51
|
Schug C, Sievert W, Urnauer S, Müller AM, Schmohl KA, Wechselberger A, Schwenk N, Lauber K, Schwaiger M, Multhoff G, Wagner E, Nelson PJ, Spitzweg C. External Beam Radiation Therapy Enhances Mesenchymal Stem Cell-Mediated Sodium-Iodide Symporter Gene Delivery. Hum Gene Ther 2018; 29:1287-1300. [PMID: 29724129 DOI: 10.1089/hum.2018.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The tumor-homing properties of mesenchymal stem cells (MSC) have led to their development as delivery vehicles for the targeted delivery of therapeutic genes such as the sodium-iodide symporter (NIS) to solid tumors. External beam radiation therapy may represent an ideal setting for the application of engineered MSC-based gene therapy, as tumor irradiation may enhance MSC recruitment into irradiated tumors through the increased production of select factors linked to MSC migration. In the present study, the irradiation of human liver cancer cells (HuH7; 1-10 Gy) showed a strong dose-dependent increase in steady-state mRNA levels of CXCL8, CXCL12, FGF2, PDGFB, TGFB1, THBS1, and VEGF (0-48 h), which was verified for most factors at the protein level (after 48 h). Radiation effects on directed MSC migration were tested in vitro using a live cell tracking migration assay and supernatants from control and irradiated HuH7 cells. A robust increase in mean forward migration index, mean center of mass, and mean directionality of MSCs toward supernatants was seen from irradiated as compared to non-irradiated tumor cells. Transferability of this effect to other tumor sources was demonstrated using the human breast adenocarcinoma cell line (MDA-MB-231), which showed a similar behavior to radiation as seen with HuH7 cells in quantitative polymerase chain reaction and migration assay. To evaluate this in a more physiologic in vivo setting, subcutaneously growing HuH7 xenograft tumors were irradiated with 0, 2, or 5 Gy followed by CMV-NIS-MSC application 24 h later. Tumoral iodide uptake was monitored using 123I-scintigraphy. The results showed increased tumor-specific dose-dependent accumulation of radioiodide in irradiated tumors. The results demonstrate that external beam radiation therapy enhances the migratory capacity of MSCs and may thus increase the therapeutic efficacy of MSC-mediated NIS radionuclide therapy.
Collapse
Affiliation(s)
- Christina Schug
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Wolfgang Sievert
- 2 Department of Radiation Oncology, Technische Universitaet Muenchen , Munich, Germany
| | - Sarah Urnauer
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Andrea M Müller
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Kathrin A Schmohl
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Alexandra Wechselberger
- 3 Clinical Biochemistry Group, Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Nathalie Schwenk
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Kirsten Lauber
- 4 Department of Radiation Oncology, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Markus Schwaiger
- 5 Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universitaet Muenchen , Munich, Germany
| | - Gabriele Multhoff
- 2 Department of Radiation Oncology, Technische Universitaet Muenchen , Munich, Germany
| | - Ernst Wagner
- 6 Department of Pharmacy, Center of Drug Research, Pharmaceutical Biotechnology, LMU Munich, Munich, Germany
| | - Peter J Nelson
- 3 Clinical Biochemistry Group, Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Christine Spitzweg
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| |
Collapse
|
52
|
Müller AM, Schmohl KA, Knoop K, Schug C, Urnauer S, Hagenhoff A, Clevert DA, Ingrisch M, Niess H, Carlsen J, Zach C, Wagner E, Bartenstein P, Nelson PJ, Spitzweg C. Hypoxia-targeted 131I therapy of hepatocellular cancer after systemic mesenchymal stem cell-mediated sodium iodide symporter gene delivery. Oncotarget 2018; 7:54795-54810. [PMID: 27458162 PMCID: PMC5342382 DOI: 10.18632/oncotarget.10758] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/10/2016] [Indexed: 12/18/2022] Open
Abstract
Adoptively transferred mesenchymal stem cells (MSCs) home to solid tumors. Biologic features within the tumor environment can be used to selectively activate transgenes in engineered MSCs after tumor invasion. One of the characteristic features of solid tumors is hypoxia. We evaluated a hypoxia-based imaging and therapy strategy to target expression of the sodium iodide symporter (NIS) gene to experimental hepatocellular carcinoma (HCC) delivered by MSCs. MSCs engineered to express transgenes driven by a hypoxia-responsive promoter showed robust transgene induction under hypoxia as demonstrated by mCherry expression in tumor cell spheroid models, or radioiodide uptake using NIS. Subcutaneous and orthotopic HCC xenograft mouse models revealed significant levels of perchlorate-sensitive NIS-mediated tumoral radioiodide accumulation by tumor-recruited MSCs using 123I-scintigraphy or 124I-positron emission tomography. Functional NIS expression was further confirmed by ex vivo123I-biodistribution analysis. Administration of a therapeutic dose of 131I in mice treated with NIS-transfected MSCs resulted in delayed tumor growth and reduced tumor perfusion, as shown by contrast-enhanced sonography, and significantly prolonged survival of mice bearing orthotopic HCC tumors. Interestingly, radioiodide uptake into subcutaneous tumors was not sufficient to induce therapeutic effects. Our results demonstrate the potential of using tumor hypoxia-based approaches to drive radioiodide therapy in non-thyroidal tumors.
Collapse
Affiliation(s)
- Andrea M Müller
- Department of Internal Medicine II, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Kathrin A Schmohl
- Department of Internal Medicine II, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Kerstin Knoop
- Department of Internal Medicine II, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Christina Schug
- Department of Internal Medicine II, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Sarah Urnauer
- Department of Internal Medicine II, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Anna Hagenhoff
- Clinical Biochemistry Group, Medizinische Klinik und Poliklinik IV, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Dirk-André Clevert
- Department of Clinical Radiology, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Michael Ingrisch
- Department of Clinical Radiology, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Hanno Niess
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Janette Carlsen
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Christian Zach
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Ernst Wagner
- Department of Pharmacy, Center of Drug Research, Pharmaceutical Biotechnology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Peter J Nelson
- Clinical Biochemistry Group, Medizinische Klinik und Poliklinik IV, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Christine Spitzweg
- Department of Internal Medicine II, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| |
Collapse
|
53
|
Marofi F, Vahedi G, Biglari A, Esmaeilzadeh A, Athari SS. Mesenchymal Stromal/Stem Cells: A New Era in the Cell-Based Targeted Gene Therapy of Cancer. Front Immunol 2017; 8:1770. [PMID: 29326689 PMCID: PMC5741703 DOI: 10.3389/fimmu.2017.01770] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
Abstract
In recent years, in light of the promising potentials of mesenchymal stromal/stem cells (MSCs) for carrying therapeutic anticancer genes, a complete revisitation on old chemotherapy-based paradigms has been established. This review attempted to bring forward and introduce the novel therapeutic opportunities of using genetically engineered MSCs. The simplicities and advantages of MSCs for medical applications make them a unique and promising option in the case of cancer therapy. Some of the superiorities of using MSCs as therapeutic gene micro-carriers are the easy cell-extraction procedures and their abundant proliferation capacity in vitro without losing their main biological properties. Targeted therapy by using MSCs as the delivery vehicles of therapeutic genes is a new approach in the treatment of various types of cancers. Some of the distinct properties of MSCs, such as tumor-tropism, non-immunogenicity, stimulatory effect on the anti-inflammatory molecules, inhibitory effect on inflammatory responses, non-toxicity against the normal tissues, and easy processes for the clinical use, have formed the basis of attention to MSCs. They can be easily used for the treatment of damaged or injured tissues, regenerative medicine, and immune disorders. This review focused on the drugability of MSCs and their potential for the delivery of candidate anticancer genes. It also briefly reviewed the vectors and methods used for MSC-mediated gene therapy of malignancies. Also, the challenges, limitations, and considerations in using MSCs for gene therapy of cancer and the new methods developed for resolution of these problems are reviewed.
Collapse
Affiliation(s)
- Faroogh Marofi
- Department of Hematology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghasem Vahedi
- Research Center for Food Hygiene and Safety, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alireza Biglari
- Department of Genetics and Molecular Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | | |
Collapse
|
54
|
The future of mesenchymal stem cell-based therapeutic approaches for cancer - From cells to ghosts. Cancer Lett 2017; 414:239-249. [PMID: 29175461 DOI: 10.1016/j.canlet.2017.11.025] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/19/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells which can differentiate into a variety of cell types including osteoblasts, adipocytes and chondrocytes. They are normally resident in adipose tissue, bone marrow and the umbilical cord, but can also be found in other tissues and are known to be recruited to sites of wound healing as well as growing tumours. The therapeutic potential of MSCs has been explored in a number of phase I/II and III clinical trials, of which several were targeted against graft-versus-host disease and to support engraftment of haematopoietic stem cells (HSCs), but currently only very few in the oncology field. There are now three clinical trials either ongoing or recruiting patients that use MSCs to treat tumour disease. In these, MSCs target gastrointestinal, lung and ovarian cancer, respectively. The first study uses MSCs loaded with a HSV-TK expression construct under the control of the CCL5 promoter, and has recently reported successful completion of Phase I/II. While no adverse side effects were seen during this study, no outcomes with respect to therapeutic benefits have been published. The other clinical trials targeting lung and ovarian cancer will be using MSCs expressing cytokines as therapeutic payload. Despite these encouraging early steps towards their clinical use, many questions are still unanswered regarding the biology of MSCs in normal and pathophysiological settings. In this review, in addition to summarising the current state of MSC-based therapeutic approaches for cancer, we will describe the remaining questions, obstacles and risks, as well as novel developments such as MSC-derived nanoghosts.
Collapse
|
55
|
Oloyo AK, Ambele MA, Pepper MS. Contrasting Views on the Role of Mesenchymal Stromal/Stem Cells in Tumour Growth: A Systematic Review of Experimental Design. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1083:103-124. [DOI: 10.1007/5584_2017_118] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
56
|
von Einem JC, Peter S, Günther C, Volk HD, Grütz G, Salat C, Stoetzer O, Nelson PJ, Michl M, Modest DP, Holch JW, Angele M, Bruns C, Niess H, Heinemann V. Treatment of advanced gastrointestinal cancer with genetically modified autologous mesenchymal stem cells - TREAT-ME-1 - a phase I, first in human, first in class trial. Oncotarget 2017; 8:80156-80166. [PMID: 29113291 PMCID: PMC5655186 DOI: 10.18632/oncotarget.20964] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/26/2017] [Indexed: 12/22/2022] Open
Abstract
PURPOSE This phase I, first in human, first in class clinical study aimed at evaluating the safety, tolerability and efficacy of treatment with genetically modified mesenchymal stromal cells (MSC) in combination with ganciclovir (GCV). MSC_apceth_101 are genetically modified autologous MSCs used as vehicles for a cell-based gene therapy in patients with advanced gastrointestinal adenocarcinoma. EXPERIMENTAL DESIGN The study design consisted of a dose-escalation 3 + 3 design. All patients (n = 6) were treated with up to three applications of MSC_apceth_101, followed by GCV infusions given on three consecutive days starting 48 hours after injection of MSC_apceth_101. Three of six patients received a total dose of 1.5 × 106 cells/kg. Two patients received three doses of 1 × 106 cells/kg, while one patient received only two doses of 1 × 106 cells/kg due to a SADR. RESULTS Six patients received MSC_apceth_101. No IMP-related serious adverse events occurred. Adverse-events related to IMP-injection were increased creatinine, cough, fever, and night sweat. TNF, IL-6, IL-8, IL-10 and sE-Selectin, showed that repeated application is immunologically safe, but induces a switch of the functional properties of monocytes to an inflammatory phenotype. Treatment induced stable disease in 4/6 patients, and progressive disease in 2/6 patients. CONCLUSION Treatment with MSC_apceth_101 in combination with GCV demonstrated acceptable safety and tolerability in patients with advanced gastrointestinal adenocarcinoma.
Collapse
Affiliation(s)
- Jobst C. von Einem
- Department of Medical Oncology and Comprehensive Cancer Center, University Hospital Grosshadern, LMU, Munich, Germany
| | | | | | - Hans-Dieter Volk
- Institute for Medical Immunology and Berlin-Brandenburg Center for Regenerative Medicine, Charité-Universitätsmedizin, Berlin, Germany
| | - Gerald Grütz
- Institute for Medical Immunology and Berlin-Brandenburg Center for Regenerative Medicine, Charité-Universitätsmedizin, Berlin, Germany
| | - Christoph Salat
- Haemato-Onkologische Schwerpunktpraxis Prof. Salat, Dr. Stoetzer, Munich, Germany
| | - Oliver Stoetzer
- Haemato-Onkologische Schwerpunktpraxis Prof. Salat, Dr. Stoetzer, Munich, Germany
| | - Peter J. Nelson
- Department of Medicine IV, University Hospital of Munich, LMU, Munich, Germany
| | - Marlies Michl
- Department of Medical Oncology and Comprehensive Cancer Center, University Hospital Grosshadern, LMU, Munich, Germany
| | - Dominik P. Modest
- Department of Medical Oncology and Comprehensive Cancer Center, University Hospital Grosshadern, LMU, Munich, Germany
| | - Julian W. Holch
- Department of Medical Oncology and Comprehensive Cancer Center, University Hospital Grosshadern, LMU, Munich, Germany
| | - Martin Angele
- Department of Surgery, University Hospital Grosshadern, LMU, Munich, Germany
| | - Christiane Bruns
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Hanno Niess
- Department of Surgery, University Hospital Grosshadern, LMU, Munich, Germany
| | - Volker Heinemann
- Department of Medical Oncology and Comprehensive Cancer Center, University Hospital Grosshadern, LMU, Munich, Germany
| |
Collapse
|
57
|
Xiang BY, Chen L, Wang XJ, Xiang C. Mesenchymal stem cells as therapeutic agents and in gene delivery for the treatment of glioma *. J Zhejiang Univ Sci B 2017; 18:737-746. [PMCID: PMC5611545 DOI: 10.1631/jzus.b1600337] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/22/2016] [Indexed: 06/13/2024]
Abstract
Mesenchymal stem cells (MSCs) are plastic-adherent cells with a characteristic surface phenotype and properties of self-renewal, differentiation, and high proliferative potential. The characteristics of MSCs and their tumor-tropic capability make them an ideal tool for use in cell-based therapies for cancer, including glioma. These cells can function either through a bystander effect or as a delivery system for genes and drugs. MSCs have been demonstrated to inhibit the growth of glioma and to improve survival following transplantation into the brain. We briefly review the current data regarding the use of MSCs in the treatment of glioma and discuss the potential strategies for development of a more specific and effective therapy.
Collapse
|
58
|
Moradian Tehrani R, Verdi J, Noureddini M, Salehi R, Salarinia R, Mosalaei M, Simonian M, Alani B, Ghiasi MR, Jaafari MR, Mirzaei HR, Mirzaei H. Mesenchymal stem cells: A new platform for targeting suicide genes in cancer. J Cell Physiol 2017; 233:3831-3845. [DOI: 10.1002/jcp.26094] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/11/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Rana Moradian Tehrani
- Department of Applied Cell SciencesSchool of Medicine, Kashan University of Medical SciencesKashanIran
| | - Javad Verdi
- Department of Applied Cell SciencesSchool of Medicine, Kashan University of Medical SciencesKashanIran
- Department of Applied Cell Sciences School of Advanced Technologies in Medicine, Tehran University of Medical SciencesTehranIran
| | - Mahdi Noureddini
- Department of Applied Cell SciencesSchool of Medicine, Kashan University of Medical SciencesKashanIran
| | - Rasoul Salehi
- Department of Genetic and Molecular BiologyIsfahan University of Medical SciencesIsfahanIran
| | - Reza Salarinia
- Department of Medical Biotechnology and Molecular SciencesSchool of MedicineNorth Khorasan University of Medical SciencesBojnurdIran
| | - Meysam Mosalaei
- Department of Genetic and Molecular BiologyIsfahan University of Medical SciencesIsfahanIran
| | - Miganosh Simonian
- Department of Genetic and Molecular BiologyIsfahan University of Medical SciencesIsfahanIran
| | - Behrang Alani
- Department of Applied Cell SciencesSchool of Medicine, Kashan University of Medical SciencesKashanIran
| | - Moosa Rahimi Ghiasi
- Department of Genetic and Molecular BiologyIsfahan University of Medical SciencesIsfahanIran
| | - Mahmoud Reza Jaafari
- School of PharmacyNanotechnology Research CenterMashhad University of Medical SciencesMashhadIran
| | - Hamed Reza Mirzaei
- Department of Clinical Laboratory SciencesSchool of Allied Medical SciencesKashan University of Medical SciencesKashanIran
- Department of Immunology, School of MedicineTehran University of Medical SciencesTehranIran
- Clinical Research DivisionFred Hutchinson Cancer Research CenterSeattleWashington
| | - Hamed Mirzaei
- Department of Medical Biotechnology, School of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
59
|
Lee HY, Hong IS. Double-edged sword of mesenchymal stem cells: Cancer-promoting versus therapeutic potential. Cancer Sci 2017; 108:1939-1946. [PMID: 28756624 PMCID: PMC5623746 DOI: 10.1111/cas.13334] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/18/2017] [Accepted: 07/22/2017] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) derived from adipose tissue, bone marrow, cord blood, and other tissues, have recently attracted much attention as potential therapeutic agents in various diseases because of their trans‐differentiation capacity. However, recent studies have suggested that MSCs also appear to contribute to tumor pathogenesis by supporting tumor microenvironments, increasing tumor growth, and eliciting antitumor immune responses. Although some studies suggest that MSCs have inhibitory effects on tumor development, they are overwhelmed by a number of studies showing that MSCs exert stimulatory effects on tumor pathogenesis. In the present review, we summarize a number of findings to provide current information about the therapeutic potential of MSCs in various diseases. We then discuss the potential roles of MSCs in tumor progression.
Collapse
Affiliation(s)
- Hwa-Yong Lee
- The Faculty of Liberal Arts, Jungwon University, Chungbuk, Korea
| | - In-Sun Hong
- Laboratory of Stem Cell Research, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, Korea
| |
Collapse
|
60
|
Kucerova L, Durinikova E, Toro L, Cihova M, Miklikova S, Poturnajova M, Kozovska Z, Matuskova M. Targeted antitumor therapy mediated by prodrug-activating mesenchymal stromal cells. Cancer Lett 2017; 408:1-9. [PMID: 28838843 DOI: 10.1016/j.canlet.2017.08.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/09/2017] [Accepted: 08/11/2017] [Indexed: 12/14/2022]
Abstract
Mesenchymal stromal cells (MSCs) were introduced as tumor-targeted vehicles suitable for delivery of the gene-directed enzyme/prodrug therapy more than 10 years ago. Over these years key properties of tumor cells and MSCs, which are crucial for the treatment efficiency, were examined; and there are some critical issues to be considered for the maximum antitumor effect. Moreover, engineered MSCs expressing enzymes capable of activating non-toxic prodrugs achieved long-term curative effect even in metastatic and hard-to-treat tumor types in pre-clinical scenario(s). These gene-modified MSCs are termed prodrug-activating MSCs throughout the text and represent promising approach for further clinical application. This review summarizes major determinants to be considered for the application of the prodrug-activating MSCs in antitumor therapy in order to maximize therapeutic efficiency.
Collapse
Affiliation(s)
- Lucia Kucerova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia.
| | - Erika Durinikova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Lenka Toro
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Marina Cihova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Svetlana Miklikova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Martina Poturnajova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Zuzana Kozovska
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Miroslava Matuskova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| |
Collapse
|
61
|
Feng M, Xiong G, Cao Z, Yang G, Zheng S, Song X, You L, Zheng L, Zhang T, Zhao Y. PD-1/PD-L1 and immunotherapy for pancreatic cancer. Cancer Lett 2017; 407:57-65. [PMID: 28826722 DOI: 10.1016/j.canlet.2017.08.006] [Citation(s) in RCA: 242] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/17/2017] [Accepted: 08/05/2017] [Indexed: 12/16/2022]
Abstract
Therapy that targets programmed death 1 or programmed death 1 ligand 1 (PD-1/PD-L1), which are known as immune checkpoints, has been recently rapidly developing as oncotherapy for various carcinomas. However, this therapy has a poor effect on the treatment of pancreatic cancer with PD-1/PD-L1 blockade monotherapy. In this review, the development and limitations of anti-PD-1/PD-L1 monotherapy in pancreatic cancer are discussed. We then consider the underlying mechanism of anti-PD-1/PD-L1 monotherapy failure, combination strategies overcoming resistance to anti-PD-1/PD-L1 immunotherapy and the prospect of targeting PD-1/PD-L1 for the immunotherapy of pancreatic cancer.
Collapse
Affiliation(s)
- Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Guangbing Xiong
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Suli Zheng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Xujun Song
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Clinical Immunology Center, Chinese Academy of Medical Science, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
62
|
Sage EK, Thakrar RM, Janes SM. Genetically modified mesenchymal stromal cells in cancer therapy. Cytotherapy 2017; 18:1435-1445. [PMID: 27745603 PMCID: PMC5082580 DOI: 10.1016/j.jcyt.2016.09.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 08/31/2016] [Accepted: 09/02/2016] [Indexed: 12/14/2022]
Abstract
The cell therapy industry has grown rapidly over the past 3 decades, and multiple clinical trials have been performed to date covering a wide range of diseases. The most frequently used cell is mesenchymal stromal cells (MSCs), which have been used largely for their anti-inflammatory actions and in situations of tissue repair and although they have demonstrated a good safety profile, their therapeutic efficacy has been limited. In addition to these characteristics MSCs are being used for their homing and engraftment properties and have been genetically modified to enable targeted delivery of a variety of therapeutic agents in both malignant and nonmalignant conditions. This review discusses the science and technology behind genetically modified MSC therapy in malignant disease and how potential problems have been overcome to enable their use in two novel clinical trials in metastatic gastrointestinal and lung cancer.
Collapse
Affiliation(s)
- Elizabeth K Sage
- Lungs for Living Research Centre, UCL Respiratory, Rayne Institute, University College London, London, United Kingdom
| | - Ricky M Thakrar
- Lungs for Living Research Centre, UCL Respiratory, Rayne Institute, University College London, London, United Kingdom; Department of Thoracic Medicine, University College London Hospital, London, United Kingdom
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, Rayne Institute, University College London, London, United Kingdom; Department of Thoracic Medicine, University College London Hospital, London, United Kingdom.
| |
Collapse
|
63
|
Role of Mesenchymal Stem Cells in Cancer Development and Their Use in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1083:45-62. [DOI: 10.1007/5584_2017_64] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
64
|
Amara I, Pramil E, Senamaud-Beaufort C, Devillers A, Macedo R, Lescaille G, Seguin J, Tartour E, Lemoine FM, Beaune P, de Waziers I. Engineered mesenchymal stem cells as vectors in a suicide gene therapy against preclinical murine models for solid tumors. J Control Release 2016; 239:82-91. [DOI: 10.1016/j.jconrel.2016.08.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/18/2016] [Accepted: 08/20/2016] [Indexed: 01/09/2023]
|
65
|
Niess H, Thomas MN, Schiergens TS, Kleespies A, Jauch KW, Bruns C, Werner J, Nelson PJ, Angele MK. Genetic engineering of mesenchymal stromal cells for cancer therapy: turning partners in crime into Trojan horses. Innov Surg Sci 2016; 1:19-32. [PMID: 31579715 PMCID: PMC6753982 DOI: 10.1515/iss-2016-0005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/03/2016] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are adult progenitor cells with a high migratory and differentiation potential, which influence a broad range of biological functions in almost every tissue of the body. Among other mechanisms, MSCs do so by the secretion of molecular cues, differentiation toward more specialized cell types, or influence on the immune system. Expanding tumors also depend on the contribution of MSCs to building a supporting stroma, but the effects of MSCs appear to go beyond the mere supply of connective tissues. MSCs show targeted "homing" toward growing tumors, which is then followed by exerting direct and indirect effects on cancer cells. Several research groups have developed novel strategies that make use of the tumor tropism of MSCs by engineering them to express a transgene that enables an attack on cancer growth. This review aims to familiarize the reader with the current knowledge about MSC biology, the existing evidence for MSC contribution to tumor growth with its underlying mechanisms, and the strategies that have been developed using MSCs to deploy an anticancer therapy.
Collapse
Affiliation(s)
- Hanno Niess
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Michael N Thomas
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Tobias S Schiergens
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Axel Kleespies
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Karl-Walter Jauch
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Christiane Bruns
- Department of General, Visceral and Cancer Surgery, Hospital of the University of Cologne, Cologne, Germany
| | - Jens Werner
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Peter J Nelson
- Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Universitaet Muenchen, Arbeitsgruppe Klinische Biochemie, Munich, Germany
| | - Martin K Angele
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| |
Collapse
|
66
|
Nowakowski A, Drela K, Rozycka J, Janowski M, Lukomska B. Engineered Mesenchymal Stem Cells as an Anti-Cancer Trojan Horse. Stem Cells Dev 2016; 25:1513-1531. [PMID: 27460260 DOI: 10.1089/scd.2016.0120] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cell-based gene therapy holds a great promise for the treatment of human malignancy. Among different cells, mesenchymal stem cells (MSCs) are emerging as valuable anti-cancer agents that have the potential to be used to treat a number of different cancer types. They have inherent migratory properties, which allow them to serve as vehicles for delivering effective therapy to isolated tumors and metastases. MSCs have been engineered to express anti-proliferative, pro-apoptotic, and anti-angiogenic agents that specifically target different cancers. Another field of interest is to modify MSCs with the cytokines that activate pro-tumorigenic immunity or to use them as carriers for the traditional chemical compounds that possess the properties of anti-cancer drugs. Although there is still controversy about the exact function of MSCs in the tumor settings, the encouraging results from the preclinical studies of MSC-based gene therapy for a large number of tumors support the initiation of clinical trials.
Collapse
Affiliation(s)
- Adam Nowakowski
- 1 NeuroRepair Department, Mossakowski Medical Research Centre , Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Drela
- 1 NeuroRepair Department, Mossakowski Medical Research Centre , Polish Academy of Sciences, Warsaw, Poland
| | - Justyna Rozycka
- 1 NeuroRepair Department, Mossakowski Medical Research Centre , Polish Academy of Sciences, Warsaw, Poland
| | - Miroslaw Janowski
- 1 NeuroRepair Department, Mossakowski Medical Research Centre , Polish Academy of Sciences, Warsaw, Poland .,2 Division of MR Research, Russel H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Barbara Lukomska
- 1 NeuroRepair Department, Mossakowski Medical Research Centre , Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
67
|
Wu J, Liu Y, Tang Y, Wang S, Wang C, Li Y, Su X, Tian J, Tian Y, Pan J, Su Y, Zhu H, Teng Z, Lu G. Synergistic Chemo-Photothermal Therapy of Breast Cancer by Mesenchymal Stem Cell-Encapsulated Yolk-Shell GNR@HPMO-PTX Nanospheres. ACS APPLIED MATERIALS & INTERFACES 2016; 8:17927-17935. [PMID: 27356586 DOI: 10.1021/acsami.6b05677] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mesenchymal stem cells (MSCs) have attracted increasing attention as vehicles for cancer treatment. Herein, MSC-based synergistic oncotherapy strategy is presented for the first time. To achieve this goal, yolk-shell structured gold nanorod embedded hollow periodic mesoporous organosilica nanospheres (GNR@HPMOs) with high paclitaxel (PTX) loading capability and excellent photothermal transfer ability upon near-infrared (NIR) light irradiation are first prepared. Cytotoxicity and migration assays show that the viability and tumor-homing capability of MSCs are well-retained after internalization of high content of PTX loaded GNR@HPMOs (denoted as GNR@HPMOs-PTX). In vitro experiments show the GNR@HPMOs-PTX loaded MSCs (GNR@HPMOs-PTX@MSCs) possess synergistic chemo-photothermal killing effects for breast cancer cells. Also, photoacoustic imaging shows that the MSCs can improve dispersion and distribution in tumor tissue for GNR@HPMOs-PTX after intratumoral injection. In vivo experiments in breast cancer model of nude mice further demonstrate that the GNR@HPMOs-PTX@MSCs significantly inhibit tumor growth, suggesting their great potential for synergistic therapy of cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaodan Su
- Key Laboratory for Organic Electronics & Information Displays and Institute of Advanced Materials, Nanjing University of Posts and Telecommunications , Nanjing 210046, P. R. China
| | - Jihong Tian
- Department of Radiotherapy, the Second Affiliated Hospital of Nanjing Medical University , Nanjing 210011, P. R. China
| | | | | | | | | | - Zhaogang Teng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University , Nanjing 210093, P. R. China
| | - Guangming Lu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University , Nanjing 210093, P. R. China
| |
Collapse
|
68
|
Błogowski W, Bodnarczuk T, Starzyńska T. Concise Review: Pancreatic Cancer and Bone Marrow-Derived Stem Cells. Stem Cells Transl Med 2016; 5:938-45. [PMID: 27217346 PMCID: PMC4922853 DOI: 10.5966/sctm.2015-0291] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 02/15/2016] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Pancreatic adenocarcinoma remains one of the most challenging diseases of modern gastroenterology, and, even though considerable effort has been put into understanding its pathogenesis, the exact molecular mechanisms underlying the development and/or systemic progression of this malignancy still remain unclear. Recently, much attention has been paid to the potential role of bone marrow-derived stem cells (BMSCs) in this malignancy. Hence, herein, we comprehensively review the most recent discoveries and current achievements and concepts in this field. Specifically, we discuss the significance of identifying pancreatic cancer stem cells and novel therapeutic approaches involving molecular interference of their metabolism. We also describe advances in the current understanding of the biochemical and molecular mechanisms responsible for BMSC mobilization during pancreatic cancer development and systemic spread. Finally, we summarize experimental, translational, and/or clinical evidence regarding the contribution of bone marrow-derived mesenchymal stem cells, endothelial progenitor cells, hematopoietic stem/progenitor cells, and pancreatic stellate cells in pancreatic cancer development/progression. We also present their potential therapeutic value for the treatment of this deadly malignancy in humans. SIGNIFICANCE Different bone marrow-derived stem cell populations contribute to the development and/or progression of pancreatic cancer, and they might also be a promising "weapon" that can be used for anticancer treatments in humans. Even though the exact role of these stem cells in pancreatic cancer development and/or progression in humans still remains unclear, this concept continues to drive a completely novel scientific avenue in pancreatic cancer research and gives rise to innovative ideas regarding novel therapeutic modalities that can be safely offered to patients.
Collapse
Affiliation(s)
- Wojciech Błogowski
- Department of Internal Medicine, University of Zielona Góra, Zielona Góra, Poland
| | - Tomasz Bodnarczuk
- Division of Internal Medicine, 109th Military Hospital, Szczecin, Poland
| | - Teresa Starzyńska
- Department of Gastroenterology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
69
|
Hagenhoff A, Bruns CJ, Zhao Y, von Lüttichau I, Niess H, Spitzweg C, Nelson PJ. Harnessing mesenchymal stem cell homing as an anticancer therapy. Expert Opin Biol Ther 2016; 16:1079-92. [PMID: 27270211 DOI: 10.1080/14712598.2016.1196179] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Mesenchymal stromal cells (MSCs) are non-hematopoietic progenitor cells that have been exploited as vehicles for cell-based cancer therapy. The general approach is based on the innate potential of adoptively applied MSC to undergo facilitated recruitment to malignant tissue. MSC from different tissue sources have been engineered using a variety of therapy genes that have shown efficacy in solid tumor models. AREAS COVERED In this review we will focus on the current developments of MSC-based gene therapy, in particular the diverse approaches that have been used for MSCs-targeted tumor therapy. We also discuss some outstanding issues and general prospects for their clinical application. EXPERT OPINION The use of modified mesenchymal stem cells as therapy vehicles for the treatment of solid tumors has progressed to the first generation of clinical trials, but the general field is still in its infancy. There are many questions that need to be addressed if this very complex therapy approach is widely applied in clinical settings. More must be understood about the mechanisms underlying tumor tropism and we need to identify the optimal source of the cells used. Outstanding issues also include the therapy transgenes used, and which tumor types represent viable targets for this therapy.
Collapse
Affiliation(s)
- Anna Hagenhoff
- a Department of Pediatrics and Pediatric Oncology Center, Klinikum rechts der Isar , Technical University , Munich , Germany
| | - Christiane J Bruns
- b Department of Surgery , Otto-von-Guericke University , Magdeburg , Germany
| | - Yue Zhao
- b Department of Surgery , Otto-von-Guericke University , Magdeburg , Germany
| | - Irene von Lüttichau
- a Department of Pediatrics and Pediatric Oncology Center, Klinikum rechts der Isar , Technical University , Munich , Germany
| | - Hanno Niess
- c Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery , University of Munich , Munich , Germany
| | - Christine Spitzweg
- d Department of Internal Medicine II , University of Munich , Munich , Germany
| | - Peter J Nelson
- e Clinical Biochemistry Group, Medizinische Klinik und Poliklinik IV , University of Munich , Munich , Germany
| |
Collapse
|
70
|
Niess H, Camaj P, Mair R, Renner A, Zhao Y, Jäckel C, Nelson PJ, Jauch KW, Bruns CJ. Overexpression of IFN-induced protein with tetratricopeptide repeats 3 (IFIT3) in pancreatic cancer: cellular "pseudoinflammation" contributing to an aggressive phenotype. Oncotarget 2016; 6:3306-18. [PMID: 25650658 PMCID: PMC4413655 DOI: 10.18632/oncotarget.2494] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 09/16/2014] [Indexed: 12/20/2022] Open
Abstract
Inflammation contributes to important traits that cancer cells acquire during malignant progression. Gene array data recently identified upregulation of interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) in aggressive pancreatic cancer cells. IFIT3 belongs to the group of interferon stimulated genes (ISG), can be induced by several cellular stress stimuli and by its tetratricopeptide repeats interacts with a multitude of cellular proteins. Upregulation of IFIT3 was confirmed in the aggressive pancreatic cancer cell line L3.6pl compared with its less aggressive cell line of origin, COLO357FG. Transgenic induction of IFIT3 expression in COLO357FG resulted in greater mass of orthotopic tumors and higher prevalence of metastases. Several important traits that mediate malignancy were altered by IFIT3: increased VEGF and IL-6 secretion, chemoresistance and decreased starvation-induced apoptosis. IFIT3 showed binding to JNK and STAT1, the latter being an important inducer of IFIT3 expression. Despite still being alterable by “classical” IFN or NFκB signaling, our findings indicate constitutive - possibly auto-regulated - upregulation of IFIT3 in L3.6pl without presence of an adequate inflammatory stimulus. The transcription factor SOX9, which is linked to regulation of hypoxia-related genes, was identified as a key mediator of upregulation of the oncogene IFIT3 and thereby sustaining a “pseudoinflammatory” cellular condition.
Collapse
Affiliation(s)
- Hanno Niess
- Department of Surgery, Medical Center of the Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Peter Camaj
- Department of Surgery, Medical Center of the Otto-von-Guericke-University, Magdeburg, Germany
| | - Ruth Mair
- Department of Surgery, Medical Center of the Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Andrea Renner
- Department of Surgery, Medical Center of the Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Yue Zhao
- Department of Surgery, Medical Center of the Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Carsten Jäckel
- Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Universitaet Muenchen, Arbeitsgruppe Klinische Biochemie, Munich, Germany
| | - Peter J Nelson
- Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Universitaet Muenchen, Arbeitsgruppe Klinische Biochemie, Munich, Germany
| | - Karl-Walter Jauch
- Department of Surgery, Medical Center of the Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Christiane J Bruns
- Department of Surgery, Medical Center of the Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
71
|
Kopaczka K, Skowron K, Kolanko E, Czekaj P. The relationship between amniotic epithelial cells and their microenvironment. J Appl Biomed 2016. [DOI: 10.1016/j.jab.2015.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
72
|
Rhee KJ, Lee JI, Eom YW. Mesenchymal Stem Cell-Mediated Effects of Tumor Support or Suppression. Int J Mol Sci 2015; 16:30015-33. [PMID: 26694366 PMCID: PMC4691158 DOI: 10.3390/ijms161226215] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/27/2015] [Accepted: 12/01/2015] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can exhibit a marked tropism towards site of tumors. Many studies have reported that tumor progression and metastasis increase by MSCs. In contrast, other studies have shown that MSCs suppress growth of tumors. MSCs contribute to tumor growth promotion by several mechanisms: (1) transition to tumor-associated fibroblasts; (2) suppression of immune response; (3) promotion of angiogenesis; (4) stimulation of epithelial-mesenchymal transition (EMT); (5) contribution to the tumor microenvironment; (6) inhibition of tumor cell apoptosis; and (7) promotion of tumor metastasis. In contrast to the tumor-promoting properties, MSCs inhibit tumor growth by increasing inflammatory infiltration, inhibiting angiogenesis, suppressing Wnt signaling and AKT signaling, and inducing cell cycle arrest and apoptosis. In this review, we will discuss potential mechanisms by which MSC mediates tumor support or suppression and then the possible tumor-specific therapeutic strategies using MSCs as delivery vehicles, based on their homing potential to tumors.
Collapse
Affiliation(s)
- Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, 1 Yonseidae-gil, Wonju 26493, Korea.
| | - Jong In Lee
- Department of Hematology-Oncology, Wonju College of Medicine, Yonsei University, 20 Ilsan-ro, Wonju 26426, Korea.
| | - Young Woo Eom
- Cell Therapy and Tissue Engineering Center, Wonju College of Medicine, Yonsei University, 20 Ilsan-ro, Wonju 26426, Korea.
| |
Collapse
|
73
|
Schmohl KA, Müller AM, Wechselberger A, Rühland S, Salb N, Schwenk N, Heuer H, Carlsen J, Göke B, Nelson PJ, Spitzweg C. Thyroid hormones and tetrac: new regulators of tumour stroma formation via integrin αvβ3. Endocr Relat Cancer 2015; 22:941-52. [PMID: 26307023 DOI: 10.1530/erc-15-0245] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2015] [Indexed: 12/18/2022]
Abstract
To improve our understanding of non-genomic, integrin αvβ3-mediated thyroid hormone action in tumour stroma formation, we examined the effects of triiodo-l-thyronine (T3), l-thyroxine (T4) and integrin-specific inhibitor tetrac on differentiation, migration and invasion of mesenchymal stem cells (MSCs) that are an integral part of the tumour's fibrovascular network. Primary human bone marrow-derived MSCs were treated with T3 or T4 in the presence of hepatocellular carcinoma (HCC) cell-conditioned medium (CM), which resulted in stimulation of the expression of genes associated with cancer-associated fibroblast-like differentiation as determined by qPCR and ELISA. In addition, T3 and T4 increased migration of MSCs towards HCC cell-CM and invasion into the centre of three-dimensional HCC cell spheroids. All these effects were tetrac-dependent and therefore integrin αvβ3-mediated. In a subcutaneous HCC xenograft model, MSCs showed significantly increased recruitment and invasion into tumours of hyperthyroid mice compared to euthyroid and, in particular, hypothyroid mice, while treatment with tetrac almost completely eliminated MSC recruitment. These studies significantly improve our understanding of the anti-tumour activity of tetrac, as well as the mechanisms that regulate MSC differentiation and recruitment in the context of tumour stroma formation, as an important prerequisite for the utilisation of MSCs as gene delivery vehicles.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Differentiation/drug effects
- Cell Line, Tumor
- Cell Lineage
- Cell Movement
- Culture Media, Conditioned
- Heterografts
- Humans
- Hyperthyroidism/chemically induced
- Hyperthyroidism/complications
- Hypothyroidism/chemically induced
- Hypothyroidism/complications
- Integrin alphaVbeta3/physiology
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms, Experimental/complications
- Liver Neoplasms, Experimental/pathology
- Male
- Mesenchymal Stem Cells/drug effects
- Mice
- Mice, Nude
- Neoplasm Invasiveness
- Neoplasm Proteins/physiology
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/physiopathology
- Specific Pathogen-Free Organisms
- Spheroids, Cellular
- Stromal Cells/pathology
- Thyroxine/analogs & derivatives
- Thyroxine/pharmacology
- Thyroxine/therapeutic use
- Thyroxine/toxicity
- Triiodothyronine/pharmacology
- Triiodothyronine/therapeutic use
- Triiodothyronine/toxicity
- Tumor Microenvironment
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Kathrin A Schmohl
- Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany
| | - Andrea M Müller
- Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany
| | - Alexandra Wechselberger
- Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany
| | - Svenja Rühland
- Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany
| | - Nicole Salb
- Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany
| | - Nathalie Schwenk
- Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany
| | - Heike Heuer
- Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany
| | - Janette Carlsen
- Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany
| | - Burkhard Göke
- Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany
| | - Peter J Nelson
- Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany
| | - Christine Spitzweg
- Department of Internal Medicine IIUniversity Hospital of Munich, Munich, GermanyMedizinische Klinik und Poliklinik IVUniversity Hospital of Munich, Munich, GermanyDepartment of Biology IILudwig-Maximilians-University, Munich, GermanyLeibniz Institute for Environmental MedicineDüsseldorf, GermanyDepartment of Nuclear MedicineUniversity Hospital of Munich, Munich, GermanyUniversity Medical Center Hamburg-EppendorfHamburg, Germany
| |
Collapse
|
74
|
Hua J, Qian DH, Song ZS. Mesenchymal stem cell transplantation for treatment of digestive diseases. Shijie Huaren Xiaohua Zazhi 2015; 23:5263-5268. [DOI: 10.11569/wcjd.v23.i33.5263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Treatment of diseases using mesenchymal stem cells (MSCs) has gained great breakthrough with the discovery of properties of MSCs since 1990s. So far, MSC transplantation in the treatment of digestive tract diseases is mainly focused on hepatic cirrhosis, liver failure, acute or chronic pancreatitis, inflammatory bowel disease and digestive tumors. In the current editorial, we rely primarily on the existing evidence to gain a comprehensive perspective toward this area.
Collapse
|
75
|
Paniccia A, Merkow J, Edil BH, Zhu Y. Immunotherapy for pancreatic ductal adenocarcinoma: an overview of clinical trials. Chin J Cancer Res 2015; 27:376-91. [PMID: 26361407 DOI: 10.3978/j.issn.1000-9604.2015.05.01] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/08/2015] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related death and current therapeutic strategies are often unsatisfactory. Identification and development of more efficacious therapies is urgently needed. Immunotherapy offered encouraging results in preclinical models during the last decades, and several clinical trials have explored its therapeutic application in PDAC. The aim of this review is to summarize the results of clinical trials conducted to evaluate the future perspective of immunotherapy in the treatment of PDAC.
Collapse
Affiliation(s)
- Alessandro Paniccia
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Justin Merkow
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Barish H Edil
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yuwen Zhu
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
76
|
Peng LH, Zhang YH, Han LJ, Zhang CZ, Wu JH, Wang XR, Gao JQ, Mao ZW. Cell Membrane Capsules for Encapsulation of Chemotherapeutic and Cancer Cell Targeting in Vivo. ACS APPLIED MATERIALS & INTERFACES 2015; 7:18628-37. [PMID: 26262951 DOI: 10.1021/acsami.5b05065] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Systemic administration of chemotherapeutic agents can cause indiscriminate drug distribution and severe toxicity. Until now, encapsulation and targeting of drugs have typically relied on synthetic vehicles, which cannot minimize the clearance by the renal system and may also increase the risk of chemical side effects. Cell membrane capsules (CMCs) provide a generic and far more natural approach to the challenges of drug encapsulation and delivery in vivo. Here aptamer AS1411, which can recognize and bind overexpressed nucleolin on a cancer cell membrane, was chemically conjugated onto CMCs. As a result, AS1411 modified CMCs showed enhanced ingestion in certain cancer cells in vitro and accumulation in mouse cancer xenografts in vivo. Chemotherapeutics and contrast agents with therapeutically significant concentrations can be packaged into CMCs by reversible permeating their plasma membranes. The systematic administration of cancer targeting CMCs loaded with doxorubicin hydrochloride can significantly inhibit tumor growth in mouse xenografts, with significantly reduced toxicity compared to free drug. These findings suggest that cancer targeting CMCs may have considerable benefits in drug delivery and cancer treatment.
Collapse
Affiliation(s)
- Li-Hua Peng
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Yuan-Hong Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Li-Jie Han
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Chen-Zhen Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Jia-He Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Xia-Rong Wang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Zheng-Wei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University , Hangzhou 310027, People's Republic of China
| |
Collapse
|
77
|
Niess H, von Einem JC, Thomas MN, Michl M, Angele MK, Huss R, Günther C, Nelson PJ, Bruns CJ, Heinemann V. Treatment of advanced gastrointestinal tumors with genetically modified autologous mesenchymal stromal cells (TREAT-ME1): study protocol of a phase I/II clinical trial. BMC Cancer 2015; 15:237. [PMID: 25879229 PMCID: PMC4393860 DOI: 10.1186/s12885-015-1241-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/20/2015] [Indexed: 01/14/2023] Open
Abstract
Background Adenocarcinoma originating from the digestive system is a major contributor to cancer-related deaths worldwide. Tumor recurrence, advanced local growth and metastasis are key factors that frequently prevent these tumors from curative surgical treatment. Preclinical research has demonstrated that the dependency of these tumors on supporting mesenchymal stroma results in susceptibility to cell-based therapies targeting this stroma. Methods/Design TREAT-ME1 is a prospective, uncontrolled, single-arm phase I/II study assessing the safety and efficacy of genetically modified autologous mesenchymal stromal cells (MSC) as delivery vehicles for a cell-based gene therapy for advanced, recurrent or metastatic gastrointestinal or hepatopancreatobiliary adenocarcinoma. Autologous bone marrow will be drawn from each eligible patient after consent for bone marrow donation has been obtained (under a separate EC-approved protocol). In the following ~10 weeks the investigational medicinal product (IMP) is developed for each patient. To this end, the patient’s MSCs are stably transfected with a gamma-retroviral, replication-incompetent and self-inactivating (SIN) vector system containing a therapeutic promoter - gene construct that allows for tumor-specific expression of the therapeutic gene. After release of the IMP the patients are enrolled after given informed consent for participation in the TREAT-ME 1 trial. In the phase I part of the study, the safety of the IMP is tested in six patients by three treatment cycles consisting of re-transfusion of MSCs at different concentrations followed by administration of the prodrug Ganciclovir. In the phase II part of the study, sixteen patients will be enrolled receiving IMP treatment. A subgroup of patients that qualifies for surgery will be treated preoperatively with the IMP to verify homing of the MSCs to tumors as to be confirmed in the surgical specimen. Discussion The TREAT-ME1 clinical study involves a highly innovative therapeutic strategy combining cell and gene therapy and is conducted at a high level of pharmaceutical quality ensuring patient safety. This patient-tailored approach represents the first clinical study worldwide utilizing genetically engineered MSCs in humans. Trial registration EU Clinical Trials Register/European Union Drug Regulating Authorities Clinical Trials Database number: 2012-003741-15
Collapse
Affiliation(s)
- Hanno Niess
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the University of Munich, Munich, Germany.
| | - Jobst C von Einem
- Department of Medical Oncology and Comprehensive Cancer Center, Hospital of the University of Munich, Munich, Germany.
| | - Michael N Thomas
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the University of Munich, Munich, Germany.
| | - Marlies Michl
- Department of Medical Oncology and Comprehensive Cancer Center, Hospital of the University of Munich, Munich, Germany.
| | - Martin K Angele
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the University of Munich, Munich, Germany.
| | - Ralf Huss
- Apceth GmbH and Co. KG, Munich, Germany.
| | | | - Peter J Nelson
- Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Universitaet Muenchen, Arbeitsgruppe Klinische Biochemie, Munich, Germany.
| | - Christiane J Bruns
- Department of Surgery, Hospital of the University of Magdeburg, Magdeburg, Germany.
| | - Volker Heinemann
- Department of Medical Oncology and Comprehensive Cancer Center, Hospital of the University of Munich, Munich, Germany.
| |
Collapse
|
78
|
Knoop K, Schwenk N, Schmohl K, Müller A, Zach C, Cyran C, Carlsen J, Böning G, Bartenstein P, Göke B, Wagner E, Nelson PJ, Spitzweg C. Mesenchymal stem cell-mediated, tumor stroma-targeted radioiodine therapy of metastatic colon cancer using the sodium iodide symporter as theranostic gene. J Nucl Med 2015; 56:600-6. [PMID: 25745085 DOI: 10.2967/jnumed.114.146662] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 01/12/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED The tumor-homing property of mesenchymal stem cells (MSCs) allows targeted delivery of therapeutic genes into the tumor microenvironment. The application of sodium iodide symporter (NIS) as a theranostic gene allows noninvasive imaging of MSC biodistribution and transgene expression before therapeutic radioiodine application. We have previously shown that linking therapeutic transgene expression to induction of the chemokine CCL5/RANTES allows a more focused expression within primary tumors, as the adoptively transferred MSC develop carcinoma-associated fibroblast-like characteristics. Although RANTES/CCL5-NIS targeting has shown efficacy in the treatment of primary tumors, it was not clear if it would also be effective in controlling the growth of metastatic disease. METHODS To expand the potential range of tumor targets, we investigated the biodistribution and tumor recruitment of MSCs transfected with NIS under control of the RANTES/CCL5 promoter (RANTES-NIS-MSC) in a colon cancer liver metastasis mouse model established by intrasplenic injection of the human colon cancer cell line LS174t. RANTES-NIS-MSCs were injected intravenously, followed by (123)I scintigraphy, (124)I PET imaging, and (131)I therapy. RESULTS Results show robust MSC recruitment with RANTES/CCL5-promoter activation within the stroma of liver metastases as evidenced by tumor-selective iodide accumulation, immunohistochemistry, and real-time polymerase chain reaction. Therapeutic application of (131)I in RANTES-NIS-MSC-treated mice resulted in a significant delay in tumor growth and improved overall survival. CONCLUSION This novel gene therapy approach opens the prospect of NIS-mediated radionuclide therapy of metastatic cancer after MSC-mediated gene delivery.
Collapse
Affiliation(s)
- Kerstin Knoop
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | - Nathalie Schwenk
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | - Kathrin Schmohl
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | - Andrea Müller
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | - Christian Zach
- Department of Nuclear Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Clemens Cyran
- Department of Clinical Radiology, Laboratory for Experimental Radiology, Ludwig-Maximilians-University, Munich, Germany
| | - Janette Carlsen
- Department of Nuclear Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Guido Böning
- Department of Nuclear Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Burkhard Göke
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | - Ernst Wagner
- Department of Pharmacy, Center of Drug Research, Pharmaceutical Biotechnology, Munich, Germany; and
| | - Peter J Nelson
- Clinical Biochemistry Group, Department of Internal Medicine and Policlinic IV, Ludwig-Maximilians-University, Munich, Germany
| | - Christine Spitzweg
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
79
|
Targeting delivery of lipocalin 2-engineered mesenchymal stem cells to colon cancer in order to inhibit liver metastasis in nude mice. Tumour Biol 2015; 36:6011-8. [PMID: 25740061 DOI: 10.1007/s13277-015-3277-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 02/18/2015] [Indexed: 01/05/2023] Open
Abstract
One of the major obstacles in cancer therapy is the lack of anticancer agent specificity to tumor tissues. The strategy of cell-based therapy is a promising therapeutic option for cancer treatment. The specific tumor-oriented migration of mesenchymal stem cells (MSCs) makes them a useful vehicle to deliver anticancer agents. In this study, we genetically manipulated bone marrow-derived mesenchymal stem cells with their lipocalin 2 (Lcn2) in order to inhibit liver metastasis of colon cancer in nude mice. Lcn2 was successfully overexpressed in transfected MSCs. The PCR results of SRY gene confirmed the presence of MSCs in cancer liver tissue. This study showed that Lcn2-engineered MSCs (MSC-Lcn2) not only inhibited liver metastasis of colon cancer but also downregulated the expression of vascular endothelial growth factor (VEGF) in the liver. Overall, MSCs by innate tropism toward cancer cells can deliver the therapeutic agent, Lcn2, and inhibit cancer metastasis. Hence, it could be a new modality for efficient targeted delivery of anticancer agent to liver metastasis.
Collapse
|
80
|
Haga H, Yan IK, Takahashi K, Wood J, Zubair A, Patel T. Tumour cell-derived extracellular vesicles interact with mesenchymal stem cells to modulate the microenvironment and enhance cholangiocarcinoma growth. J Extracell Vesicles 2015; 4:24900. [PMID: 25557794 PMCID: PMC4283029 DOI: 10.3402/jev.v4.24900] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 10/31/2014] [Accepted: 11/28/2014] [Indexed: 12/15/2022] Open
Abstract
The contributions of mesenchymal stem cells (MSCs) to tumour growth and stroma formation are poorly understood. Tumour cells can transfer genetic information and modulate cell signalling in other cells through the release of extracellular vesicles (EVs). We examined the contribution of EV-mediated inter-cellular signalling between bone marrow MSCs and tumour cells in human cholangiocarcinoma, highly desmoplastic cancers that are characterized by tumour cells closely intertwined within a dense fibrous stroma. Exposure of MSCs to tumour cell–derived EVs enhanced MSC migratory capability and expression of alpha-smooth muscle actin mRNA, in addition to mRNA expression and release of CXCL-1, CCL2 and IL-6. Conditioned media from MSCs exposed to tumour cell–derived EVs increased STAT-3 phosphorylation and proliferation in tumour cells. These effects were completely blocked by anti-IL-6R antibody. In conclusion, tumour cell–derived EVs can contribute to the generation of tumour stroma through fibroblastic differentiation of MSCs, and can also selectively modulate the cellular release of soluble factors such as IL-6 by MSCs that can, in turn, alter tumour cell proliferation. Thus, malignant cells can “educate” MSCs to induce local microenvironmental changes that enhance tumour cell growth.
Collapse
Affiliation(s)
- Hiroaki Haga
- Department of Cancer Biology, Mayo Clinic Jacksonville, FL, USA
| | - Irene K Yan
- Department of Cancer Biology, Mayo Clinic Jacksonville, FL, USA
| | - Kenji Takahashi
- Department of Cancer Biology, Mayo Clinic Jacksonville, FL, USA
| | - Joseph Wood
- Department of Cancer Biology, Mayo Clinic Jacksonville, FL, USA
| | - Abba Zubair
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | - Tushar Patel
- Department of Cancer Biology, Mayo Clinic Jacksonville, FL, USA; Department of Transplantation, Mayo Clinic Jacksonville, FL, USA;
| |
Collapse
|
81
|
Kyurkchiev D, Bochev I, Ivanova-Todorova E, Mourdjeva M, Oreshkova T, Belemezova K, Kyurkchiev S. Secretion of immunoregulatory cytokines by mesenchymal stem cells. World J Stem Cells 2014; 6:552-570. [PMID: 25426252 PMCID: PMC4178255 DOI: 10.4252/wjsc.v6.i5.552] [Citation(s) in RCA: 462] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 08/20/2014] [Accepted: 09/10/2014] [Indexed: 02/06/2023] Open
Abstract
According to the minimal criteria of the International Society of Cellular Therapy, mesenchymal stem cells (MSCs) are a population of undifferentiated cells defined by their ability to adhere to plastic surfaces when cultured under standard conditions, express a certain panel of phenotypic markers and can differentiate into osteogenic, chondrogenic and adipogenic lineages when cultured in specific inducing media. In parallel with their major role as undifferentiated cell reserves, MSCs have immunomodulatory functions which are exerted by direct cell-to-cell contacts, secretion of cytokines and/or by a combination of both mechanisms. There are no convincing data about a principal difference in the profile of cytokines secreted by MSCs isolated from different tissue sources, although some papers report some quantitative but not qualitative differences in cytokine secretion. The present review focuses on the basic cytokines secreted by MSCs as described in the literature by which the MSCs exert immunodulatory effects. It should be pointed out that MSCs themselves are objects of cytokine regulation. Hypothetical mechanisms by which the MSCs exert their immunoregulatory effects are also discussed in this review. These mechanisms may either influence the target immune cells directly or indirectly by affecting the activities of predominantly dendritic cells. Chemokines are also discussed as participants in this process by recruiting cells of the immune systems and thus making them targets of immunosuppression. This review aims to present and discuss the published data and the personal experience of the authors regarding cytokines secreted by MSCs and their effects on the cells of the immune system.
Collapse
|
82
|
Mesenchymal stem cells as cellular vehicles for prodrug gene therapy against tumors. Biochimie 2014; 105:4-11. [DOI: 10.1016/j.biochi.2014.06.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/19/2014] [Indexed: 12/15/2022]
|
83
|
Lee WYW, Zhang T, Lau CPY, Wang CC, Chan KM, Li G. Immortalized human fetal bone marrow-derived mesenchymal stromal cell expressing suicide gene for anti-tumor therapy in vitro and in vivo. Cytotherapy 2014; 15:1484-97. [PMID: 24199592 DOI: 10.1016/j.jcyt.2013.06.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 04/28/2013] [Accepted: 06/19/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND AIMS Cancer is one of the greatest health challenges facing the world today with >10 million new cases of cancer every year. The self-renewal, tumor-homing ability and low immunogenicity of mesenchymal stromal cells (MSCs) make them potential delivery candidates for suicide genes for anti-tumor therapy. However, unstable supply and short life span of adult MSCs in vitro have limited this therapeutic potential. In this study, we aimed to evaluate if immortalization of human fetal bone marrow-derived mesenchymal stromal cells by simian virus 40 (SV40-hfBMSCs) could be a stable source of MSCs for clinical application of suicide gene therapy. METHODS AND RESULTS Transduction of SV40 and herpes simplex virus thymidine kinase-IRES-green fluorescent protein (TK-GFP) did not cause significant change in the stem cell properties of hfBMSCs. The anti-tumor effect of SV40-TK-hfBMSCs in the presence of the prodrug ganciclovir was demonstrated in vitro and in nude mice bearing human prostate cancer cells, DU145 and PC3, which had been transduced with luciferase and GFP for imaging evaluation by an in vivo live imaging system (IVIS 200 imaging system; Caliper Life Sciences). Repeated injection of low doses (1 × 10(6) cells/kg) of SV40-TK-hfBMSCs was as effective as previously reported and did not cause observable harmful side effects in multiple organs. Mixed lymphocyte reaction showed that SV40-TK-hfBMSCs did not induce significant proliferation of lymphocytes isolated from healthy adults. CONCLUSIONS Taken together, immortalized hfBMSCs represent a reliable and safe source of MSCs for further clinical translational study.
Collapse
Affiliation(s)
- Wayne Y W Lee
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | | | | | | | | | | |
Collapse
|
84
|
Yang ZS, Tang XJ, Guo XR, Zou DD, Sun XY, Feng JB, Luo J, Dai LJ, Warnock GL. Cancer cell-oriented migration of mesenchymal stem cells engineered with an anticancer gene (PTEN): an imaging demonstration. Onco Targets Ther 2014; 7:441-6. [PMID: 24669193 PMCID: PMC3962313 DOI: 10.2147/ott.s59227] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have been considered to hold great potential as ideal carriers for the delivery of anticancer agents since the discovery of their tumor tropism. This study was performed to demonstrate the effects of phosphatase and tensin homolog (PTEN) engineering on MSCs' capacity for cancer cell-oriented migration. METHODS MSCs were engineered with a PTEN-bearing plasmid and the expression was confirmed with Western blotting. A human glioma cell line (DBTRG) was used as the target cell; DBTRG cell-oriented migration of MSCs was monitored with a micro speed photographic system. RESULTS The expression of transfected PTEN in MSCs was identified by immunoblotting analysis and confirmed with cell viability assessment of target cells. The DBTRG cell-oriented migration of PTEN-engineered MSCs was demonstrated by a real-time dynamic monitoring system, and a phagocytosis-like action of MSCs was also observed. CONCLUSION MSCs maintained their capacity for cancer cell-directed migration after they were engineered with anticancer genes. This study provides the first direct evidence of MSCs' tropism post-anticancer gene engineering.
Collapse
Affiliation(s)
- Zhuo-Shun Yang
- Hubei Key Laboratory of Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Xiang-Jun Tang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Xing-Rong Guo
- Hubei Key Laboratory of Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Dan-Dan Zou
- Hubei Key Laboratory of Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Xu-Yong Sun
- Guangxi Key Laboratory for Transplant Medicine, 303 Hospital of PLA, Nanning, People's Republic of China
| | - Jing-Bo Feng
- Hubei Key Laboratory of Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Jie Luo
- Hubei Key Laboratory of Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Long-Jun Dai
- Hubei Key Laboratory of Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China; ; Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Garth L Warnock
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
85
|
Hong IS, Lee HY, Kang KS. Mesenchymal stem cells and cancer: friends or enemies? Mutat Res 2014; 768:98-106. [PMID: 24512984 DOI: 10.1016/j.mrfmmm.2014.01.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 12/21/2022]
Abstract
There is increasing evidence that mesenchymal stem cells (MSCs) have the ability to migrate and engraft into tumor sites and exert stimulatory effects on cancer cell growth, invasion and even metastasis through direct and/or indirect interaction with tumor cells. However, these pro-tumorigenic effects of MSCs are still being discovered and may even involve opposing effects. MSCs can be friends or enemies of cancer cells: they may stimulate tumor development by regulating immune surveillance, growth, and angiogenesis. On the other hand, they may inhibit tumor growth by inhibiting survival signaling such as Wnt and Akt pathway. MSCs have also been proposed as an attractive candidate for the delivery of anti-tumor agents, owing to their ability to home into tumor sites and to secrete cytokines. Detailed information about the mutual interactions between tumor cells and MSCs will undoubtedly lead to safer and more effective clinical therapy for tumors. In this article, we summarize a number of findings to provide current information on the potential roles of MSCs in tumor development; we then discuss the therapeutic potential of engineered MSCs to reveal any meaningful clinical applications.
Collapse
Affiliation(s)
- In-Sun Hong
- Department of Molecular Medicine, Gachon University, Incheon, Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Hwa-Yong Lee
- Industry-academic cooperation foundation, Jungwon University, Chungbuk, Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, Seoul National University, Seoul, Republic of Korea; Department of Veterinary Public Health, Laboratory of Stem Cell and Tumor Biology, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
86
|
Moniri MR, Dai LJ, Warnock GL. The challenge of pancreatic cancer therapy and novel treatment strategy using engineered mesenchymal stem cells. Cancer Gene Ther 2014; 21:12-23. [PMID: 24384772 DOI: 10.1038/cgt.2013.83] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 12/09/2013] [Indexed: 12/26/2022]
Abstract
Mesenchymal stem cells (MSCs) have attracted significant attention in cancer research as a result of their accessibility, tumor-oriented homing capacity, and the feasibility of auto-transplantation. This review provides a comprehensive overview of current challenges in pancreatic cancer therapy, and we propose a novel strategy for using MSCs as means of delivering anticancer genes to the site of pancreas. We aim to provide a practical platform for the development of MSC-based therapy for pancreatic cancer.
Collapse
Affiliation(s)
- M R Moniri
- Department of Surgery, University of British Columbia, Vancouver BC, Canada
| | - L-J Dai
- 1] Department of Surgery, University of British Columbia, Vancouver BC, Canada [2] Hubei Key Laboratory of Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - G L Warnock
- Department of Surgery, University of British Columbia, Vancouver BC, Canada
| |
Collapse
|
87
|
Brennen WN, Denmeade SR, Isaacs JT. Mesenchymal stem cells as a vector for the inflammatory prostate microenvironment. Endocr Relat Cancer 2013; 20:R269-90. [PMID: 23975882 PMCID: PMC3994592 DOI: 10.1530/erc-13-0151] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) have an inherent tropism for sites of inflammation, which are frequently present in sites of cancer, including prostatic lesions. MSCs have been defined as CD73/CD90/CD105 triple-positive cells in the absence of hematopoietic lineage markers with the ability to differentiate into multiple mesodermal lineages, including osteoblasts, adipocytes, and chondrocytes. Our group has previously demonstrated that MSCs represent between 0.01 and 1.1% of the total cells present in human prostatectomy tissue. In addition to their multi-lineage differentiation potential, MSCs are immunoprivileged in nature and have a range of immunomodulatory effects on both the innate and adaptive arms of the immune system. MSCs have been detected in an increasing array of tissues, and evidence suggests that they are likely present in perivascular niches throughout the body. These observations suggest that MSCs represent critical mediators of the overall immune response during physiological homeostasis and likely contribute to pathophysiological conditions as well. Chronic inflammation has been suggested as an initiating event and progression factor in prostate carcinogenesis, a process in which the immunosuppressive properties of MSCs may play a role. MSCs have also been shown to influence malignant progression through a variety of other mechanisms, including effects on tumor proliferation, angiogenesis, survival, and metastasis. Additionally, human bone marrow-derived MSCs have been shown to traffic to human prostate cancer xenografts in immunocompromised murine hosts. The trafficking properties and immunoprivileged status of MSCs suggest that they can be exploited as an allogeneic cell-based vector to deliver cytotoxic or diagnostic agents for therapy.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Chemical Therapeutics Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21287, USA
| | | | | |
Collapse
|
88
|
Human mesenchymal stem cells and their paracrine factors for the treatment of brain tumors. Cancer Gene Ther 2013; 20:539-43. [PMID: 24052128 DOI: 10.1038/cgt.2013.59] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 08/16/2013] [Accepted: 08/18/2013] [Indexed: 12/20/2022]
Abstract
Glioblastoma multiforme (GBM or World Health Organization (WHO) grade IV) is the most malignant tumor of the brain. Despite conventional combination treatment of surgery, radiotherapy and chemotherapy, the survival of patients with GBM is generally <1 year. It is a great challenge to identify an effective drug that could efficiently inhibit (i) the growth of cancer cells; (ii) angiogenesis; (iii) metastasis; (iv) tumor-associated inflammation; (v) inactivate proliferative signal, (vi) induce specific apoptosis, and yet causes minimal harm to normal cells. Mesenchymal stem cells (MSCS) do possess some unique features (inherent tumor tropism; anti-inflammatory and immunosuppressive properties) that are not commonly found in current anticancer agents. These cells are known to secrete a vast array of proteins including growth factors, cytokines, chemokines and so on that regulate their biology in an autocrine or paracrine manner in accordance to the surrounding microenvironment. This review briefly summarizes the biology of MSCs and discusses their properties and new development for brain cancer treatment.
Collapse
|
89
|
Knoop K, Schwenk N, Dolp P, Willhauck MJ, Zischek C, Zach C, Hacker M, Göke B, Wagner E, Nelson PJ, Spitzweg C. Stromal targeting of sodium iodide symporter using mesenchymal stem cells allows enhanced imaging and therapy of hepatocellular carcinoma. Hum Gene Ther 2013; 24:306-16. [PMID: 23402366 DOI: 10.1089/hum.2012.104] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The tumor-homing property of mesenchymal stem cells (MSC) has lead to their use as delivery vehicles for therapeutic genes. The application of the sodium iodide symporter (NIS) as therapy gene allows noninvasive imaging of functional transgene expression by (123)I-scintigraphy or PET-imaging, as well as therapeutic application of (131)I or (188)Re. Based on the critical role of the chemokine RANTES (regulated on activation, normal T-cell expressed and presumably secreted)/CCL5 secreted by MSCs in the course of tumor stroma recruitment, use of the RANTES/CCL5 promoter should allow tumor stroma-targeted expression of NIS after MSC-mediated delivery. Using a human hepatocellular cancer (HCC) xenograft mouse model (Huh7), we investigated distribution and tumor recruitment of RANTES-NIS-engineered MSCs after systemic injection by gamma camera imaging. (123)I-scintigraphy revealed active MSC recruitment and CCL5 promoter activation in the tumor stroma of Huh7 xenografts (6.5% ID/g (123)I, biological half-life: 3.7 hr, tumor-absorbed dose: 44.3 mGy/MBq). In comparison, 7% ID/g (188)Re was accumulated in tumors with a biological half-life of 4.1 hr (tumor-absorbed dose: 128.7 mGy/MBq). Administration of a therapeutic dose of (131)I or (188)Re (55.5 MBq) in RANTES-NIS-MSC-treated mice resulted in a significant delay in tumor growth and improved survival without significant differences between (131)I and (188)Re. These data demonstrate successful stromal targeting of NIS in HCC tumors by selective recruitment of NIS-expressing MSCs and by use of the RANTES/CCL5 promoter. The resulting tumor-selective radionuclide accumulation was high enough for a therapeutic effect of (131)I and (188)Re opening the exciting prospect of NIS-mediated radionuclide therapy of metastatic cancer using genetically engineered MSCs as gene delivery vehicles.
Collapse
Affiliation(s)
- Kerstin Knoop
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
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
|
91
|
Wörmann SM, Diakopoulos KN, Lesina M, Algül H. The immune network in pancreatic cancer development and progression. Oncogene 2013; 33:2956-67. [PMID: 23851493 DOI: 10.1038/onc.2013.257] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 04/22/2013] [Accepted: 04/22/2013] [Indexed: 02/07/2023]
Abstract
The presence of stromal desmoplasia is a hallmark of spontaneous pancreatic ductal adenocarcinoma, forming a unique microenvironment that comprises many cell types. Only recently, the immune system has entered the pathophysiology of pancreatic ductal adenocarcinoma development. Tumor cells in the pancreas seem to dysbalance the immune system, thus facilitating spontaneous cancer development. This review will try to assemble all relevant data to demonstrate the implications of the immune network on spontaneous cancer development.
Collapse
Affiliation(s)
- S M Wörmann
- Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - K N Diakopoulos
- Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - M Lesina
- Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - H Algül
- Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
92
|
Abstract
Cell therapy with Multipotent Mesenchymal Stromal Cells (MSC) holds enormous promise for the treatment of a large number of degenerative and immune/inflammatory diseases. Their multilineage differentiation potential, immunoprivilege and capacity of promoting recovery of damaged tissues coupled with anti-inflammatory and immunosuppressive properties are the focus of a multitude of clinical studies currently underway. The recognized clinical potential of MSC repairing/immunomodulatory effects now encompasses graft-versus-host disease, hematologic malignancies, cardiovascular diseases, neurologic and inherited diseases, autoimmune diseases, organ transplantation, refractory wounds, and bone/cartilage defects among others. However, it has been suggested that both the need of extensive ex vivo culture for MSC clinical use, and their proangiogenic, anti-apoptotic and immunomodulatory properties may act together as tumor promoters, raising significant safety concerns. This paper will review the available data on in vitro MSC maldifferentiation and the ability of MSC to sustain tumor growth in vivo, with the aim to clarify whether MSC-based therapeutic approaches may carry actual risk of malignancies.
Collapse
|
93
|
Rapoport N, Payne A, Dillon C, Shea J, Scaife C, Gupta R. Focused ultrasound-mediated drug delivery to pancreatic cancer in a mouse model. J Ther Ultrasound 2013; 1:11. [PMID: 25516800 PMCID: PMC4265944 DOI: 10.1186/2050-5736-1-11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 05/19/2013] [Indexed: 01/14/2023] Open
Abstract
Background Many aspects of the mechanisms involved in ultrasound-mediated therapy remain obscure. In particular, the relative roles of drug and ultrasound, the effect of the time of ultrasound application, and the effect of tissue heating are not yet clear. The current study was undertaken with the goal to clarify these aspects of the ultrasound-mediated drug delivery mechanism. Methods Focused ultrasound-mediated drug delivery was performed under magnetic resonance imaging guidance (MRgFUS) in a pancreatic ductal adenocarcinoma (PDA) model grown subcutaneously in nu/nu mice. Paclitaxel (PTX) was used as a chemotherapeutic agent because it manifests high potency in the treatment of gemcitabine-resistant PDA. Poly(ethylene oxide)-co-poly(d,l-lactide) block copolymer stabilized perfluoro-15-crown-5-ether nanoemulsions were used as drug carriers. MRgFUS was applied at sub-ablative pressure levels in both continuous wave and pulsed modes, and only a fraction of the tumor was treated. Results Positive treatment effects and even complete tumor resolution were achieved by treating the tumor with MRgFUS after injection of nanodroplet encapsulated drug. The MRgFUS treatment enhanced the action of the drug presumably through enhanced tumor perfusion and blood vessel and cell membrane permeability that increased the drug supply to tumor cells. The effect of the pulsed MRgFUS treatment with PTX-loaded nanodroplets was clearly smaller than that of continuous wave MRgFUS treatment, supposedly due to significantly lower temperature increase as measured with MR thermometry and decreased extravasation. The time of the MRgFUS application after drug injection also proved to be an important factor with the best results observed when ultrasound was applied at least 6 h after the injection of drug-loaded nanodroplets. Some collateral damage was observed with particular ultrasound protocols supposedly associated with enhanced inflammation. Conclusion This presented data suggest that there exists an optimal range of ultrasound application parameters and drug injection time. Decreased tumor growth, or complete resolution, was achieved with continuous wave ultrasound pressures below or equal to 3.1 MPa and drug injection times of at least 6 h prior to treatment. Increased acoustic pressure or ultrasound application before or shortly after drug injection gave increased tumor growth when compared to other protocols.
Collapse
Affiliation(s)
- Natalya Rapoport
- Department of Bioengineering, University of Utah, 36 S. Wasatch Dr., Room 3100, Salt Lake City, UT 84112, USA
| | - Allison Payne
- Department of Radiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Christopher Dillon
- Department of Bioengineering, University of Utah, 36 S. Wasatch Dr., Room 3100, Salt Lake City, UT 84112, USA
| | - Jill Shea
- Department of Surgery, University of Utah, Salt Lake City, UT 84112, USA
| | - Courtney Scaife
- Department of Surgery, University of Utah, Salt Lake City, UT 84112, USA
| | - Roohi Gupta
- Department of Bioengineering, University of Utah, 36 S. Wasatch Dr., Room 3100, Salt Lake City, UT 84112, USA ; Current address: Department of Radiation Oncology, Fox Chase Cancer Center, P0103, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| |
Collapse
|
94
|
Keung EZ, Nelson PJ, Conrad C. Concise Review: Genetically Engineered Stem Cell Therapy Targeting Angiogenesis and Tumor Stroma in Gastrointestinal Malignancy. Stem Cells 2013; 31:227-35. [DOI: 10.1002/stem.1269] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 10/11/2012] [Indexed: 12/13/2022]
|
95
|
Hindriksen S, Bijlsma MF. Cancer Stem Cells, EMT, and Developmental Pathway Activation in Pancreatic Tumors. Cancers (Basel) 2012; 4:989-1035. [PMID: 24213498 PMCID: PMC3712732 DOI: 10.3390/cancers4040989] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 10/02/2012] [Accepted: 10/09/2012] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is a disease with remarkably poor patient survival rates. The frequent presence of metastases and profound chemoresistance pose a severe problem for the treatment of these tumors. Moreover, cross-talk between the tumor and the local micro-environment contributes to tumorigenicity, metastasis and chemoresistance. Compared to bulk tumor cells, cancer stem cells (CSC) have reduced sensitivity to chemotherapy. CSC are tumor cells with stem-like features that possess the ability to self-renew, but can also give rise to more differentiated progeny. CSC can be identified based on increased in vitro spheroid- or colony formation, enhanced in vivo tumor initiating potential, or expression of cell surface markers. Since CSC are thought to be required for the maintenance of a tumor cell population, these cells could possibly serve as a therapeutic target. There appears to be a causal relationship between CSC and epithelial-to-mesenchymal transition (EMT) in pancreatic tumors. The occurrence of EMT in pancreatic cancer cells is often accompanied by re-activation of developmental pathways, such as the Hedgehog, WNT, NOTCH, and Nodal/Activin pathways. Therapeutics based on CSC markers, EMT, developmental pathways, or tumor micro-environment could potentially be used to target pancreatic CSC. This may lead to a reduction of tumor growth, metastatic events, and chemoresistance in pancreatic cancer.
Collapse
Affiliation(s)
- Sanne Hindriksen
- Laboratory for Experimental Oncology and Radiobiology, Academic Medical Centre, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | | |
Collapse
|
96
|
Moniri MR, Sun XY, Rayat J, Dai D, Ao Z, He Z, Verchere CB, Dai LJ, Warnock GL. TRAIL-engineered pancreas-derived mesenchymal stem cells: characterization and cytotoxic effects on pancreatic cancer cells. Cancer Gene Ther 2012; 19:652-8. [PMID: 22767216 DOI: 10.1038/cgt.2012.46] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mesenchymal stem cells (MSCs) have attracted great interest in cancer therapy owing to their tumor-oriented homing capacity and the feasibility of autologous transplantation. Currently, pancreatic cancer patients face a very poor prognosis, primarily due to the lack of therapeutic strategies with an effective degree of specificity. Anticancer gene-engineered MSCs specifically target tumor sites and can produce anticancer agents locally and constantly. This study was performed to characterize pancreas-derived MSCs and investigate the effects of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-engineered MSCs on pancreatic cancer cells under different culture conditions. Pancreas-derived MSCs exhibited positive expression on CD44, CD73, CD95, CD105, negative on CD34 and differentiated into adipogenic and osteogenic cells. TRAIL expression was assessed by both enzyme-linked immunosorbent assay and western blot analysis. Different patterns of TRAIL receptor expression were observed on the pancreatic cancer cell lines, including PANC1, HP62, ASPC1, TRM6 and BXPC3. Cell viability was assessed using a real-time monitoring system. Pancreatic cancer cell death was proportionally related to conditioned media from MSC(nsTRAIL) and MSC(stTRAIL). The results suggest that MSCs exhibit intrinsic inhibition of pancreatic cancer cells and that this effect can be potentiated by TRAIL-transfection on death receptor-bearing cell types.
Collapse
Affiliation(s)
- M R Moniri
- Department of Surgery, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Bao Q, Zhao Y, Niess H, Conrad C, Schwarz B, Jauch KW, Huss R, Nelson PJ, Bruns CJ. Mesenchymal stem cell-based tumor-targeted gene therapy in gastrointestinal cancer. Stem Cells Dev 2012; 21:2355-63. [PMID: 22530882 DOI: 10.1089/scd.2012.0060] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem (or stromal) cells (MSCs) are nonhematopoietic progenitor cells that can be obtained from bone marrow aspirates or adipose tissue, expanded and genetically modified in vitro, and then used for cancer therapeutic strategies in vivo. Here, we review available data regarding the application of MSC-based tumor-targeted therapy in gastrointestinal cancer, provide an overview of the general history of MSC-based gene therapy in cancer research, and discuss potential problems associated with the utility of MSC-based therapy such as biosafety, immunoprivilege, transfection methods, and distribution in the host.
Collapse
Affiliation(s)
- Qi Bao
- Department of Surgery, University of Munich, Campus Großhadern, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Christ B, Stock P. Mesenchymal stem cell-derived hepatocytes for functional liver replacement. Front Immunol 2012; 3:168. [PMID: 22737154 PMCID: PMC3381218 DOI: 10.3389/fimmu.2012.00168] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 06/04/2012] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells represent an alternate cell source to substitute for primary hepatocytes in hepatocyte transplantation because of their multiple differentiation potential and nearly unlimited availability. They may differentiate into hepatocyte-like cells in vitro and maintain specific hepatocyte functions also after transplantation into the regenerating livers of mice or rats both under injury and non-injury conditions. Depending on the underlying liver disease their mode of action is either to replace the diseased liver tissue or to support liver regeneration through their anti-inflammatory and anti-apoptotic as well as their pro-proliferative action.
Collapse
Affiliation(s)
- Bruno Christ
- Applied Molecular Hepatology Laboratory, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig Leipzig, Germany
| | | |
Collapse
|
99
|
Selective targeting of genetically engineered mesenchymal stem cells to tumor stroma microenvironments using tissue-specific suicide gene expression suppresses growth of hepatocellular carcinoma. Ann Surg 2012; 254:767-74; discussion 774-5. [PMID: 22042469 DOI: 10.1097/sla.0b013e3182368c4f] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The use of engineered mesenchymal stem cells (MSCs) as therapeutic vehicles for the treatment of experimental pancreatic and breast cancer has been previously demonstrated. The potential application of MSCs for the treatment of hepatocellular carcinoma (HCC) has been controversial. The general approach uses engineered MSCs to target different aspects of tumor biology, including angiogenesis or the fibroblast-like stromal compartment, through the use of tissue-specific expression of therapeutic transgenes. The aim of the present study was (1) to evaluate the effect of exogenously added MSCs on the growth of HCC and (2) the establishment of an MSC-based suicide gene therapy for experimental HCC. METHODS Mesenchymal stem cells were isolated from bone marrow of C57/Bl6 p53(-/-) mice. The cells were injected into mice with HCC xenografts and the effect on tumor proliferation and angiogenesis was evaluated. The cells were then stably transfected with red fluorescent protein (RFP) or Herpes simplex virus thymidine kinase (HSV-Tk) gene under control of the Tie2 promoter/enhancer or the CCL5 promoter. Mesenchymal stem cells were injected intravenously into mice with orthotopically growing xenografts of HCC and treated with ganciclovir (GCV). RESULTS Ex vivo examination of hepatic tumors revealed tumor-specific recruitment, enhanced tumor growth, and increased microvessel density after nontherapeutic MSC injections. After their homing to the hepatic xenografts, engineered MSCs demonstrated activation of the Tie2 or CCL5 promoter as shown by RFP expression. Application of CCL5/HSV-TK transfected MSCs in combination with GCV significantly reduced tumor growth by 56.4% as compared with the control group and by 71.6% as compared with nontherapeutic MSC injections. CCL5/HSV-TK(+) transfected MSCs proved more potent in tumor inhibition as compared with Tie2/HSV-TK(+) MSCs. CONCLUSION Exogenously added MSCs are recruited to growing HCC xenografts with concomitant activation of the CCL5 or Tie2 promoters within the MSCs. Stem cell-mediated introduction of suicide genes into the tumor followed by prodrug administration was effective for treatment of experimental HCC and thus may help fill the existing gap in bridging therapies for patients suffering from advanced HCCs.
Collapse
|
100
|
Liu H, Lu K, MacAry PA, Wong KL, Heng A, Cao T, Kemeny DM. Soluble molecules are key in maintaining the immunomodulatory activity of murine mesenchymal stromal cells. J Cell Sci 2012; 125:200-8. [PMID: 22250196 DOI: 10.1242/jcs.093070] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) possess both immuno-privileged and immuno-inhibitory properties that contribute to their therapeutic effects. Ex vivo expansion is required to obtain sufficient cells for therapy, but might also alter their immunological properties. To date there has been no systematic study of MSC immunobiology during extended culture. Here, we demonstrate that both immuno-privilege and immunosuppressive properties of MSCs change with increasing passage. We demonstrate that although MSCs exhibit powerful immunosuppressive effects through secretion of transforming growth factor-β (TGF-β) and induction of interleukin-10, these effects are diminished by a concomitant increase in MSC immunogenicity. Interferon-γ treatment for 3 days induced extendedly cultured MSCs to express significantly higher levels of major histocompatibility complex class I. In vivo, this results in cells that induce significant delayed-type hypersensitivity reactions in allogeneic recipients. Importantly, these effects are alleviated by isolation of the transplanted MSCs using a semi-permeable barrier. Under these conditions, even MSCs cultured through as many as 14 passages still exhibit immuno-inhibitory effects in vivo. Furthermore, the levels of anti-inflammatory molecule TGF-β secreted by MSCs were maintained in the extended culture. These data shed light on the variable results of allogeneic MSCs in transplantation and suggest alternative strategies for prolonging the effect of allogeneic MSCs in cell-based therapy.
Collapse
Affiliation(s)
- Hua Liu
- Department of Oral & Maxillofacial Surgery, Faculty of Dentistry, National University of Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|