1
|
Curylova L, Staniczkova Zambo I, Neradil J, Kyr M, Jurackova N, Pavlova S, Polaskova K, Mudry P, Sterba J, Veselska R, Skoda J. Dysregulation of the p53 pathway provides a therapeutic target in aggressive pediatric sarcomas with stem-like traits. Cell Oncol (Dordr) 2024; 47:2317-2334. [PMID: 39630408 DOI: 10.1007/s13402-024-01020-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2024] [Indexed: 01/11/2025] Open
Abstract
PURPOSE Pediatric sarcomas are bone and soft tissue tumors that often exhibit high metastatic potential and refractory stem-like phenotypes, resulting in poor outcomes. Aggressive sarcomas frequently harbor a disrupted p53 pathway. However, whether pediatric sarcoma stemness is associated with abrogated p53 function and might be attenuated via p53 reactivation remains unclear. METHODS We utilized a unique panel of pediatric sarcoma models and tumor tissue cohorts to investigate the correlation between the expression of stemness-related transcription factors, p53 pathway dysregulations, tumorigenicity in vivo, and clinicopathological features. TP53 mutation status was assessed by next-generation sequencing. Major findings were validated via shRNA-mediated silencing and functional assays. The p53 pathway-targeting drugs were used to explore the effects and selectivity of p53 reactivation against sarcoma cells with stem-like traits. RESULTS We found that highly tumorigenic stem-like sarcoma cells exhibit dysregulated p53, making them vulnerable to drugs that restore wild-type p53 activity. Immunohistochemistry of mouse xenografts and human tumor tissues revealed that p53 dysregulations, together with enhanced expression of the stemness-related transcription factors SOX2 or KLF4, are crucial features in pediatric osteosarcoma, rhabdomyosarcoma, and Ewing's sarcoma development. p53 dysregulation appears to be an important step for sarcoma cells to acquire a fully stem-like phenotype, and p53-positive pediatric sarcomas exhibit a high frequency of early metastasis. Importantly, reactivating p53 signaling via MDM2/MDMX inhibition selectively induces apoptosis in aggressive, stem-like Ewing's sarcoma cells while sparing healthy fibroblasts. CONCLUSIONS Our results indicate that restoring canonical p53 activity provides a promising strategy for developing improved therapies for pediatric sarcomas with unfavorable stem-like traits.
Collapse
Affiliation(s)
- Lucie Curylova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, 656 91, Czech Republic
| | - Iva Staniczkova Zambo
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, 656 91, Czech Republic
- 1st Department of Pathology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jakub Neradil
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, 656 91, Czech Republic
| | - Michal Kyr
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 613 00, Czech Republic
| | - Nicola Jurackova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- 1st Department of Pathology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sarka Pavlova
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, 625 00, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, and Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 625 00, Czech Republic
| | - Kristyna Polaskova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 613 00, Czech Republic
| | - Peter Mudry
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 613 00, Czech Republic
| | - Jaroslav Sterba
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 613 00, Czech Republic
| | - Renata Veselska
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, 656 91, Czech Republic
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, Brno, 656 91, Czech Republic.
| |
Collapse
|
2
|
Alhozeel B, Pandey SK, Shteinfer-Kuzmine A, Santhanam M, Shoshan-Barmatz V. Silencing the Mitochondrial Gatekeeper VDAC1 as a Potential Treatment for Bladder Cancer. Cells 2024; 13:627. [PMID: 38607066 PMCID: PMC11012128 DOI: 10.3390/cells13070627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 04/13/2024] Open
Abstract
The strategy for treating bladder cancer (BC) depends on whether there is muscle invasion or not, with the latter mostly treated with intravesical therapy, such as with bacillus Calmette-Guérin (BCG). However, BCG treatment is unsuccessful in 70% of patients, who are then subjected to radical cystectomy. Although immune-checkpoint inhibitors have been approved as a second-line therapy for a subset of BC patients, these have failed to meet primary endpoints in clinical trials. Thus, it is crucial to find a new treatment. The mitochondrial gatekeeper protein, the voltage-dependent anion channel 1 (VDAC1), mediates metabolic crosstalk between the mitochondria and cytosol and is involved in apoptosis. It is overexpressed in many cancer types, as shown here for BC, pointing to its significance in high-energy-demanding cancer cells. The BC cell lines UM-UC3 and HTB-5 express high VDAC1 levels compared to other cancer cell lines. VDAC1 silencing in these cells using siRNA that recognizes both human and mouse VDAC1 (si-m/hVDAC1-B) reduces cell viability, mitochondria membrane potential, and cellular ATP levels. Here, we used two BC mouse models: subcutaneous UM-UC3 cells and chemically induced BC using the carcinogen N-Butyl-N-(4-hydroxybutyl) nitrosamine (BBN). Subcutaneous UM-UC3-derived tumors treated with si-m/hVDAC1 showed inhibited tumor growth and reprogrammed metabolism, as reflected in the reduced expression of metabolism-related proteins, including Glut1, hexokinase, citrate synthase, complex-IV, and ATP synthase, suggesting reduced metabolic activity. Furthermore, si-m/hVDAC1-B reduced the expression levels of cancer-stem-cell-related proteins (cytokeratin-14, ALDH1a), modifying the tumor microenvironment, including decreased angiogenesis, extracellular matrix, tumor-associated macrophages, and inhibited epithelial-mesenchymal transition. The BBN-induced BC mouse model showed a clear carcinoma, with damaged bladder morphology and muscle-invasive tumors. Treatment with si-m/hVDAC1-B encapsulated in PLGA-PEI nanoparticles that were administered intravesically directly to the bladder showed a decreased tumor area and less bladder morphology destruction and muscle invasion. Overall, the obtained results point to the potential of si-m/hVDAC1-B as a possible therapeutic tool for treating bladder cancer.
Collapse
Affiliation(s)
- Belal Alhozeel
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (B.A.); (S.K.P.); (M.S.)
| | - Swaroop Kumar Pandey
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (B.A.); (S.K.P.); (M.S.)
| | - Anna Shteinfer-Kuzmine
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Manikandan Santhanam
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (B.A.); (S.K.P.); (M.S.)
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (B.A.); (S.K.P.); (M.S.)
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| |
Collapse
|
3
|
Martin J, Islam F. Detection and Isolation of Cancer Stem Cells. CANCER STEM CELLS: BASIC CONCEPT AND THERAPEUTIC IMPLICATIONS 2023:45-69. [DOI: 10.1007/978-981-99-3185-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
The Role of Tumor Microenvironment in Regulating the Plasticity of Osteosarcoma Cells. Int J Mol Sci 2022; 23:ijms232416155. [PMID: 36555795 PMCID: PMC9788144 DOI: 10.3390/ijms232416155] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Osteosarcoma (OS) is a malignancy that is becoming increasingly common in adolescents. OS stem cells (OSCs) form a dynamic subset of OS cells that are responsible for malignant progression and chemoradiotherapy resistance. The unique properties of OSCs, including self-renewal, multilineage differentiation and metastatic potential, 149 depend closely on their tumor microenvironment. In recent years, the likelihood of its dynamic plasticity has been extensively studied. Importantly, the tumor microenvironment appears to act as the main regulatory component of OS cell plasticity. For these reasons aforementioned, novel strategies for OS treatment focusing on modulating OS cell plasticity and the possibility of modulating the composition of the tumor microenvironment are currently being explored. In this paper, we review recent studies describing the phenomenon of OSCs and factors known to influence phenotypic plasticity. The microenvironment, which can regulate OSC plasticity, has great potential for clinical exploitation and provides different perspectives for drug and treatment design for OS.
Collapse
|
5
|
Duan JJ, Wang D, Cai J, Chen JJ, Zheng XX, Chen TQ, Wang J, Zhang X, Yang QK, Yu SC. An aldehyde dehydrogenase 1A3 inhibitor attenuates the metastasis of human colorectal cancer. Cancer Lett 2022; 536:215662. [PMID: 35331786 DOI: 10.1016/j.canlet.2022.215662] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/03/2022] [Accepted: 03/18/2022] [Indexed: 12/19/2022]
Abstract
Metastasis is the leading cause of death for patients with colorectal cancer (CRC). The development of therapeutic regimens that selectively inhibit the biological processes involved in CRC cell dissemination is important. We used multiple Affymetrix DNA microarray hybridization datasets to identify genes related to metastasis and have significant prognostic value for patients with CRC. Quantitative real-time PCR, immunofluorescent and immunohistochemical staining were used to evaluate mRNA and protein expression. The function of aldehyde dehydrogenase 1A3 (ALDH1A3) in invasion was assessed by performing transwell assays and animal experiments. Real-time PCR, luciferase reporter assays, and western blotting were used to identify the genes regulated by ALDH1A3. Molecular docking, MTS assays, cellular thermal shift assays, isothermal titration calorimetry, microscale thermophoresis, and enzymatic activity assays were used to screen and verify the efficacy of the ALDH1A3-specific inhibitor YD1701 (dibenzo-30-crown10-ether). Finally, subcutaneous or orthotopic xenograft models were established to investigate the therapeutic potential of YD1701. Human ALDH1A3 was identified to correlate with a metastatic phenotype in CRC cells and a poor patient prognosis. Moreover, ALDH1A3 upregulated the expression of ZEB1 and SNAI2 by inhibiting miR-200 family members. The ALDH1A3-specific inhibitor YD1701 was screened, attenuated the invasion of CRC cells in vitro, and prolonged the survival of mice bearing subcutaneous or orthotopic xenografts. Our results show that ALDH1A3 promotes invasion and metastasis via the miR-200-ZEB1/SANI2 axis and is thus a plausible marker for predicting CRC progression. Inhibiting ALDH1A3 with the identified compound YD1701 might represent an effective therapeutic approach to prevent the metastasis of CRC.
Collapse
Affiliation(s)
- Jiang-Jie Duan
- Department of Stem Cell and Regenerative Medicine, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Di Wang
- Department of Stem Cell and Regenerative Medicine, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiao Cai
- Department of Stem Cell and Regenerative Medicine, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jun-Jie Chen
- Department of Stem Cell and Regenerative Medicine, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiao-Xia Zheng
- Department of Stem Cell and Regenerative Medicine, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Tian-Qing Chen
- Department of Stem Cell and Regenerative Medicine, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; School of Pharmacy, Shanxi Medical University, Taiyuan, 030002, Shanxi, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiao Zhang
- Department of Stem Cell and Regenerative Medicine, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Qing-Kai Yang
- Institute of Cancer Stem Cell, Cancer Center, Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China.
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
6
|
Pan S, Cesarek M, Godoy C, Co CM, Schindler C, Padilla K, Haskell A, Barreda H, Story C, Poole R, Dabney A, Gregory CA. Morpholino-driven blockade of Dkk-1 in osteosarcoma inhibits bone damage and tumour expansion by multiple mechanisms. Br J Cancer 2022; 127:43-55. [PMID: 35277659 PMCID: PMC9276700 DOI: 10.1038/s41416-022-01764-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 02/02/2022] [Accepted: 02/15/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Background
Osteosarcoma (OS) is the most common primary bone malignancy. Chemotherapy plays an essential role in OS treatment, potentially doubling 5-year event-free survival if tumour necrosis can be stimulated. The canonical Wnt inhibitor Dickkopf-1 (Dkk-1) enhances OS survival in part through upregulation of aldehyde-dehydrogenase-1A1 which neutralises reactive oxygen species originating from nutritional stress and chemotherapeutic challenge.
Methods
A vivo morpholino (DkkMo) was employed to block the expression of Dkk-1 in OS cells. Cell mitosis, gene expression and bone destruction were measured in vitro and in vivo in the presence and absence of doxorubicin (DRB).
Results
DkkMo reduced the expression of Dkk-1 and Aldh1a1, reduced expansion of OS tumours, preserved bone volume and architecture and stimulated tumour necrosis. This was observed in the presence or absence of DRB.
Conclusion
These results indicate that administration of DkkMo with or without chemotherapeutics can substantially improve OS outcome with respect to tumour expansion and osteolytic corruption of bone in experimental OS model.
Collapse
|
7
|
HER2-targeted antibody-drug conjugate induces host immunity against cancer stem cells. Cell Chem Biol 2021; 28:610-624.e5. [PMID: 33711257 DOI: 10.1016/j.chembiol.2021.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/15/2020] [Accepted: 02/17/2021] [Indexed: 11/23/2022]
Abstract
We previously tested HER2-targeted antibody-drug conjugates (ADCs) in immunocompromised (SCID) mice, precluding evaluation of host immunity, impact on cancer stem cells (CSCs), and potential benefit when combined with PD-L1 blockade. In this study, we tested HER2-targeted ADC in two immunocompetent mouse tumor models. HER2-targeted ADC specifically inhibited the growth of HER2-expressing tumors, prolonged animal survival, and reduced HER2+ and PD-L1+ cells. ADC + anti-PD-L1 antibody augmented therapeutic efficacy, modulated immune gene signatures, increased the number and function of CD3+ and CD19+ tumor-infiltrating lymphocytes (TILs), induced tumor antigen-specific immunological memory, stimulated B cell activation, differentiation, and IgG1 production both systemically and in the tumor microenvironment. In addition, ADC therapy modulated T cell subsets and their activation in TILs. Furthermore, HER2-targeted ADC reduced the number and tumorigenicity of ALDHhi CSCs. This study demonstrates that HER2-targeted ADC effectively targets ALDHhi CSCs and this effect is augmented by co-administration of anti-PD-L1 antibody.
Collapse
|
8
|
Lin J, Wang X, Wang X, Wang S, Shen R, Yang Y, Xu J, Lin J. Hypoxia increases the expression of stem cell markers in human osteosarcoma cells. Oncol Lett 2021; 21:217. [PMID: 33613706 PMCID: PMC7856697 DOI: 10.3892/ol.2021.12478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant tumor of bone. It is a common phenomenon that osteosarcoma cells have a hypoxic microenvironment. Hypoxia can dedifferentiate cells of several malignant tumor types into stem cell-like phenotypes. However, the role of hypoxia in stemness induction and the expression of cancer stem cell (CSC) markers in human osteosarcoma cells has not been reported. The present study examined the effects of hypoxia on stem-like cells in the human osteosarcoma MNNG/HOS cells. Under the incubation with 1% oxygen, the expression of CSCs markers (Oct-4, Nanog and CD133) in MNNG/HOS cells were increased. Moreover, MNNG/HOS cells cultured under hypoxic conditions were more likely to proliferate into spheres and resulted in larger xenograft tumor. Hypoxia also increased the mRNA and protein levels of hypoxia-inducible factor (HIF)-1α. Then rapamycin was used, which has been shown to lower HIF-1α protein level, to inhibit the hypoxic response. Rapamycin suppressed the expression of HIF-1α protein and CSCs markers (Oct4, Nanog and CD133) in MNNG/HOS cells. In addition, pretreatment with rapamycin reduced the efficiency of MNNG/HOS cells in forming spheres and xenograft tumors. The results demonstrated that hypoxia (1% oxygen) can dedifferentiate some of the MNNG/HOS cells into stem cell-like phenotypes, and that the mTOR signaling pathway participates in this process via regulating the expression of HIF-1α protein.
Collapse
Affiliation(s)
- Jinluan Lin
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Xinwu Wang
- Department of Orthopedics, The First Hospital of Putian City, Putian, Fujian 351100, P.R. China
| | - Xinwen Wang
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
- Department of Orthopedics, The People's Hospital of Jiangmen, Jiangmen, Guangdong 529051, P.R. China
| | - Shenglin Wang
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Rongkai Shen
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Yanbing Yang
- Department of Radiology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Jianyong Xu
- Department of Orthopedics, The People's Hospital of Guixi, Guixi, Jiangxi 335400, P.R. China
| | - Jianhua Lin
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| |
Collapse
|
9
|
Singla A, Wang J, Yang R, Geller DS, Loeb DM, Hoang BH. Wnt Signaling in Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:125-139. [PMID: 32767238 DOI: 10.1007/978-3-030-43085-6_8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Wnt molecules are a class of cysteine-rich secreted glycoproteins that participate in various developmental events during embryogenesis and adult tissue homeostasis. Since its discovery in 1982, the roles of Wnt signaling have been established in various key regulatory systems in biology. Wnt signals exert pleiotropic effects, including mitogenic stimulation, cell fate specification, and differentiation. The Wnt signaling pathway in humans has been shown to be involved in a wide variety of disorders including colon cancer, sarcoma, coronary artery disease, tetra-amelia, Mullerian duct regression, eye vascular defects, and abnormal bone mass. The canonical Wnt pathway functions by regulating the function of the transcriptional coactivator β-catenin, whereas noncanonical pathways function independent of β-catenin. Although the role of Wnt signaling is well established in epithelial malignancies, its role in mesenchymal tumors is more controversial. Some studies have suggested that Wnt signaling plays a pro-oncogenic role in various sarcomas by driving cell proliferation and motility; however, others have reported that Wnt signaling acts as a tumor suppressor by committing tumor cells to differentiate into a mature lineage. Wnt signaling pathway also plays an important role in regulating cancer stem cell function. In this review, we will discuss Wnt signaling pathway and its role in osteosarcoma.
Collapse
Affiliation(s)
- Amit Singla
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jichuan Wang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Musculoskeletal Tumor Center, Beijing Key Laboratory for Musculoskeletal Tumors, Peking University People's Hospital, Beijing, China
| | - Rui Yang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David S Geller
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David M Loeb
- Departments of Pediatrics and Developmental and Molecular Biology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bang H Hoang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
10
|
Fujiwara S, Kawamoto T, Kawakami Y, Koterazawa Y, Hara H, Takemori T, Kitayama K, Yahiro S, Kakutani K, Matsumoto T, Matsushita T, Niikura T, Koyanagi-Aoi M, Aoi T, Kuroda R, Akisue T. Acquisition of cancer stem cell properties in osteosarcoma cells by defined factors. Stem Cell Res Ther 2020; 11:429. [PMID: 33008481 PMCID: PMC7532109 DOI: 10.1186/s13287-020-01944-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 09/20/2020] [Indexed: 12/18/2022] Open
Abstract
Background Cancer stem cells (CSCs) are considered to be responsible for tumor initiation, formation, and poor prognosis of cancer patients. However, the rarity of CSCs in clinical samples makes it difficult to elucidate characteristics of CSCs, especially in osteosarcoma (OS). The aim of this study is to verify whether it is possible to generate CSC-like cells by transducing defined factors into an OS cell line. Methods We retrovirally transduced the Octamer-binding transcription factor 3/4 (OCT3/4), Kruppel-like factor 4 (KLF4), and SRY-box transcription factor 2 (SOX2) genes into the MG-63 human OS cell line (MG-OKS). Parental and GFP-transduced MG-63 cells were used as negative control. We assessed the properties of the generated cells in vitro and in vivo. Multiple comparisons among groups were made using a one-way analysis of variance (ANOVA) followed by post hoc testing with Tukey’s procedure. Results MG-OKS cells in vitro exhibited the significantly increased mRNA expression levels of CSC markers (CD24, CD26, and CD133), decreased cell growth, increased chemoresistance and cell migration, and enhanced sphere formation. Notably, MG-OKS cells cultured under osteogenic differentiation conditions showed strongly positive staining for both Alizarin Red S and alkaline phosphatase, indicating osteogenesis of the cells. Gene ontology analysis of microarray data revealed significant upregulation of epidermal-related genes. Tumors derived from MG-OKS cells in vivo were significantly larger than those from other cells in μCT analysis, and immunohistochemical staining showed that Ki-67, osteocalcin, and HIF-1α-positive cells were more frequently detected in the MG-OKS-derived tumors. Conclusions In this study, we successfully generated OS CSC-like cells with significantly enhanced CSC properties following transduction of defined factors.
Collapse
Affiliation(s)
- Shuichi Fujiwara
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Teruya Kawamoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan. .,Division of Orthopaedic Surgery, Kobe University Hospital International Clinical Cancer Research Center, Kobe, Japan.
| | - Yohei Kawakami
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yasufumi Koterazawa
- Department of iPS Cell Applications, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan.,Division of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hitomi Hara
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Toshiyuki Takemori
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kazumichi Kitayama
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Shunsuke Yahiro
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kenichiro Kakutani
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takehiko Matsushita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takahiro Niikura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Michiyo Koyanagi-Aoi
- Department of iPS Cell Applications, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan.,Center for Human Resource development for Regenerative Medicine, Kobe University Hospital, Kobe, Japan
| | - Takashi Aoi
- Department of iPS Cell Applications, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan.,Center for Human Resource development for Regenerative Medicine, Kobe University Hospital, Kobe, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Toshihiro Akisue
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.,Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
11
|
Mardani A, Gheytanchi E, Mousavie SH, Madjd Jabari Z, Shooshtarizadeh T. Clinical Significance of Cancer Stem Cell Markers CD133 and CXCR4 in Osteosarcomas. Asian Pac J Cancer Prev 2020; 21:67-73. [PMID: 31983166 PMCID: PMC7294029 DOI: 10.31557/apjcp.2020.21.1.67] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Indexed: 12/16/2022] Open
Abstract
Objective: Osteosarcomas (OS) is one the most common primary bone malignancy in humans with the lungs metastasis in most cases. Metastasis and recurrence of OS is attributed to cancer stem cells (CSCs). Our study aimed to evaluate the clinical significance of CD133 and C-X-C chemokine receptor type 4 (CXCR4) as the frequently applied markers for CSCs in OS patients. Methods: In this cross-sectional, a total of 50 tissue samples from the patients with primary OS were immunohistochemically examined to detect the expression of CD133 and CXCR4. The associations of the relative expression and clinical significance of each marker were also evaluated. Results: High level expression of CD133 was detected in 26% of OS patient tissues. Of the 12 patients who showed lung metastasis, 5 cases showed high expression of CD133 with marginal trend correlation (P=0.06). No significant correlation was observed between CD133 expression and clinicopathological factors. Only 36% of cases showed CXCR4 expression which was not significantly correlated with gender, age, tumor size, necrosis, stage and metastasis (P>0.05). Clinically, patients with concomitant CD133/CXCR4 expression had significant association with lung metastasis (P=0.05). Conclusion: Our findings showed that concomitant expression of CSC markers CD133/CXCR4 might had a synergistic effect on the OS poor prognosis. These markers could be considered as potential therapeutic candidates of OS targeted therapy.
Collapse
Affiliation(s)
- Azam Mardani
- Department of Pathology, Iran University of Medical Science, Tehran, Iran
| | - Elmira Gheytanchi
- Oncopathology Research Center, Iran University of Medical Science, Tehran, Iran
| | - Seyed Hamzeh Mousavie
- Department of Surgery, Rasool-Akram Hospital, Iran University of Medical Science, Tehran, Iran
| | - Zahra Madjd Jabari
- Oncopathology Research Center, Iran University of Medical Science, Tehran, Iran
| | | |
Collapse
|
12
|
Cancer Stem Cells and Osteosarcoma: Opportunities and Limitations. Tech Orthop 2019. [DOI: 10.1097/bto.0000000000000408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
|
13
|
Izadpanah S, Shabani P, Aghebati-Maleki A, Baghbanzadeh A, Fotouhi A, Bisadi A, Aghebati-Maleki L, Baradaran B. Prospects for the involvement of cancer stem cells in the pathogenesis of osteosarcoma. J Cell Physiol 2019; 235:4167-4182. [PMID: 31709547 DOI: 10.1002/jcp.29344] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 08/26/2019] [Indexed: 12/31/2022]
Abstract
Osteosarcoma (OS) is one of the most common bone tumors in children and adolescents that cause a high rate of mortality in this age group and tends to be metastatic, in spite of chemotherapy and surgery. The main reason for this can be returned to a small group of malignant cells called cancer stem cells (CSCs). OS-CSCs play a key role in the resistance to treatment and relapse and metastasis through self-renewal and differentiation abilities. In this review, we intend to go through the different aspects of this malignant disease, including the cancer stem cell-phenotype, methods for isolating CSCs, signaling pathways, and molecular markers in this disease, and drugs showing resistance in treatment efforts of OS.
Collapse
Affiliation(s)
- Sama Izadpanah
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parastoo Shabani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Fotouhi
- Department of Orthopedic Surgery, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Bisadi
- Department of Orthopedic Surgery, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Schiavone K, Garnier D, Heymann MF, Heymann D. The Heterogeneity of Osteosarcoma: The Role Played by Cancer Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1139:187-200. [PMID: 31134502 DOI: 10.1007/978-3-030-14366-4_11] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Osteosarcoma is the most common bone sarcoma and is one of the cancer entities characterized by the highest level of heterogeneity in humans. This heterogeneity takes place not only at the macroscopic and microscopic levels, with heterogeneous micro-environmental components, but also at the genomic, transcriptomic and epigenetic levels. Recent investigations have revealed the existence in osteosarcoma of cancer cells with stemness properties. Cancer stem cells are characterized by their specific phenotype and low cycling capacity, and are linked to drug resistance, tumour growth and the metastatic process. In addition, cancer stem cells contribute to the enrichment of tumour heterogeneity. The present manuscript will describe the main characteristic features of cancer stem cells in osteosarcoma and will discuss their impact on maintaining tumour heterogeneity. Their clinical implications will also be briefly addressed.
Collapse
Affiliation(s)
- Kristina Schiavone
- INSERM, European Associated Laboratory "Sarcoma Research Unit", Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
| | - Delphine Garnier
- INSERM, Institut de Cancérologie de l'Ouest, CRCINA, Université de Nantes, Université d'Angers, Saint Herblain, France
| | - Marie-Francoise Heymann
- INSERM, Institut de Cancérologie de l'Ouest, CRCINA, Université de Nantes, Université d'Angers, Saint Herblain, France
| | - Dominique Heymann
- INSERM, European Associated Laboratory "Sarcoma Research Unit", Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.
- INSERM, Institut de Cancérologie de l'Ouest, CRCINA, Université de Nantes, Université d'Angers, Saint Herblain, France.
| |
Collapse
|
15
|
TRAIL responses are enhanced by nuclear export inhibition in osteosarcoma. Biochem Biophys Res Commun 2019; 517:383-389. [PMID: 31362889 DOI: 10.1016/j.bbrc.2019.07.047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 07/17/2019] [Indexed: 01/10/2023]
Abstract
Tumour necrosis factor-related apoptosis inducing ligand (TRAIL) is a promising anti-tumour agent that induces apoptosis of malignant cells through activation of death receptors. Death receptor agonistic antibodies are in clinical trials as TRAIL-mimetics, however, along with TRAIL monotherapy, there is limited efficacy due to the rapid emergence of TRAIL resistance, or due to existing TRAIL-insensitive disease. TRAIL-sensitisers, which enhance TRAIL activity or overcome TRAIL resistance, may facilitate death receptor agonists as viable anti-tumour strategies. In this study we demonstrate that the nuclear export inhibitor Leptomycin B, is a potent in vitro TRAIL-sensitiser in osteosarcoma cell lines. Leptomycin B works synergistically with both TRAIL and death receptor 5 agonistic antibodies to induce apoptosis in TRAIL sensitive cell lines. Further, Leptomycin B sensitises TRAIL-insensitive cell lines to TRAIL and death receptor agonistic antibody mediated apoptosis. We also confirmed that aldehyde dehydrogenase (ALDH) positive cells are not resistant to the apoptotic effects of TRAIL and Leptomycin B, an important observation since ALDH positive cells can have enhanced tumorigenicity and are implicated in disease recurrence and metastasis. The nuclear export pathway in combination with death receptor agonists, is a potential therapeutic strategy in osteosarcoma and warrants further research on clinically relevant selective inhibitors of nuclear export.
Collapse
|
16
|
ABCG1 and Pgp identify drug resistant, self-renewing osteosarcoma cells. Cancer Lett 2019; 453:142-157. [DOI: 10.1016/j.canlet.2019.03.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/11/2018] [Accepted: 03/04/2019] [Indexed: 12/20/2022]
|
17
|
Fourneaux B, Bourdon A, Dadone B, Lucchesi C, Daigle SR, Richard E, Laroche-Clary A, Le Loarer F, Italiano A. Identifying and targeting cancer stem cells in leiomyosarcoma: prognostic impact and role to overcome secondary resistance to PI3K/mTOR inhibition. J Hematol Oncol 2019; 12:11. [PMID: 30683135 PMCID: PMC6347793 DOI: 10.1186/s13045-018-0694-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 12/27/2018] [Indexed: 12/22/2022] Open
Abstract
Background Leiomyosarcoma (LMS) is one of the most frequent soft tissue sarcoma subtypes and is characterized by a consistent deregulation of the PI3K/mTOR pathway. Cancer stem cells (CSCs) have been poorly studied in soft tissue sarcomas. In this study, we aimed to evaluate the association between CSCs, the outcome of LMS patients, and the resistance to PI3K/mTOR pathway inhibition. Methods We investigated the relationships between aldehyde dehydrogenase 1 (ALDH1) expression, a cancer stem cell marker, and the outcome of LMS patients in two independent cohorts. We assessed the impact of CSCs in resistance to PI3K/mTOR pathway inhibition using LMS cell lines, a xenograft mouse model, and human tumor samples. Results We found that enhanced ALDH1 activity is a hallmark of LMS stem cells and is an independent prognostic factor. We also identified that secondary resistance to PI3K/mTOR pathway inhibition was associated with the expansion of LMS CSCs. Interestingly, we found that EZH2 inhibition, a catalytic component of polycomb repressive complex which plays a critical role in stem cell maintenance, restored sensitivity to PI3K/mTOR pathway inhibition. Importantly, we confirmed the clinical relevance of our findings by analyzing tumor samples from patients who showed secondary resistance after treatment with a PI3Kα inhibitor. Conclusions Altogether, our findings suggest that CSCs have a strong impact on the outcome of patients with LMS and that combining PI3K/mTOR and EZH2 inhibitors may represent a promising strategy in this setting. Electronic supplementary material The online version of this article (10.1186/s13045-018-0694-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Benjamin Fourneaux
- Université de Bordeaux, Bordeaux, France.,Institut National de la Santé et de la Recherche Medicale (INSERM) U1218, Institut Bergonié, 229 Cours de l'Argonne, 33000, Bordeaux, France
| | - Aurélien Bourdon
- Institut National de la Santé et de la Recherche Medicale (INSERM) U1218, Institut Bergonié, 229 Cours de l'Argonne, 33000, Bordeaux, France
| | | | - Carlo Lucchesi
- Institut National de la Santé et de la Recherche Medicale (INSERM) U1218, Institut Bergonié, 229 Cours de l'Argonne, 33000, Bordeaux, France
| | | | - Elodie Richard
- Université de Bordeaux, Bordeaux, France.,Institut National de la Santé et de la Recherche Medicale (INSERM) U1218, Institut Bergonié, 229 Cours de l'Argonne, 33000, Bordeaux, France
| | - Audrey Laroche-Clary
- Université de Bordeaux, Bordeaux, France.,Institut National de la Santé et de la Recherche Medicale (INSERM) U1218, Institut Bergonié, 229 Cours de l'Argonne, 33000, Bordeaux, France
| | | | - Antoine Italiano
- Institut National de la Santé et de la Recherche Medicale (INSERM) U1218, Institut Bergonié, 229 Cours de l'Argonne, 33000, Bordeaux, France. .,Department of Medical Oncology, Institut Bergonié, Bordeaux, France.
| |
Collapse
|
18
|
Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States. Cancers (Basel) 2018; 10:cancers10120499. [PMID: 30544833 PMCID: PMC6316808 DOI: 10.3390/cancers10120499] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/27/2018] [Accepted: 12/04/2018] [Indexed: 01/10/2023] Open
Abstract
Oncogenic properties, along with the metabolic reprogramming necessary for tumour growth and motility, are acquired by cancer cells. Thus, tumour metabolism is becoming a target for cancer therapy. Here, cancer cell metabolism was tackled by silencing the expression of voltage-dependent anion channel 1 (VDAC1), a mitochondrial protein that controls cell energy, as well as metabolic and survival pathways and that is often over-expressed in many cancers. We demonstrated that silencing VDAC1 expression using human-specific siRNA (si-hVDAC1) inhibited cancer cell growth, both in vitro and in mouse xenograft models of human glioblastoma (U-87MG), lung cancer (A549), and triple negative breast cancer (MDA-MB-231). Importantly, treatment with si-hVDAC1 induced metabolic rewiring of the cancer cells, reversing their oncogenic properties and diverting them towards differentiated-like cells. The si-hVDAC1-treated residual “tumour” showed reprogrammed metabolism, decreased proliferation, inhibited stemness and altered expression of genes and proteins, leading to cell differentiation toward less malignant lineages. These VDAC1 depletion-mediated effects involved alterations in master transcription factors associated with cancer hallmarks, such as highly increased expression of p53 and decreased expression of HIF-1a and c-Myc that regulate signalling pathways (e.g., AMPK, mTOR). High expression of p53 and the pro-apoptotic proteins cytochrome c and caspases without induction of apoptosis points to functions for these proteins in promoting cell differentiation. These results clearly show that VDAC1 depletion similarly leads to a rewiring of cancer cell metabolism in breast and lung cancer and glioblastoma, regardless of origin or mutational status. This metabolic reprogramming results in cell growth arrest and inhibited tumour growth while encouraging cell differentiation, thus generating cells with decreased proliferation capacity. These results further suggest VDAC1 to be an innovative and markedly potent therapeutic target.
Collapse
|
19
|
Xiong Q, Wang X, Wang L, Huang Y, Tian X, Fan Y, Lin CY. BMP-2 inhibits lung metastasis of osteosarcoma: an early investigation using an orthotopic model. Onco Targets Ther 2018; 11:7543-7553. [PMID: 30464502 PMCID: PMC6214601 DOI: 10.2147/ott.s176724] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Bone morphogenetic proteins (BMPs), members of the TGF-β superfamily, are known to regulate cell proliferation, differentiation, apoptosis, chemotaxis, and angiogenesis. BMPs also participate in the development of most tissues and organs in vertebrates. Recombinant human (rh) BMPs, such as rhBMP-2, rhBMP-4, and rhBMP-7, have been recently approved to augment spinal fusion and recalcitrant long-bone non-unions because of their equivalent or superior efficacy to autogenous bone graft in enhancing bony fusion. Nonetheless, the use of BMPs is contraindicated in surgery for bone tumors because of concerns that this anabolic growth factor may cause tumor proliferation. However, we have repeatedly reported that BMP-2 is effective in inducing osteogenic differentiation of a subpopulation of osteosarcoma (OSA) cells that acquire stem cell attributes and are capable of reconstituting tumor masses, which in turn suppress the malignancy of the bone tumor. Methods 3×105/20 µL human OSA 143B cells were inoculated into 5–6 weeks old BABL/c nude mice to establish orthotopic OSA. X-ray device was used to monitor the developed tumors in animals. Necropsy was performed and the pathology of lung metastasis were tested by Haemotoxylin and Eosin. Moreover, bone formation induced by rhBMP-2 was investigated through micro-computed tomography. In addition, immunohistochemistry staining was used to evaluate the tumorigenicity and growth of OSA cells after rhBMP-2 treatment. Results In the present study, we established an orthotopic model of OSA by inoculating 143B cells into BABL/c mice, which resulted in a tumor occurrence rate of 100%. Following the treatment with rhBMP-2, lung metastasis, which contributes to poor prognosis, was significantly restricted, indicating an additional aspect of rhBMP-2 to suppress expansion of OSA. Concurrently, our micro-computed tomography and radiographic analyses showed that rhBMP-2 reduced the invasion of tumor cells into adjacent bone tissue, which in turn helped to preserve the integrity of the affected bone tissue. Finally, the growth of Ki-67-positive cells and those cells that express high levels of aldehyde dehydrogenase (ALDHbr) was found to be inhibited in the developed tumors. Conclusion On the basis of these results, we conclude that rhBMP-2 can impede the malignancy of OSA by reducing lung metastasis of the tumor. Induction of the tumor cells by rhBMP-2 also helps to preserve the impaired skeleton. These results imply that BMP-2 or BMP-2-mimetic drugs, if properly combined with traditional therapies, may provide a new therapeutic option for the treatment of OSA.
Collapse
Affiliation(s)
- Qisheng Xiong
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China, .,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China,
| | - Xuesong Wang
- Spine Department, The No 2 Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lizhen Wang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China, .,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China,
| | - Yan Huang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China, .,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China,
| | - Xiaodong Tian
- Spine Department, The No 2 Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yubo Fan
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China, .,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China,
| | - Chia-Ying Lin
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China, .,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China, .,Department of Orthopaedic Surgery, University of Cincinnati Academic Health Center, Cincinnati, OH, USA, .,Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA,
| |
Collapse
|
20
|
Otoukesh B, Boddouhi B, Moghtadaei M, Kaghazian P, Kaghazian M. Novel molecular insights and new therapeutic strategies in osteosarcoma. Cancer Cell Int 2018; 18:158. [PMID: 30349420 PMCID: PMC6192346 DOI: 10.1186/s12935-018-0654-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/01/2018] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma (OS) is one of the most prevalent malignant cancers with lower survival and poor overall prognosis mainly in children and adolescents. Identifying the molecular mechanisms and OS stem cells (OSCs) as new concepts involved in disease pathogenesis and progression may potentially lead to new therapeutic targets. Therefore, therapeutic targeting of OSCs can be one of the most important and effective strategies for the treatment of OS. This review describes the new molecular targets of OS as well as novel therapeutic approaches in the design of future investigations and treatment.
Collapse
Affiliation(s)
- Babak Otoukesh
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, 1445613131 Iran
| | - Bahram Boddouhi
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, 1445613131 Iran
| | - Mehdi Moghtadaei
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, 1445613131 Iran
| | - Peyman Kaghazian
- Department of Orthopedic and Traumatology, Universitätsklinikum Bonn, Bonn, Germany
| | - Maria Kaghazian
- Department of Biology, Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
21
|
Cancer stem cells in sarcomas: Getting to the stemness core. Biochim Biophys Acta Gen Subj 2018; 1862:2134-2139. [DOI: 10.1016/j.bbagen.2018.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 12/16/2022]
|
22
|
CSCs: regenerating optimism for osteosarcoma treatment. Oncotarget 2018; 9:31562-31563. [PMID: 30167075 PMCID: PMC6114969 DOI: 10.18632/oncotarget.25820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/16/2018] [Indexed: 01/21/2023] Open
|
23
|
Shteinfer-Kuzmine A, Amsalem Z, Arif T, Zooravlov A, Shoshan-Barmatz V. Selective induction of cancer cell death by VDAC1-based peptides and their potential use in cancer therapy. Mol Oncol 2018; 12:1077-1103. [PMID: 29698587 PMCID: PMC6026870 DOI: 10.1002/1878-0261.12313] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 03/22/2018] [Accepted: 04/15/2018] [Indexed: 01/06/2023] Open
Abstract
Mitochondrial VDAC1 mediates cross talk between the mitochondria and other parts of the cell by transporting anions, cations, ATP, Ca2+, and metabolites and serves as a key player in apoptosis. As such, VDAC1 is involved in two important hallmarks of cancer development, namely energy and metabolic reprograming and apoptotic cell death evasion. We previously developed cell‐penetrating VDAC1‐derived peptides that interact with hexokinase (HK), Bcl‐2, and Bcl‐xL to prevent the anti‐apoptotic activities of these proteins and induce cancer cell death, with a focus on leukemia and glioblastoma. In this study, we demonstrated the sensitivity of a panel of genetically characterized cancer cell lines, differing in origin and carried mutations, to VDAC1‐based peptide‐induced apoptosis. Noncancerous cell lines were less affected by the peptides. Furthermore, we constructed additional VDAC1‐based peptides with the aim of improving targeting, selectivity, and cellular stability, including R‐Tf‐D‐LP4, containing the transferrin receptor internalization sequence (Tf) that allows targeting of the peptide to cancer cells, known to overexpress the transferrin receptor. The mode of action of the VDAC1‐based peptides involves HK detachment, interfering with the action of anti‐apoptotic proteins, and thus activating multiple routes leading to an impairment of cell energy and metabolism homeostasis and the induction of apoptosis. Finally, in xenograft glioblastoma, lung, and breast cancer mouse models, R‐Tf‐D‐LP4 inhibited tumor growth while inducing massive cancer cell death, including of cancer stem cells. Thus, VDAC1‐based peptides offer an innovative new conceptual framework for cancer therapy.
Collapse
Affiliation(s)
- Anna Shteinfer-Kuzmine
- Department of Life Sciences, National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Zohar Amsalem
- Department of Life Sciences, National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tasleem Arif
- Department of Life Sciences, National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alexandra Zooravlov
- Department of Life Sciences, National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Varda Shoshan-Barmatz
- Department of Life Sciences, National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
24
|
Prince MEP, Zhou L, Moyer JS, Tao H, Lu L, Owen J, Etigen M, Zheng F, Chang AE, Xia J, Wolf G, Wicha MS, Huang S, Ren X, Li Q. Evaluation of the immunogenicity of ALDH(high) human head and neck squamous cell carcinoma cancer stem cells in vitro. Oral Oncol 2018; 59:30-42. [PMID: 27424180 DOI: 10.1016/j.oraloncology.2016.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 05/04/2016] [Accepted: 05/20/2016] [Indexed: 12/20/2022]
Abstract
OBJECTIVES To establish the concept that the antigenicity/immunogenicity of ALDH(high) human head and neck squamous cell carcinoma (HNSCC) cancer stem cells (CSC) is distinct from that of ALDH(low) non-CSCs. METHODS We generated CSC-loaded dendritic cells (DCs) to sensitize autologous peripheral blood T, B lymphocytes to react with CSCs using human HNSCC samples in vitro. RESULTS From peripheral blood collected from patients with HNSCC, we obtained PBMCs. DCs generated from the PBMC and pulsed with the lysate of ALDH(high) cells isolated from cultured HNSCC cells (CSC-DC) could sensitize autologous T, B lymphocytes in vitro, which was evident by cytokine production, CTL activity, and antibody secretion of these primed T, B cells in response to ALDH(high) CSCs. In contrast, DCs pulsed with lysate of ALDH(low) cells (ALDH(low)-DC) resulted in limited sensitization/priming of autologous T, B lymphocytes to produce IFNγ, GM-CSF; lyse CSCs, and secrete IgM and IgG in response to ALDH(high) CSCs. These results demonstrated significant differences in the antigenicity/immunogenicity between ALDH(high) CSCs vs. ALDH(low) cells isolated from the tumor specimen of patients with HNSCC, which indicates the existence of unique CSC antigens in the ALDH(high) population. CONCLUSION It is feasible to generate DCs from the PBMCs and isolate ALDH(high) CSCs from cultured tumor cells of the patients with HNSCC to prepare CSC-DC vaccines that can induce anti-HNSCC CSC cellular and humoral immunity, indicating its potential clinical application to treat patients with HNSCC.
Collapse
Affiliation(s)
- Mark E P Prince
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Li Zhou
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA.,Department of Immunology, Biotherapy Center ,Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Jeffrey S Moyer
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Huimin Tao
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA.,Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Lu
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA.,State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - John Owen
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Martin Etigen
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Fang Zheng
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA.,Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Alfred E Chang
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Jianchuan Xia
- State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Gregory Wolf
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Max S Wicha
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Shiang Huang
- Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiubao Ren
- Department of Immunology, Biotherapy Center ,Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Qiao Li
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| |
Collapse
|
25
|
Gatti M, Solari A, Pattarozzi A, Campanella C, Thellung S, Maniscalco L, De Maria R, Würth R, Corsaro A, Bajetto A, Ratto A, Ferrari A, Daga A, Barbieri F, Florio T. In vitro and in vivo characterization of stem-like cells from canine osteosarcoma and assessment of drug sensitivity. Exp Cell Res 2018; 363:48-64. [PMID: 29305964 DOI: 10.1016/j.yexcr.2018.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/22/2017] [Accepted: 01/02/2018] [Indexed: 12/19/2022]
Abstract
Cancer stem cell (CSC) self-renewing and drug resistance cause treatment failure and tumor recurrence. Osteosarcoma is an aggressive bone tumor characterized by biological and molecular heterogeneity, possibly dependent on CSCs. CSC identification in osteosarcoma and their efficient targeting are still open questions. Spontaneous canine osteosarcoma shares clinical and biological features with the human tumors, representing a model for translational studies. We characterized three CSC-enriched canine osteosarcoma cultures. In serum-free conditions, these CSC cultures grow as anchorage-independent spheroids, show mesenchymal-like properties and in vivo tumorigenicity, recapitulating the heterogeneity of the original osteosarcoma. Osteosarcoma CSCs express stem-related factors (Sox2, Oct4, CD133) and chemokine receptors and ligands (CXCR4, CXCL12) involved in tumor proliferation and self-renewal. Standard drugs for osteosarcoma treatment (doxorubicin and cisplatin) affected CSC-enriched and parental primary cultures, showing different efficacy within tumors. Moreover, metformin, a type-2 diabetes drug, significantly inhibits osteosarcoma CSC viability, migration and self-renewal and, in co-treatment with doxorubicin and cisplatin, enhances drug cytotoxicity. Collectively, we demonstrate that canine osteosarcoma primary cultures contain CSCs exhibiting distinctive sensitivity to anticancer agents, as a reliable experimental model to assay drug efficacy. We also provide proof-of-principle of metformin efficacy, alone or in combination, as pharmacological strategy to target osteosarcoma CSCs.
Collapse
Affiliation(s)
- Monica Gatti
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2, 16132 Genova, Italy
| | - Agnese Solari
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2, 16132 Genova, Italy
| | - Alessandra Pattarozzi
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2, 16132 Genova, Italy
| | - Chiara Campanella
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Piazza Borgo Pila 39, 16129 Genova, Italy
| | - Stefano Thellung
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2, 16132 Genova, Italy
| | - Lorella Maniscalco
- Department of Veterinary Sciences, University of Torino, Largo Braccini 2, 10095 Grugliasco (Torino), Italy
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Torino, Largo Braccini 2, 10095 Grugliasco (Torino), Italy
| | - Roberto Würth
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2, 16132 Genova, Italy
| | - Alessandro Corsaro
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2, 16132 Genova, Italy
| | - Adriana Bajetto
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2, 16132 Genova, Italy
| | - Alessandra Ratto
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Piazza Borgo Pila 39, 16129 Genova, Italy
| | - Angelo Ferrari
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Piazza Borgo Pila 39, 16129 Genova, Italy
| | - Antonio Daga
- IRCCS-AOU San Martino-IST, Largo Benzi 10, 16132 Genova, Italy
| | - Federica Barbieri
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2, 16132 Genova, Italy.
| | - Tullio Florio
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2, 16132 Genova, Italy.
| |
Collapse
|
26
|
Henderson T, Chen M, Darrow MA, Li CS, Chiu CL, Monjazeb AM, Murphy WJ, Canter RJ. Alterations in cancer stem-cell marker CD44 expression predict oncologic outcome in soft-tissue sarcomas. J Surg Res 2017; 223:207-214. [PMID: 29433875 DOI: 10.1016/j.jss.2017.11.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/28/2017] [Accepted: 11/03/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND Cancer stem cells (CSCs) have been shown to resist chemotherapy and promote metastasis after cytotoxic therapies. We sought to determine if the expression of CSC markers (aldehyde dehydrogenase [ALDH], CD44, and epidermal growth factor receptor [EGFR]) predicted outcomes in soft-tissue sarcoma (STS) patients. METHODS We queried an institutional database of 23 STS patients and evaluated immunohistochemical expression of CSC markers ALDH, CD44, and EGFR. The Cancer Genome Atlas (TCGA) was also queried for STS clinical and genomic data. Disease-specific (DSS) and overall survival (OS) were assessed by univariate and Kaplan-Meier analysis. RESULTS Of the 23 institutional patients, the majority was female, had high-grade tumors and had extremity tumors. With a median follow-up of 27 months, nine patients (39%) experienced distant recurrence, and four (17%) died of disease. Mean H-scores at diagnosis (±standard error of the mean) for CD44, ALDH1, and EGFR were 169 ± 27, 77 ± 15, and 144 ± 23, respectively. On univariate analysis, there was a trend for increased CD44 score to predict both worse DSS and OS (hazard ratio = 1.01, 95% confidence interval 1-1.02, P = 0.056), whereas ALDH and EGFR scores did not. Analysis of 74 TCGA STS cases with complete clinical and genomic data revealed that CD44 copy number alterations predicted worse DSS (9.89 months versus 72.5 months, P = 0.007) and a trend for worse OS (14.03 months versus 38.6 months, P = 0.12), whereas ALDH1 and EGFR copy number alteration did not. Multivariate analysis of the combined data sets was consistent with worse DSS among patients with higher CD44 expression. CONCLUSIONS Institutional and national TCGA data show the association of elevated baseline CD44 expression with worse STS outcomes. Further study of CD44 as a possible novel STS biomarker appears indicated.
Collapse
Affiliation(s)
| | - Mingyi Chen
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California
| | - Morgan A Darrow
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California
| | - Chin-Shang Li
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis, Sacramento, California
| | - Chi-Lu Chiu
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis, Sacramento, California
| | - Arta M Monjazeb
- Department of Radiation Oncology, University of California Davis, Sacramento, California
| | - William J Murphy
- Departments of Dermatology and Internal Medicine, Laboratory of Cancer Immunology, University of California Davis Medical Center, Sacramento, California
| | - Robert J Canter
- Division of Surgical Oncology, Department of Surgery, University of California Davis Medical Center, Sacramento, California.
| |
Collapse
|
27
|
Abstract
Sarcomas arise from primitive mesenchymal cells, which are classified, into two main groups: Bone and soft tissue sarcomas. We have searched all-important electronic databases including Google scholar and PubMed for the collection of latest literature pertaining to pediatric sarcomas. Latest literature confirmed that these tumors are relatively rare and represent only 1% of all malignancies but they have higher incidence in children. Pediatric sarcomas comprise about 13% of all pediatric malignancies and are ranked third in childhood cancers. The highest incidence rates are reported among rhabdomyosarcoma, osteosarcoma and Ewing's sarcomas in children. All of these neoplasms often display highly aggressive behavior with tendency to form metastases. Important globally used management avenues include surgery with systemic chemotherapy and have success rate of 70% at 5-years. Furthermore, in the cases of advanced stages, the prognosis is poor, chances of treatment failure and recurrence are quite high. Utilization of cancer stem cells is the latest approach with great potential in management of above pathological state. The present review article discuss all-important aspects of commonly found pediatric sarcomas throughout the world.
Collapse
Affiliation(s)
- Junhua Cao
- Department of Pediatric Internal Medicine, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Qi An
- Department of Pediatric Internal Medicine, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Lei Wang
- Department of Pediatric Internal Medicine, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
28
|
Li Y, Xian M, Yang B, Ying M, He Q. Inhibition of KLF4 by Statins Reverses Adriamycin-Induced Metastasis and Cancer Stemness in Osteosarcoma Cells. Stem Cell Reports 2017; 8:1617-1629. [PMID: 28552603 PMCID: PMC5470096 DOI: 10.1016/j.stemcr.2017.04.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/21/2017] [Accepted: 04/24/2017] [Indexed: 12/19/2022] Open
Abstract
Adriamycin-based combination chemotherapy is the standard first-line treatment for osteosarcoma, but tumor recurrence and metastasis occurs in most cases. Recent evidence suggests that microenvironmental stress such as chemotherapy can lead to the enrichment of cancer stem cells (CSCs), which result in cancer metastasis, recurrence, and drug resistance. However, the exact mechanisms underlying this phenomenon and how to target CSCs are still open questions. Herein, we report that Adriamycin treatment induces a stem-like phenotype and promotes metastatic potential in osteosarcoma cells through upregulating KLF4. KLF4 knockdown blocks Adriamycin-induced stemness phenotype and metastasis capacity. We further screen that statins remarkably reverse Adriamycin-induced CSC properties and metastasis by downregulating KLF4. Most strikingly, simvastatin severely impaired Adriamycin-enhanced tumorigenesis of KHOS/NP cells in vivo. These data suggest that Adriamycin-based chemotherapeutics may simulate CSCs through activation of KLF4 signaling and that selective inhibition of KLF4 with statins should be considered in the development of osteosarcoma therapeutics. Adriamycin treatment induces a stemness phenotype in osteosarcoma cells KLF4 is a key transcriptional regulator of ADR-induced osteosarcoma cancer stemness Simvastatin reverses ADR-induced CSC properties by downregulating KLF4 Simvastatin abolishes ADR-enhanced tumorigenesis of KHOS/NP cells in vivo
Collapse
Affiliation(s)
- Yangling Li
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Room 427, Hangzhou 310058, China
| | - Miao Xian
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Room 427, Hangzhou 310058, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Room 427, Hangzhou 310058, China
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Room 427, Hangzhou 310058, China.
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Room 427, Hangzhou 310058, China.
| |
Collapse
|
29
|
Overcoming Therapeutic Resistance of Bone Sarcomas: Overview of the Molecular Mechanisms and Therapeutic Targets for Bone Sarcoma Stem Cells. Stem Cells Int 2016; 2016:2603092. [PMID: 28115942 PMCID: PMC5223039 DOI: 10.1155/2016/2603092] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 10/31/2016] [Accepted: 11/16/2016] [Indexed: 02/07/2023] Open
Abstract
Bone sarcomas are heterogeneous malignant tumors that exhibit clinical, histological, and molecular heterogeneity. Recent progress in their multimodal treatment has gradually improved patient prognosis; however, drug resistance and distant metastasis remain unresolved clinical problems. Recent investigations have suggested the existence of cancer stem-like cells (CSCs) in bone sarcomas, which represent a subpopulation of tumor cells with high tumor-forming ability. The hallmarks of CSCs include tumor- and metastasis-forming potential and drug resistance, which are responsible for poor prognoses of bone sarcoma patients. Therefore, elucidation of the molecular mechanisms of CSCs and identification of therapeutic targets could contribute to novel treatment strategies for bone sarcomas and improve patient prognosis. This paper provides an overview of the accumulating knowledge on bone sarcoma stem cells and preclinical analyses to overcome their lethal phenotypes, in addition to a discussion of their potential for novel therapeutics for bone sarcomas.
Collapse
|
30
|
Re-calculating! Navigating through the osteosarcoma treatment roadblock. Pharmacol Res 2016; 117:54-64. [PMID: 27940205 DOI: 10.1016/j.phrs.2016.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/29/2016] [Accepted: 12/05/2016] [Indexed: 01/03/2023]
Abstract
The survival rates for patients with osteosarcoma have remained almost static for the past three decades. Current standard of care therapy includes chemotherapies such as doxorubicin, cisplatin, and methotrexate along with complete surgical resection and surgery with or without ifosfamide and etoposide for relapse, though outcomes are hoped to be improved through clinical trials. Additionally, increased understanding of the genetics, signaling pathways and microenvironmental factors driving the disease have led to the identification of promising agents and potential paths towards translation of an exciting array of novel targeted therapies. Here, we review the mechanism of action of these emerging therapies and how, with clinical translation, they can potentially improve the survival rates for osteosarcoma patients in the near future.
Collapse
|
31
|
Brown HK, Tellez-Gabriel M, Heymann D. Cancer stem cells in osteosarcoma. Cancer Lett 2016; 386:189-195. [PMID: 27894960 DOI: 10.1016/j.canlet.2016.11.019] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/15/2016] [Accepted: 11/17/2016] [Indexed: 12/11/2022]
Abstract
Osteosarcoma is the most common primary bone tumour in children and adolescents and advanced osteosarcoma patients with evidence of metastasis share a poor prognosis. Osteosarcoma frequently gains resistance to standard therapies highlighting the need for improved treatment regimens and identification of novel therapeutic targets. Cancer stem cells (CSC) represent a sub-type of tumour cells attributed to critical steps in cancer including tumour propagation, therapy resistance, recurrence and in some cases metastasis. Recent published work demonstrates evidence of cancer stem cell phenotypes in osteosarcoma with links to drug resistance and tumorigenesis. In this review we will discuss the commonly used isolation techniques for cancer stem cells in osteosarcoma as well as the identified biochemical and molecular markers.
Collapse
Affiliation(s)
- Hannah K Brown
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, S10 2RX, Sheffield, UK; European Associated Laboratory, INSERM-University of Sheffield, Sarcoma Research Unit, Medical School, S10 2RX, Sheffield, UK.
| | - Marta Tellez-Gabriel
- Laboratotio Hematologia Oncologica y de Transplantes, Institut Investigacions Biomèdiques (IBB) Sant Pau, Hospital de la Santa Creui Sant Pau, 08025, Barcelona, Spain
| | - Dominique Heymann
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, S10 2RX, Sheffield, UK; European Associated Laboratory, INSERM-University of Sheffield, Sarcoma Research Unit, Medical School, S10 2RX, Sheffield, UK; INSERM, UMR 957, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Equipe Ligue 2012, University of Nantes, Faculty of Medicine, 44035, Nantes, France.
| |
Collapse
|
32
|
Martinez-Cruzado L, Tornin J, Santos L, Rodriguez A, García-Castro J, Morís F, Rodriguez R. Aldh1 Expression and Activity Increase During Tumor Evolution in Sarcoma Cancer Stem Cell Populations. Sci Rep 2016; 6:27878. [PMID: 27292183 PMCID: PMC4904413 DOI: 10.1038/srep27878] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 05/25/2016] [Indexed: 12/11/2022] Open
Abstract
Tumors evolve from initial tumorigenic events into increasingly aggressive behaviors in a process usually driven by subpopulations of cancer stem cells (CSCs). Mesenchymal stromal/stem cells (MSCs) may act as the cell-of-origin for sarcomas, and CSCs that present MSC features have been identified in sarcomas due to their ability to grow as self-renewed floating spheres (tumorspheres). Accordingly, we previously developed sarcoma models using human MSCs transformed with relevant oncogenic events. To study the evolution/emergence of CSC subpopulations during tumor progression, we compared the tumorigenic properties of bulk adherent cultures and tumorsphere-forming subpopulations both in the sarcoma cell-of-origin models (transformed MSCs) and in their corresponding tumor xenograft-derived cells. Tumor formation assays showed that the tumorsphere cultures from xenograft-derived cells, but not from the cell-of-origin models, were enriched in CSCs, providing evidence of the emergence of bona fide CSCs subpopulations during tumor progression. Relevant CSC-related factors, such as ALDH1 and SOX2, were increasingly upregulated in CSCs during tumor progression, and importantly, the increased levels and activity of ALDH1 in these subpopulations were associated with enhanced tumorigenicity. In addition to being a CSC marker, our findings indicate that ALDH1 could also be useful for tracking the malignant potential of CSC subpopulations during sarcoma evolution.
Collapse
Affiliation(s)
- Lucia Martinez-Cruzado
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Juan Tornin
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Laura Santos
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Aida Rodriguez
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Javier García-Castro
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | | | - Rene Rodriguez
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
33
|
MiR-26a inhibits stem cell-like phenotype and tumor growth of osteosarcoma by targeting Jagged1. Oncogene 2016; 36:231-241. [PMID: 27270422 DOI: 10.1038/onc.2016.194] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/26/2016] [Accepted: 04/22/2016] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRNAs) are important epigenetic regulators of gene expression. Although several miRNAs have been implicated in osteosarcoma, their role in regulation of osteosarcoma cancer stem cells (CSCs) remains unknown. Here we demonstrated that miR-26a is downregulated in osteosarcoma CSCs when derived by either sarcosphere generation, chemodrug or aldehyde dehydrogenase (ALDH) activity selection. Lentiviral overexpression of miR-26a in ZOS and 143B osteosarcoma cells decreases the expression of stem cell markers and suppresses sarcosphere formation, as well as ALDH activity. Moreover, miR-26a overexpression inhibits the tumor cell growth both in vitro and in vivo. We further demonstrate that miR-26a directly target Jagged1, one of the Notch ligand, and that its tumor suppressive effects are mediated through inhibition of Jagged1/Notch signaling. Importantly, reduced miR-26a expression, as determined by in situ hybridization in patient tumors (n=92), is associated with lung metastasis and poor overall survival of osteosarcoma patients. Together, these data suggest the essential role of miR-26a/Jagged1/Notch pathway in regulating the stem cell-like traits of osteosarcoma cells and provide a potential target for osteosarcoma therapy.
Collapse
|
34
|
Osteosarcoma: Cells-of-Origin, Cancer Stem Cells, and Targeted Therapies. Stem Cells Int 2016; 2016:3631764. [PMID: 27366153 PMCID: PMC4913005 DOI: 10.1155/2016/3631764] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/10/2016] [Indexed: 12/25/2022] Open
Abstract
Osteosarcoma (OS) is the most common type of primary solid tumor that develops in bone. Although standard chemotherapy has significantly improved long-term survival over the past few decades, the outcome for those patients with metastatic or recurrent OS remains dismally poor and, therefore, novel agents and treatment regimens are urgently required. A hypothesis to explain the resistance of OS to chemotherapy is the existence of drug resistant CSCs with progenitor properties that are responsible of tumor relapses and metastasis. These subpopulations of CSCs commonly emerge during tumor evolution from the cell-of-origin, which are the normal cells that acquire the first cancer-promoting mutations to initiate tumor formation. In OS, several cell types along the osteogenic lineage have been proposed as cell-of-origin. Both the cell-of-origin and their derived CSC subpopulations are highly influenced by environmental and epigenetic factors and, therefore, targeting the OS-CSC environment and niche is the rationale for many recently postulated therapies. Likewise, some strategies for targeting CSC-associated signaling pathways have already been tested in both preclinical and clinical settings. This review recapitulates current OS cell-of-origin models, the properties of the OS-CSC and its niche, and potential new therapies able to target OS-CSCs.
Collapse
|
35
|
Deshpande N, Rangarajan A. Cancer Stem Cells: Formidable Allies of Cancer. Indian J Surg Oncol 2016; 6:400-14. [PMID: 27081258 DOI: 10.1007/s13193-015-0451-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 07/24/2015] [Indexed: 12/17/2022] Open
Abstract
Cancer stem cells (CSC) represent the subpopulation of cells within a tumour showing two fundamental properties of stem cells - self-renewal (the ability to make more of their own kind) and differentiation (the ability to generate diverse cell types present within a tissue). The CSC hypothesis posits that CSCs play an important role in tumour initiation, maintenance and progression. Furthermore, owing to their intrinsic drug resistance, they remain refractory to currently used therapy, thereby contributing to tumour relapse. Thus, targeting or taming CSCs can lead to more effective cancer treatment in the coming decades. In this review, we will discuss about the origin of CSC hypothesis, evidence showing their existence, clinical relevance and translational significance.
Collapse
Affiliation(s)
- Neha Deshpande
- Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560065 India
| | - Annapoorni Rangarajan
- Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560065 India
| |
Collapse
|
36
|
Lye KL, Nordin N, Vidyadaran S, Thilakavathy K. Mesenchymal stem cells: From stem cells to sarcomas. Cell Biol Int 2016; 40:610-8. [PMID: 26992453 DOI: 10.1002/cbin.10603] [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] [Received: 12/01/2015] [Accepted: 03/14/2016] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) have garnered vast interests in clinical settings, especially in regenerative medicine due to their unique properties-they are reliably isolated and expanded from various tissue sources; they are able to differentiate into mesodermal tissues such as bones, cartilages, adipose tissues, and muscles; and they have unique immunosuppressive properties. However, there are some concerns pertaining to the role of MSCs in the human body. On one hand, they are crucial component in the regeneration and repair of the human body. On the contrary, they are shown to transform into sarcomas. Although the exact mechanisms are still unknown, many new leads have pointed to the belief that MSCs do play a role in sarcomagenesis. This review focuses on the current updates and findings of the role of MSCs in their transformation process into sarcomas.
Collapse
Affiliation(s)
- Kwan Liang Lye
- Medical Genetics Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Norshariza Nordin
- Medical Genetics Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.,Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Sharmili Vidyadaran
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.,Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Karuppiah Thilakavathy
- Medical Genetics Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.,Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
37
|
Canter RJ, Grossenbacher SK, Ames E, Murphy WJ. Immune targeting of cancer stem cells in gastrointestinal oncology. J Gastrointest Oncol 2016; 7:S1-S10. [PMID: 27034806 DOI: 10.3978/j.issn.2078-6891.2015.066] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The cancer stem cell (CSC) hypothesis postulates that a sub-population of quiescent cells exist within tumors which are resistant to conventional cytotoxic/anti-proliferative therapies. It is these CSCs which then seed tumor relapse, even in cases of apparent complete response to systemic therapy. Therefore, therapies, such as immunotherapy, which add a specific anti-CSC strategy to standard cytoreductive treatments may provide a promising new direction for future cancer therapies. CSCs are an attractive target for immune therapies since, unlike chemotherapy or radiotherapy, immune effector cells do not specifically require target cells to be proliferating in order to effectively kill them. Although recent advances have been made in the development of novel systemic and targeted therapies for advanced gastro-intestinal (GI) malignancies, there remains an unmet need for durable new therapies for these refractory malignancies. Novel immunotherapeutic strategies targeting CSCs are in pre-clinical and clinical development across the spectrum of the immune system, including strategies utilizing adaptive immune cell-based effectors, innate immune effectors, as well as vaccine approaches. Lastly, since important CSC functions are affected by the tumor microenvironment, targeting of both cellular (myeloid derived suppressor cells and tumor-associated macrophages) and sub-cellular (cytokines, chemokines, and PD1/PDL1) components of the tumor microenvironment is under investigation in the immune targeting of CSCs. These efforts are adding to the significant optimism about the potential utility of immunotherapy to overcome cancer resistance mechanisms and cure greater numbers of patients with advanced malignancy.
Collapse
Affiliation(s)
- Robert J Canter
- 1 Division of Surgical Oncology, Department of Surgery, 2 Laboratory of Cancer Immunology, Department of Dermatology, 3 Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Steven K Grossenbacher
- 1 Division of Surgical Oncology, Department of Surgery, 2 Laboratory of Cancer Immunology, Department of Dermatology, 3 Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Erik Ames
- 1 Division of Surgical Oncology, Department of Surgery, 2 Laboratory of Cancer Immunology, Department of Dermatology, 3 Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - William J Murphy
- 1 Division of Surgical Oncology, Department of Surgery, 2 Laboratory of Cancer Immunology, Department of Dermatology, 3 Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
38
|
Opposite Effects of Soluble Factors Secreted by Adipose Tissue on Proliferating and Quiescent Osteosarcoma Cells. Plast Reconstr Surg 2016; 137:865-875. [DOI: 10.1097/01.prs.0000479989.88114.8b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
39
|
Enhancement of tumor initiation and expression of KCNMA1, MORF4L2 and ASPM genes in the adenocarcinoma of lung xenograft after vorinostat treatment. Oncotarget 2016; 6:8663-75. [PMID: 25796627 PMCID: PMC4496174 DOI: 10.18632/oncotarget.3536] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/12/2015] [Indexed: 12/15/2022] Open
Abstract
Cancer stem cells (CSCs) are usually tolerant to chemotherapy and radiotherapy and associated with tumor relapse. Suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor (HDACI), is currently being used in clinical trials of lung cancer. However, SAHA facilitates the formation of induced pluripotent stem cells from somatic cells. We hypothesized that SAHA would mediate the CSCs properties and subsequently confer a more malignant phenotype in lung cancer. Transfected H1299 lung cancer cells, which stably expresses a triple fused reporter gene (DsRedm-Fluc-tTKsr39) under the control of CMV promoter was used to establish a xenograft mouse model. After the treatment of SAHA, H1299 cell line and tumor xenografts were sorted by fluorescence-activated cell sorting (FACS) based on aldehyde dehydrogenase (ALDH) activity. We found that SAHA could suppress the growth of xenografted H1299 tumors with decreased proportion of ALDHbr lung cancer cells indicating that SAHA may target CSCs. However, SAHA significantly enhanced the tumor initiating capacity and the expression of malignant genes such as KCNMA1, MORF4L2 and ASPM in the remaining living ALDHbr cells. These findings suggested that SAHA treatment created a more drug-resistant state in residual ALDHbr cells. The in vivo imaging technique may facilitate searching and characterization of CSCs.
Collapse
|
40
|
Abstract
The cancer stem cell model in solid tumors has evolved significantly from the early paradigm shifting work highlighting parallels between the stem cell hierarchy in hematologic malignancies and solid tumors. Putative stem cells can dedifferentiated, be induced by context, and be the result of accumulated genetic mutations. The simple hypothesis that stem cell therapies will overcome the minority of cells that lead to recurrence has evolved with it. Nevertheless, the body of evidence that this field is clinically relevant in patients and patient care has grown with the complexity of the hypotheses, and numerous clinical strategies to target these cells have been identified. Herein we review this progress and highlight the work still outstanding.
Collapse
Affiliation(s)
- Wendy A Woodward
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Richard P Hill
- Princess Margaret Cancer Centre, Ontario Cancer Insitute, Toronto, ON, M5G 2M9, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 2M9, Canada
| |
Collapse
|
41
|
Yan GN, Lv YF, Guo QN. Advances in osteosarcoma stem cell research and opportunities for novel therapeutic targets. Cancer Lett 2016; 370:268-74. [DOI: 10.1016/j.canlet.2015.11.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/03/2015] [Accepted: 11/03/2015] [Indexed: 12/15/2022]
|
42
|
Ames E, Canter RJ, Grossenbacher SK, Mac S, Chen M, Smith RC, Hagino T, Perez-Cunningham J, Sckisel GD, Urayama S, Monjazeb AM, Fragoso RC, Sayers TJ, Murphy WJ. NK Cells Preferentially Target Tumor Cells with a Cancer Stem Cell Phenotype. THE JOURNAL OF IMMUNOLOGY 2015; 195:4010-9. [PMID: 26363055 DOI: 10.4049/jimmunol.1500447] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/10/2015] [Indexed: 01/02/2023]
Abstract
Increasing evidence supports the hypothesis that cancer stem cells (CSCs) are resistant to antiproliferative therapies, able to repopulate tumor bulk, and seed metastasis. NK cells are able to target stem cells as shown by their ability to reject allogeneic hematopoietic stem cells but not solid tissue grafts. Using multiple preclinical models, including NK coculture (autologous and allogeneic) with multiple human cancer cell lines and dissociated primary cancer specimens and NK transfer in NSG mice harboring orthotopic pancreatic cancer xenografts, we assessed CSC viability, CSC frequency, expression of death receptor ligands, and tumor burden. We demonstrate that activated NK cells are capable of preferentially killing CSCs identified by multiple CSC markers (CD24(+)/CD44(+), CD133(+), and aldehyde dehydrogenase(bright)) from a wide variety of human cancer cell lines in vitro and dissociated primary cancer specimens ex vivo. We observed comparable effector function of allogeneic and autologous NK cells. We also observed preferential upregulation of NK activation ligands MICA/B, Fas, and DR5 on CSCs. Blocking studies further implicated an NKG2D-dependent mechanism for NK killing of CSCs. Treatment of orthotopic human pancreatic cancer tumor-bearing NSG mice with activated NK cells led to significant reductions in both intratumoral CSCs and tumor burden. Taken together, these data from multiple preclinical models, including a strong reliance on primary human cancer specimens, provide compelling preclinical evidence that activated NK cells preferentially target cancer cells with a CSC phenotype, highlighting the translational potential of NK immunotherapy as part of a combined modality approach for refractory solid malignancies.
Collapse
Affiliation(s)
- Erik Ames
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Robert J Canter
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Steven K Grossenbacher
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Stephanie Mac
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Mingyi Chen
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Rachel C Smith
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Takeshi Hagino
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Jessica Perez-Cunningham
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Gail D Sckisel
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Shiro Urayama
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817
| | - Arta M Monjazeb
- Department of Radiation Oncology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Ruben C Fragoso
- Department of Radiation Oncology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Thomas J Sayers
- Basic Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, MD 21702; and
| | - William J Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817; Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817
| |
Collapse
|
43
|
Martins-Neves SR, Corver WE, Paiva-Oliveira DI, van den Akker BEWM, Briaire-de-Bruijn IH, Bovée JVMG, Gomes CMF, Cleton-Jansen AM. Osteosarcoma Stem Cells Have Active Wnt/β-catenin and Overexpress SOX2 and KLF4. J Cell Physiol 2015; 231:876-86. [DOI: 10.1002/jcp.25179] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Sara R. Martins-Neves
- Department of Pathology; Leiden University Medical Center; Leiden The Netherlands
- Pharmacology and Experimental Therapeutics; Institute for Biomedical Imaging and Life Sciences (IBILI); Faculty of Medicine; University of Coimbra; Coimbra Portugal
- CNC.IBILI; University of Coimbra; Coimbra Portugal
- Center of Investigation in Environment; Genetics and Oncobiology; CIMAGO - Faculty of Medicine; University of Coimbra, Coimbra; Celas Portugal
| | - Willem E. Corver
- Department of Pathology; Leiden University Medical Center; Leiden The Netherlands
| | - Daniela I. Paiva-Oliveira
- Pharmacology and Experimental Therapeutics; Institute for Biomedical Imaging and Life Sciences (IBILI); Faculty of Medicine; University of Coimbra; Coimbra Portugal
- CNC.IBILI; University of Coimbra; Coimbra Portugal
| | | | | | | | - Célia M. F. Gomes
- Pharmacology and Experimental Therapeutics; Institute for Biomedical Imaging and Life Sciences (IBILI); Faculty of Medicine; University of Coimbra; Coimbra Portugal
- CNC.IBILI; University of Coimbra; Coimbra Portugal
- Center of Investigation in Environment; Genetics and Oncobiology; CIMAGO - Faculty of Medicine; University of Coimbra, Coimbra; Celas Portugal
| | | |
Collapse
|
44
|
Alfranca A, Martinez-Cruzado L, Tornin J, Abarrategi A, Amaral T, de Alava E, Menendez P, Garcia-Castro J, Rodriguez R. Bone microenvironment signals in osteosarcoma development. Cell Mol Life Sci 2015; 72:3097-113. [PMID: 25935149 PMCID: PMC11113487 DOI: 10.1007/s00018-015-1918-y] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 02/06/2023]
Abstract
The bone is a complex connective tissue composed of many different cell types such as osteoblasts, osteoclasts, chondrocytes, mesenchymal stem/progenitor cells, hematopoietic cells and endothelial cells, among others. The interaction between them is finely balanced through the processes of bone formation and bone remodeling, which regulates the production and biological activity of many soluble factors and extracellular matrix components needed to maintain the bone homeostasis in terms of cell proliferation, differentiation and apoptosis. Osteosarcoma (OS) emerges in this complex environment as a result of poorly defined oncogenic events arising in osteogenic lineage precursors. Increasing evidence supports that similar to normal development, the bone microenvironment (BME) underlies OS initiation and progression. Here, we recapitulate the physiological processes that regulate bone homeostasis and review the current knowledge about how OS cells and BME communicate and interact, describing how these interactions affect OS cell growth, metastasis, cancer stem cell fate and therapy outcome.
Collapse
Affiliation(s)
- Arantzazu Alfranca
- Unidad de Biotecnología Celular, Área de Genética Humana, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Lucia Martinez-Cruzado
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Juan Tornin
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Ander Abarrategi
- Unidad de Biotecnología Celular, Área de Genética Humana, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, UK
| | - Teresa Amaral
- Molecular Pathology Program, Institute of Biomedical Research of Salamanca-Centro de Investigación del Cáncer, Centro de Investigación del Cáncer (IBSAL-CIC), Salamanca, Spain
- Department of Pathology and Biobank, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), CSIC-Universidad de Sevilla, Seville, Spain
| | - Enrique de Alava
- Molecular Pathology Program, Institute of Biomedical Research of Salamanca-Centro de Investigación del Cáncer, Centro de Investigación del Cáncer (IBSAL-CIC), Salamanca, Spain
- Department of Pathology and Biobank, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), CSIC-Universidad de Sevilla, Seville, Spain
| | - Pablo Menendez
- Cell Therapy Program, School of Medicine, Josep Carreras Leukemia Research Institute, University of Barcelona, Barcelona, Spain
- Instituciò Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| | - Javier Garcia-Castro
- Unidad de Biotecnología Celular, Área de Genética Humana, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Rene Rodriguez
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| |
Collapse
|
45
|
BMP-2 inhibits tumor-initiating ability in human renal cancer stem cells and induces bone formation. J Cancer Res Clin Oncol 2014; 141:1013-24. [PMID: 25431339 DOI: 10.1007/s00432-014-1883-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 11/22/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE We have previously shown that BMP-2 induces bone formation and inhibits tumorigenicity of cancer stem cells (CSCs) in a human osteosarcoma OS99-1 cell line. In this study, we sought to determine whether BMP-2 can similarly induce bone formation and inhibit the tumorigenicity of renal CSCs identified based on aldehyde dehydrogenase (ALDH) activity in renal cell carcinoma (RCC) cell lines and primary tumors. METHODS Using a xenograft model in which cells from human RCC cell lines ACHN, Caki-2, and primary tumors were grown in NOD/SCID mice, renal CSCs were identified as a subset of ALDH(br) cells. The ALDH(br) cells possessed a greater colony-forming efficiency, higher proliferative output, increased expression of stem cell marker genes Oct3/4A, Nanog, renal embryonic marker Pax-2, and greater tumorigenicity compared to cells with low ALDH activity (ALDH(lo) cells), generating new tumors with as few as 25 cells in mice. RESULTS In vitro, BMP-2 was found to inhibit the ALDH(br) cell growth, down-regulate the expression of embryonic stem cell markers, and up-regulate the transcription of osteogenic markers. In vivo, all animals receiving a low number of ALDH(br) cells (5 × 10(3)) from ACHN, Caki-2, and primary tumor xenografts treated with 30 µg BMP-2 per animal showed limited tumor growth with significant bone formation, while untreated cells developed large tumor masses without bone formation. CONCLUSIONS These results suggest that BMP-2 inhibits the tumor-initiating ability of renal CSCs and induces osseous bone formation. BMP-2 may therefore provide a beneficial strategy for human RCC treatment by targeting the CSC-enriched population.
Collapse
|
46
|
Yang J, Ren Z, Du X, Hao M, Zhou W. The role of mesenchymal stem/progenitor cells in sarcoma: update and dispute. Stem Cell Investig 2014; 1:18. [PMID: 27358864 DOI: 10.3978/j.issn.2306-9759.2014.10.01] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/10/2014] [Indexed: 12/26/2022]
Abstract
Sarcoma is the collective name for a relatively rare, yet heterogeneous group of cancers, most probably derived from mesenchymal tissues. There are currently over 50 sarcoma subtypes described underscoring the clinical and biologic diversity of this group of malignant cancers. This wide lineage range might suggest that sarcomas originate from either many committed different cell types or from a multipotent cell. Mesenchymal stem/progenitor cells (MSCs) are able to differentiate into many cell types and these multipotent cells have been isolated from several adult human tumors, making them available for research as well as potential beneficial therapeutical agents. Recent accomplishments in the field have broadened our knowledge of MSCs in relation to sarcoma origin and sarcoma treatment in therapeutic settings. However, numerous concerns and disputes have been raised about whether they are the putative originating cells of sarcoma and their questionable role in sarcomagenesis and progression. We summarize the update and dispute about MSC investigations in sarcomas including the definition, cell origin hypothesis, functional and descriptive assays, roles in sarcomagenesis and targeted therapy, with the purpose to give a comprehensive view of the role of MSCs in sarcomas.
Collapse
Affiliation(s)
- Jilong Yang
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Zhiwu Ren
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Xiaoling Du
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Mengze Hao
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Wenya Zhou
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| |
Collapse
|
47
|
Canter RJ, Ames E, Mac S, Grossenbacher SK, Chen M, Li CS, Borys D, Smith RC, Tellez J, Sayers TJ, Monjazeb AM, Murphy WJ. Anti-proliferative but not anti-angiogenic tyrosine kinase inhibitors enrich for cancer stem cells in soft tissue sarcoma. BMC Cancer 2014; 14:756. [PMID: 25301268 PMCID: PMC4200119 DOI: 10.1186/1471-2407-14-756] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/02/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Increasing studies implicate cancer stem cells (CSCs) as the source of resistance and relapse following conventional cytotoxic therapies. Few studies have examined the response of CSCs to targeted therapies, such as tyrosine kinase inhibitors (TKIs). We hypothesized that TKIs would have differential effects on CSC populations depending on their mechanism of action (anti-proliferative vs. anti-angiogenic). METHODS We exposed human sarcoma cell lines to sorafenib, regorafenib, and pazopanib and assessed cell viability and expression of CSC markers (ALDH, CD24, CD44, and CD133). We evaluated survival and CSC phenotype in mice harboring sarcoma metastases after TKI therapy. We exposed dissociated primary sarcoma tumors to sorafenib, regorafenib, and pazopanib, and we used tissue microarray (TMA) and primary sarcoma samples to evaluate the frequency and intensity of CSC markers after neoadjuvant therapy with sorafenib and pazopanib. Parametric and non-parametric statistical analyses were performed as appropriate. RESULTS After functionally validating the CSC phenotype of ALDHbright sarcoma cells, we observed that sorafenib and regorafenib were cytotoxic to sarcoma cell lines (P < 0.05), with a corresponding 1.4 - 2.8 fold increase in ALDHbright cells from baseline (P < 0.05). In contrast, we observed negligible effects on viability and CSC sub-populations with pazopanib. At low doses, there was progressive CSC enrichment in vitro after longer term exposure to sorafenib although the anti-proliferative effects were attenuated. In vivo, sorafenib improved median survival by 11 days (P < 0.05), but enriched ALDHbright cells 2.5 - 2.8 fold (P < 0.05). Analysis of primary human sarcoma samples revealed direct cytotoxicity following exposure to sorafenib and regorafenib with a corresponding increase in ALDHbright cells (P < 0.05). Again, negligible effects from pazopanib were observed. TMA analysis of archived specimens from sarcoma patients treated with sorafenib demonstrated significant enrichment for ALDHbright cells in the post-treatment resection specimen (P < 0.05), whereas clinical specimens obtained longitudinally from a patient treated with pazopanib showed no enrichment for ALDHbright cells (P > 0.05). CONCLUSIONS Anti-proliferative TKIs appear to enrich for sarcoma CSCs while anti-angiogenic TKIs do not. The rational selection of targeted therapies for sarcoma patients may benefit from an awareness of the differential impact of TKIs on CSC populations.
Collapse
Affiliation(s)
- Robert J Canter
- Department of Surgery, Division of Surgical Oncology, University of California Davis Medical Center, 4501 X Street, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Allegra A, Alonci A, Penna G, Innao V, Gerace D, Rotondo F, Musolino C. The cancer stem cell hypothesis: a guide to potential molecular targets. Cancer Invest 2014; 32:470-95. [PMID: 25254602 DOI: 10.3109/07357907.2014.958231] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Common cancer theories hold that tumor is an uncontrolled somatic cell proliferation caused by the progressive addition of random mutations in critical genes that control cell growth. Nevertheless, various contradictions related to the mutation theory have been reported previously. These events may be elucidated by the persistence of residual tumor cells, called Cancer Stem Cells (CSCs) responsible for tumorigenesis, tumor maintenance, tumor spread, and tumor relapse. Herein, we summarize the current understanding of CSCs, with a focus on the possibility to identify specific markers of CSCs, and discuss the clinical application of targeting CSCs for cancer treatment.
Collapse
|
49
|
TAMARI KEISUKE, HAYASHI KAZUHIKO, ISHII HIDESHI, KANO YOSHIHIRO, KONNO MASAMITSU, KAWAMOTO KOICHI, NISHIDA NAOHIRO, KOSEKI JUN, FUKUSUMI TAKAHITO, HASEGAWA SHINICHIRO, OGAWA HISATAKA, HAMABE ATSUSHI, MIYO MASAAKI, NOGUCHI KOZO, SEO YUJI, DOKI YUICHIRO, MORI MASAKI, OGAWA KAZUHIKO. Identification of chemoradiation-resistant osteosarcoma stem cells using an imaging system for proteasome activity. Int J Oncol 2014; 45:2349-54. [DOI: 10.3892/ijo.2014.2671] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 08/12/2014] [Indexed: 11/06/2022] Open
|
50
|
Sabbatino F, Wang Y, Wang X, Schwab JH, Ferrone S, Ferrone CR. Novel tumor antigen-specific monoclonal antibody-based immunotherapy to eradicate both differentiated cancer cells and cancer-initiating cells in solid tumors. Semin Oncol 2014; 41:685-99. [PMID: 25440613 DOI: 10.1053/j.seminoncol.2014.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
A growing body of experimental and clinical evidence strongly suggests that the resistance of cancer-initiating cells (CICs) to conventional therapies represents a major obstacle to the successful treatment of a malignant disease. To overcome this limitation a novel combinatorial tumor antigen (TA)-specific monoclonal antibody (mAb) strategy has been developed. In this strategy TA-specific mAbs are combined with chemotherapeutic agents and/or small molecules that inhibit aberrantly activated signaling pathways in cancer cells and especially in CICs. The in vitro results we have obtained indicate that this strategy is very effective in eradicating both differentiated cancer cells and CICs in several types of malignant disease. If the in vitro results have in vivo relevance, the strategy we have designed may have an impact on the treatment of malignant diseases.
Collapse
Affiliation(s)
- Francesco Sabbatino
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Yangyang Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Xinhui Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Joseph H Schwab
- Department Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|