Targeting Tumor Metabolism: From Mechanisms to Therapies II

A special issue of Biomolecules (ISSN 2218-273X). This special issue belongs to the section "Cellular Biochemistry".

Deadline for manuscript submissions: closed (15 March 2023) | Viewed by 22791

Special Issue Editors


E-Mail Website
Guest Editor
Department of Chemistry, Technological Laboratories, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-093 Aveiro, Portugal
Interests: toxicity and biological assessment of (nano)materials with biomedical applications; metabolism of immune cells and metabolic immunomodulation; metabolism of tumor cells and metabolic effects of anticancer therapies (drugs, nanomedicines, photothermal therapy); NMR spectroscopy and metabolomics
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Chemistry, Technological Laboratories, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-093 Aveiro, Portugal
Interests: metabolomics of human biofluids, tissues, and cellular systems for the biochemical characterisation of diseases (mainly prenatal/newborn disorders and cancer); metabolomics testing of bio- and/or nano-materials for assessment of in vitro and in vivo biological performance; metabolomic assessment of environmental effects on human metabolism; metabolomics for the routine quality control of products and processes in the food industry
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Following a very successful first run, we are pleased to announce the launch of a second edition of a Special Issue on Targeting Tumor Metabolism.

With the support of advanced molecular and analytical tools, our understanding of tumor metabolism has grown immensely over the past years. The intricate relationship between oncogenic events and metabolic reprogramming is currently recognized as an important cancer hallmark, which actively supports the proliferation, survival, and migration of tumor cells. Different metabolic features such as elevated glucose uptake and glycolysis, enhanced synthesis of nucleotides and lipids, or an altered redox status have been identified across several cellular and animal models, as well as in human samples. Various metabolites have been proposed as disease biomarkers with potential diagnostic or prognostic value, and a renewed interest in exploring metabolic targets for therapeutic purposes has arisen. At the same time, a number of important questions have emerged, for instance, regarding the metabolic diversity between tumor types and their histological/molecular subtypes, the metabolic crosstalk between tumor cells and other cells in the tumor microenvironment (such as immune cells, adipocytes, and fibroblasts), or the translatability of the in vitro findings to in vivo situations.

For this Special Issue, we would like to invite review papers or original research articles that address the topic of tumor metabolism and the above-mentioned questions. In particular, studies that contribute to advancing the current mechanistic understanding of cancer metabolic rewiring, as well as manuscripts providing novel application-oriented perspectives, are highly welcome.

We look forward to receiving your contributions.

Dr. Iola F. Duarte Ciceco
Prof. Ana M. Gil
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Biomolecules is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • tumor metabolism
  • metabolic pathways
  • ‘omics’ approaches
  • metabolic targets and drugs
  • tumor microenvironment
  • metabolic biomarkers

Related Special Issue

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

15 pages, 3073 KiB  
Article
Liver X Receptor Inverse Agonist GAC0001E5 Impedes Glutaminolysis and Disrupts Redox Homeostasis in Breast Cancer Cells
by Asitha Premaratne, Charles Ho, Shinjini Basu, Ashfia Fatima Khan, Tasneem Bawa-Khalfe and Chin-Yo Lin
Biomolecules 2023, 13(2), 345; https://doi.org/10.3390/biom13020345 - 10 Feb 2023
Viewed by 2302
Abstract
Liver X receptors (LXRs) are members of the nuclear receptor family of ligand-dependent transcription factors which regulate the expression of lipid and cholesterol metabolism genes. Moreover, LXRs and their ligands have been shown to inhibit tumor growth in a variety of cancers. We [...] Read more.
Liver X receptors (LXRs) are members of the nuclear receptor family of ligand-dependent transcription factors which regulate the expression of lipid and cholesterol metabolism genes. Moreover, LXRs and their ligands have been shown to inhibit tumor growth in a variety of cancers. We have previously identified the small molecule compound GAC0001E5 (1E5) as an LXR inverse agonist and a potent inhibitor of pancreatic cancer cells. Transcriptomic and metabolomic studies showed that 1E5 disrupts glutamine metabolism, an essential metabolic pathway commonly reprogrammed during malignant transformation, including in breast cancers. To determine the role of LXRs and potential application of 1E5 in breast cancer, we examined LXR expression in publicly available clinical samples, and found that LXR expression is elevated in breast tumors as compared to normal tissues. In luminal A, endocrine therapy-resistant, and triple-negative breast cancer cells, 1E5 exhibited LXR inverse agonist and “degrader” activity and strongly inhibited cell proliferation and colony formation. Treatments with 1E5 downregulated the transcription of key glutaminolysis genes, and, correspondingly, biochemical assays indicated that 1E5 lowered intracellular glutamate and glutathione levels and increased reactive oxygen species. These results indicate that novel LXR ligand 1E5 is an inhibitor of glutamine metabolism and redox homeostasis in breast cancers and suggest that modulating LXR activity and expression in tumor cells is a promising strategy for targeting metabolic reprogramming in breast cancer therapeutics. Full article
(This article belongs to the Special Issue Targeting Tumor Metabolism: From Mechanisms to Therapies II)
Show Figures

Figure 1

17 pages, 4030 KiB  
Article
Alteration of Cellular Energy Metabolism through LPAR2-Axin2 Axis in Gastric Cancer
by Hosne Ara, Utsab Subedi, Papori Sharma, Susmita Bhattarai, Sudha Sharma, Shrivats Manikandan, Xiuping Yu, Md. Shenuarin Bhuiyan, Hong Sun, Sumitra Miriyala and Manikandan Panchatcharam
Biomolecules 2022, 12(12), 1805; https://doi.org/10.3390/biom12121805 - 02 Dec 2022
Cited by 3 | Viewed by 2151
Abstract
Lysophosphatidic acid (LPA), a multifunctional endogenous phospholipid, plays a vital role in cellular homeostasis and the malignant behavior of cancer cells through G-protein-coupled receptors. However, the role of LPA in β-catenin-mediated gastric cancer is unknown. Here, we have noted the high expression of [...] Read more.
Lysophosphatidic acid (LPA), a multifunctional endogenous phospholipid, plays a vital role in cellular homeostasis and the malignant behavior of cancer cells through G-protein-coupled receptors. However, the role of LPA in β-catenin-mediated gastric cancer is unknown. Here, we have noted the high expression of LPAR2 in human gastric cancer tissues, and that LPA treatment significantly increased the proliferation, migration, and invasion of human gastric cancer cells. Results from our biochemical experiments showed that an LPA exposure increased the expression of β-catenin and its nuclear localization, increased the phosphorylation of glycogen synthase kinase 3β (GSK-3β), decreased the expression of Axin2, and increased the expression of the target genes of the β-catenin signaling pathway. The LPA2 receptor (LPAR2) antagonist significantly reduced the LPA-induced nuclear localization of β-catenin, the primary signaling event. The knockdown of LPAR2 in the gastric cancer cell lines robustly reduced the LPA-induced β-catenin activity. An LPA exposure increased the ATP production by both oxidative phosphorylation and glycolysis, and this effect was abrogated with the addition of an LPAR2 antagonist and XAV393, which stabilizes the Axin and inhibits the β-catenin signaling pathway. Based on our findings, the possibility that LPA contributes to gastric cancer initiation and progression through the β-catenin signaling pathway as well as by the dysregulation of the energy metabolism via the LPAR2 receptor and Axin2, respectively, provides a novel insight into the mechanism of and possible therapeutic targets of gastric cancer. Full article
(This article belongs to the Special Issue Targeting Tumor Metabolism: From Mechanisms to Therapies II)
Show Figures

Figure 1

14 pages, 1650 KiB  
Article
Statins Alleviate Tumor Hypoxia in Prostate Cancer Models by Decreasing Oxygen Consumption: An Opportunity for Radiosensitization?
by Donatienne d’Hose, Lionel Mignion, Loïc Hamelin, Pierre Sonveaux, Bénédicte F. Jordan and Bernard Gallez
Biomolecules 2022, 12(10), 1418; https://doi.org/10.3390/biom12101418 - 03 Oct 2022
Cited by 4 | Viewed by 1507
Abstract
Background: Because statins were found to decrease the oxygen consumption rate (OCR) of a variety of normal cells, our hypothesis was that statins may also decrease the OCR of cancer cells, alleviate tumor hypoxia and radiosensitize tumors. Methods: OCR was assessed using the [...] Read more.
Background: Because statins were found to decrease the oxygen consumption rate (OCR) of a variety of normal cells, our hypothesis was that statins may also decrease the OCR of cancer cells, alleviate tumor hypoxia and radiosensitize tumors. Methods: OCR was assessed using the Seahorse XF96 technology and EPR respirometry in PC-3 prostate cancer cells. Mitochondrial superoxide production was measured by EPR with mitoTEMPO-H as a sensing probe. Tumor pO2 was measured in vivo using low-frequency EPR oximetry to define the optimal window of reoxygenation, the time at which tumors were irradiated with a single 6 Gy dose with a Cesium-137 irradiator. Results: 24-h exposure to simvastatin and fluvastatin significantly decreased the OCR of PC-3 cancer cells. An increase in mitochondrial superoxide levels was also observed after fluvastatin exposure. The PC-3 prostate cancer model was found highly hypoxic at the basal level. When mice were treated with simvastatin or fluvastatin (daily injection of 20 mg/kg), tumor oxygenation increased 48 and 72 h after initiation of the treatment. However, despite reoxygenation, simvastatin did not sensitize the PC-3 tumor model to RT. Conclusions: exposure to statins affect tumor metabolism and tumor oxygenation, however, with limited impact on tumor growth with or without irradiation. Full article
(This article belongs to the Special Issue Targeting Tumor Metabolism: From Mechanisms to Therapies II)
Show Figures

Figure 1

20 pages, 3086 KiB  
Article
Altered L-Arginine Metabolic Pathways in Gastric Cancer: Potential Therapeutic Targets and Biomarkers
by Iwona Bednarz-Misa, Mariusz G. Fleszar, Paulina Fortuna, Łukasz Lewandowski, Magdalena Mierzchała-Pasierb, Dorota Diakowska and Małgorzata Krzystek-Korpacka
Biomolecules 2021, 11(8), 1086; https://doi.org/10.3390/biom11081086 - 23 Jul 2021
Cited by 14 | Viewed by 3586
Abstract
There is a pressing need for molecular targets and biomarkers in gastric cancer (GC). We aimed at identifying aberrations in L-arginine metabolism with therapeutic and diagnostic potential. Systemic metabolites were quantified using mass spectrometry in 293 individuals and enzymes’ gene expression was quantified [...] Read more.
There is a pressing need for molecular targets and biomarkers in gastric cancer (GC). We aimed at identifying aberrations in L-arginine metabolism with therapeutic and diagnostic potential. Systemic metabolites were quantified using mass spectrometry in 293 individuals and enzymes’ gene expression was quantified in 29 paired tumor-normal samples using qPCR and referred to cancer pathology and molecular landscape. Patients with cancer or benign disorders had reduced systemic arginine, citrulline, and ornithine and elevated symmetric dimethylarginine and dimethylamine. Citrulline and ornithine depletion was accentuated in metastasizing cancers. Metabolite diagnostic panel had 91% accuracy in detecting cancer and 70% accuracy in differentiating cancer from benign disorders. Gastric tumors had upregulated NOS2 and downregulated ASL, PRMT2, ORNT1, and DDAH1 expression. NOS2 upregulation was less and ASL downregulation was more pronounced in metastatic cancers. Tumor ASL and PRMT2 expression was inversely related to local advancement. Enzyme up- or downregulation was greater or significant solely in cardia subtype. Metabolic reprogramming in GC includes aberrant L-arginine metabolism, reflecting GC subtype and pathology, and is manifested by altered interplay of its intermediates and enzymes. Exploiting L-arginine metabolic pathways for diagnostic and therapeutic purposes is warranted. Functional studies on ASL, PRMT2, and ORNT1 in GC are needed. Full article
(This article belongs to the Special Issue Targeting Tumor Metabolism: From Mechanisms to Therapies II)
Show Figures

Figure 1

Review

Jump to: Research

70 pages, 8544 KiB  
Review
The Interplay between Dysregulated Metabolism and Epigenetics in Cancer
by Mahmoud Adel Bassal
Biomolecules 2023, 13(6), 944; https://doi.org/10.3390/biom13060944 - 05 Jun 2023
Cited by 1 | Viewed by 2615
Abstract
Cellular metabolism (or energetics) and epigenetics are tightly coupled cellular processes. It is arguable that of all the described cancer hallmarks, dysregulated cellular energetics and epigenetics are the most tightly coregulated. Cellular metabolic states regulate and drive epigenetic changes while also being capable [...] Read more.
Cellular metabolism (or energetics) and epigenetics are tightly coupled cellular processes. It is arguable that of all the described cancer hallmarks, dysregulated cellular energetics and epigenetics are the most tightly coregulated. Cellular metabolic states regulate and drive epigenetic changes while also being capable of influencing, if not driving, epigenetic reprogramming. Conversely, epigenetic changes can drive altered and compensatory metabolic states. Cancer cells meticulously modify and control each of these two linked cellular processes in order to maintain their tumorigenic potential and capacity. This review aims to explore the interplay between these two processes and discuss how each affects the other, driving and enhancing tumorigenic states in certain contexts. Full article
(This article belongs to the Special Issue Targeting Tumor Metabolism: From Mechanisms to Therapies II)
Show Figures

Figure 1

24 pages, 853 KiB  
Review
One-Carbon and Polyamine Metabolism as Cancer Therapy Targets
by Anowarul Islam, Zeeshan Shaukat, Rashid Hussain and Stephen L. Gregory
Biomolecules 2022, 12(12), 1902; https://doi.org/10.3390/biom12121902 - 19 Dec 2022
Cited by 4 | Viewed by 3256
Abstract
Cancer metabolic reprogramming is essential for maintaining cancer cell survival and rapid replication. A common target of this metabolic reprogramming is one-carbon metabolism which is notable for its function in DNA synthesis, protein and DNA methylation, and antioxidant production. Polyamines are a key [...] Read more.
Cancer metabolic reprogramming is essential for maintaining cancer cell survival and rapid replication. A common target of this metabolic reprogramming is one-carbon metabolism which is notable for its function in DNA synthesis, protein and DNA methylation, and antioxidant production. Polyamines are a key output of one-carbon metabolism with widespread effects on gene expression and signaling. As a result of these functions, one-carbon and polyamine metabolism have recently drawn a lot of interest for their part in cancer malignancy. Therapeutic inhibitors that target one-carbon and polyamine metabolism have thus been trialed as anticancer medications. The significance and future possibilities of one-carbon and polyamine metabolism as a target in cancer therapy are discussed in this review. Full article
(This article belongs to the Special Issue Targeting Tumor Metabolism: From Mechanisms to Therapies II)
Show Figures

Figure 1

48 pages, 2856 KiB  
Review
Druggable Metabolic Vulnerabilities Are Exposed and Masked during Progression to Castration Resistant Prostate Cancer
by Stephen Y. C. Choi, Caroline Fidalgo Ribeiro, Yuzhuo Wang, Massimo Loda, Stephen R. Plymate and Takuma Uo
Biomolecules 2022, 12(11), 1590; https://doi.org/10.3390/biom12111590 - 28 Oct 2022
Cited by 5 | Viewed by 3771
Abstract
There is an urgent need for exploring new actionable targets other than androgen receptor to improve outcome from lethal castration-resistant prostate cancer. Tumor metabolism has reemerged as a hallmark of cancer that drives and supports oncogenesis. In this regard, it is important to [...] Read more.
There is an urgent need for exploring new actionable targets other than androgen receptor to improve outcome from lethal castration-resistant prostate cancer. Tumor metabolism has reemerged as a hallmark of cancer that drives and supports oncogenesis. In this regard, it is important to understand the relationship between distinctive metabolic features, androgen receptor signaling, genetic drivers in prostate cancer, and the tumor microenvironment (symbiotic and competitive metabolic interactions) to identify metabolic vulnerabilities. We explore the links between metabolism and gene regulation, and thus the unique metabolic signatures that define the malignant phenotypes at given stages of prostate tumor progression. We also provide an overview of current metabolism-based pharmacological strategies to be developed or repurposed for metabolism-based therapeutics for castration-resistant prostate cancer. Full article
(This article belongs to the Special Issue Targeting Tumor Metabolism: From Mechanisms to Therapies II)
Show Figures

Figure 1

19 pages, 1491 KiB  
Review
Searching for the Metabolic Signature of Cancer: A Review from Warburg’s Time to Now
by Pierre Jacquet and Angélique Stéphanou
Biomolecules 2022, 12(10), 1412; https://doi.org/10.3390/biom12101412 - 02 Oct 2022
Cited by 7 | Viewed by 2410
Abstract
This review focuses on the evolving understanding that we have of tumor cell metabolism, particularly glycolytic and oxidative metabolism, and traces back its evolution through time. This understanding has developed since the pioneering work of Otto Warburg, but the understanding of tumor cell [...] Read more.
This review focuses on the evolving understanding that we have of tumor cell metabolism, particularly glycolytic and oxidative metabolism, and traces back its evolution through time. This understanding has developed since the pioneering work of Otto Warburg, but the understanding of tumor cell metabolism continues to be hampered by misinterpretation of his work. This has contributed to the use of the new concepts of metabolic switch and metabolic reprogramming, that are out of step with reality. The Warburg effect is often considered to be a hallmark of cancer, but is it really? More generally, is there a metabolic signature of cancer? We draw the conclusion that the signature of cancer cannot be reduced to a single factor, but is expressed at the tissue level in terms of the capacity of cells to dynamically explore a vast metabolic landscape in the context of significant environmental heterogeneities. Full article
(This article belongs to the Special Issue Targeting Tumor Metabolism: From Mechanisms to Therapies II)
Show Figures

Figure 1

Back to TopTop