New Aspects of Targeting Cancer Metabolism in Therapeutic Approach

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cellular Metabolism".

Deadline for manuscript submissions: closed (31 March 2022) | Viewed by 103331

Special Issue Editor


E-Mail Website
Guest Editor
National Cancer Center, Gyeonggi, Goyang, Korea
Interests: cancer metabolism; cancer energy metabolism; cancer therapeutic targets; cancer therapeutics development; anti-cancer drugs; transglutaminase

Special Issue Information

Dear Colleagues,

Cancer-specific metabolism was discovered about 90 years ago, now known as the Warburg effect. Since then, many researchers looking for a cure to cancer have been thwarted, because most biochemical metabolic pathways had not been discovered at the time.

Recently, cancer therapy has made a significant change by heading toward regulating the immune system, despite the fact that most cancers are not induced by mutation of the immune system. This implies a very important shift in focus, from what causes cancer to how we can cure cancer. The real matter resides in the question of how we can distinguish cancer cells from normal cells.

Cancer metabolism is quickly becoming a major drug target for the treatment of a variety of cancers. Cancer-specific metabolic inhibitor enasidenib has been approved for acute myeloid leukemia therapy (2017) by the US FDA and will likely continue to expand. A series of studies on cancer specific metabolic dependency may find a use for the list of metabolic inhibitors as therapeutic agents. That will be the ultimate answer for how we can kill only cancer cells when systemically mixed with normal cells.

This Special Issue focuses on the connection between cancer-specific metabolism and its possibility as a therapeutic target, with an emphasis on novel inhibitors and new therapeutic possibilities targeting metabolic pathways.

Dr. Soo-Youl Kim
Guest Editor

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. Cells 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 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

  • cancer metabolism
  • anticancer drug
  • tumor microenvironment
  • cancer therapy
  • cancer therapeutic target
  • cancer anabolism
  • cancer catabolism
  • cancer mitochondria
  • cancer energy metabolism
  • cancer metabolomics

Published Papers (18 papers)

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

Research

Jump to: Review

17 pages, 1643 KiB  
Article
A Heme-Binding Transcription Factor BACH1 Regulates Lactate Catabolism Suggesting a Combined Therapy for Triple-Negative Breast Cancer
by Joselyn Padilla, Bok-Soon Lee, Karen Zhai, Bethany Rentz, Tia Bobo, Norca Maritza Dowling and Jiyoung Lee
Cells 2022, 11(7), 1177; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11071177 - 31 Mar 2022
Cited by 7 | Viewed by 2919
Abstract
The oncogenic expression or mutation of tumor suppressors drives metabolic alteration, causing cancer cells to utilize diverse nutrients. Lactate is a known substrate for cancer cells, yet the regulatory mechanisms of lactate catabolism are limited. Here, we show that a heme-binding transcription factor, [...] Read more.
The oncogenic expression or mutation of tumor suppressors drives metabolic alteration, causing cancer cells to utilize diverse nutrients. Lactate is a known substrate for cancer cells, yet the regulatory mechanisms of lactate catabolism are limited. Here, we show that a heme-binding transcription factor, BACH1, negatively regulates lactate catabolic pathways in triple-negative breast cancer (TNBC) cells. BACH1 suppresses the transcriptional expression of monocarboxylate transporter 1 (MCT1) and lactate dehydrogenase B, inhibiting lactate-mediated mitochondrial metabolism. In our studies, the depletion of BACH1 either genetically or pharmacologically increased the lactate use of TNBC cells, increasing their sensitivity to MCT1 inhibition. Thus, small inhibitory molecules (SR13800 and AZD3965) blocking MCT1 better suppressed the growth of BACH1-depleted TNBC cells than did the controls. Particularly, hemin treatment degrading BACH1 proteins induced lactate catabolism in TNBC cells, generating synthetic lethality with MCT1 inhibition. Our data indicates that targeting BACH1 generates metabolic vulnerability and increases sensitivity to lactate transporter inhibition, suggesting a potential novel combination therapy for cancer patients with TNBC. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

16 pages, 28830 KiB  
Article
Targeting PGM3 as a Novel Therapeutic Strategy in KRAS/LKB1 Co-Mutant Lung Cancer
by Hyunmin Lee, Feng Cai, Neil Kelekar, Nipun K. Velupally and Jiyeon Kim
Cells 2022, 11(1), 176; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11010176 - 05 Jan 2022
Cited by 10 | Viewed by 3885
Abstract
In non-small-cell lung cancer (NSCLC), concurrent mutations in the oncogene KRAS and tumor suppressor STK11 (also known as LKB1) confer an aggressive malignant phenotype, an unfavourability towards immunotherapy, and overall poor prognoses in patients. In a previous study, we showed that murine KRAS/LKB1 [...] Read more.
In non-small-cell lung cancer (NSCLC), concurrent mutations in the oncogene KRAS and tumor suppressor STK11 (also known as LKB1) confer an aggressive malignant phenotype, an unfavourability towards immunotherapy, and overall poor prognoses in patients. In a previous study, we showed that murine KRAS/LKB1 co-mutant tumors and human co-mutant cancer cells have an enhanced dependence on glutamine-fructose-6-phosphate transaminase 2 (GFPT2), a rate-limiting enzyme in the hexosamine biosynthesis pathway (HBP), which could be targeted to reduce survival of KRAS/LKB1 co-mutants. Here, we found that KRAS/LKB1 co-mutant cells also exhibit an increased dependence on N-acetylglucosamine-phosphate mutase 3 (PGM3), an enzyme downstream of GFPT2. Genetic or pharmacologic suppression of PGM3 reduced KRAS/LKB1 co-mutant tumor growth in both in vitro and in vivo settings. Our results define an additional metabolic vulnerability in KRAS/LKB1 co-mutant tumors to the HBP and provide a rationale for targeting PGM3 in this aggressive subtype of NSCLC. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

16 pages, 4884 KiB  
Article
Oxidized Phospholipids in Tumor Microenvironment Stimulate Tumor Metastasis via Regulation of Autophagy
by Jin Kyung Seok, Eun-Hee Hong, Gabsik Yang, Hye Eun Lee, Sin-Eun Kim, Kwang-Hyeon Liu, Han Chang Kang, Yong-Yeon Cho, Hye Suk Lee and Joo Young Lee
Cells 2021, 10(3), 558; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10030558 - 04 Mar 2021
Cited by 9 | Viewed by 2967
Abstract
Oxidized phospholipids are well known to play physiological and pathological roles in regulating cellular homeostasis and disease progression. However, their role in cancer metastasis has not been entirely understood. In this study, effects of oxidized phosphatidylcholines such as 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC) on [...] Read more.
Oxidized phospholipids are well known to play physiological and pathological roles in regulating cellular homeostasis and disease progression. However, their role in cancer metastasis has not been entirely understood. In this study, effects of oxidized phosphatidylcholines such as 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC) on epithelial-mesenchymal transition (EMT) and autophagy were determined in cancer cells by immunoblotting and confocal analysis. Metastasis was analyzed by a scratch wound assay and a transwell migration/invasion assay. The concentrations of POVPC and 1-palmitoyl-2-glutaroyl-sn-glycero-phosphocholine (PGPC) in tumor tissues obtained from patients were measured by LC-MS/MS analysis. POVPC induced EMT, resulting in increase of migration and invasion of human hepatocellular carcinoma cells (HepG2) and human breast cancer cells (MCF7). POVPC induced autophagic flux through AMPK-mTOR pathway. Pharmacological inhibition or siRNA knockdown of autophagy decreased migration and invasion of POVPC-treated HepG2 and MCF7 cells. POVPC and PGPC levels were greatly increased at stage II of patient-derived intrahepatic cholangiocarcinoma tissues. PGPC levels were higher in malignant breast tumor tissues than in adjacent nontumor tissues. The results show that oxidized phosphatidylcholines increase metastatic potential of cancer cells by promoting EMT, mediated through autophagy. These suggest the positive regulatory role of oxidized phospholipids accumulated in tumor microenvironment in the regulation of tumorigenesis and metastasis. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

21 pages, 5301 KiB  
Article
Deciphering Fatty Acid Synthase Inhibition-Triggered Metabolic Flexibility in Prostate Cancer Cells through Untargeted Metabolomics
by Ju Eun Oh, Byung Hwa Jung, Jinyoung Park, Soosung Kang and Hyunbeom Lee
Cells 2020, 9(11), 2447; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9112447 - 10 Nov 2020
Cited by 12 | Viewed by 2817
Abstract
Fatty acid synthase (FAS) is a key enzyme involved in de novo lipogenesis that produces lipids that are necessary for cell growth and signal transduction, and it is known to be overexpressed, especially in cancer cells. Although lipid metabolism alteration is an important [...] Read more.
Fatty acid synthase (FAS) is a key enzyme involved in de novo lipogenesis that produces lipids that are necessary for cell growth and signal transduction, and it is known to be overexpressed, especially in cancer cells. Although lipid metabolism alteration is an important metabolic phenotype in cancer cells, the development of drugs targeting FAS to block lipid synthesis is hampered by the characteristics of cancer cells with metabolic flexibility leading to rapid adaptation and resistance. Therefore, to confirm the metabolic alterations at the cellular level during FAS inhibition, we treated LNCaP-LN3 prostate cancer cells with FAS inhibitors (Fasnall, GSK2194069, and TVB-3166). With untargeted metabolomics, we observed significant changes in a total of 56 metabolites in the drug-treated groups. Among the altered metabolites, 28 metabolites were significantly changed in all of the drug-treated groups. To our surprise, despite the inhibition of FAS, which is involved in palmitate production, the cells increase their fatty acids and glycerophospholipids contents endogenously. Also, some of the notable changes in the metabolic pathways include polyamine metabolism and energy metabolism. This is the first study to compare and elucidate the effect of FAS inhibition on cellular metabolic flexibility using three different FAS inhibitors through metabolomics. We believe that our results may provide key data for the development of future FAS-targeting drugs. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Graphical abstract

13 pages, 4035 KiB  
Article
Breast Cancer Subtypes Underlying EMT-Mediated Catabolic Metabolism
by Eunae Sandra Cho, Nam Hee Kim, Jun Seop Yun, Sue Bean Cho, Hyun Sil Kim and Jong In Yook
Cells 2020, 9(9), 2064; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9092064 - 10 Sep 2020
Cited by 14 | Viewed by 3724
Abstract
Efficient catabolic metabolism of adenosine triphosphate (ATP) and reduced nicotinamide adenine dinucleotide phosphate (NADPH) is essentially required for cancer cell survival, especially in metastatic cancer progression. Epithelial–mesenchymal transition (EMT) plays an important role in metabolic rewiring of cancer cells as well as in [...] Read more.
Efficient catabolic metabolism of adenosine triphosphate (ATP) and reduced nicotinamide adenine dinucleotide phosphate (NADPH) is essentially required for cancer cell survival, especially in metastatic cancer progression. Epithelial–mesenchymal transition (EMT) plays an important role in metabolic rewiring of cancer cells as well as in phenotypic conversion and therapeutic resistance. Snail (SNAI1), a well-known inducer of cancer EMT, is critical in providing ATP and NADPH via suppression of several gatekeeper genes involving catabolic metabolism, such as phosphofructokinase 1 (PFK1), fructose-1,6-bisphosphatase 1 (FBP1), and acetyl-CoA carboxylase 2 (ACC2). Paradoxically, PFK1 and FBP1 are counter-opposing and rate-limiting reaction enzymes of glycolysis and gluconeogenesis, respectively. In this study, we report a distinct metabolic circuit of catabolic metabolism in breast cancer subtypes. Interestingly, PFKP and FBP1 are inversely correlated in clinical samples, indicating different metabolic subsets of breast cancer. The luminal types of breast cancer consist of the pentose phosphate pathway (PPP) subset by suppression of PFKP while the basal-like subtype (also known as triple negative breast cancer, TNBC) mainly utilizes glycolysis and mitochondrial fatty acid oxidation (FAO) by loss of FBP1 and ACC2. Notably, PPP remains active via upregulation of TIGAR in the FBP1-loss basal-like subset, indicating the importance of PPP in catabolic cancer metabolism. These results indicate different catabolic metabolic circuits and thus therapeutic strategies in breast cancer subsets. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Graphical abstract

17 pages, 2411 KiB  
Article
Targeting Oxidative Phosphorylation Reverses Drug Resistance in Cancer Cells by Blocking Autophagy Recycling
by Jae-Seon Lee, Ho Lee, Hyonchol Jang, Sang Myung Woo, Jong Bae Park, Seon-Hyeong Lee, Joon Hee Kang, Hee Yeon Kim, Jaewhan Song and Soo-Youl Kim
Cells 2020, 9(9), 2013; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9092013 - 01 Sep 2020
Cited by 26 | Viewed by 4812
Abstract
The greatest challenge in cancer therapy is posed by drug-resistant recurrence following treatment. Anticancer chemotherapy is largely focused on targeting the rapid proliferation and biosynthesis of cancer cells. This strategy has the potential to trigger autophagy, enabling cancer cell survival through the recycling [...] Read more.
The greatest challenge in cancer therapy is posed by drug-resistant recurrence following treatment. Anticancer chemotherapy is largely focused on targeting the rapid proliferation and biosynthesis of cancer cells. This strategy has the potential to trigger autophagy, enabling cancer cell survival through the recycling of molecules and energy essential for biosynthesis, leading to drug resistance. Autophagy recycling contributes amino acids and ATP to restore mTOR complex 1 (mTORC1) activity, which leads to cell survival. However, autophagy with mTORC1 activation can be stalled by reducing the ATP level. We have previously shown that cytosolic NADH production supported by aldehyde dehydrogenase (ALDH) is critical for supplying ATP through oxidative phosphorylation (OxPhos) in cancer cell mitochondria. Inhibitors of the mitochondrial complex I of the OxPhos electron transfer chain and ALDH significantly reduce the ATP level selectively in cancer cells, terminating autophagy triggered by anticancer drug treatment. With the aim of overcoming drug resistance, we investigated combining the inhibition of mitochondrial complex I, using phenformin, and ALDH, using gossypol, with anticancer drug treatment. Here, we show that OxPhos targeting combined with anticancer drugs acts synergistically to enhance the anticancer effect in mouse xenograft models of various cancers, which suggests a potential therapeutic approach for drug-resistant cancer. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

15 pages, 3339 KiB  
Article
Disruption of Cancer Metabolic SREBP1/miR-142-5p Suppresses Epithelial–Mesenchymal Transition and Stemness in Esophageal Carcinoma
by Chih-Ming Huang, Chin-Sheng Huang, Tung-Nien Hsu, Mao-Suan Huang, Iat-Hang Fong, Wei-Hwa Lee and Shao-Cheng Liu
Cells 2020, 9(1), 7; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9010007 - 18 Dec 2019
Cited by 23 | Viewed by 3896
Abstract
Elevated activity of sterol regulatory element-binding protein 1 (SREBP1) has been implicated in the tumorigenesis of different cancer types. However, the functional roles of SREBP1 in esophageal cancer are not well appreciated. Here, we aimed to investigate the therapeutic potential of SREBP1 and [...] Read more.
Elevated activity of sterol regulatory element-binding protein 1 (SREBP1) has been implicated in the tumorigenesis of different cancer types. However, the functional roles of SREBP1 in esophageal cancer are not well appreciated. Here, we aimed to investigate the therapeutic potential of SREBP1 and associated signaling in esophageal cancer. Our initial bioinformatics analyses showed that SREBP1 expression was overexpressed in esophageal tumors and correlated with a significantly lower overall survival rate in patients. Additionally, tumor suppressor miR-142-5p was predicted to target SREBP1/ZEB1 and a lower miR-142-5p was correlated with poor prognosis. We then performed in vitro experiments and showed that overexpressing SREBP1 in OE33 cell line led to increased abilities of colony formation, migration, and invasion; the opposite was observed in SREBP1-silenced OE21cells and SREBP1-silencing was accompanied by the reduced mesenchymal markers, including vimentin (Vim) and ZEB1, while E-cadherin and tumor suppressor miR-142-5p were increased. Subsequently, we first demonstrated that both SREBP1 and ZEB1 were potential targets of miR-142-5p, followed by the examination of the regulatory circuit of miR-142-5p and SREBP1/ZEB1. We observed that increased miR-142-5p level led to the reduced tumorigenic properties, such as migration and tumor sphere formation, and both observations were accompanied by the reduction of ZEB1 and SREBP1, and increase of E-cadherin. We then explored the potential therapeutic agent targeting SREBP1-associated signaling by testing fatostatin (4-hydroxytamoxifen, an active metabolite of tamoxifen). We found that fatostatin suppressed the cell viability of OE21 and OE33 cells and tumor spheres. Interestingly, fatostatin treatment reduced CD133+ population in both OE21 and OE33 cells in concert of increased miR-142-5p level. Finally, we evaluated the efficacy of fatostatin using a xenograft mouse model. Mice treated with fatostatin showed a significantly lower tumor burden and better survival rate as compared to their control counterparts. The treatment of fatostatin resulted in the reduced staining of SREBP1, ZEB1, and Vim, while E-cadherin and miR-142-5p were increased. In summary, we showed that increased SREBP1 and reduced miR-142-5p were associated with increased tumorigenic properties of esophageal cancer cells and poor prognosis. Preclinical tests showed that suppression of SREBP1 using fatostatin led to the reduced malignant phenotype of esophageal cancer via the reduction of EMT markers and increased tumor suppressor, miR-142-5p. Further investigation is warranted for the clinical use of fatostatin for the treatment of esophageal malignancy. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Graphical abstract

Review

Jump to: Research

17 pages, 392 KiB  
Review
Obesity-Associated Cancers: Evidence from Studies in Mouse Models
by Ho Lee
Cells 2022, 11(9), 1472; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11091472 - 27 Apr 2022
Cited by 8 | Viewed by 2802
Abstract
Obesity, one of the major problems in modern human society, is correlated with various diseases, including type 2 diabetes mellitus (T2DM). In particular, epidemiological and experimental evidence indicates that obesity is closely linked to at least 13 different types of cancer. The mechanisms [...] Read more.
Obesity, one of the major problems in modern human society, is correlated with various diseases, including type 2 diabetes mellitus (T2DM). In particular, epidemiological and experimental evidence indicates that obesity is closely linked to at least 13 different types of cancer. The mechanisms that potentially explain the link between obesity and cancer include hyperactivation of the IGF pathway, metabolic dysregulation, dysfunctional angiogenesis, chronic inflammation, and interaction between pro-inflammatory cytokines, endocrine hormones, and adipokines. However, how the largely uniform morbidity of obesity leads to different types of cancer still needs to be investigated. To study the link between obesity and cancer, researchers have commonly used preclinical animal models, particularly mouse models. These models include monogenic models of obesity (e.g., ob/ob and db/db mice) and genetically modified mouse models of human cancers (e.g., Kras-driven pancreatic cancer, Apc-mutated colorectal cancer, and Her2/neu-overexpressing breast cancer). The experimental results obtained using these mouse models revealed strong evidence of a link between obesity and cancer and suggested their underlying mechanisms. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

21 pages, 1949 KiB  
Review
Role of Mitochondrial Stress Response in Cancer Progression
by Yu Geon Lee, Do Hong Park and Young Chan Chae
Cells 2022, 11(5), 771; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11050771 - 23 Feb 2022
Cited by 20 | Viewed by 5282
Abstract
Mitochondria are subcellular organelles that are a hub for key biological processes, such as bioenergetic, biosynthetic, and signaling functions. Mitochondria are implicated in all oncogenic processes, from malignant transformation to metastasis and resistance to chemotherapeutics. The harsh tumor environment constantly exposes cancer cells [...] Read more.
Mitochondria are subcellular organelles that are a hub for key biological processes, such as bioenergetic, biosynthetic, and signaling functions. Mitochondria are implicated in all oncogenic processes, from malignant transformation to metastasis and resistance to chemotherapeutics. The harsh tumor environment constantly exposes cancer cells to cytotoxic stressors, such as nutrient starvation, low oxygen, and oxidative stress. Excessive or prolonged exposure to these stressors can cause irreversible mitochondrial damage, leading to cell death. To survive hostile microenvironments that perturb mitochondrial function, cancer cells activate a stress response to maintain mitochondrial protein and genome integrity. This adaptive mechanism, which is closely linked to mitochondrial function, enables rapid adjustment and survival in harsh environmental conditions encountered during tumor dissemination, thereby promoting cancer progression. In this review, we describe how the mitochondria stress response contributes to the acquisition of typical malignant traits and highlight the potential of targeting the mitochondrial stress response as an anti-cancer therapeutic strategy. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

17 pages, 843 KiB  
Review
New Immunometabolic Strategy Based on Cell Type-Specific Metabolic Reprogramming in the Tumor Immune Microenvironment
by Ji-Yong Sung and Jae-Ho Cheong
Cells 2022, 11(5), 768; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11050768 - 22 Feb 2022
Cited by 14 | Viewed by 5459
Abstract
Immunometabolism is an emerging discipline in cancer immunotherapy. Tumor tissues are heterogeneous and influenced by metabolic reprogramming of the tumor immune microenvironment (TIME). In the TIME, multiple cell types interact, and the tumor and immune cells compete for limited nutrients, resulting in altered [...] Read more.
Immunometabolism is an emerging discipline in cancer immunotherapy. Tumor tissues are heterogeneous and influenced by metabolic reprogramming of the tumor immune microenvironment (TIME). In the TIME, multiple cell types interact, and the tumor and immune cells compete for limited nutrients, resulting in altered anticancer immunity. Therefore, metabolic reprogramming of individual cell types may influence the outcomes of immunotherapy. Understanding the metabolic competition for access to limited nutrients between tumor cells and immune cells could reveal the breadth and complexity of the TIME and aid in developing novel therapeutic approaches for cancer. In this review, we highlight that, when cells compete for nutrients, the prevailing cell type gains certain advantages over other cell types; for instance, if tumor cells prevail against immune cells for nutrients, the former gains immune resistance. Thus, a strategy is needed to selectively suppress such resistant tumor cells. Although challenging, the concept of cell type-specific metabolic pathway inhibition is a potent new strategy in anticancer immunotherapy. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Graphical abstract

27 pages, 7526 KiB  
Review
Amino Acid Metabolism in Cancer Drug Resistance
by Hee-Chan Yoo and Jung-Min Han
Cells 2022, 11(1), 140; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11010140 - 02 Jan 2022
Cited by 36 | Viewed by 8511
Abstract
Despite the numerous investigations on resistance mechanisms, drug resistance in cancer therapies still limits favorable outcomes in cancer patients. The complexities of the inherent characteristics of tumors, such as tumor heterogeneity and the complicated interaction within the tumor microenvironment, still hinder efforts to [...] Read more.
Despite the numerous investigations on resistance mechanisms, drug resistance in cancer therapies still limits favorable outcomes in cancer patients. The complexities of the inherent characteristics of tumors, such as tumor heterogeneity and the complicated interaction within the tumor microenvironment, still hinder efforts to overcome drug resistance in cancer cells, requiring innovative approaches. In this review, we describe recent studies offering evidence for the essential roles of amino acid metabolism in driving drug resistance in cancer cells. Amino acids support cancer cells in counteracting therapies by maintaining redox homeostasis, sustaining biosynthetic processes, regulating epigenetic modification, and providing metabolic intermediates for energy generation. In addition, amino acid metabolism impacts anticancer immune responses, creating an immunosuppressive or immunoeffective microenvironment. A comprehensive understanding of amino acid metabolism as it relates to therapeutic resistance mechanisms will improve anticancer therapeutic strategies. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

24 pages, 1318 KiB  
Review
Oxidative Stress and the Intersection of Oncogenic Signaling and Metabolism in Squamous Cell Carcinomas
by Joshua H. Choe, Simbarashe Mazambani, Tae Hoon Kim and Jung-whan Kim
Cells 2021, 10(3), 606; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10030606 - 09 Mar 2021
Cited by 3 | Viewed by 3992
Abstract
Squamous cell carcinomas (SCCs) arise from both stratified squamous and non-squamous epithelium of diverse anatomical sites and collectively represent one of the most frequent solid tumors, accounting for more than one million cancer deaths annually. Despite this prevalence, SCC patients have not fully [...] Read more.
Squamous cell carcinomas (SCCs) arise from both stratified squamous and non-squamous epithelium of diverse anatomical sites and collectively represent one of the most frequent solid tumors, accounting for more than one million cancer deaths annually. Despite this prevalence, SCC patients have not fully benefited from recent advances in molecularly targeted therapy or immunotherapy. Rather, decades old platinum-based or radiation regimens retaining limited specificity to the unique characteristics of SCC remain first-line treatment options. Historically, a lack of a consolidated perspective on genetic aberrations driving oncogenic transformation and other such factors essential for SCC pathogenesis and intrinsic confounding cellular heterogeneity in SCC have contributed to a critical dearth in effective and specific therapies. However, emerging evidence characterizing the distinct genomic, epigenetic, and metabolic landscapes of SCC may be elucidating unifying features in a seemingly heterogeneous disease. In this review, by describing distinct metabolic alterations and genetic drivers of SCC revealed by recent studies, we aim to establish a conceptual framework for a previously unappreciated network of oncogenic signaling, redox perturbation, and metabolic reprogramming that may reveal targetable vulnerabilities at their intersection. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

33 pages, 2376 KiB  
Review
Roles of Farnesyl-Diphosphate Farnesyltransferase 1 in Tumour and Tumour Microenvironments
by Nguyen Thi Ha and Chang Hoon Lee
Cells 2020, 9(11), 2352; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9112352 - 25 Oct 2020
Cited by 32 | Viewed by 5111
Abstract
Farnesyl-diphosphate farnesyltransferase 1 (FDFT1, squalene synthase), a membrane-associated enzyme, synthesizes squalene via condensation of two molecules of farnesyl pyrophosphate. Accumulating evidence has noted that FDFT1 plays a critical role in cancer, particularly in metabolic reprogramming, cell proliferation, and invasion. Based on these advances [...] Read more.
Farnesyl-diphosphate farnesyltransferase 1 (FDFT1, squalene synthase), a membrane-associated enzyme, synthesizes squalene via condensation of two molecules of farnesyl pyrophosphate. Accumulating evidence has noted that FDFT1 plays a critical role in cancer, particularly in metabolic reprogramming, cell proliferation, and invasion. Based on these advances in our knowledge, FDFT1 could be a potential target for cancer treatment. This review focuses on the contribution of FDFT1 to the hallmarks of cancer, and further, we discuss the applicability of FDFT1 as a cancer prognostic marker and target for anticancer therapy. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

31 pages, 1552 KiB  
Review
Cancer Metabolism: Phenotype, Signaling and Therapeutic Targets
by Jae Hyung Park, Woo Yang Pyun and Hyun Woo Park
Cells 2020, 9(10), 2308; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9102308 - 16 Oct 2020
Cited by 202 | Viewed by 19302
Abstract
Aberrant metabolism is a major hallmark of cancer. Abnormal cancer metabolism, such as aerobic glycolysis and increased anabolic pathways, has important roles in tumorigenesis, metastasis, drug resistance, and cancer stem cells. Well-known oncogenic signaling pathways, such as phosphoinositide 3-kinase (PI3K)/AKT, Myc, and Hippo [...] Read more.
Aberrant metabolism is a major hallmark of cancer. Abnormal cancer metabolism, such as aerobic glycolysis and increased anabolic pathways, has important roles in tumorigenesis, metastasis, drug resistance, and cancer stem cells. Well-known oncogenic signaling pathways, such as phosphoinositide 3-kinase (PI3K)/AKT, Myc, and Hippo pathway, mediate metabolic gene expression and increase metabolic enzyme activities. Vice versa, deregulated metabolic pathways contribute to defects in cellular signal transduction pathways, which in turn provide energy, building blocks, and redox potentials for unrestrained cancer cell proliferation. Studies and clinical trials are being performed that focus on the inhibition of metabolic enzymes by small molecules or dietary interventions (e.g., fasting, calorie restriction, and intermittent fasting). Similar to genetic heterogeneity, the metabolic phenotypes of cancers are highly heterogeneous. This heterogeneity results from diverse cues in the tumor microenvironment and genetic mutations. Hence, overcoming metabolic plasticity is an important goal of modern cancer therapeutics. This review highlights recent findings on the metabolic phenotypes of cancer and elucidates the interactions between signal transduction pathways and metabolic pathways. We also provide novel rationales for designing the next-generation cancer metabolism drugs. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

37 pages, 1316 KiB  
Review
Oncology Therapeutics Targeting the Metabolism of Amino Acids
by Nefertiti Muhammad, Hyun Min Lee and Jiyeon Kim
Cells 2020, 9(8), 1904; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9081904 - 15 Aug 2020
Cited by 26 | Viewed by 6977
Abstract
Amino acid metabolism promotes cancer cell proliferation and survival by supporting building block synthesis, producing reducing agents to mitigate oxidative stress, and generating immunosuppressive metabolites for immune evasion. Malignant cells rewire amino acid metabolism to maximize their access to nutrients. Amino acid transporter [...] Read more.
Amino acid metabolism promotes cancer cell proliferation and survival by supporting building block synthesis, producing reducing agents to mitigate oxidative stress, and generating immunosuppressive metabolites for immune evasion. Malignant cells rewire amino acid metabolism to maximize their access to nutrients. Amino acid transporter expression is upregulated to acquire amino acids from the extracellular environment. Under nutrient depleted conditions, macropinocytosis can be activated where proteins from the extracellular environment are engulfed and degraded into the constituent amino acids. The demand for non-essential amino acids (NEAAs) can be met through de novo synthesis pathways. Cancer cells can alter various signaling pathways to boost amino acid usage for the generation of nucleotides, reactive oxygen species (ROS) scavenging molecules, and oncometabolites. The importance of amino acid metabolism in cancer proliferation makes it a potential target for therapeutic intervention, including via small molecules and antibodies. In this review, we will delineate the targets related to amino acid metabolism and promising therapeutic approaches. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

19 pages, 1135 KiB  
Review
Peroxisome Metabolism in Cancer
by Jung-Ae Kim
Cells 2020, 9(7), 1692; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9071692 - 14 Jul 2020
Cited by 53 | Viewed by 7518
Abstract
Peroxisomes are metabolic organelles involved in lipid metabolism and cellular redox
balance. Peroxisomal function is central to fatty acid oxidation, ether phospholipid synthesis, bile acid
synthesis, and reactive oxygen species homeostasis. Human disorders caused by genetic mutations in
peroxisome genes have led to [...] Read more.
Peroxisomes are metabolic organelles involved in lipid metabolism and cellular redox
balance. Peroxisomal function is central to fatty acid oxidation, ether phospholipid synthesis, bile acid
synthesis, and reactive oxygen species homeostasis. Human disorders caused by genetic mutations in
peroxisome genes have led to extensive studies on peroxisome biology. Peroxisomal defects are linked
to metabolic dysregulation in diverse human diseases, such as neurodegeneration and age-related
disorders, revealing the significance of peroxisome metabolism in human health. Cancer is a disease
with metabolic aberrations. Despite the critical role of peroxisomes in cell metabolism, the functional
eects of peroxisomes in cancer are not as well recognized as those of other metabolic organelles,
such as mitochondria. In addition, the significance of peroxisomes in cancer is less appreciated than
it is in degenerative diseases. In this review, I summarize the metabolic pathways in peroxisomes
and the dysregulation of peroxisome metabolism in cancer. In addition, I discuss the potential of
inactivating peroxisomes to target cancer metabolism, which may pave the way for more eective
cancer treatment. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

23 pages, 1798 KiB  
Review
Role of Mitochondria-Cytoskeleton Interactions in the Regulation of Mitochondrial Structure and Function in Cancer Stem Cells
by Jungmin Kim and Jae-Ho Cheong
Cells 2020, 9(7), 1691; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9071691 - 14 Jul 2020
Cited by 18 | Viewed by 4717
Abstract
Despite the promise of cancer medicine, major challenges currently confronting the treatment of cancer patients include chemoresistance and recurrence. The existence of subpopulations of cancer cells, known as cancer stem cells (CSCs), contributes to the failure of cancer therapies and is associated with [...] Read more.
Despite the promise of cancer medicine, major challenges currently confronting the treatment of cancer patients include chemoresistance and recurrence. The existence of subpopulations of cancer cells, known as cancer stem cells (CSCs), contributes to the failure of cancer therapies and is associated with poor clinical outcomes. Of note, one of the recently characterized features of CSCs is augmented mitochondrial function. The cytoskeleton network is essential in regulating mitochondrial morphology and rearrangement, which are inextricably linked to its functions, such as oxidative phosphorylation (OXPHOS). The interaction between the cytoskeleton and mitochondria can enable CSCs to adapt to challenging conditions, such as a lack of energy sources, and to maintain their stemness. Cytoskeleton-mediated mitochondrial trafficking and relocating to the high energy requirement region are crucial steps in epithelial-to-mesenchymal transition (EMT). In addition, the cytoskeleton itself interplays with and blocks the voltage-dependent anion channel (VDAC) to directly regulate bioenergetics. In this review, we describe the regulation of cellular bioenergetics in CSCs, focusing on the cytoskeleton-mediated dynamic control of mitochondrial structure and function. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

35 pages, 2239 KiB  
Review
Cancer Biology and Prevention in Diabetes
by Swayam Prakash Srivastava and Julie E. Goodwin
Cells 2020, 9(6), 1380; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9061380 - 02 Jun 2020
Cited by 38 | Viewed by 6813
Abstract
The available evidence suggests a complex relationship between diabetes and cancer. Epidemiological data suggest a positive correlation, however, in certain types of cancer, a more complex picture emerges, such as in some site-specific cancers being specific to type I diabetes but not to [...] Read more.
The available evidence suggests a complex relationship between diabetes and cancer. Epidemiological data suggest a positive correlation, however, in certain types of cancer, a more complex picture emerges, such as in some site-specific cancers being specific to type I diabetes but not to type II diabetes. Reports share common and differential mechanisms which affect the relationship between diabetes and cancer. We discuss the use of antidiabetic drugs in a wide range of cancer therapy and cancer therapeutics in the development of hyperglycemia, especially antineoplastic drugs which often induce hyperglycemia by targeting insulin/IGF-1 signaling. Similarly, dipeptidyl peptidase 4 (DPP-4), a well-known target in type II diabetes mellitus, has differential effects on cancer types. Past studies suggest a protective role of DPP-4 inhibitors, but recent studies show that DPP-4 inhibition induces cancer metastasis. Moreover, molecular pathological mechanisms of cancer in diabetes are currently largely unclear. The cancer-causing mechanisms in diabetes have been shown to be complex, including excessive ROS-formation, destruction of essential biomolecules, chronic inflammation, and impaired healing phenomena, collectively leading to carcinogenesis in diabetic conditions. Diabetes-associated epithelial-to-mesenchymal transition (EMT) and endothelial-to-mesenchymal transition (EndMT) contribute to cancer-associated fibroblast (CAF) formation in tumors, allowing the epithelium and endothelium to enable tumor cell extravasation. In this review, we discuss the risk of cancer associated with anti-diabetic therapies, including DPP-4 inhibitors and SGLT2 inhibitors, and the role of catechol-o-methyltransferase (COMT), AMPK, and cell-specific glucocorticoid receptors in cancer biology. We explore possible mechanistic links between diabetes and cancer biology and discuss new therapeutic approaches. Full article
(This article belongs to the Special Issue New Aspects of Targeting Cancer Metabolism in Therapeutic Approach)
Show Figures

Figure 1

Back to TopTop