1
|
Wang S, Gu S, Chen J, Yuan Z, Liang P, Cui H. Mechanism of Notch Signaling Pathway in Malignant Progression of Glioblastoma and Targeted Therapy. Biomolecules 2024; 14:480. [PMID: 38672496 PMCID: PMC11048644 DOI: 10.3390/biom14040480] [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: 01/26/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of glioma and the most common primary tumor of the central nervous system. Despite significant advances in clinical management strategies and diagnostic techniques for GBM in recent years, it remains a fatal disease. The current standard of care includes surgery, radiation, and chemotherapy, but the five-year survival rate for patients is less than 5%. The search for a more precise diagnosis and earlier intervention remains a critical and urgent challenge in clinical practice. The Notch signaling pathway is a critical signaling system that has been extensively studied in the malignant progression of glioblastoma. This highly conserved signaling cascade is central to a variety of biological processes, including growth, proliferation, self-renewal, migration, apoptosis, and metabolism. In GBM, accumulating data suggest that the Notch signaling pathway is hyperactive and contributes to GBM initiation, progression, and treatment resistance. This review summarizes the biological functions and molecular mechanisms of the Notch signaling pathway in GBM, as well as some clinical advances targeting the Notch signaling pathway in cancer and glioblastoma, highlighting its potential as a focus for novel therapeutic strategies.
Collapse
Affiliation(s)
- Shenghao Wang
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China;
| | - Sikuan Gu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China; (S.G.); (J.C.); (Z.Y.)
| | - Junfan Chen
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China; (S.G.); (J.C.); (Z.Y.)
| | - Zhiqiang Yuan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China; (S.G.); (J.C.); (Z.Y.)
| | - Ping Liang
- Department of Neurosurgery, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Hongjuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China;
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China; (S.G.); (J.C.); (Z.Y.)
- Department of Neurosurgery, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| |
Collapse
|
2
|
Chen J, Cui L, Lu S, Xu S. Amino acid metabolism in tumor biology and therapy. Cell Death Dis 2024; 15:42. [PMID: 38218942 PMCID: PMC10787762 DOI: 10.1038/s41419-024-06435-w] [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: 09/27/2023] [Revised: 12/19/2023] [Accepted: 01/04/2024] [Indexed: 01/15/2024]
Abstract
Amino acid metabolism plays important roles in tumor biology and tumor therapy. Accumulating evidence has shown that amino acids contribute to tumorigenesis and tumor immunity by acting as nutrients, signaling molecules, and could also regulate gene transcription and epigenetic modification. Therefore, targeting amino acid metabolism will provide new ideas for tumor treatment and become an important therapeutic approach after surgery, radiotherapy, and chemotherapy. In this review, we systematically summarize the recent progress of amino acid metabolism in malignancy and their interaction with signal pathways as well as their effect on tumor microenvironment and epigenetic modification. Collectively, we also highlight the potential therapeutic application and future expectation.
Collapse
Affiliation(s)
- Jie Chen
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Likun Cui
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Shaoteng Lu
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Sheng Xu
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
| |
Collapse
|
3
|
Zhang Z, Huang Y, Li S, Hong L. Comprehensive analysis based on glycolytic and glutaminolytic pathways signature for predicting prognosis and immunotherapy in ovarian cancer. J Cancer 2024; 15:383-400. [PMID: 38169546 PMCID: PMC10758027 DOI: 10.7150/jca.88359] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 11/08/2023] [Indexed: 01/05/2024] Open
Abstract
Background: Our study attempts to develop and identify an aerobic glycolysis and glutamine-related genes (AGGRGs) signature for estimating prognostic effectively of ovarian cancer (OV) patients. Materials & methods: OV related data were extracted from the multiple public databases, including TCGA-OV, GSE26193, GSE63885, and ICGC-OV. A consistent clustering approach was used to characterize the subtypes associated with AGGRGs. LASSO Cox regressions was utilized to construct the prognosis signatures of AGGRGs. In addition, GSE26193, GSE63885 and ICGC-OV served as independent external cohorts to assess the reliability of the model. In vitro and in vivo experiments were conducted to study the role of AAK1 in the malignant progression and glutamine metabolism of OV, and assessed its therapeutic potential for treating OV patients. Results: OV patients could be separated into four subtypes (quiescent, glycolysis, glutaminolytic, and mixed subtypes). The survival outcome of glutaminolytic subtype was notably worse than the glycolytic subtype. Besides, we identified eight AGGRGs (AAK1, GJB6, HMGN5, LPIN3, INTS6L, PPOX, SPAG4, and ZNF316) to establish a prognostic signature for OV patients. Comprehensive analysis revealed that the signature risk score served as an independent prognostic factor for OV. Additionally, high-risk OV patients were less sensitive to platinum and, conversely, were proved to be more responsive to immunotherapy than low-risk score. In cytological experiments, we found that AAK1 could promote cancer progression and glutamine metabolism via activating the Notch3 pathway in OV cells. Furthermore, knockdown of AAK1 significantly inhibited tumor growth and weight, decreased lung metastases, and ultimately extended the survival time of the nude mice. Conclusions: The prognostic signature of AGGRGs constructed could efficiently estimate the prognosis and immunotherapy effectiveness of OV patients. In addition, AAK1 may represent a promising therapeutic target for OV.
Collapse
Affiliation(s)
- Zihui Zhang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Yuqin Huang
- Department of Gynecology and Obstetrics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, People's Republic of China
| | - Shuang Li
- Department of Gynecology and Obstetrics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, People's Republic of China
| | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| |
Collapse
|
4
|
Trejo-Solis C, Silva-Adaya D, Serrano-García N, Magaña-Maldonado R, Jimenez-Farfan D, Ferreira-Guerrero E, Cruz-Salgado A, Castillo-Rodriguez RA. Role of Glycolytic and Glutamine Metabolism Reprogramming on the Proliferation, Invasion, and Apoptosis Resistance through Modulation of Signaling Pathways in Glioblastoma. Int J Mol Sci 2023; 24:17633. [PMID: 38139462 PMCID: PMC10744281 DOI: 10.3390/ijms242417633] [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: 11/07/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Glioma cells exhibit genetic and metabolic alterations that affect the deregulation of several cellular signal transduction pathways, including those related to glucose metabolism. Moreover, oncogenic signaling pathways induce the expression of metabolic genes, increasing the metabolic enzyme activities and thus the critical biosynthetic pathways to generate nucleotides, amino acids, and fatty acids, which provide energy and metabolic intermediates that are essential to accomplish the biosynthetic needs of glioma cells. In this review, we aim to explore how dysregulated metabolic enzymes and their metabolites from primary metabolism pathways in glioblastoma (GBM) such as glycolysis and glutaminolysis modulate anabolic and catabolic metabolic pathways as well as pro-oncogenic signaling and contribute to the formation, survival, growth, and malignancy of glioma cells. Also, we discuss promising therapeutic strategies by targeting the key players in metabolic regulation. Therefore, the knowledge of metabolic reprogramming is necessary to fully understand the biology of malignant gliomas to improve patient survival significantly.
Collapse
Affiliation(s)
- Cristina Trejo-Solis
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Norma Serrano-García
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Roxana Magaña-Maldonado
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Dolores Jimenez-Farfan
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico;
| | - Elizabeth Ferreira-Guerrero
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (E.F.-G.); (A.C.-S.)
| | - Arturo Cruz-Salgado
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (E.F.-G.); (A.C.-S.)
| | | |
Collapse
|
5
|
Zarzuela L, Durán RV, Tomé M. Metabolism and signaling crosstalk in glioblastoma progression and therapy resistance. Mol Oncol 2023. [PMID: 38105543 DOI: 10.1002/1878-0261.13571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/09/2023] [Accepted: 12/15/2023] [Indexed: 12/19/2023] Open
Abstract
Glioblastoma is the most common form of primary malignant brain tumor in adults and one of the most lethal human cancers, with high recurrence and therapy resistance. Glioblastoma cells display extensive genetic and cellular heterogeneity, which precludes a unique and common therapeutic approach. The standard of care in glioblastoma patients includes surgery followed by radiotherapy plus concomitant temozolomide. As in many other cancers, cell signaling is deeply affected due to mutations or alterations in the so-called molecular drivers. Moreover, glioblastoma cells undergo metabolic adaptations to meet the new demands in terms of energy and building blocks, with an increasing amount of evidence connecting metabolic transformation and cell signaling deregulation in this type of aggressive brain tumor. In this review, we summarize some of the most common alterations both in cell signaling and metabolism in glioblastoma, presenting an integrative discussion about how they contribute to therapy resistance. Furthermore, this review aims at providing a comprehensive overview of the state-of-the-art of therapeutic approaches and clinical trials exploiting signaling and metabolism in glioblastoma.
Collapse
Affiliation(s)
- Laura Zarzuela
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain
| | - Raúl V Durán
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain
| | - Mercedes Tomé
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain
| |
Collapse
|
6
|
Glutamine Metabolism in Cancer Stem Cells: A Complex Liaison in the Tumor Microenvironment. Int J Mol Sci 2023; 24:ijms24032337. [PMID: 36768660 PMCID: PMC9916789 DOI: 10.3390/ijms24032337] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
In this review we focus on the role of glutamine in control of cancer stem cell (CSC) fate. We first provide an overview of glutamine metabolism, and then summarize relevant studies investigating how glutamine metabolism modulates the CSC compartment, concentrating on solid tumors. We schematically describe how glutamine in CSC contributes to several metabolic pathways, such as redox metabolic pathways, ATP production, non-essential aminoacids and nucleotides biosynthesis, and ammonia production. Furthermore, we show that glutamine metabolism is a key regulator of epigenetic modifications in CSC. Finally, we briefly discuss how cancer-associated fibroblasts, adipocytes, and senescent cells in the tumor microenvironment may indirectly influence CSC fate by modulating glutamine availability. We aim to highlight the complexity of glutamine's role in CSC, which supports our knowledge about metabolic heterogeneity within the CSC population.
Collapse
|
7
|
Abu Raya YS, Srebnik N, Rubinstein E, Schonberger O, Broza YY, Suschinel R, Haick H, Ionescu R. Noninvasive Pregestational Genetic Testing of Embryos Using Smart Sensors Array. ACS Sens 2022; 7:3265-3271. [PMID: 36374562 DOI: 10.1021/acssensors.2c01492] [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] [Indexed: 11/16/2022]
Abstract
Pregestational genetic testing of embryos is the conventional tool in detecting genetic disorders (fetal aneuploidy and monogenic disorders) for in vitro fertilization (IVF) procedures. The accepted clinical practice for genetic testing still depends on biopsy, which has the potential to harm the embryo. Noninvasive genetic prenatal testing has not yet been achieved. In this study, embryos with common genetic disorders created through IVF were tested with an artificially intelligent nanosensor array. Volatile organic compounds emitted by the culture fluid of embryos were analyzed with chemical gas sensors. The obtained results showed significant discrimination between the embryos with different genetic diseases and their wild-types. Embryos were obtained from the same clinical center for avoiding differences based on clinical and demographical characteristics. The achieved discrimination accuracy was 81% for PKD disease, 90% for FRAX disease, 85% for HOCM disease, 90% for BRCA disease, and 100% for HSCR disease. These proof-of-concept findings might launch the development of a noninvasive approach for early assessment of embryos by examining the culture fluid of the embryos, potentially enabling noninvasive diagnosis and screening of genetic diseases for IVF.
Collapse
Affiliation(s)
- Yasmin Shibli Abu Raya
- Department of Chemical Engineering and Russel Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, 3200003 Haifa, Israel
| | - Naama Srebnik
- In Vitro Fertilization Unit, Shaare Zedek Medical Center, Hebrew University Medical School, 9112102 Jerusalem, Israel
| | - Esther Rubinstein
- In Vitro Fertilization Unit, Shaare Zedek Medical Center, Hebrew University Medical School, 9112102 Jerusalem, Israel
| | - Oshrat Schonberger
- In Vitro Fertilization Unit, Shaare Zedek Medical Center, Hebrew University Medical School, 9112102 Jerusalem, Israel
| | - Yoav Y Broza
- Department of Chemical Engineering and Russel Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, 3200003 Haifa, Israel
| | - Raluca Suschinel
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51006 Tartu, Estonia
| | - Hossam Haick
- Department of Chemical Engineering and Russel Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, 3200003 Haifa, Israel
| | - Radu Ionescu
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51006 Tartu, Estonia
| |
Collapse
|
8
|
Suzuki S, Venkatesh D, Kanda H, Nakayama A, Hosokawa H, Lee E, Miki T, Stockwell BR, Yokote K, Tanaka T, Prives C. GLS2 Is a Tumor Suppressor and a Regulator of Ferroptosis in Hepatocellular Carcinoma. Cancer Res 2022; 82:3209-3222. [PMID: 35895807 PMCID: PMC11057045 DOI: 10.1158/0008-5472.can-21-3914] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 06/12/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022]
Abstract
Glutamine synthase 2 (GLS2) is a key regulator of glutaminolysis and has been previously implicated in activities consistent with tumor suppression. Here we generated Gls2 knockout (KO) mice that develop late-occurring B-cell lymphomas and hepatocellular carcinomas (HCC). Further, Gls2 KO mice subjected to the hepatocarcinogenic Stelic Animal Model (STAM) protocol produce larger HCC tumors than seen in wild-type (WT) mice. GLS2 has been shown to promote ferroptosis, a form of cell death characterized by iron-dependent accumulation of lipid peroxides. In line with this, GLS2 deficiency, either in cells derived from Gls2 KO mice or in human cancer cells depleted of GLS2, conferred significant resistance to ferroptosis. Mechanistically, GLS2, but not GLS1, increased lipid reactive oxygen species (ROS) production by facilitating the conversion of glutamate to α-ketoglutarate (αKG), thereby promoting ferroptosis. Ectopic expression of WT GLS2 in a human hepatic adenocarcinoma xenograft model significantly reduced tumor size; this effect was nullified by either expressing a catalytically inactive form of GLS2 or by blocking ferroptosis. Furthermore, analysis of cancer patient datasets supported a role for GLS2-mediated regulation of ferroptosis in human tumor suppression. These data suggest that GLS2 is a bona fide tumor suppressor and that its ability to favor ferroptosis by regulating glutaminolysis contributes to its tumor suppressive function. SIGNIFICANCE This study demonstrates that the key regulator of glutaminolysis, GLS2, can limit HCC in vivo by promoting ferroptosis through αKG-dependent lipid ROS, which in turn might lay the foundation for a novel therapeutic approach.
Collapse
Affiliation(s)
- Sawako Suzuki
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba, Japan
| | - Divya Venkatesh
- Department of Biological Sciences, Columbia University, New York, USA
| | - Hiroaki Kanda
- Department of Pathology, Saitama Cancer Center, Saitama, Japan
| | - Akitoshi Nakayama
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroyuki Hosokawa
- Department of Immunology, Tokai University School of Medicine, Kanagawa, Japan
| | - Eunyoung Lee
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takashi Miki
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Brent R. Stockwell
- Department of Biological Sciences, Columbia University, New York, USA
- Department of Chemistry, Columbia University, New York, USA
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba, Japan
| | - Tomoaki Tanaka
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, USA
| |
Collapse
|
9
|
Kubik J, Humeniuk E, Adamczuk G, Madej-Czerwonka B, Korga-Plewko A. Targeting Energy Metabolism in Cancer Treatment. Int J Mol Sci 2022; 23:ijms23105572. [PMID: 35628385 PMCID: PMC9146201 DOI: 10.3390/ijms23105572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer is the second most common cause of death worldwide after cardiovascular diseases. The development of molecular and biochemical techniques has expanded the knowledge of changes occurring in specific metabolic pathways of cancer cells. Increased aerobic glycolysis, the promotion of anaplerotic responses, and especially the dependence of cells on glutamine and fatty acid metabolism have become subjects of study. Despite many cancer treatment strategies, many patients with neoplastic diseases cannot be completely cured due to the development of resistance in cancer cells to currently used therapeutic approaches. It is now becoming a priority to develop new treatment strategies that are highly effective and have few side effects. In this review, we present the current knowledge of the enzymes involved in the different steps of glycolysis, the Krebs cycle, and the pentose phosphate pathway, and possible targeted therapies. The review also focuses on presenting the differences between cancer cells and normal cells in terms of metabolic phenotype. Knowledge of cancer cell metabolism is constantly evolving, and further research is needed to develop new strategies for anti-cancer therapies.
Collapse
Affiliation(s)
- Joanna Kubik
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
| | - Ewelina Humeniuk
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
- Correspondence: ; Tel.: +48-81-448-65-20
| | - Grzegorz Adamczuk
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
| | - Barbara Madej-Czerwonka
- Human Anatomy Department, Faculty of Medicine, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Agnieszka Korga-Plewko
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
| |
Collapse
|
10
|
Campos-Sandoval JA, Gómez-García MC, Santos-Jiménez JDL, Matés JM, Alonso FJ, Márquez J. Antioxidant responses related to temozolomide resistance in glioblastoma. Neurochem Int 2021; 149:105136. [PMID: 34274381 DOI: 10.1016/j.neuint.2021.105136] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/20/2021] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
Abstract
Glioblastoma remains one of the most challenging and devastating cancers, with only a very small proportion of patients achieving 5-year survival. The current standard of care consists of surgery, followed by radiation therapy with concurrent and maintenance chemotherapy with the alkylating agent temozolomide. To date, this drug is the only one that provides a significant survival benefit, albeit modest, as patients end up acquiring resistance to this drug. As a result, tumor progression and recurrence inevitably occur, leading to death. Several factors have been proposed to explain this resistance, including an upregulated antioxidant system to keep the elevated intracellular ROS levels, a hallmark of cancer cells, under control. In this review, we discuss the mechanisms of chemoresistance -including the important role of glioblastoma stem cells-with emphasis on antioxidant defenses and how agents that impair redox balance (i.e.: sulfasalazine, erastin, CB-839, withaferin, resveratrol, curcumin, chloroquine, and hydroxychloroquine) might be advantageous in combined therapies against this type of cancer.
Collapse
Affiliation(s)
- José A Campos-Sandoval
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab. Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain, and Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain.
| | - María C Gómez-García
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab. Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain, and Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Juan de Los Santos-Jiménez
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab. Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain, and Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - José M Matés
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab. Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain, and Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Francisco J Alonso
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab. Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain, and Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Javier Márquez
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab. Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain, and Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| |
Collapse
|
11
|
Jaroch K, Modrakowska P, Bojko B. Glioblastoma Metabolomics-In Vitro Studies. Metabolites 2021; 11:315. [PMID: 34068300 PMCID: PMC8153257 DOI: 10.3390/metabo11050315] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/20/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
In 2016, the WHO introduced new guidelines for the diagnosis of brain gliomas based on new genomic markers. The addition of these new markers to the pre-existing diagnostic methods provided a new level of precision for the diagnosis of glioma and the prediction of treatment effectiveness. Yet, despite this new classification tool, glioblastoma (GBM), a grade IV glioma, continues to have one of the highest mortality rates among central nervous system tumors. Metabolomics is a particularly promising tool for the analysis of GBM tumors and potential methods of treating them, as it is the only "omics" approach that is capable of providing a metabolic signature of a tumor's phenotype. With careful experimental design, cell cultures can be a useful matrix in GBM metabolomics, as they ensure stable conditions and, under proper conditions, are capable of capturing different tumor phenotypes. This paper reviews in vitro metabolomic profiling studies of high-grade gliomas, with a particular focus on sample-preparation techniques, crucial metabolites identified, cell culture conditions, in vitro-in vivo extrapolation, and pharmacometabolomics. Ultimately, this review aims to elucidate potential future directions for in vitro GBM metabolomics.
Collapse
Affiliation(s)
| | | | - Barbara Bojko
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, dr A. Jurasza 2 Street, 85-089 Bydgoszcz, Poland; (K.J.); (P.M.)
| |
Collapse
|
12
|
Zhou Z, Tian J, Zhang W, Xiang W, Ming Y, Chen L, Zhou J. Multiple strategies to improve the therapeutic efficacy of oncolytic herpes simplex virus in the treatment of glioblastoma. Oncol Lett 2021; 22:510. [PMID: 33986870 DOI: 10.3892/ol.2021.12771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/29/2021] [Indexed: 11/06/2022] Open
Abstract
Oncolytic viruses have attracted widespread attention as biological anticancer agents that can selectively kill tumor cells without affecting normal cells. Although progress has been made in therapeutic strategies, the prognosis of patients with glioblastoma (GBM) remains poor and no ideal treatment approach has been developed. Recently, oncolytic herpes simplex virus (oHSV) has been considered a promising novel treatment approach for GBM. However, the therapeutic efficacy of oHSV in GBM, with its intricate pathophysiology, remains unsatisfactory due to several obstacles, such as limited replication and attenuated potency of oHSV owing to deletions or mutations in virulence genes, and ineffective delivery of the therapeutic virus. Multiple strategies have attempted to identify the optimal strategy for the successful clinical application of oHSV. Several preclinical trials have demonstrated that engineering novel oHSVs, developing combination therapies and improving methods for delivering oHSV to tumor cells seem to hold promise for improving the efficacy of this virotherapy.
Collapse
Affiliation(s)
- Zhengjun Zhou
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Junjie Tian
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Wenyan Zhang
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Wei Xiang
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Yang Ming
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Ligang Chen
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan 646000, P.R. China.,Neurological Diseases and Brain Function Laboratory, Luzhou, Sichuan 646000, P.R. China
| | - Jie Zhou
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan 646000, P.R. China.,Neurological Diseases and Brain Function Laboratory, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
13
|
Giesen B, Nickel AC, Barthel J, Kahlert UD, Janiak C. Augmented Therapeutic Potential of Glutaminase Inhibitor CB839 in Glioblastoma Stem Cells Using Gold Nanoparticle Delivery. Pharmaceutics 2021; 13:295. [PMID: 33672398 PMCID: PMC7926460 DOI: 10.3390/pharmaceutics13020295] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Gold nanoparticles (Au NPs) are studied as delivery systems to enhance the effect of the glutaminase1 inhibitor CB839, a promising drug candidate already in clinical trials for tumor treatments. Au NPs were synthesized using a bottom-up approach and covered with polymers able to bind CB839 as a Au-polymer-CB839 conjugate. The drug loading efficiency (DLE) was determined using high-performance liquid chromatography and characterization of the CB839-loaded NPs was done with various microscopic and spectroscopic methods. Despite the chemical inertness of CB839, Au NPs were efficient carriers with a DLE of up to 12%, depending on the polymer used. The therapeutic effect of CB839 with and without Au was assessed in vitro in 2D and 3D glioblastoma (GBM) cell models using different assays based on the colony formation ability of GBM stem cells (GSCs). To avoid readout disturbances from the Au metal, viability methods which do not require optical detection were hereby optimized. These showed that Au NP delivery increased the efficacy of CB839 in GSCs, compared to CB839 alone. Fluorescent microscopy proved successful NP penetration into the GSCs. With this first attempt to combine CB839 with Au nanotechnology, we hope to overcome delivery hurdles of this pharmacotherapy and increase bioavailability in target sites.
Collapse
Affiliation(s)
- Beatriz Giesen
- Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine-Universität Düsseldorf, 40204 Düsseldorf, Germany;
| | - Ann-Christin Nickel
- Klinik für Neurochirurgie, Medizinische Fakultät, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany;
| | - Juri Barthel
- Ernst Ruska-Centrum für Mikroskopie und Spektroskopie mit Elektronen (ER-C 2), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany;
| | - Ulf Dietrich Kahlert
- Klinik für Neurochirurgie, Medizinische Fakultät, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany;
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), 40225 Düsseldorf, Germany
| | - Christoph Janiak
- Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine-Universität Düsseldorf, 40204 Düsseldorf, Germany;
| |
Collapse
|
14
|
Nguyen TL, Nokin MJ, Terés S, Tomé M, Bodineau C, Galmar O, Pasquet JM, Rousseau B, van Liempd S, Falcon-Perez JM, Richard E, Muzotte E, Rezvani HR, Priault M, Bouchecareilh M, Redonnet-Vernhet I, Calvo J, Uzan B, Pflumio F, Fuentes P, Toribio ML, Khatib AM, Soubeyran P, Murdoch PDS, Durán RV. Downregulation of Glutamine Synthetase, not glutaminolysis, is responsible for glutamine addiction in Notch1-driven acute lymphoblastic leukemia. Mol Oncol 2021; 15:1412-1431. [PMID: 33314742 PMCID: PMC8096784 DOI: 10.1002/1878-0261.12877] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/21/2020] [Accepted: 12/09/2020] [Indexed: 01/03/2023] Open
Abstract
The cellular receptor Notch1 is a central regulator of T-cell development, and as a consequence, Notch1 pathway appears upregulated in > 65% of the cases of T-cell acute lymphoblastic leukemia (T-ALL). However, strategies targeting Notch1 signaling render only modest results in the clinic due to treatment resistance and severe side effects. While many investigations reported the different aspects of tumor cell growth and leukemia progression controlled by Notch1, less is known regarding the modifications of cellular metabolism induced by Notch1 upregulation in T-ALL. Previously, glutaminolysis inhibition has been proposed to synergize with anti-Notch therapies in T-ALL models. In this work, we report that Notch1 upregulation in T-ALL induced a change in the metabolism of the important amino acid glutamine, preventing glutamine synthesis through the downregulation of glutamine synthetase (GS). Downregulation of GS was responsible for glutamine addiction in Notch1-driven T-ALL both in vitro and in vivo. Our results also confirmed an increase in glutaminolysis mediated by Notch1. Increased glutaminolysis resulted in the activation of the mammalian target of rapamycin complex 1 (mTORC1) pathway, a central controller of cell growth. However, glutaminolysis did not play any role in Notch1-induced glutamine addiction. Finally, the combined treatment targeting mTORC1 and limiting glutamine availability had a synergistic effect to induce apoptosis and to prevent Notch1-driven leukemia progression. Our results placed glutamine limitation and mTORC1 inhibition as a potential therapy against Notch1-driven leukemia.
Collapse
Affiliation(s)
- Tra Ly Nguyen
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | - Marie-Julie Nokin
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | - Silvia Terés
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | - Mercedes Tomé
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain.,Angiogenesis and Cancer Microenvironment Laboratory INSERM U1029, Université de Bordeaux, Pessac, France
| | - Clément Bodineau
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France.,Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain
| | - Oriane Galmar
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | | | - Benoit Rousseau
- Service Commun des Animaleries, University of Bordeaux, France
| | - Sebastian van Liempd
- Exosomes Laboratory and Platform of Metabolomics, CIC bioGUNE, CIBERehd, Derio, Spain
| | - Juan Manuel Falcon-Perez
- Exosomes Laboratory and Platform of Metabolomics, CIC bioGUNE, CIBERehd, Derio, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Elodie Richard
- Institut Bergonié, INSERM U1218, University of Bordeaux, France
| | | | | | - Muriel Priault
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, France
| | - Marion Bouchecareilh
- Bordeaux Research in Translational Oncology, INSERM U1053, Université de Bordeaux, France
| | - Isabelle Redonnet-Vernhet
- Maladies Héréditaires du Métabolisme, Laboratoire de Biochimie, Hôpital Pellegrin, CHU Bordeaux, France
| | - Julien Calvo
- UMR967, Inserm, CEA, Université Paris 7, Université Paris 11, Fontenay-aux-Roses, France
| | - Benjamin Uzan
- UMR967, Inserm, CEA, Université Paris 7, Université Paris 11, Fontenay-aux-Roses, France
| | - Françoise Pflumio
- UMR967, Inserm, CEA, Université Paris 7, Université Paris 11, Fontenay-aux-Roses, France
| | - Patricia Fuentes
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Spain
| | - Maria L Toribio
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Spain
| | - Abdel-Majid Khatib
- Angiogenesis and Cancer Microenvironment Laboratory INSERM U1029, Université de Bordeaux, Pessac, France
| | | | - Piedad Del Socorro Murdoch
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain.,Departamento de Bioquímica Vegetal y Biología Molecular, Universidad de Sevilla, Spain
| | - Raúl V Durán
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France.,Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain
| |
Collapse
|
15
|
Rapalink-1 Targets Glioblastoma Stem Cells and Acts Synergistically with Tumor Treating Fields to Reduce Resistance against Temozolomide. Cancers (Basel) 2020; 12:cancers12123859. [PMID: 33371210 PMCID: PMC7766508 DOI: 10.3390/cancers12123859] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Glioblastoma (GBM) resistance to standard treatment is driven by stem-like cell behavior and epithelial-like-mesenchymal transition. The main purpose of this paper was to functionally validate a novel discovered pharmacological strategy to treat GBM, the dual mTOR pathway inhibitor Rapalink-1 (RL1) using relevant stem cell models of the disease to unravel mechanistic insights. Our approach also interrogates combination studies with clinical treatment options of tumor treating fields (TTFields) and the best standard of care chemotherapy, temozolomide (TMZ). We provided clinical relevance of our experimental work through in silico evaluation on molecular data of diverse patient samples. RL1 effectively impaired motility and clonogenicity of GBM stem cells and reduced the expression of stem cell molecules. We elucidated a synergistic therapeutic potential of the inhibitor with TTFields to minimize therapy resistance toward TMZ, which supports its consideration for further translational oriented studies. Abstract Glioblastoma (GBM) is a lethal disease with limited clinical treatment options available. Recently, a new inhibitor targeting the prominent cancer signaling pathway mTOR was discovered (Rapalink-1), but its therapeutic potential on stem cell populations of GBM is unknown. We applied a collection of physiological relevant organoid-like stem cell models of GBM and studied the effect of RL1 exposure on various cellular features as well as on the expression of mTOR signaling targets and stem cell molecules. We also undertook combination treatments with this agent and clinical GBM treatments tumor treating fields (TTFields) and the standard-of-care drug temozolomide, TMZ. Low nanomolar (nM) RL1 treatment significantly reduced cell growth, proliferation, migration, and clonogenic potential of our stem cell models. It acted synergistically to reduce cell growth when applied in combination with TMZ and TTFields. We performed an in silico analysis from the molecular data of diverse patient samples to probe for a relationship between the expression of mTOR genes, and mesenchymal markers in different GBM cohorts. We supported the in silico results with correlative protein data retrieved from tumor specimens. Our study further validates mTOR signaling as a druggable target in GBM and supports RL1, representing a promising therapeutic target in brain oncology.
Collapse
|
16
|
Different Calculation Strategies Are Congruent in Determining Chemotherapy Resistance of Brain Tumors In Vitro. Cells 2020; 9:cells9122689. [PMID: 33333810 PMCID: PMC7765228 DOI: 10.3390/cells9122689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 01/14/2023] Open
Abstract
In cancer pharmacology, a drug candidate’s therapeutic potential is typically expressed as its ability to suppress cell growth. Different methods in assessing the cell phenotype and calculating the drug effect have been established. However, inconsistencies in drug response outcomes have been reported, and it is still unclear whether and to what extent the choice of data post-processing methods is responsible for that. Studies that systematically examine these questions are rare. Here, we compare three established calculation methods on a collection of nine in vitro models of glioblastoma, exposed to a library of 231 clinical drugs. The therapeutic potential of the drugs is determined on the growth curves, using growth inhibition 50% (GI50) and point-of-departure (PoD) as the criteria. An effect is detected on 36% of the drugs when relying on GI50 and on 27% when using PoD. For the area under the curve (AUC), a threshold of 9.5 or 10 could be set to discriminate between the drugs with and without an effect. GI50, PoD, and AUC are highly correlated. The ranking of substances by different criteria varies somewhat, but the group of the top 20 substances according to one criterion typically includes 17–19 top candidates according to another. In addition to generating preclinical values with high clinical potential, we present off-target appreciation of top substance predictions by interrogating the drug response data of non-cancer cells in our calculation technology.
Collapse
|
17
|
Restall IJ, Cseh O, Richards LM, Pugh TJ, Luchman HA, Weiss S. Brain Tumor Stem Cell Dependence on Glutaminase Reveals a Metabolic Vulnerability through the Amino Acid Deprivation Response Pathway. Cancer Res 2020; 80:5478-5490. [PMID: 33106333 DOI: 10.1158/0008-5472.can-19-3923] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/24/2020] [Accepted: 10/21/2020] [Indexed: 11/16/2022]
Abstract
Cancer cells can metabolize glutamine to replenish TCA cycle intermediates, leading to a dependence on glutaminolysis for cell survival. However, a mechanistic understanding of the role that glutamine metabolism has on the survival of glioblastoma (GBM) brain tumor stem cells (BTSC) has not yet been elucidated. Here, we report that across a panel of 19 GBM BTSC lines, inhibition of glutaminase (GLS) showed a variable response from complete blockade of cell growth to absolute resistance. Surprisingly, BTSC sensitivity to GLS inhibition was a result of reduced intracellular glutamate triggering the amino acid deprivation response (AADR) and not due to the contribution of glutaminolysis to the TCA cycle. Moreover, BTSC sensitivity to GLS inhibition negatively correlated with expression of the astrocytic glutamate transporters EAAT1 and EAAT2. Blocking glutamate transport in BTSCs with high EAAT1/EAAT2 expression rendered cells susceptible to GLS inhibition, triggering the AADR and limiting cell growth. These findings uncover a unique metabolic vulnerability in BTSCs and support the therapeutic targeting of upstream activators and downstream effectors of the AADR pathway in GBM. Moreover, they demonstrate that gene expression patterns reflecting the cellular hierarchy of the tissue of origin can alter the metabolic requirements of the cancer stem cell population. SIGNIFICANCE: Glioblastoma brain tumor stem cells with low astrocytic glutamate transporter expression are dependent on GLS to maintain intracellular glutamate to prevent the amino acid deprivation response and cell death.
Collapse
Affiliation(s)
- Ian J Restall
- Hotchkiss Brain Institute, Arnie Charbonneau Cancer Institute, and Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - Orsolya Cseh
- Hotchkiss Brain Institute, Arnie Charbonneau Cancer Institute, and Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - Laura M Richards
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Trevor J Pugh
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - H Artee Luchman
- Hotchkiss Brain Institute, Arnie Charbonneau Cancer Institute, and Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada.
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - Samuel Weiss
- Hotchkiss Brain Institute, Arnie Charbonneau Cancer Institute, and Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada.
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
18
|
A computational guided, functional validation of a novel therapeutic antibody proposes Notch signaling as a clinical relevant and druggable target in glioma. Sci Rep 2020; 10:16218. [PMID: 33004830 PMCID: PMC7531005 DOI: 10.1038/s41598-020-72480-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022] Open
Abstract
The Notch signaling network determines stemness in various tissues and targeting signaling activity in malignant brain cancers by gamma-secretase inhibitors (GSI) has shown promising preclinical success. However, the clinical translation remains challenging due to severe toxicity side effects and emergence of therapy resistance. Better anti-Notch directed therapies, specifically directed against the tumor promoting Notch receptor 1 signaling framework, and biomarkers predicting response to such therapy are of highest clinical need. We assessed multiple patient datasets to probe the clinical relevance Notch1 activation and possible differential distribution amongst molecular subtypes in brain cancers. We functionally assessed the biological effects of the first-in-human tested blocking antibody against Notch1 receptor (brontictuzumab, BRON) in a collection of glioma stem-like cell (GSC) models and compared its effects to genetic Notch1 inhibition as well as classical pharmacological Notch inhibitor treatment using gamma-secretase inhibitor MRK003. We also assess effects on Wingless (WNT) stem cell signaling activation, which includes the interrogation of genetic WNT inhibition models. Our computed transcriptional Notch pathway activation score is upregulated in neural stem cells, as compared to astrocytes; as well as in GSCs, as compared to differentiated glioblastoma cells. Moreover, the Notch signature is clinical predictive in our glioblastoma patient discovery and validation cohort. Notch signature is significantly increased in tumors with mutant IDH1 genome and tumors without 1p and 19q co-deletion. In GSCs with elevated Notch1 expression, BRON treatment blocks transcription of Notch pathway target genes Hes1/Hey1, significantly reduced the amount of cleaved Notch1 receptor protein and caused significantly impairment of cellular invasion. Benchmarking this phenotype to those observed with genetic Notch1 inhibition in corresponding cell models did result in higher reduction of cell invasion under chemotherapy. BRON treatment caused signs of upregulation of Wingless (WNT) stem cell signaling activity, and vice versa, blockage of WNT signaling caused induction of Notch target gene expression in our models. We extend the list of evidences that elevated Notch signal expression is a biomarker signature declaring stem cell prevalence and useful for predicting negative clinical course in glioblastoma. By using functional assays, we validated a first in man tested Notch1 receptor specific antibody as a promising drug candidate in the context of neuro oncology and propose biomarker panel to predict resistance and therapy success of this treatment option. We note that the observed phenotype seems only in part due to Notch1 blockage and the drug candidate leads to activation of off target signals. Further studies addressing a possible emergence of therapy resistance due to WNT activation need to be conducted. We further validated our 3D disease modeling technology to be of benefit for drug development projects.
Collapse
|
19
|
Take Advantage of Glutamine Anaplerosis, the Kernel of the Metabolic Rewiring in Malignant Gliomas. Biomolecules 2020; 10:biom10101370. [PMID: 32993063 PMCID: PMC7599606 DOI: 10.3390/biom10101370] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Glutamine is a non-essential amino acid that plays a key role in the metabolism of proliferating cells including neoplastic cells. In the central nervous system (CNS), glutamine metabolism is particularly relevant, because the glutamine-glutamate cycle is a way of controlling the production of glutamate-derived neurotransmitters by tightly regulating the bioavailability of the amino acids in a neuron-astrocyte metabolic symbiosis-dependent manner. Glutamine-related metabolic adjustments have been reported in several CNS malignancies including malignant gliomas that are considered ‘glutamine addicted’. In these tumors, glutamine becomes an essential amino acid preferentially used in energy and biomass production including glutathione (GSH) generation, which is crucial in oxidative stress control. Therefore, in this review, we will highlight the metabolic remodeling that gliomas undergo, focusing on glutamine metabolism. We will address some therapeutic regimens including novel research attempts to target glutamine metabolism and a brief update of diagnosis strategies that take advantage of this altered profile. A better understanding of malignant glioma cell metabolism will help in the identification of new molecular targets and the design of new therapies.
Collapse
|
20
|
Marisetty AL, Lu L, Veo BL, Liu B, Coarfa C, Kamal MM, Kassem DH, Irshad K, Lu Y, Gumin J, Henry V, Paulucci-Holthauzen A, Rao G, Baladandayuthapani V, Lang FF, Fuller GN, Majumder S. REST-DRD2 mechanism impacts glioblastoma stem cell-mediated tumorigenesis. Neuro Oncol 2020; 21:775-785. [PMID: 30953587 DOI: 10.1093/neuonc/noz030] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a lethal, heterogeneous human brain tumor, with regulatory mechanisms that have yet to be fully characterized. Previous studies have indicated that the transcriptional repressor REST (repressor element-1 silencing transcription factor) regulates the oncogenic potential of GBM stem cells (GSCs) based on level of expression. However, how REST performs its regulatory role is not well understood. METHODS We examined 2 independent high REST (HR) GSC lines using genome-wide assays, biochemical validations, gene knockdown analysis, and mouse tumor models. We analyzed in-house patient tumors and patient data present in The Cancer Genome Atlas (TCGA). RESULTS Genome-wide transcriptome and DNA-binding analyses suggested the dopamine receptor D2 (DRD2) gene, a dominant regulator of neurotransmitter signaling, as a direct target of REST. Biochemical analyses and mouse intracranial tumor models using knockdown of REST and double knockdown of REST and DRD2 validated this target and suggested that DRD2 is a downstream target of REST regulating tumorigenesis, at least in part, through controlling invasion and apoptosis. Further, TCGA GBM data support the presence of the REST-DRD2 axis and reveal that high REST/low DRD2 (HRLD) and low REST/high DRD2 (LRHD) tumors are specific subtypes, are molecularly different from the known GBM subtypes, and represent functional groups with distinctive patterns of enrichment of gene sets and biological pathways. The inverse HRLD/LRHD expression pattern is also seen in in-house GBM tumors. CONCLUSIONS These findings suggest that REST regulates neurotransmitter signaling pathways through DRD2 in HR-GSCs to impact tumorigenesis. They further suggest that the REST-DRD2 mechanism forms distinct subtypes of GBM.
Collapse
Affiliation(s)
- Anantha L Marisetty
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Li Lu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bethany L Veo
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bin Liu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Mohamed Mostafa Kamal
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dina Hamada Kassem
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Khushboo Irshad
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yungang Lu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Verlene Henry
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Ganesh Rao
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregory N Fuller
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sadhan Majumder
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
21
|
Koch K, Hartmann R, Tsiampali J, Uhlmann C, Nickel AC, He X, Kamp MA, Sabel M, Barker RA, Steiger HJ, Hänggi D, Willbold D, Maciaczyk J, Kahlert UD. A comparative pharmaco-metabolomic study of glutaminase inhibitors in glioma stem-like cells confirms biological effectiveness but reveals differences in target-specificity. Cell Death Discov 2020; 6:20. [PMID: 32337072 PMCID: PMC7162917 DOI: 10.1038/s41420-020-0258-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/27/2020] [Accepted: 03/26/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer cells upregulate anabolic processes to maintain high rates of cellular turnover. Limiting the supply of macromolecular precursors by targeting enzymes involved in biosynthesis is a promising strategy in cancer therapy. Several tumors excessively metabolize glutamine to generate precursors for nonessential amino acids, nucleotides, and lipids, in a process called glutaminolysis. Here we show that pharmacological inhibition of glutaminase (GLS) eradicates glioblastoma stem-like cells (GSCs), a small cell subpopulation in glioblastoma (GBM) responsible for therapy resistance and tumor recurrence. Treatment with small molecule inhibitors compound 968 and CB839 effectively diminished cell growth and in vitro clonogenicity of GSC neurosphere cultures. However, our pharmaco-metabolic studies revealed that only CB839 inhibited GLS enzymatic activity thereby limiting the influx of glutamine derivates into the TCA cycle. Nevertheless, the effects of both inhibitors were highly GLS specific, since treatment sensitivity markedly correlated with GLS protein expression. Strikingly, we found GLS overexpressed in in vitro GSC models as compared with neural stem cells (NSC). Moreover, our study demonstrates the usefulness of in vitro pharmaco-metabolomics to score target specificity of compounds thereby refining drug development and risk assessment.
Collapse
Affiliation(s)
- Katharina Koch
- Neurosurgery Department, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Rudolf Hartmann
- Institute of Complex Systems (ICS-6) Structural Biochemistry and JuStruct: Juelich Center for Structural Biology, Forschungszentrum Juelich, 52425 Juelich, Germany
| | - Julia Tsiampali
- Neurosurgery Department, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Constanze Uhlmann
- Neurosurgery Department, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Ann-Christin Nickel
- Neurosurgery Department, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Xiaoling He
- John van Geest Centre for Brain Repair and WT/MRC Cambridge Stem Cell Institute, Department of Clinical Neurosciences, University of Cambridge, CB2 0PY Cambridge, UK
| | - Marcel A. Kamp
- Neurosurgery Department, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Michael Sabel
- Neurosurgery Department, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Roger A. Barker
- John van Geest Centre for Brain Repair and WT/MRC Cambridge Stem Cell Institute, Department of Clinical Neurosciences, University of Cambridge, CB2 0PY Cambridge, UK
| | - Hans-Jakob Steiger
- Neurosurgery Department, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Daniel Hänggi
- Neurosurgery Department, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Dieter Willbold
- Institute of Complex Systems (ICS-6) Structural Biochemistry and JuStruct: Juelich Center for Structural Biology, Forschungszentrum Juelich, 52425 Juelich, Germany
- Institut für Physikalische Biologie, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Jaroslaw Maciaczyk
- Neurosurgery Department, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
- Neurosurgery Department, University Hospital Bonn, 53127 Bonn, Germany
| | - Ulf D. Kahlert
- Neurosurgery Department, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
- German Cancer Consortium (DKTK), Essen/Duesseldorf, Germany
| |
Collapse
|
22
|
Obara-Michlewska M, Szeliga M. Targeting Glutamine Addiction in Gliomas. Cancers (Basel) 2020; 12:cancers12020310. [PMID: 32013066 PMCID: PMC7072559 DOI: 10.3390/cancers12020310] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/16/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
The most common malignant brain tumors are those of astrocytic origin, gliomas, with the most aggressive glioblastoma (WHO grade IV) among them. Despite efforts, medicine has not made progress in terms of the prognosis and life expectancy of glioma patients. Behind the malignant phenotype of gliomas lies multiple genetic mutations leading to reprogramming of their metabolism, which gives those highly proliferating cells an advantage over healthy ones. The so-called glutamine addiction is a metabolic adaptation that supplements oxidative glycolysis in order to secure neoplastic cells with nutrients and energy in unfavorable conditions of hypoxia. The present review aims at presenting the research and clinical attempts targeting the different metabolic pathways involved in glutamine metabolism in gliomas. A brief description of the biochemistry of glutamine transport, synthesis, and glutaminolysis, etc. will forego a detailed comparison of the therapeutic strategies undertaken to inhibit glutamine utilization by gliomas.
Collapse
|
23
|
Amino Acid-Mediated Metabolism: A New Power to Influence Properties of Stem Cells. Stem Cells Int 2019; 2019:6919463. [PMID: 31885621 PMCID: PMC6915148 DOI: 10.1155/2019/6919463] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/05/2019] [Accepted: 11/15/2019] [Indexed: 01/10/2023] Open
Abstract
The self-renewal and differentiation potentials of stem cells are dependent on amino acid (AA) metabolism. We review the literature on the metabolic preference of both cancer and noncancer stem cells. The balance in AA metabolism is responsible for maintaining the functionality of noncancer stem cells, and altering the levels of AAs can influence the malignant biological behavior of cancer stem cells. AAs are considered nutrients participating in metabolism and playing a critical role in maintaining the activity of normal stem cells and the effect of therapy of cancer stem cells. Targeting AA metabolism helps inhibit the stemness of cancer stem cells and remodels the function of normal stem cells. This review summarizes the metabolic characteristics and regulation pathways of AA in different stem cells, not only from the nutritional perspective but also from the genomic perspective that have been reported in the recent five years. In addition, we briefly survey new therapeutic modalities that may help eradicate cancer stem cells by exploiting nutrient deprivation. Understanding AA uptake characteristics helps researchers define the preference for AA in different stem cells and enables clinicians make timely interventions to specifically target the cell behavior.
Collapse
|
24
|
Clinically Actionable Insights into Initial and Matched Recurrent Glioblastomas to Inform Novel Treatment Approaches. JOURNAL OF ONCOLOGY 2019; 2019:4878547. [PMID: 32082376 PMCID: PMC7012245 DOI: 10.1155/2019/4878547] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 10/07/2019] [Accepted: 10/25/2019] [Indexed: 12/31/2022]
Abstract
Glioblastoma is the most common primary adult brain tumour, and despite optimal treatment, the median survival is 12–15 months. Patients with matched recurrent glioblastomas were investigated to try to find actionable mutations. Tumours were profiled using a validated DNA-based gene panel. Copy number variations (CNVs) and single nucleotide variants (SNVs) were examined, and potentially pathogenic variants and clinically actionable mutations were identified. The results revealed that glioblastomas were IDH-wildtype (IDHWT; n = 38) and IDH-mutant (IDHMUT; n = 3). SNVs in TSC2, MSH6, TP53, CREBBP, and IDH1 were variants of unknown significance (VUS) that were predicted to be pathogenic in both subtypes. IDHWT tumours had SNVs that impacted RTK/Ras/PI(3)K, p53, WNT, SHH, NOTCH, Rb, and G-protein pathways. Many tumours had BRCA1/2 (18%) variants, including confirmed somatic mutations in haemangioblastoma. IDHWT recurrent tumours had fewer pathways impacted (RTK/Ras/PI(3)K, p53, WNT, and G-protein) and CNV gains (BRCA2, GNAS, and EGFR) and losses (TERT and SMARCA4). IDHMUT tumours had SNVs that impacted RTK/Ras/PI(3)K, p53, and WNT pathways. VUS in KLK1 was possibly pathogenic in IDHMUT. Recurrent tumours also had fewer pathways (p53, WNT, and G-protein) impacted by genetic alterations. Public datasets (TCGA and GDC) confirmed the clinical significance of findings in both subtypes. Overall in this cohort, potentially actionable variation was most often identified in EGFR, PTEN, BRCA1/2, and ATM. This study underlines the need for detailed molecular profiling to identify individual GBM patients who may be eligible for novel treatment approaches. This information is also crucial for patient recruitment to clinical trials.
Collapse
|
25
|
Bazzoni R, Bentivegna A. Role of Notch Signaling Pathway in Glioblastoma Pathogenesis. Cancers (Basel) 2019; 11:cancers11030292. [PMID: 30832246 PMCID: PMC6468848 DOI: 10.3390/cancers11030292] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/17/2019] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
Notch signaling is an evolutionarily conserved pathway that regulates important biological processes, such as cell proliferation, apoptosis, migration, self-renewal, and differentiation. In mammals, Notch signaling is composed of four receptors (Notch1–4) and five ligands (Dll1-3–4, Jagged1–2) that mainly contribute to the development and maintenance of the central nervous system (CNS). Neural stem cells (NSCs) are the starting point for neurogenesis and other neurological functions, representing an essential aspect for the homeostasis of the CNS. Therefore, genetic and functional alterations to NSCs can lead to the development of brain tumors, including glioblastoma. Glioblastoma remains an incurable disease, and the reason for the failure of current therapies and tumor relapse is the presence of a small subpopulation of tumor cells known as glioma stem cells (GSCs), characterized by their stem cell-like properties and aggressive phenotype. Growing evidence reveals that Notch signaling is highly active in GSCs, where it suppresses differentiation and maintains stem-like properties, contributing to Glioblastoma tumorigenesis and conventional-treatment resistance. In this review, we try to give a comprehensive view of the contribution of Notch signaling to Glioblastoma and its possible implication as a target for new therapeutic approaches.
Collapse
Affiliation(s)
- Riccardo Bazzoni
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Pz.le Scuro 10, 37134 Verona, Italy.
- Program in Clinical and Experimental Biomedical Sciences, University of Verona, 37134 Verona, Italy.
- NeuroMi, Milan Center for Neuroscience, Department of Neurology and Neuroscience, San Gerardo Hospital, University of Milano-Bicocca, 20900 Monza, Italy.
| | - Angela Bentivegna
- NeuroMi, Milan Center for Neuroscience, Department of Neurology and Neuroscience, San Gerardo Hospital, University of Milano-Bicocca, 20900 Monza, Italy.
- School of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy.
| |
Collapse
|
26
|
Transfection with GLS2 Glutaminase (GAB) Sensitizes Human Glioblastoma Cell Lines to Oxidative Stress by a Common Mechanism Involving Suppression of the PI3K/AKT Pathway. Cancers (Basel) 2019; 11:cancers11010115. [PMID: 30669455 PMCID: PMC6356507 DOI: 10.3390/cancers11010115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 01/23/2023] Open
Abstract
GLS-encoded glutaminase promotes tumorigenesis, while GLS2-encoded glutaminase displays tumor-suppressive properties. In glioblastoma (GBM), the most aggressive brain tumor, GLS is highly expressed and in most cases GLS2 is silenced. Previously, it was shown that transfection with a sequence encoding GAB, the main GLS2 isoform, decreased the survival, growth, and ability to migrate of human GBM cells T98G and increased their sensitivity towards an alkylating agent temozolomide (TMZ) and oxidative stress compared to the controls, by a not well-defined mechanism. In this study we report that GAB transfection inhibits growth and increases susceptibility towards TMZ and H2O2-mediated oxidative stress of two other GBM cell lines, U87MG and LN229. We also show that in GAB-transfected cells treated with H2O2, the PI3K/AKT pathway is less induced compared to the pcDNA-transfected counterparts and that pretreatment with PDGF-BB, an activator of AKT, protects GAB-transfected cells from death caused by the H2O2 treatment. In conclusion, our results show that (i) GAB suppresses the malignant phenotype of the GBM cells of different tumorigenic potentials and genetic backgrounds and (ii) the GAB-mediated increase of sensitivity to oxidative stress is causally related to the inhibition of the PI3K/AKT pathway. The upregulation of the GLS2 expression and the inhibition of the PI3K/AKT pathway may become a novel combined therapeutic strategy for anti-glioma preclinical investigations.
Collapse
|
27
|
Abstract
INTRODUCTION The kidney-type glutaminase (GLS) controlling the first step of glutamine metabolism is overexpressed in many cancer cells. Targeting inhibition of GLS shows obvious inhibitory effects on cancer cell proliferation. Therefore, extensive research and development of GLS inhibitors have been carried out in industrial and academic institutions over the past decade to address this unmet medical need. AREAS COVERED This review covers researches and patent literatures in the field of discovery and development of small molecule inhibitors of GLS for cancer therapy over the past 16 years. EXPERT OPINION The detailed ligand-receptor interaction information from their complex structure not only guides the rational drug design, but also facilitates in silico structure-based virtual ligand screening of novel GLS inhibitors. Multi-drug combination administration is of great significance both in terms of safety and efficacy.
Collapse
Affiliation(s)
- CanRong Wu
- a Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China
| | - LiXia Chen
- b Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education , Shenyang Pharmaceutical University , Shenyang , China
| | - Sanshan Jin
- c Maternal and Child Health Hospital of Hubei Province , Wuhan , China
| | - Hua Li
- a Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China.,b Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education , Shenyang Pharmaceutical University , Shenyang , China
| |
Collapse
|
28
|
Hombach-Klonisch S, Mehrpour M, Shojaei S, Harlos C, Pitz M, Hamai A, Siemianowicz K, Likus W, Wiechec E, Toyota BD, Hoshyar R, Seyfoori A, Sepehri Z, Ande SR, Khadem F, Akbari M, Gorman AM, Samali A, Klonisch T, Ghavami S. Glioblastoma and chemoresistance to alkylating agents: Involvement of apoptosis, autophagy, and unfolded protein response. Pharmacol Ther 2018; 184:13-41. [DOI: 10.1016/j.pharmthera.2017.10.017] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
29
|
Akins NS, Nielson TC, Le HV. Inhibition of Glycolysis and Glutaminolysis: An Emerging Drug Discovery Approach to Combat Cancer. Curr Top Med Chem 2018; 18:494-504. [PMID: 29788892 PMCID: PMC6110043 DOI: 10.2174/1568026618666180523111351] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 05/05/2018] [Accepted: 05/11/2018] [Indexed: 12/15/2022]
Abstract
Cancer cells have a very different metabolism from that of normal cells from which they are derived. Their metabolism is elevated, which allows them to sustain higher proliferative rate and resist some cell death signals. This phenomenon, known as the "Warburg effect", has become the focus of intensive efforts in the discovery of new therapeutic targets and new cancer drugs. Both glycolysis and glutaminolysis pathways are enhanced in cancer cells. While glycolysis is enhanced to satisfy the increasing energy demand of cancer cells, glutaminolysis is enhanced to provide biosynthetic precursors for cancer cells. It was recently discovered that there is a tyrosine phosphorylation of a specific isoform of pyruvate kinase, the M2 isoform, that is preferentially expressed in all cancer cells, which results in the generation of pyruvate through a unique enzymatic mechanism that is uncoupled from ATP production. Pyruvate produced through this unique enzymatic mechanism is converted primarily into lactic acid, rather than acetyl-CoA for the synthesis of citrate, which would normally then enter the citric acid cycle. Inhibition of key enzymes in glycolysis and glutaminolysis pathways with small molecules has provided a novel but emerging area of cancer research and has been proven effective in slowing the proliferation of cancer cells, with several inhibitors being in clinical trials. This review paper will cover recent advances in the development of chemotherapeutic agents against several metabolic targets for cancer therapy, including glucose transporters, hexokinase, pyruvate kinase M2, glutaminase, and isocitrate dehydrogenase.
Collapse
Affiliation(s)
- Nicholas S. Akins
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Mississippi 38677, USA
| | - Tanner C. Nielson
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Mississippi 38677, USA
| | - Hoang V. Le
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Mississippi 38677, USA
| |
Collapse
|
30
|
Metabolic shift in density-dependent stem cell differentiation. Cell Commun Signal 2017; 15:44. [PMID: 29052507 PMCID: PMC5649068 DOI: 10.1186/s12964-017-0173-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 05/02/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Vascular progenitor cells (VPCs) derived from embryonic stem cells (ESCs) are a valuable source for cell- and tissue-based therapeutic strategies. During the optimization of endothelial cell (EC) inductions from mouse ESCs using our staged and chemically-defined induction methods, we found that cell seeding density but not VEGF treatment between 10 ng/mL and 40 ng/mL was a significant variable directing ESCs into FLK1+ VPCs during stage 1 induction. Here, we examine potential contributions from cell-to-cell signaling or cellular metabolism in the production of VPCs from ESCs seeded at different cell densities. METHODS Using 1D 1H-NMR spectroscopy, transcriptomic arrays, and flow cytometry, we observed that the density-dependent differentiation of ESCs into FLK1+ VPCs positively correlated with a shift in metabolism and cellular growth. RESULTS Specifically, cell differentiation correlated with an earlier plateauing of exhaustive glycolysis, decreased lactate production, lower metabolite consumption, decreased cellular proliferation and an increase in cell size. In contrast, cells seeded at a lower density of 1,000 cells/cm2 exhibited increased rates of glycolysis, lactate secretion, metabolite utilization, and proliferation over the same induction period. Gene expression analysis indicated that high cell seeding density correlated with up-regulation of several genes including cell adhesion molecules of the notch family (NOTCH1 and NOTCH4) and cadherin family (CDH5) related to vascular development. CONCLUSIONS These results confirm that a distinct metabolic phenotype correlates with cell differentiation of VPCs.
Collapse
|
31
|
Maciaczyk D, Picard D, Zhao L, Koch K, Herrera-Rios D, Li G, Marquardt V, Pauck D, Hoerbelt T, Zhang W, Ouwens DM, Remke M, Jiang T, Steiger HJ, Maciaczyk J, Kahlert UD. CBF1 is clinically prognostic and serves as a target to block cellular invasion and chemoresistance of EMT-like glioblastoma cells. Br J Cancer 2017; 117:102-112. [PMID: 28571041 PMCID: PMC5520214 DOI: 10.1038/bjc.2017.157] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Glioblastoma is the most common and most lethal primary brain cancer. CBF1 (also known as Recombination signal Binding Protein for immunoglobulin kappa J, RBPJ) is the cardinal transcriptional regulator of the Notch signalling network and has been shown to promote cancer stem-like cells (CSCs) in glioblastoma. Recent studies suggest that some of the malignant properties of CSCs are mediated through the activation of pro-invasive programme of epithelial-to-mesenchymal transition (EMT). Little is known whether CBF1 is involved in the EMT-like phenotype of glioma cells. METHODS In a collection of GBM neurosphere lines, we genetically inhibited CBF1 and investigated the consequences on EMT-related properties, including in vitro invasiveness by Boyden chambers assay, chemoresistance using a clinical drug library screen and glycolytic metabolism assessing live-cell extracellular acidification rate. We also compared CBF1 expression in cells exposed to low and high oxygen tension. In silico analysis in large-scale Western and Eastern patient cohorts investigated the clinical prognostic value of CBF1 expression in low- and high-grade glioma as well as medulloblastoma. RESULTS Mean CBF1 expression is significantly increased in isocitrate dehydrogenase 1 (IDH1) R132H mutant glioblastoma and serves as prognostic marker for prolonged overall survival in brain tumours, particularly after therapy with temozolomide. Hypoxic regions of glioblastoma have higher CBF1 activation and exposure to low oxygen can induce its expression in glioma cells in vitro. CBF1 inhibition blocks EMT activators such as zinc finger E-box-binding homeobox 1 (ZEB1) and significantly reduces cellular invasion and resistance to clinically approved anticancer drugs. Moreover, we indicate that CBF1 inhibition can impede cellular glycolysis. CONCLUSIONS Mean CBF1 activation in bulk tumour samples serves as a clinical predictive biomarker in brain cancers but its intratumoral and intertumoral expression is highly heterogeneous. Microenvironmental changes such as hypoxia can stimulate the activation of CBF1 in glioblastoma. CBF1 blockade can suppress glioblastoma invasion in vitro in particular in cells undergone EMT such as those found in the hypoxic niche. Targeting CBF1 can be an effective anti-EMT therapy to impede invasive properties and chemosensitivity in those cells.
Collapse
Affiliation(s)
- D Maciaczyk
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine University Dusseldorf, Dusseldorf 40225, Germany
| | - D Picard
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Heinrich-Heine University Dusseldorf, Dusseldorf 40225, Germany.,Department of Neuropathology, Medical Faculty, Heinrich-Heine University Düsseldorf, Dusseldorf 40225, Germany.,Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - L Zhao
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine University Dusseldorf, Dusseldorf 40225, Germany
| | - K Koch
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine University Dusseldorf, Dusseldorf 40225, Germany
| | - D Herrera-Rios
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine University Dusseldorf, Dusseldorf 40225, Germany
| | - G Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing 100050, China
| | - V Marquardt
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Heinrich-Heine University Dusseldorf, Dusseldorf 40225, Germany.,Department of Neuropathology, Medical Faculty, Heinrich-Heine University Düsseldorf, Dusseldorf 40225, Germany.,Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine University Düsseldorf, Dusseldorf 40225, Germany
| | - D Pauck
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Heinrich-Heine University Dusseldorf, Dusseldorf 40225, Germany.,Department of Neuropathology, Medical Faculty, Heinrich-Heine University Düsseldorf, Dusseldorf 40225, Germany.,Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - T Hoerbelt
- Institute of Clinical Biochemistry and Pathobiochemistry, German Center for Diabetes Research (DZD), Dusseldorf, Germany
| | - W Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing 100050, China
| | - D M Ouwens
- Institute of Clinical Biochemistry and Pathobiochemistry, German Center for Diabetes Research (DZD), Dusseldorf, Germany
| | - M Remke
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Heinrich-Heine University Dusseldorf, Dusseldorf 40225, Germany.,Department of Neuropathology, Medical Faculty, Heinrich-Heine University Düsseldorf, Dusseldorf 40225, Germany.,Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - T Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing 100050, China.,Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China
| | - H J Steiger
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine University Dusseldorf, Dusseldorf 40225, Germany
| | - J Maciaczyk
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine University Dusseldorf, Dusseldorf 40225, Germany
| | - U D Kahlert
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine University Dusseldorf, Dusseldorf 40225, Germany
| |
Collapse
|
32
|
Yuan L, Sheng X, Clark LH, Zhang L, Guo H, Jones HM, Willson AK, Gehrig PA, Zhou C, Bae-Jump VL. Glutaminase inhibitor compound 968 inhibits cell proliferation and sensitizes paclitaxel in ovarian cancer. Am J Transl Res 2016; 8:4265-4277. [PMID: 27830010 PMCID: PMC5095319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/14/2016] [Indexed: 06/06/2023]
Abstract
Our overall goal was to investigate the anti-tumor activity of the glutaminase 1 (GLS1) Inhibitor compound 968 in ovarian cancer cells. The human ovarian cancer cell lines, HEY, SKOV3 and IGROV-1 were used. Cell proliferation was assessed by MTT assay after treatment with compound 968. Cell cycle progression and Annexin V expression were evaluated using Cellometer. Western blotting was performed to determine changes in GLS1, cellular stress and cell cycle checkpoints. Reactive oxygen species (ROS) and glutamate dehydrogenase (GDH) activity were assessed by ELISA assay. Compound 968 significantly inhibited cell proliferation and the expression of GLS1 in a dose-dependent manner in all three ovarian cancer cell lines. Compound 968 induced G1 phase cell cycle arrest and apoptosis. Treatment with compound 968 increased ROS levels and induced the protein expression of calnexin, binding immunoglobulin protein (BiP) and protein kinase RNA-like endoplasmic reticulum kinase (PERK). Deprivation of glutamine increased the sensitivity of cells to paclitaxel, and compound 968 sensitized cells to the anti-proliferative effects of paclitaxel. Compound 968 inhibited cell growth in ovarian cancer cells through induction of G1 phase cell cycle arrest, apoptosis and cellular stress, suggesting that targeting GLS1 provide a novel therapeutic strategy for ovarian cancer.
Collapse
Affiliation(s)
- Lingqin Yuan
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical SciencesJinan, Shandong, China
- Division of Gynecological Oncology, University of North CarolinaChapel Hill, NC, USA
| | - Xiugui Sheng
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical SciencesJinan, Shandong, China
| | - Leslie H Clark
- Division of Gynecological Oncology, University of North CarolinaChapel Hill, NC, USA
| | - Lu Zhang
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical SciencesJinan, Shandong, China
- Division of Gynecological Oncology, University of North CarolinaChapel Hill, NC, USA
| | - Hui Guo
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical SciencesJinan, Shandong, China
- Division of Gynecological Oncology, University of North CarolinaChapel Hill, NC, USA
| | - Hannah M Jones
- Division of Gynecological Oncology, University of North CarolinaChapel Hill, NC, USA
| | - Adam K Willson
- Division of Gynecological Oncology, University of North CarolinaChapel Hill, NC, USA
| | - Paola A Gehrig
- Division of Gynecological Oncology, University of North CarolinaChapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North CarolinaChapel Hill, NC, USA
| | - Chunxiao Zhou
- Division of Gynecological Oncology, University of North CarolinaChapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North CarolinaChapel Hill, NC, USA
| | - Victoria L Bae-Jump
- Division of Gynecological Oncology, University of North CarolinaChapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North CarolinaChapel Hill, NC, USA
| |
Collapse
|
33
|
Kahlert UD, Mooney SM, Natsumeda M, Steiger HJ, Maciaczyk J. Targeting cancer stem-like cells in glioblastoma and colorectal cancer through metabolic pathways. Int J Cancer 2016; 140:10-22. [PMID: 27389307 DOI: 10.1002/ijc.30259] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/29/2016] [Accepted: 07/04/2016] [Indexed: 12/12/2022]
Abstract
Cancer stem-like cells (CSCs) are thought to be the main cause of tumor occurrence, progression and therapeutic resistance. Strong research efforts in the last decade have led to the development of several tailored approaches to target CSCs with some very promising clinical trials underway; however, until now no anti-CSC therapy has been approved for clinical use. Given the recent improvement in our understanding of how onco-proteins can manipulate cellular metabolic networks to promote tumorigenesis, cancer metabolism research may well lead to innovative strategies to identify novel regulators and downstream mediators of CSC maintenance. Interfering with distinct stages of CSC-associated metabolics may elucidate novel, more efficient strategies to target this highly malignant cell population. Here recent discoveries regarding the metabolic properties attributed to CSCs in glioblastoma (GBM) and malignant colorectal cancer (CRC) were summarized. The association between stem cell markers, the response to hypoxia and other environmental stresses including therapeutic insults as well as developmentally conserved signaling pathways with alterations in cellular bioenergetic networks were also discussed. The recent developments in metabolic imaging to identify CSCs were also summarized. This summary should comprehensively update basic and clinical scientists on the metabolic traits of CSCs in GBM and malignant CRC.
Collapse
Affiliation(s)
- U D Kahlert
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| | - S M Mooney
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - M Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - H-J Steiger
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| | - J Maciaczyk
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| |
Collapse
|
34
|
Desbats MA, Giacomini I, Prayer-Galetti T, Montopoli M. Iron granules in plasma cells. J Clin Pathol 1982; 10:281. [PMID: 32211323 PMCID: PMC7068907 DOI: 10.3389/fonc.2020.00281] [Citation(s) in RCA: 99] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/18/2020] [Indexed: 01/16/2023]
Abstract
Resistance of cancer cells to chemotherapy is the first cause of cancer-associated death. Thus, new strategies to deal with the evasion of drug response and to improve clinical outcomes are needed. Genetic and epigenetic mechanisms associated with uncontrolled cell growth result in metabolism reprogramming. Cancer cells enhance anabolic pathways and acquire the ability to use different carbon sources besides glucose. An oxygen and nutrient-poor tumor microenvironment determines metabolic interactions among normal cells, cancer cells and the immune system giving rise to metabolically heterogeneous tumors which will partially respond to metabolic therapy. Here we go into the best-known cancer metabolic profiles and discuss several studies that reported tumors sensitization to chemotherapy by modulating metabolic pathways. Uncovering metabolic dependencies across different chemotherapy treatments could help to rationalize the use of metabolic modulators to overcome therapy resistance.
Collapse
Affiliation(s)
- Maria Andrea Desbats
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Isabella Giacomini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Monica Montopoli
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- *Correspondence: Monica Montopoli
| |
Collapse
|