Tumor Metabolome: Therapeutic Opportunities Targeting Cancer Metabolic Reprogramming

A special issue of Cancers (ISSN 2072-6694).

Deadline for manuscript submissions: closed (31 July 2020) | Viewed by 63412

Special Issue Editors

1. Departamento de Biología Molecular y Bioquímica, Canceromics Lab, Universidad de Málaga, 29071 Málaga, Spain
2. Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Málaga, Spain
Interests: canceromics; cancer metabolic reprogramming; gene therapy; tumor nitrogen metabolism
Special Issues, Collections and Topics in MDPI journals
1. Canceromics Lab, Departmento de Biología Molecular y Bioquímica, Faculty of Sciences, University of Malaga, 29071 Málaga, Spain
2. Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Málaga, Spain
Interests: antioxidant enzymes; cancer metabolism; combined therapies against cancer; oxidative stress; ROS; tumor-specific isoenzymes

Special Issue Information

Dear Colleagues,

Cancer cells have metabolic requirements that separate them from normal cells and render them vulnerable to drugs that target these processes. The altered metabolism exhibited by most tumor cells is a hallmark of cancer. Pioneering works studying the differential metabolic traits of cancer cells firstly classified tumors as glucose and nitrogen traps, thus reflecting their ability to avidly consume both nutrients. In recent years, metabolic studies using omics technologies are more precisely unveiling the tumor metabolome and cancer-specific metabolomic signatures.

Despite the sound advances in knowledge and the rapid progress in the field of cancer metabolic reprogramming, this information needs to be translated into a more successful class of cancer therapeutic strategies. Furthermore, the mechanims that control the metabolic phenotype of specific tumors have not been fully understood, and the biological functions of many cancer-relevant proteins have yet to be explored. These issues need to be addressed in order to improve drug design and to overcome the drug resistance phenomenon frequently found in cancer therapy. In this Special Issue of Cancers, we aim to stimulate discussions on these topics, whilst allowing an updated account of relevant works on the tumor metabolome by bringing together expert opinions from across the field. We welcome submissions that cover any relevant topic, including metabolite addiction in cancer cells, cancer metabolic rewiring, cancer metabolic remodelling by tumor-specific isoenzymes, cancer-specific bionergetic alterations, new metabolic functions of tumor suppresor genes and oncogenes, mechanisms of metabolic dysregulation in cancers, and new therapeutic approaches targeting cancer metabolism.

Prof. Dr. Javier Marquez
Prof. Dr. José M. Matés
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. Cancers is an international peer-reviewed open access semimonthly 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 2900 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

  • Oncometabolites
  • Glutaminolysis
  • Tumor metabolome
  • Cancer metabolic reprogramming
  • Tumor-specific isoenzymes
  • Cancer biomarkers
  • Therapeutic targets

Published Papers (13 papers)

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

Editorial

Jump to: Research, Review, Other

5 pages, 228 KiB  
Editorial
Tumor Metabolome: Therapeutic Opportunities Targeting Cancer Metabolic Reprogramming
by Javier Márquez and José M. Matés
Cancers 2021, 13(2), 314; https://doi.org/10.3390/cancers13020314 - 16 Jan 2021
Cited by 2 | Viewed by 1510
Abstract
The study of cancer metabolism is regaining center stage and becoming a hot topic in tumor biology and clinical research, after a period where such kind of experimental approaches were somehow forgotten or disregarded in favor of powerful functional genomic and proteomic studies [...] Read more.
The study of cancer metabolism is regaining center stage and becoming a hot topic in tumor biology and clinical research, after a period where such kind of experimental approaches were somehow forgotten or disregarded in favor of powerful functional genomic and proteomic studies [...] Full article

Research

Jump to: Editorial, Review, Other

18 pages, 13026 KiB  
Article
Glutamic-Pyruvic Transaminase 1 Facilitates Alternative Fuels for Hepatocellular Carcinoma Growth—A Small Molecule Inhibitor, Berberine
by Wei Guo, Hor-Yue Tan, Sha Li, Ning Wang and Yibin Feng
Cancers 2020, 12(7), 1854; https://doi.org/10.3390/cancers12071854 - 09 Jul 2020
Cited by 20 | Viewed by 3776
Abstract
Metabolic reprogramming is an essential hallmark of cancer. Besides the “Warburg effect”, cancer cells also actively reprogram amino acid metabolism to satisfy high nutritional demands in a nutrient-poor environment. In the glucose–alanine cycle, exogenous alanine taken up by hepatocytes is converted to pyruvate [...] Read more.
Metabolic reprogramming is an essential hallmark of cancer. Besides the “Warburg effect”, cancer cells also actively reprogram amino acid metabolism to satisfy high nutritional demands in a nutrient-poor environment. In the glucose–alanine cycle, exogenous alanine taken up by hepatocytes is converted to pyruvate via glutamic-pyruvic transaminases (GPTs). However, the precise role of the glucose–alanine cycle in hepatocellular carcinoma (HCC) remains elusive. The current study revealed that alanine, as an alternative energy source, induced the metabolic reprogramming of HCC cells via activation of the downstream glucose–alanine cycle and thus promoted HCC growth in nutrient-depleted conditions. Further overexpression and loss-of-function studies indicated that GPT1 was an essential regulator for alanine-supplemented HCC growth. Combining molecular docking and metabolomics analyses, our study further identified a naturally occurring alkaloid, berberine (BBR), as the GPT1 inhibitor in HCC. Mechanically, BBR-mediated metabolic reprogramming of alanine-supplemented HCC via GPT1 suppression attenuated adenosine triphosphate (ATP) production and thus suppressed HCC growth. In conclusion, our study suggests that GPT1-mediated alanine–glucose conversion may be a potential molecular target for HCC therapy. Further demonstration of BBR-mediated metabolic reprogramming of HCC would contribute to the development of this Chinese medicine-derived compound as an adjuvant therapy for HCC. Full article
Show Figures

Graphical abstract

15 pages, 1540 KiB  
Article
Human Prostate Cancer Is Characterized by an Increase in Urea Cycle Metabolites
by Andras Franko, Yaping Shao, Martin Heni, Jörg Hennenlotter, Miriam Hoene, Chunxiu Hu, Xinyu Liu, Xinjie Zhao, Qingqing Wang, Andreas L. Birkenfeld, Tilman Todenhöfer, Arnulf Stenzl, Andreas Peter, Hans-Ulrich Häring, Rainer Lehmann, Guowang Xu and Stefan Z. Lutz
Cancers 2020, 12(7), 1814; https://doi.org/10.3390/cancers12071814 - 06 Jul 2020
Cited by 22 | Viewed by 3254
Abstract
Despite it being the most common incident of cancer among men, the pathophysiological mechanisms contributing to prostate cancer (PCa) are still poorly understood. Altered mitochondrial metabolism is postulated to play a role in the development of PCa. To determine the key metabolites (which [...] Read more.
Despite it being the most common incident of cancer among men, the pathophysiological mechanisms contributing to prostate cancer (PCa) are still poorly understood. Altered mitochondrial metabolism is postulated to play a role in the development of PCa. To determine the key metabolites (which included mitochondrial oncometabolites), benign prostatic and cancer tissues of patients with PCa were analyzed using capillary electrophoresis and liquid chromatography coupled with mass spectrometry. Gene expression was studied using real-time PCR. In PCa tissues, we found reduced levels of early tricarboxylic acid cycle metabolites, whereas the contents of urea cycle metabolites including aspartate, argininosuccinate, arginine, proline, and the oncometabolite fumarate were higher than that in benign controls. Fumarate content correlated positively with the gene expression of oncogenic HIF1α and NFκB pathways, which were significantly higher in the PCa samples than in the benign controls. Furthermore, data from the TCGA database demonstrated that prostate cancer patients with activated NFκB pathway had a lower survival rate. In summary, our data showed that fumarate content was positively associated with carcinogenic genes. Full article
Show Figures

Graphical abstract

12 pages, 1484 KiB  
Article
Metabolic Landscape of a Genetically Engineered Mouse Model of IDH1 Mutant Glioma
by Victor Ruiz-Rodado, Tomohiro Seki, Tyrone Dowdy, Adrian Lita, Meili Zhang, Sue Han, Chunzhang Yang, Murali K. Cherukuri, Mark R. Gilbert and Mioara Larion
Cancers 2020, 12(6), 1633; https://doi.org/10.3390/cancers12061633 - 19 Jun 2020
Cited by 9 | Viewed by 5230
Abstract
Understanding the metabolic reprogramming of aggressive brain tumors has potential applications for therapeutics as well as imaging biomarkers. However, little is known about the nutrient requirements of isocitrate dehydrogenase 1 (IDH1) mutant gliomas. The IDH1 mutation involves the acquisition of a neomorphic enzymatic [...] Read more.
Understanding the metabolic reprogramming of aggressive brain tumors has potential applications for therapeutics as well as imaging biomarkers. However, little is known about the nutrient requirements of isocitrate dehydrogenase 1 (IDH1) mutant gliomas. The IDH1 mutation involves the acquisition of a neomorphic enzymatic activity which generates D-2-hydroxyglutarate from α-ketoglutarate. In order to gain insight into the metabolism of these malignant brain tumors, we conducted metabolic profiling of the orthotopic tumor and the contralateral regions for the mouse model of IDH1 mutant glioma; as well as to examine the utilization of glucose and glutamine in supplying major metabolic pathways such as glycolysis and tricarboxylic acid (TCA). We also revealed that the main substrate of 2-hydroxyglutarate is glutamine in this model, and how this re-routing impairs its utilization in the TCA. Our 13C tracing analysis, along with hyperpolarized magnetic resonance experiments, revealed an active glycolytic pathway similar in both regions (tumor and contralateral) of the brain. Therefore, we describe the reprogramming of the central carbon metabolism associated with the IDH1 mutation in a genetically engineered mouse model which reflects the tumor biology encountered in glioma patients. Full article
Show Figures

Figure 1

17 pages, 4257 KiB  
Article
Metabolic Reprogramming in Metastatic Melanoma with Acquired Resistance to Targeted Therapies: Integrative Metabolomic and Proteomic Analysis
by Laura Soumoy, Corentin Schepkens, Mohammad Krayem, Ahmad Najem, Vanessa Tagliatti, Ghanem E. Ghanem, Sven Saussez, Jean-Marie Colet and Fabrice Journe
Cancers 2020, 12(5), 1323; https://doi.org/10.3390/cancers12051323 - 22 May 2020
Cited by 12 | Viewed by 3522
Abstract
Treatments of metastatic melanoma underwent an impressive development over the past few years, with the emergence of small molecule inhibitors targeting mutated proteins, such as BRAF, NRAS, or cKIT. However, since a significant proportion of patients acquire resistance to these therapies, new strategies [...] Read more.
Treatments of metastatic melanoma underwent an impressive development over the past few years, with the emergence of small molecule inhibitors targeting mutated proteins, such as BRAF, NRAS, or cKIT. However, since a significant proportion of patients acquire resistance to these therapies, new strategies are currently being considered to overcome this issue. For this purpose, melanoma cell lines with mutant BRAF, NRAS, or cKIT and with acquired resistances to BRAF, MEK, or cKIT inhibitors, respectively, were investigated using both 1H-NMR-based metabonomic and protein microarrays. The 1H-NMR profiles highlighted a similar go and return pattern in the metabolism of the BRAF, NRAS, and cKIT mutated cell lines. Indeed, melanoma cells exposed to mutation-specific inhibitors underwent metabolic disruptions following acute exposure but partially recovered their basal metabolism in long-term exposure, most likely acquiring resistance skills. The protein microarrays inquired about the potential cellular mechanisms used by the resistant cells to escape drug treatment, by showing decreased levels of proteins linked to the drug efficacy, especially in the downstream part of the MAPK signaling pathway. Integrating metabonomic and proteomic findings revealed some metabolic pathways (i.e., glutaminolysis, choline metabolism, glutathione production, glycolysis, oxidative phosphorylation) and key proteins (i.e., EPHA2, DUSP4, and HIF-1A) as potential targets to discard drug resistance. Full article
Show Figures

Graphical abstract

15 pages, 4542 KiB  
Article
ACLY (ATP Citrate Lyase) Mediates Radioresistance in Head and Neck Squamous Cell Carcinomas and is a Novel Predictive Radiotherapy Biomarker
by Eva-Leonne Göttgens, Corina NAM van den Heuvel, Monique C de Jong, Johannes HAM Kaanders, William PJ Leenders, Marleen Ansems, Johan Bussink and Paul N Span
Cancers 2019, 11(12), 1971; https://doi.org/10.3390/cancers11121971 - 07 Dec 2019
Cited by 18 | Viewed by 3589
Abstract
Radiotherapy is an important treatment modality of head and neck squamous cell carcinomas (HNSCC). Multiple links have been described between the metabolic activity of tumors and their clinical outcome. Here we test the hypothesis that metabolic features determine radiosensitivity, explaining the relationship between [...] Read more.
Radiotherapy is an important treatment modality of head and neck squamous cell carcinomas (HNSCC). Multiple links have been described between the metabolic activity of tumors and their clinical outcome. Here we test the hypothesis that metabolic features determine radiosensitivity, explaining the relationship between metabolism and clinical outcome. Radiosensitivity of 14 human HNSCC cell lines was determined using colony forming assays and the expression profile of approximately 200 metabolic and cancer-related genes was generated using targeted RNA sequencing by single molecule molecular inversion probes. Results: Correlation between radiosensitivity data and expression profiles yielded 18 genes associated with radiosensitivity or radioresistance, of which adenosine triphosphate (ATP) citrate lyase (ACLY) was of particular interest. Pharmacological inhibition of ACLY caused an impairment of DNA damage repair, specifically homologous recombination, and lead to radiosensitization in HNSCC cell lines. Examination of a The Cancer Genome Atlas (TCGA) cohort of HNSCC patients revealed that high expression of ACLY was predictive for radiotherapy failure, as it was only associated with poor overall survival in patients who received radiotherapy (hazard ratio of 2.00, 95% CI: 1.12–3.55; p = 0.0184). These data were further validated in an independent cohort of HNSCC patients treated with chemoradiation. Furthermore, patients with poor locoregional control after radiotherapy have significantly higher nuclear ACLY protein levels. Together, we here show that ACLY affects DNA damage repair, and is a predictive factor for radiotherapy outcome in HNSCC. Full article
Show Figures

Figure 1

21 pages, 2162 KiB  
Article
The Epithelial to Mesenchymal Transition Promotes Glutamine Independence by Suppressing GLS2 Expression
by Esmeralda Ramirez-Peña, James Arnold, Vinita Shivakumar, Robiya Joseph, Geraldine Vidhya Vijay, Petra den Hollander, Neeraja Bhangre, Paul Allegakoen, Rishika Prasad, Zachary Conley, José M. Matés, Javier Márquez, Jeffrey T. Chang, Suhas Vasaikar, Rama Soundararajan, Arun Sreekumar and Sendurai A. Mani
Cancers 2019, 11(10), 1610; https://doi.org/10.3390/cancers11101610 - 22 Oct 2019
Cited by 25 | Viewed by 5048
Abstract
Identifying bioenergetics that facilitate the epithelial to mesenchymal transition (EMT) in breast cancer cells may uncover targets to treat incurable metastatic disease. Metastasis is the number one cause of cancer-related deaths; therefore, it is urgent to identify new treatment strategies to prevent the [...] Read more.
Identifying bioenergetics that facilitate the epithelial to mesenchymal transition (EMT) in breast cancer cells may uncover targets to treat incurable metastatic disease. Metastasis is the number one cause of cancer-related deaths; therefore, it is urgent to identify new treatment strategies to prevent the initiation of metastasis. To characterize the bioenergetics of EMT, we compared metabolic activities and gene expression in cells induced to differentiate into the mesenchymal state with their epithelial counterparts. We found that levels of GLS2, which encodes a glutaminase, are inversely associated with EMT. GLS2 down-regulation was correlated with reduced mitochondrial activity and glutamine independence even in low-glucose conditions. Restoration of GLS2 expression in GLS2-negative breast cancer cells rescued mitochondrial activity, enhanced glutamine utilization, and inhibited stem-cell properties. Additionally, inhibition of expression of the transcription factor FOXC2, a critical regulator of EMT in GLS2-negative cells, restored GLS2 expression and glutamine utilization. Furthermore, in breast cancer patients, high GLS2 expression is associated with improved survival. These findings suggest that epithelial cancer cells rely on glutamine and that cells induced to undergo EMT become glutamine independent. Moreover, the inhibition of EMT leads to a GLS2-directed metabolic shift in mesenchymal cancer cells, which may make these cells susceptible to chemotherapies. Full article
Show Figures

Graphical abstract

19 pages, 3238 KiB  
Article
HSF1 Regulates Mevalonate and Cholesterol Biosynthesis Pathways
by Hyeji Kang, Taerim Oh, Young Yil Bahk, Geon-Hee Kim, Sang-Yeon Kan, Dong Hoon Shin, Ji Hyung Kim and Ji-Hong Lim
Cancers 2019, 11(9), 1363; https://doi.org/10.3390/cancers11091363 - 13 Sep 2019
Cited by 17 | Viewed by 3749
Abstract
Heat shock factor 1 (HSF1) is an essential transcription factor in cellular adaptation to various stresses such as heat, proteotoxic stress, metabolic stress, reactive oxygen species, and heavy metals. HSF1 promotes cancer development and progression, and increased HSF1 levels are frequently observed in [...] Read more.
Heat shock factor 1 (HSF1) is an essential transcription factor in cellular adaptation to various stresses such as heat, proteotoxic stress, metabolic stress, reactive oxygen species, and heavy metals. HSF1 promotes cancer development and progression, and increased HSF1 levels are frequently observed in multiple types of cancers. Increased activity in the mevalonate and cholesterol biosynthesis pathways, which are very important for cancer growth and progression, is observed in various cancers. However, the functional role of HSF1 in the mevalonate and cholesterol biosynthesis pathways has not yet been investigated. Here, we demonstrated that the activation of RAS-MAPK signaling through the overexpression of H-RasV12 increased HSF1 expression and the cholesterol biosynthesis pathway. In addition, the activation of HSF1 was also found to increase cholesterol biosynthesis. Inversely, the suppression of HSF1 by the pharmacological inhibitor KRIBB11 and short-hairpin RNA (shRNA) reversed H-RasV12-induced cholesterol biosynthesis. From the standpoint of therapeutic applications for hepatocellular carcinoma (HCC) treatment, HSF1 inhibition was shown to sensitize the antiproliferative effects of simvastatin in HCC cells. Overall, our findings demonstrate that HSF1 is a potential target for statin-based HCC treatment. Full article
Show Figures

Figure 1

19 pages, 2289 KiB  
Article
Integrative Metabolomic and Transcriptomic Analysis for the Study of Bladder Cancer
by Alba Loras, Cristian Suárez-Cabrera, M. Carmen Martínez-Bisbal, Guillermo Quintás, Jesús M. Paramio, Ramón Martínez-Máñez, Salvador Gil and José Luis Ruiz-Cerdá
Cancers 2019, 11(5), 686; https://doi.org/10.3390/cancers11050686 - 16 May 2019
Cited by 29 | Viewed by 4342
Abstract
Metabolism reprogramming is considered a hallmark of cancer. The study of bladder cancer (BC) metabolism could be the key to developing new strategies for diagnosis and therapy. This work aimed to identify tissue and urinary metabolic signatures as biomarkers of BC and get [...] Read more.
Metabolism reprogramming is considered a hallmark of cancer. The study of bladder cancer (BC) metabolism could be the key to developing new strategies for diagnosis and therapy. This work aimed to identify tissue and urinary metabolic signatures as biomarkers of BC and get further insight into BC tumor biology through the study of gene-metabolite networks and the integration of metabolomics and transcriptomics data. BC and control tissue samples (n = 44) from the same patients were analyzed by High-Resolution Magic Angle Spinning Nuclear Magnetic Resonance and microarrays techniques. Besides, urinary profiling study (n = 35) was performed in the same patients to identify a metabolomic profile, linked with BC tissue hallmarks, as a potential non-invasive approach for BC diagnosis. The metabolic profile allowed for the classification of BC tissue samples with a sensitivity and specificity of 100%. The most discriminant metabolites for BC tissue samples reflected alterations in amino acids, glutathione, and taurine metabolic pathways. Transcriptomic data supported metabolomic results and revealed a predominant downregulation of metabolic genes belonging to phosphorylative oxidation, tricarboxylic acid cycle, and amino acid metabolism. The urinary profiling study showed a relation with taurine and other amino acids perturbed pathways observed in BC tissue samples, and classified BC from non-tumor urine samples with good sensitivities (91%) and specificities (77%). This urinary profile could be used as a non-invasive tool for BC diagnosis and follow-up. Full article
Show Figures

Figure 1

Review

Jump to: Editorial, Research, Other

27 pages, 925 KiB  
Review
Metabolic Escape Routes of Cancer Stem Cells and Therapeutic Opportunities
by Alice Turdo, Gaetana Porcelli, Caterina D’Accardo, Simone Di Franco, Francesco Verona, Stefano Forte, Dario Giuffrida, Lorenzo Memeo, Matilde Todaro and Giorgio Stassi
Cancers 2020, 12(6), 1436; https://doi.org/10.3390/cancers12061436 - 31 May 2020
Cited by 14 | Viewed by 3722
Abstract
Although improvement in early diagnosis and treatment ameliorated life expectancy of cancer patients, metastatic disease still lacks effective therapeutic approaches. Resistance to anticancer therapies stems from the refractoriness of a subpopulation of cancer cells—termed cancer stem cells (CSCs)—which is endowed with tumor initiation [...] Read more.
Although improvement in early diagnosis and treatment ameliorated life expectancy of cancer patients, metastatic disease still lacks effective therapeutic approaches. Resistance to anticancer therapies stems from the refractoriness of a subpopulation of cancer cells—termed cancer stem cells (CSCs)—which is endowed with tumor initiation and metastasis formation potential. CSCs are heterogeneous and diverge by phenotypic, functional and metabolic perspectives. Intrinsic as well as extrinsic stimuli dictated by the tumor microenvironment (TME)have critical roles in determining cell metabolic reprogramming from glycolytic toward an oxidative phenotype and vice versa, allowing cancer cells to thrive in adverse milieus. Crosstalk between cancer cells and the surrounding microenvironment occurs through the interchange of metabolites, miRNAs and exosomes that drive cancer cells metabolic adaptation. Herein, we identify the metabolic nodes of CSCs and discuss the latest advances in targeting metabolic demands of both CSCs and stromal cells with the scope of improving current therapies and preventing cancer progression. Full article
Show Figures

Figure 1

24 pages, 968 KiB  
Review
Targeting Glutamine Addiction in Gliomas
by Marta Obara-Michlewska and Monika Szeliga
Cancers 2020, 12(2), 310; https://doi.org/10.3390/cancers12020310 - 29 Jan 2020
Cited by 58 | Viewed by 5722
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 [...] Read more.
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. Full article
Show Figures

Figure 1

21 pages, 1147 KiB  
Review
Lactate Dehydrogenases as Metabolic Links between Tumor and Stroma in the Tumor Microenvironment
by Deepshikha Mishra and Debabrata Banerjee
Cancers 2019, 11(6), 750; https://doi.org/10.3390/cancers11060750 - 29 May 2019
Cited by 160 | Viewed by 13151 | Correction
Abstract
Cancer is a metabolic disease in which abnormally proliferating cancer cells rewire metabolic pathways in the tumor microenvironment (TME). Molecular reprogramming in the TME helps cancer cells to fulfill elevated metabolic demands for bioenergetics and cellular biosynthesis. One of the ways through which [...] Read more.
Cancer is a metabolic disease in which abnormally proliferating cancer cells rewire metabolic pathways in the tumor microenvironment (TME). Molecular reprogramming in the TME helps cancer cells to fulfill elevated metabolic demands for bioenergetics and cellular biosynthesis. One of the ways through which cancer cell achieve this is by regulating the expression of metabolic enzymes. Lactate dehydrogenase (LDH) is the primary metabolic enzyme that converts pyruvate to lactate and vice versa. LDH also plays a significant role in regulating nutrient exchange between tumor and stroma. Thus, targeting human lactate dehydrogenase for treating advanced carcinomas may be of benefit. LDHA and LDHB, two isoenzymes of LDH, participate in tumor stroma metabolic interaction and exchange of metabolic fuel and thus could serve as potential anticancer drug targets. This article reviews recent research discussing the roles of lactate dehydrogenase in cancer metabolism. As molecular regulation of LDHA and LDHB in different cancer remains obscure, we also review signaling pathways regulating LDHA and LDHB expression. We highlight on the role of small molecule inhibitors in targeting LDH activity and we emphasize the development of safer and more effective LDH inhibitors. We trust that this review will also generate interest in designing combination therapies based on LDH inhibition, with LDHA being targeted in tumors and LDHB in stromal cells for better treatment outcome. Full article
Show Figures

Graphical abstract

Other

27 pages, 2885 KiB  
Perspective
Tumor Cell-Intrinsic Immunometabolism and Precision Nutrition in Cancer Immunotherapy
by Elisabet Cuyàs, Sara Verdura, Begoña Martin-Castillo, Tomás Alarcón, Ruth Lupu, Joaquim Bosch-Barrera and Javier A. Menendez
Cancers 2020, 12(7), 1757; https://doi.org/10.3390/cancers12071757 - 02 Jul 2020
Cited by 16 | Viewed by 5874
Abstract
One of the greatest challenges in the cancer immunotherapy field is the need to biologically rationalize and broaden the clinical utility of immune checkpoint inhibitors (ICIs). The balance between metabolism and immune response has critical implications for overcoming the major weaknesses of ICIs, [...] Read more.
One of the greatest challenges in the cancer immunotherapy field is the need to biologically rationalize and broaden the clinical utility of immune checkpoint inhibitors (ICIs). The balance between metabolism and immune response has critical implications for overcoming the major weaknesses of ICIs, including their lack of universality and durability. The last decade has seen tremendous advances in understanding how the immune system’s ability to kill tumor cells requires the conspicuous metabolic specialization of T-cells. We have learned that cancer cell-associated metabolic activities trigger shifts in the abundance of some metabolites with immunosuppressory roles in the tumor microenvironment. Yet very little is known about the tumor cell-intrinsic metabolic traits that control the immune checkpoint contexture in cancer cells. Likewise, we lack a comprehensive understanding of how systemic metabolic perturbations in response to dietary interventions can reprogram the immune checkpoint landscape of tumor cells. We here review state-of-the-art molecular- and functional-level interrogation approaches to uncover how cell-autonomous metabolic traits and diet-mediated changes in nutrient availability and utilization might delineate new cancer cell-intrinsic metabolic dependencies of tumor immunogenicity. We propose that clinical monitoring and in-depth molecular evaluation of the cancer cell-intrinsic metabolic traits involved in primary, adaptive, and acquired resistance to cancer immunotherapy can provide the basis for improvements in therapeutic responses to ICIs. Overall, these approaches might guide the use of metabolic therapeutics and dietary approaches as novel strategies to broaden the spectrum of cancer patients and indications that can be effectively treated with ICI-based cancer immunotherapy. Full article
Show Figures

Graphical abstract

Back to TopTop