1
|
Shams F, Pourjabbar B, Hashemi N, Farahmandian N, Golchin A, Nuoroozi G, Rahimpour A. Current progress in engineered and nano-engineered mesenchymal stem cells for cancer: From mechanisms to therapy. Biomed Pharmacother 2023; 167:115505. [PMID: 37716113 DOI: 10.1016/j.biopha.2023.115505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023] Open
Abstract
Mesenchymal stem cells (MSCs), as self-renewing multipotent stromal cells, have been considered promising agents for cancer treatment. A large number of studies have demonstrated the valuable properties of MSC-based treatment, such as low immunogenicity and intrinsic tumor-trophic migratory properties. To enhance the potency of MSCs for therapeutic purposes, equipping MSCs with targeted delivery functions using genetic engineering is highly beneficial. Genetically engineered MSCs can express tumor suppressor agents such as pro-apoptotic, anti-proliferative, anti-angiogenic factors and act as ideal delivery vehicles. MSCs can also be loaded with nanoparticle drugs for increased efficacy and externally moderated targeting. Moreover, exosomes secreted by MSCs have important physiological properties, so they can contribute to intercellular communication and transfer cargo into targeted tumor cells. The precise role of genetically modified MSCs in tumor environments is still up for debate, but the beginning of clinical trials has been confirmed by promising results from preclinical investigations of MSC-based gene therapy for a wide range of malignancies. This review highlights the advanced techniques of engineering/nano-engineering and MSC-derived exosomes in tumor-targeted therapy.
Collapse
Affiliation(s)
- Forough Shams
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, 1968917313 Tehran, Iran
| | - Bahareh Pourjabbar
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nader Hashemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, 1968917313 Tehran, Iran
| | - Navid Farahmandian
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Golchin
- Cellular & Molecular Research Center, Cellular & Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia 57157993313, Iran; Department of Clinical Biochemistry & Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia 57157993313, Islamic Republic of Iran
| | - Ghader Nuoroozi
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Rahimpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Mehralizadeh H, Nazari A, Oruji F, Roostaie M, Hosseininozari G, Yazdani O, Esbati R, Roudini K. Cytokine sustained delivery for cancer therapy; special focus on stem cell- and biomaterial- based delivery methods. Pathol Res Pract 2023; 247:154528. [PMID: 37257247 DOI: 10.1016/j.prp.2023.154528] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 06/02/2023]
Abstract
As immune regulators, cytokines serve critical role as signaling molecules in response to danger, tissue damage, or injury. Importantly, due to their vital role in immunological surveillance, cytokine therapy has become a promising therapeutics for cancer therapy. Cytokines have, however, been used only in certain clinical settings. Two key characteristics of cytokines contribute to this clinical translational challenge: first, they are highly pleiotropic, and second, in healthy physiology, they are typically secreted and act very locally in tissues. Systemic administration of the cytokines can consequently result in serious side effects. Thus, scientists have sought various strategies to circumvent theses hurdles. Recent in vivo reports signify that cytokine delivery platforms can increase their safety and therapeutic efficacy in tumor xenografts. Meanwhile, cytokine delivery using multipotent stem cells, in particular mesenchymal stem/stromal cells (MSCs), and also a diversity of particles and biomaterials has demonstrated greater capability in this regards. Herein, we take a glimpse into the recent advances in cytokine sustained delivery using stem cells and also biomaterials to ease safe and effective treatments of a myriad of human tumors.
Collapse
Affiliation(s)
| | - Ahmad Nazari
- Tehran University of Medical Sciences, Tehran, Iran
| | - Farshid Oruji
- College of Medicine, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Minoo Roostaie
- School of Medicine, Islamic Azad University Tehran Medical Branch, Tehran, Iran
| | - Ghazaleh Hosseininozari
- Department of Cell and Molecular biology, Babol Branch, Islamic Azad University, Babol, Iran
| | - Omid Yazdani
- Department of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Romina Esbati
- Department of Medicine, Shahid Beheshti University, Tehran, Iran.
| | - Kamran Roudini
- Department of Internal Medicine, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Iran.
| |
Collapse
|
3
|
Quiroz-Reyes AG, Gonzalez-Villarreal CA, Limon-Flores AY, Delgado-Gonzalez P, Martinez-Rodriguez HG, Said-Fernandez SL, Soto-Dominguez A, Rivas-Estilla AM, Islas JF, Molina-De la Garza JF, Garza-Treviño EN. Mesenchymal Stem Cells Genetically Modified by Lentivirus-Express Soluble TRAIL and Interleukin-12 Inhibit Growth and Reduced Metastasis-Relate Changes in Lymphoma Mice Model. Biomedicines 2023; 11:biomedicines11020595. [PMID: 36831131 PMCID: PMC9953195 DOI: 10.3390/biomedicines11020595] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
BACKGROUND Cancer treatment has many side effects; therefore, more efficient treatments are needed. Mesenchymal stem cells (MSC) have immunoregulatory properties, tumor site migration and can be genetically modified. Some proteins, such as soluble TRAIL (sTRAIL) and interleukin-12 (IL-12), have shown antitumoral potential, thus its combination in solid tumors could increase their activity. MATERIALS AND METHODS Lentiviral transduction of bone marrow MSC with green fluorescent protein (GFP) and transgenes (sTRAIL and IL-12) was confirmed by fluorescence microscopy and Western blot. Soluble TRAIL levels were quantified by ELISA. Lymphoma L5178Y cells express a reporter gene (GFP/mCherry), and TRAIL receptor (DR5). RESULTS An in vivo model showed that combined treatment with MSC expressing sTRAIL+IL-12 or IL-12 alone significantly reduced tumor volume and increased survival in BALB/c mice (p < 0.05) with only one application. However, at the histological level, only MSC expressing IL-12 reduced tumor cell infiltration significantly in the right gastrocnemius compared with the control group (p < 0.05). It presented less tissue dysplasia confirmed by fluorescence and hematoxylin-eosin dye; nevertheless, treatment not inhibited hepatic metastasis. CONCLUSIONS MSC expressing IL-12, is or combination with BM-MSC expressing sTRAIL represents an antitumor strategy for lymphoma tumors since they increase survival and reduce tumor development. However, the combination did not show significative additive effect. The localized application did not inhibit metastasis but reduced morphological alterations of tissue associated with liver metastasis.
Collapse
Affiliation(s)
- Adriana G. Quiroz-Reyes
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon (UANL), Monterrey 64460, Mexico
| | - Carlos A. Gonzalez-Villarreal
- Department of Basic Sciences, Laboratory of Molecular Genetics, University of Monterrey (UDEM), Monterrey 66238, Mexico
| | - Alberto Y. Limon-Flores
- Department and Service of Immunology, School of Medicine, Autonomous University of Nuevo Leon (UANL), San Nicolás de los Garza 64460, Mexico
| | - Paulina Delgado-Gonzalez
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon (UANL), Monterrey 64460, Mexico
| | - Herminia G. Martinez-Rodriguez
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon (UANL), Monterrey 64460, Mexico
| | - Salvador L. Said-Fernandez
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon (UANL), Monterrey 64460, Mexico
| | - Adolfo Soto-Dominguez
- Department of Histology, School of Medicine, Autonomous University of Nuevo Leon (UANL), San Nicolás de los Garza 64460, Mexico
| | - Ana M. Rivas-Estilla
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon (UANL), Monterrey 64460, Mexico
| | - Jose F. Islas
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon (UANL), Monterrey 64460, Mexico
| | - Juan F. Molina-De la Garza
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon (UANL), Monterrey 64460, Mexico
| | - Elsa N. Garza-Treviño
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon (UANL), Monterrey 64460, Mexico
- Correspondence: ; Tel.: +52-818-3294-173
| |
Collapse
|
4
|
Azimifar MA, Hashemi M, Babaei N, Salmasi Z, Doosti A. Interleukin gene delivery for cancer gene therapy: In vitro and in vivo studies. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:128-136. [PMID: 36742134 PMCID: PMC9869882 DOI: 10.22038/ijbms.2022.66890.14668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/12/2022] [Indexed: 02/07/2023]
Abstract
Cytokine-mediated cancer therapy has the potential to enhance immunotherapeutic approaches and cancer elimination plans through the endowing of the immune system by providing improved anticancer immunity. Despite the encouraging pioneer studies on interleukins (ILs), the influence of ILs-originated therapeutics is still restricted by a class of potent immunoregulatory cytokines, systemic dose-limiting toxicities, ILs pleiotropy, and administration issues. During previous years, the area of transferring genes encoding immunostimulatory ILs was fundamentally widened to overcome these challenges and expedite ILs-based tumor regression. Numerous viral and non-viral delivery systems are currently available to act as crucial elements of the gene therapy toolbox. Moreover, cell-based cancer therapies are recruiting MSCs in the role of versatile gene delivery platforms to design one of the promising therapeutic approaches. These formulated gene carrier systems can provide possible alternatives to diminish dose-limiting adverse effects, promote administration, and enhance the therapeutic activity of ILs-derived treatment modalities in cancer treatment. This review provides a discussion on the advances of ILs gene delivery systems while focusing on the developing platforms in preclinical cancer immunogene therapy studies.
Collapse
Affiliation(s)
- Mohammad Amin Azimifar
- Department of Cell Molecular Biology, Bushehr Branch, Islamic Azad University, Bushehr, Iran
| | - Maryam Hashemi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nahid Babaei
- Department of Cell Molecular Biology, Bushehr Branch, Islamic Azad University, Bushehr, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Doosti
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
5
|
Razeghian E, Margiana R, Chupradit S, Bokov DO, Abdelbasset WK, Marofi F, Shariatzadeh S, Tosan F, Jarahian M. Mesenchymal Stem/Stromal Cells as a Vehicle for Cytokine Delivery: An Emerging Approach for Tumor Immunotherapy. Front Med (Lausanne) 2021; 8:721174. [PMID: 34513882 PMCID: PMC8430327 DOI: 10.3389/fmed.2021.721174] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/30/2021] [Indexed: 12/22/2022] Open
Abstract
Pro-inflammatory cytokines can effectively be used for tumor immunotherapy, affecting every step of the tumor immunity cycle. Thereby, they can restore antigen priming, improve the effector immune cell frequencies in the tumor microenvironment (TME), and eventually strengthen their cytolytic function. A renewed interest in the anticancer competencies of cytokines has resulted in a substantial promotion in the number of trials to address the safety and efficacy of cytokine-based therapeutic options. However, low response rate along with the high toxicity associated with high-dose cytokine for reaching desired therapeutic outcomes negatively affect their clinical utility. Recently, mesenchymal stem/stromal cells (MSCs) due to their pronounced tropism to tumors and also lower immunogenicity have become a promising vehicle for cytokine delivery for human malignancies. MSC-based delivery of the cytokine can lead to the more effective immune cell-induced antitumor response and provide sustained release of target cytokines, as widely evidenced in a myriad of xenograft models. In the current review, we offer a summary of the novel trends in cytokine immunotherapy using MSCs as a potent and encouraging carrier for antitumor cytokines, focusing on the last two decades' animal reports.
Collapse
Affiliation(s)
- Ehsan Razeghian
- Human Genetics Division, Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Cipto Mangunkusumo Hospital, The National Referral Hospital, Central Jakarta, Indonesia
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Dmitry O. Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russia
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siavash Shariatzadeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Foad Tosan
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
6
|
Challenges and advances in clinical applications of mesenchymal stromal cells. J Hematol Oncol 2021; 14:24. [PMID: 33579329 PMCID: PMC7880217 DOI: 10.1186/s13045-021-01037-x] [Citation(s) in RCA: 331] [Impact Index Per Article: 82.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stromal cells (MSCs), also known as mesenchymal stem cells, have been intensely investigated for clinical applications within the last decades. However, the majority of registered clinical trials applying MSC therapy for diverse human diseases have fallen short of expectations, despite the encouraging pre-clinical outcomes in varied animal disease models. This can be attributable to inconsistent criteria for MSCs identity across studies and their inherited heterogeneity. Nowadays, with the emergence of advanced biological techniques and substantial improvements in bio-engineered materials, strategies have been developed to overcome clinical challenges in MSC application. Here in this review, we will discuss the major challenges of MSC therapies in clinical application, the factors impacting the diversity of MSCs, the potential approaches that modify MSC products with the highest therapeutic potential, and finally the usage of MSCs for COVID-19 pandemic disease.
Collapse
|
7
|
Nguyen KG, Vrabel MR, Mantooth SM, Hopkins JJ, Wagner ES, Gabaldon TA, Zaharoff DA. Localized Interleukin-12 for Cancer Immunotherapy. Front Immunol 2020; 11:575597. [PMID: 33178203 PMCID: PMC7593768 DOI: 10.3389/fimmu.2020.575597] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/08/2020] [Indexed: 12/30/2022] Open
Abstract
Interleukin-12 (IL-12) is a potent, pro-inflammatory type 1 cytokine that has long been studied as a potential immunotherapy for cancer. Unfortunately, IL-12's remarkable antitumor efficacy in preclinical models has yet to be replicated in humans. Early clinical trials in the mid-1990's showed that systemic delivery of IL-12 incurred dose-limiting toxicities. Nevertheless, IL-12's pleiotropic activity, i.e., its ability to engage multiple effector mechanisms and reverse tumor-induced immunosuppression, continues to entice cancer researchers. The development of strategies which maximize IL-12 delivery to the tumor microenvironment while minimizing systemic exposure are of increasing interest. Diverse IL-12 delivery systems, from immunocytokine fusions to polymeric nanoparticles, have demonstrated robust antitumor immunity with reduced adverse events in preclinical studies. Several localized IL-12 delivery approaches have recently reached the clinical stage with several more at the precipice of translation. Taken together, localized delivery systems are supporting an IL-12 renaissance which may finally allow this potent cytokine to fulfill its considerable clinical potential. This review begins with a brief historical account of cytokine monotherapies and describes how IL-12 went from promising new cure to ostracized black sheep following multiple on-study deaths. The bulk of this comprehensive review focuses on developments in diverse localized delivery strategies for IL-12-based cancer immunotherapies. Advantages and limitations of different delivery technologies are highlighted. Finally, perspectives on how IL-12-based immunotherapies may be utilized for widespread clinical application in the very near future are offered.
Collapse
Affiliation(s)
- Khue G Nguyen
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Maura R Vrabel
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Siena M Mantooth
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Jared J Hopkins
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Ethan S Wagner
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Taylor A Gabaldon
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - David A Zaharoff
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
8
|
Chulpanova DS, Solovyeva VV, James V, Arkhipova SS, Gomzikova MO, Garanina EE, Akhmetzyanova ER, Tazetdinova LG, Khaiboullina SF, Rizvanov AA. Human Mesenchymal Stem Cells Overexpressing Interleukin 2 Can Suppress Proliferation of Neuroblastoma Cells in Co-Culture and Activate Mononuclear Cells In Vitro. Bioengineering (Basel) 2020; 7:bioengineering7020059. [PMID: 32560387 PMCID: PMC7356660 DOI: 10.3390/bioengineering7020059] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 12/20/2022] Open
Abstract
High-dose recombinant interleukin 2 (IL2) therapy has been shown to be successful in renal cell carcinoma and metastatic melanoma. However, systemic administration of high doses of IL2 can be toxic, causing capillary leakage syndrome and stimulating pro-tumor immune response. One of the strategies to reduce the systemic toxicity of IL2 is the use of mesenchymal stem cells (MSCs) as a vehicle for the targeted delivery of IL2. Human adipose tissue-derived MSCs were transduced with lentivirus encoding IL2 (hADSCs-IL2) or blue fluorescent protein (BFP) (hADSCs-BFP). The proliferation, immunophenotype, cytokine profile and ultrastructure of hADSCs-IL2 and hADSCs-BFP were determined. The effect of hADSCs on activation of peripheral blood mononuclear cells (PBMCs) and proliferation and viability of SH-SY5Y neuroblastoma cells after co-culture with native hADSCs, hADSCs-BFP or hADSCs-IL2 on plastic and Matrigel was evaluated. Ultrastructure and cytokine production by hADSCs-IL2 showed modest changes in comparison with hADSCs and hADSCs-BFP. Conditioned medium from hADSC-IL2 affected tumor cell proliferation, increasing the proliferation of SH-SY5Y cells and also increasing the number of late-activated T-cells, natural killer (NK) cells, NKT-cells and activated T-killers. Conversely, hADSC-IL2 co-culture led to a decrease in SH-SY5Y proliferation on plastic and Matrigel. These data show that hADSCs-IL2 can reduce SH-SY5Y proliferation and activate PBMCs in vitro. However, IL2-mediated therapeutic effects of hADSCs could be offset by the increased expression of pro-oncogenes, as well as the natural ability of hADSCs to promote the progression of some tumors.
Collapse
Affiliation(s)
- Daria S. Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (V.V.S.); (S.S.A.); (M.O.G.); (E.E.G.); (E.R.A.); (L.G.T.); (S.F.K.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, 117997 Moscow, Russia
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (V.V.S.); (S.S.A.); (M.O.G.); (E.E.G.); (E.R.A.); (L.G.T.); (S.F.K.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, 117997 Moscow, Russia
| | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham LE12 5RD, UK;
| | - Svetlana S. Arkhipova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (V.V.S.); (S.S.A.); (M.O.G.); (E.E.G.); (E.R.A.); (L.G.T.); (S.F.K.)
| | - Marina O. Gomzikova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (V.V.S.); (S.S.A.); (M.O.G.); (E.E.G.); (E.R.A.); (L.G.T.); (S.F.K.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, 117997 Moscow, Russia
| | - Ekaterina E. Garanina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (V.V.S.); (S.S.A.); (M.O.G.); (E.E.G.); (E.R.A.); (L.G.T.); (S.F.K.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, 117997 Moscow, Russia
| | - Elvira R. Akhmetzyanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (V.V.S.); (S.S.A.); (M.O.G.); (E.E.G.); (E.R.A.); (L.G.T.); (S.F.K.)
| | - Leysan G. Tazetdinova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (V.V.S.); (S.S.A.); (M.O.G.); (E.E.G.); (E.R.A.); (L.G.T.); (S.F.K.)
| | - Svetlana F. Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (V.V.S.); (S.S.A.); (M.O.G.); (E.E.G.); (E.R.A.); (L.G.T.); (S.F.K.)
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (V.V.S.); (S.S.A.); (M.O.G.); (E.E.G.); (E.R.A.); (L.G.T.); (S.F.K.)
- Correspondence: ; Tel.: +7-905-316-7599
| |
Collapse
|
9
|
LI A, ZHANG T, GAO J. [Progress on utilizing mesenchymal stem cells as cellular delivery system for targeting delivery of as drug/gene for anti-tumor therapy]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2020; 49:20-34. [PMID: 32621413 PMCID: PMC8800717 DOI: 10.3785/j.issn.1008-9292.2020.02.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/15/2020] [Indexed: 06/11/2023]
Abstract
Mesenchymal stem cells (MSCs) have the inherent tumor-homing ability with the attraction of multiple chemokines released by tumor tissues or tumor microenvironments, which can be utilized as promising cellular carriers for targeted delivery of anti-tumor drugs and genes. In most circumstances, large amount of systemicly administrated MSCs will be firstly trapped by lungs, following with re-distribution and homing to tumor tissues after lung clearance. Several approaches like enhanced interactions between chemokines and receptors on MSCs or reducing the retention of MSCs by changes of administration methods are firstly reviewed for improving the homing of MSCs towards tumor tissues. Additionally, the potentials and gains of utilizing MSCs to carry several chemotherapeutics, such as doxorubicin, paclitaxel and gemcitabine are summarized, showing the advantages of overcoming the short half-life and poor tumor targeting of these chemotherapeutics. Moreover, the applications of MSCs to protect and deliver therapeutic genes to tumor sites for selectively tumor cells eliminating or promoting immune system are highlighted. In addition, the potentials of using MSCs for tumor-targeting delivery of diagnostic and therapeutic agents are addressed. We believed that the continuous improvement and optimization of this stem cells-based cellular delivery system will provide a novel delivery strategy and option for tumor treatment.
Collapse
|
10
|
Genetically engineered mesenchymal stem cells: targeted delivery of immunomodulatory agents for tumor eradication. Cancer Gene Ther 2020; 27:854-868. [PMID: 32418986 DOI: 10.1038/s41417-020-0179-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/28/2020] [Accepted: 05/05/2020] [Indexed: 12/17/2022]
Abstract
Cancer immunotherapy emerged as a novel therapeutic option that employs enhanced or amended native immune system to create a robust response against malignant cells. The systemic therapies with immune-stimulating cytokines have resulted in substantial dose-limiting toxicities. Targeted cytokine immunotherapy is being explored to overcome the heterogeneity of malignant cells and tumor cell defense with a remarkable reduction of systemic side effects. Cell-based strategies, such as dendritic cells (DCs), fibroblasts or mesenchymal stem cells (MSCs) seek to minimize the numerous toxic side effects of systemic administration of cytokines for extended periods of time. The usual toxicities comprised of a vascular leak, hypotension, and respiratory insufficiency. Natural and strong tropism of MSCs toward malignant cells made them an ideal systemic delivery vehicle to direct the proposed therapeutic genes to the vicinity of a tumor where their expression could evoke an immune reaction against the tumor. Compared with other methods, the delivery of cytokines via engineered MSCs is safer and renders a more practical, and promising strategy. Large numbers of genes code for cytokines have been utilized to reengineer MSCs as therapeutic cells. This review highlights the recent findings on the cytokine gene therapy for human malignancies by focusing on MSCs application in cancer immunotherapy.
Collapse
|
11
|
Hombach AA, Geumann U, Günther C, Hermann FG, Abken H. IL7-IL12 Engineered Mesenchymal Stem Cells (MSCs) Improve A CAR T Cell Attack Against Colorectal Cancer Cells. Cells 2020; 9:cells9040873. [PMID: 32260097 PMCID: PMC7226757 DOI: 10.3390/cells9040873] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 02/07/2023] Open
Abstract
Chimeric antigen receptor (CAR) redirected T cells are efficacious in the treatment of leukemia/lymphoma, however, showed less capacities in eliminating solid tumors which is thought to be partly due to the lack of cytokine support in the tumor lesion. In order to deliver supportive cytokines, we took advantage of the inherent ability of mesenchymal stem cells (MSCs) to actively migrate to tumor sites and engineered MSCs to release both IL7 and IL12 to promote homeostatic expansion and Th1 polarization. There is a mutual interaction between engineered MSCs and CAR T cells; in presence of CAR T cell released IFN-γ and TNF-α, chronic inflammatory Th2 MSCs shifted towards a Th17/Th1 pattern with IL2 and IL15 release that mutually activated CAR T cells with extended persistence, amplification, killing and protection from activation induced cell death. MSCs releasing IL7 and IL12 were superior over non-modified MSCs in supporting the CAR T cell response and improved the anti-tumor attack in a transplant tumor model. Data demonstrate the first use of genetically modified MSCs as vehicles to deliver immuno-modulatory proteins to the tumor tissue in order to improve the efficacy of CAR T cells in the treatment of solid malignancies.
Collapse
Affiliation(s)
- Andreas A. Hombach
- Center for Molecular Medicine Cologne, Tumor Genetics, University of Cologne, and Department I Internal Medicine, University Hospital Cologne, D-50931 Cologne, Germany;
| | - Ulf Geumann
- Apceth Biopharma GmbH, D-81377 Munich, Germany; (U.G.); (F.G.H.)
| | | | - Felix G. Hermann
- Apceth Biopharma GmbH, D-81377 Munich, Germany; (U.G.); (F.G.H.)
| | - Hinrich Abken
- Center for Molecular Medicine Cologne, Tumor Genetics, University of Cologne, and Department I Internal Medicine, University Hospital Cologne, D-50931 Cologne, Germany;
- Department for Genetic Immunotherapy, Regensburg Center for Interventional Immunology, and University Hospital Regensburg, D-93053 Regensburg, Germany
- Correspondence: ; Tel.: +49-941-944-381-11; Fax: +49-941-944-381-13
| |
Collapse
|
12
|
Chulpanova DS, Kitaeva KV, Tazetdinova LG, James V, Rizvanov AA, Solovyeva VV. Application of Mesenchymal Stem Cells for Therapeutic Agent Delivery in Anti-tumor Treatment. Front Pharmacol 2018; 9:259. [PMID: 29615915 PMCID: PMC5869248 DOI: 10.3389/fphar.2018.00259] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/08/2018] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are non-hematopoietic progenitor cells, which can be isolated from different types of tissues including bone marrow, adipose tissue, tooth pulp, and placenta/umbilical cord blood. There isolation from adult tissues circumvents the ethical concerns of working with embryonic or fetal stem cells, whilst still providing cells capable of differentiating into various cell lineages, such as adipocytes, osteocytes and chondrocytes. An important feature of MSCs is the low immunogenicity due to the lack of co-stimulatory molecules expression, meaning there is no need for immunosuppression during allogenic transplantation. The tropism of MSCs to damaged tissues and tumor sites makes them a promising vector for therapeutic agent delivery to tumors and metastatic niches. MSCs can be genetically modified by virus vectors to encode tumor suppressor genes, immunomodulating cytokines and their combinations, other therapeutic approaches include MSCs priming/loading with chemotherapeutic drugs or nanoparticles. MSCs derived membrane microvesicles (MVs), which play an important role in intercellular communication, are also considered as a new therapeutic agent and drug delivery vector. Recruited by the tumor, MSCs can exhibit both pro- and anti-oncogenic properties. In this regard, for the development of new methods for cancer therapy using MSCs, a deeper understanding of the molecular and cellular interactions between MSCs and the tumor microenvironment is necessary. In this review, we discuss MSC and tumor interaction mechanisms and review the new therapeutic strategies using MSCs and MSCs derived MVs for cancer treatment.
Collapse
Affiliation(s)
- Daria S Chulpanova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Kristina V Kitaeva
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Leysan G Tazetdinova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Albert A Rizvanov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Valeriya V Solovyeva
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
13
|
Promotion of Cell-Based Therapy: Special Focus on the Cooperation of Mesenchymal Stem Cell Therapy and Gene Therapy for Clinical Trial Studies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1119:103-118. [PMID: 30155859 DOI: 10.1007/5584_2018_256] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Regenerative medicine (RM) is a promising new field of medicine that has mobilized several new tools to repair or replace lost or damaged cells or tissues by stimulating natural regenerative mechanisms nearby cell and tissue-based therapy approaches. However, mesenchymal stem cell (MSC) based therapy has been shown to be safe and effective to a certain degree in multiple clinical trial studies (CTSs) of several diseases, in most MSC CTSs the efficacy of treatment has been reported low. Therefore, researchers have focused on efficacy enhancing of MSC to improve migratory and homing, survival, stemness, differentiation and other therapeutic applicable properties by using different approaches. Gene therapy is one of the experimental technique tools that uses genes to change cells for therapeutic and investigation purposes. In this study has been focused on genetically modified MSCs for use in RM with an emphasis on CTSs. We highlight the basic concept of genetic modifications and also discuss recent clinical studies aspects. Recently reviewed studies show that MSC therapy with assistant gene therapy can be used in cancer therapy, heart diseases, Fanconi anemia and several other diseases.
Collapse
|
14
|
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: 84] [Impact Index Per Article: 10.5] [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
|
15
|
Quaranta P, Focosi D, Freer G, Pistello M. Tweaking Mesenchymal Stem/Progenitor Cell Immunomodulatory Properties with Viral Vectors Delivering Cytokines. Stem Cells Dev 2016; 25:1321-41. [PMID: 27476883 DOI: 10.1089/scd.2016.0145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal Stem Cells (MSCs) can be found in various body sites. Their main role is to differentiate into cartilage, bone, muscle, and fat cells to allow tissue maintenance and repair. During inflammation, MSCs exhibit important immunomodulatory properties that are not constitutive, but require activation, upon which they may exert immunosuppressive functions. MSCs are defined as "sensors of inflammation" since they modulate their ability of interfering with the immune system both in vitro and in vivo upon interaction with different factors. MSCs may influence immune responses through different mechanisms, such as direct cell-to-cell contact, release of soluble factors, and through the induction of anergy and apoptosis. Human MSCs are defined as plastic-adherent cells expressing specific surface molecules. Lack of MHC class II antigens makes them appealing as allogeneic tools for the therapy of both autoimmune diseases and cancer. MSC therapeutic potential could be highly enhanced by the expression of exogenous cytokines provided by transduction with viral vectors. In this review, we attempt to summarize the results of a great number of in vitro and in vivo studies aimed at improving the ability of MSCs as immunomodulators in the therapy of autoimmune, degenerative diseases and cancer. We will also compare results obtained with different vectors to deliver heterologous genes to these cells.
Collapse
Affiliation(s)
- Paola Quaranta
- 1 Department of Translational Research and New Technologies in Medicine and Surgery, Virology Section and Retrovirus Center, University of Pisa , Pisa, Italy
| | - Daniele Focosi
- 2 North-Western Tuscany Blood Bank, Pisa University Hospital , Pisa, Italy
| | - Giulia Freer
- 1 Department of Translational Research and New Technologies in Medicine and Surgery, Virology Section and Retrovirus Center, University of Pisa , Pisa, Italy .,3 Virology Unit, Pisa University Hospital , Pisa, Italy
| | - Mauro Pistello
- 1 Department of Translational Research and New Technologies in Medicine and Surgery, Virology Section and Retrovirus Center, University of Pisa , Pisa, Italy .,3 Virology Unit, Pisa University Hospital , Pisa, Italy
| |
Collapse
|
16
|
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
|
17
|
Sun Z, Li T, Wen H, Wang H, Ji W, Ma Y. Immunological effect induced by mesenchymal stem cells in a rat liver transplantation model. Exp Ther Med 2015; 10:401-406. [PMID: 26622328 DOI: 10.3892/etm.2015.2551] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 11/12/2014] [Indexed: 12/17/2022] Open
Abstract
The aim of the present study was to investigate the immunological effect induced by bone marrow mesenchymal stem cells (MSCs) in rats that had undergone an orthotopic liver transplantation (OLT). MSCs were isolated and cultured from the bone marrow tissue of Lewis rats. In total, 42 rat OLT models were established and equally distributed into three groups. Group A received an OLT only, group B were also intramuscularly injected with tacrolimus (FK506), while group C were not only administered FK506, but also received MSCs. On day 7 post-surgery, the blood levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST) and total bilirubin (TBIL) were measured. In addition, pathological changes were observed in the liver, levels of immune cytokines, including transforming growth factor (TGF)-α1, interleukin (IL)-10 and IL-12, were determined using immunohistochemistry, MSC homing was assessed and the survival times of the patients were recorded. Liver function, as assessed by the levels of ALT, AST and TBIL, was shown to improve in group C when compared with groups B and A (both P<0.01). In addition, survival analysis revealed that the survival times in groups B (median, 44 days) and C (median, 63 days) were significantly longer compared with group A (median, 11 days; both P<0.01). The survival rate of group C was also higher compared with group B (P<0.01). Pathological examination demonstrated strong acute rejection in group A, a mild acute rejection in group B and the mildest reaction in group C. In addition, immunohistochemistry revealed that TGF-α1 and IL-10 expression was stronger in groups C and B, with group C exhibiting more significant expression than group B. By contrast, expression levels of IL-12 in groups A, B and C were positive, weak-positive and negative, respectively. Therefore, postoperative immunosuppression induced by MSCs is important for the alleviation of immune rejection from recipient-to-graft, and may induce immune tolerance in rat OLT models.
Collapse
Affiliation(s)
- Zhenqiang Sun
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, Ürümqi, Xinjiang 830011, P.R. China ; Research Laboratory of Disease Genomics, Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Tao Li
- Department of Digestive and Vascular Surgery Center, First Affiliated Hospital, Xinjiang Medical University, Ürümqi, Xinjiang 830054, P.R. China
| | - Hao Wen
- Department of Digestive and Vascular Surgery Center, First Affiliated Hospital, Xinjiang Medical University, Ürümqi, Xinjiang 830054, P.R. China ; Xinjiang Organ Transplant Research Institute, First Affiliated Hospital, Xinjiang Medical University, Ürümqi, Xinjiang 830054, P.R. China
| | - Haijiang Wang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, Ürümqi, Xinjiang 830011, P.R. China
| | - Weizheng Ji
- Oncology Center, First Affiliated Hospital, Xinjiang Medical University, Ürümqi, Xinjiang 830054, P.R. China
| | - Yan Ma
- Stem Cell Laboratory of Medical Research Center, First Affiliated Hospital, Xinjiang Medical University, Ürümqi, Xinjiang 830054, P.R. China
| |
Collapse
|