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
|
Mercer-Smith AR, Findlay IA, Bomba HN, Hingtgen SD. Intravenously Infused Stem Cells for Cancer Treatment. Stem Cell Rev Rep 2021; 17:2025-2041. [PMID: 34138421 DOI: 10.1007/s12015-021-10192-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2021] [Indexed: 01/14/2023]
Abstract
Despite the recent influx of immunotherapies and small molecule drugs to treat tumors, cancer remains a leading cause of death in the United States, in large part due to the difficulties of treating metastatic cancer. Stem cells, which are inherently tumoritropic, provide a useful drug delivery vehicle to target both primary and metastatic tumors. Intravenous infusions of stem cells carrying or secreting therapeutic payloads show significant promise in the treatment of cancer. Stem cells may be engineered to secrete cytotoxic products, loaded with oncolytic viruses or nanoparticles containing small molecule drugs, or conjugated with immunotherapies. Herein we describe these preclinical and clinical studies, discuss the distribution and migration of stem cells following intravenous infusion, and examine both the limitations of and the methods to improve the migration and therapeutic efficacy of tumoritropic, therapeutic stem cells.
Collapse
Affiliation(s)
- Alison R Mercer-Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA
| | - Ingrid A Findlay
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA
| | - Hunter N Bomba
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA. .,Department of Neurosurgery, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA.
| |
Collapse
|
3
|
Tong Q, Qiu N, Ji J, Ye L, Zhai G. Research Progress in Bioinspired Drug Delivery Systems. Expert Opin Drug Deliv 2020; 17:1269-1288. [PMID: 32543953 DOI: 10.1080/17425247.2020.1783235] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION To tackle challenges associated with traditional drug carriers, investigators have explored cells, cellular membrane, and macromolecular components including proteins and exosomes for the fabrication of delivery vehicles, owing to their excellent biocompatibility, lower toxicity, lower immunogenicity and similarities with the host. Biomacromolecule- and biomimetic nanoparticle (NP)-based drug/gene carriers are drawing immense attention, and biomimetic drug delivery systems (BDDSs) have been conceived and constructed. AREAS COVERED This review focuses on BDDS based on mammalian cells, including blood cells, cancer cells, adult stem cells, endogenous proteins, pathogens and extracellular vesicles (EVs). EXPERT OPINION Compared with traditional drug delivery systems (DDSs), BDDSs are based on biological nanocarriers, exhibiting superior biocompatibility, fewer side effects, natural targeting, and diverse modifications. In addition to directly employing natural biomaterials such as cells, proteins, pathogens and EVs as carriers, BDDSs offer these advantages by mimicking the structure of natural nanocarriers through bioengineering technologies. Furthermore, BDDSs demonstrate fewer limitations and irregularities than natural materials and can overcome several shortcomings associated with natural carriers. Although research remains ongoing to resolve these limitations, it is anticipated that BDDSs possess the potential to overcome challenges associated with traditional DDS, with a promising future in the treatment of human diseases.
Collapse
Affiliation(s)
- Qirong Tong
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Na Qiu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| |
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
|
Choi SS, Yoon K, Choi SA, Yoon SB, Kim SU, Lee HJ. Tumor-specific gene therapy for pancreatic cancer using human neural stem cells encoding carboxylesterase. Oncotarget 2018; 7:75319-75327. [PMID: 27659534 PMCID: PMC5342743 DOI: 10.18632/oncotarget.12173] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 09/13/2016] [Indexed: 01/14/2023] Open
Abstract
Advanced pancreatic cancer is one of the most lethal malignant human diseases lacking effective treatment. Its extremely low survival rate necessitates development of novel therapeutic approach. Human neural stem cells (NSCs) are known to have tumor-tropic effect. We genetically engineered them to express rabbit carboxyl esterase (F3.CE), which activates prodrug CPT-11(irinotecan) into potent metabolite SN-38. We found significant inhibition of the growth of BxPC3 human pancreatic cancer cell line in vitro by F3.CE in presence of CPT-11. Apoptosis was also markedly increased in BxPC3 cells treated with F3.CE and CPT-11. The ligand VEGF and receptor VEGF-1(Flt1) were identified to be the relevant tumor-tropic chemoattractant. We confirmed in vivo that in mice injected with BxPC3 on their skin, there was significant reduction of tumor size in those treated with both F3.CE and BxPC3 adjacent to the cancer mass. Administration of F3.CE in conjunction with CPT-11 could be a new possibility as an effective treatment regimen for patients suffering from advanced pancreatic cancer.
Collapse
Affiliation(s)
- Sung S Choi
- Biomedical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
| | - Kichul Yoon
- Biomedical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea.,Seoul Adventist Hospital, Seoul, Korea.,Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Seon-A Choi
- Futuristic Animal Resource & Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Korea
| | - Seung-Bin Yoon
- Futuristic Animal Resource & Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Korea
| | - Seung U Kim
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Hong J Lee
- Biomedical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
| |
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
|
Parker Kerrigan BC, Shimizu Y, Andreeff M, Lang FF. Mesenchymal stromal cells for the delivery of oncolytic viruses in gliomas. Cytotherapy 2017; 19:445-457. [PMID: 28233640 DOI: 10.1016/j.jcyt.2017.02.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 01/24/2017] [Accepted: 02/06/2017] [Indexed: 02/08/2023]
Abstract
Mesenchymal stromal cells (MSCs) are a type of adult stem cell that has been exploited for the treatment of a variety of diseases, including cancer. In particular, MSCs have been studied extensively for their ability to treat glioblastoma (GBM), the most common and deadly form of brain cancer in adults. MSCs are attractive therapeutics because they can be obtained relatively easily from patients, are capable of being expanded numerically in vitro, can be easily engineered and are inherently capable of homing to tumors, making them ideal vehicles for delivering biological antitumoral agents. Oncolytic viruses are promising biological therapeutic agents that have been used in the treatment of GBMs, and MSCs are currently being explored as a means of delivering these viruses. Here we review the role of MSCs in the treatment of GBMs, focusing on the intersection of MSCs and oncolytic viruses.
Collapse
Affiliation(s)
- Brittany C Parker Kerrigan
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Yuzaburo Shimizu
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Neurosurgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
8
|
Krassikova LS, Karshieva SS, Cheglakov IB, Belyavsky AV. Combined treatment, based on lysomustine administration with mesenchymal stem cells expressing cytosine deaminase therapy, leads to pronounced murine Lewis lung carcinoma growth inhibition. J Gene Med 2016; 18:220-33. [DOI: 10.1002/jgm.2894] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/18/2016] [Accepted: 07/20/2016] [Indexed: 12/22/2022] Open
Affiliation(s)
- Lyudmila S. Krassikova
- Pushchino State Institute of Natural Sciences; Pushchino Russia
- Engelhardt Institute of Molecular Biology RAS; Moscow Russia
| | - Saida S. Karshieva
- Engelhardt Institute of Molecular Biology RAS; Moscow Russia
- N. N. Blokhin Cancer Research Center; Russia
| | - Ivan B. Cheglakov
- Engelhardt Institute of Molecular Biology RAS; Moscow Russia
- N. N. Blokhin Cancer Research Center; Russia
| | | |
Collapse
|
9
|
Abstract
Stem cell-based drug delivery for cancer therapy has steadily gained momentum in the past decade as several studies have reported stem cells' inherent tropism towards tumors. Since this science is still in its early stages and there are many factors that could significantly impact tumor tropism of stem cells, some contradictory results have been observed. This review starts by examining a number of proof-of-concept studies that demonstrate the potential application of stem cells in cancer therapy. Studies that illustrate stem cells' tumor tropism and discuss the technical difficulties that could impact the therapeutic outcome are also highlighted. The discussion also emphasizes stem cell imaging/tracking, as it plays a crucial role in performing reliable dose-response studies and evaluating the therapeutic outcome of treatment protocols. In each section, the pros and cons associated with each method are highlighted, limitations are underlined, and potential solutions are discussed. The overall intention is to familiarize the reader with important practical issues related to stem cell cancer tropism and in vivo tracking, underline the shortcomings, and emphasize critical factors that need to be considered for effective translation of this science into the clinic.
Collapse
|
10
|
Newman WC, Amankulor NA. Targeted Treatment of Experimental Spinal Cord Glioma With Dual Gene-Engineered Human Neural Stem Cells. Neurosurgery 2016; 78:N15-7. [PMID: 27191809 DOI: 10.1227/01.neu.0000484056.27923.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
11
|
Ropper AE, Zeng X, Haragopal H, Anderson JE, Aljuboori Z, Han I, Abd-El-Barr M, Lee HJ, Sidman RL, Snyder EY, Viapiano MS, Kim SU, Chi JH, Teng YD. Targeted Treatment of Experimental Spinal Cord Glioma With Dual Gene-Engineered Human Neural Stem Cells. Neurosurgery 2015; 79:481-91. [DOI: 10.1227/neu.0000000000001174] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Abstract
BACKGROUND
There are currently no satisfactory treatments or experimental models showing autonomic dysfunction for intramedullary spinal cord gliomas (ISCG).
OBJECTIVE
To develop a rat model of ISCG and investigate whether genetically engineered human neural stem cells (F3.hNSCs) could be developed into effective therapies for ISCG.
METHODS
Immunodeficient/Rowett Nude rats received C6 implantation of G55 human glioblastoma cells (10K/each). F3.hNSCs engineered to express either cytosine deaminase gene only (i.e., F3.CD) or dual genes of CD and thymidine kinase (i.e., F3.CD-TK) converted benign 5-fluorocytosine and ganciclovir into oncolytic 5-fluorouracil and ganciclovir-triphosphate, respectively. ISCG rats received injection of F3.CD-TK, F3.CD, or F3.CD-TK debris near the tumor epicenter 7 days after G55 seeding, followed with 5-FC (500 mg/kg/5 mL) and ganciclovir administrations (25 mg/kg/1 mL/day × 5/each repeat, intraperitoneal injection). Per humane standards for animals, loss of weight-bearing stepping in the hindlimb was used to determine post-tumor survival. Also evaluated were autonomic functions and tumor growth rate in vivo.
RESULTS
ISCG rats with F3.CD-TK treatment survived significantly longer (37.5 ± 4.78 days) than those receiving F3.CD (21.5 ± 1.75 days) or F3.CD-TK debris (19.3 ± 0.85 days; n = 4/group; P <.05, median rank test), with significantly improved autonomic function and reduced tumor growth rate. F3.DC-TK cells migrated diffusively into ISCG clusters to mediate oncolytic effect.
CONCLUSION
Dual gene-engineered human neural stem cell regimen markedly prolonged survival in a rat model that emulates somatomotor and autonomic dysfunctions of human cervical ISCG. F3.CD-TK may provide a novel approach to treating clinical ISCG.
Collapse
Affiliation(s)
- Alexander E. Ropper
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Xiang Zeng
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Hariprakash Haragopal
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Jamie E. Anderson
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Zaid Aljuboori
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Inbo Han
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Muhammad Abd-El-Barr
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hong Jun Lee
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
| | - Richard L. Sidman
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
| | - Evan Y. Snyder
- Stem Cell Center, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Mariano S. Viapiano
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Seung U. Kim
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - John H. Chi
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Yang D. Teng
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Department of PM&R, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
12
|
Krasikova LS, Karshieva SS, Cheglakov IB, Belyavsky AV. Mesenchymal stem cells expressing cytosine deaminase inhibit growth of murine melanoma B16F10 in vivo. Mol Biol 2015. [DOI: 10.1134/s0026893315060126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
13
|
Salgado AJ, Sousa JC, Costa BM, Pires AO, Mateus-Pinheiro A, Teixeira FG, Pinto L, Sousa N. Mesenchymal stem cells secretome as a modulator of the neurogenic niche: basic insights and therapeutic opportunities. Front Cell Neurosci 2015. [PMID: 26217178 PMCID: PMC4499760 DOI: 10.3389/fncel.2015.00249] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neural stem cells (NSCs) and mesenchymal stem cells (MSCs) share few characteristics apart from self-renewal and multipotency. In fact, the neurogenic and osteogenic stem cell niches derive from two distinct embryonary structures; while the later originates from the mesoderm, as all the connective tissues do, the first derives from the ectoderm. Therefore, it is highly unlikely that stem cells isolated from one niche could form terminally differentiated cells from the other. Additionally, these two niches are associated to tissues/systems (e.g., bone and central nervous system) that have markedly different needs and display diverse functions within the human body. Nevertheless they do share common features. For instance, the differentiation of both NSCs and MSCs is intimately associated with the bone morphogenetic protein family. Moreover, both NSCs and MSCs secrete a panel of common growth factors, such as nerve growth factor (NGF), glial derived neurotrophic factor (GDNF), and brain derived neurotrophic factor (BDNF), among others. But it is not the features they share but the interaction between them that seem most important, and worth exploring; namely, it has already been shown that there are mutually beneficially effects when these cell types are co-cultured in vitro. In fact the use of MSCs, and their secretome, become a strong candidate to be used as a therapeutic tool for CNS applications, namely by triggering the endogenous proliferation and differentiation of neural progenitors, among other mechanisms. Quite interestingly it was recently revealed that MSCs could be found in the human brain, in the vicinity of capillaries. In the present review we highlight how MSCs and NSCs in the neurogenic niches interact. Furthermore, we propose directions on this field and explore the future therapeutic possibilities that may arise from the combination/interaction of MSCs and NSCs.
Collapse
Affiliation(s)
- Antonio J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho Braga, Portugal ; ICVS/3B's, PT Government Associate Laboratory Braga/Guimarães, Portugal
| | - Joao C Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho Braga, Portugal ; ICVS/3B's, PT Government Associate Laboratory Braga/Guimarães, Portugal
| | - Bruno M Costa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho Braga, Portugal ; ICVS/3B's, PT Government Associate Laboratory Braga/Guimarães, Portugal
| | - Ana O Pires
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho Braga, Portugal ; ICVS/3B's, PT Government Associate Laboratory Braga/Guimarães, Portugal
| | - António Mateus-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho Braga, Portugal ; ICVS/3B's, PT Government Associate Laboratory Braga/Guimarães, Portugal
| | - F G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho Braga, Portugal ; ICVS/3B's, PT Government Associate Laboratory Braga/Guimarães, Portugal
| | - Luisa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho Braga, Portugal ; ICVS/3B's, PT Government Associate Laboratory Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho Braga, Portugal ; ICVS/3B's, PT Government Associate Laboratory Braga/Guimarães, Portugal
| |
Collapse
|
14
|
Kapoor M, Burgess DJ. Targeted Delivery of Nucleic Acid Therapeutics via Nonviral Vectors. ADVANCES IN DELIVERY SCIENCE AND TECHNOLOGY 2015. [DOI: 10.1007/978-3-319-11355-5_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
15
|
Zhu D, Chen C, Purwanti YI, Du S, Lam DH, Wu C, Zeng J, Toh HC, Wang S. Induced Pluripotent Stem Cell-Derived Neural Stem Cells Transduced with Baculovirus Encoding CD40 Ligand for Immunogene Therapy in Mouse Models of Breast Cancer. Hum Gene Ther 2014; 25:747-58. [DOI: 10.1089/hum.2013.160] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Detu Zhu
- Department of Biological Sciences, National University of Singapore, Singapore 117543
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Can Chen
- Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Yovita Ida Purwanti
- Department of Biological Sciences, National University of Singapore, Singapore 117543
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Shouhui Du
- Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Dang Hoang Lam
- Department of Biological Sciences, National University of Singapore, Singapore 117543
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Chunxiao Wu
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Jieming Zeng
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | | | - Shu Wang
- Department of Biological Sciences, National University of Singapore, Singapore 117543
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| |
Collapse
|
16
|
Kim HS, Yi BR, Hwang KA, Kim SU, Choi KC. Anticancer effects of the engineered stem cells transduced with therapeutic genes via a selective tumor tropism caused by vascular endothelial growth factor toward HeLa cervical cancer cells. Mol Cells 2013; 36:347-54. [PMID: 24008363 PMCID: PMC3887992 DOI: 10.1007/s10059-013-0153-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 07/22/2013] [Accepted: 07/23/2013] [Indexed: 10/26/2022] Open
Abstract
The aim of the present study was to investigate the therapeutic efficacy of genetically engineered stem cells (GESTECs) expressing bacterial cytosine deaminase (CD) and/or human interferon-beta (IFN-β) gene against HeLa cervical cancer and the migration factors of the GESTECs toward the cancer cells. Anticancer effect of GESTECs was examined in a co-culture with HeLa cells using MTT assay to measure cell viability. A transwell migration assay was performed so as to assess the migration capability of the stem cells to cervical cancer cells. Next, several chemoattractant ligands and their receptors related to a selective migration of the stem cells toward HeLa cells were determined by real-time PCR. The cell viability of HeLa cells was decreased in response to 5-fluorocytosine (5-FC), a prodrug, indicating that 5-fluorouracil (5-FU), a toxic metabolite, was converted from 5-FC by CD gene and it caused the cell death in a co-culture system. When IFN-β was additionally expressed with CD gene by these GESTECs, the anticancer activity was significantly increased. In the migration assay, the GESTECs selectively migrated to HeLa cervical cancer cells. As results of real-time PCR, chemoattractant ligands such as MCP-1, SCF, and VEGF were expressed in HeLa cells, and several receptors such as uPAR, VEGFR2, and c-kit were produced by the GESTECs. These GESTECs transduced with CD gene and IFN-β may provide a potential of a novel gene therapy for anticervical cancer treatments via their selective tumor tropism derived from VEGF and VEGFR2 expressions between HeLa cells and the GESTECs.
Collapse
Affiliation(s)
- Hye-Sun Kim
- Laboratory of Veterinary Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 361-763, Korea
| | - Bo-Rim Yi
- Laboratory of Veterinary Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 361-763, Korea
| | - Kyung-A Hwang
- Laboratory of Veterinary Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 361-763, Korea
| | - Seung U. Kim
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| | - Kyung-Chul Choi
- Laboratory of Veterinary Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 361-763, Korea
| |
Collapse
|
17
|
Abstract
Development of antitumor preparations with low toxicity and high selectivity of action is one of the top priorities of cancer gene therapy. Mesenchymal stem cells possess natural tropism towards tumors, a property that makes possible their use as a vehicle for targeted delivery of therapeutic genes into tumors of various etiologies. At present, genes encoding enzymes (cytosine deaminase, thymidine kinase, carboxyl esterase), cytokines (IL-2, IL-4, IL-12, IFN-beta) and apoptosis inducing factors (TRAIL) are used as therapeutic genes. Mesenchymal stem cells, as demonstrated using experimental models of tumors of various etiologies as well as animals with metastases in brain and lungs, are able to successfully deliver therapeutic genes into tumors and produce significant antitumor effect. However, to effectively use this therapeutic strategy in clinic, one still has to solve a number of technical problems.
Collapse
|
18
|
Kang W, Seol HJ, Seong DH, Kim J, Kim Y, Kim SU, Nam DH, Joo KM. Adenosine potentiates the therapeutic effects of neural stem cells expressing cytosine deaminase against metastatic brain tumors. Oncol Rep 2013; 30:1101-6. [PMID: 23828015 DOI: 10.3892/or.2013.2584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 01/07/2013] [Indexed: 11/06/2022] Open
Abstract
Tumor-tropic properties of neural stem cells (NSCs) provide a novel approach with which to deliver targeting therapeutic genes to brain tumors. Previously, we developed a therapeutic strategy against metastatic brain tumors using a human NSC line (F3) expressing cytosine deaminase (F3.CD). F3.CD converts systemically administered 5-fluorocytosine (5-FC), a blood-brain barrier permeable nontoxic prodrug, into the anticancer agent 5-fluorouracil (5-FU). In this study, we potentiated a therapeutic strategy of treatment with nucleosides in order to chemically facilitate the endogenous conversion of 5-FU to its toxic metabolite 5-FU ribonucleoside (5-FUR). In vitro, 5-FUR showed superior cytotoxic activity against MDA-MB-435 cancer cells when compared to 5-FU. Although adenosine had little cytotoxic activity, the addition of adenosine significantly potentiated the in vitro cytotoxicity of 5-FU. When MDA-MB‑435 cells were co-cultured with F3.CD cells, F3.CD cells and 5-FC inhibited the growth of MDA-MB-435 cells more significantly in the presence of adenosine. Facilitated 5-FUR production by F3.CD was confirmed by an HPLC analysis of the conditioned media derived from F3.CD cells treated with 5-FC and adenosine. In vivo systemic adenosine treatment also significantly potentiated the therapeutic effects of F3.CD cells and 5-FC in an MDA-MB-435 metastatic brain tumor model. Simple adenosine addition improved the antitumor activity of the NSCs carrying the therapeutic gene. Our results demonstrated an increased therapeutic potential, and thereby, clinical applicability of NSC-based gene therapy.
Collapse
Affiliation(s)
- Wonyoung Kang
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Zhu D, Lam DH, Purwanti YI, Goh SL, Wu C, Zeng J, Fan W, Wang S. Systemic delivery of fusogenic membrane glycoprotein-expressing neural stem cells to selectively kill tumor cells. Mol Ther 2013; 21:1621-30. [PMID: 23752308 DOI: 10.1038/mt.2013.123] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Accepted: 05/10/2013] [Indexed: 12/29/2022] Open
Abstract
Intravenously injected neural stem cells (NSCs) can infiltrate both primary and metastatic tumor sites; thus, they are attractive tumor-targeting vehicles for delivering anticancer agents. However, because the systemic distribution of the injected NSCs involves normal organs and might induce off-target actions leading to unintended side effects, clinical applications of this approach is impeded. Given that the vesicular stomatitis virus glycoprotein (VSV-G) can promote the formation of multinucleated syncytia to kill cells in a pH-dependent manner, we engineered a pH sensor of VSV-G and generated a novel VSV-G mutant that efficiently promotes syncytium formation at the tumor extracellular pH (pHe) but not at pH 7.4. Using transduced NSCs derived from induced pluripotent stem cells (iPSCs), the VSV-G mutant was delivered into mice with metastatic breast cancers in the lung through tail vein injection. Compared with the conventional stem cell-based gene therapy that uses the herpes simplex virus thymidine kinase (HSVtk) suicide gene, this treatment did not display toxicity to normal non-targeted organs while retaining therapeutic effects in tumor-bearing organs. Our findings demonstrate the effectiveness of a new approach for achieving tumor-selective killing effects following systemic stem cell administration. Its potential in stem cell-based gene therapy for metastatic cancer is worthy of further exploration.
Collapse
Affiliation(s)
- Detu Zhu
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Current status of gene therapy for brain tumors. Transl Res 2013; 161:339-54. [PMID: 23246627 PMCID: PMC3733107 DOI: 10.1016/j.trsl.2012.11.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 11/14/2012] [Accepted: 11/16/2012] [Indexed: 01/06/2023]
Abstract
Glioblastoma (GBM) is the most common and deadliest primary brain tumor in adults, with current treatments having limited impact on disease progression. Therefore the development of alternative treatment options is greatly needed. Gene therapy is a treatment strategy that relies on the delivery of genetic material, usually transgenes or viruses, into cells for therapeutic purposes, and has been applied to GBM with increasing promise. We have included selectively replication-competent oncolytic viruses within this strategy, although the virus acts directly as a complex biologic anti-tumor agent rather than as a classic gene delivery vehicle. GBM is a good candidate for gene therapy because tumors remain locally within the brain and only rarely metastasize to other tissues; the majority of cells in the brain are post-mitotic, which allows for specific targeting of dividing tumor cells; and tumors can often be accessed neurosurgically for administration of therapy. Delivery vehicles used for brain tumors include nonreplicating viral vectors, normal adult stem/progenitor cells, and oncolytic viruses. The therapeutic transgenes or viruses are typically cytotoxic or express prodrug activating suicide genes to kill glioma cells, immunostimulatory to induce or amplify anti-tumor immune responses, and/or modify the tumor microenvironment such as blocking angiogenesis. This review describes current preclinical and clinical gene therapy strategies for the treatment of glioma.
Collapse
|
21
|
Bovenberg MSS, Degeling MH, Tannous BA. Advances in stem cell therapy against gliomas. Trends Mol Med 2013; 19:281-91. [PMID: 23537753 DOI: 10.1016/j.molmed.2013.03.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 03/04/2013] [Accepted: 03/04/2013] [Indexed: 12/17/2022]
Abstract
Malignant gliomas are one of the most lethal cancers, and despite extensive research very little progress has been made in improving prognosis. Multimodality treatment combining surgery, radiation, and chemotherapy is the current gold standard, but effective treatment remains difficult due to the invasive nature and high recurrence of gliomas. Stem cell-based therapy using neural, mesenchymal, or hematopoietic stem cells may be an alternative approach because it is tumor selective and allows targeted therapy that spares healthy brain tissue. Stem cells can be used to establish a long-term antitumor response by stimulating the immune system and delivering prodrug, metabolizing genes, or oncolytic viruses. In this review, we discuss current trends and the latest developments in stem cell therapy against malignant gliomas from both the experimental laboratory and the clinic.
Collapse
Affiliation(s)
- M Sarah S Bovenberg
- Experimental Therapeutics and Molecular Imaging Laboratory, Neuroscience Center, Department of Neurology, Massachusetts General Hospital, and Program in Neuroscience, Harvard Medical School, Boston, MA 02129, USA
| | | | | |
Collapse
|
22
|
|