bims-glucam Biomed News
on Glutamine cancer metabolism
Issue of 2022‒08‒07
seven papers selected by
Sreeparna Banerjee
Middle East Technical University


  1. Mol Cancer Ther. 2022 Aug 05. pii: MCT-22-0282. [Epub ahead of print]
      Glutamine is a conditionally essential amino acid consumed by rapidly proliferating cancer cells, which deprives the same fuel from immune cells and contributes to tumor immune evasion. As such, the broad antagonism of glutamine in tumors and the tumor microenvironment may lead to direct antitumor activity and stimulation of antitumoral immune responses. DRP-104 (sirpiglenastat) was designed as a novel prodrug of the broad-acting glutamine antagonist 6-diazo-5-oxo-L-norleucine (DON). DRP-104 is an inactive form that is preferentially enzymatically converted to DON within tumors. Metabolomic profiling of tumors treated with DRP-104 revealed widespread changes indicative of the disruption of tumor anabolism and canonical cancer metabolism pathways; including altered glutamine metabolism while several immuno-suppressive metabolites were decreased. Gene expression profiling revealed broad immunological modulation, confirmed by flow cytometry indicating that DRP-104 treatment resulted in substantial and broad changes in various immune cell infiltrates, such as increased TIL, T, NK and NKT cells. Functionally, T cells became more proliferative and less exhausted; TAMs were polarized to the M1 phenotype; MDSCs and pro-tumorigenic proteins were decreased in TME. Finally, DRP-104 demonstrated significant antitumor activity as a monotherapy, which was further enhanced in combination with checkpoint blockade therapies, leading to improved survival and long-term durable cures. In summary, DRP-104 broadly remodels the tumor microenvironment by inducing extensive tumor metabolism effects and enhancing the infiltration and function of multiple immune cells distinct from those obtained by checkpoint inhibitor therapy. This unique mechanism of action supports the ongoing clinical development of DRP-104 alone and in combination with checkpoint inhibitors.
    DOI:  https://doi.org/10.1158/1535-7163.MCT-22-0282
  2. J Control Release. 2022 Jul 27. pii: S0168-3659(22)00453-9. [Epub ahead of print]
      The growth and rapid proliferation of tumor cells depend on both glycolysis and glutamine metabolism, leading to metabolic compensation. Here, dual inhibition on the metabolic plasticity by Glucose oxidase and Telaglenastat loaded liposome (Lip@GOx&Tel) were studied for intervening metabolic pathway on energy and material against breast cancer. Lip@GOx&Tel targeting inhibited the two nutrient supply mechanisms employed by tumor cells, reducing the supply of ATP production and biosynthesis precursors essential necessary for tumor, thereby eliciting anti-tumor and anti-metastasis effect. Meanwhile, Lip@GOx&Tel ingeniously amplify the therapeutic effect by up-regulating ROS and down-regulating GSH to disrupt redox homeostasis, thus resulting in inspiring 82% tumor suppression rate on 4 T1 tumor model. Moreover, our study solved the limitation of combination between protein drugs and small molecule drugs in vivo by using liposome nanoparticles with clinical translation value. In short, this work provides a unique perspective of nanomedicine for treating diseases from metabolic intervention.
    Keywords:  Breast cancer; Glutamine-addiction; Liposome nanomedicine; Metabolic intervention; Target delivery
    DOI:  https://doi.org/10.1016/j.jconrel.2022.07.034
  3. Biomater Adv. 2022 Jul;pii: S2772-9508(22)00077-2. [Epub ahead of print]138 212800
      Mass spectrometry-based metabolomics plays a vital role in discovering new markers and revealing the unpredictable biological effects of external stimuli. However, the current metabolomics research on materials is still in its infancy, and in-depth research on possible toxic mechanisms is lacking. In this study, a nanocomposite of gold nanoparticles (AuNPs)-zeolite-imidazole framework-8 (ZIF-8) (Au@ZIF-8) was designed to investigate its effects on metabolism in mouse RAW 264.7 macrophages. The successful synthesis of Au@ZIF-8 was confirmed by transmission electron microscopy (TEM) and elemental analysis. The changes in the metabolic activity of mouse RAW 264.7 macrophages at different concentrations of Au@ZIF-8 and different treatment times were investigated, and their influence on the morphological changes and behavior of RAW 264.7 cells was discussed. In addition, ultrahigh-performance liquid chromatography quadrupole-orbital high-resolution mass spectrometry (UHPLC/Q-Orbitrap HRMS) was used to study the metabolic effects of Au@ZIF-8 on RAW 264.7 cells, and the results showed different metabolites being expressed at different reaction times. After 4, 8 and 24 h of treatment, the differential expression of 14, 16, and 16 metabolites, respectively, was detected. Twenty-five candidate key metabolites were identified from the results of the expression patterns and metabolic pathways. These metabolites are related to glutamine metabolism, the tricarboxylic acid cycle and glycolytic metabolic pathways, which may provide insight into the treatment of diseases caused and progressed by glutamine metabolism. This study also indicates the effectiveness of high-resolution LC-MS in revealing the nanotoxicity mechanism of Au@ZIF-8.
    Keywords:  Au@ZIF-8; Macrophage; Metabolic; Metabolomics
    DOI:  https://doi.org/10.1016/j.bioadv.2022.212800
  4. Front Pharmacol. 2022 ;13 935553
      Metabolic reprogramming is an emerging hallmark of tumor cells. In order to survive in the nutrient-deprived environment, tumor cells rewire their metabolic phenotype to provide sufficient energy and build biomass to sustain their transformed state and promote malignant behaviors. Amino acids are the main compositions of protein, which provide key intermediate substrates for the activation of signaling pathways. Considering that cells can synthesize arginine via argininosuccinate synthase 1 (ASS1), arginine is regarded as a non-essential amino acid, making arginine depletion as a promising therapeutic strategy for ASS1-silencing tumors. In this review, we summarize the current knowledge of expression pattern of ASS1 and related signaling pathways in cancer and its potential role as a novel therapeutic target in cancer. Besides, we outline how ASS1 affects metabolic regulation and tumor progression and further discuss the role of ASS1 in arginine deprivation therapy. Finally, we review approaches to target ASS1 for cancer therapies.
    Keywords:  amino acid; arginine; metabolic reprogramming; prognosis; resistance
    DOI:  https://doi.org/10.3389/fphar.2022.935553
  5. Front Oncol. 2022 ;12 901951
      Glioblastoma multiforme (GBM), a highly invasive and incurable tumor, is the humans' foremost, commonest, and deadliest brain cancer. As in other cancers, distinct combinations of genetic alterations (GA) in GBM induce a diversity of metabolic phenotypes resulting in enhanced malignancy and altered sensitivity to current therapies. Furthermore, GA as a hallmark of cancer, dysregulated cell metabolism in GBM has been recently linked to the acquired GA. Indeed, Numerous point mutations and copy number variations have been shown to drive glioma cells' metabolic state, affecting tumor growth and patient outcomes. Among the most common, IDH mutations, EGFR amplification, mutation, PTEN loss, and MGMT promoter mutation have emerged as key patterns associated with upregulated glycolysis and OXPHOS glutamine addiction and altered lipid metabolism in GBM. Therefore, current Advances in cancer genetic and metabolic profiling have yielded mechanistic insights into the metabolism rewiring of GBM and provided potential avenues for improved therapeutic modalities. Accordingly, actionable metabolic dependencies are currently used to design new treatments for patients with glioblastoma. Herein, we capture the current knowledge of genetic alterations in GBM, provide a detailed understanding of the alterations in metabolic pathways, and discuss their relevance in GBM therapy.
    Keywords:  GBM; genetic alteration; glioma therapy; glycolysis; metabolic genes
    DOI:  https://doi.org/10.3389/fonc.2022.901951
  6. Metab Eng. 2022 Jul 31. pii: S1096-7176(22)00095-7. [Epub ahead of print]
      Cancer cells adapt their intracellular energy metabolism to the oxygen-deprived tumor microenvironment (TME) to ensure tumor progression. This adaptive mechanism has focused attention on the metabolic phenotypes of tumor cells under hypoxic TME for developing novel cancer therapies. Although widely used monolayer (2D) culture does not fully reflect in vivo hypoxic TME, spheroid (3D) culture can produce a milieu similar to the TME in vivo. However, how different metabolic phenotypes are expressed in 3D cultures mimicking tumor hypoxia compared with 2D cultures under hypoxia remains unclear. To address this issue, we investigated the metabolic phenotypes of 2D- and 3D-cultured cancer cells by 13C-metabolic flux analysis (13C-MFA). Principal component analysis of 13C mass isotopomer distributions clearly demonstrated distinct metabolic phenotypes of 3D-cultured cells. 13C-MFA clarified that 3D culture significantly upregulated pyruvate carboxylase flux in line with the pyruvate carboxylase protein expression level. On the other hand, 3D culture downregulated glutaminolytic flux. Consistent with our findings, 3D-cultured cells are more resistant to a glutaminase inhibitor than 2D-cultured cells. This study suggests the importance of considering the metabolic characteristics of the particular in vitro model used for research on cancer metabolism.
    Keywords:  (13)C-metabolic flux analysis; 3D culture; Cancer metabolism; Hypoxic tumor microenvironment; Spheroid
    DOI:  https://doi.org/10.1016/j.ymben.2022.07.008
  7. Trends Cancer. 2022 Jul 28. pii: S2405-8033(22)00156-X. [Epub ahead of print]
      Metastasis is responsible for 90% of deaths in patients with cancer. Understanding the role of metabolism during metastasis has been limited by the development of robust and sensitive technologies that capture metabolic processes in metastasizing cancer cells. We discuss the current technologies available to study (i) metabolism in primary and metastatic cancer cells and (ii) metabolic interactions between cancer cells and the tumor microenvironment (TME) at different stages of the metastatic cascade. We identify advantages and disadvantages of each method and discuss how these tools and technologies will further improve our understanding of metastasis. Studies investigating the complex metabolic rewiring of different cells using state-of-the-art metabolomic technologies have the potential to reveal novel biological processes and therapeutic interventions for human cancers.
    Keywords:  analytical techniques; cancer metabolism; cancer metastasis; metabolomics
    DOI:  https://doi.org/10.1016/j.trecan.2022.07.003