1
|
Martínez-Magaña CJ, Murbartián J. Estrogen receptor α regulates the IKKs/NF-kB activity involved in the development of mechanical allodynia induced by REM sleep deprivation in rats. Brain Res 2024; 1845:149269. [PMID: 39384127 DOI: 10.1016/j.brainres.2024.149269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/11/2024] [Accepted: 10/06/2024] [Indexed: 10/11/2024]
Abstract
Several signaling pathways that converge in NF-kB activation have been linked to developing and maintaining different types of pathological pain. In addition, some mechanisms implied in the establishment of chronic pain have been demonstrated to have a sex-dependent correlation. This study aimed to determine if the IKKs/NF-kB signaling pathway is involved in establishing REM sleep deprivation (REMSD) induced mechanical allodynia in rats and its possible regulation depending on estradiol and estrogen receptors. Intrathecal administration of BMS-345541 or minocycline, two drugs that reduce the IKKs/NF-kB activity, avoided the development of mechanical allodynia in female but not in male rats subjected to 48 h of REMSD. Ovariectomy in female rats abolished the effect of BMS-345541 and minocycline. Meanwhile, the 17-β-estradiol restitution restored it. Intrathecal administration of MPP, a selective ERα antagonist, but not PHTPP, a selective ERβ antagonist, avoided the effect of BMS-345541 in female rats without hormonal manipulation. In addition, the transient run-down of ERα in female rats abolished the effect of BMS-345541. All data suggest an important role of ERα as a regulator of the IKKs/NF-kB activity. REMSD increased the ERα protein expression in the dorsal root ganglia and the dorsal spinal cord in females but not in male rats. Interestingly, ERα activation or ERα overexpression allowed the effect of BMS-345541 in male rats. Data suggest an important regulatory role of ERα in the IKKs/NF-kB activity on establishing mechanical allodynia induced by REMSD in female rats.
Collapse
Affiliation(s)
| | - Janet Murbartián
- Pharmacobiology Department, Cinvestav, South Campus, Mexico City, Mexico.
| |
Collapse
|
2
|
Mokbel K. GD2 in Breast Cancer: A Potential Biomarker and Therapeutic Target. Cancer Genomics Proteomics 2024; 21:549-556. [PMID: 39467630 PMCID: PMC11534030 DOI: 10.21873/cgp.20471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/27/2024] [Accepted: 09/11/2024] [Indexed: 10/30/2024] Open
Abstract
Expression of disialoganglioside GD2 in normal tissues is primarily limited to the central nervous system, peripheral sensory nerve fibers, dermal melanocytes, lymphocytes, and mesenchymal stem cells. Its widespread overexpression in various cancer types allows it to be classified as a tumor-associated antigen with potential diagnostic and therapeutic implications. This article reviews the synthesis pathways of GD2 and its role in cancer cell adhesion, proliferation, and metastasis with a focus on breast cancer. GD2 appears to be overexpressed on the outer membrane of most breast cancer cells and breast cancer stem cells (BCSCs) and is closely linked to epithelial-mesenchymal transition (EMT). GD3 synthase (GD3S) is considered to be the rate-determining step in GD2 synthesis. Clinical studies indicate that GD2 expression is increased in 35-70% of breast cancer samples, with higher levels in triple-negative breast cancer (TNBC). This overexpression correlates with more aggressive tumor features and worse prognosis. Therapeutic targeting of GD2 with monoclonal antibodies (moABs) like dinutuximab and naxitamab has demonstrated anti-cancer activity in preclinical cancer models and human clinical trials against high-risk neuroblastoma reducing tumor growth and enhancing survival. GD2-specific chimeric antigen receptor (CAR) T-cell therapy and GD3S inhibition present other promising therapeutic strategies to improve clinical outcomes. Furthermore, GD2-targeted vaccines are currently being investigated in cancer therapy. This narrative review article underscores the critical role of GD2 in breast cancer pathogenesis and highlights the promising therapeutic opportunities it offers. It advocates for the initiation of clinical trials to further explore the potential of GD2-targeted treatment in combination with standard breast cancer therapies.
Collapse
Affiliation(s)
- Kefah Mokbel
- The London Breast Institute, Princess Grace Hospital, London, U.K.
| |
Collapse
|
3
|
Karampuri A, Jakkula BK, Perugu S. ResisenseNet hybrid neural network model for predicting drug sensitivity and repurposing in breast Cancer. Sci Rep 2024; 14:23949. [PMID: 39397003 PMCID: PMC11471817 DOI: 10.1038/s41598-024-71076-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/23/2024] [Indexed: 10/15/2024] Open
Abstract
Breast cancer remains a leading cause of mortality among women worldwide, with drug resistance driven by transcription factors and mutations posing significant challenges. To address this, we present ResisenseNet, a predictive model for drug sensitivity and resistance. ResisenseNet integrates transcription factor expression, genomic markers, drugs, and molecular descriptors, employing a hybrid architecture of 1D-CNN + LSTM and DNN to effectively learn long-range and temporal patterns from amino acid sequences and transcription factor data. The model demonstrated exceptional predictive accuracy, achieving a validation accuracy of 0.9794 and a loss value of 0.042. Comprehensive validation included comparisons with state-of-the-art models and ablation studies, confirming the robustness of the developed architecture. ResisenseNet has been applied to repurpose existing anticancer drugs across 14 different cancers, with a focus on breast cancer. Among the malignancies studied, drugs targeting Low-grade Glioma (LGG) and Lung Adenocarcinoma (LUAD) showed increased sensitivity to breast cancer as per ResisenseNet's assessment. Further evaluation of the predicted sensitive drugs revealed that 14 had no prior history of anticancer activity against breast cancer. These drugs target key signaling pathways involved in breast cancer, presenting novel therapeutic opportunities. ResisenseNet addresses drug resistance by filtering ineffective compounds and enhancing chemotherapy for breast cancer. In vitro studies on sensitive drugs provide valuable insights into breast cancer prognosis, contributing to improved treatment strategies.
Collapse
Affiliation(s)
- Anush Karampuri
- Department of Biotechnology, National Institute of Technology, Warangal, 500604, India
| | - Bharath Kumar Jakkula
- Department of Biotechnology, National Institute of Technology, Warangal, 500604, India
| | - Shyam Perugu
- Department of Biotechnology, National Institute of Technology, Warangal, 500604, India.
| |
Collapse
|
4
|
Bhat AM, Mohapatra BC, Luan H, Mushtaq I, Chakraborty S, Kumar S, Wu W, Nolan B, Dutta S, Storck MD, Schott M, Meza JL, Lele SM, Lin MF, Cook LM, Corey E, Morrissey C, Coulter DW, Rowley MJ, Natarajan A, Datta K, Band V, Band H. GD2 and its biosynthetic enzyme GD3 synthase promote tumorigenesis in prostate cancer by regulating cancer stem cell behavior. Sci Rep 2024; 14:13523. [PMID: 38866755 PMCID: PMC11169677 DOI: 10.1038/s41598-024-60052-3] [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: 05/23/2023] [Accepted: 04/18/2024] [Indexed: 06/14/2024] Open
Abstract
While better management of loco-regional prostate cancer (PC) has greatly improved survival, advanced PC remains a major cause of cancer deaths. Identification of novel targetable pathways that contribute to tumor progression in PC could open new therapeutic options. The di-ganglioside GD2 is a target of FDA-approved antibody therapies in neuroblastoma, but the role of GD2 in PC is unexplored. Here, we show that GD2 is expressed in a small subpopulation of PC cells in a subset of patients and a higher proportion of metastatic tumors. Variable levels of cell surface GD2 expression were seen on many PC cell lines, and the expression was highly upregulated by experimental induction of lineage progression or enzalutamide resistance in CRPC cell models. GD2high cell fraction was enriched upon growth of PC cells as tumorspheres and GD2high fraction was enriched in tumorsphere-forming ability. CRISPR-Cas9 knockout (KO) of the rate-limiting GD2 biosynthetic enzyme GD3 Synthase (GD3S) in GD2high CRPC cell models markedly impaired the in vitro oncogenic traits and growth as bone-implanted xenograft tumors and reduced the cancer stem cell and epithelial-mesenchymal transition marker expression. Our results support the potential role of GD3S and its product GD2 in promoting PC tumorigenesis by maintaining cancer stem cells and suggest the potential for GD2 targeting in advanced PC.
Collapse
Affiliation(s)
- Aaqib M Bhat
- Eppley Institute for Research in Cancer and Allied Diseases, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bhopal C Mohapatra
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Haitao Luan
- Eppley Institute for Research in Cancer and Allied Diseases, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Insha Mushtaq
- Eppley Institute for Research in Cancer and Allied Diseases, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
- Departments of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Incyte Corporation, Wilmington, DE, USA
| | - Sukanya Chakraborty
- Eppley Institute for Research in Cancer and Allied Diseases, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Siddhartha Kumar
- Eppley Institute for Research in Cancer and Allied Diseases, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Wangbin Wu
- Eppley Institute for Research in Cancer and Allied Diseases, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Ben Nolan
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Samikshan Dutta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Micah Schott
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jane L Meza
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Subodh M Lele
- Departments of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ming-Fong Lin
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Leah M Cook
- Departments of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Donald W Coulter
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
- Incyte Corporation, Wilmington, DE, USA
| | - M Jordan Rowley
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Vimla Band
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA.
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, 985805 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Departments of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
5
|
Bhat AM, Mohapatra BC, Luan H, Mushtaq I, Chakraborty S, Kumar S, Wu W, Nolan B, Dutta S, Stock MD, Schott M, Meza JL, Lele SM, Lin MF, Cook LM, Corey E, Morrissey C, Coulter DW, Rowley J, Natarajan A, Datta K, Band V, Band H. GD2 and its biosynthetic enzyme GD3 synthase promote tumorigenesis in prostate cancer by regulating cancer stem cell behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.18.533299. [PMID: 36993422 PMCID: PMC10055271 DOI: 10.1101/2023.03.18.533299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
While better management of loco-regional prostate cancer (PC) has greatly improved survival, advanced PC remains a major cause of cancer deaths. Identification of novel targetable pathways that contribute to tumor progression in PC could open new therapeutic options. The di-ganglioside GD2 is a target of FDA-approved antibody therapies in neuroblastoma, but the role of GD2 in PC is unexplored. Here, we show that GD2 is expressed in a small subpopulation of PC cells in a subset of patients and a higher proportion of metastatic tumors. Variable levels of cell surface GD2 expression were seen on many PC cell lines, and the expression was highly upregulated by experimental induction of lineage progression or enzalutamide resistance in CRPC cell models. GD2high cell fraction was enriched upon growth of PC cells as tumorspheres and GD2high fraction was enriched in tumorsphere-forming ability. CRISPR-Cas9 knockout (KO) of the rate-limiting GD2 biosynthetic enzyme GD3 Synthase (GD3S) in GD2high CRPC cell models markedly impaired the in vitro oncogenic traits and growth as bone-implanted xenograft tumors and reduced the cancer stem cell (CSC) and epithelial-mesenchymal transition (EMT) marker expression. Our results support the potential role of GD3S and its product GD2 in promoting PC tumorigenesis by maintaining cancer stem cells and suggest the potential for GD2 targeting in advanced PC.
Collapse
|
6
|
Philippova J, Shevchenko J, Sennikov S. GD2-targeting therapy: a comparative analysis of approaches and promising directions. Front Immunol 2024; 15:1371345. [PMID: 38558810 PMCID: PMC10979305 DOI: 10.3389/fimmu.2024.1371345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Disialoganglioside GD2 is a promising target for immunotherapy with expression primarily restricted to neuroectodermal and epithelial tumor cells. Although its role in the maintenance and repair of neural tissue is well-established, its functions during normal organism development remain understudied. Meanwhile, studies have shown that GD2 plays an important role in tumorigenesis. Its functions include proliferation, invasion, motility, and metastasis, and its high expression and ability to transform the tumor microenvironment may be associated with a malignant phenotype. Structurally, GD2 is a glycosphingolipid that is stably expressed on the surface of tumor cells, making it a suitable candidate for targeting by antibodies or chimeric antigen receptors. Based on mouse monoclonal antibodies, chimeric and humanized antibodies and their combinations with cytokines, toxins, drugs, radionuclides, nanoparticles as well as chimeric antigen receptor have been developed. Furthermore, vaccines and photoimmunotherapy are being used to treat GD2-positive tumors, and GD2 aptamers can be used for targeting. In the field of cell therapy, allogeneic immunocompetent cells are also being utilized to enhance GD2 therapy. Efforts are currently being made to optimize the chimeric antigen receptor by modifying its design or by transducing not only αβ T cells, but also γδ T cells, NK cells, NKT cells, and macrophages. In addition, immunotherapy can combine both diagnostic and therapeutic methods, allowing for early detection of disease and minimal residual disease. This review discusses each immunotherapy method and strategy, its advantages and disadvantages, and highlights future directions for GD2 therapy.
Collapse
Affiliation(s)
| | | | - Sergey Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| |
Collapse
|
7
|
Cao S, Hu X, Ren S, Wang Y, Shao Y, Wu K, Yang Z, Yang W, He G, Li X. The biological role and immunotherapy of gangliosides and GD3 synthase in cancers. Front Cell Dev Biol 2023; 11:1076862. [PMID: 36824365 PMCID: PMC9941352 DOI: 10.3389/fcell.2023.1076862] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Gangliosides are a large subfamily of glycosphingolipids that broadly exist in the nervous system and interact with signaling molecules in the lipid rafts. GD3 and GD2 are two types of disialogangliosides (GDs) that include two sialic acid residues. The expression of GD3 and GD2 in various cancers is mostly upregulated and is involved in tumor proliferation, invasion, metastasis, and immune responses. GD3 synthase (GD3S, ST8SiaI), a subclass of sialyltransferases, regulates the biosynthesis of GD3 and GD2. GD3S is also upregulated in most tumors and plays an important role in the development and progression of tumors. Many clinical trials targeting GD2 are ongoing and various immunotherapy studies targeting gangliosides and GD3S are gradually attracting much interest and attention. This review summarizes the function, molecular mechanisms, and ongoing clinical applications of GD3, GD2, and GD3S in abundant types of tumors, which aims to provide novel targets for future cancer therapy.
Collapse
Affiliation(s)
- Shangqi Cao
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xu Hu
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Shangqing Ren
- 2Robotic Minimally Invasive Surgery Center, Sichuan Academy of Medical Sciences and Sichuan Provincial Peoples Hospital, Chengdu, China
| | - Yaohui Wang
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yanxiang Shao
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Kan Wu
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Zhen Yang
- 3Department of Urology, Chengdu Second People’s Hospital, Chengdu, China
| | - Weixiao Yang
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Gu He
- 4State Key Laboratory of Biotherapy and Department of Pharmacy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China,*Correspondence: Gu He, ; Xiang Li,
| | - Xiang Li
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China,*Correspondence: Gu He, ; Xiang Li,
| |
Collapse
|
8
|
Troschke-Meurer S, Zumpe M, Meißner L, Siebert N, Grabarczyk P, Forkel H, Maletzki C, Bekeschus S, Lode HN. Chemotherapeutics Used for High-Risk Neuroblastoma Therapy Improve the Efficacy of Anti-GD2 Antibody Dinutuximab Beta in Preclinical Spheroid Models. Cancers (Basel) 2023; 15:cancers15030904. [PMID: 36765861 PMCID: PMC9913527 DOI: 10.3390/cancers15030904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Anti-disialoganglioside GD2 antibody ch14.18/CHO (dinutuximab beta, DB) improved the outcome of patients with high-risk neuroblastoma (HR-NB) in the maintenance phase. We investigated chemotherapeutic compounds used in newly diagnosed patients in combination with DB. Vincristine, etoposide, carboplatin, cisplatin, and cyclophosphamide, as well as DB, were used at concentrations achieved in pediatric clinical trials. The effects on stress ligand and checkpoint expression by neuroblastoma cells and on activation receptors of NK cells were determined by using flow cytometry. NK-cell activity was measured with a CD107a/IFN-γ assay. Long-term cytotoxicity was analyzed in three spheroid models derived from GD2-positive neuroblastoma cell lines (LAN-1, CHLA 20, and CHLA 136) expressing a fluorescent near-infrared protein. Chemotherapeutics combined with DB in the presence of immune cells improved cytotoxic efficacy up to 17-fold compared to in the controls, and the effect was GD2-specific. The activating stress and inhibitory checkpoint ligands on neuroblastoma cells were upregulated by the chemotherapeutics up to 9- and 5-fold, respectively, and activation receptors on NK cells were not affected. The CD107a/IFN-γ assay revealed no additional activation of NK cells by the chemotherapeutics. The synergistic effect of DB with chemotherapeutics seems primarily attributed to the combined toxicity of antibody-dependent cellular cytotoxicity and chemotherapy, which supports further clinical evaluation in frontline induction therapy.
Collapse
Affiliation(s)
- Sascha Troschke-Meurer
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Ferdinand-Sauerbruch Strasse 1, 17475 Greifswald, Germany
| | - Maxi Zumpe
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Ferdinand-Sauerbruch Strasse 1, 17475 Greifswald, Germany
| | - Lena Meißner
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Ferdinand-Sauerbruch Strasse 1, 17475 Greifswald, Germany
| | - Nikolai Siebert
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Ferdinand-Sauerbruch Strasse 1, 17475 Greifswald, Germany
| | - Piotr Grabarczyk
- Department of Internal Medicine, Clinic III—Hematology, Oncology, University Medicine Greifswald, Ferdinand-Sauerbruch Strasse 1, 17475 Greifswald, Germany
| | - Hannes Forkel
- Department of Internal Medicine, Clinic III—Hematology, Oncology, University Medicine Greifswald, Ferdinand-Sauerbruch Strasse 1, 17475 Greifswald, Germany
| | - Claudia Maletzki
- Department of Medicine, Clinic III—Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057 Rostock, Germany
| | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Holger N. Lode
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Ferdinand-Sauerbruch Strasse 1, 17475 Greifswald, Germany
- Correspondence: ; Tel.: +49-3834-86-6300; Fax: +49-3834-86-6410
| |
Collapse
|
9
|
Liu Z, Zhou J, Yang X, Liu Y, Zou C, Lv W, Chen C, Cheng KKY, Chen T, Chang LJ, Wu D, Mao J. Safety and antitumor activity of GD2-Specific 4SCAR-T cells in patients with glioblastoma. Mol Cancer 2023; 22:3. [PMID: 36617554 PMCID: PMC9827625 DOI: 10.1186/s12943-022-01711-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/29/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND This study aimed to validate whether infusion of GD2-specific fourth-generation safety-designed chimeric antigen receptor (4SCAR)-T cells is safe and whether CAR-T cells exert anti-glioblastoma (GBM) activity. METHODS A total of eight patients with GD2-positive GBM were enrolled and infused with autologous GD2-specific 4SCAR-T cells, either through intravenous administration alone or intravenous combined with intracavitary administration. RESULTS 4SCAR-T cells expanded for 1-3 weeks and persisted at a low frequency in peripheral blood. Of the eight evaluable patients, four showed a partial response for 3 to 24 months, three had progressive disease for 6 to 23 months, and one had stable disease for 4 months after infusion. For the entire cohort, the median overall survival was 10 months from the infusion. GD2 antigen loss and infiltrated T cells were observed in the tumor resected after infusion. CONCLUSION Both single and combined infusions of GD2-specific 4SCAR-T cells in targeting GBM were safe and well tolerated, with no severe adverse events. In addition, GD2-specific 4SCAR-T cells partially mediate antigen loss and activate immune responses in the tumor microenvironment. Validation of our findings in a larger prospective trial is warranted. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03170141 . Registered 30 May 2017.
Collapse
Affiliation(s)
- Zhuohao Liu
- grid.488521.2Department of Neurosurgery, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong China ,grid.284723.80000 0000 8877 7471Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong China ,grid.284723.80000 0000 8877 7471The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong China ,grid.440218.b0000 0004 1759 7210Present Address: Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong China
| | - Jiayi Zhou
- grid.284723.80000 0000 8877 7471Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong China ,grid.284723.80000 0000 8877 7471The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong China
| | - Xinzhi Yang
- grid.488521.2Department of Neurosurgery, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong China ,grid.284723.80000 0000 8877 7471The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong China ,grid.440218.b0000 0004 1759 7210Present Address: Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong China
| | - Yuchen Liu
- grid.489184.8Shenzhen Geno-Immune Medical Institute, Shenzhen, Guangdong China
| | - Chang Zou
- grid.10784.3a0000 0004 1937 0482School of Medicine, Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong China
| | - Wen Lv
- grid.440218.b0000 0004 1759 7210Present Address: Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong China
| | - Cheng Chen
- grid.440218.b0000 0004 1759 7210Present Address: Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong China
| | - Kenneth King-yip Cheng
- grid.16890.360000 0004 1764 6123Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Tao Chen
- grid.284723.80000 0000 8877 7471Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong China ,grid.284723.80000 0000 8877 7471The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong China
| | - Lung-Ji Chang
- grid.489184.8Shenzhen Geno-Immune Medical Institute, Shenzhen, Guangdong China
| | - Dinglan Wu
- grid.284723.80000 0000 8877 7471Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong China ,grid.284723.80000 0000 8877 7471The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong China
| | - Jie Mao
- grid.440218.b0000 0004 1759 7210Present Address: Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong China
| |
Collapse
|
10
|
Kondapuram SK, Coumar MS. Pan-cancer gene expression analysis: Identification of deregulated autophagy genes and drugs to target them. Gene X 2022; 844:146821. [PMID: 35985410 DOI: 10.1016/j.gene.2022.146821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/07/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022] Open
Abstract
Identifying suitable deregulated targets in autophagy pathway is essential for developing autophagy modulating cancer therapies. With this aim, we systematically analyzed the expression levels of genes that contribute to the execution of autophagy in 21 cancers. Deregulated genes for 21 cancers were analyzed using the level 3 mRNA data from TCGAbiolinks. A total of 574 autophagy genes were mapped to the deregulated genes across 21 cancers. PPI network, cluster analysis, gene enrichment, gene ontology, KEGG pathway, patient survival, protein expression and cMap analysis were performed. Among the autophagy genes, 260 were upregulated, and 43 were downregulated across pan-cancer. The upregulated autophagy genes - CDKN2A and BIRC5 - were the most frequent signatures in cancers and could be universal cancer biomarkers. Significant involvement of autophagy process was found in 8 cancers (CHOL, HNSC, GBM, KICH, KIRC, KIRP, LIHC and SARC). Fifteen autophagy hub genes (ATP6V0C, BIRC5, HDAC1, IL4, ITGB1, ITGB4, MAPK3, mTOR, cMYC, PTK2, SRC, TCIRG1, TP63, TP73 and ULK1) were found to be linked with patients survival and also expressed in cancer patients tissue samples, making them as potential drug targets for these cancers. The deregulated autophagy genes were further used to identify drugs Losartan, BMS-345541, Embelin, Abexinostat, Panobinostat, Vorinostat, PD-184352, PP-1, XMD-1150, Triplotide, Doxorubicin and Ouabain, which could target one or more autophagy hub genes. Overall, our findings shed light on the most frequent cancer-associated autophagy genes, potential autophagy targets and molecules for cancer treatment. These findings can accelerate autophagy modulation in cancer therapy.
Collapse
Affiliation(s)
- Sree Karani Kondapuram
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry- 605014, India
| | - Mohane Selvaraj Coumar
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry- 605014, India.
| |
Collapse
|
11
|
Di Francesco B, Verzella D, Capece D, Vecchiotti D, Di Vito Nolfi M, Flati I, Cornice J, Di Padova M, Angelucci A, Alesse E, Zazzeroni F. NF-κB: A Druggable Target in Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:3557. [PMID: 35884618 PMCID: PMC9319319 DOI: 10.3390/cancers14143557] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 02/01/2023] Open
Abstract
Acute Myeloid Leukemia (AML) is an aggressive hematological malignancy that relies on highly heterogeneous cytogenetic alterations. Although in the last few years new agents have been developed for AML treatment, the overall survival prospects for AML patients are still gloomy and new therapeutic options are still urgently needed. Constitutive NF-κB activation has been reported in around 40% of AML patients, where it sustains AML cell survival and chemoresistance. Given the central role of NF-κB in AML, targeting the NF-κB pathway represents an attractive strategy to treat AML. This review focuses on current knowledge of NF-κB's roles in AML pathogenesis and summarizes the main therapeutic approaches used to treat NF-κB-driven AML.
Collapse
|
12
|
Kasprowicz A, Sophie GD, Lagadec C, Delannoy P. Role of GD3 Synthase ST8Sia I in Cancers. Cancers (Basel) 2022; 14:cancers14051299. [PMID: 35267607 PMCID: PMC8909605 DOI: 10.3390/cancers14051299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 12/31/2022] Open
Abstract
Simple Summary The carbohydrate moiety of cell surface glycolipids is modified in cancers of neuro–ectoderm origin, leading to the expression of more complex structures with two or more sialic acid residues. These alterations result from the upregulation of the ST8SIA1 gene that encodes GD3 synthase, the enzyme controlling the biosynthesis of complex gangliosides, and are usually associated with a more aggressive phenotype and a poor outcome for patients, making GD3 synthase an interesting target for cancer therapy. This review reports our general knowledge of GD3 synthase gene expression and regulation, its role in both epithelial–mesenchymal transition (EMT) and cancer progression, and the different approaches targeting GD3S expression in cancers. Abstract GD3 synthase controls the biosynthesis of complex gangliosides, bearing two or more sialic acid residues. Disialylated gangliosides GD3 and GD2 are tumor-associated carbohydrate antigens (TACA) in neuro–ectoderm-derived cancers, and are directly involved in cell malignant properties, i.e., migration, invasion, stemness, and epithelial–mesenchymal transition. Since GD3 and GD2 levels are directly linked to GD3 synthase expression and activity, targeting GD3 synthase appears to be a promising strategy through which to interfere with ganglioside-associated malignant properties. We review here the current knowledge on GD3 synthase expression and regulation in cancers, and the consequences of complex ganglioside expression on cancer cell signaling and properties, highlighting the relationships between GD3 synthase expression and epithelial–mesenchymal transition and stemness. Different strategies were used to modulate GD3 synthase expression in cancer cells in vitro and in animal models, such as inhibitors or siRNA/lncRNA, which efficiently reduced cancer cell malignant properties and the proportion of GD2 positive cancer stem cells, which are associated with high metastatic properties, resistance to therapy, and cancer relapse. These data show the relevance of targeting GD3 synthase in association with conventional therapies, to decrease the number of cancer stem cells in tumors.
Collapse
Affiliation(s)
- Angelina Kasprowicz
- University of Lille, CNRS, UMR 8576-UGSF-Unité de Glycosylation Structurale et Fonctionnelle, F-59000 Lille, France;
| | - Groux-Degroote Sophie
- University of Lille, CNRS, UMR 8576-UGSF-Unité de Glycosylation Structurale et Fonctionnelle, F-59000 Lille, France;
- Correspondence: (G.-D.S.); (P.D.)
| | - Chann Lagadec
- University of Lille, CNRS, Inserm, CHU Lille UMR9020-U1277-CANTHER Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France;
| | - Philippe Delannoy
- University of Lille, CNRS, UMR 8576-UGSF-Unité de Glycosylation Structurale et Fonctionnelle, F-59000 Lille, France;
- Correspondence: (G.-D.S.); (P.D.)
| |
Collapse
|
13
|
Variable Expression of the Disialoganglioside GD2 in Breast Cancer Molecular Subtypes. Cancers (Basel) 2021; 13:cancers13215577. [PMID: 34771738 PMCID: PMC8582848 DOI: 10.3390/cancers13215577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/22/2021] [Accepted: 11/03/2021] [Indexed: 02/03/2023] Open
Abstract
Simple Summary GD2 is an antigen that is tumor-specific and can be used as a target for specific immunotherapies. Since the knowledge about GD2 in breast cancer is limited, we analyzed the frequency of GD2 expression in breast cancer using two different staining methods and the impact of GD2 expression on the survival of breast cancer patients. GD2 expression was found in more than 50% of breast cancer cases, with the highest frequency in hormone receptor-positive tumors. GD2 expression was not significantly associated with patient outcome. Unlike previous studies with smaller sample sizes that lacked correlation with clinical data, this study includes a larger cohort and associations with survival data and shows that GD2 is expressed on human breast cancer cells, providing a potential target for immunotherapies (e.g., anti-GD2 antibodies or GD2 CAR T cells), that are currently undergoing clinical testing. Abstract The disialoganglioside GD2 is a tumor-associated antigen that may allow for the application of targeted immunotherapies (anti-GD2 antibodies, GD2 CAR T cells) in patients with neuroblastoma and other solid tumors. We retrospectively investigated GD2 expression in a breast cancer cohort, using immunohistochemistry (IHC) and immunofluorescence (IF) on tissue microarrays (TMAs), and its impact on survival. GD2 expression on IHC (n = 568) and IF (n = 503) was investigated in relation to subtypes and patient outcome. Overall, 50.2% of the 568 IHC-assessed samples and 69.8% of the 503 IF-assessed samples were GD2-positive. The highest proportion of GD2-positive tumors was observed in luminal tumors. Significantly fewer GD2-positive cases were detected in triple-negative breast cancer (TNBC) compared with other subtypes. The proportion of GD2-expressing tumors were significantly lower in HER2-positive breast cancer in comparison with luminal tumors on IF staining (but not IHC). GD2 expression of IHC or IF was not significantly associated with disease-free or overall survival, in either the overall cohort or in individual subtypes. However, GD2 expression can be seen in more than 50% of breast cancer cases, with the highest frequency in hormone receptor-positive tumors. With this high expression frequency, patients with GD2-positive advanced breast cancer of all subtypes may benefit from GD2-targeting immunotherapies, which are currently subject to clinical testing.
Collapse
|
14
|
Shao C, Anand V, Andreeff M, Battula VL. Ganglioside GD2: a novel therapeutic target in triple-negative breast cancer. Ann N Y Acad Sci 2021; 1508:35-53. [PMID: 34596246 DOI: 10.1111/nyas.14700] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022]
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease characterized by lack of hormone receptor expression and is known for high rates of recurrence, distant metastases, and poor clinical outcomes. TNBC cells lack targetable receptors; hence, there is an urgent need for targetable markers for the disease. Breast cancer stem-like cells (BCSCs) are a fraction of cells in primary tumors that are associated with tumorigenesis, metastasis, and resistance to chemotherapy. Targeting BCSCs is thus an effective strategy for preventing cancer metastatic spread and sensitizing tumors to chemotherapy. The CD44hi CD24lo phenotype is a well-established phenotype for identification of BCSCs, but CD44 and CD24 are not targetable markers owing to their expression in normal tissues. The ganglioside GD2 has been shown to be upregulated in primary TNBC tumors compared with normal breast tissue and has been shown to identify BCSCs. In this review, we discuss GD2 as a BCSC- and tumor-specific marker in TNBC; epithelial-to-mesenchymal transition and the signaling pathways that are upstream and downstream of GD2 and the role of these pathways in tumorigenesis and metastasis in TNBC; direct and indirect approaches for targeting GD2; and ongoing clinical trials and treatments directed against GD2 as well as future directions for these strategies.
Collapse
Affiliation(s)
- Claire Shao
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivek Anand
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Venkata Lokesh Battula
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
15
|
Fasanya HO, Dopico PJ, Yeager Z, Fan ZH, Siemann DW. Using a combination of gangliosides and cell surface vimentin as surface biomarkers for isolating osteosarcoma cells in microfluidic devices. J Bone Oncol 2021; 28:100357. [PMID: 33912384 PMCID: PMC8065304 DOI: 10.1016/j.jbo.2021.100357] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/18/2021] [Accepted: 03/01/2021] [Indexed: 12/28/2022] Open
Abstract
Background Osteosarcoma (OS) is the most common primary bone tumor and the third leading cause of pediatric cancer deaths. Liquid biopsies are an alternative to current diagnostic imaging modalities that can be used to monitor treatment efficacy and the development of metastases. This study addresses the use of novel biomarkers to detect circulating osteosarcoma cells. Procedures Flow cytometry was used to evaluate the relative expression of epithelial cell adhesion molecule (EpCAM), ganglioside 2 and 3 (GD2/3), and cell surface vimentin (CSV) on a panel of OS cell lines. A microfluidic device was used to affirm the efficacy of GD2/3 and CSV to capture CTCs. Once captured, CTCs on the device are enumerated and the capture efficiency for each marker is measured. Patient samples were captured using the LFAM chip. Results We report the evaluation of GD2, GD3, and CSV as markers for OS cell capture in cell lines and in patient samples. The results of our capture studies correlate with our flow cytometry data and have shown a low capture efficiency of OS cells using EpCAM antibodies, while showing a moderate capture efficiency of OS cells using the GD2, GD3, and CSV antibodies independently. The combination of biomarkers demonstrate a high capture efficiency of approximately 80%. This is further supported by the detection of 1-1.5 CTCs per mL of blood using GD2 + CSV in OS patient samples. Conclusions The combination of GD2 + CSV significantly increased the capture efficacy of OS cells. The detection of CTCs through routine blood sampling may be used clinically for earlier detection of metastases and monitoring the therapeutic effect of treatments in metastatic osteosarcomas.
Collapse
Key Words
- CK, Cytokeratin
- CSV, Cell Surface Vimentin
- CTC, Circulating Tumor Cell
- Circulating tumor cells
- DAPI, 4′,6-diamidino-2-phenylindole
- EpCAM, Epithelial Cell Adhesion Molecule
- GD2, Ganglioside 2
- GD3, Ganglioside 3
- Ganglioside GD2
- Ganglioside GD3
- IHC, Immunohistochemistry
- OS, Osteosarcoma
- Osteosarcoma
- PET, Positron Emission Tomography
- Vimentin
- mL, Milliliter
Collapse
Affiliation(s)
- Henrietta O. Fasanya
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
- College of Medicine MD-PhD Program, University of Florida, Gainesville, FL, USA
- Corresponding authors at: Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL, USA (Z.H. Fan) Department of Radiation Oncology, University of Florida, Gainesville, FL, USA (H.O. Fasanya).
| | - Pablo J. Dopico
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL, USA
| | - Zachary Yeager
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL, USA
| | - Z. Hugh Fan
- J. Crayton Pruitt Family Department of Biomedical Engineering, Gainesville, FL, USA
- Corresponding authors at: Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL, USA (Z.H. Fan) Department of Radiation Oncology, University of Florida, Gainesville, FL, USA (H.O. Fasanya).
| | - Dietmar W. Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
- Corresponding authors at: Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL, USA (Z.H. Fan) Department of Radiation Oncology, University of Florida, Gainesville, FL, USA (H.O. Fasanya).
| |
Collapse
|
16
|
Zheng Q, Zhang M, Zhou F, Zhang L, Meng X. The Breast Cancer Stem Cells Traits and Drug Resistance. Front Pharmacol 2021; 11:599965. [PMID: 33584277 PMCID: PMC7876385 DOI: 10.3389/fphar.2020.599965] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
Drug resistance is a major challenge in breast cancer (BC) treatment at present. Accumulating studies indicate that breast cancer stem cells (BCSCs) are responsible for the BC drugs resistance, causing relapse and metastasis in BC patients. Thus, BCSCs elimination could reverse drug resistance and improve drug efficacy to benefit BC patients. Consequently, mastering the knowledge on the proliferation, resistance mechanisms, and separation of BCSCs in BC therapy is extremely helpful for BCSCs-targeted therapeutic strategies. Herein, we summarize the principal BCSCs surface markers and signaling pathways, and list the BCSCs-related drug resistance mechanisms in chemotherapy (CT), endocrine therapy (ET), and targeted therapy (TT), and display therapeutic strategies for targeting BCSCs to reverse drug resistance in BC. Even more importantly, more attention should be paid to studies on BCSC-targeted strategies to overcome the drug resistant dilemma of clinical therapies in the future.
Collapse
Affiliation(s)
- Qinghui Zheng
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Mengdi Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou, China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xuli Meng
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
17
|
Rodrigues FS, Miranda VS, Carneiro-Lobo TC, Scalabrini LC, Kruspig B, Levantini E, Murphy DJ, Bassères DS. IKKβ Kinase Promotes Stemness, Migration, and Invasion in KRAS-Driven Lung Adenocarcinoma Cells. Int J Mol Sci 2020; 21:E5806. [PMID: 32823550 PMCID: PMC7460870 DOI: 10.3390/ijms21165806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/25/2020] [Accepted: 07/31/2020] [Indexed: 12/11/2022] Open
Abstract
KRAS oncogenic mutations are widespread in lung cancer and, because direct targeting of KRAS has proven to be challenging, KRAS-driven cancers lack effective therapies. One alternative strategy for developing KRAS targeted therapies is to identify downstream targets involved in promoting important malignant features, such as the acquisition of a cancer stem-like and metastatic phenotype. Based on previous studies showing that KRAS activates nuclear factor kappa-B (NF-κB) through inhibitor of nuclear factor kappa-B kinase β (IKKβ) to promote lung tumourigenesis, we hypothesized that inhibition of IKKβ would reduce stemness, migration and invasion of KRAS-mutant human lung cancer cells. We show that KRAS-driven lung tumoursphere-derived cells exhibit stemness features and increased IKKβ kinase activity. IKKβ targeting by different approaches reduces the expression of stemness-associated genes, tumoursphere formation, and self-renewal, and preferentially impairs the proliferation of KRAS-driven lung tumoursphere-derived cells. Moreover, we show that IKKβ targeting reduces tumour cell migration and invasion, potentially by regulating both expression and activity of matrix metalloproteinase 2 (MMP2). In conclusion, our results indicate that IKKβ is an important mediator of KRAS-induced stemness and invasive features in lung cancer, and, therefore, might constitute a promising strategy to lower recurrence rates, reduce metastatic dissemination, and improve survival of lung cancer patients with KRAS-driven disease.
Collapse
Affiliation(s)
- Felipe Silva Rodrigues
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, 05508-000 São Paulo, Brazil; (F.S.R.); (V.S.M.); (T.C.C.-L.); (L.C.S.)
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (B.K.); (D.J.M.)
| | - Vanessa Silva Miranda
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, 05508-000 São Paulo, Brazil; (F.S.R.); (V.S.M.); (T.C.C.-L.); (L.C.S.)
| | - Tatiana Correa Carneiro-Lobo
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, 05508-000 São Paulo, Brazil; (F.S.R.); (V.S.M.); (T.C.C.-L.); (L.C.S.)
| | - Luiza Coimbra Scalabrini
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, 05508-000 São Paulo, Brazil; (F.S.R.); (V.S.M.); (T.C.C.-L.); (L.C.S.)
| | - Björn Kruspig
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (B.K.); (D.J.M.)
| | - Elena Levantini
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
- Istituto di Tecnologie Biomediche, Consiglio Nazionale dele Ricerche, 56124 Pisa, Italy
| | - Daniel J. Murphy
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (B.K.); (D.J.M.)
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Daniela Sanchez Bassères
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, 05508-000 São Paulo, Brazil; (F.S.R.); (V.S.M.); (T.C.C.-L.); (L.C.S.)
| |
Collapse
|
18
|
Reikvam H. Inhibition of NF-κB Signaling Alters Acute Myelogenous Leukemia Cell Transcriptomics. Cells 2020; 9:E1677. [PMID: 32664684 PMCID: PMC7408594 DOI: 10.3390/cells9071677] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
Acute myelogenous leukemia (AML) is an aggressive hematological malignancy. The pathophysiology of the disease depends on cytogenetic abnormalities, gene mutations, aberrant gene expressions, and altered epigenetic regulation. Although new pharmacological agents have emerged during the last years, the prognosis is still dismal and new therapeutic strategies are needed. The transcription factor nuclear factor-κB (NF-κB) is regarded a possible therapeutic target. In this study, we investigated the alterations in the global gene expression profile (GEP) in primary AML cells derived from 16 consecutive patients after exposure to the NF-κB inhibitor BMS-345541. We identified a profound and highly discriminative transcriptomic profile associated with NF-κB inhibition. Bioinformatical analyses identified cytokine/interleukin signaling, metabolic regulation, and nucleic acid binding/transcription among the major biological functions influenced by NF-κB inhibition. Furthermore, several key genes involved in leukemogenesis, among them RUNX1 and CEBPA, in addition to NFKB1 itself, were influenced by NF-κB inhibition. Finally, we identified a significant impact of NF-κB inhibition on the expression of genes included in a leukemic stem cell (LSC) signature, indicating possible targeting of LSCs. We conclude that NF-κB inhibition significantly altered the expression of genes central to the leukemic process.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Cell Line, Tumor
- Down-Regulation/genetics
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Gene Ontology
- Gene Regulatory Networks
- Genes, Neoplasm
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Male
- Middle Aged
- NF-kappa B/metabolism
- Signal Transduction
- Transcriptome/genetics
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Håkon Reikvam
- Institute of Clinical Science, University of Bergen, 5020 Bergen, Norway;
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| |
Collapse
|
19
|
Advances in molecular mechanisms of drugs affecting abnormal glycosylation and metastasis of breast cancer. Pharmacol Res 2020; 155:104738. [PMID: 32151681 DOI: 10.1016/j.phrs.2020.104738] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 12/27/2022]
Abstract
Breast cancer remains the leading cause of cancer-related death among women worldwide, and its incidence is also increasing. High recurrence rate and metastasis rate are the key causes of poor prognosis and death. It is suggested that abnormal glycosylation plays an important role in the growth, invasion, metastasis and resistance to therapy of breast cancer cells. Meanwhile, it can be used as the biomarkers for the early detection and prognosis of breast cancer and the potential attractive targets for drug treatment. However, only a few attentions have been paid to the molecular mechanism of abnormal glycosylation in the epithelial-mesenchymal transition (EMT) of breast cancer cells and the related intervention of drugs. This manuscript thus investigated the relationship between abnormal glycosylation, the EMT, and breast cancer metastasis. Then, the process of abnormal glycosylation, the classification and their molecular regulatory mechanisms of breast cancer were analyzed in detail. Last, potential drugs are introduced in different categories, which are expected to reverse or intervene the abnormal glycosylation of breast cancer. This review is conducive to an in-depth understanding of the metastasis and drug resistance of breast cancer cells, which will provide new ideas for the clinical regulation of glycosylation and related drug treatments in breast cancer.
Collapse
|
20
|
Zhu Q, Shen Y, Chen X, He J, Liu J, Zu X. Self-Renewal Signalling Pathway Inhibitors: Perspectives on Therapeutic Approaches for Cancer Stem Cells. Onco Targets Ther 2020; 13:525-540. [PMID: 32021295 PMCID: PMC6970631 DOI: 10.2147/ott.s224465] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/07/2019] [Indexed: 12/24/2022] Open
Abstract
The poor survival and prognosis of individuals with cancer are often attributed to tumour relapse and metastasis, which may be due to the presence of cancer stem cells (CSCs). CSCs have the characteristics of self-renewal, differentiation potential, high carcinogenicity, and drug resistance. In addition, CSCs exhibit many characteristics similar to those of embryonic or tissue stem cells while displaying persistent abnormal activation of self-renewal pathways associated with development and tissue homeostasis, including the Wnt, Notch, Hedgehog (Hh), TGF-β, JAK/STAT3, and NF-κB pathways. Therefore, we can eliminate CSCs by targeting these self-renewal pathways to constrain stem cell replication, survival and differentiation. At the same time, we cannot neglect the ping-pong effect of the tumour microenvironment, which releases cytokines and promotes self-renewal pathways in CSCs. Recently, meaningful progress has been made in the study of inhibitors of self-renewal pathways in tumours. This review primarily summarizes several representative and novel agents targeting these self-renewal signalling pathways and the tumour microenvironment and that represent a promising strategy for treating refractory and recurrent cancer.
Collapse
Affiliation(s)
- Qingyun Zhu
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yingying Shen
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Xiguang Chen
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Jun He
- Department of Spine Surgery, The Affiliated Nanhua Hospital of University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Jianghua Liu
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Xuyu Zu
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, People's Republic of China
| |
Collapse
|
21
|
Lian H, Han YP, Zhang YC, Zhao Y, Yan S, Li QF, Wang BC, Wang JJ, Meng W, Yang J, Wang QH, Mao WW, Ma J. Integrative analysis of gene expression and DNA methylation through one-class logistic regression machine learning identifies stemness features in medulloblastoma. Mol Oncol 2019; 13:2227-2245. [PMID: 31385424 PMCID: PMC6763787 DOI: 10.1002/1878-0261.12557] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/12/2019] [Accepted: 08/05/2019] [Indexed: 12/16/2022] Open
Abstract
Most human cancers develop from stem and progenitor cell populations through the sequential accumulation of various genetic and epigenetic alterations. Cancer stem cells have been identified from medulloblastoma (MB), but a comprehensive understanding of MB stemness, including the interactions between the tumor immune microenvironment and MB stemness, is lacking. Here, we employed a trained stemness index model based on an existent one‐class logistic regression (OCLR) machine‐learning method to score MB samples; we then obtained two stemness indices, a gene expression‐based stemness index (mRNAsi) and a DNA methylation‐based stemness index (mDNAsi), to perform an integrated analysis of MB stemness in a cohort of primary cancer samples (n = 763). We observed an inverse trend between mRNAsi and mDNAsi for MB subgroup and metastatic status. By applying the univariable Cox regression analysis, we found that mRNAsi significantly correlated with overall survival (OS) for all MB patients, whereas mDNAsi had no significant association with OS for all MB patients. In addition, by combining the Lasso‐penalized Cox regression machine‐learning approach with univariate and multivariate Cox regression analyses, we identified a stemness‐related gene expression signature that accurately predicted survival in patients with Sonic hedgehog (SHH) MB. Furthermore, positive correlations between mRNAsi and prognostic copy number aberrations in SHH MB, including MYCN amplifications and GLI2 amplifications, were detected. Analyses of the immune microenvironment revealed unanticipated correlations of MB stemness with infiltrating immune cells. Lastly, using the Connectivity Map, we identified potential drugs targeting the MB stemness signature. Our findings based on stemness indices might advance the development of objective diagnostic tools for quantitating MB stemness and lead to novel biomarkers that predict the survival of patients with MB or the efficacy of strategies targeting MB stem cells.
Collapse
Affiliation(s)
- Hao Lian
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Yi-Peng Han
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Yu-Chao Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science, Fudan University, Shanghai, China
| | - Yang Zhao
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Shan Yan
- Huamu Community Health Service Center, Shanghai, China
| | - Qi-Feng Li
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Bao-Cheng Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Jia-Jia Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Wei Meng
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Jian Yang
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Qin-Hua Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Wei-Wei Mao
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Jie Ma
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
22
|
Pasquel-Dávila DS, Yanez-Vaca SA, Espinosa-Hidalgo ND, Cuadros Buenaventura EG. Gangliosides generalities and role in cancer therapies. BIONATURA 2019. [DOI: 10.21931/rb/cs/2019.02.01.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Gangliosides are located in the plasma membrane; this confers them the ability to interact with other molecules in order to participate in important cellular processes. Some gangliosides presence or absence in the cell surface is associated with either normal condition or pathologies. Particularly in cancer, gangliosides play a critical role in pathological events like cellular malignancy, tumor formation, and metastasis, defining gangliosides as good candidates to be used as cellular markers. When specific gangliosides are exhibited, immunotherapy could be applied in order to inhibit tumorigenesis or induce an immunogenic response. Novel cancer treatments such as NGcGM3/VSSP vaccines, valproic acid, BMS-345541 inhibitor of GD2 and immunotherapies using 1E10 and 14F7 monoclonal antibodies are described. On this review, there will be studied the gangliosides that allowed developing biological techniques that can give immunogenicity to cancer cells
Collapse
Affiliation(s)
| | - Sabrina A. Yanez-Vaca
- School of Biological Science and Engineering, Yachay Tech University, Urcuquí – Ecuador
| | | | | |
Collapse
|
23
|
Intra-articular administration of IκBα kinase inhibitor suppresses mouse knee osteoarthritis via downregulation of the NF-κB/HIF-2α axis. Sci Rep 2018; 8:16475. [PMID: 30405206 PMCID: PMC6220282 DOI: 10.1038/s41598-018-34830-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 10/18/2018] [Indexed: 12/02/2022] Open
Abstract
Activation of NF-κB signaling promotes osteoarthritis (OA) through the transcriptional induction of Hif-2α and catabolic enzymes. This study sought to examine whether inhibiting IκBα kinase (IKK) could suppress the development of surgically-induced OA of the knee in a mouse model. We employed BMS-345541 (4(2′-aminoethyl) amino-1, 8-dimethylimidazo (1,2-a) quinoxaline) as a selective inhibitor of the subunits of IKK. OA was created by resecting the medial collateral ligament and the medial meniscus in the knees of mice. The mice were then treated with an intra-articular injection of BMS-345541 (50 nM to 500 µM) or vehicle three times a week for 8 weeks. We found that the intra-articular administration of 500 nM and 5 µM BMS-345541 significantly suppressed OA development. In the BMS-345541-treated cartilage, there was a decrease in the phosphorylation of IκBα and the expression of Hif-2α, Mmp13, and Adamts5. In human articular chondrocytes, the IL-1β-enhanced expression of Hif-2α and catabolic factors were decreased by BMS-345541 treatment in dose-dependent manner. We conclude that the intra-articular administration of BMS-345541 at some concentrations may suppress the development of OA by downregulating signaling through the NF-κB–Hif-2α axis.
Collapse
|
24
|
Nguyen K, Yan Y, Yuan B, Dasgupta A, Sun J, Mu H, Do KA, Ueno NT, Andreeff M, Battula VL. ST8SIA1 Regulates Tumor Growth and Metastasis in TNBC by Activating the FAK-AKT-mTOR Signaling Pathway. Mol Cancer Ther 2018; 17:2689-2701. [PMID: 30237308 DOI: 10.1158/1535-7163.mct-18-0399] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/23/2018] [Accepted: 09/05/2018] [Indexed: 01/16/2023]
Abstract
Breast cancer stem-like cells (BCSC) are implicated in cancer recurrence and metastasis of triple-negative breast cancer (TNBC). We have recently discovered that ganglioside GD2 expression defines BCSCs and that ST8SIA1 regulates GD2 expression and BCSC function. In this report, we show that ST8SIA1 is highly expressed in primary TNBC; its expression is positively correlated with the expression of several BCSC-associated genes such as BCL11A, FOXC1, CXCR4, PDGFRβ, SOX2, and mutations in p53. CRISPR knockout of ST8SIA1 completely inhibited BCSC functions, including in vitro tumorigenesis and mammosphere formation. Mechanistic studies discovered activation of the FAK-AKT-mTOR signaling pathway in GD2+ BCSCs, and its tight regulation by ST8SIA1. Finally, knockout of ST8SIA1 completely blocked in vivo tumor growth and metastasis by TNBC cells. In summary, these data demonstrate the mechanism by which ST8SIA1 regulates tumor growth and metastasis in TNBC and identifies it as a novel therapeutic target.
Collapse
Affiliation(s)
- Khoa Nguyen
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuanqing Yan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bin Yuan
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abhishek Dasgupta
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey Sun
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Mu
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - V Lokesh Battula
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
25
|
Yesilkanal AE, Rosner MR. Targeting Raf Kinase Inhibitory Protein Regulation and Function. Cancers (Basel) 2018; 10:cancers10090306. [PMID: 30181452 PMCID: PMC6162369 DOI: 10.3390/cancers10090306] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/24/2018] [Accepted: 08/30/2018] [Indexed: 12/22/2022] Open
Abstract
Raf Kinase Inhibitory Protein (RKIP) is a highly conserved kinase inhibitor that functions as a metastasis suppressor in a variety of cancers. Since RKIP can reprogram tumor cells to a non-metastatic state by rewiring kinase networks, elucidating the mechanism by which RKIP acts not only reveals molecular mechanisms that regulate metastasis, but also represents an opportunity to target these signaling networks therapeutically. Although RKIP is often lost during metastatic progression, the mechanism by which this occurs in tumor cells is complex and not well understood. In this review, we summarize our current understanding of RKIP regulation in tumors and consider experimental and computational strategies for recovering or mimicking its function by targeting mediators of metastasis.
Collapse
Affiliation(s)
- Ali Ekrem Yesilkanal
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA.
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA.
| | - Marsha Rich Rosner
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA.
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
26
|
Bai X, Ni J, Beretov J, Graham P, Li Y. Cancer stem cell in breast cancer therapeutic resistance. Cancer Treat Rev 2018; 69:152-163. [PMID: 30029203 DOI: 10.1016/j.ctrv.2018.07.004] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/11/2018] [Accepted: 07/12/2018] [Indexed: 12/15/2022]
Abstract
Development of therapeutic resistance and metastasis is a major challenge with current breast cancer (BC) therapy. Mounting evidence suggests that a subpopulation of cancer stem cells (CSCs) contribute to the cancer therapeutic resistance and metastasis, leading to the recurrence and death in patients. Breast cancer stem cells (BCSCs) are not only a consequence of mutations that overactivate the self-renewal ability of normal stem cells or committed progenitors but also a result of the de-differentiation of cancer cells induced by somatic mutations or microenvironmental components under treatment. Eradication of BCSCs may bring hope and relief to patients whose lives are threatened by recurrent BCs. Therefore, a better understanding of the generation, regulatory mechanisms, and identification of CSCs in BC therapeutic resistance and metastasis will be imperative for developing BCSC-targeted strategies. Here we summarize the latest studies about cell surface markers and signalling pathways that sustain the stemness of BCSC and discuss the associations of mechanisms behind these traits with phenotype and behavior changes in BCSCs. More importantly, their implications for future study are also evaluated and potential BCSC-targeted strategies are proposed to break through the limitation of current therapies.
Collapse
Affiliation(s)
- Xupeng Bai
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia; St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Jie Ni
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia; St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Julia Beretov
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia; St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia; Anatomical Pathology, NSW Health Pathology, St. George Hospital, Kogarah, NSW 2217, Australia
| | - Peter Graham
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia; St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Yong Li
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia; St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia; School of Basic Medical Sciences, Zhengzhou University, Henan 450001, China.
| |
Collapse
|
27
|
Nguyen K, Battula VL. Targeting NFκB signaling in GD2 + BCSCs. Aging (Albany NY) 2017; 9:1847-1848. [PMID: 28858852 PMCID: PMC5611972 DOI: 10.18632/aging.101274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 08/02/2017] [Indexed: 11/25/2022]
Affiliation(s)
- Khoa Nguyen
- Section of Molecular Hematology and Therapy, Leukemia Department, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - V Lokesh Battula
- Section of Molecular Hematology and Therapy, Leukemia Department, UT MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Breast Medical Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|