model. The levels of luciferase-labeled MSCs in erbB2-expressing targeted sites were evaluated by measuring luciferase activity using luciferase assay and imaging. Results The expression of AR enhanced binding of MSC-AR to erbB2-expressing cells in vitro, compared to unmodified MSCs. Furthermore, we have tested the properties of erbB2-targeted MSCs in vivo and demonstrated an increased retention of MSC-AR in lungs expressing erbB2. We have also confirmed increased numbers of erbB2-targeted MSCs in ovarian tumors, compared to unmodified MSC. The kinetic of tumor targeting by ip injected MSC was also investigated. Conclusion These data demonstrate that targeting abilities of MSCs can be enhanced via introduction of artificial receptors. The application of this strategy for tumor cell-based delivery could increase a number of cell carriers in tumors and enhance efficacy of cell-based therapy.
Collapse
Affiliation(s)
- Svetlana Komarova
- Division of Human Gene Therapy, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294- 2172, USA.
| | | | | | | | | |
Collapse
|
45
|
Grisendi G, Bussolari R, Cafarelli L, Petak I, Rasini V, Veronesi E, De Santis G, Spano C, Tagliazzucchi M, Barti-Juhasz H, Scarabelli L, Bambi F, Frassoldati A, Rossi G, Casali C, Morandi U, Horwitz EM, Paolucci P, Conte P, Dominici M. Adipose-derived mesenchymal stem cells as stable source of tumor necrosis factor-related apoptosis-inducing ligand delivery for cancer therapy. Cancer Res 2010; 70:3718-29. [PMID: 20388793 DOI: 10.1158/0008-5472.can-09-1865] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Adipose-derived mesenchymal stromal/stem cells (AD-MSC) may offer efficient tools for cell-based gene therapy approaches. In this study, we evaluated whether AD-MSC could deliver proapoptotic molecules for cancer treatment. Human AD-MSCs were isolated and transduced with a retroviral vector encoding full-length human tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a proapoptotic ligand that induces apoptosis in a variety of human cancers but not normal tissues. Although several studies have documented the antitumor activity of recombinant human TRAIL, its use in vivo is limited by a short half-life in plasma due to a rapid clearance by the kidney. We found that these limitations can be overcome using stably transduced AD-MSC, which could serve as a constant source of TRAIL production. AD-MSC armed with TRAIL targeted a variety of tumor cell lines in vitro, including human cervical carcinoma, pancreatic cancer, colon cancer, and, in combination with bortezomib, TRAIL-resistant breast cancer cells. Killing activity was associated with activation of caspase-8 as expected. When injected i.v. or s.c. into mice, AD-MSC armed with TRAIL localized into tumors and mediated apoptosis without significant apparent toxicities to normal tissues. Collectively, our results provide preclinical support for a model of TRAIL-based cancer therapy relying on the use of adipose-derived mesenchymal progenitors as cellular vectors.
Collapse
Affiliation(s)
- Giulia Grisendi
- Department of Oncology, Hematology and Respiratory Diseases, Plastic Surgery Unit, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mesenchymal stem cells as therapeutic tools and gene carriers in liver fibrosis and hepatocellular carcinoma. Gene Ther 2010; 17:692-708. [PMID: 20220785 DOI: 10.1038/gt.2010.10] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem (stromal) cells (MSCs) are a source of circulating progenitors that are able to generate cells of all mesenchymal lineages and to cover cellular demands of injured tissues. The extent of their transdifferentiation plasticity remains controversial. Cells with MSC properties have been obtained from diverse tissues after purification and expansion in vitro. These cellular populations are heterogeneous and under certain conditions show pluripotent-like properties. MSCs present immunosuppressive and anti-inflammatory features and high migratory capacity toward inflamed or remodeling tissues. In this study we review available data regarding factors and signaling axes involved in the chemoattraction and engraftment of MSCs to an injured tissue or to a tissue undergoing active remodeling. Moreover, experimental evidence in support of uses of MSCs as vehicles of therapeutic genes is discussed. Because of its regenerative capacity and its particular immune properties, the liver is a good model to analyze the potential of MSC-based therapies. Finally, the potential application of MSCs and genetically modified MSCs in liver fibrosis and hepatocellular carcinoma (HCC) is proposed in view of available evidence.
Collapse
|
47
|
García-Castro J, Alemany R, Cascalló M, Martínez-Quintanilla J, Arriero MDM, Lassaletta A, Madero L, Ramírez M. Treatment of metastatic neuroblastoma with systemic oncolytic virotherapy delivered by autologous mesenchymal stem cells: an exploratory study. Cancer Gene Ther 2010; 17:476-83. [PMID: 20168350 DOI: 10.1038/cgt.2010.4] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Treatment of metastatic tumors with engineered adenoviruses that replicate selectively in tumor cells is a new therapeutic approach in cancer. Systemic administration of these oncolytic adenoviruses lack metastatic targeting ability. The tumor stroma engrafting property of intravenously injected mesenchymal stem cells (MSCs) may allow the use of MSCs as cellular vehicles for targeted delivery. In this work, we study the safety and the efficacy of infusing autologous MSCs infected with ICOVIR-5, a new oncolytic adenovirus, for treating metastatic neuroblastoma. Four children with metastatic neuroblastoma refractory to front-line therapies received several doses of autologous MSCs carrying ICOVIR-5, under an approved preliminary study. The tolerance to the treatment was excellent. A complete clinical response was documented in one case, and the child is in complete remission 3 years after this therapy. We postulate that MSCs can deliver oncolytic adenoviruses to metastatic tumors with very low systemic toxicity and with beneficial antitumor effects.
Collapse
Affiliation(s)
- J García-Castro
- Servicio de Oncohematología y Trasplante, Hospital Universitario Niño Jesús, Avenida Menéndez Pelayo 65, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Lentivirus-modified human umbilical cord mesenchymal stem cells maintain their pluripotency. Biotechnol Appl Biochem 2010; 55:53-62. [DOI: 10.1042/ba20090210] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
49
|
Abstract
Oncolytic adenoviruses (Ads) constitute a promising new class of anticancer agent. They are based on the well-studied adenoviral vector system, which lends itself to concept-driven design to generate oncolytic variants. The first oncolytic Ad was approved as a drug in China in 2005, although clinical efficacy observed in human trials has failed to reach the high expectations that were based on studies in animal models. Current obstacles to the full realization of efficacy of this class of anticancer agent include (i) limited efficiency of infection and specific replication in tumor cells, (ii) limited vector spread within the tumor, (iii) imperfect animal models and methods of in vivo imaging, and (iv) an incomplete understanding of the interaction of these agents with the host. In this review, we discuss recent advances in the field of oncolytic Ads and potential ways to overcome current obstacles to their clinical application and efficacy.
Collapse
|
50
|
Dembinski JL, Spaeth EL, Fueyo J, Gomez-Manzano C, Studeny M, Andreeff M, Marini FC. Reduction of nontarget infection and systemic toxicity by targeted delivery of conditionally replicating viruses transported in mesenchymal stem cells. Cancer Gene Ther 2009; 17:289-97. [PMID: 19876078 DOI: 10.1038/cgt.2009.67] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The fiber-modified adenoviral vector Delta-24-RGD (D24RGD) offers vast therapeutic potential. Direct injection of D24RGD has been used to successfully target ovarian tumors in mice. However, systemic toxicity, especially in the liver, profoundly limits the efficacy of direct viral vector delivery. Mesenchymal stem cells (MSC) have the ability to function as a vector for targeted gene therapy because of their preferential engraftment into solid tumors and participation in tumor stroma formation. We show that MSC-guided delivery of D24RGD is specific and efficient and reduces the overall systemic toxicity in mice to negligible levels compared with D24RGD alone. In our model, we found efficient targeted delivery of MSC-D24RGD to both breast and ovarian cell lines. Furthermore, immunohistochemical staining for adenoviral hexon protein confirmed negligible levels of systemic toxicity in mice that were administered MSC-D24RGD compared with those that were administered D24RGD. These data suggest that delivery of D24RGD through MSC not only increases the targeted delivery efficiency, but also reduces the systemic exposure of the virus, thereby reducing overall systemic toxicity to the host and ultimately enhancing its value as an anti-tumor therapeutic candidate.
Collapse
Affiliation(s)
- J L Dembinski
- Department of Stem Cell Transplantation and Cellular Therapy, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|