1
|
Bardhan M, Muneer MA, Khare A, Minesh Shah R, Kaur A, Vasipalli SS, Suresh V, Podder V, Ahluwalia M, Odia Y, Chen Z. Advances in stem cell-based therapeutic transfers for glioblastoma treatment. Expert Rev Neurother 2025:1-17. [PMID: 40245098 DOI: 10.1080/14737175.2025.2490543] [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: 09/23/2024] [Revised: 02/25/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025]
Abstract
INTRODUCTION Glioblastoma (GBM), a highly malignant brain tumor, has a poor prognosis despite standard treatments like surgery, chemotherapy, and radiation. Glioblastoma stem cells (GSCs) play a critical role in recurrence and therapy resistance. Stem cell-based therapies have emerged as innovative approaches, leveraging the tumor-targeting abilities of stem cells to deliver treatments directly to GBM. AREAS COVERED This review focuses on using intact stem cells or subtypes for GBM therapy, excluding antigenic characteristics. The stem cell-based therapies explored include neural, mesenchymal, glioblastoma, hematopoietic and adipose-derived stem cells that have been investigated in both clinical and preclinical settings. A systematic search in PubMed, EMBASE, ClinicalTrials.gov, and Scopus had identified research up until January 2024. Key mechanisms reviewed include immune modulation, angiogenesis inhibition, and apoptosis induction. Discussion of completed and ongoing trials include emphasis on safety, efficacy, challenges, and study design limitations. EXPERT OPINION Stem cell-based therapies hold promise for treating GBM by targeting GSCs and improving treatment outcomes. Despite some potential advantages, challenges such as tumorigenesis risks, delivery complexities, and sustained therapeutic effects persist. Future research should prioritize optimizing stem cell modifications, combining them with current treatments, and conducting large-scale trials to ensure safety and efficacy. Integrating stem cell therapies into GBM treatment could provide more effective and less invasive options for patients.
Collapse
Affiliation(s)
- Mainak Bardhan
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | | | - Abhinav Khare
- All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| | | | - Anmol Kaur
- Lady Hardinge Medical College, New Delhi, India
| | - Sonit Sai Vasipalli
- Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Vinay Suresh
- King George's Medical University, Lucknow, India
| | - Vivek Podder
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Manmeet Ahluwalia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Yazmin Odia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Zhijian Chen
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| |
Collapse
|
2
|
Benmelouka AY, Munir M, Sayed A, Attia MS, Ali MM, Negida A, Alghamdi BS, Kamal MA, Barreto GE, Ashraf GM, Meshref M, Bahbah EI. Neural Stem Cell-Based Therapies and Glioblastoma Management: Current Evidence and Clinical Challenges. Int J Mol Sci 2021; 22:2258. [PMID: 33668356 PMCID: PMC7956497 DOI: 10.3390/ijms22052258] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 02/05/2023] Open
Abstract
Gliomas, which account for nearly a quarter of all primary CNS tumors, present significant contemporary therapeutic challenges, particularly the highest-grade variant (glioblastoma multiforme), which has an especially poor prognosis. These difficulties are due to the tumor's aggressiveness and the adverse effects of radio/chemotherapy on the brain. Stem cell therapy is an exciting area of research being explored for several medical issues. Neural stem cells, normally present in the subventricular zone and the hippocampus, preferentially migrate to tumor masses. Thus, they have two main advantages: They can minimize the side effects associated with systemic radio/chemotherapy while simultaneously maximizing drug delivery to the tumor site. Another feature of stem cell therapy is the variety of treatment approaches it allows. Stem cells can be genetically engineered into expressing a wide variety of immunomodulatory substances that can inhibit tumor growth. They can also be used as delivery vehicles for oncolytic viral vectors, which can then be used to combat the tumorous mass. An alternative approach would be to combine stem cells with prodrugs, which can subsequently convert them into the active form upon migration to the tumor mass. As with any therapeutic modality still in its infancy, much of the research regarding their use is primarily based upon knowledge gained from animal studies, and a number of ongoing clinical trials are currently investigating their effectiveness in humans. The aim of this review is to highlight the current state of stem cell therapy in the treatment of gliomas, exploring the different mechanistic approaches, clinical applicability, and the existing limitations.
Collapse
Affiliation(s)
| | - Malak Munir
- Faculty of Medicine, Ain Shams University, Cairo 11591, Egypt; (M.M.); (A.S.)
| | - Ahmed Sayed
- Faculty of Medicine, Ain Shams University, Cairo 11591, Egypt; (M.M.); (A.S.)
| | - Mohamed Salah Attia
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Mohamad M. Ali
- Faculty of Medicine, Al-Azhar University, Damietta 34511, Egypt; (M.M.A.); (E.I.B.)
| | - Ahmed Negida
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2UP, UK;
- Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Badrah S. Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; or
| | - Mohammad Amjad Kamal
- West China School of Nursing/Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China;
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia
- Novel Global Community Educational Foundation, 7 Peterlee Place, Hebersham, NSW 2770, Australia
| | - George E. Barreto
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 32310, Chile
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; or
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | | | - Eshak I. Bahbah
- Faculty of Medicine, Al-Azhar University, Damietta 34511, Egypt; (M.M.A.); (E.I.B.)
| |
Collapse
|
3
|
Tu GXE, Ho YK, Ng ZX, Teo KJ, Yeo TT, Too HP. A facile and scalable in production non-viral gene engineered mesenchymal stem cells for effective suppression of temozolomide-resistant (TMZR) glioblastoma growth. Stem Cell Res Ther 2020; 11:391. [PMID: 32917269 PMCID: PMC7488524 DOI: 10.1186/s13287-020-01899-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/28/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) serve as an attractive vehicle for cell-directed enzyme prodrug therapy (CDEPT) due to their unique tumour-nesting ability. Such approach holds high therapeutic potential for treating solid tumours including glioblastoma multiforme (GBM), a devastating disease with limited effective treatment options. Currently, it is a common practice in research and clinical manufacturing to use viruses to deliver therapeutic genes into MSCs. However, this is limited by the inherent issues of safety, high cost and demanding manufacturing processes. The aim of this study is to identify a facile, scalable in production and highly efficient non-viral method to transiently engineer MSCs for prolonged and exceptionally high expression of a fused transgene: yeast cytosine deaminase::uracil phosphoribosyl-transferase::green fluorescent protein (CD::UPRT::GFP). METHODS MSCs were transfected with linear polyethylenimine using a cpg-free plasmid encoding the transgene in the presence of a combination of fusogenic lipids and β tubulin deacetylase inhibitor (Enhancer). Process scalability was evaluated in various planar vessels and microcarrier-based bioreactor. The transfection efficiency was determined with flow cytometry, and the therapeutic efficacy of CD::UPRT::GFP expressing MSCs was evaluated in cocultures with temozolomide (TMZ)-sensitive or TMZ-resistant human glioblastoma cell lines. In the presence of 5-fluorocytosine (5FC), the 5-fluorouracil-mediated cytotoxicity was determined by performing colometric MTS assay. In vivo antitumor effects were examined by local injection into subcutaneous TMZ-resistant tumors implanted in the athymic nude mice. RESULTS At > 90% transfection efficiency, the phenotype, differentiation potential and tumour tropism of MSCs were unaltered. High reproducibility was observed in all scales of transfection. The therapeutically modified MSCs displayed strong cytotoxicity towards both TMZ-sensitive and TMZ-resistant U251-MG and U87-MG cell lines only in the presence of 5FC. The effectiveness of this approach was further validated with other well-characterized and clinically annotated patient-derived GBM cells. Additionally, a long-term suppression (> 30 days) of the growth of a subcutaneous TMZ-resistant U-251MG tumour was demonstrated. CONCLUSIONS Collectively, this highly efficient non-viral workflow could potentially enable the scalable translation of therapeutically engineered MSC for the treatment of TMZ-resistant GBM and other applications beyond the scope of this study.
Collapse
Affiliation(s)
- Geraldine Xue En Tu
- Department of Biochemistry, National University of Singapore, Singapore, 117596, Singapore
| | - Yoon Khei Ho
- Department of Biochemistry, National University of Singapore, Singapore, 117596, Singapore.
| | - Zhi Xu Ng
- Division of Neurosurgery, Department of General Surgery, Khoo Teck Puat Hospital, Singapore, 768828, Singapore
| | - Ke Jia Teo
- Division of Neurosurgery, Department of General Surgery, National University Hospital, National University Health Systems, Singapore, Singapore
| | - Tseng Tsai Yeo
- Division of Neurosurgery, Department of General Surgery, National University Hospital, National University Health Systems, Singapore, Singapore
| | - Heng-Phon Too
- Department of Biochemistry, National University of Singapore, Singapore, 117596, Singapore
| |
Collapse
|
4
|
Tamura R, Miyoshi H, Yoshida K, Okano H, Toda M. Recent progress in the research of suicide gene therapy for malignant glioma. Neurosurg Rev 2019; 44:29-49. [PMID: 31781985 DOI: 10.1007/s10143-019-01203-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022]
Abstract
Malignant glioma, which is characterized by diffuse infiltration into the normal brain parenchyma, is the most aggressive primary brain tumor with dismal prognosis. Over the past 40 years, the median survival has only slightly improved. Therefore, new therapeutic modalities must be developed. In the 1990s, suicide gene therapy began attracting attention for the treatment of malignant glioma. Some clinical trials used a viral vector for suicide gene transduction; however, it was found that viral vectors cannot cover the large invaded area of glioma cells. Interest in this therapy was recently revived because some types of stem cells possess a tumor-tropic migratory capacity, which can be used as cellular delivery vehicles. Immortalized, clonal neural stem cell (NSC) line has been used for patients with recurrent high-grade glioma, which showed safety and efficacy. Embryonic and induced pluripotent stem cells may be considered as sources of NSC because NSC is difficult to harvest, and ethical issues have been raised. Mesenchymal stem cells are alternative candidates for cellular vehicle and are easily harvested from the bone marrow. In addition, a new type of nonlytic, amphotropic retroviral replicating vector encoding suicide gene has shown efficacy in patients with recurrent high-grade glioma in a clinical trial. This replicating viral capacity is another possible candidate as delivery vehicle to tackle gliomas. Herein, we review the concept of suicide gene therapy, as well as recent progress in preclinical and clinical studies in this field.
Collapse
Affiliation(s)
- Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Miyoshi
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
5
|
The inhibiting effect of neural stem cells on proliferation and invasion of glioma cells. Oncotarget 2017; 8:76949-76960. [PMID: 29100360 PMCID: PMC5652754 DOI: 10.18632/oncotarget.20270] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 06/02/2017] [Indexed: 12/11/2022] Open
Abstract
The invasive and infiltrative nature of tumor cells leads to the poor prognosis of glioma. Currently, novel therapeutic means to eliminate the tumor cells without damaging the normal brain tissue are still strongly demanded. Significant attentions had been paid to stem cell-based therapy and neural stem cell (NSC) had been considered as one of the efficient delivery vehicles for targeting therapeutic genes. However, whether the NSCs could directly affect glioma cells remains to be seen. In this study, both rat and human glioma cells (C6 and U251) were co-cultured with normal rat embryonic NSCs directly or in-directly. We found the survival, proliferation, invasion and migration of glioma cells were significantly inhibited, while the differentiation was not affected in the in vitro co-culture system. In nude mice, although no significant difference was observed in the tumor growth, survival status and time of tumor-bearing mice were significantly promoted when U251 cells were subcutaneously injected with NSCs. In coincidence with the suppression of glioma cell growth in vitro, expression of mutant p53 and phosphorylation of AKT, ERK1/2 decreased while the level of caspase-3 increased significantly. Our results suggested that normal NSCs could possess direct anti-glioma properties via inhibiting the glioma cell viability, proliferation, invasion and migration. It could be a very promising candidate for glioma treatment.
Collapse
|
6
|
Barish ME, Herrmann K, Tang Y, Argalian Herculian S, Metz M, Aramburo S, Tirughana R, Gutova M, Annala A, Moats RA, Goldstein L, Rockne RC, Gutierrez J, Brown CE, Ghoda L, Aboody KS. Human Neural Stem Cell Biodistribution and Predicted Tumor Coverage by a Diffusible Therapeutic in a Mouse Glioma Model. Stem Cells Transl Med 2017; 6:1522-1532. [PMID: 28481046 PMCID: PMC5689763 DOI: 10.1002/sctm.16-0397] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 02/20/2017] [Indexed: 12/20/2022] Open
Abstract
Engineered neural stem cells (NSCs) intrinsically migrating to brain tumors offer a promising mechanism for local therapeutic delivery. However, difficulties in quantitative assessments of NSC migration and in estimates of tumor coverage by diffusible therapeutics have impeded development and refinement of NSC-based therapies. To address this need, we developed techniques by which conventional serial-sectioned formalin-fixed paraffin-embedded (FFPE) brains can be analyzed in their entirety across multiple test animals. We considered a conventional human glioblastoma model: U251 glioma cells orthotopically engrafted in immunodeficient mice receiving intracerebral (i.c.) or intravenous (i.v.) administrations of NSCs expressing a diffusible enzyme to locally catalyze chemotherapeutic formation. NSC migration to tumor sites was dose-dependent, reaching 50%-60% of total administered NSCs for the i.c route and 1.5% for the i.v. route. Curiously, the most efficient NSC homing was seen with smaller NSC doses, implying existence of rate-limiting process active during administration and/or migration. Predicted tumor exposure to a diffusing therapeutic (assuming a 50 µm radius of action) could reach greater than 50% of the entire tumor volume for i.c. and 25% for i.v. administration. Within individual sections, coverage of tumor area could be as high as 100% for i.c. and 70% for i.v. routes. Greater estimated therapeutic coverage was observed for larger tumors and for larger tumor regions in individual sections. Overall, we have demonstrated a framework within which investigators may rationally evaluate NSC migration to, and integration into, brain tumors, and therefore enhance understanding of mechanisms that both promote and limit this therapeutic modality. Stem Cells Translational Medicine 2017;6:1522-1532.
Collapse
Affiliation(s)
- Michael E Barish
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Kelsey Herrmann
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Yang Tang
- Department of Radiology, University of Southern California, Los Angeles, California, USA
| | - Siranush Argalian Herculian
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Marianne Metz
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Soraya Aramburo
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Revathiswari Tirughana
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Margarita Gutova
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Alexander Annala
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Rex A Moats
- Department of Radiology, University of Southern California, Los Angeles, California, USA.,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Leanne Goldstein
- Department of Information Sciences, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Russell C Rockne
- Department of Information Sciences, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Jennifer Gutierrez
- Department of Information Sciences, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Christine E Brown
- Department of Hematology/HCT, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA.,Department of Immuno-Oncology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Lucy Ghoda
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Karen S Aboody
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA.,Department of Division of Neurosurgery, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| |
Collapse
|
7
|
Gardeck AM, Sheehan J, Low WC. Immune and viral therapies for malignant primary brain tumors. Expert Opin Biol Ther 2017; 17:457-474. [DOI: 10.1080/14712598.2017.1296132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Andrew M. Gardeck
- Departments of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - Jordan Sheehan
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Walter C. Low
- Departments of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
8
|
Portnow J, Synold TW, Badie B, Tirughana R, Lacey SF, D'Apuzzo M, Metz MZ, Najbauer J, Bedell V, Vo T, Gutova M, Frankel P, Chen M, Aboody KS. Neural Stem Cell-Based Anticancer Gene Therapy: A First-in-Human Study in Recurrent High-Grade Glioma Patients. Clin Cancer Res 2016; 23:2951-2960. [PMID: 27979915 DOI: 10.1158/1078-0432.ccr-16-1518] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 11/08/2016] [Accepted: 11/29/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Human neural stem cells (NSC) are inherently tumor tropic, making them attractive drug delivery vehicles. Toward this goal, we retrovirally transduced an immortalized, clonal NSC line to stably express cytosine deaminase (HB1.F3.CD.C21; CD-NSCs), which converts the prodrug 5-fluorocytosine (5-FC) to 5-fluorouracil (5-FU).Experimental Design: Recurrent high-grade glioma patients underwent intracranial administration of CD-NSCs during tumor resection or biopsy. Four days later, patients began taking oral 5-FC every 6 hours for 7 days. Study treatment was given only once. A standard 3 + 3 dose escalation schema was used to increase doses of CD-NSCs from 1 × 107 to 5 × 107 and 5-FC from 75 to 150 mg/kg/day. Intracerebral microdialysis was performed to measure brain levels of 5-FC and 5-FU. Serial blood samples were obtained to assess systemic drug concentrations as well as to perform immunologic correlative studies.Results: Fifteen patients underwent study treatment. We saw no dose-limiting toxicity (DLT) due to the CD-NSCs. There was 1 DLT (grade 3 transaminitis) possibly related to 5-FC. We did not see development of anti-CD-NSC antibodies and did not detect CD-NSCs or replication-competent retrovirus in the systemic circulation. Intracerebral microdialysis revealed that CD-NSCs produced 5-FU locally in the brain in a 5-FC dose-dependent manner. Autopsy data indicate that CD-NSCs migrated to distant tumor sites and were nontumorigenic.Conclusions: Collectively, our results from this first-in-human study demonstrate initial safety and proof of concept regarding the ability of NSCs to target brain tumors and locally produce chemotherapy. Clin Cancer Res; 23(12); 2951-60. ©2016 AACR.
Collapse
Affiliation(s)
- Jana Portnow
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, California.
| | | | - Behnam Badie
- Division of Neurosurgery, City of Hope, Duarte, California
| | | | - Simon F Lacey
- Clinical Immunobiology Correlative Studies Laboratory, City of Hope, Duarte, California
| | | | - Marianne Z Metz
- Department of Developmental & Stem Cell Biology, City of Hope, Duarte, California
| | - Joseph Najbauer
- Department of Developmental & Stem Cell Biology, City of Hope, Duarte, California
| | | | - Tien Vo
- Department of Developmental & Stem Cell Biology, City of Hope, Duarte, California
| | - Margarita Gutova
- Department of Developmental & Stem Cell Biology, City of Hope, Duarte, California
| | - Paul Frankel
- Division of Biostatistics, City of Hope, Duarte, California
| | - Mike Chen
- Division of Neurosurgery, City of Hope, Duarte, California
| | - Karen S Aboody
- Division of Neurosurgery, City of Hope, Duarte, California.,Department of Developmental & Stem Cell Biology, City of Hope, Duarte, California
| |
Collapse
|
9
|
Current Perspective of Stem Cell Therapy in Neurodegenerative and Metabolic Diseases. Mol Neurobiol 2016; 54:7276-7296. [PMID: 27815831 DOI: 10.1007/s12035-016-0217-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 10/12/2016] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases have been an unsolved riddle for quite a while; to date, there are no proper and effective curative treatments and only palliative and symptomatic treatments are available to treat these illnesses. The absence of therapeutic treatments for neurodegenerative ailments has huge economic hit and strain on the society. Pharmacotherapies and various surgical procedures like deep brain stimulation are being given to the patient, but they are only effective for the symptoms and not for the diseases. This paper reviews the recent studies and development of stem cell therapy for neurodegenerative disorders. Stem cell-based treatment is a promising new way to deal with neurodegenerative diseases. Stem cell transplantation can advance useful recuperation by delivering trophic elements that impel survival and recovery of host neurons in animal models and patients with neurodegenerative maladies. Several mechanisms, for example, substitution of lost cells, cell combination, release of neurotrophic factor, proliferation of endogenous stem cell, and transdifferentiation, may clarify positive remedial results. With the current advancements in the stem cell therapies, a new hope for the cure has come out since they have potential to be a cure for the same. This review compiles stem cell therapy recent conceptions in neurodegenerative and neurometabolic diseases and updates in this field. Graphical Absract ᅟ.
Collapse
|
10
|
Gabashvili AN, Baklaushev VP, Grinenko NF, Mel'nikov PA, Cherepanov SA, Levinsky AB, Chehonin VP. Antitumor Activity of Rat Mesenchymal Stem Cells during Direct or Indirect Co-Culturing with C6 Glioma Cells. Bull Exp Biol Med 2016; 160:519-524. [PMID: 26902362 DOI: 10.1007/s10517-016-3211-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Indexed: 10/22/2022]
Abstract
The tumor-suppressive effect of rat mesenchymal stem cells against low-differentiated rat C6 glioma cells during their direct and indirect co-culturing and during culturing of C6 glioma cells in the medium conditioned by mesenchymal stem cells was studied in an in vitro experiment. The most pronounced antitumor activity of mesenchymal stem cells was observed during direct co-culturing with C6 glioma cells. The number of live C6 glioma cells during indirect co-culturing and during culturing in conditioned medium was slightly higher than during direct co-culturing, but significantly differed from the control (C6 glioma cells cultured in medium conditioned by C6 glioma cells). The cytotoxic effect of medium conditioned by mesenchymal stem cells was not related to medium depletion by glioma cells during their growth. The medium conditioned by other "non-stem" cells (rat astrocytes and fibroblasts) produced no tumor-suppressive effect. Rat mesenchymal stem cells, similar to rat C6 glioma cells express connexin 43, the main astroglial gap junction protein. During co-culturing, mesenchymal stem cells and glioma C6 cells formed functionally active gap junctions. Gap junction blockade with connexon inhibitor carbenoxolone attenuated the antitumor effect observed during direct co-culturing of C6 glioma cells and mesenchymal stem cells to the level produced by conditioned medium. Cell-cell signaling mediated by gap junctions can be a mechanism of the tumor-suppressive effect of mesenchymal stem cells against C6 glioma cells. This phenomenon can be used for the development of new methods of cell therapy for high-grade malignant gliomas.
Collapse
Affiliation(s)
- A N Gabashvili
- Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N. I. Pirogov National Research Medical University, Moscow, Russia.
| | - V P Baklaushev
- Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N. I. Pirogov National Research Medical University, Moscow, Russia
- Federal Research-and-Clinical Center, Federal Medico-Biological Agency, Moscow, Russia
| | - N F Grinenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky Federal Medical Research Center of Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - P A Mel'nikov
- Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N. I. Pirogov National Research Medical University, Moscow, Russia
| | - S A Cherepanov
- Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N. I. Pirogov National Research Medical University, Moscow, Russia
| | - A B Levinsky
- Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N. I. Pirogov National Research Medical University, Moscow, Russia
| | - V P Chehonin
- Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N. I. Pirogov National Research Medical University, Moscow, Russia
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky Federal Medical Research Center of Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
11
|
NAMBA HIROKI, KAWAJI HIROSHI, YAMASAKI TOMOHIRO. Use of genetically engineered stem cells for glioma therapy. Oncol Lett 2016; 11:9-15. [PMID: 26870161 PMCID: PMC4726949 DOI: 10.3892/ol.2015.3860] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 09/24/2015] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma, the most common and most malignant type of primary brain tumor, is associated with poor prognosis, even when treated using combined therapies, including surgery followed by concomitant radiotherapy with temozolomide-based chemotherapy. The invasive nature of this type of tumor is a major reason underlying treatment failure. The tumor-tropic ability of neural and mesenchymal stem cells offers an alternative therapeutic approach, where these cells may be used as vehicles for the invasion of tumors. Stem cell-based therapy is particularly attractive due to its tumor selectivity, meaning that the stem cells are able to target tumor cells without harming healthy brain tissue, as well as the extensive tumor tropism of stem cells when delivering anti-tumor substances, even to distant tumor microsatellites. Stem cells have previously been used to deliver cytokine genes, suicide genes and oncolytic viruses. The present review will summarize current trends in experimental studies of stem cell-based gene therapy against gliomas, and discuss the potential concerns for translating these promising strategies into clinical use.
Collapse
Affiliation(s)
- HIROKI NAMBA
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - HIROSHI KAWAJI
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - TOMOHIRO YAMASAKI
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| |
Collapse
|
12
|
Truong SN, Van Pham P. Stem cell technology and engineering for cancer treatment. BIOMEDICAL RESEARCH AND THERAPY 2015. [DOI: 10.7603/s40730-015-0013-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
13
|
Gabashvili AN, Baklaushev VP, Grinenko NF, Levinskii AB, Mel'nikov PA, Cherepanov SA, Chekhonin VP. Functionally Active Gap Junctions between Connexin 43-Positive Mesenchymal Stem Cells and Glioma Cells. Bull Exp Biol Med 2015; 159:173-179. [PMID: 26033611 DOI: 10.1007/s10517-015-2916-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Indexed: 12/14/2022]
Abstract
The formation of functional gap junctions between mesenchymal stem cells and cells of low-grade rat glioma C6 cells was studied in in vitro experiments. Immunocytochemical analysis with antibodies to connexin 43 extracellular loop 2 showed that mesenchymal stem cells as well as C6 glioma cells express the main astroglial gap junction protein connexin 43. Analysis of migration activity showed that mesenchymal stem cells actively migrate towards C6 glioma cells. During co-culturing, mesenchymal stem cells and glioma C6 form functionally active gap junctions mediating the transport of cytoplasmic dye from glioma cells to mesenchymal stem cells in the opposite direction. Fluorometry showed that the intensity of transport of low-molecular substances through heterologous gap junctions between mesenchymal stem cells and glioma cells is similar to that through homologous gap junctions between glioma cells. This phenomenon can be used for the development of new methods of cell therapy of high-grade gliomas.
Collapse
Affiliation(s)
- A N Gabashvili
- Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N. I. Pirogov Russian National Research Medical University, Moscow, Russia,
| | | | | | | | | | | | | |
Collapse
|
14
|
Huang TT, Parab S, Burnett R, Diago O, Ostertag D, Hofman FM, Espinoza FL, Martin B, Ibañez CE, Kasahara N, Gruber HE, Pertschuk D, Jolly DJ, Robbins JM. Intravenous administration of retroviral replicating vector, Toca 511, demonstrates therapeutic efficacy in orthotopic immune-competent mouse glioma model. Hum Gene Ther 2015; 26:82-93. [PMID: 25419577 DOI: 10.1089/hum.2014.100] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Toca 511 (vocimagene amiretrorepvec), a nonlytic, amphotropic retroviral replicating vector (RRV), encodes and delivers a functionally optimized yeast cytosine deaminase (CD) gene to tumors. In orthotopic glioma models treated with Toca 511 and 5-fluorocytosine (5-FC) the CD enzyme within infected cells converts 5-FC to 5-fluorouracil (5-FU), resulting in tumor killing. Toca 511, delivered locally either by intratumoral injection or by injection into the resection bed, in combination with subsequent oral extended-release 5-FC (Toca FC), is under clinical investigation in patients with recurrent high-grade glioma (HGG). If feasible, intravenous administration of vectors is less invasive, can easily be repeated if desired, and may be applicable to other tumor types. Here, we present preclinical data that support the development of an intravenous administration protocol. First we show that intravenous administration of Toca 511 in a preclinical model did not lead to widespread or uncontrolled replication of the RVV. No, or low, viral DNA was found in the blood and most of the tissues examined 180 days after Toca 511 administration. We also show that RRV administered intravenously leads to efficient infection and spread of the vector carrying the green fluorescent protein (GFP)-encoding gene (Toca GFP) through tumors in both immune-competent and immune-compromised animal models. However, initial vector localization within the tumor appeared to depend on the mode of administration. Long-term survival was observed in immune-competent mice when Toca 511 was administered intravenously or intracranially in combination with 5-FC treatment, and this combination was well tolerated in the preclinical models. Enhanced survival could also be achieved in animals with preexisting immune response to vector, supporting the potential for repeated administration. On the basis of these and other supporting data, a clinical trial investigating intravenous administration of Toca 511 in patients with recurrent HGG is currently open and enrolling.
Collapse
|
15
|
Okura H, Smith CA, Rutka JT. Gene therapy for malignant glioma. MOLECULAR AND CELLULAR THERAPIES 2014; 2:21. [PMID: 26056588 PMCID: PMC4451964 DOI: 10.1186/2052-8426-2-21] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 06/27/2014] [Indexed: 01/01/2023]
Abstract
Glioblastoma multiforme (GBM) is the most frequent and devastating primary brain tumor in adults. Despite current treatment modalities, such as surgical resection followed by chemotherapy and radiotherapy, only modest improvements in median survival have been achieved. Frequent recurrence and invasiveness of GBM are likely due to the resistance of glioma stem cells to conventional treatments; therefore, novel alternative treatment strategies are desperately needed. Recent advancements in molecular biology and gene technology have provided attractive novel treatment possibilities for patients with GBM. Gene therapy is defined as a technology that aims to modify the genetic complement of cells to obtain therapeutic benefit. To date, gene therapy for the treatment of GBM has demonstrated anti-tumor efficacy in pre-clinical studies and promising safety profiles in clinical studies. However, while this approach is obviously promising, concerns still exist regarding issues associated with transduction efficiency, viral delivery, the pathologic response of the brain, and treatment efficacy. Tumor development and progression involve alterations in a wide spectrum of genes, therefore a variety of gene therapy approaches for GBM have been proposed. Improved viral vectors are being evaluated, and the potential use of gene therapy alone or in synergy with other treatments against GBM are being studied. In this review, we will discuss the most commonly studied gene therapy approaches for the treatment of GBM in preclinical and clinical studies including: prodrug/suicide gene therapy; oncolytic gene therapy; cytokine mediated gene therapy; and tumor suppressor gene therapy. In addition, we review the principles and mechanisms of current gene therapy strategies as well as advantages and disadvantages of each.
Collapse
Affiliation(s)
- Hidehiro Okura
- The Arthur and Sonia Labatt Brain Tumour Research Centre, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, 17th Floor, Toronto, ON M5G 0A4 Canada ; Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Christian A Smith
- The Arthur and Sonia Labatt Brain Tumour Research Centre, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, 17th Floor, Toronto, ON M5G 0A4 Canada
| | - James T Rutka
- The Arthur and Sonia Labatt Brain Tumour Research Centre, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, 17th Floor, Toronto, ON M5G 0A4 Canada ; Department of Surgery, University of Toronto, 149 College Street, 5th Floor, Toronto, Ontario M5T 1P5 Canada ; Division of Neurosurgery, The Hospital for Sick Children, Suite 1503, 555 University Avenue, Toronto, Ontario M5G 1X8 Canada
| |
Collapse
|
16
|
PD-L1 expression by neurons nearby tumors indicates better prognosis in glioblastoma patients. J Neurosci 2013; 33:14231-45. [PMID: 23986257 DOI: 10.1523/jneurosci.5812-12.2013] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of brain tumor. In general, tumor growth requires disruption of the tissue microenvironment, yet how this affects glioma progression is unknown. We studied program death-ligand (PD-L)1 in neurons and gliomas in tumors from GBM patients and associated the findings with clinical outcome. Remarkably, we found that upregulation of PD-L1 by neurons in tumor-adjacent brain tissue (TABT) associated positively with GBM patient survival, whereas lack of neuronal PD-L1 expression was associated with high PD-L1 in tumors and unfavorable prognosis. To understand the molecular mechanism of PD-L1 signaling in neurons, we investigated PD-L1 function in cerebellar and cortical neurons and its impact on gliomas. We discovered that neuronal PD-L1-induced caspase-dependent apoptosis of glioma cells. Because interferon (IFN)-β induces PD-L1 expression, we studied the functional consequences of neuronal Ifnb gene deletion on PD-L1 signaling and function. Ifnb-/- neurons lacked PD-L1 and were defective in inducing glioma cell death; this effect was reversed on PD-L1 gene transfection. Ifnb-/- mice with intracerebral isografts survived poorly. Similar to the observations in GBM patients, better survival in wild-type mice was associated with high neuronal PD-L1 in TABT and downregulation of PD-L1 in tumors, which was defective in Ifnb-/- mice. Our data indicated that neuronal PD-L1 signaling in brain cells was important for GBM patient survival. Reciprocal PD-L1 regulation in TABT and tumor tissue could be a prognostic biomarker for GBM. Understanding the complex interactions between tumor and adjacent stromal tissue is important in designing targeted GBM therapies.
Collapse
|
17
|
Kwiatkowska A, Nandhu MS, Behera P, Chiocca EA, Viapiano MS. Strategies in gene therapy for glioblastoma. Cancers (Basel) 2013; 5:1271-305. [PMID: 24202446 PMCID: PMC3875940 DOI: 10.3390/cancers5041271] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 10/15/2013] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive form of brain cancer, with a dismal prognosis and extremely low percentage of survivors. Novel therapies are in dire need to improve the clinical management of these tumors and extend patient survival. Genetic therapies for GBM have been postulated and attempted for the past twenty years, with variable degrees of success in pre-clinical models and clinical trials. Here we review the most common approaches to treat GBM by gene therapy, including strategies to deliver tumor-suppressor genes, suicide genes, immunomodulatory cytokines to improve immune response, and conditionally-replicating oncolytic viruses. The review focuses on the strategies used for gene delivery, including the most common and widely used vehicles (i.e., replicating and non-replicating viruses) as well as novel therapeutic approaches such as stem cell-mediated therapy and nanotechnologies used for gene delivery. We present an overview of these strategies, their targets, different advantages, and challenges for success. Finally, we discuss the potential of gene therapy-based strategies to effectively attack such a complex genetic target as GBM, alone or in combination with conventional therapy.
Collapse
Affiliation(s)
- Aneta Kwiatkowska
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
18
|
Lee JY, Lee DH, Kim HA, Choi SA, Lee HJ, Park CK, Phi JH, Wang KC, Kim SU, Kim SK. Double suicide gene therapy using human neural stem cells against glioblastoma: double safety measures. J Neurooncol 2013; 116:49-57. [PMID: 24113876 DOI: 10.1007/s11060-013-1264-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 09/22/2013] [Indexed: 11/30/2022]
|
19
|
Development and application of neural stem cells for treating various human neurological diseases in animal models. Lab Anim Res 2013; 29:131-7. [PMID: 24106507 PMCID: PMC3791346 DOI: 10.5625/lar.2013.29.3.131] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/08/2013] [Accepted: 07/12/2013] [Indexed: 12/12/2022] Open
Abstract
Stem cells derived from adult tissues or the inner cell mass (ICM) of embryos in the mammalian blastocyst (BL) stage are capable of self-renewal and have remarkable potential for undergoing lineage-specific differentiation under in vitro culturing conditions. In particular, neural stem cells (NSCs) that self-renew and differentiate into major cell types of the brain exist in the developing and adult central nervous system (CNS). The exact function and distribution of NSCs has been assessed, and they represent an interesting population that includes astrocytes, oligodendrocytes, and neurons. Many researchers have demonstrated functional recovery in animal models of various neurological diseases such as stroke, Parkinson's disease (PD), brain tumors, and metastatic tumors. The safety and efficacy of stem cell-based therapies (SCTs) are also being evaluated in humans. The therapeutic efficacy of NSCs has been shown in the brain disorder-induced animal models, and animal models may be well established to perform the test before clinical stage. Taken together, data from the literature have indicated that therapeutic NSCs may be useful for selectively treating diverse types of human brain diseases without incurring adverse effects.
Collapse
|
20
|
Jin G, Zhou Y, Chai Q, Zhu G, Xu F, Liu F. VP22 and cytosine deaminase fusion gene modified tissue-engineered neural stem cells for glioma therapy. J Cancer Res Clin Oncol 2013; 139:475-83. [PMID: 23180018 DOI: 10.1007/s00432-012-1347-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 10/31/2012] [Indexed: 12/20/2022]
Abstract
PURPOSE The herpes simplex virus type 1 tegument protein VP22 has the remarkable property of intercellular trafficking, thus making it a promising tool for improving gene transfer efficiency. METHODS To investigate whether the fusion of VP22 to the cytosine deaminase (CD) suicide gene could enhance the therapeutic efficiency of neural stem cells (NSCs) in the treatment for C6 glioma, the lentiviral vectors pHIV-VP(22)-EGFP, pHIV-CD, and pHIV-VP(22)-CD were constructed based on the pHIV-EGFP vector. After packaging, vectors were transduced into rat NSCs. RESULTS Fluorescence-activated cell sorting analysis revealed that the fusion of VP22-EGFP increased the expression rate of EGFP in NSCs compared with lenti-EGFP transduced cells. Under incubation with the prodrug 5-fluorocytosine (5-FC), the survival rates of C6 cells co-cultured with NSCs/VP(22)-CD (NSCs transduced with lenti-VP(22)-CD) decreased tremendously compared with those of C6 and NSCs/CD. Similar results were also observed in vivo; a significant reduction in tumor volumes in C6 glioma-bearing rats was observed in the NSCs/VP(22)-CD therapy group when compared with other control groups. CONCLUSIONS Our results reveal that VP22 increases the transduction efficiency of lentivirus into NSCs and enhances the therapeutic efficacy of CD-engineered rat NSCs in the treatment for C6 glioma, demonstrating that VP22 might be a useful tool for the gene therapy of engineered NSCs and providing a potential novel strategy for enhancing the effectiveness of gene therapy in other diseases.
Collapse
Affiliation(s)
- Guishan Jin
- Brain Tumor Research Center, Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Tiantan Xili 6, Chongwen District, Beijing 100050, People's Republic of China
| | | | | | | | | | | |
Collapse
|
21
|
Yi BR, Hwang KA, Kang NH, Kim SU, Jeung EB, Kim HC, Choi KC. Synergistic effects of genetically engineered stem cells expressing cytosine deaminase and interferon-β via their tumor tropism to selectively target human hepatocarcinoma cells. Cancer Gene Ther 2012; 19:644-51. [PMID: 22790964 DOI: 10.1038/cgt.2012.45] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Stem cells have received a great deal of attention for their clinical and therapeutic potential for treating human diseases and disorders. Recent studies have shown that it is possible to genetically engineered stem cells (GESTECs) to produce suicide enzymes that convert non-toxic prodrugs to toxic metabolites, selectively migrate toward tumor sites and reduce tumor growth. In this study, we evaluated whether these GESTECs are capable of migrating to hepatocarcinoma cells and examined the potential therapeutic efficacy of gene-directed enzyme prodrug therapy against liver cancer cells in cellular and animal models. A modified transwell migration assay was performed to determine the migratory capacity of GESTECs to Hep3B hepatocarcinoma cells. GESTECs, that is, HB1.F3.CD or HB1.F3.CD.interferon-β (IFN-β) cells, engineered to express a suicide gene, cytosine deaminase (CD), selectively migrated toward liver cancer cells. Treatment of Hep3B, human liver cancer cells, with the prodrug 5-fluorocytosine (5-FC) in the presence of HB1.F3.CD or HB1.F3.CD.IFN-β cells resulted in the inhibition of Hep3B cell growth. In a xenografted mouse model injected with hepatocarcinoma, we investigated the therapeutic effect of these stem cells. For 9 weeks, the xenografted mice were treated with HB1.F3.CD or HB1.F3.CD.IFN-β in the presence of 5-FC. A growth of tumor mass was inhibited about 40-50% in the mice treated with GESTECs and a prodrug. In addition, we further confirmed the cytotoxic effect on tumor cells by histological analysis and migratory effect of therapeutic stem cells. Taken together, GESTECs expressing a fusion gene encoding CD and IFN-β may exert a synergistic antitumor effect on this type of tumor.
Collapse
Affiliation(s)
- B-R Yi
- Laboratory of Veterinary Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
22
|
Kosaka H, Ichikawa T, Kurozumi K, Kambara H, Inoue S, Maruo T, Nakamura K, Hamada H, Date I. Therapeutic effect of suicide gene-transferred mesenchymal stem cells in a rat model of glioma. Cancer Gene Ther 2012; 19:572-8. [DOI: 10.1038/cgt.2012.35] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
23
|
Fei S, Qi X, Kedong S, Guangchun J, Jian L, Wei Q. The antitumor effect of mesenchymal stem cells transduced with a lentiviral vector expressing cytosine deaminase in a rat glioma model. J Cancer Res Clin Oncol 2012; 138:347-57. [PMID: 22139383 DOI: 10.1007/s00432-011-1104-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/21/2011] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Mesenchymal stem cells (MSCs) have been recognized as promising delivery vehicles for gene therapy of gliomas. The purpose of this study was to evaluate the antitumor effect of cytosine deaminase (CD)-expressing MSCs in a rat C6 glioma model. METHODS Lentiviral vectors expressing cytosine deaminase (CD) or enhanced green fluorescent protein (eGFP) were constructed and transduced into rat MSCs to generate MSC-CD/eGFP cells. By intracranially injecting C6 glioma cells (1 × 10(6)) alone or in combination with parental MSCs (1 × 10(6) or 2 × 10(6)) or MSC-CD/eGFP cells (1 × 10(6) or 2 × 10(6)) into rats, we examined the effect of engineered MSCs on tumor growth, tumor cell apoptosis, and rat survival in the presence of 5-fluorocytosine (5-FC). RESULTS MSC-CD/eGFP cells were largely localized at the junction of the tumor with normal tissue. The mean survival time of rats co-injected with C6 glioma cells and MSC-CD/eGFP cells was significantly extended (C6 + MSC-CD/eGFP (1:1), 32.3 days; C6 + MSC-CD/eGFP (1:2), 45.9 days) when compared with rats injected with C6 glioma cells alone (15.3 days) or those co-injected with C6 glioma cells and parental cells (C6 + MSCs (1:1), 16.0 days; C6 + MSCs (1:2), 16.6 days). MSC-CD/eGFP-mediated gene therapy significantly reduced the tumor volume in C6 glioma-bearing rats. On day 14 after cell injection, the reduction in the mean tumor volume in rats co-injected with C6 + MSC-CD/eGFP cells (1:1 and 1:2) was 77.24 and 83.28%, respectively. In addition, MSC-CD/eGFP-mediated gene therapy promoted tumor cell apoptosis in rat C6 gliomas. CONCLUSION Genetically engineered MSCs have good therapeutic efficacy against experimental gliomas in rats.
Collapse
Affiliation(s)
- Song Fei
- Department of Neurosurgery, Second Hospital Affiliated to Dalian Medical University, Dalian, 116027, China.
| | | | | | | | | | | |
Collapse
|
24
|
Torimura T, Ueno T, Taniguchi E, Masuda H, Iwamoto H, Nakamura T, Inoue K, Hashimoto O, Abe M, Koga H, Barresi V, Nakashima E, Yano H, Sata M. Interaction of endothelial progenitor cells expressing cytosine deaminase in tumor tissues and 5-fluorocytosine administration suppresses growth of 5-fluorouracil-sensitive liver cancer in mice. Cancer Sci 2012; 103:542-8. [PMID: 22151662 DOI: 10.1111/j.1349-7006.2011.02182.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The drug delivery system to tumors is a critical factor in upregulating the effect of anticancer drugs and reducing adverse events. Recent studies indicated selective migration of bone marrow-derived endothelial progenitor cells (EPC) into tumor tissues. Cytosine deaminase (CD) transforms nontoxic 5-fluorocytosine (5-FC) into the highly toxic 5-fluorouracil (5-FU). We investigated the antitumor effect of a new CD/5-FC system with CD cDNA transfected EPC for hepatocellular carcinoma (HCC) in mice. We used human hepatoma cell lines (HuH-7, HLF, HAK1-B, KYN-2, KIM-1) and a rat EPC cell line (TR-BME-2). Escherichia coli CD cDNA was transfected into TR-BME-2 (CD-TR-BME). The inhibitory effect of 5-FU on the proliferation of hepatoma cell lines and the inhibitory effect of 5-FU secreted by CD-TR-BME and 5-FC on the proliferation of co-cultured hepatoma cells were evaluated by a tetrazolium-based assay. In mouse subcutaneous xenograft models of KYN-2 and HuH-7, CD-TR-BME was transplanted intravenously followed by 5-FC injection intraperitoneally. HuH-7 cells were the most sensitive to 5-FU and KYN-2 cells were the most resistant. CD-TR-BME secreted 5-FU and inhibited HuH-7 proliferation in a 5-FC dose-dependent manner. CD-TR-BME were recruited into the tumor tissues and some were incorporated into tumor vessels. Tumor growth of HuH-7 was significantly suppressed during 5-FC administration. No bodyweight loss, ALT abnormality or bone marrow suppression was observed. These findings suggest that our new CD/5-FC system with CD cDNA transfected EPC could be an effective and safe treatment for suppression of 5-FU-sensitive HCC growth.
Collapse
Affiliation(s)
- Takuji Torimura
- Liver Cancer Division, Research Center for Innovative Cancer Therapy and Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ramnaraine ML, Mathews WE, Clohisy DR. Lentivirus transduction of human osteoclast precursor cells and differentiation into functional osteoclasts. Bone 2012; 50:97-103. [PMID: 21989297 PMCID: PMC3246560 DOI: 10.1016/j.bone.2011.09.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 08/22/2011] [Accepted: 09/10/2011] [Indexed: 02/06/2023]
Abstract
Gene transfer into stem cells has been an ongoing priority as a treatment for genetic disease and cancer for more than two decades. Methods described herein, form the basis for providing the cell source to determine if osteoclast precursor cells (OcP) can be used as therapeutic gene delivery systems in vivo. Osteoclasts and tumor associated macrophages or OcP, support survival, tumor progression and osteolysis in bone cancers. Two sources of precursor cells are compared: CD14+ cells, the standard OcP, found abundantly in peripheral blood and CD34+ cells, hematopoietic stem cells that are rare, but which can be expanded into OcP. Our findings characterize cell yield at each step of the transduction process and thus provide essential data for planning future in vivo experiments. In addition we demonstrate that essential functions of OcP are preserved following lentiviral transduction. Specifically, neither the transduction method nor the lentiviral transduction influence the OcP's ability to form osteoclasts, express the marker gene, EGFP, or resorb bone. Finally, we conclude that CD34+ cells yield significantly more transduced cells and form functionally superior osteoclasts in vitro. This study represents a step towards considering human gene therapy for bone cancer by demonstrating successful transduction of human OcP for use as cellular delivery vehicles to sites of bone cancer.
Collapse
Affiliation(s)
- Margaret L Ramnaraine
- Department of Orthopedic Surgery, University of Minnesota, 420 Delaware Street SE, MMC 806, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
26
|
Therapeutic effect of genetically modified human neural stem cells encoding cytosine deaminase on experimental glioma. Biochem Biophys Res Commun 2012; 417:534-40. [DOI: 10.1016/j.bbrc.2011.11.155] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 11/30/2011] [Indexed: 11/18/2022]
|
27
|
Binello E, Germano IM. Stem cells as therapeutic vehicles for the treatment of high-grade gliomas. Neuro Oncol 2011; 14:256-65. [PMID: 22166262 DOI: 10.1093/neuonc/nor204] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stem cells have generated great interest in the past decade as potential tools for cell-based treatment of human high-grade gliomas. Thus far, 3 types of stem cells have been tested as vehicles for various therapeutic agents: embryonic, neural, and mesenchymal. The types of therapeutic approaches and/or agents examined in the context of stem cell-based delivery include cytokines, enzyme/prodrug suicide combinations, viral particles, matrix metalloproteinases, and antibodies. Each strategy has specific advantages and disadvantages. Irrespective of the source and/or type of stem cell, there are several areas of concern for their translation to the clinical setting, such as migration in the adult human brain, potential teratogenesis, immune rejection, and regulatory and ethical issues. Nonetheless, a clinical trial is under way using neural stem cell-based delivery of an enzyme/prodrug suicide combination for recurrent high-grade glioma. A proposed future direction could encompass the use of stem cells as vehicles for delivery of agents targeting glioma stem cells, which have been implicated in the resistance of high-grade glioma to treatment. Overall, stem cells are providing an unprecedented opportunity for cell-based approaches in the treatment of high-grade gliomas, which have a persistently dismal prognosis and mandate a continued search for therapeutic options.
Collapse
Affiliation(s)
- Emanuela Binello
- Department of Neurosurgery, Mount Sinai School of Medicine, 1 Gustave L. Levy Place, Box 1136, New York, NY 10029, USA
| | | |
Collapse
|
28
|
Kim YH, Cho SH, Lee SJ, Choi SA, Phi JH, Kim SK, Wang KC, Cho BK, Kim CY. Growth-inhibitory effect of neurotrophin-3-secreting adipose tissue-derived mesenchymal stem cells on the D283-MED human medulloblastoma cell line. J Neurooncol 2011; 106:89-98. [PMID: 21720807 DOI: 10.1007/s11060-011-0656-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Accepted: 06/24/2011] [Indexed: 01/21/2023]
Abstract
Medulloblastoma (MBL), the most common malignant pediatric brain tumor, is incurable in about one-third of patients and can lead to long-term disabilities despite current multimodal treatments. The purpose of this study was to demonstrate in vitro biological effects of neurotrophins-3 (NT-3) on MBL cells and to evaluate the growth-inhibitory effect of neurotrophin-3 (NT-3)-secreting stem cells on tumor cells. We confirmed by western blotting that D283-MED cells express tyrosine kinase C, a specific receptor for NT-3. Analyzing the biological effects of NT-3 on MBL cells, we evaluated autophagy, apoptosis, senescence, and differentiation of tumor cells with NT-3. The NT-3 induced a concentration-dependent increase in apoptosis in the tumor cell line (P < 0.001). The high concentrations of NT-3 increased the expression of class III β-tubulin (P < 0.001) and decreased the expression of Nestin (P < 0.05). NT-3-secreting stem cells were produced by nucleofecting pIRES2.EGFP-NT3 into human adipose tissue-derived mesenchymal stem cells (hAT-MSCs) and their tropic property toward MBL cells was confirmed by migration assay. Double-layered co-culture experiments with the NT-3-secreting hAT-MSCs and D283-MED MBL cells were performed, and NT-3-induced cell death was studied by 3-(4,5-dimethylathiazol-2-yl)-2,5-dephenyl-tetrazolium bromide (MTT) assay. Consequently, the high concentrations of NT-3-secreting hAT-MSCs significantly (P < 0.05) increased the death of D283-MED cells in vitro. The present study demonstrated that both apoptotic cell death and neuronal differentiation of tumor cells were the mechanisms of growth-inhibitory effect of NT-3-secreting hAT-MSCs on MBL cell line.
Collapse
Affiliation(s)
- Young-Hoon Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Roger M, Clavreul A, Venier-Julienne MC, Passirani C, Montero-Menei C, Menei P. The potential of combinations of drug-loaded nanoparticle systems and adult stem cells for glioma therapy. Biomaterials 2011; 32:2106-16. [DOI: 10.1016/j.biomaterials.2010.11.056] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 11/14/2010] [Indexed: 12/16/2022]
|
30
|
|
31
|
Waerzeggers Y, Monfared P, Viel T, Winkeler A, Jacobs AH. Mouse models in neurological disorders: applications of non-invasive imaging. Biochim Biophys Acta Mol Basis Dis 2010; 1802:819-39. [PMID: 20471478 DOI: 10.1016/j.bbadis.2010.04.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 04/26/2010] [Accepted: 04/29/2010] [Indexed: 12/14/2022]
Abstract
Neuroimaging techniques represent powerful tools to assess disease-specific cellular, biochemical and molecular processes non-invasively in vivo. Besides providing precise anatomical localisation and quantification, the most exciting advantage of non-invasive imaging techniques is the opportunity to investigate the spatial and temporal dynamics of disease-specific functional and molecular events longitudinally in intact living organisms, so called molecular imaging (MI). Combining neuroimaging technologies with in vivo models of neurological disorders provides unique opportunities to understand the aetiology and pathophysiology of human neurological disorders. In this way, neuroimaging in mouse models of neurological disorders not only can be used for phenotyping specific diseases and monitoring disease progression but also plays an essential role in the development and evaluation of disease-specific treatment approaches. In this way MI is a key technology in translational research, helping to design improved disease models as well as experimental treatment protocols that may afterwards be implemented into clinical routine. The most widely used imaging modalities in animal models to assess in vivo anatomical, functional and molecular events are positron emission tomography (PET), magnetic resonance imaging (MRI) and optical imaging (OI). Here, we review the application of neuroimaging in mouse models of neurodegeneration (Parkinson's disease, PD, and Alzheimer's disease, AD) and brain cancer (glioma).
Collapse
Affiliation(s)
- Yannic Waerzeggers
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck Institute for Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society and the Faculty of Medicine of the University of Cologne, Cologne, Germany
| | | | | | | | | |
Collapse
|
32
|
Kim KY, Kim SU, Leung PCK, Jeung EB, Choi KC. Influence of the prodrugs 5-fluorocytosine and CPT-11 on ovarian cancer cells using genetically engineered stem cells: tumor-tropic potential and inhibition of ovarian cancer cell growth. Cancer Sci 2010; 101:955-62. [PMID: 20704576 PMCID: PMC11159652 DOI: 10.1111/j.1349-7006.2009.01485.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024] Open
Abstract
Recent studies have shown that genetically engineered stem cells (GESTECs) to produce suicide enzymes that convert non-toxic prodrugs to toxic metabolites selectively migrate toward tumor sites and reduce tumor growth. In the present study, we evaluated whether these GESTECs were capable of migrating to human ovarian cancer cells and examined the potential therapeutic efficacy of the gene-directed enzyme prodrug therapy against ovarian cancer cells in vitro. The expression of cytosine deaminase (CD) or carboxyl esterase (CE) mRNA of GESTECs was confirmed by RT-PCR. A modified transwell migration assay was performed to determine the migratory capacity of GESTECs to ovarian cancer cells. GESTECs (HB1.F3.CD or HB1.F3.CE cells) engineered to express a suicide gene (CD or CE) selectively migrated toward ovarian cancer cells. A [(3)H] thymidine incorporation assay was conducted to measure the proliferative index. Treatment of human epithelial ovarian cancer cell line (SKOV-3, an ovarian adenocarcinoma derived from the ascites of an ovarian cancer patient) with the prodrugs 5-fluorocytosine (5-FC) or camptothecin-11 (CPT-11) in the presence of HB1.F3.CD or HB1.F3.CE cells resulted in the inhibition of ovarian cancer cell growth. Based on the data presented herein, we suggest that GESTECs expressing CD/CE may have a potent advantage to selectively treat ovarian cancers.
Collapse
Affiliation(s)
- Ki-Yon Kim
- Department of Obstetrics and Gynecology, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|
33
|
Kim JH, Lee JE, Kim SU, Cho KG. Stereological Analysis on Migration of Human Neural Stem Cells in the Brain of Rats Bearing Glioma. Neurosurgery 2010; 66:333-42; discussion 342. [DOI: 10.1227/01.neu.0000363720.07070.a8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Jae-Ho Kim
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, Korea (Jae-Ho Kim) (Cho)
| | - Jong-Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea (Lee)
| | - Seung U. Kim
- Medical Research Institute, Chungang University College of Medicine, Seoul, Korea (Seung U. Kim)
| | - Kyung-Gi Cho
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
34
|
Achanta P, Roman NIS, Quiñones-Hinojosa A. Gliomagenesis and the use of neural stem cells in brain tumor treatment. Anticancer Agents Med Chem 2010; 10:121-30. [PMID: 20184546 PMCID: PMC2981502 DOI: 10.2174/187152010790909290] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Accepted: 12/29/2009] [Indexed: 01/08/2023]
Abstract
The role of neural stem cells (NSCs) in both the physiological and pathological processes in the brain has been refined through recent studies within the neuro-oncological field. Alterations in NSC regulatory mechanisms may be fundamental for the development and progression of malignant gliomas. A subpopulation of cells within the tumor known as brain tumor stem cells (BTSCs) have been shown to share key properties with NSCs. The BTSC hypothesis has significantly contributed to a potential understanding as to why brain tumors hold such dismal prognosis. On the other hand, the normal NSCs possess the capacity to migrate extensively towards the tumor bulk as well as to lingering neoplastic regions of the brain. The tropism of NSCs towards brain tumors may provide an additional tool for the treatment of brain cancer. The creation of potential therapies through the use of NSCs has been studied and includes the delivery of gene products to specific locations of the central nervous system selectively targeting malignant brain tumor cells and maximizing the efficiency of their delivery. Here, the proposed mechanisms of how brain tumors emerge, the molecular pathways interrupted in NSC pathogenesis and the most recent preclinical results in the use of NSCs for glioma treatment are reviewed.
Collapse
Affiliation(s)
- Pragathi Achanta
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, CRB II, Room 272, 1550 Orleans Street, Baltimore, MD, 21231, USA
| | - Neda I Sedora Roman
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, CRB II, Room 272, 1550 Orleans Street, Baltimore, MD, 21231, USA
- University of Puerto Rico School of Medicine, Office A-873, PO BOX 365067, San Juan PR 00936-5067
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, CRB II, Room 272, 1550 Orleans Street, Baltimore, MD, 21231, USA
| |
Collapse
|
35
|
Kim KY, Kim SU, Leung PCK, Jeung EB, Choi KC. Influence of the prodrugs 5-fluorocytosine and CPT-11 on ovarian cancer cells using genetically engineered stem cells: tumor-tropic potential and inhibition of ovarian cancer cell growth. Cancer Sci 2010. [DOI: 10.1111/j.1349-7006.2010.01485.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
36
|
Mercapide J, Rappa G, Anzanello F, King J, Fodstad O, Lorico A. Primary gene-engineered neural stem/progenitor cells demonstrate tumor-selective migration and antitumor effects in glioma. Int J Cancer 2010; 126:1206-15. [PMID: 19653275 DOI: 10.1002/ijc.24809] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The prognosis of patients with glioblastoma multiforme (GBM) is generally poor after surgical tumor resection. With the aim of developing new adjuvant therapeutic strategies, we have investigated primary neural stem/progenitor cells (NSPC) in co-cultures with glioma cells, and in a model of gene therapy on aggressively growing malignant glioma. NSPC exhibited tropism towards medium conditioned by glioma cells, and in adherent low-cell density co-culture, were attracted to, and fused with, tumor cells. Similarly, within 24-48 hr of co-culture in suspension, NSPC-tumor hybrids were observed, representing 2-3% of the total cell population. NSPC were then coinjected into mouse brain with GBM cells, employing NSPC expressing cyclophosphamide (CPA)-activating enzyme cytochrome p450 2B6 (CYP2B6), which catalyzes CPA prodrug transformation into membrane diffusible DNA-alkylating metabolites. Upon CPA administration, NSPC containing CYP2B6 elicited substantial impairment of tumor growth. When implanted intracerebrally at a distant site from the tumor, gene-engineered NSPC specifically targeted GBM grafts, after traveling through brain parenchyma, and hindered tumor growth through local activation of CPA. Directed migration of primary NSPC corresponded closely with intracerebral and tumoral pattern of expression of vascular endothelial growth factor, which is a motility factor for NSPC. Overall, these findings indicate that therapeutic gene delivery mediated by primary NSPC is a potentially valid strategy for treatment of high-grade gliomas.
Collapse
Affiliation(s)
- Javier Mercapide
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | | | | | | | | | | |
Collapse
|
37
|
Biological Horizons for Targeting Brain Malignancy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 671:93-104. [DOI: 10.1007/978-1-4419-5819-8_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
38
|
Ito S, Natsume A, Shimato S, Ohno M, Kato T, Chansakul P, Wakabayashi T, Kim SU. Human neural stem cells transduced with IFN-β and cytosine deaminase genes intensify bystander effect in experimental glioma. Cancer Gene Ther 2009; 17:299-306. [DOI: 10.1038/cgt.2009.80] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
39
|
Abstract
Primary malignant brain cancer, one of the most deadly diseases, has a high rate of recurrence after treatment. Studies in the past several years have led to the hypothesis that the root of the recurrence may be brain tumor stem cells (BTSCs), stem-like subpopulation of cells that are responsible for propagating the tumor. Current treatments combining surgery and chemoradiotherapy could not eliminate BTSCs because these cells are highly infiltrative and possess several properties that can reduce the damages caused by radiation or anti-cancer drugs. BTSCs are similar to NSCs in molecular marker expression and multi-lineage differentiation potential. Genetic analyses of Drosophila CNS neoplasia, mouse glioma models, and human glioma tissues have revealed a link between increased NSC self-renewal and brain tumorigenesis. Furthermore, data from various rodent models of malignant brain tumors have provided compelling evidence that multipotent NSCs and lineage-restricted neural progenitor cells (NPCs) could be the cell origin of brain tumors. Thus, the first event of brain tumorigenesis might be the occurrence of oncogenic mutations in the stem cell self-renewal pathway in an NSC or NPC. These mutations convert the NSC or NPC to a BTSC, which then initiates and sustains the growth of the tumor. The self-renewal of BTSCs is controlled by several evolutionarily conserved signaling pathways and requires an intact vascular niche. Targeting these pathways and the vascular niche could be a principle in novel brain tumor therapies aimed to eliminate BTSCs.
Collapse
|
40
|
Kim SU, de Vellis J. Stem cell-based cell therapy in neurological diseases: a review. J Neurosci Res 2009; 87:2183-200. [PMID: 19301431 DOI: 10.1002/jnr.22054] [Citation(s) in RCA: 310] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human neurological disorders such as Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis (ALS), Alzheimer's disease, multiple sclerosis (MS), stroke, and spinal cord injury are caused by a loss of neurons and glial cells in the brain or spinal cord. Cell replacement therapy and gene transfer to the diseased or injured brain have provided the basis for the development of potentially powerful new therapeutic strategies for a broad spectrum of human neurological diseases. However, the paucity of suitable cell types for cell replacement therapy in patients suffering from neurological disorders has hampered the development of this promising therapeutic approach. In recent years, neurons and glial cells have successfully been generated from stem cells such as embryonic stem cells, mesenchymal stem cells, and neural stem cells, and extensive efforts by investigators to develop stem cell-based brain transplantation therapies have been carried out. We review here notable experimental and preclinical studies previously published involving stem cell-based cell and gene therapies for Parkinson's disease, Huntington's disease, ALS, Alzheimer's disease, MS, stroke, spinal cord injury, brain tumor, and lysosomal storage diseases and discuss the future prospects for stem cell therapy of neurological disorders in the clinical setting. There are still many obstacles to be overcome before clinical application of cell therapy in neurological disease patients is adopted: 1) it is still uncertain what kind of stem cells would be an ideal source for cellular grafts, and 2) the mechanism by which transplantation of stem cells leads to an enhanced functional recovery and structural reorganization must to be better understood. Steady and solid progress in stem cell research in both basic and preclinical settings should support the hope for development of stem cell-based cell therapies for neurological diseases.
Collapse
Affiliation(s)
- Seung U Kim
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, British Columbia, Canada.
| | | |
Collapse
|
41
|
Rath P, Shi H, Maruniak JA, Litofsky NS, Maria BL, Kirk MD. Stem cells as vectors to deliver HSV/tk gene therapy for malignant gliomas. Curr Stem Cell Res Ther 2009; 4:44-9. [PMID: 19149629 DOI: 10.2174/157488809787169138] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The prognosis of patients diagnosed with malignant gliomas including glioblastoma multiforme (GBM) is poor and there is an urgent need to develop and translate novel therapies into the clinic. Neural stem cells display remarkable tropism toward GBMs and thus may provide a platform to deliver oncolytic agents to improve survival. First we provide a brief review of clinical trials that have used intra-tumoral herpes simplex virus thymidine kinase (HSV/tk) gene therapy to treat brain tumors. Then, we review recent evidence that neural stem cells can be used to deliver HSV/tk to GBMs in animal models. While previous clinical trials used viruses or non-migratory vector-producing cells to deliver HSV/tk, the latter approaches were not effective in humans, primarily because of satellite tumor cells that escaped surgical resection and survived due to low efficiency delivery of HSV/tk. To enhance delivery of HSV/tk to kill gliomas cells, recent animal studies have focused on the ability of neural stem cells, transduced with HSV/tk, to migrate efficiently and selectively to regions occupied by GBM cells. This approach holds the promise of targeting GBM cells that have infiltrated the brain well beyond the original site of the tumor epicenter.
Collapse
Affiliation(s)
- Prakash Rath
- Division of Biological Sciences, University of Missouri-Columbia, Columbia, MO 65211, USA
| | | | | | | | | | | |
Collapse
|
42
|
Oh MC, Lim DA. Novel treatment strategies for malignant gliomas using neural stem cells. Neurotherapeutics 2009; 6:458-64. [PMID: 19560736 PMCID: PMC5084182 DOI: 10.1016/j.nurt.2009.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Accepted: 05/15/2009] [Indexed: 01/18/2023] Open
Abstract
Recent studies in stem cell biology have refined our understanding of the origin and progression of cancer. Identification and characterization of endogenous neural stem cells (NSCs), especially those in the adult human brain, have inspired new ideas for selectively targeting and destroying malignant gliomas. Gliomas consist of a heterogeneous population of cells, and some of these cells have characteristics of cancer stem cells. These brain tumor stem cells (BTSCs) share certain characteristics with normal NSCs. It is still unclear, however, whether malignant gliomas in human patients originate from these aberrant BTSCs. Nonetheless, the cellular and molecular similarities between BTSCs and normal NSCs suggest a common research landscape underlying both normal and cancer stem cell biology, wherein findings of one field are relevant to the other. Furthermore, the natural tropism of NSCs to gliomas has generated the idea that modified NSCs can deliver modified genes to selectively destroy malignant brain tumor cells, and even BTSCs, while leaving healthy surrounding neurons intact. These studies and others on the basic biology of both BTSCs and NSCs will be crucial to expanding our treatment strategies for malignant gliomas.
Collapse
Affiliation(s)
- Michael C Oh
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California 94143-0112, USA.
| | | |
Collapse
|
43
|
Kosztowski T, Zaidi HA, Quiñones-Hinojosa A. Applications of neural and mesenchymal stem cells in the treatment of gliomas. Expert Rev Anticancer Ther 2009; 9:597-612. [PMID: 19445577 PMCID: PMC2705652 DOI: 10.1586/era.09.22] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In addition to stem cells providing a better understanding about the biology and origins of gliomas, new therapeutic approaches have been developed based on the use of stem cells as delivery vehicles. The unique ability of stem cells to track down tumor cells makes them a very appealing therapeutic modality. This review introduces neural and mesenchymal stem cells, discusses the advances that have been made in the utilization of these stem cells as therapies and in diagnostic imaging (to track the advancement of the stem cells towards the tumor cells), and concludes by addressing various challenges and concerns regarding these therapies.
Collapse
Affiliation(s)
- Thomas Kosztowski
- The Johns Hopkins Hospital, Department of Neurosurgery, Johns Hopkins University, CRB II, 1550 Orleans Street, Room 247, Baltimore, MD 21231, USA Tel.: +1 410 502 2906
| | - Hasan A Zaidi
- The Johns Hopkins Hospital, Department of Neurosurgery, Johns Hopkins University, CRB II, 1550 Orleans Street, Room 247, Baltimore, MD 21231, USA Tel.: +1 410 502 2906
| | - Alfredo Quiñones-Hinojosa
- The Johns Hopkins Hospital, Department of Neurosurgery, Johns Hopkins University, CRB II, 1550 Orleans Street, Room 247, Baltimore, MD 21231, USA Tel.: +1 410 502 2906
| |
Collapse
|
44
|
Germano IM, Binello E. Gene therapy as an adjuvant treatment for malignant gliomas: from bench to bedside. J Neurooncol 2009; 93:79-87. [PMID: 19430884 PMCID: PMC11766529 DOI: 10.1007/s11060-009-9869-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Accepted: 03/16/2009] [Indexed: 01/02/2023]
Abstract
Malignant brain tumors, including high-grade gliomas, are among the most lethal of all cancers. Despite considerable advances, including multi-modality treatments with surgery, radiotherapy, and chemotherapy, the overall prognosis for patients with this disease remains dismal. Currently available treatments necessitate the development of more effective tumor-selective therapies. The use of gene therapy for brain tumor therapy is promising as it can be delivered in situ and selectively targets brain tumor cells while sparing the adjacent normal brain tissue. In this article, we summarize the laboratory and clinical work using viral, cell-based, and synthetic vectors, as well as other strategies focused on potentiate gene delivery. Although tangible results on patients' survival remains to be further documented, significant advances in therapeutic gene transfer strategies have been made. The enthusiasm of this progress needs to be tempered by the realistic assessment of the challenges needed to be overcome. Finally, as the field of gene delivery progresses, advances must be made in identifying genes and proteins key to the treatment of malignant gliomas. Due to the great heterogeneity of malignant glioma cells, only approaches combining different strategies may be ultimately successful in defeating this disease.
Collapse
Affiliation(s)
- Isabelle M Germano
- Department of Neurosurgery, Mt. Sinai School of Medicine, New York, NY 10029, USA.
| | | |
Collapse
|
45
|
Gilad AA, van Laarhoven HWM, McMahon MT, Walczak P, Heerschap A, Neeman M, van Zijl PCM, Bulte JWM. Feasibility of concurrent dual contrast enhancement using CEST contrast agents and superparamagnetic iron oxide particles. Magn Reson Med 2009; 61:970-4. [PMID: 19189296 PMCID: PMC2743130 DOI: 10.1002/mrm.21928] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Accepted: 11/19/2008] [Indexed: 11/06/2022]
Abstract
A major challenge for cellular and molecular MRI is to study interactions between two different cell populations or biological processes. We studied the possibility to simultaneously image contrast agents based on two different MRI contrast mechanisms: chemical exchange saturation transfer (CEST) and enhancement of T2 relaxation. Various amounts of superparamagnetic iron oxide (SPIO) nanoparticles were mixed with a fixed concentration (250 microM) of the CEST agent poly-L-lysine. T2 maps, CEST maps, and frequency-dependent saturation spectra were then measured. Color-coded overlay maps demonstrated the feasibility of concurrent dual contrast enhancement. We found that at concentrations lower than 5 microg(Fe)/mL both contrast agents can be imaged simultaneously. At higher concentrations, the iron-based agent can be used to "shut off" the signal arising from the CEST agent. These initial findings are a first step toward using dual CEST/T2 contrast imaging for studying multiple cellular or molecular targets simultaneously in vivo.
Collapse
Affiliation(s)
- Assaf A Gilad
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Neurosection, Baltimore, Maryland 21231, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Hovakimyan M, Weinreich K, Haas SJP, Cattaneo E, Rolfs A, Wree A. In vitro characterization of embryionic ST14A-cells. Int J Neurosci 2009; 118:1489-501. [PMID: 18853328 DOI: 10.1080/00207450701769257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The embryonic striatal temperature sensitive immortalized ST14A-cell line was characterized in vitro by immunocytochemistry when cultured at 33 degrees C and at nonpermissive temperature of 39 degrees C for up to 14 days. At 33 degrees C in DMEM/10% FCS, cells proliferated, were extensively expressing the neural progenitor cell markers nestin and vimentin contrary to neuronal markers. However, when cultured at 39 degrees C the proliferation was delayed and cells began to increase the expression of neuronal markers, followed by a decrease of nestin and vimentin. In serum-free medium the process of neuronal differentiation became more obvious, indicating the potential to use these cells for experimental restorative therapies.
Collapse
|
47
|
Joo KM, Park IH, Shin JY, Jin J, Kang BG, Kim MH, Lee SJ, Jo MY, Kim SU, Nam DH. Human neural stem cells can target and deliver therapeutic genes to breast cancer brain metastases. Mol Ther 2009; 17:570-5. [PMID: 19127251 DOI: 10.1038/mt.2008.290] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The tumor-tropic properties of neural stem cells (NSCs) led to the development of a novel strategy for delivering therapeutic genes to tumors in the brain. To apply this strategy to the treatment of brain metastases, we made a human NSC line expressing cytosine deaminase (F3.CD), which converts 5-fluorocytosine (5-FC) into 5-fluorouracil, an anticancer agent. In vitro, the F3.CD cells significantly inhibited the growth of tumor cell lines in the presence of the prodrug 5-FC. In vivo, MDA-MB-435 human breast cancer cells were implanted into the brain of immune-deficient mouse stereotactically, and F3.CD cells were injected into the contralateral hemisphere followed by systemic 5-FC administration. The F3.CD cells migrated selectively into the brain metastases located in the opposite hemisphere and resulted in significantly reduced volumes. The F3.CD and 5-FC treatment also decreased both tumor volume and number of tumor mass significantly, when immune-deficient mouse had MDA-MB-435 cells injected into the internal carotid artery and F3.CD cells were transplanted into the contralateral brain hemisphere stereotactically. Taken together, brain transplantation of human NSCs, encoding the suicide enzyme CD, combined with systemic administration of the prodrug 5-FC, is an effective treatment regimen for brain metastases of tumors.
Collapse
Affiliation(s)
- Kyeung Min Joo
- Department of Neurosurgery, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Amos PJ, Shang H, Bailey AM, Taylor A, Katz AJ, Peirce SM. IFATS collection: The role of human adipose-derived stromal cells in inflammatory microvascular remodeling and evidence of a perivascular phenotype. Stem Cells 2008; 26:2682-90. [PMID: 18436860 PMCID: PMC2672107 DOI: 10.1634/stemcells.2008-0030] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A growing body of literature suggests that human adipose-derived stromal cells (hASCs) possess developmental plasticity both in vitro and in vivo, and might represent a viable cell source for therapeutic angiogenesis and tissue engineering. We investigate their phenotypic similarity to perivascular cell types, ability to contribute to in vivo microvascular remodeling, and ability to modulate vascular stability. We evaluated hASC surface expression of vascular and stem/progenitor cell markers in vitro, as well as any effects of platelet-derived growth factor B chain (PDGF-BB) and vascular endothelial growth factor 165 on in vitro hASC migration. To ascertain in vivo behavior of hASCs in an angiogenic environment, hASCs were isolated, expanded in culture, labeled with a fluorescent marker, and injected into adult nude rat mesenteries that were stimulated to undergo microvascular remodeling. Ten, 30, and 60 days after injection, tissues from anesthetized animals were harvested and processed with immunohistochemical techniques to determine hASC quantity, positional fate in relation to microvessels, and expression of endothelial and perivascular cell markers. After 60 days, 29% of hASCs exhibited perivascular morphologies compared with 11% of injected human lung fibroblasts. hASCs exhibiting perivascular morphologies also expressed markers characteristic of vascular pericytes: smooth muscle alpha-actin (10%) and neuron-glia antigen 2 (8%). In tissues treated with hASCs, vascular density was significantly increased over age-matched controls lacking hASCs. This study demonstrates that hASCs express pericyte lineage markers in vivo and in vitro, exhibit increased migration in response to PDGF-BB in vitro, exhibit perivascular morphology when injected in vivo, and contribute to increases in microvascular density during angiogenesis by migrating toward vessels. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Peter J. Amos
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Hulan Shang
- Department of Plastic Surgery, University of Virginia, Charlottesville, VA 22908
| | - Alexander M. Bailey
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Alyssa Taylor
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Adam J. Katz
- Department of Plastic Surgery, University of Virginia, Charlottesville, VA 22908
| | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
49
|
Dietrich J, Imitola J, Kesari S. Mechanisms of Disease: the role of stem cells in the biology and treatment of gliomas. ACTA ACUST UNITED AC 2008; 5:393-404. [PMID: 18521117 DOI: 10.1038/ncponc1132] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Accepted: 11/01/2007] [Indexed: 12/26/2022]
Abstract
The study of neural stem cell and progenitor cell biology has improved our understanding of the biology of brain tumors in a developmental context. Recent work has demonstrated that brain tumors may harbor small subpopulations of cells that share characteristics of neural stem cells. There is still an ongoing debate about the specific role of these stem-like cells in cancer initiation, development and progression. Nonetheless, the concept of cancer stem cells has offered a new paradigm to understand tumor biology and resistance to current treatment modalities. Molecular aberrations in these cancer stem cells might be crucial targets for therapeutic intervention, with the hope of achieving more durable clinical responses. Recent studies have demonstrated that endogenous and transplanted neural stem cells and progenitor cells show a marked tropism to brain tumors. Although the mechanisms that govern these processes are poorly understood, the use of neural stem cells and progenitor cells as delivery vehicles for molecules toxic to tumors offers a promising experimental treatment strategy. This Review summarizes recent advances in the basic understanding of neural stem cell and cancer stem cell biology and the progress towards translating these novel concepts into the clinic.
Collapse
Affiliation(s)
- Jörg Dietrich
- Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
50
|
Aboody KS, Najbauer J, Danks MK. Stem and progenitor cell-mediated tumor selective gene therapy. Gene Ther 2008; 15:739-52. [PMID: 18369324 DOI: 10.1038/gt.2008.41] [Citation(s) in RCA: 215] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The poor prognosis for patients with aggressive or metastatic tumors and the toxic side effects of currently available treatments necessitate the development of more effective tumor-selective therapies. Stem/progenitor cells display inherent tumor-tropic properties that can be exploited for targeted delivery of anticancer genes to invasive and metastatic tumors. Therapeutic genes that have been inserted into stem cells and delivered to tumors with high selectivity include prodrug-activating enzymes (cytosine deaminase, carboxylesterase, thymidine kinase), interleukins (IL-2, IL-4, IL-12, IL-23), interferon-beta, apoptosis-promoting genes (tumor necrosis factor-related apoptosis-inducing ligand) and metalloproteinases (PEX). We and others have demonstrated that neural and mesenchymal stem cells can deliver therapeutic genes to elicit a significant antitumor response in animal models of intracranial glioma, medulloblastoma, melanoma brain metastasis, disseminated neuroblastoma and breast cancer lung metastasis. Most studies reported reduction in tumor volume (up to 90%) and increased survival of tumor-bearing animals. Complete cures have also been achieved (90% disease-free survival for >1 year of mice bearing disseminated neuroblastoma tumors). As we learn more about the biology of stem cells and the molecular mechanisms that mediate their tumor-tropism and we identify efficacious gene products for specific tumor types, the clinical utility of cell-based delivery strategies becomes increasingly evident.
Collapse
Affiliation(s)
- K S Aboody
- Division of Hematology/Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA.
| | | | | |
Collapse
|