151
|
Mortezazadeh T, Gholibegloo E, Khoobi M, Alam NR, Haghgoo S, Mesbahi A. In vitro and in vivo characteristics of doxorubicin-loaded cyclodextrine-based polyester modified gadolinium oxide nanoparticles: a versatile targeted theranostic system for tumour chemotherapy and molecular resonance imaging. J Drug Target 2019; 28:533-546. [PMID: 31842616 DOI: 10.1080/1061186x.2019.1703188] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
β-Cyclodextrine-based polyester was coated on the surface of gadolinium oxide nanoparticles (NPs) and then functionalised with folic acid to produce an efficient pH-sensitive targeted theranostic system (Gd2O3@PCD-FA) for doxorubicin delivery and magnetic resonance imaging (MRI). Gd2O3@PCD-FA was fully characterised by FTIR, vibrating sample magnetometer, TGA, XRD, SEM and TEM analyses. The dissolution profile of DOX showed a pH sensitive release. No significant toxicity was observed for the targeted NPs (Gd2O3@PCD-FA) and DOX-loaded NPs inhibiting M109 cells viability more efficiently than free DOX. Moreover, the negligible hemolytic activity of the targeted NPs showed their appropriate hemocompatibility. The preferential uptake was observed for the developed Gd2O3@PCD-FA-DOX NPs in comparison with Dotarem using T1- and T2-weighted MRI in the presence of folate receptor-positive and folate receptor-negative cancer cells (M109 and 4T1, respectively). Furthermore, in vivo studies revealed that Gd2O3@PCD-FA-DOX not only exhibited considerably relaxivity performance as a contrast agent for MRI, but also improved in vivo anti-tumour efficacy of the system. The results suggest that Gd2O3@PCD-FA-DOX improves its therapeutic efficacy in the treatment of solid tumours and also reduces the adverse effects, so it could be proposed as a promising drug delivery system for chemotherapy and molecular imaging diagnosis in MRI.
Collapse
Affiliation(s)
- Tohid Mortezazadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Gholibegloo
- Biomaterials Group, The Institute of Pharmaceutical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Khoobi
- Biomaterials Group, The Institute of Pharmaceutical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Nader Riyahi Alam
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical, Sciences, Tehran, Iran
| | - Soheila Haghgoo
- Pharmaceutical Department, Food and Drug Laboratory Research Center, Food and Drug Organization (FDO), Ministry of Health, Tehran, Iran
| | - Asghar Mesbahi
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
152
|
Lathia J, Liu H, Matei D. The Clinical Impact of Cancer Stem Cells. Oncologist 2019; 25:123-131. [PMID: 32043793 DOI: 10.1634/theoncologist.2019-0517] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/23/2019] [Indexed: 01/10/2023] Open
Abstract
Patients with cancer can go though many stages in their disease, including diagnosis, recurrence, metastasis, and treatment failure. Cancer stem cells (CSCs) are a subgroup of cells within tumors that may explain the mechanism by which tumors recur and progress. CSCs can both self-renew and produce progenitor cells of more differentiated cancer cells as well as heterogeneously demonstrate resistance and the abilities to migrate and metastasize. These "stemness" characteristics are often the result of dysregulation of one or more pathways, which can be detected by various biomarkers. Although there has been considerable laboratory research conducted on CSCs, its relevance to the practicing oncologist may seem questionable. We sought to determine the clinical impact of CSCs on patients. A systematic literature search was conducted to identify analyses containing survival information based on the expression of known CSC biomarkers in any cancer. Overall, 234 survival analyses were identified, of which 82% reported that high expression of CSC biomarker(s) resulted in poor overall survival and/or disease-free survival compared with low or no expression of the biomarker. Elevated stemness biomarker levels were also associated with decreased tumor differentiation, altered TNM stage, and increased metastasis. This analysis would suggest that CSCs have a clinical impact on patients and that practicing oncologists need to start considering incorporating CSC-targeting therapies into their patients' treatment regimens. IMPLICATIONS FOR PRACTICE: Cancer stem cells (CSCs) may occur at any stage of cancer and are implicated in the occurrence of resistance, recurrence, and metastasis. A systematic literature analysis has shown that the presence of CSCs, identified via the upregulation of stemness pathway biomarkers, results in reduced survival across all cancers studied. Several CSC-targeting agents are currently approved, and several others are in clinical trials. Future treatment regimens will likely include CSC-targeting agents to enable the elimination of these holdouts to current therapies.
Collapse
Affiliation(s)
- Justin Lathia
- Cancer Impact Area, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Huiping Liu
- Department of Pharmacology and Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Daniela Matei
- Department of Obstetrics and Gynecology and Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
153
|
Bartram I, Jeschke JM. Do cancer stem cells exist? A pilot study combining a systematic review with the hierarchy-of-hypotheses approach. PLoS One 2019; 14:e0225898. [PMID: 31834886 PMCID: PMC6910685 DOI: 10.1371/journal.pone.0225898] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 11/14/2019] [Indexed: 12/13/2022] Open
Abstract
The phenomenon of cancer cell heterogeneity has been explained by different hypotheses, each entailing different therapy strategies. The most recent is the cancer stem cell model, which says that tumourigenicity and self-renewal are restricted to rare stem cell-like cancer cells. Since its conception, conflicting evidence has been published. In this study, we tested the applicability of a new approach developed in the field of ecology, the hierarchy-of-hypotheses approach, for the Cancer Stem Cell hypothesis. This approach allows to structure a broad concept into more specific sub-hypotheses, which in turn can be connected to available empirical studies. To generate a dataset with empirical studies, we conducted a systematic literature review in the Web of Science limited to the first 1000 publications returned by the search. From this pool, 51 publications were identified that tested whether a cell sub-population had cancer stem cell properties. By classifying the studies according to: (1) assessed indicators, (2) experimental assays and (3) model cancer cells used, we built a hierarchical structure of sub-hypotheses. The empirical tests from the selected studies were subsequently assigned to this hierarchy of hypotheses, and the percentage of supporting, undecided and questioning evidence was calculated for each sub-hypothesis, as well as additional experimental characteristics. Our approach successfully allowed us to determine that within our dataset, the empirical support for the CSC hypothesis was only 49.0%. The support of different sub-hypotheses was highly variable. Most noticeable, the conception that putative cancer stem cells are a rare subset of cells could not be confirmed by most studies (13.5% support). The empirical support varied also between types of cancer, animal models and cell isolation method used. For the first time, this study showed the applicability of the hierarchy-of-hypotheses approach for synthesizing and evaluating empirical evidence for a broad hypothesis in the field of bio-medical research.
Collapse
Affiliation(s)
- Isabelle Bartram
- Department of Biology, Chemistry, Pharmacy, Institute of Biology, Freie Universität Berlin, Berlin, Germany
| | - Jonathan M. Jeschke
- Department of Biology, Chemistry, Pharmacy, Institute of Biology, Freie Universität Berlin, Berlin, Germany
- Leibniz-Institute of Freshwater Ecology and Inland Fisheries (IGB), Freie Universität Berlin, Berlin, Germany
- Berlin-Brandenburg Institute of Advanced Biodiversity Research (BBIB), Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
154
|
Razi E, Radak M, Mahjoubin-Tehran M, Talebi S, Shafiee A, Hajighadimi S, Moradizarmehri S, Sharifi H, Mousavi N, Sarvizadeh M, Nejati M, Taghizadeh M, Ghasemi F. Cancer stem cells as therapeutic targets of pancreatic cancer. Fundam Clin Pharmacol 2019; 34:202-212. [PMID: 31709581 DOI: 10.1111/fcp.12521] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 11/02/2019] [Accepted: 11/06/2019] [Indexed: 12/11/2022]
Abstract
The discovery of stem cells and their potential abilities in self-renewal and differentiation has opened a new horizon in medicine. Scientists have found a small population of stem cells in some types of cancers with the same functions as normal stem cells. There are two models for tumor progression: clonal (stochastic) and cancer stem cell (CSCs) models. According to the first model, all transformed cells in the tumor have carcinogenic potential and are able to proliferate and produce the same cells. The latter model, which has received more attention recently, considers the role of CSCs in drug resistance and tumor metastasis. Following the model, researchers have found that targeting CSCs may be a promising way in cancer therapy. This review describes CSC characteristics in general, while also focusing on CSC properties in the context of pancreatic cancer.
Collapse
Affiliation(s)
- Ebrahim Razi
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Mehran Radak
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Talebi
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alimohammad Shafiee
- Division of General Internal Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Sarah Hajighadimi
- Division of General Internal Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Sanaz Moradizarmehri
- Division of General Internal Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Hossein Sharifi
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Nousin Mousavi
- Department of Surgery, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mostafa Sarvizadeh
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Majid Nejati
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
155
|
Lasso P, Llano Murcia M, Sandoval TA, Urueña C, Barreto A, Fiorentino S. Breast Tumor Cells Highly Resistant to Drugs Are Controlled Only by the Immune Response Induced in an Immunocompetent Mouse Model. Integr Cancer Ther 2019; 18:1534735419848047. [PMID: 31056957 PMCID: PMC6505237 DOI: 10.1177/1534735419848047] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: The tumor cells responsible for metastasis are highly
resistant to chemotherapy and have characteristics of stem cells, with a high
capacity for self-regeneration and the use of detoxifying mechanisms that
participate in drug resistance. In vivo models of highly resistant cells allow
us to evaluate the real impact of the immune response in the control of cancer.
Materials and Methods: A tumor population derived from the 4T1
breast cancer cell line that was stable in vitro and highly aggressive in vivo
was obtained, characterized, and determined to exhibit cancer stem cell (CSC)
phenotypes (CD44+, CD24+, ALDH+,
Oct4+, Nanog+, Sox2+, and high self-renewal
capacity). Orthotopic transplantation of these cells allowed us to evaluate
their in vivo susceptibility to chemo and immune responses induced after
vaccination. Results: The immune response induced after vaccination
with tumor cells treated with doxorubicin decreased the formation of tumors and
macrometastasis in this model, which allowed us to confirm the immune response
relevance in the control of highly chemotherapy-resistant ALDH+ CSCs
in an aggressive tumor model in immunocompetent animals.
Conclusions: The antitumor immune response was the main element
capable of controlling tumor progression as well as metastasis in a highly
chemotherapy-resistant aggressive breast cancer model.
Collapse
Affiliation(s)
- Paola Lasso
- 1 Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | | | | | | | | |
Collapse
|
156
|
Jia P, Wu Y, Du H, Yang L, Zhang Z, Ma T, Li S, Yuan S, Lu L, Zha X. I-8, a novel inhibitor of mutant IDH1, inhibits cancer progression in vitro and in vivo. Eur J Pharm Sci 2019; 140:105072. [PMID: 31518680 DOI: 10.1016/j.ejps.2019.105072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/01/2019] [Accepted: 09/09/2019] [Indexed: 11/15/2022]
Abstract
Isocitrate dehydrogenase 1 mutations have been discovered in an array of hematologic malignancies and solid tumors. These mutations could cause the production of high levels of 2-hydroxyglutarate, which in turn implicated in epigenetic changes and impaired cell differentiation. Here, we described the characterization of compound I-8, a novel mutant IDH1 inhibitor, both in vitro and in vivo. Compound I-8 specifically inhibited 2-HG production, reduced histone methylation levels, induced differentiation and depleted stem characteristics in engineered and endogenous IDH1 mutant cells. In addition, oral administration of I-8 also significantly suppressed 2-HG production and histone methylation with dose of 150 mg/kg. And I-8 treatment also could induce differentiation and attenuate stem characteristics in tumor tissue. Together, these studies indicated that compound I-8 has clinical potential in tumor therapies as a effective mutant IDH1 inhibitor, and provided scientific guidance for the development of mutant IDH1 inhibitor in the future.
Collapse
Affiliation(s)
- Panli Jia
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Yao Wu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Hongzhi Du
- School of Pharmacy, Hubei University of Chinese Medicine, Hubei, China
| | - Lijun Yang
- Department of Pharmaceutical Engineering and Department of Biochemical Engineering, China Pharmaceutical University, Nanjing, China
| | - Zhibo Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Tianfang Ma
- Department of Pharmaceutical Engineering and Department of Biochemical Engineering, China Pharmaceutical University, Nanjing, China
| | - Sun Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Shengtao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.
| | - Ligong Lu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China.
| | - Xiaoming Zha
- Department of Pharmaceutical Engineering and Department of Biochemical Engineering, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
157
|
Zheng P, Eskandari A, Lu C, Laws K, Aldous L, Suntharalingam K. Biophysical analysis of cancer stem cell-potent copper(ii) coordination complexes. Dalton Trans 2019; 48:5892-5896. [PMID: 30632590 DOI: 10.1039/c8dt04706e] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Copper(ii) coordination complexes, 1 and 2, containing nonsteroidal anti-inflammatory drugs (NSAIDs) potently kill breast cancer stem cells (CSCs) and bulk breast cancer cells. Although detailed biological studies have been conducted to shed light on their mechanism of cytotoxicity, little is known about their molecular level mechanism of action. This biophysical study, aided by the preparation of a fluorophore-containing analogue, 3, reveals that the complexes operate by undergoing reduction to a copper(i) form and releasing the associated NSAIDs.
Collapse
Affiliation(s)
- Puyi Zheng
- Department of Chemistry, King's College London, London, UK.
| | | | | | | | | | | |
Collapse
|
158
|
Hwang JH, Yoon J, Cho YH, Cha PH, Park JC, Choi KY. A mutant KRAS-induced factor REG4 promotes cancer stem cell properties via Wnt/β-catenin signaling. Int J Cancer 2019; 146:2877-2890. [PMID: 31605540 DOI: 10.1002/ijc.32728] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/22/2019] [Accepted: 10/01/2019] [Indexed: 01/02/2023]
Abstract
Mutant KRAS provides a driving force for enhancement of cancer stem cells (CSCs) characteristics contributing transformation of colorectal cancer (CRC) cells harboring adenomatous polyposis coli (APC) mutations. Here, we identified the factors mediating the promotion of CSCs properties induced by KRAS mutation through microarray analyses of genes specifically induced in CRC spheroids harboring both KRAS and APC mutations. Among them, REG4 was identified as a key factor since CRISPR/Cas9-mediated knockout of REG4 most significantly affected the stem cell characteristics in which CSCs markers were effectively suppressed. We show that REG4 mediates promotion of CSCs properties via Wnt/β-catenin signaling in various in vitro studies including tumor organoid systems. Furthermore, expression patterns of CSCs markers and REG4 correlated in intestinal tumors from Apcmin/+ /KrasG12D LA2 mice and in CRC patient tissues harboring both KRAS and APC mutations. The role of REG4 in the tumor-initiating capacity accompanied by enhancement of CSCs characteristics was also revealed by NSG mice xenograft system. Collectively, our study highlights the importance of REG4 in promoting CSCs properties induced by KRAS mutation, and provides a new therapeutic strategy for CRC harboring both APC and KRAS mutations.
Collapse
Affiliation(s)
- Jeong-Ha Hwang
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, South Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.,Department of Biomaterials Science and Engineering, Yonsei University, Seoul, South Korea
| | - Junyong Yoon
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, South Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Yong-Hee Cho
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, South Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Pu-Hyeon Cha
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, South Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Jong-Chan Park
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, South Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Kang-Yell Choi
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, South Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.,CK Biotechnology Inc., Seoul, South Korea
| |
Collapse
|
159
|
Susman S, Pîrlog R, Leucuța D, Mitre AO, Padurean VA, Melincovici C, Moldovan I, Crișan D, Florian SI. The role of p-Stat3 Y705 immunohistochemistry in glioblastoma prognosis. Diagn Pathol 2019; 14:124. [PMID: 31690341 PMCID: PMC6829927 DOI: 10.1186/s13000-019-0903-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 10/14/2019] [Indexed: 12/27/2022] Open
Abstract
Background In spite of the multimodal treatment used today, glioblastoma is still the most aggressive and lethal cerebral tumour. To increase survival in these patients, novel therapeutic targets must be discovered. Signal transducer and activator of transcription 3 (Stat3), a transcription factor that controls normal cell differentiation and survival is also involved in neoplastic celltransformation. In this study we evaluated the immunohistochemical expression of pY705-Stat3 in patients with primary glioblastoma and determined its prognostic role by correlating it with survival. Methods This retrospective study included 94 patients diagnosed with glioblastoma. We determined the localization, number of positive cells, and marker intensity for pY705-Stat3 in these patients with the use of immunohistochemistry. The prognostic role was determined by correlating pY705-Stat3 expression on formalin-fixed paraffin-embedded tumour tissues with the patient’s survival in univariate and multivariate COX regressions. Results We found a statistically significant difference in survival between the patients with more than 20% pY705-Stat3 positive cells and those with less than 20% pY705-Stat3 positive cells (8.9 months median survival versus 13.7 months medial survival, p < 0.001). On multivariate analyses with the COX proportional hazards regression model including pY705-Stat3 expression, age and relapse status, pY705-Stat3 status was an independent prognostic factor in glioblastoma (P < 0.001). Conclusion The results obtained show that the immunohistochemical expression of pY705-Stat3 correlates with survival in glioblastoma. This study identifies Stat3 as a possible target for existing or new developed Stat3 inhibitors.
Collapse
Affiliation(s)
- Sergiu Susman
- Department of Morphological Sciences, Iuliu Hațieganu University of Medicine and Pharmacy, 6 Pasteur Street, 400349, Cluj-Napoca, Romania. .,Department of Pathology, Imogen Research Centre, Cluj-Napoca, Romania.
| | - Radu Pîrlog
- Department of Morphological Sciences, Iuliu Hațieganu University of Medicine and Pharmacy, 6 Pasteur Street, 400349, Cluj-Napoca, Romania
| | - Daniel Leucuța
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Andrei Otto Mitre
- Department of Neurosurgery, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Carmen Melincovici
- Department of Morphological Sciences, Iuliu Hațieganu University of Medicine and Pharmacy, 6 Pasteur Street, 400349, Cluj-Napoca, Romania
| | - Ioana Moldovan
- Department of Morphological Sciences, Iuliu Hațieganu University of Medicine and Pharmacy, 6 Pasteur Street, 400349, Cluj-Napoca, Romania
| | - Doinița Crișan
- Department of Morphological Sciences, Iuliu Hațieganu University of Medicine and Pharmacy, 6 Pasteur Street, 400349, Cluj-Napoca, Romania.,Department of Pathology, Emergency County Hospital, Cluj-Napoca, Romania
| | - Stefan Ioan Florian
- Department of Neurosurgery, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Neurosurgery, Emergency County Hospital, Cluj-Napoca, Romania
| |
Collapse
|
160
|
The Metabolic Inhibitor CPI-613 Negates Treatment Enrichment of Ovarian Cancer Stem Cells. Cancers (Basel) 2019; 11:cancers11111678. [PMID: 31671803 PMCID: PMC6896080 DOI: 10.3390/cancers11111678] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
One of the most significant therapeutic challenges in the treatment of ovarian cancer is the development of recurrent platinum-resistant disease. Cancer stem cells (CSCs) are postulated to contribute to recurrent and platinum-resistant ovarian cancer (OvCa). Drugs that selectively target CSCs may augment the standard of care cytotoxics and have the potential to prevent and/or delay recurrence. Increased reliance on metabolic pathway modulation in CSCs relative to non-CSCs offers a possible therapeutic opportunity. We demonstrate that treatment with the metabolic inhibitor CPI-613 (devimistat, an inhibitor of tricarboxylic acid (TCA) cycle) in vitro decreases CD133+ and CD117+ cell frequency relative to untreated OvCa cells, with negligible impact on non-CSC cell viability. Additionally, sphere-forming capacity and tumorigenicity in vivo are reduced in the CPI-613 treated cells. Collectively, these results suggest that treatment with CPI-613 negatively impacts the ovarian CSC population. Furthermore, CPI-613 impeded the unintended enrichment of CSC following olaparib or carboplatin/paclitaxel treatment. Collectively, our results suggest that CPI-613 preferentially targets ovarian CSCs and could be a candidate to augment current treatment strategies to extend either progression-free or overall survival of OvCa.
Collapse
|
161
|
Huang TH, Wu ATH, Cheng TS, Lin KT, Lai CJ, Hsieh HW, Chang PMH, Wu CW, Huang CYF, Chen KY. In silico identification of thiostrepton as an inhibitor of cancer stem cell growth and an enhancer for chemotherapy in non-small-cell lung cancer. J Cell Mol Med 2019; 23:8184-8195. [PMID: 31638335 PMCID: PMC6850923 DOI: 10.1111/jcmm.14689] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/15/2019] [Accepted: 07/23/2019] [Indexed: 12/15/2022] Open
Abstract
Cancer stem cells (CSCs) play an important role in cancer treatment resistance and disease progression. Identifying an effective anti-CSC agent may lead to improved disease control. We used CSC-associated gene signatures to identify drug candidates that may inhibit CSC growth by reversing the CSC gene signature. Thiostrepton, a natural cyclic oligopeptide antibiotic, was the top-ranked candidate. In non-small-cell lung cancer (NSCLC) cells, thiostrepton inhibited CSC growth in vitro and reduced protein expression of cancer stemness markers, including CD133, Nanog and Oct4A. In addition, metastasis-associated Src tyrosine kinase signalling, cell migration and epithelial-to-mesenchymal transition (EMT) were all inhibited by thiostrepton. Mechanistically, thiostrepton treatment led to elevated levels of tumour suppressor miR-98. Thiostrepton combined with gemcitabine synergistically suppressed NSCLC cell growth and induced apoptosis. The inhibition of NSCLC tumours and CSC growth by thiostrepton was also demonstrated in vivo. Our findings indicate that thiostrepton, an established drug identified in silico, is an inhibitor of CSC growth and a potential enhancer of chemotherapy in NSCLC.
Collapse
Affiliation(s)
- Tse-Hung Huang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan.,School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan.,School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.,Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Alexander T H Wu
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Tai-Shan Cheng
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Kuan-Ting Lin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Chia-Jou Lai
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Hao-Wen Hsieh
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Peter Mu-Hsin Chang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Wen Wu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kuan-Yu Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
162
|
Koroth J, Nirgude S, Tiwari S, Gopalakrishnan V, Mahadeva R, Kumar S, Karki SS, Choudhary B. Investigation of anti-cancer and migrastatic properties of novel curcumin derivatives on breast and ovarian cancer cell lines. Altern Ther Health Med 2019; 19:273. [PMID: 31638975 PMCID: PMC6802352 DOI: 10.1186/s12906-019-2685-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 09/13/2019] [Indexed: 01/08/2023]
Abstract
Background Curcumin is known for its multitude of medicinal properties, including anti-cancer and migrastatic activity. Efforts to overcome poor bioavailability, stability, and side effects associated with the higher dose of curcumin has led to the development of newer derivatives of curcumin. Thus, the focus of this study is to screen novel curcumin derivatives, namely ST03 and ST08, which have not been reported before, for their cytotoxicity and migrastatic property on cancer cells. Methods Anti-cancer activity of ST03 and ST08 was carried out using standard cytotoxicity assays viz., LDH, MTT, and Trypan blue on both solid and liquid cancer types. Flow cytometric assays and western blotting was used to investigate the cell death mechanisms. Transwell migration assay was carried out to check for migrastatic properties of the compounds. Results Both the compounds, ST03 and ST08, showed ~ 100 fold higher potency on liquid and solid tumour cell lines compared to its parent compound curcumin. They induced cytotoxicity by activating the intrinsic pathway of apoptosis in the breast (MDA-MB-231) and ovarian cancer cell lines (PA-1) bearing metastatic and stem cell properties, respectively. Moreover, ST08 also showed inhibition on breast cancer cell migration by inhibiting MMP1 (matrix metalloproteinase 1). Conclusion Both ST03 and ST08 exhibit anti-cancer activity at nanomolar concentration. They induce cell death by activating the intrinsic pathway of apoptosis. Also, they inhibit migration of the cancer cells by inhibiting MMP1 in breast cancer cells.
Collapse
|
163
|
Nasr MA, Salah RA, Abd Elkodous M, Elshenawy SE, El-Badri N. Dysregulated MicroRNA Fingerprints and Methylation Patterns in Hepatocellular Carcinoma, Cancer Stem Cells, and Mesenchymal Stem Cells. Front Cell Dev Biol 2019; 7:229. [PMID: 31681762 PMCID: PMC6811506 DOI: 10.3389/fcell.2019.00229] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the top causes of cancer mortality worldwide. Although HCC has been researched extensively, there is still a need for novel and effective therapeutic interventions. There is substantial evidence that initiation of carcinogenesis in liver cirrhosis, a leading cause of HCC, is mediated by cancer stem cells (CSCs). CSCs were also shown to be responsible for relapse and chemoresistance in several cancers, including HCC. MicroRNAs (miRNAs) constitute important epigenetic markers that regulate carcinogenesis by acting post-transcriptionally on mRNAs, contributing to the progression of HCC. We have previously shown that co-culture of cancer cells with mesenchymal stem cells (MSCs) could induce the reprogramming of MSCs into CSC-like cells. In this review, we evaluate the available data concerning the epigenetic regulation of miRNAs through methylation and the possible role of this regulation in stem cell and somatic reprogramming in HCC.
Collapse
Affiliation(s)
- Mohamed A Nasr
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 6th of October City, Egypt
| | - Radwa Ayman Salah
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 6th of October City, Egypt
| | - M Abd Elkodous
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 6th of October City, Egypt
| | - Shimaa E Elshenawy
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 6th of October City, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 6th of October City, Egypt
| |
Collapse
|
164
|
Sridharan S, Howard CM, Tilley AMC, Subramaniyan B, Tiwari AK, Ruch RJ, Raman D. Novel and Alternative Targets Against Breast Cancer Stemness to Combat Chemoresistance. Front Oncol 2019; 9:1003. [PMID: 31681564 PMCID: PMC6805781 DOI: 10.3389/fonc.2019.01003] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/18/2019] [Indexed: 12/15/2022] Open
Abstract
Breast cancer stem cells (BCSCs) play a vital role in tumor progression and metastasis. They are heterogeneous and inherently radio- and chemoresistant. They have the ability to self-renew and differentiate into non-BCSCs. These determinants of BCSCs including the plasticity between the mesenchymal and epithelial phenotypes often leads to minimal residual disease (MRD), tumor relapse, and therapy failure. By studying the resistance mechanisms in BCSCs, a combinatorial therapy can be formulated to co-target BCSCs and bulk tumor cells. This review addresses breast cancer stemness and molecular underpinnings of how the cancer stemness can lead to pharmacological resistance. This might occur through rewiring of signaling pathways and modulated expression of various targets that support survival and self-renewal, clonogenicity, and multi-lineage differentiation into heterogeneous bulk tumor cells following chemotherapy. We explore emerging novel and alternative molecular targets against BC stemness and chemoresistance involving survival, drug efflux, metabolism, proliferation, cell migration, invasion, and metastasis. Strategic targeting of such vulnerabilities in BCSCs may overcome the chemoresistance and increase the longevity of the metastatic breast cancer patients.
Collapse
Affiliation(s)
- Sangita Sridharan
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Cory M. Howard
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | | | | | - Amit K. Tiwari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
| | - Randall J. Ruch
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Dayanidhi Raman
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| |
Collapse
|
165
|
Kim JH, Choi HS, Kim SL, Lee DS. The PAK1-Stat3 Signaling Pathway Activates IL-6 Gene Transcription and Human Breast Cancer Stem Cell Formation. Cancers (Basel) 2019; 11:cancers11101527. [PMID: 31658701 PMCID: PMC6826853 DOI: 10.3390/cancers11101527] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 11/16/2022] Open
Abstract
Cancer stem cells (CSCs) have unique properties, including self-renewal, differentiation, and chemoresistance. In this study, we found that p21-activated kinase (PAK1) inhibitor (Group I, PAK inhibitor, IPA-3) and inactivator (ivermectin) treatments inhibit cell proliferation and that tumor growth of PAK1-knockout cells in a mouse model is significantly reduced. IPA-3 and ivermectin inhibit CSC formation. PAK1 physically interacts with Janus Kinase 2 (JAK2), and JAK2 inhibitor (TG101209) treatment inhibits mammosphere formation and reduces the nuclear PAK1 protein level. PAK1 interacts with signal transducer and activator of transcription 3 (Stat3), and PAK1 and Stat3 colocalize in the nucleus. We show through electrophoretic mobility shift assay (EMSA), chromatin immunoprecipitation (ChIP), and reporter assays that the PAK1/Stat3 complex binds to the IL-6 promoter and regulates the transcription of the IL-6 gene. Inhibition of PAK1 and JAK2 in mammospheres reduces the nuclear pStat3 and extracellular IL-6 levels. PAK1 inactivation inhibits CSC formation by decreasing pStat3 and extracellular IL-6 levels. Our results reveal that JAK2/PAK1 dysregulation inhibits the Stat3 signaling pathway and CSC formation, the PAK1/Stat3 complex regulates IL-6 gene expression, PAK1/Stat3 signaling regulates CSC formation, and PAK1 may be an important target for treating breast cancer.
Collapse
Affiliation(s)
- Ji-Hyang Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Korea.
| | - Hack Sun Choi
- Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, SARI, Jeju 63243, Korea.
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Korea.
| | - Su-Lim Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Korea.
- School of Biomaterials Science and Technology, College of Applied Life Science, Jeju National University, Jeju 63243, Korea.
| | - Dong-Sun Lee
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Korea.
- Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, SARI, Jeju 63243, Korea.
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Korea.
- School of Biomaterials Science and Technology, College of Applied Life Science, Jeju National University, Jeju 63243, Korea.
- Practical Translational Research Center, Jeju National University, Jeju 63243, Korea.
| |
Collapse
|
166
|
Abstract
An incomplete view of the mechanisms that drive metastasis, the primary cause of cancer-related death, has been a major barrier to development of effective therapeutics and prognostic diagnostics. Increasing evidence indicates that the interplay between microenvironment, genetic lesions, and cellular plasticity drives the metastatic cascade and resistance to therapies. Here, using melanoma as a model, we outline the diversity and trajectories of cell states during metastatic dissemination and therapy exposure, and highlight how understanding the magnitude and dynamics of nongenetic reprogramming in space and time at single-cell resolution can be exploited to develop therapeutic strategies that capitalize on nongenetic tumor evolution.
Collapse
Affiliation(s)
- Florian Rambow
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Herestraat 49, 3000 Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KULeuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Herestraat 49, 3000 Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KULeuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Colin R Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
167
|
Siddiqui Z, Srivastava AN, Sankhwar SN, Zaidi N, Fatima N, Singh S, Yusuf M. Oct-4: a prognostic biomarker of urinary bladder cancer in North India. Ther Adv Urol 2019; 11:1756287219875576. [PMID: 31632462 PMCID: PMC6769204 DOI: 10.1177/1756287219875576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/19/2019] [Indexed: 12/29/2022] Open
Abstract
Background The objective of this study was to evaluate Octamer-binding transcription factor 4 (Oct-4), neutrophil to lymphocyte ratio (NLR) and body mass index (BMI) as independent prognostic biomarkers for prediction of urinary bladder cancer (UBC) outcomes. With the advancement in prognostic biomarker discovery, tumor recurrence is difficult to accurately predict in UBC. UBC is costly to treat due to the requirement of frequent invasive follow-up sessions. Therefore, it is of utmost importance to evaluate good prognostic biomarkers for UBC surveillance. Methods We studied 39 UBC tissue samples. Oct-4 protein expression was evaluated semiquantitatively by immunohistochemistry (IHC). Complete blood count data and body weight as well as the height of the patients were retrieved and recorded before the date of the first transurethral resection of bladder tumor (TURBT). The follow-up period was 48 months for recurrence-free survival (RFS), progression-free survival (PFS), and overall survival (OS). Results Oct-4 expression profile was found to be significantly associated with gender (p = 0.028), tumor grade (p = 0.038), tumor stage (p = 0.003), lymph node status (p = 0.029), recurrence (p = 0.004), progression (p = 0.011), and treatment modality (p = 0.016). Tumor grade and progression were found significant with NLR values (tumor grade, p = 0.006; progression, p = 0.038) and BMI (tumor grade, p = 0.036; progression, p = 0.014). Moreover, BMI was also significantly associated with UBC recurrence (p = 0.014). Kaplan-Meier survival analysis showed poor prognosis with both high Oct-4 expression (RFS, p = 0.001; PFS, p = 0.004; OS, p = 0.014) and high NLR values (RFS, p = 0.049; PFS, p = 0.004; OS, p = 0.005). Patients with high BMI too had poor RFS (p = 0.025) and poor PFS (p = 0.032). Furthermore, multivariate Cox regression analysis, indicated Oct-4 as an independent prognostic biomarker for RFS (HR = 0.240, 95% CI, 0.072-0.804, p = 0.021). Conclusions We conclude that the expression profile of Oct-4 will be beneficial in prediction of UBC recurrence, and could have profound implications on the development of new therapeutic targets for UBC treatment.
Collapse
Affiliation(s)
- Zainab Siddiqui
- Department of Pathology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, India; Department of Biotechnology, Dr. A. P. J. Abdul Kalam Technical University, Lucknow, India
| | - Anand N Srivastava
- Department of Pathology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, Uttar Pradesh, India
| | - Satya N Sankhwar
- Department of Urology, King George's Medical University, Lucknow, India
| | - Noorin Zaidi
- Department of Pathology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, India
| | - Naseem Fatima
- Department of Pathology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, India
| | - Shivangi Singh
- Research Metabolic Unit, Era's Lucknow Medical College and Hospital, Era University, Lucknow, India
| | - Mohd Yusuf
- Department of Pathology, King George's Medical University, Lucknow, India
| |
Collapse
|
168
|
Eugenio-Pérez D, Briones-Herrera A, Martínez-Klimova E, Pedraza-Chaverri J. Divide et Impera: Drp1-mediated Mitochondrial Fission in Glioma Malignancy. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2019; 92:423-433. [PMID: 31543706 PMCID: PMC6747948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Mitochondria are pivotal organelles involved in vital cellular functions, including energy generation, reactive oxygen species and calcium signaling, as well as intermediate biosynthesis. They are dynamic organelles that adapt their shape, size, and distribution to changes in intracellular conditions, being able to divide, fuse, or move along the cell, processes known as mitochondrial dynamics. Mitochondrial dynamics are involved in cell division and migration, as well as maintenance of pluripotency in stem (non-differentiated) cells. Thus, its central role in carcinogenesis is not surprising. Particularly, mitochondrial dynamics have been found to be pivotal to the development of gliomas, a lethal group of tumors developed from glial cells, which are nervous system cells that provide support to neurons. Unfortunately, prognosis of glioma patients is poor, most of them do not survive more than five years after diagnosis. In this context, it is fundamental to understand the cellular mechanisms involved in this pathology, in order to develop an appropriate clinical approach. As previously mentioned, mitochondrial dynamics is central to glioma development, particularly, mitochondrial division (fission) and one of its central effectors, dynamin-related protein 1 (Drp1), have been observed to be enhanced in gliomas and involved in the maintenance of stem cells (which initiate and maintain the tumor), as well as in migration and invasiveness, being central to gliomagenesis. In this review, we discuss the findings on mitochondrial fission role in these processes, further, we analyze the potential use of Drp1 as a novel prognostic biomarker in glioma patients.
Collapse
Affiliation(s)
| | | | | | - José Pedraza-Chaverri
- To whom all correspondence should be addressed: José Pedraza-Chaverri, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, 04510 Mexico;
| |
Collapse
|
169
|
Stafman LL, Williams AP, Marayati R, Aye JM, Markert HR, Garner EF, Quinn CH, Lallani SB, Stewart JE, Yoon KJ, Whelan K, Beierle EA. Focal Adhesion Kinase Inhibition Contributes to Tumor Cell Survival and Motility in Neuroblastoma Patient-Derived Xenografts. Sci Rep 2019; 9:13259. [PMID: 31519958 PMCID: PMC6744403 DOI: 10.1038/s41598-019-49853-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/24/2019] [Indexed: 12/29/2022] Open
Abstract
Patient-derived xenografts (PDXs) provide an opportunity to evaluate the effects of therapies in an environment that more closely resembles the human condition than that seen with long-term passage cell lines. In the current studies, we investigated the effects of FAK inhibition on two neuroblastoma PDXs in vitro. Cells were treated with two small molecule inhibitors of FAK, PF-573,228 (PF) and 1,2,4,5-benzentetraamine tetrahydrochloride (Y15). Following FAK inhibition, cell survival and proliferation decreased significantly and cell cycle arrest was seen in both cell lines. Migration and invasion assays were used to determine the effect of FAK inhibition on cell motility, which decreased significantly in both cell lines in the presence of either inhibitor. Finally, tumor cell stemness following FAK inhibition was evaluated with extreme limiting dilution assays as well as with immunoblotting and quantitative real-time PCR for the expression of stem cell markers. FAK inhibition decreased formation of tumorspheres and resulted in a corresponding decrease in established stem cell markers. FAK inhibition decreased many characteristics of the malignant phenotype, including cancer stem cell like features in neuroblastoma PDXs, making FAK a candidate for further investigation as a potential target for neuroblastoma therapy.
Collapse
Affiliation(s)
- Laura L Stafman
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, AL, 35205, USA
| | - Adele P Williams
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, AL, 35205, USA
| | - Raoud Marayati
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, AL, 35205, USA
| | - Jamie M Aye
- Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Alabama, Birmingham, AL, 35233, USA
| | - Hooper R Markert
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, AL, 35205, USA
| | - Evan F Garner
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, AL, 35205, USA
| | - Colin H Quinn
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, AL, 35205, USA
| | - Shoeb B Lallani
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Jerry E Stewart
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, AL, 35205, USA
| | - Karina J Yoon
- Department of Pharmacology and Toxicology, University of Alabama, Birmingham, AL, 35233, USA
| | - Kimberly Whelan
- Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Alabama, Birmingham, AL, 35233, USA
| | - Elizabeth A Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, AL, 35205, USA.
| |
Collapse
|
170
|
Howard N, Clementino M, Kim D, Wang L, Verma A, Shi X, Zhang Z, DiPaola RS. New developments in mechanisms of prostate cancer progression. Semin Cancer Biol 2019; 57:111-116. [DOI: 10.1016/j.semcancer.2018.09.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/28/2018] [Accepted: 09/06/2018] [Indexed: 01/07/2023]
|
171
|
Cordella M, Tabolacci C, Senatore C, Rossi S, Mueller S, Lintas C, Eramo A, D'Arcangelo D, Valitutti S, Facchiano A, Facchiano F. Theophylline induces differentiation and modulates cytoskeleton dynamics and cytokines secretion in human melanoma-initiating cells. Life Sci 2019; 230:121-131. [PMID: 31125565 DOI: 10.1016/j.lfs.2019.05.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/22/2022]
Abstract
AIMS Cutaneous melanoma is the most aggressive skin cancer, derived from neoplastic transformation of melanocytes. Since several evidences highlighted the importance of a hierarchical model of differentiation among cancer cells, closely related to resistance mechanisms and tumor relapse, we investigated the effects of theophylline (Theo), a methylxanthine commonly used in treatment of respiratory diseases, on melanoma cells with different degree of differentiation, including patient-derived melanoma-initiating cells. MATERIALS AND METHODS The antiproliferative and antimetastatic effects of Theo was demonstrated by cell counting, adhesion and migration assays on A375 and SK-MEL-30 cells. Further, Theo ability to reduce cell growth was highly significant in A375-derived spheroids and in two patient-derived melanoma-initiating cells (MICs). In order to identify pathways potentially involved in the antineoplastic properties of Theo, a comparative mass spectrometry proteomic analysis was used. Then, melanin content, tyrosinase and tissue transglutaminase activities as differentiation markers and actin re-organization through confocal microscopy were evaluated. Furthermore, a secretome profile of MICs after Theo treatments was performed by multiplex immunoassay. KEY FINDINGS Obtained results demonstrate inhibitory effects of Theo on melanoma cell proliferation and migration, mainly in MICs, together with the induction of differentiation parameters. Moreover, our data indicate that the known anti-melanoma effect of Theo is due also to its ability to interfere with cytoskeleton dynamics and to induce the secretion of inflammatory molecules involved in recruitment of immunosuppressive cells in tumor microenvironment. SIGNIFICANCE Data strongly suggest that Theo supplement, either as drug or as dietary supply, may represent a potent additional weapon against melanoma.
Collapse
Affiliation(s)
- Martina Cordella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Cinzia Senatore
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Carla Lintas
- Center for Neurodevelopmental Disorders, Laboratory of Molecular Psychiatry and Neurogenetics, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Adriana Eramo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Salvatore Valitutti
- Cancer Research Center of Toulouse, Toulouse, France; Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, Toulouse, France
| | | | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
172
|
Rodriguez-Ramirez C, Nör JE. p53 and Cell Fate: Sensitizing Head and Neck Cancer Stem Cells to Chemotherapy. Crit Rev Oncog 2019; 23:173-187. [PMID: 30311573 DOI: 10.1615/critrevoncog.2018027353] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Head and neck cancers are deadly diseases that are diagnosed annually in approximately half a million individuals worldwide. Growing evidence supporting a role for cancer stem cells (CSCs) in the pathobiology of head and neck cancers has led to increasing interest in identifying therapeutics to target these cells. Apart from the canonical tumor-suppressor functions of p53, emerging research supports a significant role for this protein in physiological stem cell and CSC maintenance and reprogramming. Therefore, p53 has become a promising target to sensitize head and neck CSCs to chemotherapy. In this review, we highlight the role of p53 in stem cell maintenance and discuss potential implications of targeting p53 to treat patients with head and neck cancers.
Collapse
Affiliation(s)
- Christie Rodriguez-Ramirez
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109
| | - Jacques E Nör
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109; Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI; Department of Otolaryngology, University of Michigan School of Medicine, Ann Arbor, MI; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
173
|
Detection and clinical significance of circulating tumor cells in patients with nasopharyngeal carcinoma. Oncol Lett 2019; 18:2537-2547. [PMID: 31452741 DOI: 10.3892/ol.2019.10560] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 09/28/2018] [Indexed: 12/11/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is the most common cancer type originating in the nasopharynx, and varies notably from other cancer types of the head and neck in its occurrence, causes, clinical behavior and treatment. Significant effort has been made into understanding the biological properties of circulating tumor cells (CTCs), with previous studies demonstrating the critical role CTCs serve in the metastatic spread of carcinoma. However, associations between NPC and CTCs have not been completely elucidated. Therefore, in the present study, the CanPatrol™ CTC-enrichment technique and classical in situ hybridization assay were utilized to acquire, identify and classify CTCs from patients with NPC. Subsequently, the correlation between CTCs and the clinical indexes, progression-free survival (PFS), N-cadherin gene expression and the response to therapy were investigated. The present study then determined whether the Wnt/β-catenin signaling pathway served a role in therapy for NPC cells. Collectively, the research demonstrated that CTCs could be detected in patients with NPC. Additionally, CTCs exhibited a statistically significant association with the Epstein-Barr virus infection prior to therapy and Eastern Cooperative Oncology Group score following therapy. Furthermore, co-treatment with cisplatin and paclitaxel significantly decreased the number of CTCs. In addition, mesenchymal CTCs may serve as a predictor of PFS. Finally, the present study demonstrated that cisplatin combined with paclitaxel induced apoptosis and decreased the tumor markers in NPC cells through the Wnt/β-catenin signaling pathway. In conclusion, these data indicated that CTCs may serve as a biomarker in monitoring the therapeutic efficacy of treatments for NPC. Furthermore, the Wnt/β-catenin signaling pathway served a therapeutic role in the treatment of NPC.
Collapse
|
174
|
Shokraii F, Moharrami M, Motamed N, Shahhoseini M, Totonchi M, Ezzatizadeh V, Firouzi J, Khosravani P, Ebrahimi M. Histone Modification Marks Strongly Regulate CDH1 Promoter in Prostospheres as A Model of Prostate Cancer Stem Like Cells. CELL JOURNAL 2019; 21:124-134. [PMID: 30825285 PMCID: PMC6397603 DOI: 10.22074/cellj.2019.5702] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 10/02/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Cadherin-1 (CDH1) plays an important role in the metastasis, while expression of this protein is under control of epigenetic changes on its gene promoter. Therefore we evaluated both DNA methylation (DNAmet) and histone modification marks of CDH1 in prostate cancer stem like cells (PCSLCs). MATERIALS AND METHODS In this experimental study, we isolated PCSLCs using cell surface marker and prostaspheroid formation, respectively. The cells isolated from both methods were characterized and then the levels of H3K4me2, H3K27me3, H3K9me2/3 and H3K9ac as well as DNAmet were assessed in CDH1 promoter of the isolated cells. RESULTS The CD44+ CD49hi cells were not validated as PCSLCs. However, prostaspheres overexpressed stemness related genes and had higher ability of invasion potential, associated with reduction in CDH1 expression. Epigenetic status analysis showed that CDH1 promoter was hypo-methylated. Histone modifications of H3K9ac and H3K4me3 were significantly reduced, in parallel with an increased level of H3K27me3. CONCLUSION Our results suggest that slight decrease of DNAmet of the CpG island in CDH1 promoter does not significantly contribute to the change of CDH1 expression. Therefore, histone modifications are responsible in repressing CDH1 in PCSLCs.
Collapse
Affiliation(s)
- Fatemeh Shokraii
- Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Moharrami
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Nasrin Motamed
- School of Biology, College of Science, University of Tehran, Tehran, Iran. Electronic Adress:
| | - Maryam Shahhoseini
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mehdi Totonchi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Vahid Ezzatizadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Medical Genetics, Royesh Medical Laboratory Centre, Tehran, Iran
| | - Javad Firouzi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Pardis Khosravani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. Electronic Address:
| |
Collapse
|
175
|
Goh JXH, Tan LTH, Goh JK, Chan KG, Pusparajah P, Lee LH, Goh BH. Nobiletin and Derivatives: Functional Compounds from Citrus Fruit Peel for Colon Cancer Chemoprevention. Cancers (Basel) 2019; 11:E867. [PMID: 31234411 PMCID: PMC6627117 DOI: 10.3390/cancers11060867] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 12/19/2022] Open
Abstract
The search for effective methods of cancer treatment and prevention has been a continuous effort since the disease was discovered. Recently, there has been increasing interest in exploring plants and fruits for molecules that may have potential as either adjuvants or as chemopreventive agents against cancer. One of the promising compounds under extensive research is nobiletin (NOB), a polymethoxyflavone (PMF) extracted exclusively from citrus peel. Not only does nobiletin itself exhibit anti-cancer properties, but its derivatives are also promising chemopreventive agents; examples of derivatives with anti-cancer activity include 3'-demethylnobiletin (3'-DMN), 4'-demethylnobiletin (4'-DMN), 3',4'-didemethylnobiletin (3',4'-DMN) and 5-demethylnobiletin (5-DMN). In vitro studies have demonstrated differential efficacies and mechanisms of NOB and its derivatives in inhibiting and killing of colon cancer cells. The chemopreventive potential of NOB has also been well demonstrated in several in vivo colon carcinogenesis animal models. NOB and its derivatives target multiple pathways in cancer progression and inhibit several of the hallmark features of colorectal cancer (CRC) pathophysiology, including arresting the cell cycle, inhibiting cell proliferation, inducing apoptosis, preventing tumour formation, reducing inflammatory effects and limiting angiogenesis. However, these substances have low oral bioavailability that limits their clinical utility, hence there have been numerous efforts exploring better drug delivery strategies for NOB and these are part of this review. We also reviewed data related to patents involving NOB to illustrate the extensiveness of each research area and its direction of commercialisation. Furthermore, this review also provides suggested directions for future research to advance NOB as the next promising candidate in CRC chemoprevention.
Collapse
Affiliation(s)
- Joanna Xuan Hui Goh
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, 47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Loh Teng-Hern Tan
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Joo Kheng Goh
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Kok Gan Chan
- Division of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia.
- International Genome Centre, Jiangsu University, Zhenjiang 212013, China.
| | - Priyia Pusparajah
- Medical Health and Translational Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia.
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Asian Centre for Evidence Synthesis in Population, Implementation and Clinical Outcomes (PICO), Health and Well-being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Bandar Sunway 47500, Malaysia.
| | - Bey-Hing Goh
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, 47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Asian Centre for Evidence Synthesis in Population, Implementation and Clinical Outcomes (PICO), Health and Well-being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Bandar Sunway 47500, Malaysia.
| |
Collapse
|
176
|
O'Reilly D, Buchanan P. Calcium channels and cancer stem cells. Cell Calcium 2019; 81:21-28. [PMID: 31163289 DOI: 10.1016/j.ceca.2019.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 12/24/2022]
Abstract
Cancer stem cells (CSC's) have emerged as a key area of investigation due to associations with cancer development and treatment resistance, related to their ability to remain quiescent, self-renew and terminally differentiate. Targeting CSC's in addition to the tumour bulk could ensure complete removal of the cancer, lessening the risk of relapse and improving patient survival. Understanding the mechanisms supporting the functions of CSC's is essential to highlight targets for the development of therapeutic strategies. Changes in intracellular calcium through calcium channel activity is fundamental for integral cellular processes such as proliferation, migration, differentiation and survival in a range of cell types, under both normal and pathological conditions. Here in we highlight how calcium channels represent a key mechanism involved in CSC function. It is clear that expression and or function of a number of channels involved in calcium entry and intracellular store release are altered in CSC's. Correlating with aberrant proliferation, self-renewal and differentiation, which in turn promoted cancer progression and treatment resistance. Research outlined has demonstrated that targeting altered calcium channels in CSC populations can reduce their stem properties and induce terminal differentiation, sensitising them to existing cancer treatments. Overall this highlights calcium channels as emerging novel targets for CSC therapies.
Collapse
Affiliation(s)
- Debbie O'Reilly
- National Institute of Cellular Biotechnology, Dublin City University, Dublin, Ireland; School of Nursing and Human science, Dublin City University, Dublin, Ireland
| | - Paul Buchanan
- National Institute of Cellular Biotechnology, Dublin City University, Dublin, Ireland; School of Nursing and Human science, Dublin City University, Dublin, Ireland.
| |
Collapse
|
177
|
Gupta R, Bhatt LK, Johnston TP, Prabhavalkar KS. Colon cancer stem cells: Potential target for the treatment of colorectal cancer. Cancer Biol Ther 2019; 20:1068-1082. [PMID: 31050577 DOI: 10.1080/15384047.2019.1599660] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite incessant research, colon cancer still is one of the most common causes of fatalities in both men and women worldwide. Also, nearly 50% of patients with colorectal cancer show tumor recurrence. Recent investigations have highlighted the involvement of colon cancer stem cells (CCSCs) in cancer relapse and chemoresistance. CCSCs deliver a significant protumorigenic niche through persistent overexpression of self-renewal capabilities. Moreover, CSCs cross network with stromal cells, immune infiltrates, and cyotokine-chemokine, which potentiate their aggressive proliferative potential. Targeting CCSCs through small molecule inhibitors, miRNAs, and monoclonal antibodies (mAbs) in in vivo studies has generated compelling evidence for the effectiveness of these various treatments. This review effectively compiles the role of CCSC surface markers and dysregulated and/or upregulated pathways in the pathogenesis of colorectal cancer that can be used to target CCSCs for effective colorectal cancer treatment.
Collapse
Affiliation(s)
- Riya Gupta
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| | - Lokesh Kumar Bhatt
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| | - Thomas P Johnston
- b Division of Pharmacology and Pharmaceutical Sciences , University of Missouri-Kansas City , Kansas City , MO , USA
| | - Kedar S Prabhavalkar
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| |
Collapse
|
178
|
Yoon HJ, Kim DH, Kim SJ, Jang JH, Surh YJ. Src-mediated phosphorylation, ubiquitination and degradation of Caveolin-1 promotes breast cancer cell stemness. Cancer Lett 2019; 449:8-19. [DOI: 10.1016/j.canlet.2019.01.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/02/2019] [Accepted: 01/14/2019] [Indexed: 12/19/2022]
|
179
|
Javed Z, Farooq HM, Ullah M, Iqbal MZ, Raza Q, Sadia H, Pezzani R, Salehi B, Sharifi-Rad J, Cho WC. Wnt Signaling: A Potential Therapeutic Target in Head and
Neck Squamous Cell Carcinoma. Asian Pac J Cancer Prev 2019; 20:995-1003. [PMID: 31030466 PMCID: PMC6948882 DOI: 10.31557/apjcp.2019.20.4.995] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 04/15/2019] [Indexed: 12/24/2022] Open
Abstract
Cellular maintenance and development are two fundamental mechanisms regulated by the canonical Wnt signaling pathway. Wnt/beta-catenin signaling pathway controls a myriad of cellular processes that are essential for normal cell functioning. Cell cycle progression, differentiation, fate determination, and migration are generally orchestrated by canonical Wnt signaling. Altered Wnt/beta-catenin signaling has been considered a promoting event for different types of cancers and the oncogenic potential of Wnt signaling have been discussed in many cancer types, including breast, colon, pancreatic as well as head and neck. Furthermore, Wnt signaling is critical for the maintenance and stemness of both the normal as well as cancer stem cells. This review sheds new light on Wnt signaling and explains how it can regulate normal physiological processes and curtail the development of cancer. It depicts the vital functions of Wnt signaling in the stem cell growth and differentiation by focusing on current druggable targets that have been ascribed by recent studies. Thus, Wnt signaling pathway retains a tremendous potential in eradicating head and neck squamous cell carcinoma.
Collapse
Affiliation(s)
| | | | | | - Muhammad Zaheer Iqbal
- Institute of Biochemistry and Biotechnology, University of Veterinary and Animal Sciences, Pakistan,
| | - Qamar Raza
- Center for Excellence in Molecular Biology, University of The Punjab, Lahore,
| | | | - Raffaele Pezzani
- OU Endocrinology, Department of Medicine (DIMED), University of Padova, via Ospedale 105,
- AIROB, Associazione Italiana per la Ricerca Oncologica di Base, Padova, Italy,
| | - Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam,
| | - Javad Sharifi-Rad
- Food Safety Research Center (salt), Semnan University of Medical Sciences, Semnan, Iran,
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China.
| |
Collapse
|
180
|
Moon HJ, Park SY, Lee SH, Kang CD, Kim SH. Nonsteroidal Anti-inflammatory Drugs Sensitize CD44-Overexpressing Cancer Cells to Hsp90 Inhibitor Through Autophagy Activation. Oncol Res 2019; 27:835-847. [PMID: 30982499 PMCID: PMC7848457 DOI: 10.3727/096504019x15517850319579] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recently, novel therapeutic strategies have been designed with the aim of killing cancer stem-like cells (CSCs), and considerable interest has been generated in the development of specific therapies that target stemness-related marker of CSCs. In this study, nonsteroidal anti-inflammatory drugs (NSAIDs) significantly potentiated Hsp90 inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG)-mediated cytotoxicity through apoptotic and autophagic cell death induction, but COX-2-inhibitory function was not required for NSAID-induced autophagy in CD44-overexpressing human chronic myeloid leukemia K562 (CD44highK562) cells. Importantly, we found that treatment with NSAIDs resulted in a dose-dependent increase in LC3-II level and decrease in p62 level and simultaneous reduction in multiple stemness-related markers including CD44, Oct4, c-Myc, and mutant p53 (mutp53) in CD44highK562 cells, suggesting that NSAIDs could induce autophagy, which might mediate degradation of stemness-related marker proteins. Activation of AMPK and inhibition of Akt/mTOR/p70S6K/4EBP1 participated in NSAID-induced autophagy in CD44highK562 cells. In addition, treatment of CD44highK562 cells with NSAIDs inhibited expression of HSF1/Hsps, which resulted in suppression of 17-AAG-induced activation of Hsp70, leading to reversal of 17-AAG resistance and sensitization of CD44highK562 cells to 17-AAG by NSAIDs. In conclusion, combining NSAIDs with Hsp90 inhibitor may offer one of the most promising strategies for eradication of CD44-overexpressing CSCs.
Collapse
Affiliation(s)
- Hyun-Jung Moon
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, South Korea
| | - So-Young Park
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, South Korea
| | - Su-Hoon Lee
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, South Korea
| | - Chi-Dug Kang
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, South Korea
| | - Sun-Hee Kim
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, South Korea
| |
Collapse
|
181
|
Rezalotfi A, Ahmadian E, Aazami H, Solgi G, Ebrahimi M. Gastric Cancer Stem Cells Effect on Th17/Treg Balance; A Bench to Beside Perspective. Front Oncol 2019; 9:226. [PMID: 31024835 PMCID: PMC6464032 DOI: 10.3389/fonc.2019.00226] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 03/13/2019] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer stem cells (GCSCs), a small population among tumor cells, are responsible for tumor initiation, development, metastasis, and recurrence. They play a crucial role in immune evasion, immunomodulation, and impairment of effector immunity and believed to be emerged to change the balance of the immune system, importantly CD4+ T cells in the chronic inflamed tumor site. However, different subtypes of innate and adaptive immune cells are involved in the formation of the immune system in the tumor microenvironment, we would look at T cells in this study. Tumor microenvironment induces differentiation of CD4+ T cells into different subsets of T cells, mainly suppressive regulatory T cells (Treg), and T helper 17 (Th17) cells, although their exact role in tumor immunity is still under debate depending on tumor types and stages. Counterbalance between Th17 and Treg cells in the gastrointestinal system result in the homeostasis and normal function of the immune system, particularly mucosal immunity. Recent data demonstrated a high infiltration of Th17 and Treg cells into the gastric tumor site and proved that tumor microenvironment might disturb the balance between Th17 and Treg. It is possible to assume an association between activation of CSCs which contribute to metastasis in late stages, and the imbalanced Th17/Treg cells observed in advanced gastric cancer patients. This review intends to clarify the importance of gastric tumor microenvironment specifically CSCs in relation to Th17/Tregs balance firstly and to highlight the relevance of imbalanced Th17/Treg subsets in determining the stages and behavior of the tumor secondly. Finally, the present study suggests a clinical approach looking at the plasticity of T cells with a focus on Th17 as a promising dedicated arm in cancer immunotherapy.
Collapse
Affiliation(s)
- Alaleh Rezalotfi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Elmira Ahmadian
- Faculty of Biological Sciences and Technology, Department of Animal Sciences, Shahid Beheshti University, Tehran, Iran
| | - Hossein Aazami
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Students Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Solgi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
182
|
Petpiroon N, Bhummaphan N, Soonnarong R, Chantarawong W, Maluangnont T, Pongrakhananon V, Chanvorachote P. Ti 0.8O 2 Nanosheets Inhibit Lung Cancer Stem Cells by Inducing Production of Superoxide Anion. Mol Pharmacol 2019; 95:418-432. [PMID: 30737252 DOI: 10.1124/mol.118.114447] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/02/2019] [Indexed: 02/14/2025] Open
Abstract
Recent research into the cancer stem cell (CSC) concept has driven progress in the understanding of cancer biology and has revealed promising CSC-specific targets for drug discovery efforts. As malignancies of lung cancer have been shown to be strongly associated with activities of CSCs, we examined the effects of Ti0.8O2 nanosheets on these cells. Here we show that the nanosheets target lung CSCs but not normal primary dermal papilla (DP) stem cells. Whereas Ti0.8O2 caused a dramatic apoptosis along with a decrease in CSC phenotypes, in primary human DP cells such effects of nanosheets have been minimal. Nanosheets reduced the ability of lung cancer cells to generate three-dimensional tumor spheroids, lung CSC markers (CD133 and ALDH1A1), and CSC transcription factors (Nanog and Oct-4). Ti0.8O2 nanosheets reduced CSC signaling through mechanisms involving suppression of protein kinase B (AKT) and Notch-1 pathways. In addition, the nanosheets inhibited the migration and invasive activities of lung cancer cells and reduced epithelial-to-mesenchymal transition (EMT) markers as N-cadherin, vimentin, and Slug, as well as metastasis-related integrins (integrin-αv and integrin-β1). Importantly, we found that the selectivity of the Ti0.8O2 nanosheets in targeting cancer cells was mediated by induction of cellular superoxide anion in cancerous but not normal cells. Inhibition of nanosheet-induced superoxide anion restored the suppression of CSC and EMT in cancer cells. These findings demonstrate a promising distinctive effect of Ti0.8O2 nanosheets on lung CSC that may lead to opportunities to use such a nanomaterial in cancer therapy.
Collapse
Affiliation(s)
- Nalinrat Petpiroon
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences (N.P., W.C., V.P., P.C.), Interdisciplinary Program of Biomedical Sciences, Faculty of Graduate School (N.B.), Interdisciplinary Program of Pharmacology Graduate School (R.S.), and Cell-based Drug and Health Products Development Research Unit (N.P., N.B., R.S., W.C., V.P., P.C.), Chulalongkorn, University, Bangkok, Thailand; and College of Nanotechnology, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand (T.M.)
| | - Narumol Bhummaphan
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences (N.P., W.C., V.P., P.C.), Interdisciplinary Program of Biomedical Sciences, Faculty of Graduate School (N.B.), Interdisciplinary Program of Pharmacology Graduate School (R.S.), and Cell-based Drug and Health Products Development Research Unit (N.P., N.B., R.S., W.C., V.P., P.C.), Chulalongkorn, University, Bangkok, Thailand; and College of Nanotechnology, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand (T.M.)
| | - Rapeepun Soonnarong
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences (N.P., W.C., V.P., P.C.), Interdisciplinary Program of Biomedical Sciences, Faculty of Graduate School (N.B.), Interdisciplinary Program of Pharmacology Graduate School (R.S.), and Cell-based Drug and Health Products Development Research Unit (N.P., N.B., R.S., W.C., V.P., P.C.), Chulalongkorn, University, Bangkok, Thailand; and College of Nanotechnology, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand (T.M.)
| | - Wipa Chantarawong
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences (N.P., W.C., V.P., P.C.), Interdisciplinary Program of Biomedical Sciences, Faculty of Graduate School (N.B.), Interdisciplinary Program of Pharmacology Graduate School (R.S.), and Cell-based Drug and Health Products Development Research Unit (N.P., N.B., R.S., W.C., V.P., P.C.), Chulalongkorn, University, Bangkok, Thailand; and College of Nanotechnology, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand (T.M.)
| | - Tosapol Maluangnont
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences (N.P., W.C., V.P., P.C.), Interdisciplinary Program of Biomedical Sciences, Faculty of Graduate School (N.B.), Interdisciplinary Program of Pharmacology Graduate School (R.S.), and Cell-based Drug and Health Products Development Research Unit (N.P., N.B., R.S., W.C., V.P., P.C.), Chulalongkorn, University, Bangkok, Thailand; and College of Nanotechnology, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand (T.M.)
| | - Varisa Pongrakhananon
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences (N.P., W.C., V.P., P.C.), Interdisciplinary Program of Biomedical Sciences, Faculty of Graduate School (N.B.), Interdisciplinary Program of Pharmacology Graduate School (R.S.), and Cell-based Drug and Health Products Development Research Unit (N.P., N.B., R.S., W.C., V.P., P.C.), Chulalongkorn, University, Bangkok, Thailand; and College of Nanotechnology, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand (T.M.)
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences (N.P., W.C., V.P., P.C.), Interdisciplinary Program of Biomedical Sciences, Faculty of Graduate School (N.B.), Interdisciplinary Program of Pharmacology Graduate School (R.S.), and Cell-based Drug and Health Products Development Research Unit (N.P., N.B., R.S., W.C., V.P., P.C.), Chulalongkorn, University, Bangkok, Thailand; and College of Nanotechnology, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand (T.M.)
| |
Collapse
|
183
|
Zahedpanah M, Takanlu JS, Nikbakht M, Rad F, Farhid F, Mousavi SA, Rad S, Fumani HK, Hosseini Rad SMA, Mohammadi S. Microvesicles of osteoblasts modulate bone marrow mesenchymal stem cell-induced apoptosis to curcumin in myeloid leukemia cells. J Cell Physiol 2019; 234:18707-18719. [PMID: 30916405 DOI: 10.1002/jcp.28511] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/16/2019] [Accepted: 02/20/2019] [Indexed: 11/07/2022]
Abstract
Microvesicles (MVs) derived from bone marrow niche components have an important role in genetic reprogramming and subsequent drugs induce apoptosis in leukemic cells. Here, we have found that undertreatment of curcumin or daunorubicin, the cross-talk through MVs of KG-1-bone marrow mesenchymal stem cells (BMSCs), significantly downregulates the expression of the survival gene osteopontin (OPN), CXCL-12, IL-6 (interleukin-6), STAT-3, and VCAM-1 (vascular cell adhesion molecule 1) in treated-KG-1 cells as well as exclusively upregulates CXCL-12 in BMSCs. Drug treated-cell populations' MVs of both single cultured osteoblasts (OBs) and cocultured KG-1 + BMSCs + OBs similarly upregulate survival mediators' OPN, CXCL-12, IL-6, STAT-3, and VCAM-1 in treated-KG-1 cells. Likewise, isolated MVs from KG-1 cells or communication between KG-1, BMSCs, and OBs treated by drugs increase the expression of genes OPN, CXCL-12, IL-6, STAT3, and VCAM-1 by OBs. MVs derived from KG-1 + BMSCs + OBs reduce drug-induced apoptosis in KG-1 cells. This suggests MVs-mediated information transfer is a procedure whereby OBs could overcome BMSCs-induced apoptosis in drug-treated-KG-1 cells.
Collapse
Affiliation(s)
- Mahdi Zahedpanah
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Javid Sabour Takanlu
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | - Mohsen Nikbakht
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | - Fariba Rad
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Fatemeh Farhid
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Asadollah Mousavi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | - Soroush Rad
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | - Hosein Kamranzadeh Fumani
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | | | - Saeed Mohammadi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| |
Collapse
|
184
|
Dihydrotanshinone-Induced NOX5 Activation Inhibits Breast Cancer Stem Cell through the ROS/Stat3 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9296439. [PMID: 31019654 PMCID: PMC6451810 DOI: 10.1155/2019/9296439] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022]
Abstract
Cancer stem cells (CSCs) are known to mediate metastasis and recurrence and are therefore a promising therapeutic target. In this study, we found that dihydrotanshinone (DHTS) inhibits CSC formation. DHTS inhibited mammosphere formation in a dose-dependent manner and showed significant tumor growth inhibition in a xenograft model. This compound reduced the CD44high/CD24low- and aldehyde dehydrogenase- (ALDH-) expressing cell population and the self-renewal-related genes Nanog, SOX2, OCT4, C-Myc, and CD44. DHTS induced NOX5 activation by increasing calcium, and NOX5 activation induced reactive oxygen species (ROS) production. ROS production reduced the nuclear phosphorylation levels of Stat3 and secreted IL-6 levels in the mammospheres. DHTS deregulated the dynamic equilibrium from non-stem cancer cells to CSCs by dephosphorylating Stat3 and decreasing IL-6 secretion and inhibiting CSC formation. These novel findings showed that DHTS-induced ROS deregulated the Stat3/IL-6 pathway and induced CSC death. NOX5 activation by DHTS inhibits CSC formation through ROS/Stat3/IL-6 signaling, and DHTS may be a promising potential therapeutic agent against breast CSCs.
Collapse
|
185
|
Kutwin M, Sawosz E, Jaworski S, Wierzbicki M, Strojny B, Grodzik M, Ewa Sosnowska M, Trzaskowski M, Chwalibog A. Nanocomplexes of Graphene Oxide and Platinum Nanoparticles against Colorectal Cancer Colo205, HT-29, HTC-116, SW480, Liver Cancer HepG2, Human Breast Cancer MCF-7, and Adenocarcinoma LNCaP and Human Cervical Hela B Cell Lines. MATERIALS 2019; 12:ma12060909. [PMID: 30893818 PMCID: PMC6470683 DOI: 10.3390/ma12060909] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/11/2019] [Accepted: 03/15/2019] [Indexed: 12/12/2022]
Abstract
Inefficient drug administration into cancer cells is related to the chemoresistance of cancer cells caused by genetic mutations including genes involved in drug transport, enzyme metabolism, and/or DNA damage repair. The objective of the present study was to evaluate the properties of platinum (NP-Pt), graphene oxide (GO), and the nanocomplex of GO functionalized with platinum nanoparticles (GO-NP-Pt) against several genetically, phenotypically, and metabolically different cancer cell lines: Colo205, HT-29, HTC-116, SW480, HepG2, MCF-7, LNCaP, and Hela B. The anticancer effects toward the cancer cell lines were evaluated by 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxyanilide salt (XTT) and bromodeoxyuridine (BrdU) assays and measurements of cell apoptosis and morphology deformations. The NP-Pt and GO could effectively be introduced to cancer cells, but more effective delivery was observed after GO-NP-Pt treatment. The delivery of the GO-NP-Pt nanocomplex significantly decreased the viability of Colo 205 and HepG2 cells, but did not increase the cytotoxicity of other investigated cancer cells. The nanocomplex GO-NP-Pt also significantly increased the apoptosis of Colo 205 and HepG2 cancer cells. The obtained results suggest that the nanocomplex GO-NP-Pt is a remarkable nanostructure that can improve the delivery of Pt nanoparticles into cancer cells and has potential anticancer applications.
Collapse
Affiliation(s)
- Marta Kutwin
- Department of Animal Nutrition and Biotechnology, Faculty of Animal Science, Warsaw University of Life Sciences, 02-786 Warsaw, Poland.
| | - Ewa Sawosz
- Department of Animal Nutrition and Biotechnology, Faculty of Animal Science, Warsaw University of Life Sciences, 02-786 Warsaw, Poland.
| | - Sławomir Jaworski
- Department of Animal Nutrition and Biotechnology, Faculty of Animal Science, Warsaw University of Life Sciences, 02-786 Warsaw, Poland.
| | - Mateusz Wierzbicki
- Department of Animal Nutrition and Biotechnology, Faculty of Animal Science, Warsaw University of Life Sciences, 02-786 Warsaw, Poland.
| | - Barbara Strojny
- Department of Animal Nutrition and Biotechnology, Faculty of Animal Science, Warsaw University of Life Sciences, 02-786 Warsaw, Poland.
| | - Marta Grodzik
- Department of Animal Nutrition and Biotechnology, Faculty of Animal Science, Warsaw University of Life Sciences, 02-786 Warsaw, Poland.
| | - Malwina Ewa Sosnowska
- Department of Animal Nutrition and Biotechnology, Faculty of Animal Science, Warsaw University of Life Sciences, 02-786 Warsaw, Poland.
| | - Maciej Trzaskowski
- Centre for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland.
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, 00-645 Warsaw, Poland.
| | - André Chwalibog
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark.
| |
Collapse
|
186
|
Weng YS, Tseng HY, Chen YA, Shen PC, Al Haq AT, Chen LM, Tung YC, Hsu HL. MCT-1/miR-34a/IL-6/IL-6R signaling axis promotes EMT progression, cancer stemness and M2 macrophage polarization in triple-negative breast cancer. Mol Cancer 2019; 18:42. [PMID: 30885232 PMCID: PMC6421700 DOI: 10.1186/s12943-019-0988-0] [Citation(s) in RCA: 278] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/26/2019] [Indexed: 02/07/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC) is a poor prognostic breast cancer with the highest mutations and limited therapeutic choices. Cytokine networking between cancer cells and the tumor microenvironment (TME) maintains the self-renewing subpopulation of breast cancer stem cells (BCSCs) that mediate tumor heterogeneity, resistance and recurrence. Immunotherapy of those factors combined with targeted therapy or chemoagents may advantage TNBC treatment. Results We found that the oncogene Multiple Copies in T-cell Malignancy 1 (MCT-1/MCTS1) expression is a new poor-prognosis marker in patients with aggressive breast cancers. Overexpressing MCT-1 perturbed the oncogenic breast epithelial acini morphogenesis and stimulated epithelial-mesenchymal transition and matrix metalloproteinase activation in invasive TNBC cells, which were repressed after MCT-1 gene silencing. As mammary tumor progression was promoted by oncogenic MCT-1 activation, tumor-promoting M2 macrophages were enriched in TME, whereas M2 macrophages were decreased and tumor-suppressive M1 macrophages were increased as the tumor was repressed via MCT-1 knockdown. MCT-1 stimulated interleukin-6 (IL-6) secretion that promoted monocytic THP-1 polarization into M2-like macrophages to increase TNBC cell invasiveness. In addition, MCT-1 elevated the soluble IL-6 receptor levels, and thus, IL-6R antibodies antagonized the effect of MCT-1 on promoting M2-like polarization and cancer cell invasion. Notably, MCT-1 increased the features of BCSCs, which were further advanced by IL-6 but prevented by tocilizumab, a humanized IL-6R antibody, thus MCT-1 knockdown and tocilizumab synergistically inhibited TNBC stemness. Tumor suppressor miR-34a was induced upon MCT-1 knockdown that inhibited IL-6R expression and activated M1 polarization. Conclusions The MCT-1 pathway is a novel and promising therapeutic target for TNBC. Electronic supplementary material The online version of this article (10.1186/s12943-019-0988-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yueh-Shan Weng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Hong-Yu Tseng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Yen-An Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Pei-Chun Shen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Aushia Tanzih Al Haq
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Li-Mei Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Yi-Chung Tung
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsin-Ling Hsu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan.
| |
Collapse
|
187
|
Liskova A, Kubatka P, Samec M, Zubor P, Mlyncek M, Bielik T, Samuel SM, Zulli A, Kwon TK, Büsselberg D. Dietary Phytochemicals Targeting Cancer Stem Cells. Molecules 2019; 24:molecules24050899. [PMID: 30836718 PMCID: PMC6429493 DOI: 10.3390/molecules24050899] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022] Open
Abstract
There is an increasing awareness of the importance of a diet rich in fruits and vegetables for human health. Cancer stem cells (CSCs) are characterized as a subpopulation of cancer cells with aberrant regulation of self-renewal, proliferation or apoptosis leading to cancer progression, invasiveness, metastasis formation, and therapy resistance. Anticancer effects of phytochemicals are also directed to target CSCs. Here we provide a comprehensive review of dietary phytochemicals targeting CSCs. Moreover, we evaluate and summarize studies dealing with effects of dietary phytochemicals on CSCs of various malignancies in preclinical and clinical research. Dietary phytochemicals have a significant impact on CSCs which may be applied in cancer prevention and treatment. However, anticancer effects of plant derived compounds have not yet been fully investigated in clinical research.
Collapse
Affiliation(s)
- Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Pavol Zubor
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Milos Mlyncek
- Department of Obstetrics and Gynecology Faculty Hospital Nitra Constantine the Philosopher University, 949 01 Nitra, Slovakia.
| | - Tibor Bielik
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha 24144, Qatar.
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia.
| | - Taeg Kyu Kwon
- Department of Immunology and School of Medicine, Keimyung University, Dalseo-Gu, Daegu 426 01, Korea.
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha 24144, Qatar.
| |
Collapse
|
188
|
Phenethyl Isothiocyanate Exposure Promotes Oxidative Stress and Suppresses Sp1 Transcription Factor in Cancer Stem Cells. Int J Mol Sci 2019; 20:ijms20051027. [PMID: 30818757 PMCID: PMC6429440 DOI: 10.3390/ijms20051027] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/05/2019] [Accepted: 02/22/2019] [Indexed: 12/11/2022] Open
Abstract
Aldehyde dehydrogenase 1 (ALDH1) is a cytosolic marker of cancer stem cells (CSCs), which are a sub-population within heterogeneous tumor cells. CSCs associate with therapy-resistance, self-renewal, malignancy, tumor-relapse, and reduced patient-survival window. ALDH1-mediated aldehyde scavenging helps CSCs to survive a higher level of oxidative stress than regular cancer cells. Cruciferous vegetable-derived phenethyl isothiocyanate (PEITC) selectively induces reactive oxygen species (ROS), leading to apoptosis of cancer cells, but not healthy cells. However, this pro-oxidant role of PEITC in CSCs is poorly understood and is investigated here. In a HeLa CSCs model (hCSCs), the sphere-culture and tumorsphere assay showed significantly enriched ALDHhi CSCs from HeLa parental cells (p < 0.05). Aldefluor assay and cell proliferation assay revealed that PEITC treatments resulted in a reduced number of ALDHhi hCSCs in a concentration-dependent manner (p < 0.05). In the ROS assay, PEITC promoted oxidative stress in hCSCs (p ≤ 0.001). Using immunoblotting and flow cytometry techniques, we reported that PEITC suppressed the cancer-associated transcription factor (Sp1) and a downstream multidrug resistance protein (P-glycoprotein) (both, p < 0.05). Furthermore, PEITC-treatment of hCSCs, prior to xenotransplantation in mice, lowered the in vivo tumor-initiating potential of hCSCs. In summary, PEITC treatment suppressed the proliferation of ALDH1 expressing cancer stem cells as well as key factors that are involved with drug-resistance, while promoting oxidative stress and apoptosis in hCSCs.
Collapse
|
189
|
Zarrintaj P, Mostafapoor F, Milan PB, Saeb MR. Theranostic Platforms Proposed for Cancerous Stem Cells: A Review. Curr Stem Cell Res Ther 2019; 14:137-145. [DOI: 10.2174/1574888x13666181002152247] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 08/29/2018] [Accepted: 09/04/2018] [Indexed: 01/06/2023]
Abstract
It is next-to-impossible not to accept that cancer takes a position as the main cause of the global burden of disease, for it is hard to ignore the outnumbered people dying from cancer. Looking at the statistics proves that progress in cancer therapy is always beyond cancer in a race of pessimism about the future; for various kinds of cancers yearly cause death in the world, whereas the conventional and even modern therapies often exhibit lack of reliability in the treatment of cancer. In principle, various reasons are identified for cancer resistance and recurrence. Recognizing the cells/tissue from which cancer takes origin enables its early detection, and optimistically saying, protection of patients against death. It has been recognized that cancer stem cells are responsible for cancer cell proliferation and metastasis. Conventional therapies cannot eradicate the cancer stem cell; therefore, cancer recurrence is unavoidable. In this regards, designing smart platforms with specific properties is an essential step in cancer treatment. Theranostic platforms have facilitated the cancer diagnosis and treatment, simultaneously. In this respect, several types of smart materials have been designed to detect and cure cancer. Cancer stem cell as a root of the cancerous tumor should be eradicated to achieve the complete treatment; hence, cancer stem cell mechanism must be known precisely to design an appropriate platform making possible to encounter with cancer stem cell. In this review paper, various therapeutic and diagnostic techniques of cancerous stem cell are discussed to pave a way for designing proper platforms for cancer eradication.
Collapse
Affiliation(s)
- Payam Zarrintaj
- Color and Polymer Research Center (CPRC), Amirkabir University of Technology, P.O. Box 15875-4413, Tehran, Iran
| | - Farnaz Mostafapoor
- Department of Polymer Processing, Iran Polymer and Petrochemical Institute, P.O. Box: 14965-115, Tehran, Iran
| | - Peiman Brouki Milan
- Cellular and Molecular Research Center (CMRC), Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Saeb
- Color and Polymer Research Center (CPRC), Amirkabir University of Technology, P.O. Box 15875-4413, Tehran, Iran
| |
Collapse
|
190
|
Ghasemi F, Sarabi PZ, Athari SS, Esmaeilzadeh A. Therapeutics strategies against cancer stem cell in breast cancer. Int J Biochem Cell Biol 2019; 109:76-81. [PMID: 30772480 DOI: 10.1016/j.biocel.2019.01.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/21/2019] [Accepted: 01/28/2019] [Indexed: 01/06/2023]
Abstract
Breast cancer is known as a most prevalent cancer and second deadly cancer, among women worldwide. Due to the high incidence rate of breast cancer and limitations of conventional therapy it seemed essential to look for new targets in cancer cells and directly target them such as target therapy on breast cancer stem cells. In this review we indicate some of therapeutic uses of cancer stem cells in breast cancer. Some strategies are targeting surface specific markers and activated signaling pathways in their microenvironment such as Notch, Hedgehog, Wnt/b-catenin, PI3K/Akt, NF-kB, BMP and TGF-β and their maintenance and drug resistance, using various miRNAs, enhancement of CSCs apoptosis, differentiation therapy, blocking epithelial to mesenchymal transition and using different natural compounds. Recent studies have shown that cancer stem cells play major roles in target therapy on breast cancer. The new manipulation approaches of cancer stem cells can be used as target therapy of breast cancer that were highlighted for immunotherapy of cancer.
Collapse
Affiliation(s)
- Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Parisa Zia Sarabi
- Molecular and Medicine Research Center, Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
191
|
Scalable Culture Strategies for the Expansion of Patient-Derived Cancer Stem Cell Lines. Stem Cells Int 2019; 2019:8347595. [PMID: 30918523 PMCID: PMC6409046 DOI: 10.1155/2019/8347595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 11/15/2018] [Indexed: 12/26/2022] Open
Abstract
Cancer stem cells (CSCs) have recently raised great interest as a promising biological system for designing effective cancer therapies. The scarcity of CSCs in vivo and the consequent low numbers obtained from biopsies represent a major hurdle to the development of such strategies. It is therefore necessary to design robust scalable methods to enable efficient expansion of bona fide CSCs in vitro. Here, we evaluated the applicability of computer-controlled bioreactors combined with 3D aggregate culture and microcarrier technology, widely used in stem cell bioprocessing, for the expansion and enrichment of CSCs isolated from different types of solid tumors—colorectal cancer (CRC) and non-small-cell lung cancer (NSCLC) from two patients. Results show that these culture strategies improved cell expansion and CSC enrichment. Both patient-derived CSC lines were able to grow on microcarriers, the best results being achieved for PPlus 102-L, Pro-F 102-L, Fact 102-L, and CGEN 102-L beads (5-fold and 40-fold increase in total cell concentration for CRC and NSCLC cells, respectively, in 6 days). As for 3D aggregate culture strategy, the cell proliferation profile was donor dependent. NSCLC cells were the only cells able to form aggregates and proliferate, and the flat-bottom bioreactor vessel equipped with a trapezoid-shaped paddle impeller was the most efficient configuration for cell growth (21-fold increase in cell concentration achieved in 8 days). Serum-free medium promotes CSC enrichment in both 3D aggregate and microcarrier cultures. The protocols developed herein for CSC expansion have the potential to be transferred to clinical and industrial settings, providing key insights to guide bioprocess design towards the production of enriched CSC cultures in higher quantity and improved quality.
Collapse
|
192
|
Dolatabadi S, Jonasson E, Lindén M, Fereydouni B, Bäcksten K, Nilsson M, Martner A, Forootan A, Fagman H, Landberg G, Åman P, Ståhlberg A. JAK-STAT signalling controls cancer stem cell properties including chemotherapy resistance in myxoid liposarcoma. Int J Cancer 2019; 145:435-449. [PMID: 30650179 PMCID: PMC6590236 DOI: 10.1002/ijc.32123] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 12/03/2018] [Accepted: 01/07/2019] [Indexed: 12/14/2022]
Abstract
Myxoid liposarcoma (MLS) shows extensive intratumoural heterogeneity with distinct subpopulations of tumour cells. Despite improved survival of MLS patients, existing therapies have shortcomings as they fail to target all tumour cells. The nature of chemotherapy‐resistant cells in MLS remains unknown. Here, we show that MLS cell lines contained subpopulations of cells that can form spheres, efflux Hoechst dye and resist doxorubicin, all properties attributed to cancer stem cells (CSCs). By single‐cell gene expression, western blot, phospho‐kinase array, immunoprecipitation, immunohistochemistry, flow cytometry and microarray analysis we showed that a subset of MLS cells expressed JAK–STAT genes with active signalling. JAK1/2 inhibition via ruxolitinib decreased, while stimulation with LIF increased, phosphorylation of STAT3 and the number of cells with CSC properties indicating that JAK–STAT signalling controlled the number of cells with CSC features. We also show that phosphorylated STAT3 interacted with the SWI/SNF complex. We conclude that MLS contains JAK–STAT‐regulated subpopulations of cells with CSC features. Combined doxorubicin and ruxolitinib treatment targeted both proliferating cells as well as cells with CSC features, providing new means to circumvent chemotherapy resistance in treatment of MLS patients. What's new? Despite improved survival of patients, existing therapies for Myxoid liposarcoma (MLS) present shortcomings as they fail to target all tumour cells. The nature of chemotherapy‐resistant cells in MLS remains unknown, however. Here, the authors show that myxoid liposarcomas are heterogeneous and contain subpopulations of cells with stem cell properties, including chemotherapy resistance. Moreover, JAK‐STAT signalling is active in MLS and regulates the size of the cancer stem cells‐like subpopulation via the SWI/SNF complex. The results shed light on the mechanisms of therapy resistance in MLS and point to JAK‐STAT inhibitors as a new avenue for targeted MLS therapies.
Collapse
Affiliation(s)
- Soheila Dolatabadi
- Sahlgrenska Cancer Center, Department of Pathology and GeneticsInstitute of Biomedicine, Sahlgrenska Academy at University of GothenburgGothenburgSweden
| | - Emma Jonasson
- Sahlgrenska Cancer Center, Department of Pathology and GeneticsInstitute of Biomedicine, Sahlgrenska Academy at University of GothenburgGothenburgSweden
| | - Malin Lindén
- Sahlgrenska Cancer Center, Department of Pathology and GeneticsInstitute of Biomedicine, Sahlgrenska Academy at University of GothenburgGothenburgSweden
| | - Bentolhoda Fereydouni
- Sahlgrenska Cancer Center, Department of Pathology and GeneticsInstitute of Biomedicine, Sahlgrenska Academy at University of GothenburgGothenburgSweden
| | - Karin Bäcksten
- Sahlgrenska Cancer Center, Department of Pathology and GeneticsInstitute of Biomedicine, Sahlgrenska Academy at University of GothenburgGothenburgSweden
| | - Malin Nilsson
- TIMM Laboratory, Sahlgrenska Cancer CenterUniversity of GothenburgGothenburgSweden
| | - Anna Martner
- TIMM Laboratory, Sahlgrenska Cancer CenterUniversity of GothenburgGothenburgSweden
| | - Amin Forootan
- Sahlgrenska Cancer Center, Department of Pathology and GeneticsInstitute of Biomedicine, Sahlgrenska Academy at University of GothenburgGothenburgSweden
- MultiD Analysis ABGothenburgSweden
| | - Henrik Fagman
- Sahlgrenska Cancer Center, Department of Pathology and GeneticsInstitute of Biomedicine, Sahlgrenska Academy at University of GothenburgGothenburgSweden
- Department of Clinical Pathology and GeneticsSahlgrenska University HospitalGothenburgSweden
| | - Göran Landberg
- Sahlgrenska Cancer Center, Department of Pathology and GeneticsInstitute of Biomedicine, Sahlgrenska Academy at University of GothenburgGothenburgSweden
| | - Pierre Åman
- Sahlgrenska Cancer Center, Department of Pathology and GeneticsInstitute of Biomedicine, Sahlgrenska Academy at University of GothenburgGothenburgSweden
| | - Anders Ståhlberg
- Sahlgrenska Cancer Center, Department of Pathology and GeneticsInstitute of Biomedicine, Sahlgrenska Academy at University of GothenburgGothenburgSweden
- Department of Clinical Pathology and GeneticsSahlgrenska University HospitalGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
| |
Collapse
|
193
|
Alblihy A, Mesquita KA, Sadiq MT, Madhusudan S. Development and implementation of precision therapies targeting base-excision DNA repair in BRCA1-associated tumors. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1567266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Adel Alblihy
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals, Nottingham, UK
| | - Katia A. Mesquita
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals, Nottingham, UK
| | - Maaz T. Sadiq
- Department of Oncology, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| | - Srinivasan Madhusudan
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals, Nottingham, UK
- Department of Oncology, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| |
Collapse
|
194
|
Selected Office Based Anticancer Treatment Strategies. JOURNAL OF ONCOLOGY 2019; 2019:7462513. [PMID: 30766601 PMCID: PMC6350558 DOI: 10.1155/2019/7462513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/06/2019] [Indexed: 12/16/2022]
Abstract
Over the years, the treatment of patients with cancer has varied widely as much because of recent advancements in science and medicine as the philosophies that belie their use. This paper briefly describes many of the prevailing approaches in use today with an attempt to offer some perspective of how to apply these disparate methodologies so that they may be more effectively integrated, resulting in consistently better clinical responses.
Collapse
|
195
|
B591, a novel specific pan-PI3K inhibitor, preferentially targets cancer stem cells. Oncogene 2019; 38:3371-3386. [PMID: 30635656 PMCID: PMC6756013 DOI: 10.1038/s41388-018-0674-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 12/06/2018] [Accepted: 12/18/2018] [Indexed: 12/17/2022]
Abstract
Cancer stem cells (CSCs) have been implicated in metastasis, relapse, and therapeutic resistance of cancer, so successful cancer therapy may therefore require the development of drugs against CSCs or combining anti-CSCs drugs with conventional therapies. The phosphoinositide 3-kinase (PI3K) signaling pathway is one of the most frequently activated signaling pathways in human cancer, playing a central role in tumorigenesis as well as the maintenance of CSCs. Here, we designed and identified B591, a dihydrobenzofuran-imidazolium salt, as a novel specific pan-PI3K inhibitor with potent inhibitory activity against class I PI3K isoforms, which showed effective inhibition of cellular PI3K/mTOR signaling pathway and robust antitumor activity in a set of cancer cell lines. Notably, compared with bulk tumor cell populations, B591 exhibited more potency in suppressing CSCs survival and inducing CSCs apoptosis, and presence of B591 effectively eliminated paclitaxel-enriched CSCs. B591 diminished self-renewal capacity and decreased the expression of epithelial-mesenchymal transition (EMT) markers of CSCs. In vivo, B591 preferentially decreased CSCs levels in mouse xenograft model of human breast cancer as evidenced especially by remarkable reduction of tumor-initiating ability. Consistent with the preferential targeting of CSCs, B591 effectively inhibited breast tumor metastasis and delayed tumor regrowth following paclitaxel treatment. Taken together, our findings establish B591, a novel PI3K inhibitor, as a strong candidate for clinical evaluation as a CSCs targeting agent.
Collapse
|
196
|
Lu L, Shen X, Tao B, Lin C, Li K, Luo Z, Cai K. The nanoparticle-facilitated autophagy inhibition of cancer stem cells for improved chemotherapeutic effects on glioblastomas. J Mater Chem B 2019; 7:2054-2062. [DOI: 10.1039/c8tb03165g] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Paclitaxel (PTX) and chloroquine (CQ) loaded bovine serum albumin (BSA) nanoparticles can achieve efficient glioma therapyviaautophagy inhibition.
Collapse
Affiliation(s)
- Lu Lu
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Xinkun Shen
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Bailong Tao
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Chuanchuan Lin
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Ke Li
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Zhong Luo
- School of Life Science
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Kaiyong Cai
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| |
Collapse
|
197
|
Parejo S, Tschan MP, Muraro MG, Garattini E, Spagnoli GC, Schläfli AM. Assessing Autophagy During Retinoid Treatment of Breast Cancer Cells. Methods Mol Biol 2019; 2019:237-256. [PMID: 31359401 DOI: 10.1007/978-1-4939-9585-1_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Retinoids are derived from vitamin A through a multi-step process. Within a target cell, retinoids regulate gene expression by activating the retinoid acid receptors (RAR) and retinoid x receptors (RXR), which are ligand-dependent transcription factors. Besides its therapeutic use in dermatological disorders, all-trans retinoic acid (ATRA) is successfully utilized to treat acute promyelocytic leukemia (APL) patients. The use of ATRA in APL patients is the first example of clinically useful differentiation therapy. Therapeutic strategies aiming at cancer cell differentiation have great potential for solid tumors, including breast cancer. The few clinical studies conducted with ATRA in breast cancer are rather disappointing. However, these studies did not take into account the heterogeneity of the disease and were conducted on unselected cohorts of patients.We recently showed that ATRA treatment of breast cancer cells induces autophagy, a highly conserved process aiming at degrading and recycling superfluous or harmful cellular components. In addition, autophagy inhibition significantly increases the therapeutic activity of ATRA. This finding is of fundamental importance, since autophagy has a dual role in cancer. Whereas autophagy may be a protective mechanism during the initial phases of cancer development, it may support cancer cell survival in already established tumors. Furthermore, autophagy can lower or enhance therapeutic efficiency, depending on the tumor type and the anticancer agent considered. Therefore, it is important to investigate the role of autophagy in the context of specific tumors and therapeutic approaches. Accurate autophagy studies are challenging given the dynamic nature of the process and the difficulty of measuring the rate of autophagosome degradation (autophagic flux). In this chapter, we provide protocols for a careful assessment of the autophagic flux in ATRA treated 2D and 3D breast cancer cultures.
Collapse
Affiliation(s)
- Sarah Parejo
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Manuele G Muraro
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Enrico Garattini
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giulio C Spagnoli
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
- National Research Council, Institute of Translational Pharmacology, Rome, Italy
| | - Anna M Schläfli
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
198
|
Sridharan S, Howard CM, Tilley AMC, Subramaniyan B, Tiwari AK, Ruch RJ, Raman D. Novel and Alternative Targets Against Breast Cancer Stemness to Combat Chemoresistance. Front Oncol 2019. [PMID: 31681564 DOI: 10.3389/fonc.2019.01003.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
Abstract
Breast cancer stem cells (BCSCs) play a vital role in tumor progression and metastasis. They are heterogeneous and inherently radio- and chemoresistant. They have the ability to self-renew and differentiate into non-BCSCs. These determinants of BCSCs including the plasticity between the mesenchymal and epithelial phenotypes often leads to minimal residual disease (MRD), tumor relapse, and therapy failure. By studying the resistance mechanisms in BCSCs, a combinatorial therapy can be formulated to co-target BCSCs and bulk tumor cells. This review addresses breast cancer stemness and molecular underpinnings of how the cancer stemness can lead to pharmacological resistance. This might occur through rewiring of signaling pathways and modulated expression of various targets that support survival and self-renewal, clonogenicity, and multi-lineage differentiation into heterogeneous bulk tumor cells following chemotherapy. We explore emerging novel and alternative molecular targets against BC stemness and chemoresistance involving survival, drug efflux, metabolism, proliferation, cell migration, invasion, and metastasis. Strategic targeting of such vulnerabilities in BCSCs may overcome the chemoresistance and increase the longevity of the metastatic breast cancer patients.
Collapse
Affiliation(s)
- Sangita Sridharan
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Cory M Howard
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Augustus M C Tilley
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | | | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
| | - Randall J Ruch
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Dayanidhi Raman
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| |
Collapse
|
199
|
Velasco-Velázquez MA, Velázquez-Quesada I, Vásquez-Bochm LX, Pérez-Tapia SM. Targeting Breast Cancer Stem Cells: A Methodological Perspective. Curr Stem Cell Res Ther 2019; 14:389-397. [PMID: 30147014 DOI: 10.2174/1574888x13666180821155701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/01/2018] [Accepted: 08/09/2018] [Indexed: 02/07/2023]
Abstract
Cancer Stem Cells (CSCs) constitute a subpopulation at the top of the tumor cell hierarchy that contributes to tumor heterogeneity and is uniquely capable of seeding new tumors. Because of their biological properties, CSCs have been pointed out as therapeutic targets for the development of new therapies against breast cancer. The identification of drugs that selectively target breast CSCs requires a clear understanding of their biological functions and the experimental methods to evaluate such hallmarks. Herein, we review the methods to study breast CSCs properties and discuss their value in the preclinical evaluation of CSC-targeting drugs.
Collapse
Affiliation(s)
- Marco A Velasco-Velázquez
- Departamento de Farmacologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Ciudad de Mexico, Mexico
- Unidad Periférica de Investigación en Biomedicina Traslacional, Facultad de Medicina, UNAM, Ciudad de México, México
| | - Inés Velázquez-Quesada
- Departamento de Farmacologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Ciudad de Mexico, Mexico
- Unidad de Desarrollo e Investigacion en Bioprocesos, ENCB, Instituto Politecnico Nacional, Ciudad de Mexico, Mexico
| | - Luz X Vásquez-Bochm
- Departamento de Farmacologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Ciudad de Mexico, Mexico
- Posgrado en Ciencias Químicas, UNAM, Ciudad de México, México
| | - Sonia M Pérez-Tapia
- Unidad de Desarrollo e Investigacion en Bioprocesos, ENCB, Instituto Politecnico Nacional, Ciudad de Mexico, Mexico
| |
Collapse
|
200
|
Jadaun A, Sharma S, Verma R, Dixit A. Pinostrobin inhibits proliferation and induces apoptosis in cancer stem-like cells through a reactive oxygen species-dependent mechanism. RSC Adv 2019; 9:12097-12109. [PMID: 35516989 PMCID: PMC9063484 DOI: 10.1039/c8ra08380k] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/19/2019] [Indexed: 12/30/2022] Open
Abstract
Current treatments and targeted therapies for malignancies are limited due to their severe toxicity and the development of resistance against such treatments, which leads to relapse. Past evidence has indicated that a number of plant-derived dietary agents possess biological activity against highly tumorigenic and resistant cell populations associated with cancer relapse. These subpopulations, termed cancer stem-like cells (CSCs), have been targeted with plant-derived dietary flavonoids. The present study was undertaken to assess the anti-proliferative potential of pinostrobin, a dietary flavonoid, against CSCs. Sphere-forming cells were developed from HeLa cell lines using specific culture conditions. The existence of a CSC population was confirmed by the morphological examination and analysis of surface markers using confocal microscopy and flow cytometry. The effect of pinostrobin on the cell viability of the CSC population, evaluated through MTT reduction assays and the expression levels of surface markers (CD44+ and CD24+), was studied through various biological assays. HeLa-derived CSCs showed higher CD44+ and lower CD24+ expression. Pinostrobin inhibited the self-renewal capacity and sphere formation efficiency of CSCs in a dose-dependent manner. Increased ROS production, and decreased mitochondrial membrane potential and CD44+ expression indicated that pinostrobin promoted ROS-mediated apoptosis in CSCs. These results thus demonstrate the therapeutic potential and effectiveness of pinostrobin in the chemoprevention and relapse of cancer by targeting the CSC population. Thus, pinostrobin, in combination with currently available chemo and radiation therapies, could possibly be used as a safe strategy to alleviate adverse treatment effects, together with enhancing the efficacy. The anti-proliferative potential of pinostrobin, a dietary flavonoid, is evaluated against cancer stem-like cells.![]()
Collapse
Affiliation(s)
- Alka Jadaun
- Gene Regulation Laboratory
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi-110067
- India
| | - Sapna Sharma
- Gene Regulation Laboratory
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi-110067
- India
| | - Radha Verma
- Gene Regulation Laboratory
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi-110067
- India
| | - Aparna Dixit
- Gene Regulation Laboratory
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi-110067
- India
| |
Collapse
|