Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Journal of China Pharmaceutical University ; (6): 392-399, 2022.
Article in Chinese | WPRIM | ID: wpr-939964

ABSTRACT

@#Starvation therapy is an emerging oncological treatment that targets the abnormally elevated nutrient uptake and metabolic pathways to inhibit and kill tumors. In addition to glucose, the targets of starvation therapy also include other nutrients in tumor cells.However, concerns like ineffective targeting and drug tolerance probably have an impact on their clinical translation.Nanomaterial-assisted starvation treatment has been developing quickly in recent years to address these concerns.In this review, several exemplary nanomedicines for starvation therapy and combined starvation therapy with other therapies were offered.They target nutrients other than glucose metabolism, including lactic acid, amino acids, and lipids, using nanomaterials to improve the efficacy of starvation therapy.This review provides reference for further development of nanomedicines with starvation treatment effect.

2.
Acta Pharmaceutica Sinica ; (12): 1801-1807, 2022.
Article in Chinese | WPRIM | ID: wpr-929452

ABSTRACT

The mitochondrial enzyme glutaminase C (GAC) is highly expressed in a variety of cancer cells, resulting in increased glutamine metabolism and cancer development. Therefore, GAC has become a potential target for anti-tumor drug development. However, current GAC inhibitors shared similar structural characteristics, few new scaffolds were reported. By conducting a prokaryotic Escherichia coli expression system, human GAC protein of high-purity was obtained through lysozyme digestion combined with ultrasound dissociation, and cobalt magnetic beads purification, Moreover, we performed studies to validate interaction between small molecules and GAC protein through thermal shift assay, drug affinity responsive target stability assay, protein crosslinking and GAC enzyme activity detection. Meanwhile, a comprehensive small molecule-protein interaction confirmation and systematic pharmacodynamic study in vitro were carried out on compound C19, which was a reported GAC inhibitor screened from the Enamine database. Results showed that C19 directly bind to GAC protein, disturbed GAC tetramers formation, and inhibited its enzyme catalytic activity. By interfering GAC function, C19 dose-dependently suppressed GAC-mediated glutamine metabolism, reduced glutamate in cancer cells, and thus alleviated A549 and NCI-H1299 non-small cell lung cancer cell growth. Together, C19 was identified as a lead compound, providing a new strategy for the structural design of drugs targeting GAC.

3.
Frontiers of Medicine ; (4): 679-692, 2021.
Article in English | WPRIM | ID: wpr-922507

ABSTRACT

Metabolic reprogramming, such as abnormal utilization of glucose, addiction to glutamine, and increased de-novo lipid synthesis, extensively occurs in proliferating cancer cells, but the underneath rationale has remained to be elucidated. Based on the concept of the degree of reduction of a compound, we have recently proposed a calculation termed as potential of electron transfer (PET), which is used to characterize the degree of electron redistribution coupled with metabolic transformations. When this calculation is combined with the assumed model of electron balance in a cellular context, the enforced selective reprogramming could be predicted by examining the net changes of the PET values associated with the biochemical pathways in anaerobic metabolism. Some interesting properties of PET in cancer cells were also discussed, and the model was extended to uncover the chemical nature underlying aerobic glycolysis that essentially results from energy requirement and electron balance. Enabling electron transfer could drive metabolic reprogramming in cancer metabolism. Therefore, the concept and model established on electron transfer could guide the treatment strategies of tumors and future studies on cellular metabolism.


Subject(s)
Humans , Electrons , Energy Metabolism , Glucose , Glycolysis , Neoplasms
4.
J Cancer Res Ther ; 2019 May; 15(3): 631-637
Article | IMSEAR | ID: sea-213396

ABSTRACT

Background: According to Darwin's theory of evolution, complex creatures evolve from more simplistic ancestors. Dollo's law of irreversibility states that evolution is irreversible. However, cancer cells tend to follow anti-Dollo's law. Unfavorable conditions such as hypoxia, acidic pH and low nutrients cause the cancer cells to switch their lifestyle atavistically in order to survive. They start behaving like a unicellular organism. There is a switch from normal metabolism to Warburg effect and finally cannibalism. Cannibalism is a cell eating cell phenomenon. It is defined as a large cell enclosing a smaller one within its cytoplasm and is known by odd names such as “bird's eye cells” or “signet ring cells.” Smaller tumor cells are found in the cytoplasm of larger tumor cells with crescent-shaped nucleus. Cannibalistic cells (CCs) are a feature of aggressive tumors. These cell types are vulnerable to metastasis. Aim: The aim of this study is to identify CCs in various histological grades of oral squamous cell carcinoma (OSCC) and to relate them with the pattern of invasion, lymphocytic response (LR), and mitotic figures (Mfs). The purpose of the article is to establish it as a marker of aggressiveness and metastasis and as an evidence of de-evolution and retroversion of multicellularity. Materials and Methods: Sixty-five histologically confirmed cases of OSCC were studied. Pattern of invasion, LR, number of CCs, and Mfs were recorded on 5 μ hematoxylin and eosin-stained tissue sections. ANOVA and t-test were applied; P < 0.05 was considered statistically significant. Results: CCs were more in sections with patchy LR, increased Mfs, and grade IV pattern of invasion. Conclusion: With increase in dedifferentiation, tumor cells start behaving like unicellular organisms with cell eating cell characteristics

5.
Journal of Medical Postgraduates ; (12): 468-473, 2019.
Article in Chinese | WPRIM | ID: wpr-818262

ABSTRACT

In the past decades, the targeted therapeutic strategies of anti-cancer drugs based on metabolic regulation has been progressing. The study found that the regulation of over-activated metabolic pathways and the subsequent changes brought to metabolic homeostasis can effectively inhibit tumor growth and metastasis. However, the mechanistic link between cancer metabolism and cell fates has remained unclear. As the advancements of biological mass spectrometry and functional omics, researchers have discovered that endogenous metabolites can interact with multiple proteins as functional ligands, and thus affect the survival and proliferation of cancer cells. Nevertheless, the the direct targets and regulatory mechanisms of most functional metabolites in tumors are still unknown. The missing recognition of them has impeded further exploration of the development of precise targeted drug design based metabolic the phenomenon of tumor metabolic reprogramming. Therefore, the capability of elucidating the direct targets of endogenous metabolites in vivo not only helps to develop drugs based on the leading compounds targeting tumor metabolic, but also provides new ideas for personalized medicines of tumor patients. This review thus focuses on the characteristics of cancer metabolism and how endogenous metabolites affects tumor survival, and introduces current target identification approaches applicable to endogenous compounds, in hope to provide thoughts for developing precise treatment strategies based on cancer metabolism.

6.
Clinical Nutrition Research ; : 153-160, 2018.
Article in English | WPRIM | ID: wpr-716069

ABSTRACT

Cancer metabolism is considered as one of major cancer hallmarks. It is important to understand cancer-specific metabolic changes and its impact on cancer biology to identify therapeutic potentials. Among cancer-specific metabolic changes, a role of serine metabolism has been discovered in various cancer types. Upregulation of serine synthesis pathway (SSP) supports cell proliferation and metastasis. The change of serine metabolism is, in part, mediated by epigenetic modifiers, such as Euchromatic histone-lysine N-methyltransferase 2 and Lysine Demethylase 4C. On the other hand, SSP also influences epigenetic landscape such as methylation status of nucleic acids and histone proteins via affecting S-adenosyl methionine production. In the review, we highlight recent evidences on interactions between SSP and epigenetic regulation in cancer. It may provide an insight on roles and regulation of SSP in cancer metabolism and the potential of serine metabolism for cancer therapy.


Subject(s)
Biology , Cell Proliferation , Epigenomics , Hand , Histone-Lysine N-Methyltransferase , Histones , Lysine , Metabolism , Methionine , Methylation , Neoplasm Metastasis , Nucleic Acids , Serine , Up-Regulation
7.
Biomolecules & Therapeutics ; : 45-56, 2018.
Article in English | WPRIM | ID: wpr-739646

ABSTRACT

Cancer is the leading cause of human deaths worldwide. Understanding the biology underlying the evolution of cancer is important for reducing the economic and social burden of cancer. In addition to genetic aberrations, recent studies demonstrate metabolic rewiring, such as aerobic glycolysis, glutamine dependency, accumulation of intermediates of glycolysis, and upregulation of lipid and amino acid synthesis, in several types of cancer to support their high demands on nutrients for building blocks and energy production. Moreover, oncogenic mutations are known to be associated with metabolic reprogramming in cancer, and these overall changes collectively influence tumor-microenvironment interactions and cancer progression. Accordingly, several agents targeting metabolic alterations in cancer have been extensively evaluated in preclinical and clinical settings. Additionally, metabolic reprogramming is considered a novel target to control cancers harboring un-targetable oncogenic alterations such as KRAS. Focusing on lung cancer, here, we highlight recent findings regarding metabolic rewiring in cancer, its association with oncogenic alterations, and therapeutic strategies to control deregulated metabolism in cancer.


Subject(s)
Humans , Biology , Carcinoma, Non-Small-Cell Lung , Glutamine , Glycolysis , Lung Neoplasms , Metabolism , Up-Regulation
8.
Tumor ; (12): 901-906, 2018.
Article in Chinese | WPRIM | ID: wpr-848351

ABSTRACT

Oxidative stress in tumor microenvironment is mainly derived from tumor epithelial cells, which is widely distributed in tumor tissues. Oxidative stress in tumor cells is mainly produced by the accumulation of reactive oxygen species (ROS). Low concentrations of ROS can activate the host cells in tumor microenvironment, and promote the glucose metabolism of cancers by inducing autophagy of mitochondria, changing key enzymes and genomes of glucose metabolism, and activating signaling pathways, so as to maintain the high energy requirement of tumors. High concentrations of ROS can inhibit the occurrence and development of tumors by inducing apoptosis of tumor cells. In recent years, whether anti-oxidant or pro-oxidant can fight against cancer has become a hot topic in cancer therapy. This review focuses on the role of oxidative stress in glucose metabolism of cancer and on the relationship between oxidation-antioxidation balance and tumor therapy, in order to provide new ideas for tumor targeting therapy.

9.
Fudan University Journal of Medical Sciences ; (6): 402-407,440, 2018.
Article in Chinese | WPRIM | ID: wpr-695815

ABSTRACT

Cancer is one of the leading causes responsible for resident's death worldwide.The abnormal status of cancer metabolism and metabolic target antitumor therapy are becoming the hotpot in cancer research.Shortly after the close of 2017 American Society of Clinical Oncology (ASCO) annual meeting,the progress in the field of cancer metabolism and nutrition support could be reviewed in four aspects:clinical investigation of drugs targeting metabolic pathway,metabolism based radiological technologies,role and mechanism of metabolic molecules involved in tumor initiation and progress,and clinical nutrition support.

10.
Journal of International Oncology ; (12): 234-237, 2018.
Article in Chinese | WPRIM | ID: wpr-693484

ABSTRACT

Abnormal fatty acid metabolism is one of the unique metabolic ways in which malignant cells maintain their growth needs and plays a crucial role in tumor progression.Acetyl-CoA carboxylase (ACC) is the rate-limiting enzyme in fatty acid synthesis and oxidative metabolism.More and more researches confirm that ACC is highly expressed in many tumors,and is closely related to tumor progression and prognosis of patients,which makes ACC as a potential marker for clinical diagnosis and prognosis.Tumor cell fatty acid synthesis can be blocked and fatty acid β oxidation can be stimulated by inhibiting the activity of ACC,resulting in serious lipid consumption of tumor,and then inhibit tumor growth and proliferation.Investigating the effect and molecular mechanisms of ACC in the genesis and development of tumors can provide a new insight into cancer targeted molecular therapy.

11.
Protein & Cell ; (12): 216-237, 2018.
Article in English | WPRIM | ID: wpr-756949

ABSTRACT

The tricarboxylic acid (TCA) cycle is a central route for oxidative phosphorylation in cells, and fulfills their bioenergetic, biosynthetic, and redox balance requirements. Despite early dogma that cancer cells bypass the TCA cycle and primarily utilize aerobic glycolysis, emerging evidence demonstrates that certain cancer cells, especially those with deregulated oncogene and tumor suppressor expression, rely heavily on the TCA cycle for energy production and macromolecule synthesis. As the field progresses, the importance of aberrant TCA cycle function in tumorigenesis and the potentials of applying small molecule inhibitors to perturb the enhanced cycle function for cancer treatment start to evolve. In this review, we summarize current knowledge about the fuels feeding the cycle, effects of oncogenes and tumor suppressors on fuel and cycle usage, common genetic alterations and deregulation of cycle enzymes, and potential therapeutic opportunities for targeting the TCA cycle in cancer cells. With the application of advanced technology and in vivo model organism studies, it is our hope that studies of this previously overlooked biochemical hub will provide fresh insights into cancer metabolism and tumorigenesis, subsequently revealing vulnerabilities for therapeutic interventions in various cancer types.


Subject(s)
Animals , Humans , Citric Acid Cycle , Molecular Targeted Therapy , Methods , Neoplasms , Drug Therapy , Genetics , Metabolism , Pathology , Oncogenes , Genetics , Tumor Suppressor Proteins , Metabolism
12.
Yonsei Medical Journal ; : 1143-1149, 2018.
Article in English | WPRIM | ID: wpr-718498

ABSTRACT

Various molecular targeted therapies and diagnostic modalities have been developed for the treatment of hepatocellular carcinoma (HCC); however, HCC still remains a difficult malignancy to cure. Recently, the focus has shifted to cancer metabolism for the diagnosis and treatment of various cancers, including HCC. In addition to conventional diagnostics, the measurement of enhanced tumor cell metabolism using F-18 fluorodeoxyglucose (18F-FDG) for increased glycolysis or C-11 acetate for fatty acid synthesis by positron emission tomography/computed tomography (PET/CT) is well established for clinical management of HCC. Unlike tumors displaying the Warburg effect, HCCs vary substantially in terms of 18F-FDG uptake, which considerably reduces the sensitivity for tumor detection. Accordingly, C-11 acetate has been proposed as a complementary radiotracer for detecting tumors that are not identified by 18F-FDG. In addition to HCC diagnosis, since the degree of 18F-FDG uptake converted to standardized uptake value (SUV) correlates well with tumor aggressiveness, 18F-FDG PET/CT scans can predict patient outcomes such as treatment response and survival with an inverse relationship between SUV and survival. The loss of tumor suppressor genes or activation of oncogenes plays an important role in promoting HCC development, and might be involved in the “metabolic reprogramming” of cancer cells. Mutations in various genes such as TERT, CTNNB1, TP53, and Axin1 are responsible for the development of HCC. Some microRNAs (miRNAs) involved in cancer metabolism are deregulated in HCC, indicating that the modulation of genes/miRNAs might affect HCC growth or metastasis. In this review, we will discuss cancer metabolism as a mechanism for treatment resistance, as well as an attractive potential therapeutic target in HCC.


Subject(s)
Humans , Carcinoma, Hepatocellular , Diagnosis , Drug Resistance , Electrons , Fluorodeoxyglucose F18 , Genes, Tumor Suppressor , Glycolysis , Metabolism , MicroRNAs , Molecular Targeted Therapy , Neoplasm Metastasis , Oncogenes , Positron Emission Tomography Computed Tomography
13.
Clinics ; 66(supl.1): 33-43, 2011. ilus, graf
Article in English | LILACS | ID: lil-593147

ABSTRACT

Cellular energy metabolism is one of the main processes affected during the transition from normal to cancer cells, and it is a crucial determinant of cell proliferation or cell death. As a support for rapid proliferation, cancer cells choose to use glycolysis even in the presence of oxygen (Warburg effect) to fuel macromolecules for the synthesis of nucleotides, fatty acids, and amino acids for the accelerated mitosis, rather than fuel the tricarboxylic acid cycle and oxidative phosphorylation. Mitochondria biogenesis is also reprogrammed in cancer cells, and the destiny of those cells is determined by the balance between energy and macromolecule supplies, and the efficiency of buffering of the cumulative radical oxygen species. In glioblastoma, the most frequent and malignant adult brain tumor, a metabolic shift toward aerobic glycolysis is observed, with regulation by well known genes as integrants of oncogenic pathways such as phosphoinositide 3-kinase/protein kinase, MYC, and hypoxia regulated gene as hypoxia induced factor 1. The expression profile of a set of genes coding for glycolysis and the tricarboxylic acid cycle in glioblastoma cases confirms this metabolic switch. An understanding of how the main metabolic pathways are modified by cancer cells and the interactions between oncogenes and tumor suppressor genes with these pathways may enlighten new strategies in cancer therapy. In the present review, the main metabolic pathways are compared in normal and cancer cells, and key regulations by the main oncogenes and tumor suppressor genes are discussed. Potential therapeutic targets of the cancer energetic metabolism are enumerated, highlighting the astrocytomas, the most common brain cancer.


Subject(s)
Humans , Brain Neoplasms , Glutaminase , Glutamine , Oncogenes/physiology , Brain Neoplasms , Cell Proliferation , Cell Transformation, Neoplastic , Citric Acid Cycle/physiology , Glycolysis/physiology , Pentose Phosphate Pathway/physiology , Stem Cells , Stem Cells
14.
Journal of the Korean Medical Association ; : 113-120, 2009.
Article in Korean | WPRIM | ID: wpr-8084

ABSTRACT

In the 1920's, Warburg reported an observation that cancer cells depend on glycolysis even in the presence of available oxygen likely due to impaired function of mitochondria. Since then, this Warburg s effect has been the most important hypothesis in cancer metabolism and is considered as a seventh hallmark of many human cancers. Aerobic glycolysis was originally attributable to increased bioenergetic needs in rapidly proliferating cancer cells. Recently, biosynthetic aspects of aerobic glycolysis, which reprograms cancer metabolism to synthesize macromolecules such as nucleotides, fatty acids, amino acids, etc., are under active investigation. Introduction of positron emission tomography (PET) and metabolic radiotracers including F-18 flurorodeoxyglucose (FDG) and C-11 acetate made it possible to image cancer metabolism in vivo and to renew the interests on this issue. Studies have found that cancer cells with highly glycolysis features are associated with resistance to many chemotherapeutic regimens and radiation treatment. Therefore, development of glycolytic inhibitors can have an incremental effect to conventional treatments. In addition, functional imaging with metabolic radiotracers will continuously play important roles in detecting cancers and monitoring therapeutic responses to novel anti-metabolic approaches to cancer cells.


Subject(s)
Humans , Amino Acids , Electrons , Energy Metabolism , Fatty Acids , Glycolysis , Mitochondria , Nucleotides , Oxygen , Positron-Emission Tomography
SELECTION OF CITATIONS
SEARCH DETAIL