251
|
Laws K, Bineva-Todd G, Eskandari A, Lu C, O'Reilly N, Suntharalingam K. A Copper(II) Phenanthroline Metallopeptide That Targets and Disrupts Mitochondrial Function in Breast Cancer Stem Cells. Angew Chem Int Ed Engl 2017; 57:287-291. [DOI: 10.1002/anie.201710910] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Kristine Laws
- Department of Chemistry; King's College London; London SE1 1DB UK
| | | | - Arvin Eskandari
- Department of Chemistry; King's College London; London SE1 1DB UK
| | - Chunxin Lu
- Department of Chemistry; King's College London; London SE1 1DB UK
| | - Nicola O'Reilly
- Peptide Chemistry; The Francis Crick Institute; London NW1 1AT UK
| | | |
Collapse
|
252
|
Singh M, Venugopal C, Tokar T, Brown KR, McFarlane N, Bakhshinyan D, Vijayakumar T, Manoranjan B, Mahendram S, Vora P, Qazi M, Dhillon M, Tong A, Durrer K, Murty N, Hallet R, Hassell JA, Kaplan DR, Cutz JC, Jurisica I, Moffat J, Singh SK. RNAi screen identifies essential regulators of human brain metastasis-initiating cells. Acta Neuropathol 2017; 134:923-940. [PMID: 28766011 DOI: 10.1007/s00401-017-1757-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/30/2022]
Abstract
Brain metastases (BM) are the most common brain tumor in adults and are a leading cause of cancer mortality. Metastatic lesions contain subclones derived from their primary lesion, yet their functional characterization is limited by a paucity of preclinical models accurately recapitulating the metastatic cascade, emphasizing the need for a novel approach to BM and their treatment. We identified a unique subset of stem-like cells from primary human patient brain metastases, termed brain metastasis-initiating cells (BMICs). We now establish a BMIC patient-derived xenotransplantation (PDXT) model as an investigative tool to comprehensively interrogate human BM. Using both in vitro and in vivo RNA interference screens of these BMIC models, we identified SPOCK1 and TWIST2 as essential BMIC regulators. SPOCK1 in particular is a novel regulator of BMIC self-renewal, modulating tumor initiation and metastasis from the lung to the brain. A prospective cohort of primary lung cancer specimens showed that SPOCK1 was overexpressed only in patients who ultimately developed BM. Protein-protein interaction network mapping between SPOCK1 and TWIST2 identified novel pathway interactors with significant prognostic value in lung cancer patients. Of these genes, INHBA, a TGF-β ligand found mutated in lung adenocarcinoma, showed reduced expression in BMICs with knockdown of SPOCK1. In conclusion, we have developed a useful preclinical model of BM, which has served to identify novel putative BMIC regulators, presenting potential therapeutic targets that block the metastatic process, and transform a uniformly fatal systemic disease into a locally controlled and eminently more treatable one.
Collapse
Affiliation(s)
- Mohini Singh
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Chitra Venugopal
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Tomas Tokar
- Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, ON, Canada
| | - Kevin R Brown
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre, Toronto, ON, Canada
| | - Nicole McFarlane
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - David Bakhshinyan
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Thusyanth Vijayakumar
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Branavan Manoranjan
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Sujeivan Mahendram
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Parvez Vora
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Maleeha Qazi
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Manvir Dhillon
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Amy Tong
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre, Toronto, ON, Canada
| | - Kathrin Durrer
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre, Toronto, ON, Canada
| | - Naresh Murty
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Robin Hallet
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - John A Hassell
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - David R Kaplan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - Jean-Claude Cutz
- Anatomic Pathology, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Igor Jurisica
- Princess Margaret Cancer Centre, IBM Life Sciences Discovery Centre, University Health Network, Toronto, ON, Canada
- Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, ON, Canada
| | - Jason Moffat
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre, Toronto, ON, Canada
| | - Sheila K Singh
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.
- Department of Surgery, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
253
|
Zhang Q, Yang Z, Shan J, Liu L, Liu C, Shen J, Chen X, Xu Y, Chen J, Ma Q, Yang L, Qian C. MicroRNA-449a maintains self-renewal in liver cancer stem-like cells by targeting Tcf3. Oncotarget 2017; 8:110187-110200. [PMID: 29299140 PMCID: PMC5746375 DOI: 10.18632/oncotarget.22705] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 10/29/2017] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) are thought to be responsible for tumor invasion, metastasis, and recurrence. We previously showed that the pluripotency factor Nanog not only serves as a novel biomarker of CSCs but also potentially plays a crucial role in maintaining the self-renewal ability of liver CSCs. However, how CSCs maintain Nanog gene expression has not been elucidated. Here, we demonstrated that microRNA-449a (miR-449a) is overexpressed in poorly differentiated hepatocellular carcinoma tissues, drug-resistant liver cancer cells, cultured liver tumorspheres, and Nanog-positive liver cancer cells. The upregulation of miR-449a in non-CSCs increased stemness, whereas the downregulation of miR-449a in Nanog-positive CSCs reduced stemness. Furthermore, transcription factor 3 (TCF3), a target of miR-449a, could downregulate Nanog expression, and restoring TCF3 expression in miR-449a-expressing Nanog-negative cells abrogated cellular stemness. These data establish that the miR449a-TCF3-Nanog axis maintains stemness in liver CSCs.
Collapse
Affiliation(s)
- Qianzhen Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, China.,College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Zhi Yang
- Center of Biological Therapy, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Juanjuan Shan
- Center of Biological Therapy, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Limei Liu
- Center of Biological Therapy, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Chungang Liu
- Center of Biological Therapy, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Junjie Shen
- Center of Biological Therapy, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Xuejiao Chen
- Center of Biological Therapy, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yanmin Xu
- Center of Biological Therapy, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Jun Chen
- Center of Biological Therapy, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Qinghua Ma
- Center of Biological Therapy, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, China.,College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Cheng Qian
- Center of Biological Therapy, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
254
|
Kim JS, Shin DH, Kim JS. Dual-targeting immunoliposomes using angiopep-2 and CD133 antibody for glioblastoma stem cells. J Control Release 2017; 269:245-257. [PMID: 29162480 DOI: 10.1016/j.jconrel.2017.11.026] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 10/12/2017] [Accepted: 11/17/2017] [Indexed: 01/10/2023]
Abstract
Glioblastoma stem cells (GSCs), which are identified as subpopulation of CD133+/ALDH1+, are known to show resistance to the most of chemotherapy and radiation therapy, leading to the recurrence of tumor in glioblastoma multiforme (GBM) patients. Also, delivery of temozolomide (TMZ), a mainline treatment of GBM, to the GBM site is hampered by various barriers including the blood-brain barrier (BBB). A dual-targeting immunoliposome encapsulating TMZ (Dual-LP-TMZ) was developed by using angiopep-2 (An2) and anti-CD133 monoclonal antibody (CD133 mAb) for BBB transcytosis and specific delivery to GSCs, respectively. The size, zeta potential and drug encapsulation efficiency of Dual-LP-TMZ were 203.4nm in diameter, -1.6mV and 99.2%, respectively. The in vitro cytotoxicity of Dual-LP-TMZ against U87MG GSCs was increased by 425- and 181-folds when compared with that of free TMZ and non-targeted TMZ liposome (LP-TMZ) (10.3μM vs. 4380μM and 1869μM in IC50, respectively). Apoptosis and anti-migration ability of Dual-LP-TMZ in U87MG GSCs were also significantly enhanced comparing with those of free TMZ or LP-TMZ. In vivo study clearly showed a significant reduction in tumor size after intravenous administrations of Dual-LP-TMZ to the orthotopically-implanted brain tumor mice when compared with free TMZ or LP-TMZ. Increased life span (ILS) and median survival time (MST) of tumor-bearing mice were also increased when treated with Dual-LP-TMZ (211.2% in ILS and 49.2days in MST) than with free TMZ (0% in ILS and 23.3day in MST). These data indicate that conjugation of both An2 peptide and CD133 mAb to TMZ-encapsulating liposome is very effective in delivering the TMZ to GSCs via BBB, suggesting a potential use of Dual-LP-TMZ as a therapeutic modality for GBM.
Collapse
Affiliation(s)
- Jung Seok Kim
- Research Center for Cell Fate Control (RCCFC), Drug Information Research Institute (DIRI), College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Dae Hwan Shin
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Jin-Seok Kim
- Research Center for Cell Fate Control (RCCFC), Drug Information Research Institute (DIRI), College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea.
| |
Collapse
|
255
|
Olejniczak A, Szaryńska M, Kmieć Z. In vitro characterization of spheres derived from colorectal cancer cell lines. Int J Oncol 2017; 52:599-612. [PMID: 29207035 DOI: 10.3892/ijo.2017.4206] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/08/2017] [Indexed: 11/06/2022] Open
Abstract
Spherical cultures (SCs) can be regarded in cancer research as a link between in vitro investigations on cancer lines and in vivo studies of tumor development. SCs are believed to mimic tumor architecture and to be enriched in cancer stem cell-like cells (CSC-like cells). In the present study we characterized colonospheres derived from colorectal cancer (CRC) cell lines, and we confirmed the ability of HCT116 and HT29 cell lines to form spheres within serum-free medium, however, the detailed analysis presented the major differences concerning their characteristics including morphology, phenotype, proliferative potential, distribution in the cell cycle phases and spherogenicity. Moreover, after we magnetically separated CD133+ and CD133- cells we could conduct the analogical analysis as we performed for the original cells. We observed that all cellular fractions unveiled sphere formation capacity, even when cultured in limited number of cells per well and only SCs originated from CD133+ fraction resembled morphologically the parental spheres. Both CD133+ and CD133- subsets derived from HCT116 line were more enriched in cells in G0/G1 phase of the cell cycle in comparison to their HT29 analogues. Additionally, proliferative potential also varied amongst all studied fractions. Surprisingly, 3-D invasion assay revealed that only HCT116-derived populations were able to migrate into extended regions of Matrigel Matrix confirming their higher aggressiveness. Our results provided comprehensive characterization of CRC cell lines culture in adherent and spherical forms and, what seems to be the most advantageous, the comparison of two distinct fractions after magnetic separation. As we found the specific features of cells presented line- and expansion mode-dependency, thus, such complete description might appear crucial before CRC lines would be involved into sophisticated assays, especially focused on potentially novel therapeutic agents targeting CSC-like cells.
Collapse
Affiliation(s)
- Agata Olejniczak
- Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Magdalena Szaryńska
- Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Zbigniew Kmieć
- Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
256
|
Ramos EK, Hoffmann AD, Gerson SL, Liu H. New Opportunities and Challenges to Defeat Cancer Stem Cells. Trends Cancer 2017; 3:780-796. [PMID: 29120754 PMCID: PMC5958547 DOI: 10.1016/j.trecan.2017.08.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 12/13/2022]
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer cells that are capable of self-renewal, proliferation, differentiation, plastic adaptation, and immune regulation, thereby mediating tumorigenesis, metastasis, and therapy resistance. CSCs are associated with cancer progression and clinical outcome in cancer patients. Successful targeting of CSCs will therefore be necessary to eradicate and cure cancer. Functional regulators of stem cell (stemness) signaling pathways in human cancers have brought new opportunities to target CSCs and reframe cancer-targeting strategies in clinical settings. However, challenges remain due to a lack of complete understanding of CSC plasticity/heterogeneity and the limited efficacy of individual stemness inhibitors in cancer treatment. In this article we review CSC signaling pathways and the current state of CSC-targeting therapeutics in combinatory treatments in clinical trials.
Collapse
Affiliation(s)
- Erika K Ramos
- Driskill Graduate Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department Pharmacology, Northwestern University, Chicago, IL, USA; These authors equally contributed to the manuscript preparation
| | - Andrew D Hoffmann
- Department Pharmacology, Northwestern University, Chicago, IL, USA; These authors equally contributed to the manuscript preparation
| | - Stanton L Gerson
- The Case Comprehensive Cancer Center, Cleveland, OH, USA; The National Center for Regenerative Medicine, Cleveland, OH, USA; Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
| | - Huiping Liu
- Department Pharmacology, Northwestern University, Chicago, IL, USA; The Case Comprehensive Cancer Center, Cleveland, OH, USA; The National Center for Regenerative Medicine, Cleveland, OH, USA; Department of Medicine, Division of Hematology and Oncology, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
257
|
Hsieh HY, Chuang HC, Shen FH, Detroja K, Hsin LW, Chen CS. Targeting breast cancer stem cells by novel HDAC3-selective inhibitors. Eur J Med Chem 2017; 140:42-51. [DOI: 10.1016/j.ejmech.2017.08.069] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/30/2017] [Accepted: 08/30/2017] [Indexed: 10/18/2022]
|
258
|
Tanno B, Leonardi S, Babini G, Giardullo P, De Stefano I, Pasquali E, Saran A, Mancuso M. Nanog-driven cell-reprogramming and self-renewal maintenance in Ptch1 +/- granule cell precursors after radiation injury. Sci Rep 2017; 7:14238. [PMID: 29079783 PMCID: PMC5660207 DOI: 10.1038/s41598-017-14506-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/11/2017] [Indexed: 12/31/2022] Open
Abstract
Medulloblastoma (MB) is the most common pediatric brain tumor, comprising four distinct molecular variants, one of which characterized by activation of the Sonic Hedgehog (SHH) pathway, driving 25–30% of sporadic MB. SHH-dependent MBs arise from granule cell precursors (GCPs), are fatal in 40–70% of cases and radioresistance strongly contributes to poor prognosis and tumor recurrence. Patched1 heterozygous (Ptch1+/−) mice, carrying a germ-line heterozygous inactivating mutation in the Ptch1 gene, the Shh receptor and negative regulator of the pathway, are uniquely susceptible to MB development after radiation damage in neonatal cerebellum. Here, we irradiated ex-vivo GCPs isolated from cerebella of neonatal WT and Ptch1+/− mice. Our results highlight a less differentiated status of Ptch1-mutated cells after irradiation, influencing DNA damage response. Increased expression levels of pluripotency genes Nanog, Oct4 and Sal4, together with greater clonogenic potential, clearly suggest that radiation induces expansion of the stem-like cell compartment through cell-reprogramming and self-renewal maintenance, and that this mechanism is strongly dependent on Nanog. These results contribute to clarify the molecular mechanisms that control radiation-induced Shh-mediated tumorigenesis and may suggest Nanog as a potential target to inhibit for adjuvant radiotherapy in treatment of SHH-dependent MB.
Collapse
Affiliation(s)
- Barbara Tanno
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy
| | - Simona Leonardi
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy
| | | | - Paola Giardullo
- Department of Radiation Physics, Guglielmo Marconi University, Rome, Italy.,Department of Sciences, Roma Tre University, Rome, Italy
| | - Ilaria De Stefano
- Department of Radiation Physics, Guglielmo Marconi University, Rome, Italy
| | - Emanuela Pasquali
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy
| | - Anna Saran
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy.
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy.
| |
Collapse
|
259
|
Li W, Ma H, Zhang J, Zhu L, Wang C, Yang Y. Unraveling the roles of CD44/CD24 and ALDH1 as cancer stem cell markers in tumorigenesis and metastasis. Sci Rep 2017; 7:13856. [PMID: 29062075 PMCID: PMC5653849 DOI: 10.1038/s41598-017-14364-2] [Citation(s) in RCA: 279] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/10/2017] [Indexed: 12/18/2022] Open
Abstract
CD44/CD24 and ALDH1 are widely used cancer stem cell (CSC) markers in breast cancer. However, their expression is not always consistent even in the same subtype of breast cancer. Systematic comparison of their functions is still lacking. We investigated the expression of CD44, CD24 and ALDH1 in different subtypes of breast cancer cells, and explored their relationship with cancer progression. We defined a parameter CD44/CD24 ratio to present the expression level of CD44 and CD24 and found that high CD44/CD24 ratio and ALDH1+ are both indicators for cancer malignancy, but play different functions during tumor progression. High CD44/CD24 ratio is more related to cell proliferation and tumorigenesis, which is confirmed by mammosphere formation and tumorigenesis in xenotransplanted mice. ALDH1+ is a stronger indicator for cell migration and tumor metastasis. Suppression of CD44 and ALDH1 by siRNA led to decreased tumorigenicity and cell migration capacity. The combination of high CD44/CD24 ratio and ALDH1+ would be a more reliable way to characterize CSCs. Moreover, both high CD44/CD24 ratio and ALDH1+ were conserved during metastasis, from the primary tumors to the circulating tumor cells (CTCs) and the distant metastases, suggesting the significant value of these CSC markers in assisting cancer detection, prognostic evaluation, and even cancer therapeutics.
Collapse
Affiliation(s)
- Wenzhe Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.,University of Chinese Academy of Sciences, 19 A Yuquan Rd, Shijingshan District, Beijing, P.R. China, 100049
| | - Huailei Ma
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, 19 A Yuquan Rd, Shijingshan District, Beijing, P.R. China, 100049
| | - Jin Zhang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China. .,University of Chinese Academy of Sciences, 19 A Yuquan Rd, Shijingshan District, Beijing, P.R. China, 100049.
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China. .,University of Chinese Academy of Sciences, 19 A Yuquan Rd, Shijingshan District, Beijing, P.R. China, 100049.
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China. .,University of Chinese Academy of Sciences, 19 A Yuquan Rd, Shijingshan District, Beijing, P.R. China, 100049.
| |
Collapse
|
260
|
Hodgkinson N, Kruger CA, Abrahamse H. Targeted photodynamic therapy as potential treatment modality for the eradication of colon cancer and colon cancer stem cells. Tumour Biol 2017; 39:1010428317734691. [PMID: 28990490 DOI: 10.1177/1010428317734691] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer is commonly treated by tumour resection, as chemotherapy and radiation have proven to be less effective, especially if the tumour has metastasized. Resistance to therapies occurs in almost all patients with colorectal cancer, especially in those with metastatic tumours. Cancer stem cells have the ability to self-renew, and their slow rate of cycling enhances resistance to treatment and increases the likelihood of tumour recurrence. Most metastatic tumours are unable to be surgically removed, thus creating a need for treatment modalities that target cancers directly and destroy cancer stem cells. Photodynamic therapy involves a photosensitizer that when exposed to a light source of a particular wavelength becomes excited and produces a form of oxygen that kills cancer cells. Photodynamic therapy is currently being investigated as a treatment modality for colorectal cancer, and new studies are exploring enhancing photodynamic therapy efficacy with the aid of drug carriers and immune conjugates. These modifications could prove effective in targeting cancer stem cells that are thought to be resistant to photodynamic therapy. In order for photodynamic therapy to be an effective treatment in colorectal cancer, it requires treatment of both primary tumours and the metastatic secondary disease that is caused by colon cancer stem cells. This review focuses on current photodynamic therapy treatments available for colorectal cancer and highlights proposed actively targeted photosynthetic drug uptake mechanisms specifically mediated towards colon cancer stem cells, as well as identify the gaps in research which need to be investigated in order to develop a combinative targeted photodynamic therapy regime that can effectively control colorectal cancer primary and metastatic tumour growth by eliminating colon cancer stem cells.
Collapse
Affiliation(s)
- Natasha Hodgkinson
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| | - Cherie A Kruger
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| |
Collapse
|
261
|
Hu K, Yin F, Yu M, Sun C, Li J, Liang Y, Li W, Xie M, Lao Y, Liang W, Li ZG. In-Tether Chiral Center Induced Helical Peptide Modulators Target p53-MDM2/MDMX and Inhibit Tumor Growth in Stem-Like Cancer Cell. Theranostics 2017; 7:4566-4576. [PMID: 29158845 PMCID: PMC5695149 DOI: 10.7150/thno.19840] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023] Open
Abstract
Inhibition of the interaction between p53 and MDM2/MDMX has attracted significant attention in anticancer therapy development. We designed a series of in-tether chiral center-induced helical stabilized peptides, among which MeR/PhR effectively reactivated p53. The activation of p53 inhibits cell proliferation and induces apoptosis in both the MCF-7 normal tumor cell line and the PA-1 pluripotent cancer cell line with only minimal cellular toxicity towards normal cells or cancer cell lines with p53 mutations. The in vivo bioactivity study of the peptide in the ovarian teratocarcinoma (PA-1) xenograft model showed a tumor growth rate inhibition of 70% with a dosage of 10 mg/kg (one injection every other day). This is the first application of a stabilized peptide modulator targeting stem-like cancer cell both in vitro and in vivo and provides references to cancer stem cell therapy.
Collapse
Affiliation(s)
- Kuan Hu
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Feng Yin
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Mengyin Yu
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Chengjie Sun
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Jingxu Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Yujie Liang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Wenjun Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Mingsheng Xie
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Yuanzhi Lao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wei Liang
- Department of Radiation Oncology, the First Affiliated Hospital, Anhui Medical University, Hefei, 230022, China
| | - Zi-gang Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| |
Collapse
|
262
|
Liu Z, Delavan B, Roberts R, Tong W. Lessons Learned from Two Decades of Anticancer Drugs. Trends Pharmacol Sci 2017; 38:852-872. [DOI: 10.1016/j.tips.2017.06.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 06/09/2017] [Accepted: 06/19/2017] [Indexed: 02/08/2023]
|
263
|
Gallardo-Pérez JC, Adán-Ladrón de Guevara A, Marín-Hernández A, Moreno-Sánchez R, Rodríguez-Enríquez S. HPI/AMF inhibition halts the development of the aggressive phenotype of breast cancer stem cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017. [DOI: 10.1016/j.bbamcr.2017.06.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
264
|
Pützer BM, Solanki M, Herchenröder O. Advances in cancer stem cell targeting: How to strike the evil at its root. Adv Drug Deliv Rev 2017; 120:89-107. [PMID: 28736304 DOI: 10.1016/j.addr.2017.07.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/10/2017] [Accepted: 07/16/2017] [Indexed: 12/18/2022]
Abstract
Cancer progression to metastatic stages is still unmanageable and the promise of effective anti-metastatic therapy remains largely unmet, emphasizing the need to develop novel therapeutics. The special focus here is on cancer stem cells (CSC) as the seed of tumor initiation, epithelial-mesenchymal transition, chemoresistance and, as a consequence, drivers of metastatic dissemination. We report on targeted therapies gearing towards the CSC's internal and membrane-anchored markers using agents such as antibody derivatives, nucleic therapeutics, small molecules and genetic payloads. Another emphasis lies on novel proceedings envisaged to deliver current and prospective therapies to the target sites using newest viral and non-viral vector technologies. In this review, we summarize recent progress and remaining challenges in therapeutic strategies to combat CSC.
Collapse
Affiliation(s)
- Brigitte M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany.
| | - Manish Solanki
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| | - Ottmar Herchenröder
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| |
Collapse
|
265
|
Downregulation of DNA repair proteins and increased DNA damage in hypoxic colon cancer cells is a therapeutically exploitable vulnerability. Oncotarget 2017; 8:86296-86311. [PMID: 29156796 PMCID: PMC5689686 DOI: 10.18632/oncotarget.21145] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/04/2017] [Indexed: 01/08/2023] Open
Abstract
Surgical removal of colorectal cancer (CRC) liver metastases generates areas of tissue hypoxia. Hypoxia imposes a stem-like phenotype on residual tumor cells and promotes tumor recurrence. Moreover, in primary CRC, gene expression signatures reflecting hypoxia and a stem-like phenotype are highly expressed in the aggressive Consensus Molecular Subtype 4 (CMS4). Therapeutic strategies eliminating hypoxic stem-like cells may limit recurrence following resection of primary tumors or metastases. Here we show that expression of DNA repair genes is strongly suppressed in CMS4 and inversely correlated with hypoxia-inducible factor-1 alpha (HIF1α) and HIF-2α co-expression signatures. Tumors with high expression of HIF signatures and low expression of repair proteins showed the worst survival. In human tumors, expression of the repair proteins RAD51, KU70 and RIF1 was strongly suppressed in hypoxic peri-necrotic tumor areas. Experimentally induced hypoxia in patient derived colonospheres in vitro or in vivo (through vascular clamping) was sufficient to downregulate repair protein expression and caused DNA damage. Hypoxia-induced DNA damage was prevented by expressing the hydroperoxide-scavenging enzyme glutathione peroxidase-2 (GPx2), indicating that reactive oxygen species mediate hypoxia-induced DNA damage. Finally, the hypoxia-activated prodrug Tirapazamine greatly augmented DNA damage and reduced the fraction of stem-like (Aldefluorbright) tumor cells in vitro, and in vivo following vascular clamping. We conclude that decreased expression of DNA repair proteins and increased DNA damage in hypoxic tumor areas may be therapeutically exploited with hypoxia-activated prodrugs, and that such drugs reduce the fraction of Aldefluorbright (stem-like) tumor cells.
Collapse
|
266
|
Xiao Y, Zhu S, Yin W, Liu X, Hu Y. IGFBP-4 expression is adversely associated with lung cancer prognosis. Oncol Lett 2017; 14:6876-6880. [PMID: 29163706 DOI: 10.3892/ol.2017.7014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 04/21/2017] [Indexed: 02/07/2023] Open
Abstract
Insulin-like growth factor binding protein-4 (IGFBP-4) was reported to be associated with prognosis in several types of cancer; however, to the best of our knowledge, whether it is correlated with lung cancer has yet to be reported. In the present study, 102 pairs of lung cancer tissues and surrounding non-cancerous tissues (SNCTs) were collected. The IGFBP-4 levels in tissues were detected with immunohistochemistry. The relevance of IGFBP-4 to the survival of patients was assessed. The IGFBP-4 gene was knocked down, and its function in the proliferation of lung cancer cells was measured. The percentage of lung cancer tissues with higher IGFBP-4 expression than SNCTs (51.9%) was increased compared with the percentage with similar (11.76%) or lower (36.27%) IGFBP-4 expression. Patients with higher IGFBP-1 expression exhibited a shorter median survival time. IGFBP-1 was associated with metastasis, lung cancer stages and malignancy, but not with age, sex or tumor size. Lung cancer cells with stably knocked down IGFBP-4 showed an inhibitory proliferation rate. The present study identified that IGFBP-4 was adversely associated with the prognosis of lung cancer patients. IGFBP-4 knockdown prohibited lung cancer cell growth. The present study provides a potential marker for lung cancer diagnosis and a possible target for lung cancer therapy.
Collapse
Affiliation(s)
- Yang Xiao
- Department of Respiratory Medicine, Central Hospital of Wuhan, Wuhan, Hubei 430014, P.R. China
| | - Shan Zhu
- Department of Respiratory Medicine, Central Hospital of Wuhan, Wuhan, Hubei 430014, P.R. China
| | - Wen Yin
- Department of Respiratory Medicine, Central Hospital of Wuhan, Wuhan, Hubei 430014, P.R. China
| | - Xiaofan Liu
- Department of Respiratory Medicine, Central Hospital of Wuhan, Wuhan, Hubei 430014, P.R. China
| | - Yi Hu
- Department of Respiratory Medicine, Central Hospital of Wuhan, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
267
|
Chen X, Song Q, Xia L, Xu X. Synergy of Dendritic Cell Vaccines and Avasimibe in Treatment of Head and Neck Cancer in Mice. Med Sci Monit 2017; 23:4471-4476. [PMID: 28918429 PMCID: PMC5614335 DOI: 10.12659/msm.905814] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The main purpose of this study was to explore the antitumor effect and mechanisms of ACAT1 inhibitor combined with CSCs-DC vaccine. MATERIAL AND METHODS We isolated HNSCC CSCs and gained CSCs antigens, then used CSCs antigens to load dendritic cells (DC) and generated a CSCs-DC vaccine. We treated mice after surgical excision of established SCC7 tumors with CSCs-DC vaccine and/or ACAT1 inhibitor, and recorded local tumor relapse and host survival. T cells and B cells were harvested from mice treated with CSCs-DC vaccine and/or ACAT1 inhibitor. We tested antibody production and the death rate of CSCs killed by T cells. RESULTS The tumors in the combined treatment group were smaller than in all other groups (P<0.01). The average survival time of the combined treatment group was 82 days and was the longest of all groups. Analysis of IgG levels secreted by B cell and CTL activity in spleens of mice found that results of the combined treatment group were the highest, and the results of the CSCs-DC group were lower than in the combined treatment group. The ACAT1 inhibitor group results were lower than in the CSCs-DC group and the combined treatment group results, but higher than in the PBS group, and the difference was statistically significant. CONCLUSIONS ACAT1 inhibitor enhanced the therapeutic effect of CSCs-DC vaccine in the treatment of the mouse HNSCC postoperative recurrence model. ACAT1 may play an important role in cancer immunotherapy.
Collapse
Affiliation(s)
- Xin Chen
- Department of Oncology, Wuhan University, Renmin Hospital, Wuhan, Hubei, China (mainland)
| | - Qibin Song
- Department of Oncology, Wuhan University, Renmin Hospital, Wuhan, Hubei, China (mainland)
| | - Leiming Xia
- Departmentof Hematology and Oncology, No. 1 People's Hospital of Hefei, Hefei, Anhui, China (mainland)
| | - Ximing Xu
- Department of Oncology, Wuhan University, Renmin Hospital, Wuhan, Hubei, China (mainland)
| |
Collapse
|
268
|
Schöning JP, Monteiro M, Gu W. Drug resistance and cancer stem cells: the shared but distinct roles of hypoxia-inducible factors HIF1α and HIF2α. Clin Exp Pharmacol Physiol 2017; 44:153-161. [PMID: 27809360 DOI: 10.1111/1440-1681.12693] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 12/19/2022]
Abstract
Chemotherapy resistance is a major contributor to poor treatment responses and tumour relapse, the development of which has been strongly linked to the action of cancer stem cells (CSCs). Mounting evidence suggests that CSCs are reliant on low oxygen conditions and hypoxia-inducible factors 1α and 2α (HIF1α and HIF2α) to maintain their stem cell features. Research in the last decade has begun to clarify the functional differences between the two HIFα subtypes (HIFαs). Here, we review and discuss these differences in relation to CSC-associated drug resistance. Both HIFαs contribute to CSC survival but play different roles -HIF1α being more responsible for survival functions and HIF2α for stemness traits such as self-renewal - and are sensitive to different degrees of hypoxia. Failure to account for physiologically relevant oxygen concentrations in many studies may influence the current understanding of the roles of HIFαs. We also discuss how hypoxia and HIFαs contribute to CSC drug resistance via promotion of ABC drug transporters Breast cancer resistance protein (BCRP), MDR1, and MRP1 and through maintenance of quiescence. Additionally, we explore the PI3K/AKT cell survival pathway that may support refractory cancer by promoting CSCs and activating both HIF1α and HIF2α. Accordingly, HIF1α and HIF2α inhibition, potentially via PI3K/AKT inhibitors, could reduce chemotherapy resistance and prevent cancer relapse.
Collapse
Affiliation(s)
- Jennifer Petra Schöning
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Michael Monteiro
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
269
|
Moriya C, Taniguchi H, Miyata K, Nishiyama N, Kataoka K, Imai K. Inhibition of PRDM14 expression in pancreatic cancer suppresses cancer stem-like properties and liver metastasis in mice. Carcinogenesis 2017; 38:638-648. [PMID: 28498896 DOI: 10.1093/carcin/bgx040] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 05/04/2017] [Indexed: 01/04/2023] Open
Abstract
Pancreatic cancer is one of the most lethal types of cancer, with aggressive properties characterized by metastasis, recurrence and drug resistance. Cancer stem cells are considered to be responsible for these properties. PRDM14, a transcriptional regulator that maintains pluripotency in embryonic stem cells, is overexpressed in some cancers. Here, we assessed PRDM14 expression and the effects of PRDM14 knockdown on cancer stem-like phenotypes in pancreatic cancer. We observed that PRDM14 protein was overexpressed in pancreatic cancer tissues compared with normal pancreatic tissues. Using lentiviral shRNA-transduced pancreatic cancer cells, we found that PRDM14 knockdown decreased sphere formation, number of side population and cell surface marker-positive cells and subcutaneous xenograft tumors and liver metastasis in mice. This was accompanied by upregulation of some microRNAs (miRNAs), including miR-125a-3p. miR-125a-3p, a tumor suppressor that is down-regulated in pancreatic cancer, has been suggested to regulate the expression of the Src-family kinase, Fyn. In PRDM14-knockdown cells, Fyn was expressed at lower levels and downstream proteins were less activated. These changes were considered to cause suppression of the above cancer phenotypes. In addition, we used small interfering RNA (siRNA)-based therapy targeting PRDM14 in a mouse model of liver metastasis induced using MIA-PaCa2 cells, and this treatment significantly decreased metastasis and in vitro migration. Taken together, these results suggest that targeting the overexpression of PRDM14 suppresses cancer stem-like phenotypes, including liver metastasis, via miRNA regulation and siRNA-based therapy targeting it shows promise as a treatment for patients with pancreatic cancer.
Collapse
Affiliation(s)
| | | | - Kanjiro Miyata
- Center for Antibody and Vaccine Therapy, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan, Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Nobuhiro Nishiyama
- Polymer Chemistry Division, Chemical Resources Laboratory, Tokyo Institute of Technology, Yokohama 226-8503, Japan and
| | - Kazunori Kataoka
- Center for Antibody and Vaccine Therapy, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan, Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Kohzoh Imai
- Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
270
|
Deshmukh A, Binju M, Arfuso F, Newsholme P, Dharmarajan A. Role of epigenetic modulation in cancer stem cell fate. Int J Biochem Cell Biol 2017; 90:9-16. [DOI: 10.1016/j.biocel.2017.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/31/2017] [Accepted: 07/11/2017] [Indexed: 01/16/2023]
|
271
|
Pindiprolu SKSS, Krishnamurthy PT, Chintamaneni PK, Karri VVSR. Nanocarrier based approaches for targeting breast cancer stem cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:885-898. [PMID: 28826237 DOI: 10.1080/21691401.2017.1366337] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Breast cancer stem cells (BCSCs) are heterogeneous subpopulation of tumour initiating cells within breast tumours. They are spared even after chemotherapy and responsible for tumour relapse. Targeting BCSCs is, therefore, necessary to achieve radical cure in breast cancer. Despite the availability of agents targeting BCSCs, their clinical application is limited due to their off-target effects and bioavailability issues. Nanotechnology based drug carriers (nanocarriers) offer various advantages to deliver anti-BCSCs agents specifically to their target sites by overcoming their bioavailability issues. In this review, we describe various strategies for targeting BCSCs using nanocarriers.
Collapse
Affiliation(s)
- Sai Kiran S S Pindiprolu
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Praveen T Krishnamurthy
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Pavan Kumar Chintamaneni
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Veera Venkata Satyanarayana Reddy Karri
- b Department of Pharmaceutics , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| |
Collapse
|
272
|
Abstract
Metastasis, the dissemination of cancer cells from primary tumors, represents a major hurdle in the treatment of cancer. The epithelial-mesenchymal transition (EMT) has been studied in normal mammalian development for decades, and it has been proposed as a critical mechanism during cancer progression and metastasis. EMT is tightly regulated by several internal and external cues that orchestrate the shifting from an epithelial-like phenotype into a mesenchymal phenotype, relying on a delicate balance between these two stages to promote metastatic development. EMT is thought to be induced in a subset of metastatic cancer stem cells (MCSCs), bestowing this population with the ability to spread throughout the body and contributing to therapy resistance. The EMT pathway is of increasing interest as a novel therapeutic avenue in the treatment of cancer, and could be targeted to prevent tumor cell dissemination in early stage patients or to eradicate existing metastatic cells in advanced stages. In this review, we describe the sequence of events and defining mechanisms that take place during EMT, and how these interactions drive cancer cell progression into metastasis. We summarize clinical interventions focused on targeting various aspects of EMT and their contribution to preventing cancer dissemination.
Collapse
Affiliation(s)
- Mohini Singh
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, L8S 4K1, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Nicolas Yelle
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, L8S 4K1, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Chitra Venugopal
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, L8S 4K1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Sheila K Singh
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, L8S 4K1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada.
| |
Collapse
|
273
|
Yin C, Qi X. Pregnancy promotes pituitary tumors by increasing the rate of the cell cycle. Oncol Lett 2017; 14:4873-4877. [PMID: 29085495 DOI: 10.3892/ol.2017.6756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 03/23/2017] [Indexed: 12/18/2022] Open
Abstract
Pituitary tumors may secrete hormones that affect pregnancy. Pregnancy also induces pituitary tumor growth; however, how pregnancy increases the growth of pituitary tumors remains unclear. The present study investigated pregnant female mice with subcutaneous pituitary tumors. The time of tumor occurrence and tumor weight were detected in pregnant and control mice. Tumor weights were measured at the end of the experiment. Blood was collected from pregnant and control mice. Brain-derived neurotrophic factor (BDNF) levels in the blood were detected using an ELISA kit. The in vitro effects of BDNF on pituitary tumor AtT-20 cell proliferation and cell cycle were investigated. It was revealed that pregnancy promoted the growth of pituitary tumors. In comparison to non-pregnant mice, the pregnant mice exhibited increased BDNF levels in the blood. In vitro BDNF treatment was able to increase the rate of proliferation of pituitary tumor cells. Additional cell cycle analysis revealed that BDNF was able to alter the cell cycle distribution of pituitary tumor cells. These results indicated that pregnancy was able to increase the BDNF level and promote the growth of pituitary tumor cells by increasing the rate of the cell cycle, leading to increased tumor growth rate in vivo. The present study provides insights into how pregnancy affects the growth of pituitary tumors. Therefore, it may be beneficial to perform pituitary tumor diagnosis or therapy on pregnant patients.
Collapse
Affiliation(s)
- Changjiang Yin
- Department of Neurosurgery, Shandong Traffic Hospital, Jinan, Shandong 250031, P.R. China
| | - Xiaoxia Qi
- Department of Obstetrics and Gynecology, Maternal and Child Health Care of Shandong, Jinan, Shandong 250031, P.R. China
| |
Collapse
|
274
|
MacDonagh L, Gallagher MF, Ffrench B, Gasch C, Breen E, Gray SG, Nicholson S, Leonard N, Ryan R, Young V, O'Leary JJ, Cuffe S, Finn SP, O'Byrne KJ, Barr MP. Targeting the cancer stem cell marker, aldehyde dehydrogenase 1, to circumvent cisplatin resistance in NSCLC. Oncotarget 2017; 8:72544-72563. [PMID: 29069808 PMCID: PMC5641151 DOI: 10.18632/oncotarget.19881] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/03/2017] [Indexed: 12/15/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for a large proportion of cancer deaths and is characterized by low treatment response rates and poor overall prognosis. In the absence of specific treatable mutations, cisplatin-based chemotherapy plays an important role in the treatment of this disease. Unfortunately, the development of resistance has become a major therapeutic challenge in the use of this cytotoxic drug. Elucidating the mechanisms underlying this resistance phenotype, may result in the development of novel agents that enhance sensitivity to cisplatin in lung cancer patients. In this study, targeting the cancer stem cell activity of aldehyde dehydrogenase 1 (ALDH1) was investigated as a strategy to overcome chemoresistance in NSCLC. Tumors from NSCLC patients showed an increase in their profile of pluripotent stemness genes. Cisplatin exposure induced the emergence or expansion of an ALDH1-positive subpopulation in cisplatin sensitive and resistant NSCLC cell lines, respectively, further enhancing cisplatin resistance. Using the Aldefluor assay and FACS analysis, ALDH1 subpopulations were isolated and evaluated in terms of stem cell characteristics. Only ALDH1-positive cells exhibited asymmetric division, cisplatin resistance and increased expression of stem cell factors in vitro. Xenograft studies in NOD/SCID mice demonstrated efficient tumorigenesis from low cell numbers of ALDH1-positive and ALDH1-negative subpopulations. Targeting ALDH1 with Diethylaminobenzaldehyde (DEAB) and Disulfiram, significantly re-sensitized resistant lung cancer cells to the cytotoxic effects of cisplatin. Our data demonstrate the existence of a lung CSC population and suggest a role for targeting ALDH1 as a potential therapeutic strategy in re-sensitizing NSCLC cells to the cytotoxic effects of cisplatin.
Collapse
Affiliation(s)
- Lauren MacDonagh
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital & Trinity College, Dublin, Ireland
| | - Michael F Gallagher
- Histopathology Department, Trinity College Dublin, Sir Patrick Dun Laboratories & Central Pathology Laboratory, St. James's Hospital, Pathology Research Laboratory, Coombe Women and Infant's University Hospital, Dublin, Ireland
| | - Brendan Ffrench
- Histopathology Department, Trinity College Dublin, Sir Patrick Dun Laboratories & Central Pathology Laboratory, St. James's Hospital, Pathology Research Laboratory, Coombe Women and Infant's University Hospital, Dublin, Ireland
| | - Claudia Gasch
- Histopathology Department, Trinity College Dublin, Sir Patrick Dun Laboratories & Central Pathology Laboratory, St. James's Hospital, Pathology Research Laboratory, Coombe Women and Infant's University Hospital, Dublin, Ireland
| | - Eamon Breen
- Flow Cytometry Facility, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital & Trinity College, Dublin, Ireland
| | - Steven G Gray
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital & Trinity College, Dublin, Ireland
| | | | - Niamh Leonard
- Histopathology Department, St. James's Hospital, Dublin, Ireland
| | - Ronan Ryan
- Department of Cardiothoracic Surgery, St. James's Hospital, Dublin, Ireland
| | - Vincent Young
- Department of Cardiothoracic Surgery, St. James's Hospital, Dublin, Ireland
| | - John J O'Leary
- Histopathology Department, Trinity College Dublin, Sir Patrick Dun Laboratories & Central Pathology Laboratory, St. James's Hospital, Pathology Research Laboratory, Coombe Women and Infant's University Hospital, Dublin, Ireland
| | - Sinead Cuffe
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital & Trinity College, Dublin, Ireland
| | - Stephen P Finn
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital & Trinity College, Dublin, Ireland.,Histopathology Department, St. James's Hospital, Dublin, Ireland
| | - Kenneth J O'Byrne
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital & Trinity College, Dublin, Ireland.,Cancer & Ageing Research Program, Queensland University of Technology, Brisbane, Australia
| | - Martin P Barr
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital & Trinity College, Dublin, Ireland
| |
Collapse
|
275
|
Lu C, Eskandari A, Cressey PB, Suntharalingam K. Cancer Stem Cell and Bulk Cancer Cell Active Copper(II) Complexes with Vanillin Schiff Base Derivatives and Naproxen. Chemistry 2017; 23:11366-11374. [PMID: 28658520 DOI: 10.1002/chem.201701939] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Indexed: 01/03/2023]
Abstract
Four copper(II) complexes, 1-4 containing regioisomeric vanillin Schiff base derivatives and the nonsteroidal anti-inflammatory drug (NSAID), naproxen, were synthesised and characterised. All complexes effectively cleave DNA in cell-free systems, with 4 displaying the highest nuclease activity. DNA binding studies suggest that 4 binds to DNA via the grooves prior to inducing oxidative DNA cleavage. Three of the complexes (1, 3, and 4) indiscriminately kill cancer stem cell (CSC)-enriched cells (HMLER-shEcad) and bulk cancer cells (HMLER) at micromolar concentrations. The most effective complex, 4 also reduced the formation and size of mammospheres to a similar extent as salinomycin, a well-established CSC-potent agent. Mechanistic studies show that 4 is readily taken up by CSCs, elevates intracellular reactive oxygen species (ROS) levels, causes DNA damage, and induces caspase-dependent apoptosis. Furthermore, 4 inhibits cyclooxygenase-2 (COX-2) expression and causes COX-2-dependent CSC death. The advantage of 4 over bulk cancer cell- or CSC-selective agents is that it has the potential to remove whole tumor populations (bulk cancer cells and CSCs) with a single dose.
Collapse
Affiliation(s)
- Chunxin Lu
- Department of Chemistry, King's College London, London, SE1 1DB, United Kingdom
| | - Arvin Eskandari
- Department of Chemistry, King's College London, London, SE1 1DB, United Kingdom
| | - Paul B Cressey
- Department of Chemistry, King's College London, London, SE1 1DB, United Kingdom
| | | |
Collapse
|
276
|
A New Chapter for Mesenchymal Stem Cells: Decellularized Extracellular Matrices. Stem Cell Rev Rep 2017; 13:587-597. [DOI: 10.1007/s12015-017-9757-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
277
|
Lee SY, Kim JK, Jeon HY, Ham SW, Kim H. CD133 Regulates IL-1β Signaling and Neutrophil Recruitment in Glioblastoma. Mol Cells 2017; 40:515-522. [PMID: 28736425 PMCID: PMC5547221 DOI: 10.14348/molcells.2017.0089] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/29/2017] [Accepted: 06/29/2017] [Indexed: 12/31/2022] Open
Abstract
CD133, a pentaspan transmembrane glycoprotein, is generally used as a cancer stem cell marker in various human malignancies, but its biological function in cancer cells, especially in glioma cells, is largely unknown. Here, we demonstrated that forced expression of CD133 increases the expression of IL-1β and its downstream chemokines, namely, CCL3, CXCL3 and CXCL5, in U87MG glioma cells. Although there were no apparent changes in cell growth and sphere formation in vitro and tumor growth in vivo, in vitro trans-well studies and in vivo tumor xenograft assays showed that neutrophil recruitment was markedly increased by the ectopic expression of CD133. In addition, the clinical relevance between CD133 expression and IL-1β gene signature was established in patients with malignant gliomas. Thus, these results imply that glioma cells expressing CD133 are capable of modulating tumor microenvironment through the IL-1β signaling pathway.
Collapse
Affiliation(s)
- Seon Yong Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841,
Korea
| | - Jun-Kyum Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841,
Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841,
Korea
| | - Hee-Young Jeon
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841,
Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841,
Korea
| | - Seok Won Ham
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841,
Korea
| | - Hyunggee Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841,
Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841,
Korea
| |
Collapse
|
278
|
Singh VK, Saini A, Chandra R. The Implications and Future Perspectives of Nanomedicine for Cancer Stem Cell Targeted Therapies. Front Mol Biosci 2017; 4:52. [PMID: 28785557 PMCID: PMC5520001 DOI: 10.3389/fmolb.2017.00052] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/07/2017] [Indexed: 12/14/2022] Open
Abstract
Cancer stem cells (CSCs) are believed to exhibit distinctive self-renewal, proliferation, and differentiation capabilities, and thus play a significant role in various aspects of cancer. CSCs have significant impacts on the progression of tumors, drug resistance, recurrence and metastasis in different types of malignancies. Due to their primary role, most researchers have focused on developing anti-CSC therapeutic strategies, and tremendous efforts have been put to explore methods for selective eradication of these therapeutically resistant CSCs. In recent years, many reports have shown the use of CSCs-specific approaches such as ATP-binding cassette (ABC) transporters, blockade of self-renewal and survival of CSCs, CSCs surface markers targeted drugs delivery and eradication of the tumor microenvironment. Also, various therapeutic agents such as small molecule drugs, nucleic acids, and antibodies are said to destroy CSCs selectively. Targeted drug delivery holds the key to the success of most of the anti-CSCs based drugs/therapies. The convention CSCs-specific therapeutic agents, suffer from various problems. For instance, limited water solubility, small circulation time and inconsistent stability of conventional therapeutic agents have significantly limited their efficacy. Recent advancement in the drug delivery technology has demonstrated that specially designed nanocarrier-based drug delivery approaches (nanomedicine) can be useful in delivering sufficient amount of drug molecules even in the most interiors of CSCs niches and thus can overcome the limitations associated with the conventional free drug delivery methods. The nanomedicine has also been promising in designing effective therapeutic regime against pump-mediated drug resistance (ATP-driven) and reduces detrimental effects on normal stem cells. Here we focus on the biological processes regulating CSCs' drug resistance and various strategies developed so far to deal with them. We also review the various nanomedicine approaches developed so far to overcome these CSCs related issues and their future perspectives.
Collapse
Affiliation(s)
- Vimal K. Singh
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological UniversityNew Delhi, India
| | - Abhishek Saini
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological UniversityNew Delhi, India
| | - Ramesh Chandra
- Department of Chemistry, University of DelhiNew Delhi, India
| |
Collapse
|
279
|
Gilbert AN, Walker K, Tran AN, Boyd NH, Gillespie GY, Singh RK, Hjelmeland AB. Modeling Physiologic Microenvironments in Three-Dimensional Microtumors Maintains Brain Tumor Initiating Cells. JOURNAL OF CANCER STEM CELL RESEARCH 2017; 5:e1004. [PMID: 29075651 PMCID: PMC5653320 DOI: 10.14343/jcscr.2017.5e1004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Development of effective novel anti-tumor treatments will require improved in vitro models that incorporate physiologic microenvironments and maintain intratumoral heterogeneity, including tumor initiating cells. Brain tumor initiating cells (BTIC) are a target for cancer therapy, because BTICs are highly tumorigenic and contribute to tumor angiogenesis, invasion, and therapeutic resistance. Current leading studies rely on BTIC isolation from patient-derived xenografts followed by propagation as neurospheres. As this process is expensive and time-consuming, we determined whether three-dimensional microtumors were an alternative in vitro method for modeling tumor growth via BITC maintenance and/or enrichment. Brain tumor cells were grown as neurospheres or as microtumors produced using the human-derived biomatrix HuBiogel™ and maintained with physiologically relevant microenvironments. BITC percentages were determined using cell surface marker expression, label retention, and neurosphere formation capacity. Our data demonstrate that expansion of brain tumor cells as hypoxic and nutrient-restricted microtumors significantly increased the percentage of both CD133+ and CFSEhigh cells. We further demonstrate that BTIC-marker positive cells isolated from microtumors maintained neurosphere formation capacity in the in vitro limiting dilution assay and tumorigenic potential in vivo. These data demonstrate that microtumors can be a useful three-dimensional biological model for the study of BTIC maintenance and targeting.
Collapse
Affiliation(s)
| | - Kiera Walker
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Anh Nhat Tran
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Nathaniel H. Boyd
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - G. Yancey Gillespie
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL
| | | | - Anita B. Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
280
|
Boo L, Ho WY, Mohd Ali N, Yeap SK, Ky H, Chan KG, Yin WF, Satharasinghe DA, Liew WC, Tan SW, Cheong SK, Ong HK. Phenotypic and microRNA transcriptomic profiling of the MDA-MB-231 spheroid-enriched CSCs with comparison of MCF-7 microRNA profiling dataset. PeerJ 2017; 5:e3551. [PMID: 28717596 PMCID: PMC5511503 DOI: 10.7717/peerj.3551] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 06/16/2017] [Indexed: 12/16/2022] Open
Abstract
Breast cancer spheroids have been widely used as in vitro models of cancer stem cells (CSCs), yet little is known about their phenotypic characteristics and microRNAs (miRNAs) expression profiles. The objectives of this research were to evaluate the phenotypic characteristics of MDA-MB-231 spheroid-enriched cells for their CSCs properties and also to determine their miRNAs expression profile. Similar to our previously published MCF-7 spheroid, MDA-MB-231 spheroid also showed typical CSCs characteristics namely self-renewability, expression of putative CSCs-related surface markers and enhancement of drug resistance. From the miRNA profile, miR-15b, miR-34a, miR-148a, miR-628 and miR-196b were shown to be involved in CSCs-associated signalling pathways in both models of spheroids, which highlights the involvement of these miRNAs in maintaining the CSCs features. In addition, unique clusters of miRNAs namely miR-205, miR-181a and miR-204 were found in basal-like spheroid whereas miR-125, miR-760, miR-30c and miR-136 were identified in luminal-like spheroid. Our results highlight the roles of miRNAs as well as novel perspectives of the relevant pathways underlying spheroid-enriched CSCs in breast cancer.
Collapse
Affiliation(s)
- Lily Boo
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Selangor, Malaysia
| | - Wan Yong Ho
- Department of Biomedical Sciences, University of Nottingham, Semenyih, Selangor, Malaysia
| | - Norlaily Mohd Ali
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Selangor, Malaysia
| | | | - Huynh Ky
- Department of Genetics and Plant Breeding, Cantho University, Cantho, Vietnam
| | - Kok Gan Chan
- Division of Genetics and Molecular Biology, University of Malaya, Kuala Lumpur, Malaysia
| | - Wai Fong Yin
- Division of Genetics and Molecular Biology, University of Malaya, Kuala Lumpur, Malaysia
| | - Dilan Amila Satharasinghe
- Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Peradeniya, Central, Sri Lanka
| | - Woan Charn Liew
- Institute of Bioscience, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Sheau Wei Tan
- Institute of Bioscience, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Soon Keng Cheong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Selangor, Malaysia.,Cryocord Sdn Bhd, Cyberjaya, Selangor, Malaysia
| | - Han Kiat Ong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Selangor, Malaysia
| |
Collapse
|
281
|
Li X, Lu J, Kan Q, Li X, Fan Q, Li Y, Huang R, Slipicevic A, Dong HP, Eide L, Wang J, Zhang H, Berge V, Goscinski MA, Kvalheim G, Nesland JM, Suo Z. Metabolic reprogramming is associated with flavopiridol resistance in prostate cancer DU145 cells. Sci Rep 2017; 7:5081. [PMID: 28698547 PMCID: PMC5506068 DOI: 10.1038/s41598-017-05086-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 05/24/2017] [Indexed: 01/19/2023] Open
Abstract
Flavopiridol (FP) is a pan-cyclin dependent kinase inhibitor, which shows strong efficacy in inducing cancer cell apoptosis. Although FP is potent against most cancer cells in vitro, unfortunately it proved less efficacious in clinical trials in various aggressive cancers. To date, the molecular mechanisms of the FP resistance are mostly unknown. Here, we report that a small fraction human prostate cancer DU145 cells can survive long-term FP treatment and emerge as FP-resistant cells (DU145FP). These DU145FP cells show accumulated mitochondrial lesions with stronger glycolytic features, and they proliferate in slow-cycling and behave highly migratory with strong anti-apoptotic potential. In addition, the cells are less sensitive to cisplatin and docetaxel-induced apoptotic pressure, and over-express multiple stem cell associated biomarkers. Our studies collectively uncover for the first time that FP-resistant prostate cancer cells show metabolic remodeling, and the metabolic plasticity might be required for the FP resistance-associated cancer cell stemness up-regulation.
Collapse
Affiliation(s)
- Xiaoran Li
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
- Department of Pathology, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0318, Norway
| | - Jie Lu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Quancheng Kan
- Department of Clinical Pharmacology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Xiaoli Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Qiong Fan
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0316, Norway
| | - Yaqing Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruixia Huang
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
| | - Ana Slipicevic
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
| | - Hiep Phuc Dong
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
| | - Lars Eide
- Department of Medical Biochemistry, University of Oslo and Oslo University Hospital, Oslo, 0372, Norway
| | - Junbai Wang
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
| | - Hongquan Zhang
- Laboratory of Molecular Cell Biology and Tumor Biology, Department of Anatomy, Histology and Embryology, Peking University Health Science Center, Beijing, 100191, China
| | - Viktor Berge
- Department of Urology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
| | - Mariusz Adam Goscinski
- Departments of Surgery, The Norwegian Radium Hospital, Oslo University Hospital, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0379, Norway
| | - Gunnar Kvalheim
- Department of Cell Therapy, Cancer Institute, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
| | - Jahn M Nesland
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
- Department of Pathology, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0318, Norway
| | - Zhenhe Suo
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway.
- Department of Pathology, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0318, Norway.
| |
Collapse
|
282
|
Kiro NE, Hamblin MR, Abrahamse H. Photobiomodulation of breast and cervical cancer stem cells using low-intensity laser irradiation. Tumour Biol 2017; 39:1010428317706913. [PMID: 28653884 PMCID: PMC5564223 DOI: 10.1177/1010428317706913] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Breast and cervical cancers are dangerous threats with regard to the health of women. The two malignancies have reached the highest record in terms of cancer-related deaths among women worldwide. Despite the use of novel strategies with the aim to treat and cure advanced stages of cancer, post-therapeutic relapse believed to be caused by cancer stem cells is one of the challenges encountered during tumor therapy. Therefore, further attention should be paid to cancer stem cells when developing novel anti-tumor therapeutic approaches. Low-intensity laser irradiation is a form of phototherapy making use of visible light in the wavelength range of 630-905 nm. Low-intensity laser irradiation has shown remarkable results in a wide range of medical applications due to its biphasic dose and wavelength effect at a cellular level. Overall, this article focuses on the cellular responses of healthy and cancer cells after treatment with low-intensity laser irradiation alone or in combination with a photosensitizer as photodynamic therapy and the influence that various wavelengths and fluencies could have on the therapeutic outcome. Attention will be paid to the biomodulative effect of low-intensity laser irradiation on cancer stem cells.
Collapse
Affiliation(s)
- N E Kiro
- 1 Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| | - M R Hamblin
- 1 Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa.,2 Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA.,3 Department of Dermatology, Harvard Medical School, Boston, MA, USA.,4 Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - H Abrahamse
- 1 Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| |
Collapse
|
283
|
Liang Y, Goyette S, Hyder SM. Cholesterol biosynthesis inhibitor RO 48-8071 reduces progesterone receptor expression and inhibits progestin-dependent stem cell-like cell growth in hormone-dependent human breast cancer cells. BREAST CANCER-TARGETS AND THERAPY 2017; 9:487-494. [PMID: 28744156 PMCID: PMC5511027 DOI: 10.2147/bctt.s140265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Clinical trials and studies have shown that postmenopausal women undergoing combination hormone replacement therapy containing estrogen and progestin have an increased risk of breast cancer compared with women taking estrogen or placebo alone. Using animal models, we have previously shown that synthetic progestins, including medroxyprogesterone acetate (MPA), which is widely used clinically, accelerate breast cancer tumor growth and promote metastasis. Furthermore, we have found that MPA elevates CD44 protein expression and aldehyde dehydrogenase (ALDH) activity, two markers of cancer stem cells (CSCs), and increases mammosphere formation, another hallmark of stem cells, in hormone-dependent T47-D human breast cancer cells. Herein, we show that RO 48-8071 (RO), an inhibitor of cholesterol synthesis, reduced MPA-induced CD44 protein expression in two hormone-dependent human breast cancer cell lines, T47-D and BT-474. Because we have previously shown that MPA induction of CD44 is progesterone receptor (PR) dependent, we examined RO’s effects on PR protein and mRNA expressions in T47-D cells. PR mRNA levels remained unchanged after RO treatment; however, RO significantly reduced the protein expression of both PR receptor isoforms, PR-A and PR-B. Using the proteasome inhibitor MG-132, we demonstrated that RO decreases PR protein expression in T47-D cells via the proteasomal degradation pathway. Importantly, treatment of T47-D cells with RO abolished MPA-induced mammosphere formation. Based on our observations, we contend that RO may represent a novel means of preventing MPA-induced CSC expansion. RO could be used clinically to both treat and prevent hormone-dependent breast cancers, which represent the majority of human breast cancers. RO may also have clinical utility in reducing resistance to antihormone therapy.
Collapse
Affiliation(s)
- Yayun Liang
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Sandy Goyette
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Salman M Hyder
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
284
|
Characterization of the new human pleomorphic undifferentiated sarcoma TP53-null cell line mfh-val2. Cytotechnology 2017; 69:539-550. [PMID: 28676915 DOI: 10.1007/s10616-017-0112-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/21/2016] [Indexed: 01/01/2023] Open
Abstract
Pleomorphic undifferentiated sarcoma (PUS), also called malignant fibrous histiocytoma, is a soft tissue sarcoma which occurs predominantly in the extremities. Its origin is a poorly defined mesenchymal cell, which derives to histiocytic and fibroblastic cells. The patient, a 58 year-old man, presented a lesion located in the forearm composed by spindle cells and multinucleated giant cells, which expressed vimentin and adopted a histological pattern formed by irregular-swirling fascicles. Cells were cultured in vitro and a new cell line was established. We characterized this new cell line by histological analyses, cytogenetics (using G-bands and spectral karyotype technique) and cytometric analyses. Cells were grown in culture for more than 100 passages. They had elongated or polygonal morphology. The cells presented a saturation rate of 70,980 cells/cm2, a plating efficiency of 21.5% and a mitotic index of 21 mitoses per field. The cell line was tumorigenic in nude mice. The ploidy study using flow cytometry revealed an aneuploid peak with a DNA index of 1.43. A side population was detected, demonstrating the presence of stem and progenitor cells. Cytogenetics showed a hypotriploid range with many clonal unbalanced rearrangements. Loss of p53 gene was evidenced by MLPA. We describe, for the first time, the characterization of a new human PUS TP53-null cell line called mfh-val2. Mfh-val2 presents a wide number of applications as a TP53-null cell line and a great interest in order to characterize genetic alterations influencing the oncogenesis or progression of PUS and to advance in the biological investigation of this tumor.
Collapse
|
285
|
Hubbard JM, Grothey A. Napabucasin: An Update on the First-in-Class Cancer Stemness Inhibitor. Drugs 2017; 77:1091-1103. [PMID: 28573435 DOI: 10.1007/s40265-017-0759-4] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Napabucasin (BBI608) is an orally administered small molecule that blocks stem cell activity in cancer cells by targeting the signal transducer and activator of transcription 3 pathway. The signal transducer and activator of transcription 3 pathway is over-activated in many types of cancer and has been shown to be an important pathway in cancer stem cell-mediated propagation of cancer. Cancer stem cells are a subpopulation of cancer cells considered to be the primary source of tumor growth, metastasis, and resistance to conventional therapies, and thus, responsible for cancer relapse. This review describes the clinical development program of this first-in-class cancer stemness inhibitor, including preclinical discovery, early clinical trials, current phase III clinical trial evaluation, and future therapeutic combinations. The therapeutic potential of napabucasin was first reported in a preclinical study that demonstrated the potent anti-tumor and anti-metastatic activity of napabucasin in several different cancer types, both in vitro and in vivo. In mouse models, napabucasin was effective both as a monotherapy and in combination with other agents; in particular, synergy was observed with paclitaxel in vivo. Napabucasin clinical trials have demonstrated encouraging anti-tumor activity as monotherapy and in combination with conventional therapeutics, with no significant pharmacokinetic interactions when used in combination therapies. Adverse events attributed to napabucasin have been predominantly mild, although some patients have experienced grade 3 gastrointestinal adverse events. More severe adverse events required reduced or discontinued dosing of napabucasin or medication to reverse or manage symptoms. In conclusion, napabucasin may prove useful in targeting cancer stem cells, with the potential to suppress metastasis and prevent relapse in patients with varying cancer types.
Collapse
Affiliation(s)
- Joleen M Hubbard
- Department of Medical Oncology, Mayo Clinic Rochester, 200 First Street NW, Rochester, MN, 55905, USA
| | - Axel Grothey
- Department of Medical Oncology, Mayo Clinic Rochester, 200 First Street NW, Rochester, MN, 55905, USA.
| |
Collapse
|
286
|
Kharkar PS. Cancer stem cell (CSC) inhibitors: a review of recent patents (2012-2015). Expert Opin Ther Pat 2017; 27:753-761. [PMID: 28460551 DOI: 10.1080/13543776.2017.1325465] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 04/27/2017] [Indexed: 02/08/2023]
Abstract
Cancer stem cells (CSCs) mediate tumor initiation and maintenance. These cells are chemoresistant and possess characteristics such as self-renewal, pluripotency, plasticity and differentiation. They have aberrant or altered signaling pathways depending on tumor microenvironment, tumor type, etc. CSCs are responsible for highly aggressive and invasive form of the disease following chemo- and/or radiotherapy. Eliminating CSCs is likely to improve the survival rate in patients. Several anti-CSC strategies and associated targets have been proposed and validated till date. Areas covered: The main emphasis is on the patent applications/patents filed/granted in the last few years (2012-2015). The anti-CSC agents are discussed under two broad headings - small- and macromolecules. Different subclasses are further elaborated, e.g., kinase inhibitors, polypeptides, etc. Expert opinion: Clinical development of small- and macromolecular anti-CSC therapeutics is underway. Few of these agents act on validated targets such as kinases. Potential problems with these agents can be envisaged based on our understanding of target biology. Other issues governing the choice of small- versus macromolecules include druggability of the target, ease of its modulation and the presence of compensatory mechanisms. Drug repurposing can be attempted to discover newer anti-CSC drugs quickly.
Collapse
Affiliation(s)
- Prashant S Kharkar
- a Department of Pharmaceutical Chemistry, SPP School of Pharmacy and Technology Management , SVKM's NMIMS , Mumbai , India
| |
Collapse
|
287
|
Cai J, Fang L, Huang Y, Li R, Xu X, Hu Z, Zhang L, Yang Y, Zhu X, Zhang H, Wu J, Huang Y, Li J, Zeng M, Song E, He Y, Zhang L, Li M. Simultaneous overactivation of Wnt/β-catenin and TGFβ signalling by miR-128-3p confers chemoresistance-associated metastasis in NSCLC. Nat Commun 2017. [PMID: 28627514 PMCID: PMC5481840 DOI: 10.1038/ncomms15870] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer chemoresistance and metastasis are tightly associated features. However, whether they share common molecular mechanisms and thus can be targeted with one common strategy remain unclear in non-small cell lung cancer (NSCLC). Here, we report that high levels of microRNA-128-3p (miR-128-3p) is key to concomitant development of chemoresistance and metastasis in residual NSCLC cells having survived repeated chemotherapy and correlates with chemoresistance, aggressiveness and poor prognosis in NSCLC patients. Mechanistically, miR-128-3p induces mesenchymal and stemness-like properties through downregulating multiple inhibitors of Wnt/β-catenin and TGF-β pathways, leading to their overactivation. Importantly, antagonism of miR-128-3p potently reverses metastasis and chemoresistance of highly malignant NSCLC cells, which could be completely reversed by restoring Wnt/β-catenin and TGF-β activities. Notably, correlations among miR-128-3p levels, activated β-catenin and TGF-β signalling, and pro-epithelial-to-mesenchymal transition/pro-metastatic protein levels are validated in NSCLC patient specimens. These findings suggest that miR-128-3p might be a potential target against both metastasis and chemoresistance in NSCLC.
Collapse
Affiliation(s)
- Junchao Cai
- Department of Microbiology, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China.,Guangdong Engineering and Technology Research Center for Disease-Model Animals, Sun Yat-sen University, Guangzhou 510006, China
| | - Lishan Fang
- Department of Microbiology, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China.,Central Laboratory of The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Yongbo Huang
- State Key Laboratory of Respiratory Diseases and Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Rong Li
- Department of Microbiology, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Xiaonan Xu
- Department of Microbiology, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Zhihuang Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Le Zhang
- Department of Microbiology, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Yi Yang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xun Zhu
- Department of Microbiology, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Heng Zhang
- Neurosurgery Intensive Care Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jueheng Wu
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Yan Huang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Jun Li
- Department of Biochemistry, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510080, China
| | - Musheng Zeng
- State Key Laboratory of Oncology in South China, Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Erwei Song
- Department of Breast Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yukai He
- Department of Medicine and Department of Biochemistry and Molecular Biology, Georgia Cancer Center, Augusta University, Augusta, Georgia 30912, USA
| | - Li Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Mengfeng Li
- Department of Microbiology, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| |
Collapse
|
288
|
Nestvold J, Wang MY, Camilio KA, Zinöcker S, Tjelle TE, Lindberg A, Haug BE, Kvalheim G, Sveinbjørnsson B, Rekdal Ø. Oncolytic peptide LTX-315 induces an immune-mediated abscopal effect in a rat sarcoma model. Oncoimmunology 2017; 6:e1338236. [PMID: 28920000 PMCID: PMC5593701 DOI: 10.1080/2162402x.2017.1338236] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/26/2017] [Accepted: 05/27/2017] [Indexed: 02/07/2023] Open
Abstract
LTX 315 is an oncolytic peptide with potent immunological properties. In the present study, we demonstrate that intratumoral treatment with LTX-315 resulted in a complete regression and systemic immune response in a rat fibrosarcoma model. The treatment was T-cell dependent, and also resulted in an abscopal effect as demonstrated by the regression of distal non-treated lesions. Significant infiltration of CD8+ T cells was observed in both treated and non-treated lesions, as shown by immunohistochemical and flow cytometric analysis. LTX-315 rapidly killed the cells in vitro with a lytic mode of action followed by the subsequent release of Danger-Associated Molecular Pattern (DAMP) molecules such as HMGB1, ATP and Cytochrome c. Together, our data demonstrate that LTX-315 represents a new approach to cancer immunotherapy, which has the potential as a novel immunotherapeutic agent.
Collapse
Affiliation(s)
- Janne Nestvold
- Department of Transplantation Medicine, Institute for Surgical Research, Oslo University Hospital, NO-0732 Oslo, Norway
| | - Meng-Yu Wang
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, NO-0379 Oslo, Norway.,Department of Cellular Therapy, Oslo University Hospital, NO-0379 Oslo, Norway
| | - Ketil A Camilio
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, NO-0379 Oslo, Norway.,Lytix Biopharma, Gaustadalléen 21, NO-0349 Oslo, Norway
| | - Severin Zinöcker
- Department of Anatomy, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, N-0317, Oslo, Norway
| | - Torunn Elisabeth Tjelle
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, N-0317, Oslo, Norway
| | - Alf Lindberg
- Lytix Biopharma, Gaustadalléen 21, NO-0349 Oslo, Norway
| | - Bengt Erik Haug
- Department of Chemistry and Centre for Pharmacy, University of Bergen, Allégaten 41, NO-5007 Bergen, Norway
| | - Gunnar Kvalheim
- Department of Cellular Therapy, Oslo University Hospital, NO-0379 Oslo, Norway
| | - Baldur Sveinbjørnsson
- Lytix Biopharma, Gaustadalléen 21, NO-0349 Oslo, Norway.,Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, NO-9037 Tromsø, Norway
| | - Øystein Rekdal
- Lytix Biopharma, Gaustadalléen 21, NO-0349 Oslo, Norway.,Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, NO-9037 Tromsø, Norway
| |
Collapse
|
289
|
Peng Y, He G, Tang D, Xiong L, Wen Y, Miao X, Hong Z, Yao H, Chen C, Yan S, Lu L, Yang Y, Li Q, Deng X. Lovastatin Inhibits Cancer Stem Cells and Sensitizes to Chemo- and Photodynamic Therapy in Nasopharyngeal Carcinoma. J Cancer 2017; 8:1655-1664. [PMID: 28775785 PMCID: PMC5535721 DOI: 10.7150/jca.19100] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 03/31/2017] [Indexed: 02/06/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus-associated malignancy occurring at high incidence in Southeast Asia and southern China. In spite of the good response to radio- and chemo-therapy at the early stage, resistance and recurrence develop in NPC patients in the advanced setting. Cancer stem cells (CSCs) play an important role in drug resistance and cancer recurrence. Here we report that lovastatin, a natural compound and a lipophilic statin that has already been used in the clinic to treat hypercholesterolemia, inhibited the CSC properties and induced apoptosis and cell cycle arrest in sphere-forming cells derived from the 5-8F and 6-10B NPC cell lines. Furthermore, lovastatin conferred enhanced sensitivity to the chemotherapeutic and photodynamic agents in NPC CSCs. Together our findings suggest that targeting CSCs by lovastatin in combination with routine chemotherapeutic drugs or photodynamic therapy might be a promising approach to the treatment of NPC.
Collapse
Affiliation(s)
- Yikun Peng
- Department of Otorhinolaryngology-Head and Neck Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Guangchun He
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Da Tang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Xiong
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiongying Miao
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Hui Yao
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Chao Chen
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Shichao Yan
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Lu Lu
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Yingke Yang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Qinglong Li
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiyun Deng
- Medical College, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
290
|
Parrales A, Ranjan A, Iwakuma T. Unsaturated fatty acids regulate stemness of ovarian cancer cells through NF-κB. Stem Cell Investig 2017; 4:49. [PMID: 28607923 DOI: 10.21037/sci.2017.05.07] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 04/25/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Alejandro Parrales
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Atul Ranjan
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
291
|
Giovannetti E, van der Borden CL, Frampton AE, Ali A, Firuzi O, Peters GJ. Never let it go: Stopping key mechanisms underlying metastasis to fight pancreatic cancer. Semin Cancer Biol 2017; 44:43-59. [PMID: 28438662 DOI: 10.1016/j.semcancer.2017.04.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/12/2017] [Accepted: 04/18/2017] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive neoplasm, predicted to become the second leading cause of cancer-related deaths before 2030. This dismal trend is mainly due to lack of effective treatments against its metastatic behavior. Therefore, a better understanding of the key mechanisms underlying metastasis should provide new opportunities for therapeutic purposes. Genomic analyses revealed that aberrations that fuel PDAC tumorigenesis and progression, such as SMAD4 loss, are also implicated in metastasis. Recently, microRNAs have been shown to play a regulatory role in the metastatic behavior of many tumors, including PDAC. In particular, miR-10 and miR-21 have appeared as master regulators of the metastatic program, while members of the miR-200 family are involved in the epithelial-to-mesenchymal switch, favoring cell migration and invasiveness. Several studies have also found a close relationship between cancer stem cells (CSCs) and biological features of metastasis, and the CSC markers ALDH1, ABCG2 and c-Met are expressed at high levels in metastatic PDAC cells. Emerging evidence reveals that exosomes are involved in the modulation of the tumor microenvironment and can initiate PDAC pre-metastatic niche formation in the liver and lungs. In this review, we provide an overview of the role of all these pivotal factors in the metastatic behavior of PDAC, and discuss their potential exploitation in the clinic to improve current therapeutics and identify new drug targets.
Collapse
Affiliation(s)
- E Giovannetti
- Lab Medical Oncology, Dept. Medical Oncology, VU University Medical Center (VUmc), Amsterdam, The Netherlands; Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, Pisa, Italy
| | - C L van der Borden
- Lab Medical Oncology, Dept. Medical Oncology, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - A E Frampton
- HPB Surgical Unit, Dept. of Surgery & Cancer, Imperial College, Hammersmith Hospital Campus, London, UK
| | - A Ali
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, KP, Pakistan; Institute of Cancer Sciences, University of Glasgow, UK
| | - O Firuzi
- Lab Medical Oncology, Dept. Medical Oncology, VU University Medical Center (VUmc), Amsterdam, The Netherlands; Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - G J Peters
- Lab Medical Oncology, Dept. Medical Oncology, VU University Medical Center (VUmc), Amsterdam, The Netherlands.
| |
Collapse
|
292
|
Singh D, Minz AP, Sahoo SK. Nanomedicine-mediated drug targeting of cancer stem cells. Drug Discov Today 2017; 22:952-959. [DOI: 10.1016/j.drudis.2017.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/03/2017] [Accepted: 04/12/2017] [Indexed: 12/16/2022]
|
293
|
Amey CL, Karnoub AE. Targeting Cancer Stem Cells-A Renewed Therapeutic Paradigm. ONCOLOGY & HEMATOLOGY REVIEW 2017; 13:45-55. [PMID: 33959299 PMCID: PMC8098671 DOI: 10.17925/ohr.2017.13.01.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Metastasis is often accompanied by radio- and chemotherapeutic resistance to anticancer treatments and is the major cause of death in cancer patients. Better understanding of how cancer cells circumvent therapeutic insults and how disseminated cancer clones generate life-threatening metastases would therefore be paramount to the development of effective therapeutic approaches for clinical management of malignant disease. Mounting reports over the past two decades have provided evidence for the existence of a minor population of highly malignant cells within liquid and solid tumors, which are capable of self-renewing and of regenerating secondary growths with the heterogeneity of the primary tumors from which they derive. These cells, called tumor-initiating cells or cancer stem cells (CSCs) exhibit increased resistance to standard radio- and chemotherapies and appear to have mechanisms that enable them to evade immune surveillance. CSCs are therefore considered to be responsible for systemic residual disease after cancer therapy, as well as for disease relapse. How CSCs develop, the nature of the interactions they establish with their microenvironment, their phenotypic and functional characteristics, as well as their molecular dependencies have all taken center stage in cancer therapy. Indeed, improved understanding of CSC biology is critical to the development of important CSC-based anti-neoplastic approaches that have the potential to radically improve cancer management. Here, we summarize some of the most pertinent elements regarding CSC development and properties, and highlight some of the clinical modalities in current development as anti-CSC therapeutics.
Collapse
Affiliation(s)
| | - Antoine E Karnoub
- Department of Pathology, Beth Israel Deaconess Cancer Center and Harvard Medical School, Boston, Massachusetts, US; Harvard Stem Cell Institute, Cambridge, Massachusetts, US; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, US
| |
Collapse
|
294
|
Goyette S, Liang Y, Mafuvadze B, Cook MT, Munir M, Hyder SM. Natural and synthetic progestins enrich cancer stem cell-like cells in hormone-responsive human breast cancer cell populations in vitro. BREAST CANCER-TARGETS AND THERAPY 2017; 9:347-357. [PMID: 28579829 PMCID: PMC5446973 DOI: 10.2147/bctt.s135371] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Clinical trials and studies have shown that combination estrogen/progestin hormone replacement therapy, but not estrogen therapy alone or placebo, increases breast cancer risk in postmenopausal women. Using animal models, we have previously shown that both natural and synthetic progestins (including medroxyprogesterone acetate [MPA], a synthetic progestin used widely in the clinical setting) accelerate the development of breast tumors in vivo and increase their metastasis to lymph nodes. Based on these observations, we have hypothesized that progestin-induced breast cancer tumor growth and metastasis may be mediated by an enrichment of the cancer stem cell (CSC) pool. In this study, we used T47-D and BT-474 hormone-responsive human breast cancer cells to examine the effects of progestin on phenotypic and functional markers of CSCs in vitro. Both natural and synthetic progestins (10 nM) significantly increased protein expression of CD44, an important CSC marker in tumor cells. MPA increased the levels of both CD44 variants v3 and v6 associated with stem cell functions. This induction of CD44 was blocked by the antiprogestin RU-486, suggesting that this process is progesterone receptor (PR) dependent. CD44 induction was chiefly progestin dependent. Because RU-486 can bind other steroid receptors, we treated PR-negative T47-DCO-Y cells with MPA and found that MPA failed to induce CD44 protein expression, confirming that PR is essential for progestin-mediated CD44 induction in T47-D cells. Further, MPA treatment of T47-D cells significantly increased the activity of aldehyde dehydrogenase (ALDH), another CSC marker. Finally, two synthetic progestins, MPA and norethindrone, significantly increased the ability of T47-D cells to form mammospheres, suggesting that enrichment of the CD44high, ALDHbright subpopulation of cancer cells induced by MPA exposure is of functional significance. Based on our observations, we contend that exposure of breast cancer cells to synthetic progestins leads to an enrichment of the CSC pool, supporting the development of progestin-accelerated tumors in vivo.
Collapse
Affiliation(s)
- Sandy Goyette
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Yayun Liang
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Benford Mafuvadze
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Matthew T Cook
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Moiz Munir
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Salman M Hyder
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
295
|
Lu CS, Shieh GS, Wang CT, Su BH, Su YC, Chen YC, Su WC, Wu P, Yang WH, Shiau AL, Wu CL. Chemotherapeutics-induced Oct4 expression contributes to drug resistance and tumor recurrence in bladder cancer. Oncotarget 2017; 8:30844-30858. [PMID: 27244887 PMCID: PMC5458172 DOI: 10.18632/oncotarget.9602] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 04/23/2016] [Indexed: 12/18/2022] Open
Abstract
Cancer cells initially characterized as sensitive to chemotherapy may acquire resistance to chemotherapy and lead to tumor recurrence through the expansion of drug-resistant population. Acquisition of drug resistance to conventional chemotherapy is a major obstacle in the treatment of recurrent cancer. Here we investigated whether anticancer drugs induced Oct4 expression, thereby contributing to acquired drug resistance and tumor recurrence in bladder cancer. We identified a positive correlation of Oct4 expression with tumor recurrence in 122 clinical specimens of superficial high-grade (stages T1-2) bladder transitional cell carcinoma (TCC). Increased Oct4 levels in bladder tumors were associated with short recurrence-free intervals in the patients. Chemotherapy induced Oct4 expression in bladder cancer cells. Notably, treatment with cisplatin increased CD44-positive bladder cancer cells expressing Oct4, representing cancer stem-like cell subpopulation. Forced expression of Oct4 reduced, whereas knockdown of Oct4 enhanced, drug sensitivity in bladder cancer cells. Furthermore, tumor cells overexpressing Oct4 responded poorly to cisplatin in vivo. In regard to clinical relevance, inhibition of Oct4 by all-trans retinoic acid (ATRA) synergistically increased sensitivity to cisplatin in bladder cancer cells. Furthermore, the combination of cisplatin and ATRA was superior to cisplatin alone in suppressing tumor growth. Therefore, our results provide evidence that Oct4 increases drug resistance and implicate that inhibition of Oct4 may be a therapeutic strategy to circumvent drug resistance.
Collapse
Affiliation(s)
- Chia-Sing Lu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Gia-Shing Shieh
- Department of Urology, Tainan Hospital, Ministry of Health and Welfare, Executive Yuan, Tainan, Taiwan
| | - Chung-Teng Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bing-Hua Su
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Chu Su
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Cheng Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wu-Chou Su
- Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pensee Wu
- Institute for Science & Technology in Medicine, Keele University, Keele, United Kingdom
| | - Wen-Horng Yang
- Department of Urology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ai-Li Shiau
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chao-Liang Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
296
|
Seo Y, Kim YS, Lee KE, Park TH, Kim Y. Anti-cancer stemness and anti-invasive activity of bitter taste receptors, TAS2R8 and TAS2R10, in human neuroblastoma cells. PLoS One 2017; 12:e0176851. [PMID: 28467517 PMCID: PMC5414998 DOI: 10.1371/journal.pone.0176851] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 04/18/2017] [Indexed: 01/01/2023] Open
Abstract
Neuroblastoma (NB) originates from immature neuronal cells and currently has a poor clinical outcome. NB cells possess cancer stem cells (CSCs) characteristics that facilitate the initiation of a tumor, as well as its metastasis. Human bitter taste receptors, referred to as TAS2Rs, are one of five types of basic taste receptors and they belong to a family of G-protein coupled receptors. The recent finding that taste receptors are expressed in non-gustatory tissues suggest that they mediate additional functions distinct from taste perception. While it is generally admitted that the recognition of bitter tastes may be associated with a self-defense system to prevent the ingestion of poisonous food compounds, this recognition may also serve as a disease-related function in the human body. In particular, the anti-cancer stemness and invasion effects of TAS2Rs on NB cells remain poorly understood. In the present study, endogenous expression of TAS2R8 and TAS2R10 in SK-N-BE(2)C and SH-SY5Y cells was examined. In addition, higher levels of TAS2R8 and TAS2R10 expression were investigated in more differentiated SY5Y cells. Both TAS2Rs were up-regulated following the induction of neuronal cell differentiation by retinoic acid. In addition, ectopic transfection of the two TAS2Rs induced neurite elongation in the BE(2)C cells, and down-regulated CSCs markers (including DLK1, CD133, Notch1, and Sox2), and suppressed self-renewal characteristics. In particular, TAS2RS inhibited tumorigenicity. Furthermore, when TAS2Rs was over-expressed, cell migration, cell invasion, and matrix metalloproteinases activity were inhibited. Expression levels of hypoxia-inducible factor-1α, a well-known regulator of tumor metastasis, as well as its downstream targets, vascular endothelial growth factor and glucose transporter-1, were also suppressed by TAS2Rs. Taken together, these novel findings suggest that TAS2Rs targets CSCs by suppressing cancer stemness characteristics and NB cell invasion, thereby highlighting the chemotherapeutic potential of bitter taste receptors.
Collapse
Affiliation(s)
- Yoona Seo
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Yoo-Sun Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Kyung Eun Lee
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Yuri Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
297
|
Aponte PM, Caicedo A. Stemness in Cancer: Stem Cells, Cancer Stem Cells, and Their Microenvironment. Stem Cells Int 2017; 2017:5619472. [PMID: 28473858 PMCID: PMC5394399 DOI: 10.1155/2017/5619472] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 01/31/2017] [Accepted: 02/19/2017] [Indexed: 02/06/2023] Open
Abstract
Stemness combines the ability of a cell to perpetuate its lineage, to give rise to differentiated cells, and to interact with its environment to maintain a balance between quiescence, proliferation, and regeneration. While adult Stem Cells display these properties when participating in tissue homeostasis, Cancer Stem Cells (CSCs) behave as their malignant equivalents. CSCs display stemness in various circumstances, including the sustaining of cancer progression, and the interaction with their environment in search for key survival factors. As a result, CSCs can recurrently persist after therapy. In order to understand how the concept of stemness applies to cancer, this review will explore properties shared between normal and malignant Stem Cells. First, we provide an overview of properties of normal adult Stem Cells. We thereafter elaborate on how these features operate in CSCs. We then review the organization of microenvironment components, which enables CSCs hosting. We subsequently discuss Mesenchymal Stem/Stromal Cells (MSCs), which, although their stemness properties are limited, represent essential components of the Stem Cell niche and tumor microenvironment. We next provide insights of the therapeutic strategies targeting Stem Cell properties in tumors and the use of state-of-the-art techniques in future research. Increasing our knowledge of the CSCs microenvironment is key to identifying new therapeutic solutions.
Collapse
Affiliation(s)
- Pedro M. Aponte
- Colegio de Ciencias Biológicas y Ambientales, Universidad San Francisco de Quito (USFQ), 170901 Quito, Ecuador
- Colegio de Ciencias de la Salud, Escuela de Medicina Veterinaria, Universidad San Francisco de Quito (USFQ), 170901 Quito, Ecuador
- Mito-Act Research Consortium, Quito, Ecuador
| | - Andrés Caicedo
- Mito-Act Research Consortium, Quito, Ecuador
- Colegio de Ciencias de la Salud, Escuela de Medicina, Universidad San Francisco de Quito (USFQ), 170901 Quito, Ecuador
- Colegio de Ciencias Biológicas y Ambientales, Instituto de Microbiología, Universidad San Francisco de Quito (USFQ), 170901 Quito, Ecuador
| |
Collapse
|
298
|
Eun K, Ham SW, Kim H. Cancer stem cell heterogeneity: origin and new perspectives on CSC targeting. BMB Rep 2017; 50:117-125. [PMID: 27998397 PMCID: PMC5422023 DOI: 10.5483/bmbrep.2017.50.3.222] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Indexed: 12/14/2022] Open
Abstract
Most of the cancers are still incurable human diseases. According to recent findings, especially targeting cancer stem cells (CSCs) is the most promising therapeutic strategy. CSCs take charge of a cancer hierarchy, harboring stem cell-like properties involving self-renewal and aberrant differentiation potential. Most of all, the presence of CSCs is closely associated with tumorigenesis and therapeutic resistance. Despite the numerous efforts to target CSCs, current anti-cancer therapies are still impeded by CSC-derived cancer malignancies; increased metastases, tumor recurrence, and even acquired resistance against the anti-CSC therapies developed in experimental models. One of the most forceful underlying reasons is a “cancer heterogeneity” due to “CSC plasticity” A comprehensive understanding of CSC-derived heterogeneity will provide novel insights into the establishment of efficient targeting strategies to eliminate CSCs. Here, we introduce findings on mechanisms of CSC reprogramming and CSC plasticity, which give rise to phenotypically varied CSCs. Also, we suggest concepts to improve CSC-targeted therapy in order to overcome therapeutic resistance caused by CSC plasticity and heterogeneity.
Collapse
Affiliation(s)
- Kiyoung Eun
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Seok Won Ham
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Hyunggee Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| |
Collapse
|
299
|
Ingallina C, Costa PM, Ghirga F, Klippstein R, Wang JT, Berardozzi S, Hodgins N, Infante P, Pollard SM, Botta B, Al-Jamal KT. Polymeric glabrescione B nanocapsules for passive targeting of Hedgehog-dependent tumor therapy in vitro. Nanomedicine (Lond) 2017; 12:711-728. [PMID: 28322108 PMCID: PMC5986025 DOI: 10.2217/nnm-2016-0388] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/26/2017] [Indexed: 01/09/2023] Open
Abstract
AIM With the purpose of delivering high doses of glabrescione B (GlaB) to solid tumors after systemic administration, long-circulating GlaB-loaded oil-cored polymeric nanocapsules (NC-GlaB) were formulated. MATERIALS & METHODS Synthesis of GlaB and its encapsulation in nanocapsules (NCs) was performed. Empty and GlaB-loaded NCs were assessed for their physico-chemical properties, in vitro cytotoxicity and in vivo biodistribution. RESULTS GlaB was efficiently loaded into NCs (∽90%), which were small (∽160 nm), homogeneous and stable upon storage. Further, GlaB and NC-GlaB demonstrated specific activities against the cancer stem cells. Preliminary studies in tumor-bearing mice supported the ability of NC to accumulate in pancreatic tumors. CONCLUSION This study provides early evidence that NC-GlaB has the potential to be utilized in a preclinical setting and justifies the need to perform therapeutic experiments in mice.
Collapse
Affiliation(s)
- Cinzia Ingallina
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Rome, Italy
| | - Pedro M Costa
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
| | - Francesca Ghirga
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Rebecca Klippstein
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
| | - Julie T Wang
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
| | - Simone Berardozzi
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Rome, Italy
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Naomi Hodgins
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
| | - Paola Infante
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Steven M Pollard
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Bruno Botta
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Rome, Italy
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin−Wilkins Building, London, SE1 9NH, UK
| |
Collapse
|
300
|
Therapeutic resistance and cancer recurrence mechanisms: Unfolding the story of tumour coming back. J Biosci 2017; 41:497-506. [PMID: 27581940 DOI: 10.1007/s12038-016-9624-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer recurrence is believed to be one of the major reasons for the failure of cancer treatment strategies. This biological phenomenon could arise from the incomplete eradication of tumour cells after chemo- and radiotherapy. Recent developments in the design of models reflecting cancer recurrence and in vivo imaging techniques have led researchers to gain a deeper and more detailed insight into the mechanisms underlying tumour relapse. Here, we provide an overview of three important drivers of recurrence including cancer stem cells (CSCs), neosis, and phoenix rising. The survival of cancer stem cells is well recognized as one of the primary causes of therapeutic resistance in malignant cells. CSCs have a relatively latent metabolism and show resistance to therapeutic agents through a variety of routes. Neosis has proven to be as an important mechanism behind tumour self-proliferation after treatment which gives rise to the expansion of tumour cells in the injured site via production of Raju cells. Phoenix rising is a prorecurrence pathway through which apoptotic cancer cells send strong signals to the neighbouring diseased cells leading to their multiplication. The mechanisms involved in therapeutic resistance and tumour recurrence have not yet been fully understood and mostly remain unexplained. Without doubt, an improved understanding of the cellular machinery contributing to recurrence will pave the way for the development of novel, sophisticated and effective antitumour therapeutic strategies which can eradicate tumour without the threat of relapse.
Collapse
|