1
|
Haskell A, Pan S, Reese R, Powers A, Lopez MG, Lomeli S, Story C, Benton J, Blazier JC, Kaunas R, Gregory CA. Antisense mediated blockade of Dickkopf 1 attenuates tumor survival, metastases and bone damage in experimental osteosarcoma. Sci Rep 2025; 15:1878. [PMID: 39805917 PMCID: PMC11730318 DOI: 10.1038/s41598-024-84037-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy. The canonical Wnt inhibitor Dickkopf-1 (Dkk-1) has been implicated in bone destruction, tumor survival and metastases during OS. We examined the role of Dkk-1 in OS disease progression and explored strategies for targeting its activity. Dkk-1 enhances OS survival by amplifying a non-canonical Wnt pathway that upregulates aldehyde dehydrogenase 1A1. Targeting of Dkk-1 transcription with a vivo morpholino (DkkMo) reduced OS survival and enhanced osteogenic activity of OS in vitro. DkkMo as a single agent slowed tumor expansion, increased tumor necrosis, inhibited metastases and preserved bone in a PDX model of OS. DkkMo also reduced the frequency of dividing tumor cells and reinitiated a regenerative osteogenic phenotype in tumors and stroma while reducing infiltration of inflammatory cells. These findings indicate that DkkMo has the potential to safely target osteosarcoma growth, survival, metastases and bone destruction.
Collapse
Affiliation(s)
- Andrew Haskell
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, TX, 77807, USA
| | - Simin Pan
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, TX, 77807, USA
| | - Robert Reese
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, TX, USA
| | - Anthony Powers
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, TX, USA
| | - Megan G Lopez
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, TX, 77807, USA
| | - Sebastian Lomeli
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, TX, 77807, USA
| | - Christopher Story
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, TX, 77807, USA
| | - Joshua Benton
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, TX, 77807, USA
| | - J Chris Blazier
- Texas A&M Institute for Genome Sciences and Society, College Station, TX, USA
| | - Roland Kaunas
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, TX, 77807, USA
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, TX, USA
| | - Carl A Gregory
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, TX, 77807, USA.
| |
Collapse
|
2
|
Nguyen AL, Facey COB, Boman BM. The Significance of Aldehyde Dehydrogenase 1 in Cancers. Int J Mol Sci 2024; 26:251. [PMID: 39796106 PMCID: PMC11720537 DOI: 10.3390/ijms26010251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/16/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
The goal of this paper is to discuss the role of ALDH isozymes in different cancers, review advances in ALDH1-targeting cancer therapies, and explore a mechanism that explains how ALDH expression becomes elevated during cancer development. ALDH is often overexpressed in cancer, and each isoform has a unique expression pattern and a distinct role in different cancers. The abnormal expression of ALDHs in different cancer types (breast, colorectal, lung, gastric, cervical, melanoma, prostate, and renal) is presented and correlated with patient prognosis. ALDH plays a significant role in various cellular functions, such as metabolism, oxidative stress response, detoxification, and cellular differentiation. Among the ALDH families, ALDH1 has gained considerable attention as a cancer stem cell (CSC) marker due to its significant role in the maintenance of stemness and the differentiation of stem cells (SCs), along with its involvement in tumorigenesis. A description of the cellular mechanisms and physiology of ALDH1 that underlies cancer development is provided. Moreover, current advances in ALDH1-targeting cancer therapies are discussed.
Collapse
Affiliation(s)
- Anh L. Nguyen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
| | - Caroline O. B. Facey
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
| | - Bruce M. Boman
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
3
|
Galoian K, Bilbao D, Denny C, Campos Gallego N, Roberts E, Martinez D, Temple H. Targeting cancer stem cells by TPA leads to inhibition of refractory sarcoma and extended overall survival. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200905. [PMID: 39640862 PMCID: PMC11617462 DOI: 10.1016/j.omton.2024.200905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/23/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Refractory cancer recurrence in patients is a serious challenge in modern medicine. Tumor regrowth in a more aggressive and invasive drug-resistant form is caused by a specific sub-population of tumor cells defined as cancer stem cells (CSCs). While the role of CSCs in cancer relapse is recognized, the signaling pathways of CSCs-driven chemoresistance are less well understood. Moreover, there are no effective therapeutic strategies that involve specific inhibition of CSCs responsible for cancer recurrence and drug resistance. There is a clinical need to develop new therapies for patients with refractory sarcomas, particularly fibrosarcoma. These aggressive tumors, with poor overall survival, do not respond to conventional therapies. Standard systemic chemotherapy for these tumors includes doxorubicin (DOX). A Tyr peptide analog (TPA), developed in our laboratory, specifically targets CSCs by drastically reducing expression of the polycomb group protein enhancer of zester (EZH2) and its downstream targets, specifically ALDH1A1 and Nanog. In vivo experiments demonstrated that TPA inhibited tumor growth in nu/nu mice with relapsed DOX-treated fibrosarcoma 7-fold and led to improved overall (2-fold) survival. In an experimental metastatic model, the combination of TPA with DOX treatment extended overall survival 3-fold, suggesting that targeting CSC can become an effective strategy in the treatment of refractory/relapse fibrosarcoma.
Collapse
Affiliation(s)
- Karina Galoian
- Department of Orthopedic Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Daniel Bilbao
- Department of Pathology and Laboratory Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Carina Denny
- Department of Orthopedic Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | | | - Evan Roberts
- Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Daniel Martinez
- Department of Orthopedic Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - H.T. Temple
- Department of Orthopedic Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
4
|
Jan N, Sofi S, Qayoom H, Haq BU, Shabir A, Mir MA. Targeting breast cancer stem cells through retinoids: A new hope for treatment. Crit Rev Oncol Hematol 2023; 192:104156. [PMID: 37827439 DOI: 10.1016/j.critrevonc.2023.104156] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/09/2023] [Accepted: 10/06/2023] [Indexed: 10/14/2023] Open
Abstract
Breast cancer is a complex and diverse disease accounting for nearly 30% of all cancers diagnosed in females. But unfortunately, patients develop resistance to the existing chemotherapeutic regimen, resulting in approximately 90% treatment failure. With over half a million deaths annually, it is imperative to explore new therapeutic approaches to combat the disease. Within a breast tumor, a small sub-population of heterogeneous cells, with a unique ability of self-renew and differentiation and responsible for tumor formation, initiation, and recurrence are referred to as breast cancer stem cells (BCSCs). These BCSCs have been identified as one of the main contributors to chemoresistance in breast cancer, making them an attractive target for developing novel therapeutic strategies. These cells exhibit surface biomarkers such as CD44+, CD24-/LOW, ALDH, CD133, and CD49f phenotypes. Higher expression of CD44+ and ALDH activity has been associated with the formation of tumors in various cancers. Moreover, the abnormal regulation of signaling pathways, including Hedgehog, Notch, β-catenin, JAK/STAT, and P13K/AKT/mTOR, leads to the formation of cancer stem cells, resulting in the development of tumors. The growing drug resistance in BC is a significant challenge, highlighting the need for new therapeutic strategies to combat this dreadful disease. Retinoids, a large group of synthetic derivatives of vitamin A, have been studied as chemopreventive agents in clinical trials and have been shown to regulate various crucial biological functions including vision, development, inflammation, and metabolism. On a cellular level, the retinoid activity has been well characterized and translated and is known to induce differentiation and apoptosis, which play important roles in the outcome of the transformation of tissues into malignant. Retinoids have been investigated extensively for their use in the treatment and prevention of cancer due to their high receptor-binding affinity to directly modulate gene expression programs. Therefore, in this study, we aim to summarize the current understanding of BCSCs, their biomarkers, and the associated signaling pathways. Retinoids, such as Adapalene, a third-generation retinoid, have shown promising anti-cancer potential and may serve as therapeutic agents to target BCSCs.
Collapse
Affiliation(s)
- Nusrat Jan
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Shazia Sofi
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Hina Qayoom
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Burhan Ul Haq
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Aisha Shabir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India.
| |
Collapse
|
5
|
Estupiñán Ó, Rey V, Tornín J, Murillo D, Gallego B, Huergo C, Blanco-Lorenzo V, Victoria González M, Rodríguez A, Moris F, González J, Ayllón V, Ramos-Mejía V, Bigas A, Rodríguez R. Abrogation of stemness in osteosarcoma by the mithramycin analog EC-8042 is mediated by its ability to inhibit NOTCH-1 signaling. Biomed Pharmacother 2023; 162:114627. [PMID: 37018985 DOI: 10.1016/j.biopha.2023.114627] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/15/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Osteosarcomas are frequently associated to a poor prognosis and a modest response to current treatments. EC-8042 is a well-tolerated mithramycin analog that has demonstrated an efficient ability to eliminate tumor cells, including cancer stem cell subpopulations (CSC), in sarcomas. In transcriptomic and protein expression analyses, we identified NOTCH1 signaling as one of the main pro-stemness pathways repressed by EC-8042 in osteosarcomas. Overexpression of NOTCH-1 resulted in a reduced anti-tumor effect of EC-8042 in CSC-enriched 3D tumorspheres cultures. On the other hand, the depletion of the NOTCH-1 downstream target HES-1 was able to enhance the action of EC-8042 on CSCs. Moreover, HES1 depleted cells failed to recover after treatment withdrawal and showed reduced tumor growth potential in vivo. In contrast, mice xenografted with NOTCH1-overexpressing cells responded worse than parental cells to EC-8042. Finally, we found that active NOTCH1 levels in sarcoma patients was associated to advanced disease and lower survival. Overall, these data highlight the relevant role that NOTCH1 signaling plays in mediating stemness in osteosarcoma. Moreover, we demonstrate that EC-8042 is powerful inhibitor of NOTCH signaling and that the anti-CSC activity of this mithramycin analog highly rely on its ability to repress this pathway.
Collapse
|
6
|
Tornín J, Mateu-Sanz M, Rey V, Murillo D, Huergo C, Gallego B, Rodríguez A, Rodríguez R, Canal C. Cold plasma and inhibition of STAT3 selectively target tumorigenicity in osteosarcoma. Redox Biol 2023; 62:102685. [PMID: 36989573 PMCID: PMC10074989 DOI: 10.1016/j.redox.2023.102685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Osteosarcoma (OS) is a malignant type of bone cancer that arises in periods of increased bone formation. Curative strategies for these types of tumors have remained essentially unchanged for decades and the overall survival for most advanced cases is still dismally low. This is in part due to the existence of drug resistant Cancer Stem Cells (CSC) with progenitor properties that are responsible for tumor relapse and metastasis. In the quest for therapeutic alternatives for OS, Cold Atmospheric Plasmas and Plasma-Treated Liquids (PTL) have come to the limelight as a source of Reactive Oxygen and Nitrogen Species displaying selectivity towards a variety of cancer cell lines. However, their effects on CSC subpopulations and in vivo tumor growth have been barely studied to date. By employing bioengineered 3D tumor models and in vivo assays, here we show that low doses of PTL increase the levels of pro-stemness factors and the self-renewal ability of OS cells, coupled to an enhanced in vivo tumor growth potential. This could have critical implications to the field. By proposing a combined treatment, our results demonstrate that the deleterious pro-stemness signals mediated by PTL can be abrogated when this is combined with the STAT3 inhibitor S3I-201, resulting in a strong suppression of in vivo tumor growth. Overall, our study unveils an undesirable stem cell-promoting function of PTL in cancer and supports the use of combinatorial strategies with STAT3 inhibitors as an efficient treatment for OS avoiding critical side effects. We anticipate our work to be a starting point for wider studies using relevant 3D tumor models to evaluate the effects of plasma-based therapies on tumor subpopulations of different cancer types. Furthermore, combination with STAT3 inhibition or other suitable cancer type-specific targets can be relevant to consolidate the development of the field.
Collapse
|
7
|
Jung HS, Koo S, Won M, An S, Park H, Sessler JL, Han J, Kim JS. Cu(ii)-BODIPY photosensitizer for CAIX overexpressed cancer stem cell therapy. Chem Sci 2023; 14:1808-1819. [PMID: 36819853 PMCID: PMC9930985 DOI: 10.1039/d2sc03945a] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 01/14/2023] [Indexed: 01/21/2023] Open
Abstract
Chemoresistance originating from cancer stem cells (CSCs) is a major cause of cancer treatment failure and highlights the need to develop CSC-targeting therapies. Although enormous progress in both photodynamic therapy (PDT) and chemodynamic therapy (CDT) has been made in recent decades, the efficacy of these modalities against CSC remains limited. Here, we report a new generation photosensitizer, CA9-BPS-Cu(ii), a system that combines three subunits within a single molecule, namely a copper catalyst for CDT, a boron dipyrromethene photosensitizer for PDT, and acetazolamide for CSC targeting via carbonic anhydrase-9 (CA9) binding. A therapeutic effect in MDA-MB-231 cells was observed that is ascribed to elevated oxidative stress mediated by a combined CDT/PDT effect, as well as through copper-catalysed glutathione oxidation. The CSC targeting ability of CA9-BPS-Cu(ii) was evident from the enhanced affinity of CA9-BPS-Cu(ii) towards CD133-positive MDA-MB-231 cells where CA9 is overexpressed vs. CD133-negative cells. Moreover, the efficacy of CA9-BPS-Cu(ii) was successfully demonstrated in a xenograft mouse tumour model.
Collapse
Affiliation(s)
- Hyo Sung Jung
- Department of Biological Sciences, Hyupsung University Hwasung-si 18330 Korea
| | - Seyoung Koo
- Department of Chemistry, Korea University Seoul 02841 Korea
| | - Miae Won
- Department of Chemistry, Korea University Seoul 02841 Korea
| | - Seeun An
- Department of Biological Sciences, Hyupsung University Hwasung-si 18330 Korea
| | - Haebeen Park
- Department of Biological Sciences, Hyupsung University Hwasung-si 18330 Korea
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin Austin Texas 78712-1224 USA
| | - Jiyou Han
- Department of Biological Sciences, Hyupsung University Hwasung-si 18330 Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University Seoul 02841 Korea
| |
Collapse
|
8
|
Eid RA, Alaa Edeen M, Shedid EM, Kamal ASS, Warda MM, Mamdouh F, Khedr SA, Soltan MA, Jeon HW, Zaki MSA, Kim B. Targeting Cancer Stem Cells as the Key Driver of Carcinogenesis and Therapeutic Resistance. Int J Mol Sci 2023; 24:ijms24021786. [PMID: 36675306 PMCID: PMC9861138 DOI: 10.3390/ijms24021786] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 01/18/2023] Open
Abstract
The emerging concept of cancer stem cells (CSCs) as the key driver behind carcinogenesis, progression, and diversity has displaced the prior model of a tumor composed of cells with similar subsequently acquired mutations and an equivalent capacity for renewal, invasion, and metastasis. This significant change has shifted the research focus toward targeting CSCs to eradicate cancer. CSCs may be characterized using cell surface markers. They are defined by their capacity to self-renew and differentiate, resist conventional therapies, and generate new tumors following repeated transplantation in xenografted mice. CSCs' functional capabilities are governed by various intracellular and extracellular variables such as pluripotency-related transcription factors, internal signaling pathways, and external stimuli. Numerous natural compounds and synthetic chemicals have been investigated for their ability to disrupt these regulatory components and inhibit stemness and terminal differentiation in CSCs, hence achieving clinical implications. However, no cancer treatment focuses on the biological consequences of these drugs on CSCs, and their functions have been established. This article provides a biomedical discussion of cancer at the time along with an overview of CSCs and their origin, features, characterization, isolation techniques, signaling pathways, and novel targeted therapeutic approaches. Additionally, we highlighted the factors endorsed as controlling or helping to promote stemness in CSCs. Our objective was to encourage future studies on these prospective treatments to develop a framework for their application as single or combined therapeutics to eradicate various forms of cancer.
Collapse
Affiliation(s)
- Refaat A. Eid
- Pathology Department, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia
| | - Muhammad Alaa Edeen
- Cell Biology, Histology & Genetics Division, Biology Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
- Correspondence: (M.A.E.); (B.K.)
| | - Eslam M. Shedid
- Biotechnology Division, Zoology Department, Faculty of Science, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Al Shaimaa S. Kamal
- Biotechnology Department, Faculty of Agriculture, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Mona M. Warda
- Biotechnology Division, Zoology Department, Faculty of Science, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Farag Mamdouh
- Biotechnology Division, Zoology Department, Faculty of Science, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Sohila A. Khedr
- Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta 31733, Egypt
| | - Mohamed A. Soltan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, Ismailia 41611, Egypt
| | - Hee Won Jeon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Mohamed Samir A. Zaki
- Anatomy Department, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia
- Department of Histology and Cell Biology, College of Medicine, Zagazig University, Zagazig 31527, Egypt
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Correspondence: (M.A.E.); (B.K.)
| |
Collapse
|
9
|
Exploring the Use of Cold Atmospheric Plasma to Overcome Drug Resistance in Cancer. Biomedicines 2023; 11:biomedicines11010208. [PMID: 36672716 PMCID: PMC9855365 DOI: 10.3390/biomedicines11010208] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
Drug resistance is a major problem in cancer treatment, as it limits the effectiveness of pharmacological agents and can lead to disease progression. Cold atmospheric plasma (CAP) is a technology that uses ionized gas (plasma) to generate reactive oxygen and nitrogen species (RONS) that can kill cancer cells. CAP is a novel approach for overcoming drug resistance in cancer. In recent years, there has been a growing interest in using CAP to enhance the effectiveness of chemotherapy drugs. In this review, we discuss the mechanisms behind this phenomenon and explore its potential applications in cancer treatment. Going through the existing literature on CAP and drug resistance in cancer, we highlight the challenges and opportunities for further research in this field. Our review suggests that CAP could be a promising option for overcoming drug resistance in cancer and warrants further investigation.
Collapse
|
10
|
Cancer Stem Cell Markers in Rhabdomyosarcoma in Children. Diagnostics (Basel) 2022; 12:diagnostics12081895. [PMID: 36010245 PMCID: PMC9406733 DOI: 10.3390/diagnostics12081895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: The aim of the present study was to assess the cancer stem cell (CSC) markers CD24, CD44, CD133, and ALDH1A1 in rhabdomyosarcoma (RMS) in children and to define their prognostic role in this group of patients. (2) Methods: The study material was archival tissue specimens collected from 49 patients under 18 years of age and who had been diagnosed with RMS. Immunohistochemistry (IHC) was used to evaluate the expression of the selected CSC markers in the tumor tissue. Expression was evaluated using a semiquantitative IRS scale based on the one developed by Remmele and Stenger and was correlated with the clinical and pathomorphological parameters of prognostic importance in RMS. (3) Results: Expression of the selected CSC markers CD24, CD44, CD133, and ALDH1A1 was demonstrated in 83.7%, 55.1%, 81.6%, and 100% of the RMS patients, respectively. The expression of all of the assessed CSC markers was statistically significantly higher in the study group versus the control group. No significant correlation was found between the expression of the selected CSC markers and clinical and pathological prognostic factors that were analyzed. The expression of the CSC markers did not have a significant influence on RMS survival rates. (4) Conclusions: The results of the conducted study confirm the expression of selected CSC markers in rhabdomyosarcoma tissue in children. The study did not support the prognostic relevance of the expression of any of the assessed CSC markers. However, further studies are needed to fully understand the relevance of the selected CSC markers in RMS carcinogenesis.
Collapse
|
11
|
Kim J, Park A, Hwang J, Zhao X, Kwak J, Kim HW, Ku M, Yang J, Kim TI, Jeong KS, Choi U, Lee H, Shin SJ. KS10076, a chelator for redox-active metal ions, induces ROS-mediated STAT3 degradation in autophagic cell death and eliminates ALDH1 + stem cells. Cell Rep 2022; 40:111077. [PMID: 35858554 DOI: 10.1016/j.celrep.2022.111077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 03/17/2022] [Accepted: 06/20/2022] [Indexed: 12/21/2022] Open
Abstract
Redox-active metal ions are pivotal for rapid metabolism, proliferation, and aggression across cancer types, and this presents metal chelation as an attractive cancer cell-targeting strategy. Here, we identify a metal chelator, KS10076, as a potent anti-cancer drug candidate. A metal-bound KS10076 complex with redox potential for generating hydrogen peroxide and superoxide anions induces intracellular reactive oxygen species (ROS). The elevation of ROS by KS10076 promotes the destabilization of signal transducer and activator of transcription 3, removes aldehyde dehydrogenase isoform 1-positive cancer stem cells, and subsequently induces autophagic cell death. Bioinformatic analysis of KS10076 susceptibility in pan-cancer cells shows that KS10076 potentially targets cancer cells with increased mitochondrial function. Furthermore, patient-derived organoid models demonstrate that KS10076 efficiently represses cancer cells with active KRAS, and fluorouracil resistance, which suggests clinical advantages.
Collapse
Affiliation(s)
- Jaehee Kim
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Areum Park
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Jieon Hwang
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Xianghua Zhao
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jaesung Kwak
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Hyun Woo Kim
- Chemical Data-Driven Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Minhee Ku
- Department of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Severance of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jaemoon Yang
- Department of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Severance of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Tae Il Kim
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kyu-Sung Jeong
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Uyeong Choi
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyuk Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea.
| | - Sang Joon Shin
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
12
|
Gallego B, Murillo D, Rey V, Huergo C, Estupiñán Ó, Rodríguez A, Tornín J, Rodríguez R. Addressing Doxorubicin Resistance in Bone Sarcomas Using Novel Drug-Resistant Models. Int J Mol Sci 2022; 23:ijms23126425. [PMID: 35742867 PMCID: PMC9224263 DOI: 10.3390/ijms23126425] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 02/04/2023] Open
Abstract
Bone sarcomas have not shown a significant improvement in survival for decades, due, in part, to the development of resistance to current systemic treatments, such as doxorubicin. To better understand those mechanisms mediating drug-resistance we generated three osteosarcoma and one chondrosarcoma cell lines with a stable doxorubicin-resistant phenotype, both in vitro and in vivo. These resistant strains include a pioneer model generated from a patient-derived chondrosarcoma line. The resistant phenotype was characterized by a weaker induction of apoptosis and DNA damage after doxorubicin treatment and a lower migratory capability. In addition, all resistant lines expressed higher levels of ABC pumps; meanwhile, no clear trends were found in the expression of anti-apoptotic and stem cell-related factors. Remarkably, upon the induction of resistance, the proliferation potential was reduced in osteosarcoma lines but enhanced in the chondrosarcoma model. The exposure of resistant lines to other anti-tumor drugs revealed an increased response to cisplatin and/or methotrexate in some models. Finally, the ability to retain the resistant phenotype in vivo was confirmed in an osteosarcoma model. Altogether, this work evidenced the co-existence of common and case-dependent phenotypic traits and mechanisms associated with the development of resistance to doxorubicin in bone sarcomas.
Collapse
Affiliation(s)
- Borja Gallego
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n 33011 Oviedo, Spain; (B.G.); (D.M.); (V.R.); (C.H.); (Ó.E.); (A.R.); (J.T.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en Oncología (CIBERONC), 28029 Madrid, Spain
| | - Dzohara Murillo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n 33011 Oviedo, Spain; (B.G.); (D.M.); (V.R.); (C.H.); (Ó.E.); (A.R.); (J.T.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Verónica Rey
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n 33011 Oviedo, Spain; (B.G.); (D.M.); (V.R.); (C.H.); (Ó.E.); (A.R.); (J.T.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en Oncología (CIBERONC), 28029 Madrid, Spain
| | - Carmen Huergo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n 33011 Oviedo, Spain; (B.G.); (D.M.); (V.R.); (C.H.); (Ó.E.); (A.R.); (J.T.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Óscar Estupiñán
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n 33011 Oviedo, Spain; (B.G.); (D.M.); (V.R.); (C.H.); (Ó.E.); (A.R.); (J.T.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Aida Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n 33011 Oviedo, Spain; (B.G.); (D.M.); (V.R.); (C.H.); (Ó.E.); (A.R.); (J.T.)
| | - Juan Tornín
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n 33011 Oviedo, Spain; (B.G.); (D.M.); (V.R.); (C.H.); (Ó.E.); (A.R.); (J.T.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - René Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n 33011 Oviedo, Spain; (B.G.); (D.M.); (V.R.); (C.H.); (Ó.E.); (A.R.); (J.T.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en Oncología (CIBERONC), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-985-101-399
| |
Collapse
|
13
|
Pan S, Cesarek M, Godoy C, Co CM, Schindler C, Padilla K, Haskell A, Barreda H, Story C, Poole R, Dabney A, Gregory CA. Morpholino-driven blockade of Dkk-1 in osteosarcoma inhibits bone damage and tumour expansion by multiple mechanisms. Br J Cancer 2022; 127:43-55. [PMID: 35277659 PMCID: PMC9276700 DOI: 10.1038/s41416-022-01764-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 02/02/2022] [Accepted: 02/15/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Background
Osteosarcoma (OS) is the most common primary bone malignancy. Chemotherapy plays an essential role in OS treatment, potentially doubling 5-year event-free survival if tumour necrosis can be stimulated. The canonical Wnt inhibitor Dickkopf-1 (Dkk-1) enhances OS survival in part through upregulation of aldehyde-dehydrogenase-1A1 which neutralises reactive oxygen species originating from nutritional stress and chemotherapeutic challenge.
Methods
A vivo morpholino (DkkMo) was employed to block the expression of Dkk-1 in OS cells. Cell mitosis, gene expression and bone destruction were measured in vitro and in vivo in the presence and absence of doxorubicin (DRB).
Results
DkkMo reduced the expression of Dkk-1 and Aldh1a1, reduced expansion of OS tumours, preserved bone volume and architecture and stimulated tumour necrosis. This was observed in the presence or absence of DRB.
Conclusion
These results indicate that administration of DkkMo with or without chemotherapeutics can substantially improve OS outcome with respect to tumour expansion and osteolytic corruption of bone in experimental OS model.
Collapse
|
14
|
Mohamed MAN, Ibrahim BB, El Sheikh SAM, Magid MSA. Stem Cell Marker Aldehyde Dehydrogenase 1A1 Expression in Triple-negative Breast Carcinoma. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Increasing evidence has proposed that tumor contains tumor initiating cells or cancer stem cells (CSCs) are responsible for its progression and relapse. Aldehyde dehydrogenase 1A1 (ALDH1A1) has recently been identified as a marker for cancer stem cells in some human malignancies including breast cancer.Triple negative breast carcinomas (TNBCs) are group of primary breast tumors with aggressive clinical behavior that have no targeted therapy at present.
Aim: The assessment of immunohistochemical expression of ALDH1A1 in triple negative breast carcinoma and its correlation with the clinicopathological features of TNBC.
Material and Methods: This study consisted of 60 cases of TNBC. Immunohistochemical reactions were carried out by using ALDH1A1 monoclonal antibody. A total score of ALDH1A1 expression is obtained by multiplying the score of staining intensity & percentage of stained cells to obtain score ranging from 0 up to 300. Due to the relatively large number of positive cases, a statistical analysis was performed with a negative (score ≤ 10) and positive (score > 10) cutoff [1].
Results: Evaluation of the results of immunostaining for ALDH1A1 showed 88.3% of totalcases (53 cases) having a positive cytoplasmic reactivity. Statistical analysis for a possible correlation between ALDH1A1 expression and prognostic clinicopathological parameters; age, size, tumor grade, histologic subtypes, lymphovascular invasion, intraductal components, tumor infiltrating lymphocytes and TNM stage grouping revealed a non-significant correlation.
Conclusion: ALDH1A1 couldn’t be used solely as a diagnostic or prognostic marker In TNBCs. Further research combining with other biomarkers and with a greater number of patients is necessary to confirm the role of ALDH1A1 in TNBC.
Key words: Cancer stem cells (CSCs), Aldehyde dehydrogenase 1A1 (ALDH1A1), Triple Negative Breast cancer (TNBC).
Collapse
|
15
|
ALDH1A1 Gene Expression and Cellular Copper Levels between Low and Highly Metastatic Osteosarcoma Provide a Case for Novel Repurposing with Disulfiram and Copper. Sarcoma 2022; 2022:7157507. [PMID: 35125923 PMCID: PMC8816591 DOI: 10.1155/2022/7157507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/07/2022] [Indexed: 12/24/2022] Open
Abstract
Aldehyde dehydrogenase 1A1 (ALDH) is a cancer stem cell marker highly expressed in metastatic cells. Disulfiram (Dis) is an FDA-approved antialcoholism drug that inhibits ALDH and has been studied as a candidate for drug repurposing in multiple neoplasia. Dis cytotoxicity in cancer cells has been shown to be copper-dependent, in part due to Dis's ability to function as a bivalent metal ion chelator of copper (Cu). The objectives of this research were to test ALDH expression levels and Cu concentrations in sarcoma patient tumors and human osteosarcoma (OS) cell lines with differing metastatic phenotypes. We also sought to evaluate Dis + Cu combination therapy in human OS cells. Intracellular Cu was inversely proportional to the metastatic phenotype in human OS cell lines (SaOS2 > LM2 > LM7). Nonmetastatic human sarcoma tumors demonstrated increased Cu concentrations compared with metastatic tumors. qPCR demonstrated that ALDH expression was significantly increased in highly metastatic LM2 and LM7 human OS cell lines compared with low metastatic SaOS2. Tumor cells from sarcoma patients with metastatic disease displayed significantly increased ALDH expression compared with tumor cells from patients without metastatic disease. Serum Cu concentration in canine OS versus normal canine patients demonstrated similar trends. Dis demonstrated selective cytotoxicity compared with human multipotential stromal cells (MSCs): Dis-treated OS cells demonstrated increased apoptosis, whereas MSCs did not. CuCl2 combined with Dis and low-dose doxorubicin resulted in a superior cytotoxic effect in both SaOS2 and LM7 cell lines. In summary, ALDH gene expression and Cu levels are altered between low and highly metastatic human OS cells, canine samples, and patient tumors. Our findings support the feasibility of a repurposed drug strategy for Dis and Cu in combination with low-dose anthracycline to specifically target metastatic OS cells.
Collapse
|
16
|
Liu CC, Wu CL, Lin MX, Sze CI, Gean PW. Disulfiram Sensitizes a Therapeutic-Resistant Glioblastoma to the TGF-β Receptor Inhibitor. Int J Mol Sci 2021; 22:ijms221910496. [PMID: 34638842 PMCID: PMC8508702 DOI: 10.3390/ijms221910496] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/16/2021] [Accepted: 09/24/2021] [Indexed: 02/08/2023] Open
Abstract
Despite neurosurgery following radiation and chemotherapy, residual glioblastoma (GBM) cells develop therapeutic resistance (TR) leading to recurrence. The GBM heterogeneity confers TR. Therefore, an effective strategy must target cancer stem cells (CSCs) and other malignant cancer cells. TGF-β and mesenchymal transition are the indicators for poor prognoses. The activity of aldehyde dehydrogenases (ALDHs) is a functional CSC marker. However, the interplay between TGF-β and ALDHs remains unclear. We developed radiation-resistant and radiation-temozolomide-resistant GBM models to investigate the underlying mechanisms conferring TR. Galunisertib is a drug targeting TGF-β receptors. Disulfiram (DSF) is an anti-alcoholism drug which functions by inhibiting ALDHs. The anti-tumor effects of combining DSF and Galunisertib were evaluated by in vitro cell grow, wound healing, Transwell assays, and in vivo orthotopic GBM model. Mesenchymal-like phenotype was facilitated by TGF-β in TR GBM. Additionally, TR activated ALDHs. DSF inhibited TR-induced cell migration and tumor sphere formation. However, DSF did not affect the tumor growth in vivo. Spectacularly, DSF sensitized TR GBM to Galunisertib both in vitro and in vivo. ALDH activity positively correlated with TGF-β-induced mesenchymal properties in TR GBM. CSCs and mesenchymal-like GBM cells targeted together by combining DSF and Galunisertib may be a good therapeutic strategy for recurrent GBM patients.
Collapse
Affiliation(s)
- Chan-Chuan Liu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan;
| | - Cheng-Lin Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng-Kung University, Tainan 701, Taiwan;
- Department of Pathology, College of Medicine, National Cheng Kung University Hospital, National Cheng-Kung University, Tainan 701, Taiwan
| | - Meng-Xuan Lin
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan;
| | - Chun-I Sze
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan;
- Department of Pathology, College of Medicine, National Cheng Kung University Hospital, National Cheng-Kung University, Tainan 701, Taiwan
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan;
- Correspondence: (C.-I.S.); (P.-W.G.)
| | - Po-Wu Gean
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan;
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan
- Department of Biotechnology and Bioindustry Sciences, National Cheng-Kung University, Tainan 701, Taiwan
- Correspondence: (C.-I.S.); (P.-W.G.)
| |
Collapse
|
17
|
Estupiñán Ó, Niza E, Bravo I, Rey V, Tornín J, Gallego B, Clemente-Casares P, Moris F, Ocaña A, Blanco-Lorenzo V, Rodríguez-Santamaría M, Vallina-Álvarez A, González MV, Rodríguez A, Hermida-Merino D, Alonso-Moreno C, Rodríguez R. Mithramycin delivery systems to develop effective therapies in sarcomas. J Nanobiotechnology 2021; 19:267. [PMID: 34488783 PMCID: PMC8419920 DOI: 10.1186/s12951-021-01008-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Sarcomas comprise a group of aggressive malignancies with very little treatment options beyond standard chemotherapy. Reposition of approved drugs represents an attractive approach to identify effective therapeutic compounds. One example is mithramycin (MTM), a natural antibiotic which has demonstrated a strong antitumour activity in several tumour types, including sarcomas. However, its widespread use in the clinic was limited by its poor toxicity profile. RESULTS In order to improve the therapeutic index of MTM, we have loaded MTM into newly developed nanocarrier formulations. First, polylactide (PLA) polymeric nanoparticles (NPs) were generated by nanoprecipitation. Also, liposomes (LIP) were prepared by ethanol injection and evaporation solvent method. Finally, MTM-loaded hydrogels (HG) were obtained by passive loading using a urea derivative non-peptidic hydrogelator. MTM-loaded NPs and LIP display optimal hydrodynamic radii between 80 and 105 nm with a very low polydispersity index (PdI) and encapsulation efficiencies (EE) of 92 and 30%, respectively. All formulations show a high stability and different release rates ranging from a fast release in HG (100% after 30 min) to more sustained release from NPs (100% after 24 h) and LIP (40% after 48 h). In vitro assays confirmed that all assayed MTM formulations retain the cytotoxic, anti-invasive and anti-stemness potential of free MTM in models of myxoid liposarcoma, undifferentiated pleomorphic sarcoma and chondrosarcoma. In addition, whole genome transcriptomic analysis evidenced the ability of MTM, both free and encapsulated, to act as a multi-repressor of several tumour-promoting pathways at once. Importantly, the treatment of mice bearing sarcoma xenografts showed that encapsulated MTM exhibited enhanced therapeutic effects and was better tolerated than free MTM. CONCLUSIONS Overall, these novel formulations may represent an efficient and safer MTM-delivering alternative for sarcoma treatment.
Collapse
Affiliation(s)
- Óscar Estupiñán
- Sarcomas and Experimental Therapeutics Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, 33011, Oviedo, Spain.,CIBER en Oncología (CIBERONC), 28029, Madrid, Spain
| | - Enrique Niza
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008, Albacete, Spain.,Universidad de Castilla-La Mancha, Facultad de Farmacia de Albacete, 02008, Albacete, Spain
| | - Iván Bravo
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008, Albacete, Spain.,Universidad de Castilla-La Mancha, Facultad de Farmacia de Albacete, 02008, Albacete, Spain
| | - Verónica Rey
- Sarcomas and Experimental Therapeutics Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, 33011, Oviedo, Spain
| | - Juan Tornín
- Sarcomas and Experimental Therapeutics Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, 33011, Oviedo, Spain.,Materials Science and Engineering Department, Universitat Politècnica de Catalunya (UPC), Escola d'Enginyeria Barcelona Est (EEBE), 08019, Barcelona, Spain.,Institut de Recerca Sant Joan de Déu, 08034, Barcelona, Spain
| | - Borja Gallego
- Sarcomas and Experimental Therapeutics Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011, Oviedo, Spain
| | - Pilar Clemente-Casares
- Universidad de Castilla-La Mancha, Facultad de Farmacia de Albacete, 02008, Albacete, Spain.,Centro Regional de Investigaciones Biomédicas (CRIB), UCLM, 02008, Albacete, Spain
| | | | - Alberto Ocaña
- CIBER en Oncología (CIBERONC), 28029, Madrid, Spain.,Experimental Therapeutics Unit, Hospital Clínico San Carlos, IdISSC, 28040, Madrid, Spain
| | - Verónica Blanco-Lorenzo
- Sarcomas and Experimental Therapeutics Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011, Oviedo, Spain.,Servicio de Anatomía Patológica, Hospital Universitario Central de Asturias, 33011, Oviedo, Spain
| | - Mar Rodríguez-Santamaría
- Sarcomas and Experimental Therapeutics Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011, Oviedo, Spain
| | - Aitana Vallina-Álvarez
- Instituto Universitario de Oncología del Principado de Asturias, 33011, Oviedo, Spain.,Servicio de Anatomía Patológica, Hospital Universitario Central de Asturias, 33011, Oviedo, Spain
| | - M Victoria González
- Sarcomas and Experimental Therapeutics Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, 33011, Oviedo, Spain.,CIBER en Oncología (CIBERONC), 28029, Madrid, Spain.,Departamento de Cirugía, Universidad de Oviedo, 33006, Oviedo, Spain
| | - Aida Rodríguez
- Sarcomas and Experimental Therapeutics Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011, Oviedo, Spain
| | - Daniel Hermida-Merino
- Netherlands Organisation for Scientific Research (NWO), DUBBLE@ESRF, 38000, Grenoble, France
| | - Carlos Alonso-Moreno
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008, Albacete, Spain. .,Universidad de Castilla-La Mancha, Facultad de Farmacia de Albacete, 02008, Albacete, Spain.
| | - René Rodríguez
- Sarcomas and Experimental Therapeutics Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011, Oviedo, Spain. .,Instituto Universitario de Oncología del Principado de Asturias, 33011, Oviedo, Spain. .,CIBER en Oncología (CIBERONC), 28029, Madrid, Spain.
| |
Collapse
|
18
|
Context-dependent modulation of aggressiveness of pediatric tumors by individual oncogenic RAS isoforms. Oncogene 2021; 40:4955-4966. [PMID: 34172934 PMCID: PMC8342309 DOI: 10.1038/s41388-021-01904-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/25/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023]
Abstract
A prototypic pediatric cancer that frequently shows activation of RAS signaling is embryonal rhabdomyosarcoma (ERMS). ERMS also show aberrant Hedgehog (HH)/GLI signaling activity and can be driven by germline mutations in this pathway. We show, that in ERMS cell lines derived from sporadic tumors i.e. from tumors not caused by an inherited genetic variant, HH/GLI signaling plays a subordinate role, because oncogenic mutations in HRAS, KRAS, or NRAS (collectively named oncRAS) inhibit the main HH target GLI1 via the MEK/ERK-axis, but simultaneously increase proliferation and tumorigenicity. oncRAS also modulate expression of stem cell markers in an isoform- and context-dependent manner. In Hh-driven murine ERMS that are caused by a Patched mutation, oncHRAS and mainly oncKRAS accelerate tumor development, whereas oncNRAS induces a more differentiated phenotype. These features occur when the oncRAS mutations are induced at the ERMS precursor stage, but not when induced in already established tumors. Moreover, in contrast to what is seen in human cell lines, oncRAS mutations do not alter Hh signaling activity and marginally affect expression of stem cell markers. Together, all three oncRAS mutations seem to be advantageous for ERMS cell lines despite inhibition of HH signaling and isoform-specific modulation of stem cell markers. In contrast, oncRAS mutations do not inhibit Hh-signaling in Hh-driven ERMS. In this model, oncRAS mutations seem to be advantageous for specific ERMS populations that occur within a specific time window during ERMS development. In addition, this window may be different for individual oncRAS isoforms, at least in the mouse.
Collapse
|
19
|
Menéndez ST, Gallego B, Murillo D, Rodríguez A, Rodríguez R. Cancer Stem Cells as a Source of Drug Resistance in Bone Sarcomas. J Clin Med 2021; 10:jcm10122621. [PMID: 34198693 PMCID: PMC8232081 DOI: 10.3390/jcm10122621] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/16/2022] Open
Abstract
Bone sarcomas are commonly characterized by a high degree of intra-tumor heterogeneity, which in part is due to the presence of subpopulations of tumor cells presenting stem cell properties. Similar to normal stem cells, these cancer stem cells (CSCs) display a drug resistant phenotype and therefore are responsible for relapses and tumor dissemination. Drug resistance in bone sarcomas could be enhanced/modulated during tumor evolution though the acquisition of (epi)-genetic alterations and the adaptation to changing microenvironments, including drug treatments. Here we summarize findings supporting the involvement of pro-stemness signaling in the development of drug resistance in bone sarcomas. This include the activation of well-known pro-stemness pathways (Wnt/β-Cat, NOTCH or JAT/STAT pathways), changes in the metabolic and autophagic activities, the alteration of epigenetic pathways, the upregulation of specific non-coding RNAs and the crosstalk with different microenvironmental factors. This altered signaling is expected to be translated to the clinic in the form of biomarkers of response and new therapies able to overcome drug resistance.
Collapse
Affiliation(s)
- Sofía T. Menéndez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma s/n, 33011 Oviedo, Spain; (B.G.); (D.M.); (A.R.)
- Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, 33006 Oviedo, Spain
- CIBER en Oncología (CIBERONC), 28029 Madrid, Spain
- Correspondence: (S.T.M.); (R.R.)
| | - Borja Gallego
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma s/n, 33011 Oviedo, Spain; (B.G.); (D.M.); (A.R.)
| | - Dzohara Murillo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma s/n, 33011 Oviedo, Spain; (B.G.); (D.M.); (A.R.)
| | - Aida Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma s/n, 33011 Oviedo, Spain; (B.G.); (D.M.); (A.R.)
| | - René Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Avenida de Roma s/n, 33011 Oviedo, Spain; (B.G.); (D.M.); (A.R.)
- Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, 33006 Oviedo, Spain
- CIBER en Oncología (CIBERONC), 28029 Madrid, Spain
- Correspondence: (S.T.M.); (R.R.)
| |
Collapse
|
20
|
Estupiñán Ó, Rendueles C, Suárez P, Rey V, Murillo D, Morís F, Gutiérrez G, Blanco-López MDC, Matos M, Rodríguez R. Nano-Encapsulation of Mithramycin in Transfersomes and Polymeric Micelles for the Treatment of Sarcomas. J Clin Med 2021; 10:jcm10071358. [PMID: 33806182 PMCID: PMC8037461 DOI: 10.3390/jcm10071358] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/13/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Sarcomas are aggressive tumors which often show a poor response to current treatments. As a promising therapeutic alternative, we focused on mithramycin (MTM), a natural antibiotic with a promising anti-tumor activity but also a relevant systemic toxicity. Therefore, the encapsulation of MTM in nano-delivery systems may represent a way to increase its therapeutic window. Here, we designed novel transfersomes and PLGA polymeric micelles by combining different membrane components (phosphatidylcholine, Span 60, Tween 20 and cholesterol) to optimize the nanoparticle size, polydispersity index (PDI) and encapsulation efficiency (EE). Using both thin film hydration and the ethanol injection methods we obtained MTM-loaded transferosomes displaying an optimal hydrodynamic diameter of 100–130 nm and EE values higher than 50%. Additionally, we used the emulsion/solvent evaporation method to synthesize polymeric micelles with a mean size of 228 nm and a narrow PDI, capable of encapsulating MTM with EE values up to 87%. These MTM nano-delivery systems mimicked the potent anti-tumor activity of free MTM, both in adherent and cancer stem cell-enriched tumorsphere cultures of myxoid liposarcoma and chondrosarcoma models. Similarly to free MTM, nanocarrier-delivered MTM efficiently inhibits the signaling mediated by the pro-oncogenic factor SP1. In summary, we provide new formulations for the efficient encapsulation of MTM which may constitute a safer delivering alternative to be explored in future clinical uses.
Collapse
Affiliation(s)
- Óscar Estupiñán
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (Ó.E.); (V.R.); (D.M.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en Oncología (CIBERONC), 28029 Madrid, Spain
- Department of Chemical and Environmental Engineering, University of Oviedo, 33006 Oviedo, Spain; (C.R.); (P.S.); (G.G.)
| | - Claudia Rendueles
- Department of Chemical and Environmental Engineering, University of Oviedo, 33006 Oviedo, Spain; (C.R.); (P.S.); (G.G.)
| | - Paula Suárez
- Department of Chemical and Environmental Engineering, University of Oviedo, 33006 Oviedo, Spain; (C.R.); (P.S.); (G.G.)
| | - Verónica Rey
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (Ó.E.); (V.R.); (D.M.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Dzohara Murillo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (Ó.E.); (V.R.); (D.M.)
| | | | - Gemma Gutiérrez
- Department of Chemical and Environmental Engineering, University of Oviedo, 33006 Oviedo, Spain; (C.R.); (P.S.); (G.G.)
- Asturias University Institute of Biotechnology, University of Oviedo, 33006 Oviedo, Spain;
| | - María del Carmen Blanco-López
- Asturias University Institute of Biotechnology, University of Oviedo, 33006 Oviedo, Spain;
- Department of Physical and Analytical Chemistry, University of Oviedo, 33006 Oviedo, Spain
| | - María Matos
- Department of Chemical and Environmental Engineering, University of Oviedo, 33006 Oviedo, Spain; (C.R.); (P.S.); (G.G.)
- Asturias University Institute of Biotechnology, University of Oviedo, 33006 Oviedo, Spain;
- Correspondence: (M.M.); (R.R.)
| | - René Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (Ó.E.); (V.R.); (D.M.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en Oncología (CIBERONC), 28029 Madrid, Spain
- Correspondence: (M.M.); (R.R.)
| |
Collapse
|
21
|
Grünewald TGP, Alonso M, Avnet S, Banito A, Burdach S, Cidre‐Aranaz F, Di Pompo G, Distel M, Dorado‐Garcia H, Garcia‐Castro J, González‐González L, Grigoriadis AE, Kasan M, Koelsche C, Krumbholz M, Lecanda F, Lemma S, Longo DL, Madrigal‐Esquivel C, Morales‐Molina Á, Musa J, Ohmura S, Ory B, Pereira‐Silva M, Perut F, Rodriguez R, Seeling C, Al Shaaili N, Shaabani S, Shiavone K, Sinha S, Tomazou EM, Trautmann M, Vela M, Versleijen‐Jonkers YMH, Visgauss J, Zalacain M, Schober SJ, Lissat A, English WR, Baldini N, Heymann D. Sarcoma treatment in the era of molecular medicine. EMBO Mol Med 2020; 12:e11131. [PMID: 33047515 PMCID: PMC7645378 DOI: 10.15252/emmm.201911131] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Sarcomas are heterogeneous and clinically challenging soft tissue and bone cancers. Although constituting only 1% of all human malignancies, sarcomas represent the second most common type of solid tumors in children and adolescents and comprise an important group of secondary malignancies. More than 100 histological subtypes have been characterized to date, and many more are being discovered due to molecular profiling. Owing to their mostly aggressive biological behavior, relative rarity, and occurrence at virtually every anatomical site, many sarcoma subtypes are in particular difficult-to-treat categories. Current multimodal treatment concepts combine surgery, polychemotherapy (with/without local hyperthermia), irradiation, immunotherapy, and/or targeted therapeutics. Recent scientific advancements have enabled a more precise molecular characterization of sarcoma subtypes and revealed novel therapeutic targets and prognostic/predictive biomarkers. This review aims at providing a comprehensive overview of the latest advances in the molecular biology of sarcomas and their effects on clinical oncology; it is meant for a broad readership ranging from novices to experts in the field of sarcoma.
Collapse
Affiliation(s)
- Thomas GP Grünewald
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
- Division of Translational Pediatric Sarcoma ResearchGerman Cancer Research Center (DKFZ), Hopp Children's Cancer Center (KiTZ), German Cancer Consortium (DKTK)HeidelbergGermany
- Institute of PathologyHeidelberg University HospitalHeidelbergGermany
| | - Marta Alonso
- Program in Solid Tumors and BiomarkersFoundation for the Applied Medical ResearchUniversity of Navarra PamplonaPamplonaSpain
| | - Sofia Avnet
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Ana Banito
- Pediatric Soft Tissue Sarcoma Research GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Stefan Burdach
- Department of Pediatrics and Children's Cancer Research Center (CCRC)Technische Universität MünchenMunichGermany
| | - Florencia Cidre‐Aranaz
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | - Gemma Di Pompo
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | | | | | | | | | | | - Merve Kasan
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | | | | | - Fernando Lecanda
- Division of OncologyAdhesion and Metastasis LaboratoryCenter for Applied Medical ResearchUniversity of NavarraPamplonaSpain
| | - Silvia Lemma
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Dario L Longo
- Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TurinItaly
| | | | | | - Julian Musa
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Shunya Ohmura
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | | | - Miguel Pereira‐Silva
- Department of Pharmaceutical TechnologyFaculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Francesca Perut
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Rene Rodriguez
- Instituto de Investigación Sanitaria del Principado de AsturiasOviedoSpain
- CIBER en oncología (CIBERONC)MadridSpain
| | | | - Nada Al Shaaili
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Shabnam Shaabani
- Department of Drug DesignUniversity of GroningenGroningenThe Netherlands
| | - Kristina Shiavone
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Snehadri Sinha
- Department of Oral and Maxillofacial DiseasesUniversity of HelsinkiHelsinkiFinland
| | | | - Marcel Trautmann
- Division of Translational PathologyGerhard‐Domagk‐Institute of PathologyMünster University HospitalMünsterGermany
| | - Maria Vela
- Hospital La Paz Institute for Health Research (IdiPAZ)MadridSpain
| | | | | | - Marta Zalacain
- Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TurinItaly
| | - Sebastian J Schober
- Department of Pediatrics and Children's Cancer Research Center (CCRC)Technische Universität MünchenMunichGermany
| | - Andrej Lissat
- University Children′s Hospital Zurich – Eleonoren FoundationKanton ZürichZürichSwitzerland
| | - William R English
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Nicola Baldini
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| | - Dominique Heymann
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
- Université de NantesInstitut de Cancérologie de l'OuestTumor Heterogeneity and Precision MedicineSaint‐HerblainFrance
| |
Collapse
|
22
|
Vermani L, Kumar R, Kannan RR, Deka MK, Talukdar A, Kumar NS. Expression pattern of ALDH1, E-cadherin, Vimentin and Twist in early and late onset sporadic colorectal cancer. Biomark Med 2020; 14:1371-1382. [PMID: 33064013 DOI: 10.2217/bmm-2020-0206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/30/2020] [Indexed: 12/21/2022] Open
Abstract
Aim: To evaluate the expression pattern of ALDH1 (aldehyde dehydrogenase 1), E-cadherin, Vimentin and Twist in early and late onset sporadic colorectal cancer (CRC) and to study association of their expression with the occurrence of CRC at a young age. Materials & methods: Immunohistochemistry of ALDH1, E-cadherin, Vimentin and Twist was performed on 103 pretreated CRC biopsy samples. Results: ALDH1 expression was found to have strong correlation with early onset CRC (p < 0.0001). Conclusion: High ALDH1 expression correlates with the early onset of CRC. ALDH1 over-expression correlates with poor overall survival in colon cancer.
Collapse
Affiliation(s)
- Litika Vermani
- Mizoram University, Biotechnology, Aizawl, Mizoram 796004, India
| | - Rajeev Kumar
- Cachar Cancer Hospital & Research Centre, Research Silchar, 788015, India
| | | | - Monoj K Deka
- Silchar Medical College & Hospital, 788015, India
| | - Anuradha Talukdar
- Cachar Cancer Hospital & Research Centre, Research Silchar, 788015, India
| | | |
Collapse
|
23
|
Althobiti M, El Ansari R, Aleskandarany M, Joseph C, Toss MS, Green AR, Rakha EA. The prognostic significance of ALDH1A1 expression in early invasive breast cancer. Histopathology 2020; 77:437-448. [PMID: 32369651 DOI: 10.1111/his.14129] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/28/2020] [Indexed: 12/29/2022]
Abstract
AIMS Aldehyde dehydrogenase family 1 member A1 (ALDH1A1) is reportedly a key ALDH isozyme linked to the cancer stem cells (CSC) of many solid tumours, where it is involved in self-renewal, differentiation and self-protection. In this study, the prognostic significance of ALDH1A1 expression in early invasive breast cancer (BC) and its role as a BC stem cell (BCSC) were evaluated. METHODS AND RESULTS ALDH1A1 expression was assessed, using immunohistochemistry and tissue microarrays, in a large well-characterised BC cohort. ALDH1A1 mRNA expression was also assessed at transcriptomic levels, utilising data from the Molecular Taxonomy of Breast Cancer International Consortium. The associations of ALDH1A1 with clinicopathological parameters, other stem cell markers and patient outcomes were determined. ALDH1A1 was expressed in 71% of BC cases at both the protein and mRNA levels. High ALDH1A1 expression was associated with poor prognostic features, including high grade, poor Nottingham Prognostic Index (NPI), lymph node metastasis and highly proliferative ER+ (luminal B) and triple-negative (TNBC) subtypes. ALDH1A1 expression was positively correlated with the expression of CD44, CD24, TWIST, SOX9, EPCAM and CD133. The high immunoexpression of ALDH1A1 was significantly associated with poor BC-specific survival (P < 0.001), and specifically in the luminal B and TNBC subtypes (P = 0.042 and P = 0.003, respectively). The immunoexpression of ALDH1A1 was an independent predictor of poor prognosis (P = 0.015). CONCLUSIONS ALDH1A1, as assessed using immunohistochemistry, seems to act as a BCSC marker associated not only with other BCSC markers but also with poor prognostic characteristics and poor outcomes, particularly in the luminal B and TNBC subtypes.
Collapse
Affiliation(s)
- Maryam Althobiti
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK.,The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK.,Department of Clinical Laboratory Science, College of Applied Medical Science, Shaqra, Saudi Arabia
| | - Rokaya El Ansari
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK.,The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Mohammed Aleskandarany
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK.,The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Chitra Joseph
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK.,The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Michael S Toss
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK.,The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK.,The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK.,The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| |
Collapse
|
24
|
Menendez ST, Rey V, Martinez-Cruzado L, Gonzalez MV, Morales-Molina A, Santos L, Blanco V, Alvarez C, Estupiñan O, Allonca E, Rodrigo JP, García-Castro J, Garcia-Pedrero JM, Rodriguez R. SOX2 Expression and Transcriptional Activity Identifies a Subpopulation of Cancer Stem Cells in Sarcoma with Prognostic Implications. Cancers (Basel) 2020; 12:cancers12040964. [PMID: 32295077 PMCID: PMC7226033 DOI: 10.3390/cancers12040964] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/07/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023] Open
Abstract
Stemness in sarcomas is coordinated by the expression of pluripotency factors, like SOX2, in cancer stem cells (CSC). The role of SOX2 in tumor initiation and progression has been well characterized in osteosarcoma. However, the pro-tumorigenic features of SOX2 have been scarcely investigated in other sarcoma subtypes. Here, we show that SOX2 depletion dramatically reduced the ability of undifferentiated pleomorphic sarcoma (UPS) cells to form tumorspheres and to initiate tumor growth. Conversely, SOX2 overexpression resulted in increased in vivo tumorigenicity. Moreover, using a reporter system (SORE6) which allows to monitor viable cells expressing SOX2 and/or OCT4, we found that SORE6+ cells were significantly more tumorigenic than the SORE6- subpopulation. In agreement with this findings, SOX2 expression in sarcoma patients was associated to tumor grade, differentiation, invasive potential and lower patient survival. Finally, we studied the effect of a panel of anti-tumor drugs on the SORE6+ cells of the UPS model and patient-derived chondrosarcoma lines. We found that the mithramycin analogue EC-8042 was the most efficient in reducing SORE6+ cells in vitro and in vivo. Overall, this study demonstrates that SOX2 is a pro-tumorigenic factor with prognostic potential in sarcoma. Moreover, SORE6 transcriptional activity is a bona fide CSC marker in sarcoma and constitutes an excellent biomarker for evaluating the efficacy of anti-tumor treatments on CSC subpopulations.
Collapse
Affiliation(s)
- Sofia T. Menendez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en oncología (CIBERONC), 28029 Madrid, Spain
| | - Veronica Rey
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Lucia Martinez-Cruzado
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - M. Victoria Gonzalez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en oncología (CIBERONC), 28029 Madrid, Spain
- Departamento de Cirugía, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Alvaro Morales-Molina
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | - Laura Santos
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Verónica Blanco
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Servicio de Anatomía Patológica of the Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Carlos Alvarez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Servicio de Oncología Médica of the Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Oscar Estupiñan
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en oncología (CIBERONC), 28029 Madrid, Spain
| | - Eva Allonca
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en oncología (CIBERONC), 28029 Madrid, Spain
| | - Juan Pablo Rodrigo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en oncología (CIBERONC), 28029 Madrid, Spain
| | - Javier García-Castro
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | - Juana Maria Garcia-Pedrero
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en oncología (CIBERONC), 28029 Madrid, Spain
| | - Rene Rodriguez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en oncología (CIBERONC), 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
25
|
Serial Xenotransplantation in NSG Mice Promotes a Hybrid Epithelial/Mesenchymal Gene Expression Signature and Stemness in Rhabdomyosarcoma Cells. Cancers (Basel) 2020; 12:cancers12010196. [PMID: 31941033 PMCID: PMC7016569 DOI: 10.3390/cancers12010196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/13/2019] [Accepted: 01/09/2020] [Indexed: 02/07/2023] Open
Abstract
Serial xenotransplantation of sorted cancer cells in immunodeficient mice remains the most complex test of cancer stem cell (CSC) phenotype. However, we have demonstrated in various sarcomas that putative CSC surface markers fail to identify CSCs, thereby impeding the isolation of CSCs for subsequent analyses. Here, we utilized serial xenotransplantation of unsorted rhabdomyosarcoma cells in NOD/SCID gamma (NSG) mice as a proof-of-principle platform to investigate the molecular signature of CSCs. Indeed, serial xenotransplantation steadily enriched for rhabdomyosarcoma stem-like cells characterized by enhanced aldehyde dehydrogenase activity and increased colony and sphere formation capacity in vitro. Although the expression of core pluripotency factors (SOX2, OCT4, NANOG) and common CSC markers (CD133, ABCG2, nestin) was maintained over the passages in mice, gene expression profiling revealed gradual changes in several stemness regulators and genes linked with undifferentiated myogenic precursors, e.g., SOX4, PAX3, MIR145, and CDH15. Moreover, we identified the induction of a hybrid epithelial/mesenchymal gene expression signature that was associated with the increase in CSC number. In total, 60 genes related to epithelial or mesenchymal traits were significantly altered upon serial xenotransplantation. In silico survival analysis based on the identified potential stemness-associated genes demonstrated that serial xenotransplantation of unsorted rhabdomyosarcoma cells in NSG mice might be a useful tool for the unbiased enrichment of CSCs and the identification of novel CSC-specific targets. Using this approach, we provide evidence for a recently proposed link between the hybrid epithelial/mesenchymal phenotype and cancer stemness.
Collapse
|
26
|
Bayat Mokhtari R, Baluch N, Morgatskaya E, Kumar S, Sparaneo A, Muscarella LA, Zhao S, Cheng HL, Das B, Yeger H. Human bronchial carcinoid tumor initiating cells are targeted by the combination of acetazolamide and sulforaphane. BMC Cancer 2019; 19:864. [PMID: 31470802 PMCID: PMC6716820 DOI: 10.1186/s12885-019-6018-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
Background Bronchial carcinoids are neuroendocrine tumors that present as typical (TC) and atypical (AC) variants, the latter being more aggressive, invasive and metastatic. Studies of tumor initiating cell (TIC) biology in bronchial carcinoids has been hindered by the lack of appropriate in-vitro and xenograft models representing the bronchial carcinoid phenotype and behavior. Methods Bronchial carcinoid cell lines (H727, TC and H720, AC) were cultured in serum-free growth factor supplemented medium to form 3D spheroids and serially passaged up to the 3rd generation permitting expansion of the TIC population as verified by expression of stemness markers, clonogenicity in-vitro and tumorigenicity in both subcutaneous and orthotopic (lung) models. Acetazolamide (AZ), sulforaphane (SFN) and the AZ + SFN combination were evaluated for targeting TIC in bronchial carcinoids. Results Data demonstrate that bronchial carcinoid cell line 3rd generation spheroid cells show increased drug resistance, clonogenicity, and tumorigenic potential compared with the parental cells, suggesting selection and expansion of a TIC fraction. Gene expression and immunolabeling studies demonstrated that the TIC expressed stemness factors Oct-4, Sox-2 and Nanog. In a lung orthotopic model bronchial carcinoid, cell line derived spheroids, and patient tumor derived 3rd generation spheroids when supported by a stroma, showed robust tumor formation. SFN and especially the AZ + SFN combination were effective in inhibiting tumor cell growth, spheroid formation and in reducing tumor formation in immunocompromised mice. Conclusions Human bronchial carcinoid tumor cells serially passaged as spheroids contain a higher fraction of TIC exhibiting a stemness phenotype. This TIC population can be effectively targeted by the combination of AZ + SFN. Our work portends clinical relevance and supports the therapeutic use of the novel AZ+ SFN combination that may target the TIC population of bronchial carcinoids.
Collapse
Affiliation(s)
- Reza Bayat Mokhtari
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada. .,Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay St., Rm 15.9714, Toronto, Ontario, M5G 0A4, Canada.
| | - Narges Baluch
- Department of Pediatrics, Queen's University, 76 Stuart St, Kingston, ON, K7L 2V7, Canada
| | - Evgeniya Morgatskaya
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sushil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Williams Science Hall 3035, Department of Pharmaceutical Sciences 601 S. Saddle Creek Rd, Omaha, NE, 68106, USA
| | - Angelo Sparaneo
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, viale Cappuccini, 71013, San Giovanni Rotondo, FG, Italy
| | - Lucia Anna Muscarella
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, viale Cappuccini, 71013, San Giovanni Rotondo, FG, Italy
| | - Sheyun Zhao
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hai-Ling Cheng
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, ON, M5S 3G9, Canada
| | - Bikul Das
- Thoreau Laboratory for Global Health, M2D2, University of Massachusetts-Lowell, Innovation Hub, 110 Canal St, Lowell, MA, 01852, USA.,KaviKrishna Laboratory, Indian Institute of Technology Complex, Guwahati, India
| | - Herman Yeger
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay St., Rm 15.9714, Toronto, Ontario, M5G 0A4, Canada
| |
Collapse
|
27
|
Hermida-Prado F, Villaronga MÁ, Granda-Díaz R, Del-Río-Ibisate N, Santos L, Hermosilla MA, Oro P, Allonca E, Agorreta J, Garmendia I, Tornín J, Perez-Escuredo J, Fuente R, Montuenga LM, Morís F, Rodrigo JP, Rodríguez R, García-Pedrero JM. The SRC Inhibitor Dasatinib Induces Stem Cell-Like Properties in Head and Neck Cancer Cells that are Effectively Counteracted by the Mithralog EC-8042. J Clin Med 2019; 8:jcm8081157. [PMID: 31382448 PMCID: PMC6722627 DOI: 10.3390/jcm8081157] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 12/19/2022] Open
Abstract
The frequent dysregulation of SRC family kinases (SFK) in multiple cancers prompted various inhibitors to be actively tested in preclinical and clinical trials. Disappointingly, dasatinib and saracatinib failed to demonstrate monotherapeutic efficacy in patients with head and neck squamous cell carcinomas (HNSCC). Deeper functional and mechanistic knowledge of the actions of these drugs is therefore needed to improve clinical outcome and to develop more efficient combinational strategies. Even though the SFK inhibitors dasatinib and saracatinib robustly blocked cell migration and invasion in HNSCC cell lines, this study unveils undesirable stem cell-promoting functions that could explain the lack of clinical efficacy in HNSCC patients. These deleterious effects were targeted by the mithramycin analog EC-8042 that efficiently eliminated cancer stem cells (CSC)-enriched tumorsphere cultures as well as tumor bulk cells and demonstrated potent antitumor activity in vivo. Furthermore, combination treatment of dasatinib with EC-8042 provided favorable complementary anti-proliferative, anti-invasive, and anti-CSC functions without any noticeable adverse interactions of both agents. These findings strongly support combinational strategies with EC-8042 for clinical testing in HNSCC patients. These data may have implications on ongoing dasatinib-based trials.
Collapse
Affiliation(s)
- Francisco Hermida-Prado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain
- Ciber de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - M Ángeles Villaronga
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain
- Ciber de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Rocío Granda-Díaz
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain
- Ciber de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Nagore Del-Río-Ibisate
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain
- Ciber de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Laura Santos
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain
| | | | - Patricia Oro
- EntreChem SL, Vivero Ciencias de la Salud, 33011 Oviedo, Spain
| | - Eva Allonca
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain
- Ciber de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Jackeline Agorreta
- Ciber de Cáncer, CIBERONC, 28029 Madrid, Spain
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology, Anatomy and Physiology, University of Navarra, and Navarra's Health Research Institute (IDISNA), 31008 Pamplona, Spain
| | - Irati Garmendia
- Ciber de Cáncer, CIBERONC, 28029 Madrid, Spain
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology, Anatomy and Physiology, University of Navarra, and Navarra's Health Research Institute (IDISNA), 31008 Pamplona, Spain
| | - Juan Tornín
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain
| | | | - Rocío Fuente
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | - Luis M Montuenga
- Ciber de Cáncer, CIBERONC, 28029 Madrid, Spain
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology, Anatomy and Physiology, University of Navarra, and Navarra's Health Research Institute (IDISNA), 31008 Pamplona, Spain
| | - Francisco Morís
- EntreChem SL, Vivero Ciencias de la Salud, 33011 Oviedo, Spain
| | - Juan P Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain
- Ciber de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - René Rodríguez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain.
- Ciber de Cáncer, CIBERONC, 28029 Madrid, Spain.
| | - Juana M García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain.
- Ciber de Cáncer, CIBERONC, 28029 Madrid, Spain.
| |
Collapse
|
28
|
TRAIL responses are enhanced by nuclear export inhibition in osteosarcoma. Biochem Biophys Res Commun 2019; 517:383-389. [PMID: 31362889 DOI: 10.1016/j.bbrc.2019.07.047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 07/17/2019] [Indexed: 01/10/2023]
Abstract
Tumour necrosis factor-related apoptosis inducing ligand (TRAIL) is a promising anti-tumour agent that induces apoptosis of malignant cells through activation of death receptors. Death receptor agonistic antibodies are in clinical trials as TRAIL-mimetics, however, along with TRAIL monotherapy, there is limited efficacy due to the rapid emergence of TRAIL resistance, or due to existing TRAIL-insensitive disease. TRAIL-sensitisers, which enhance TRAIL activity or overcome TRAIL resistance, may facilitate death receptor agonists as viable anti-tumour strategies. In this study we demonstrate that the nuclear export inhibitor Leptomycin B, is a potent in vitro TRAIL-sensitiser in osteosarcoma cell lines. Leptomycin B works synergistically with both TRAIL and death receptor 5 agonistic antibodies to induce apoptosis in TRAIL sensitive cell lines. Further, Leptomycin B sensitises TRAIL-insensitive cell lines to TRAIL and death receptor agonistic antibody mediated apoptosis. We also confirmed that aldehyde dehydrogenase (ALDH) positive cells are not resistant to the apoptotic effects of TRAIL and Leptomycin B, an important observation since ALDH positive cells can have enhanced tumorigenicity and are implicated in disease recurrence and metastasis. The nuclear export pathway in combination with death receptor agonists, is a potential therapeutic strategy in osteosarcoma and warrants further research on clinically relevant selective inhibitors of nuclear export.
Collapse
|
29
|
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer cells with self-renewal capacity, that fuel tumor growth and contribute to the heterogeneous nature of tumors. First identified in hematological malignancies, CSC populations have to date been proposed in solid tumors in various organs. In vitro and in vivo assays, mouse genetic models, and more recently single-cell sequencing technologies and other '-omics' methodologies have not only facilitated the identification of novel CSC populations but also revealed and clarified novel properties of CSCs. Increasingly, both cell-autonomous and CSC niche factors are recognized as important contributors of CSC properties. The deepened understanding of CSC properties and characteristics would enable and facilitate the rational design of CSC-specific therapeutics that would, ideally, have high selectivity for cancer cells, eliminate tumor bulk, and prevent tumor recurrence. Addressing these issues would form some of the key challenges of the CSC research field in the coming years.
Collapse
|
30
|
|
31
|
Rey V, Menendez ST, Estupiñan O, Rodriguez A, Santos L, Tornin J, Martinez-Cruzado L, Castillo D, Ordoñez GR, Costilla S, Alvarez-Fernandez C, Astudillo A, Braña A, Rodriguez R. New Chondrosarcoma Cell Lines with Preserved Stem Cell Properties to Study the Genomic Drift During In Vitro/In Vivo Growth. J Clin Med 2019; 8:jcm8040455. [PMID: 30987403 PMCID: PMC6518242 DOI: 10.3390/jcm8040455] [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: 03/04/2019] [Revised: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 12/11/2022] Open
Abstract
For the cancer genomics era, there is a need for clinically annotated close-to-patient cell lines suitable to investigate altered pathways and serve as high-throughput drug-screening platforms. This is particularly important for drug-resistant tumors like chondrosarcoma which has few models available. Here we established and characterized new cell lines derived from two secondary (CDS06 and CDS11) and one dedifferentiated (CDS-17) chondrosarcomas as well as another line derived from a CDS-17-generated xenograft (T-CDS17). These lines displayed cancer stem cell-related and invasive features and were able to initiate subcutaneous and/or orthotopic animal models. Different mutations in Isocitrate Dehydrogenase-1 (IDH1), Isocitrate Dehydrogenase-2 (IDH2), and Tumor Supressor P53 (TP53) and deletion of Cyclin Dependent Kinase Inhibitor 2A (CDKN2A) were detected both in cell lines and tumor samples. In addition, other mutations in TP53 and the amplification of Mouse Double Minute 2 homolog (MDM2) arose during cell culture in CDS17 cells. Whole exome sequencing analysis of CDS17, T-CDS17, and matched patient samples confirmed that cell lines kept the most relevant mutations of the tumor, uncovered new mutations and revealed structural variants that emerged during in vitro/in vivo growth. Altogether, this work expanded the panel of clinically and genetically-annotated chondrosarcoma lines amenable for in vivo studies and cancer stem cell (CSC) characterization. Moreover, it provided clues of the genetic drift of chondrosarcoma cells during the adaptation to grow conditions.
Collapse
Affiliation(s)
- Veronica Rey
- University Central Hospital of Asturias-Health and Research Institute of Asturias (ISPA), 33011 Oviedo, Spain.
- University Institute of Oncology of Asturias, 33011 Oviedo, Spain.
| | - Sofia T Menendez
- University Central Hospital of Asturias-Health and Research Institute of Asturias (ISPA), 33011 Oviedo, Spain.
- University Institute of Oncology of Asturias, 33011 Oviedo, Spain.
- CIBER in Oncology (CIBERONC), 28029 Madrid, Spain.
| | - Oscar Estupiñan
- University Central Hospital of Asturias-Health and Research Institute of Asturias (ISPA), 33011 Oviedo, Spain.
- University Institute of Oncology of Asturias, 33011 Oviedo, Spain.
- CIBER in Oncology (CIBERONC), 28029 Madrid, Spain.
| | - Aida Rodriguez
- University Central Hospital of Asturias-Health and Research Institute of Asturias (ISPA), 33011 Oviedo, Spain.
| | - Laura Santos
- University Central Hospital of Asturias-Health and Research Institute of Asturias (ISPA), 33011 Oviedo, Spain.
| | - Juan Tornin
- University Central Hospital of Asturias-Health and Research Institute of Asturias (ISPA), 33011 Oviedo, Spain.
| | - Lucia Martinez-Cruzado
- University Central Hospital of Asturias-Health and Research Institute of Asturias (ISPA), 33011 Oviedo, Spain.
| | - David Castillo
- Disease Research and Medicine (DREAMgenics) S.L., 33011 Oviedo, Spain.
| | - Gonzalo R Ordoñez
- Disease Research and Medicine (DREAMgenics) S.L., 33011 Oviedo, Spain.
| | - Serafin Costilla
- Department of Radiology of the Servicio de Radiología of the University Central Hospital of Asturias, 33011 Oviedo, Spain.
| | - Carlos Alvarez-Fernandez
- Department of Medical Oncology of the Servicio de Radiología of the University Central Hospital of Asturias, 33011 Oviedo, Spain.
| | - Aurora Astudillo
- Department of Pathology of the Servicio de Radiología of the University Central Hospital of Asturias, 33011 Oviedo, Spain.
| | - Alejandro Braña
- Department of Traumatology of the University Central Hospital of Asturias, 33011 Oviedo, Spain.
| | - Rene Rodriguez
- University Central Hospital of Asturias-Health and Research Institute of Asturias (ISPA), 33011 Oviedo, Spain.
- University Institute of Oncology of Asturias, 33011 Oviedo, Spain.
- CIBER in Oncology (CIBERONC), 28029 Madrid, Spain.
| |
Collapse
|
32
|
Estupiñan O, Santos L, Rodriguez A, Fernandez‐Nevado L, Costales P, Perez‐Escuredo J, Hermosilla MA, Oro P, Rey V, Tornin J, Allonca E, Fernandez‐Garcia MT, Alvarez‐Fernandez C, Braña A, Astudillo A, Menendez ST, Moris F, Rodriguez R. The multikinase inhibitor EC‐70124 synergistically increased the antitumor activity of doxorubicin in sarcomas. Int J Cancer 2019; 145:254-266. [DOI: 10.1002/ijc.32081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/22/2018] [Accepted: 12/05/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Oscar Estupiñan
- Hospital Universitario Central de Asturias ‐ Instituto de Investigación Sanitaria del Principado de Asturias Oviedo Spain
- Instituto Universitario de Oncología del Principado de Asturias Oviedo Spain
- CIBER en oncología (CIBERONC) Madrid Spain
| | - Laura Santos
- Hospital Universitario Central de Asturias ‐ Instituto de Investigación Sanitaria del Principado de Asturias Oviedo Spain
| | - Aida Rodriguez
- Hospital Universitario Central de Asturias ‐ Instituto de Investigación Sanitaria del Principado de Asturias Oviedo Spain
| | - Lucia Fernandez‐Nevado
- Hospital Universitario Central de Asturias ‐ Instituto de Investigación Sanitaria del Principado de Asturias Oviedo Spain
| | | | | | | | | | - Veronica Rey
- Hospital Universitario Central de Asturias ‐ Instituto de Investigación Sanitaria del Principado de Asturias Oviedo Spain
- Instituto Universitario de Oncología del Principado de Asturias Oviedo Spain
| | - Juan Tornin
- Hospital Universitario Central de Asturias ‐ Instituto de Investigación Sanitaria del Principado de Asturias Oviedo Spain
- Instituto Universitario de Oncología del Principado de Asturias Oviedo Spain
| | - Eva Allonca
- Hospital Universitario Central de Asturias ‐ Instituto de Investigación Sanitaria del Principado de Asturias Oviedo Spain
| | | | | | - Alejandro Braña
- Servicio de Traumatología of the Hospital Universitario Central de Asturias Oviedo Spain
| | - Aurora Astudillo
- Servicio de Anatomía Patológica of the Hospital Universitario Central de Asturias Oviedo Spain
| | - Sofia T Menendez
- Hospital Universitario Central de Asturias ‐ Instituto de Investigación Sanitaria del Principado de Asturias Oviedo Spain
- Instituto Universitario de Oncología del Principado de Asturias Oviedo Spain
- CIBER en oncología (CIBERONC) Madrid Spain
| | | | - Rene Rodriguez
- Hospital Universitario Central de Asturias ‐ Instituto de Investigación Sanitaria del Principado de Asturias Oviedo Spain
- Instituto Universitario de Oncología del Principado de Asturias Oviedo Spain
- CIBER en oncología (CIBERONC) Madrid Spain
| |
Collapse
|
33
|
Hermawan A, Putri H. Current report of natural product development against breast cancer stem cells. Int J Biochem Cell Biol 2018; 104:114-132. [DOI: 10.1016/j.biocel.2018.09.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 02/08/2023]
|
34
|
Cancer stem cells in sarcomas: Getting to the stemness core. Biochim Biophys Acta Gen Subj 2018; 1862:2134-2139. [DOI: 10.1016/j.bbagen.2018.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 12/16/2022]
|
35
|
Álvarez-Teijeiro S, García-Inclán C, Villaronga MÁ, Casado P, Hermida-Prado F, Granda-Díaz R, Rodrigo JP, Calvo F, Del-Río-Ibisate N, Gandarillas A, Morís F, Hermsen M, Cutillas P, García-Pedrero JM. Factors Secreted by Cancer-Associated Fibroblasts that Sustain Cancer Stem Properties in Head and Neck Squamous Carcinoma Cells as Potential Therapeutic Targets. Cancers (Basel) 2018; 10:cancers10090334. [PMID: 30227608 PMCID: PMC6162704 DOI: 10.3390/cancers10090334] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/09/2018] [Accepted: 09/12/2018] [Indexed: 12/15/2022] Open
Abstract
This study investigates for the first time the crosstalk between stromal fibroblasts and cancer stem cell (CSC) biology in head and neck squamous cell carcinomas (HNSCC), with the ultimate goal of identifying effective therapeutic targets. The effects of conditioned media from cancer-associated fibroblasts (CAFs) and normal fibroblasts (NFs) on the CSC phenotype were assessed by combining functional and expression analyses in HNSCC-derived cell lines. Further characterization of CAFs and NFs secretomes by mass spectrometry was followed by pharmacologic target inhibition. We demonstrate that factors secreted by CAFs but not NFs, in the absence of serum/supplements, robustly increased anchorage-independent growth, tumorsphere formation, and CSC-marker expression. Modulators of epidermal growth factor receptor (EGFR), insulin-like growth factor receptor (IGFR), and platelet-derived growth factor receptor (PDGFR) activity were identified as paracrine cytokines/factors differentially secreted between CAFs and NFs, in a mass spectrometry analysis. Furthermore, pharmacologic inhibition of EGFR, IGFR, and PDGFR significantly reduced CAF-induced tumorsphere formation and anchorage-independent growth suggesting a role of these receptor tyrosine kinases in sustaining the CSC phenotype. These findings provide novel insights into tumor stroma⁻CSC communication, and potential therapeutic targets to effectively block the CAF-enhanced CSC niche signaling circuit.
Collapse
Affiliation(s)
- Saúl Álvarez-Teijeiro
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain.
- CIBERONC, 28029 Madrid, Spain.
| | - Cristina García-Inclán
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain.
| | - M Ángeles Villaronga
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain.
- CIBERONC, 28029 Madrid, Spain.
| | - Pedro Casado
- Cell Signalling & Proteomics Group, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Francisco Hermida-Prado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain.
| | - Rocío Granda-Díaz
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain.
| | - Juan P Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain.
- CIBERONC, 28029 Madrid, Spain.
| | - Fernando Calvo
- Tumour Microenvironment Team, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Nagore Del-Río-Ibisate
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain.
| | - Alberto Gandarillas
- Cell Cycle, Stem Cell Fate and Cancer Lab Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain.
| | - Francisco Morís
- EntreChem SL, Vivero Ciencias de la Salud, 33011 Oviedo, Spain.
| | - Mario Hermsen
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain.
- CIBERONC, 28029 Madrid, Spain.
| | - Pedro Cutillas
- Cell Signalling & Proteomics Group, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Juana M García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias; Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011 Oviedo, Spain.
- CIBERONC, 28029 Madrid, Spain.
| |
Collapse
|
36
|
Golan H, Shukrun R, Caspi R, Vax E, Pode-Shakked N, Goldberg S, Pleniceanu O, Bar-Lev DD, Mark-Danieli M, Pri-Chen S, Jacob-Hirsch J, Kanter I, Trink A, Schiby G, Bilik R, Kalisky T, Harari-Steinberg O, Toren A, Dekel B. In Vivo Expansion of Cancer Stemness Affords Novel Cancer Stem Cell Targets: Malignant Rhabdoid Tumor as an Example. Stem Cell Reports 2018; 11:795-810. [PMID: 30122444 PMCID: PMC6135722 DOI: 10.1016/j.stemcr.2018.07.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/21/2018] [Accepted: 07/23/2018] [Indexed: 12/14/2022] Open
Abstract
Cancer stem cell (CSC) identification relies on transplantation assays of cell subpopulations sorted from fresh tumor samples. Here, we attempt to bypass limitations of abundant tumor source and predetermined immune selection by in vivo propagating patient-derived xenografts (PDX) from human malignant rhabdoid tumor (MRT), a rare and lethal pediatric neoplasm, to an advanced state in which most cells behave as CSCs. Stemness is then probed by comparative transcriptomics of serial PDXs generating a gene signature of epithelial to mesenchymal transition, invasion/motility, metastasis, and self-renewal, pinpointing putative MRT CSC markers. The relevance of these putative CSC molecules is analyzed by sorting tumorigenic fractions from early-passaged PDX according to one such molecule, deciphering expression in archived primary tumors, and testing the effects of CSC molecule inhibition on MRT growth. Using this platform, we identify ALDH1 and lysyl oxidase (LOX) as relevant targets and provide a larger framework for target and drug discovery in rare pediatric cancers. Human malignant rhabdoid tumor (MRT) can be propagated in vivo as tumor xenografts Long-term propagated PDX enrich for CSC frequency with no need for immune selection Distinct gene signature in stem-like MRT xenografts reveals putative CSC biomarkers Screening of putative CSC biomarkers allows identification of therapeutic targets
Collapse
Affiliation(s)
- Hana Golan
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan 52621, Israel; Division of Pediatric Hemato-oncology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan 52621, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Rachel Shukrun
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan 52621, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Revital Caspi
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan 52621, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Einav Vax
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan 52621, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Naomi Pode-Shakked
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan 52621, Israel; Dr. Pinchas Borenstein Talpiot Medical Leadership Program 2013, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sanja Goldberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel
| | - Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan 52621, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dekel D Bar-Lev
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel
| | - Michal Mark-Danieli
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Sara Pri-Chen
- The Maurice and Gabriela Goldschleger Eye Research Institute, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Jasmine Jacob-Hirsch
- Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Itamar Kanter
- Faculty of Engineering and Nanotechnology Institute, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Ariel Trink
- Faculty of Engineering and Nanotechnology Institute, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Ginette Schiby
- Department of Pathology, Sheba Medical Center, Ramat-Gan 52621, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ron Bilik
- Department of Pediatric Surgery, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Tomer Kalisky
- Faculty of Engineering and Nanotechnology Institute, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan 52621, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Amos Toren
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan 52621, Israel; Division of Pediatric Hemato-oncology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan 52621, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan 52621, Israel; Division of Pediatric Nephrology, Safra Children's Hospital, Sheba Medical Center, Ramat-Gan 52621, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel.
| |
Collapse
|
37
|
Tornin J, Hermida-Prado F, Padda RS, Gonzalez MV, Alvarez-Fernandez C, Rey V, Martinez-Cruzado L, Estupiñan O, Menendez ST, Fernandez-Nevado L, Astudillo A, Rodrigo JP, Lucien F, Kim Y, Leong HS, Garcia-Pedrero JM, Rodriguez R. FUS-CHOP Promotes Invasion in Myxoid Liposarcoma through a SRC/FAK/RHO/ROCK-Dependent Pathway. Neoplasia 2017; 20:44-56. [PMID: 29190494 PMCID: PMC5747526 DOI: 10.1016/j.neo.2017.11.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 01/13/2023] Open
Abstract
Deregulated SRC/FAK signaling leads to enhanced migration and invasion in many types of tumors. In myxoid and round cell liposarcoma (MRCLS), an adipocytic tumor characterized by the expression of the fusion oncogene FUS-CHOP, SRC have been found as one of the most activated kinases. Here we used a cell-of-origin model of MRCLS and an MRCLS cell line to thoroughly characterize the mechanisms of cell invasion induced by FUS-CHOP using in vitro (3D spheroid invasion assays) and in vivo (chicken chorioallantoic membrane model) approaches. FUS-CHOP expression activated SRC-FAK signaling and increased the invasive ability of MRCLS cells. In addition, FAK expression was found to significantly correlate with tumor aggressiveness in sarcoma patient samples. The involvement of SRC/FAK activation in FUS-CHOP-mediated invasion was further confirmed using the SRC inhibitor dasatinib, the specific FAK inhibitor PF-573228, and FAK siRNA. Notably, dasatinib and PF573228 could also efficiently block the invasion of cancer stem cell subpopulations. Downstream of SRC/FAK signaling, we found that FUS-CHOP expression increases the levels of the RHO/ROCK downstream effector phospho-MLC2 (T18/S19) and that this activation was prevented by dasatinib or PF573228. Moreover, the ROCK inhibitor RKI-1447 was able to completely abolish invasion in FUS-CHOP-expressing cells. These data uncover the involvement of SRC/FAK/RHO/ROCK signaling axis in FUS-CHOP-mediated invasion, thus providing a rationale for testing inhibitors of this pathway as potential novel antimetastatic agents for MRCLS treatment.
Collapse
Affiliation(s)
- Juan Tornin
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Francisco Hermida-Prado
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain
| | - Ranjit Singh Padda
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, ON, Canada
| | - M Victoria Gonzalez
- CIBER de Cáncer (CIBERONC), Madrid, Spain; Departamento de Cirugía, Universidad de Oviedo and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | | | - Veronica Rey
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Lucia Martinez-Cruzado
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Oscar Estupiñan
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Sofia T Menendez
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain
| | - Lucia Fernandez-Nevado
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Aurora Astudillo
- Servicio de Anatomía Patológica, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Juan P Rodrigo
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain
| | | | - Yohan Kim
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, ON, Canada; Department of Urology, Mayo Clinic, Rochester, MN
| | - Hon S Leong
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, ON, Canada; Department of Urology, Mayo Clinic, Rochester, MN
| | - Juana Maria Garcia-Pedrero
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain.
| | - Rene Rodriguez
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
38
|
Zhu ZW, Chen L, Liu JX, Huang JW, Wu G, Zheng YF, Yao KT. A novel three-dimensional tumorsphere culture system for the efficient and low-cost enrichment of cancer stem cells with natural polymers. Exp Ther Med 2017; 15:85-92. [PMID: 29387183 PMCID: PMC5769308 DOI: 10.3892/etm.2017.5419] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 08/01/2017] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) are considered to serve a key role in tumor progression, recurrence and metastasis. Tumorsphere culture is the most important method for enriching CSCs and is widely used in basic research and drug screening. However, the traditional suspension cell culture system has several disadvantages, including low efficiency, high cost and difficult procedure, making it difficult to produce tumorspheres on a large scale. In the present study, two biomaterials, methylcellulose (MC) and gellan gum (GG), were used to construct a novel culture system based on the traditional system. Subsequently, the characteristics of the novel three-dimensional (3D) culture system were evaluated, the design scheme was optimized, and the morphological and biological features of the tumorspheres cultured in this 3D system were compared with the traditional system. The results revealed that the tumorspheres cultured in the novel 3D system presented a higher seeding density and improved morphology, while maintaining stem-like properties. This evidence suggests that a simple, efficient and low-cost culture system that produces tumorspheres on a large scale was successfully constructed, which can be widely used in various aspects of stem cell research.
Collapse
Affiliation(s)
- Zhen-Wei Zhu
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Oncology, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 518000, P.R. China
| | - Lin Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jing-Xian Liu
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jun-Wen Huang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Gang Wu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Yan-Fang Zheng
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Kai-Tai Yao
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Oncology, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
39
|
Martinez-Cruzado L, Tornin J, Rodriguez A, Santos L, Allonca E, Fernandez-Garcia MT, Astudillo A, Garcia-Pedrero JM, Rodriguez R. Trabectedin and Campthotecin Synergistically Eliminate Cancer Stem Cells in Cell-of-Origin Sarcoma Models. Neoplasia 2017; 19:460-470. [PMID: 28494349 PMCID: PMC5421973 DOI: 10.1016/j.neo.2017.03.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/21/2017] [Accepted: 03/24/2017] [Indexed: 12/16/2022] Open
Abstract
Trabectedin has been approved for second-line treatment of soft tissue sarcomas. However, its efficacy to target sarcoma initiating cells has not been addressed yet. Here, we used pioneer models of myxoid/round cell liposarcoma (MRCLS) and undifferentiated pleomorphic sarcoma (UPS) developed from transformed human mesenchymal stromal/stem cells (MSCs) to evaluate the effect of trabectedin in the cell type responsible for initiating sarcomagenesis and their derived cancer stem cells (CSC) subpopulations. We found that low nanomolar concentrations of trabectedin efficiently inhibited the growth of sarcoma-initiating cells, induced cell cycle arrest, DNA damage and apoptosis. Interestingly, trabectedin treatment repressed the expression of multiple genes responsible for the development of the CSC phenotype, including pluripotency factors, CSC markers and related signaling pathways. Accordingly, trabectedin induced apoptosis and reduced the survival of CSC-enriched tumorsphere cultures with the same efficiency that inhibits the growth of bulk tumor population. In vivo, trabectedin significantly reduced the mitotic index of MRCLS xenografts and inhibited tumor growth at a similar extent to that observed in doxorubicin-treated tumors. Combination of trabectedin with campthotecin (CPT), a chemotherapeutic drug that shows a robust anti-tumor activity when combined with alkylating agents, resulted in a very strong synergistic inhibition of tumor cell growth and highly increased DNA damage and apoptosis induction. Importantly, the enhanced anti-tumor activity of this combination was also observed in CSC subpopulations. These data suggest that trabectedin and CPT combination may constitute a novel strategy to effectively target both the cell-of-origin and CSC subpopulations in sarcoma.
Collapse
Affiliation(s)
- Lucia Martinez-Cruzado
- Hospital Universitario Central de Asturias - Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Asturias; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Juan Tornin
- Hospital Universitario Central de Asturias - Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Asturias; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Aida Rodriguez
- Hospital Universitario Central de Asturias - Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Asturias
| | - Laura Santos
- Hospital Universitario Central de Asturias - Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Asturias
| | - Eva Allonca
- Hospital Universitario Central de Asturias - Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Asturias; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | | | - Aurora Astudillo
- Servicio de Anatomía Patológica, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Juana Maria Garcia-Pedrero
- Hospital Universitario Central de Asturias - Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Asturias; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER en oncología (CIBERONC), Madrid, Spain
| | - Rene Rodriguez
- Hospital Universitario Central de Asturias - Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Asturias; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER en oncología (CIBERONC), Madrid, Spain.
| |
Collapse
|