Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Research

Blockade of glutamine-dependent cell survival augments antitumor efficacy of CPI-613 in head and neck cancer

Authors: Liwei Lang, Fang Wang, Zhichun Ding, Xiangdong Zhao, Reid Loveless, Jin Xie, Chloe Shay, Peng Qiu, Yonggang Ke, Nabil F. Saba, Yong Teng

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

Alterations in metabolism are one of the emerging hallmarks of cancer cells and targeting dysregulated cancer metabolism provides a new approach to developing more selective therapeutics. However, insufficient blockade critical metabolic dependencies of cancer allows the development of metabolic bypasses, thus limiting therapeutic benefits.

Methods

A series of head and neck squamous cell carcinoma (HNSCC) cell lines and animal models were used to determine the efficacy of CPI-613 and CB-839 when given alone or in combination. Glutaminase 1 (GLS1) depletion was achieved by lentiviral shRNAs. Cell viability and apoptosis were determined in HNSCC cells cultured in 2D culture dish and SeedEZ™ 3D scaffold. Molecular alterations were examined by Western blotting and immunohistochemistry. Metabolic changes were assessed by glucose uptake, lactate production, glutathione levels, and oxygen consumption rate.

Results

We show here that HNSCC cells display strong addiction to glutamine. CPI-613, a novel lipoate analog, redirects cellular activity towards tumor-promoting glutaminolysis, leading to low anticancer efficacy in HNSCC cells. Mechanistically, CPI-613 inhibits the tricarboxylic acid cycle by blocking the enzyme activities of pyruvate dehydrogenase and alpha-ketoglutarate dehydrogenase, which upregulates GLS1 and eventually promotes the compensatory role of glutaminolysis in cancer cell survival. Most importantly, the addition of a GLS1 inhibitor CB-839 to CPI-613 treatment abrogates the metabolic dependency of HNSCC cells on glutamine, achieving a synergistic anticancer effect in glutamine-addicted HNSCC.

Conclusions

These findings uncover the critical role of GLS1-mediated glutaminolysis in CPI-613 treatment and suggest that the CB-839 and CPI-613 combination may potentiate synergistic anticancer activity for HNSCC therapeutic gain.
Appendix
Available only for authorised users
Literature
1.
go back to reference Martinez-Outschoorn UE, Peiris-Pagés M, Pestell RG, Sotgia F, Lisanti MP. Cancer metabolism: a therapeutic perspective. Nat Rev Clin Oncol. 2017;14:11–31.PubMedCrossRef Martinez-Outschoorn UE, Peiris-Pagés M, Pestell RG, Sotgia F, Lisanti MP. Cancer metabolism: a therapeutic perspective. Nat Rev Clin Oncol. 2017;14:11–31.PubMedCrossRef
3.
go back to reference Stuart SD, Schauble A, Gupta S, Kennedy AD, Keppler BR, Bingham PM, et al. A strategically designed small molecule attacks alpha-ketoglutarate dehydrogenase in tumor cells through a redox process. Cancer Metab. 2014;2:1–15.CrossRef Stuart SD, Schauble A, Gupta S, Kennedy AD, Keppler BR, Bingham PM, et al. A strategically designed small molecule attacks alpha-ketoglutarate dehydrogenase in tumor cells through a redox process. Cancer Metab. 2014;2:1–15.CrossRef
4.
go back to reference Yamamoto M, Inohara H, Nakagawa T. Targeting metabolic pathways for head and neck cancers therapeutics. Cancer Metastasis Rev. 2017;36:503–14.PubMedCrossRef Yamamoto M, Inohara H, Nakagawa T. Targeting metabolic pathways for head and neck cancers therapeutics. Cancer Metastasis Rev. 2017;36:503–14.PubMedCrossRef
5.
go back to reference Hsieh YT, Chen YF, Lin SC, Chang KW, Li WC. Targeting cellular metabolism modulates head and neck oncogenesis. Int J Mol Sci. 2019;20:3960.PubMedCentralCrossRef Hsieh YT, Chen YF, Lin SC, Chang KW, Li WC. Targeting cellular metabolism modulates head and neck oncogenesis. Int J Mol Sci. 2019;20:3960.PubMedCentralCrossRef
6.
go back to reference Bingham PM, Stuart SD, Zachar Z. Lipoic acid and lipoic acid analogs in cancer metabolism and chemotherapy. Expert Rev Clin Pharmacol. 2014;7:837–46.PubMedCrossRef Bingham PM, Stuart SD, Zachar Z. Lipoic acid and lipoic acid analogs in cancer metabolism and chemotherapy. Expert Rev Clin Pharmacol. 2014;7:837–46.PubMedCrossRef
7.
go back to reference Gao L, Xu Z, Huang Z, Tang Y, Yang D, Huang J, et al. CPI-613 rewires lipid metabolism to enhance pancreatic cancer apoptosis via the AMPK-ACC signaling. J Exp Clin Cancer Res. 2020;39:1–12.CrossRef Gao L, Xu Z, Huang Z, Tang Y, Yang D, Huang J, et al. CPI-613 rewires lipid metabolism to enhance pancreatic cancer apoptosis via the AMPK-ACC signaling. J Exp Clin Cancer Res. 2020;39:1–12.CrossRef
8.
go back to reference Lee KC. Shorr R, Maturo C, W Boteju L, Sheldon a. formation and anti-tumor activity of uncommon in vitro and in vivo metabolites of CPI-613, a novel anti-tumor compound that selectively alters tumor energy metabolism. Drug Metab Lett. 2011;5:163–82.PubMedCrossRef Lee KC. Shorr R, Maturo C, W Boteju L, Sheldon a. formation and anti-tumor activity of uncommon in vitro and in vivo metabolites of CPI-613, a novel anti-tumor compound that selectively alters tumor energy metabolism. Drug Metab Lett. 2011;5:163–82.PubMedCrossRef
9.
go back to reference Lee KC, Maturo C, Perera CN, Luddy J, Rodriguez R, Shorr R. Translational assessment of mitochondrial dysfunction of pancreatic cancer from in vitro gene microarray and animal efficacy studies, to early clinical studies, via the novel tumor-specific anti-mitochondrial agent, CPI-613. Ann Transl Med. 2014;2:91.PubMedPubMedCentral Lee KC, Maturo C, Perera CN, Luddy J, Rodriguez R, Shorr R. Translational assessment of mitochondrial dysfunction of pancreatic cancer from in vitro gene microarray and animal efficacy studies, to early clinical studies, via the novel tumor-specific anti-mitochondrial agent, CPI-613. Ann Transl Med. 2014;2:91.PubMedPubMedCentral
10.
go back to reference Alistar A, Morris BB, Desnoyer R, Klepin HD, Hosseinzadeh K, Clark C, et al. Safety and tolerability of the first-in-class agent CPI-613 in combination with modified FOLFIRINOX in patients with metastatic pancreatic cancer: a single-Centre, open-label, dose-escalation, phase 1 trial. Lancet Oncol. 2017;18:770–8.PubMedPubMedCentralCrossRef Alistar A, Morris BB, Desnoyer R, Klepin HD, Hosseinzadeh K, Clark C, et al. Safety and tolerability of the first-in-class agent CPI-613 in combination with modified FOLFIRINOX in patients with metastatic pancreatic cancer: a single-Centre, open-label, dose-escalation, phase 1 trial. Lancet Oncol. 2017;18:770–8.PubMedPubMedCentralCrossRef
11.
go back to reference Bellio C, DiGloria C, Spriggs DR, Foster R, Growdon WB, Rueda BR. The metabolic inhibitor CPI-613 negates treatment enrichment of ovarian cancer stem cells. Cancers. 2019;11:1678.PubMedCentralCrossRef Bellio C, DiGloria C, Spriggs DR, Foster R, Growdon WB, Rueda BR. The metabolic inhibitor CPI-613 negates treatment enrichment of ovarian cancer stem cells. Cancers. 2019;11:1678.PubMedCentralCrossRef
12.
go back to reference Jin L, Alesi G, Kang S. Glutaminolysis as a target for cancer therapy. Oncogene. 2016;35:3619–25.PubMedCrossRef Jin L, Alesi G, Kang S. Glutaminolysis as a target for cancer therapy. Oncogene. 2016;35:3619–25.PubMedCrossRef
14.
go back to reference Xiang Y, Stine ZE, Xia J, Lu Y, O’Connor RS, Altman BJ, et al. Targeted inhibition of tumor-specific glutaminase diminishes cell-autonomous tumorigenesis. J Clin Invest. 2015;125:2293–306.PubMedPubMedCentralCrossRef Xiang Y, Stine ZE, Xia J, Lu Y, O’Connor RS, Altman BJ, et al. Targeted inhibition of tumor-specific glutaminase diminishes cell-autonomous tumorigenesis. J Clin Invest. 2015;125:2293–306.PubMedPubMedCentralCrossRef
15.
go back to reference Matés JM, Campos-Sandoval JA, Márquez J. Glutaminase isoenzymes in the metabolic therapy of cancer. Biochim Biophys Acta Rev Cancer. 1870;2018:158–64. Matés JM, Campos-Sandoval JA, Márquez J. Glutaminase isoenzymes in the metabolic therapy of cancer. Biochim Biophys Acta Rev Cancer. 1870;2018:158–64.
16.
go back to reference Saha SK, Islam S, Abdullah-Al-Wadud M, Islam S, Ali F, Park KS. Multiomics analysis reveals that GLS and GLS2 differentially modulate the clinical outcomes of cancer. J Clin Med. 2019;8:355.PubMedCentralCrossRef Saha SK, Islam S, Abdullah-Al-Wadud M, Islam S, Ali F, Park KS. Multiomics analysis reveals that GLS and GLS2 differentially modulate the clinical outcomes of cancer. J Clin Med. 2019;8:355.PubMedCentralCrossRef
17.
go back to reference Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, et al. C-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature. 2009;458:762–5.PubMedPubMedCentralCrossRef Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, et al. C-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature. 2009;458:762–5.PubMedPubMedCentralCrossRef
18.
go back to reference Wang JB, Erickson JW, Fuji R, Ramachandran S, Gao P, Dinavahi R, et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 2010;18:207–19.PubMedPubMedCentralCrossRef Wang JB, Erickson JW, Fuji R, Ramachandran S, Gao P, Dinavahi R, et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 2010;18:207–19.PubMedPubMedCentralCrossRef
19.
go back to reference He L, Gao L, Shay C, Lang L, Lv F, Teng Y. Histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the EGFR-Arf1 axis. J Exp Clin Cancer Res. 2019;38:1–10.CrossRef He L, Gao L, Shay C, Lang L, Lv F, Teng Y. Histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the EGFR-Arf1 axis. J Exp Clin Cancer Res. 2019;38:1–10.CrossRef
20.
go back to reference Saba NF, Chen ZG, Haigentz M, Bossi P, Rinaldo A, Rodrigo JP, et al. Targeting the EGFR and immune pathways in squamous cell carcinoma of the head and neck (SCCHN): forging a new alliance. Mol Cancer Ther. 2019;18:1909–15.PubMedPubMedCentralCrossRef Saba NF, Chen ZG, Haigentz M, Bossi P, Rinaldo A, Rodrigo JP, et al. Targeting the EGFR and immune pathways in squamous cell carcinoma of the head and neck (SCCHN): forging a new alliance. Mol Cancer Ther. 2019;18:1909–15.PubMedPubMedCentralCrossRef
21.
go back to reference Lang L, Xiong Y, Prieto-Dominguez N, Loveless R, Jensen C, Shay C, et al. FGF19/FGFR4 signaling axis confines and switches the role of melatonin in head and neck cancer metastasis. J Exp Clin Cancer Res. 2021;40:1–14.CrossRef Lang L, Xiong Y, Prieto-Dominguez N, Loveless R, Jensen C, Shay C, et al. FGF19/FGFR4 signaling axis confines and switches the role of melatonin in head and neck cancer metastasis. J Exp Clin Cancer Res. 2021;40:1–14.CrossRef
22.
go back to reference Lang L, Shay C, Zhao X, Xiong Y, Wang X, Teng Y. Simultaneously inactivating Src and AKT by saracatinib/capivasertib co-delivery nanoparticles to improve the efficacy of anti-Src therapy in head and neck squamous cell carcinoma. J Hematol Oncol. 2019;12:1–14.CrossRef Lang L, Shay C, Zhao X, Xiong Y, Wang X, Teng Y. Simultaneously inactivating Src and AKT by saracatinib/capivasertib co-delivery nanoparticles to improve the efficacy of anti-Src therapy in head and neck squamous cell carcinoma. J Hematol Oncol. 2019;12:1–14.CrossRef
23.
go back to reference Golias T, Papandreou I, Sun R, Kumar B, Brown NV, Swanson BJ, et al. Hypoxic repression of pyruvate dehydrogenase activity is necessary for metabolic reprogramming and growth of model tumours. Sci Rep. 2016;6:1–11.CrossRef Golias T, Papandreou I, Sun R, Kumar B, Brown NV, Swanson BJ, et al. Hypoxic repression of pyruvate dehydrogenase activity is necessary for metabolic reprogramming and growth of model tumours. Sci Rep. 2016;6:1–11.CrossRef
24.
go back to reference Gao L, Lang L, Zhao X, Shay C, Shull AY, Teng Y. FGF19 amplification reveals an oncogenic dependency upon autocrine FGF19/FGFR4 signaling in head and neck squamous cell carcinoma. Oncogene. 2019;38:2394–404.PubMedCrossRef Gao L, Lang L, Zhao X, Shay C, Shull AY, Teng Y. FGF19 amplification reveals an oncogenic dependency upon autocrine FGF19/FGFR4 signaling in head and neck squamous cell carcinoma. Oncogene. 2019;38:2394–404.PubMedCrossRef
25.
go back to reference Chou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006;58:621–81.PubMedCrossRef Chou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006;58:621–81.PubMedCrossRef
26.
go back to reference Zhao X, Lang L, He L, Gao L, Chyan D, Xiong Y, et al. Intracellular reduction in ATP levels contributes to CYT 997-induced suppression of metastasis of head and neck squamous carcinoma. J Cell Mol Med. 2019;23:1174–82.PubMedCrossRef Zhao X, Lang L, He L, Gao L, Chyan D, Xiong Y, et al. Intracellular reduction in ATP levels contributes to CYT 997-induced suppression of metastasis of head and neck squamous carcinoma. J Cell Mol Med. 2019;23:1174–82.PubMedCrossRef
27.
go back to reference Oppermann H, Schnabel L, Meixensberger J, Gaunitz F. Pyruvate attenuates the anti-neoplastic effect of carnosine independently from oxidative phosphorylation. Oncotarget. 2016;7:85848.PubMedPubMedCentralCrossRef Oppermann H, Schnabel L, Meixensberger J, Gaunitz F. Pyruvate attenuates the anti-neoplastic effect of carnosine independently from oxidative phosphorylation. Oncotarget. 2016;7:85848.PubMedPubMedCentralCrossRef
29.
30.
go back to reference Li B, Cao Y, Meng G, Qian L, Xu T, Yan C, et al. Targeting glutaminase 1 attenuates stemness properties in hepatocellular carcinoma by increasing reactive oxygen species and suppressing Wnt/beta-catenin pathway. EBioMedicine. 2019;39:239–54.PubMedCrossRef Li B, Cao Y, Meng G, Qian L, Xu T, Yan C, et al. Targeting glutaminase 1 attenuates stemness properties in hepatocellular carcinoma by increasing reactive oxygen species and suppressing Wnt/beta-catenin pathway. EBioMedicine. 2019;39:239–54.PubMedCrossRef
31.
go back to reference Pardee TS, Lee K, Luddy J, Maturo C, Rodriguez R, Isom S, et al. A phase I study of the first-in-class antimitochondrial metabolism agent, CPI-613, in patients with advanced hematologic malignancies. Clin Cancer Res. 2014;20:5255–64.PubMedPubMedCentralCrossRef Pardee TS, Lee K, Luddy J, Maturo C, Rodriguez R, Isom S, et al. A phase I study of the first-in-class antimitochondrial metabolism agent, CPI-613, in patients with advanced hematologic malignancies. Clin Cancer Res. 2014;20:5255–64.PubMedPubMedCentralCrossRef
32.
go back to reference Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B, et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol Cancer Ther. 2014;13:890–901.PubMedCrossRef Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B, et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol Cancer Ther. 2014;13:890–901.PubMedCrossRef
33.
go back to reference Galan-Cobo A, Sitthideatphaiboon P, Qu X, Poteete A, Pisegna MA, Tong P, et al. LKB1 and KEAP1/NRF2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in KRAS-mutant lung adenocarcinoma. Cancer Res. 2019;79:3251–67.PubMedPubMedCentralCrossRef Galan-Cobo A, Sitthideatphaiboon P, Qu X, Poteete A, Pisegna MA, Tong P, et al. LKB1 and KEAP1/NRF2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in KRAS-mutant lung adenocarcinoma. Cancer Res. 2019;79:3251–67.PubMedPubMedCentralCrossRef
34.
go back to reference Peterse EF, Niessen B, Addie RD, de Jong Y, Cleven AH, Kruisselbrink AB, et al. Targeting glutaminolysis in chondrosarcoma in context of the IDH1/2 mutation. Br J Cancer. 2018;118:1074–83.PubMedPubMedCentralCrossRef Peterse EF, Niessen B, Addie RD, de Jong Y, Cleven AH, Kruisselbrink AB, et al. Targeting glutaminolysis in chondrosarcoma in context of the IDH1/2 mutation. Br J Cancer. 2018;118:1074–83.PubMedPubMedCentralCrossRef
35.
go back to reference Jin H, Wang S, Zaal EA, Wang C, Wu H, Bosma A, et al. A powerful drug combination strategy targeting glutamine addiction for the treatment of human liver cancer. Elife. 2020;9:e56749.PubMedPubMedCentralCrossRef Jin H, Wang S, Zaal EA, Wang C, Wu H, Bosma A, et al. A powerful drug combination strategy targeting glutamine addiction for the treatment of human liver cancer. Elife. 2020;9:e56749.PubMedPubMedCentralCrossRef
36.
go back to reference Li Y, Li X, Li X, Zhong Y, Ji Y, Yu D, et al. PDHA1 gene knockout in prostate cancer cells results in metabolic reprogramming towards greater glutamine dependence. Oncotarget. 2016;7:53837.PubMedPubMedCentralCrossRef Li Y, Li X, Li X, Zhong Y, Ji Y, Yu D, et al. PDHA1 gene knockout in prostate cancer cells results in metabolic reprogramming towards greater glutamine dependence. Oncotarget. 2016;7:53837.PubMedPubMedCentralCrossRef
37.
go back to reference Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE, Grandis JR. Head and neck squamous cell carcinoma. Nat Rev Dis Primers. 2020;6:1–22.CrossRef Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE, Grandis JR. Head and neck squamous cell carcinoma. Nat Rev Dis Primers. 2020;6:1–22.CrossRef
38.
go back to reference Miranda-Galvis M, Loveless R, Kowalski LP, Teng Y. Impacts of environmental factors on head and neck cancer pathogenesis and progression. Cells. 2021;10:389.PubMedPubMedCentralCrossRef Miranda-Galvis M, Loveless R, Kowalski LP, Teng Y. Impacts of environmental factors on head and neck cancer pathogenesis and progression. Cells. 2021;10:389.PubMedPubMedCentralCrossRef
39.
go back to reference Gao L, Zhao X, Lang L, Shay C, Yeudall WA, Teng Y. Autophagy blockade sensitizes human head and neck squamous cell carcinoma towards CYT997 through enhancing excessively high reactive oxygen species-induced apoptosis. J Mol Med (Berl). 2018;96:929–38.CrossRef Gao L, Zhao X, Lang L, Shay C, Yeudall WA, Teng Y. Autophagy blockade sensitizes human head and neck squamous cell carcinoma towards CYT997 through enhancing excessively high reactive oxygen species-induced apoptosis. J Mol Med (Berl). 2018;96:929–38.CrossRef
40.
go back to reference Philip PA, Buyse ME, Alistar AT, Lima CM, Luther S, et al. A phase III open-label trial to evaluate efficacy and safety of CPI-613 plus modified FOLFIRINOX (mFFX) versus FOLFIRINOX (FFX) in patients with metastatic adenocarcinoma of the pancreas. Future Oncol. 2019;15:3189–96.PubMedPubMedCentralCrossRef Philip PA, Buyse ME, Alistar AT, Lima CM, Luther S, et al. A phase III open-label trial to evaluate efficacy and safety of CPI-613 plus modified FOLFIRINOX (mFFX) versus FOLFIRINOX (FFX) in patients with metastatic adenocarcinoma of the pancreas. Future Oncol. 2019;15:3189–96.PubMedPubMedCentralCrossRef
41.
go back to reference Lang L, Chemmalakuzhy R, Shay C, Teng Y. PFKP signaling at a glance: an emerging mediator of cancer cell metabolism. Adv Exp Med Biol. 2019;1134:243–58.PubMedCrossRef Lang L, Chemmalakuzhy R, Shay C, Teng Y. PFKP signaling at a glance: an emerging mediator of cancer cell metabolism. Adv Exp Med Biol. 2019;1134:243–58.PubMedCrossRef
42.
go back to reference Qie S, Chu C, Li W, Wang C, Sang N. ErbB2 activation upregulates glutaminase 1 expression which promotes breast cancer cell proliferation. J Cell Biochem. 2014;115:498–509.PubMedPubMedCentralCrossRef Qie S, Chu C, Li W, Wang C, Sang N. ErbB2 activation upregulates glutaminase 1 expression which promotes breast cancer cell proliferation. J Cell Biochem. 2014;115:498–509.PubMedPubMedCentralCrossRef
43.
go back to reference Chen Z, Li D, Xu N, Fang J, Yu Y, Hou W, et al. Novel 1, 3, 4-selenadiazole-containing kidney-type glutaminase inhibitors showed improved cellular uptake and antitumor activity. J Med Chem. 2018;62:589–603.PubMedCrossRef Chen Z, Li D, Xu N, Fang J, Yu Y, Hou W, et al. Novel 1, 3, 4-selenadiazole-containing kidney-type glutaminase inhibitors showed improved cellular uptake and antitumor activity. J Med Chem. 2018;62:589–603.PubMedCrossRef
44.
go back to reference Yang KY, Wu CR, Zheng MZ, Tang RT, Li XZ, Chen LX, et al. Physapubescin I from husk tomato suppresses SW1990 cancer cell growth by targeting kidney-type glutaminase. Bioorg Chem. 2019;92:103186.PubMedCrossRef Yang KY, Wu CR, Zheng MZ, Tang RT, Li XZ, Chen LX, et al. Physapubescin I from husk tomato suppresses SW1990 cancer cell growth by targeting kidney-type glutaminase. Bioorg Chem. 2019;92:103186.PubMedCrossRef
45.
go back to reference Biancur DE, Paulo JA, Małachowska B, Del Rey MQ, Sousa CM, Wang X, et al. Compensatory metabolic networks in pancreatic cancers upon perturbation of glutamine metabolism. Nat Commun. 2017;8:1–15.CrossRef Biancur DE, Paulo JA, Małachowska B, Del Rey MQ, Sousa CM, Wang X, et al. Compensatory metabolic networks in pancreatic cancers upon perturbation of glutamine metabolism. Nat Commun. 2017;8:1–15.CrossRef
46.
go back to reference Xu X, Meng Y, Li L, Xu P, Wang J, Li Z, et al. Overview of the development of glutaminase inhibitors: achievements and future directions. J Med Chem. 2018;62:1096–115.PubMedCrossRef Xu X, Meng Y, Li L, Xu P, Wang J, Li Z, et al. Overview of the development of glutaminase inhibitors: achievements and future directions. J Med Chem. 2018;62:1096–115.PubMedCrossRef
47.
go back to reference Momcilovic M, Bailey ST, Lee JT, Fishbein MC, Braas D, Go J, et al. The GSK3 signaling axis regulates adaptive glutamine metabolism in lung squamous cell carcinoma. Cancer Cell. 2018;33:905–21.PubMedPubMedCentralCrossRef Momcilovic M, Bailey ST, Lee JT, Fishbein MC, Braas D, Go J, et al. The GSK3 signaling axis regulates adaptive glutamine metabolism in lung squamous cell carcinoma. Cancer Cell. 2018;33:905–21.PubMedPubMedCentralCrossRef
48.
go back to reference Meric-Bernstam F, Lee RJ, Carthon BC, Iliopoulos O, Mier JW, Patel MR, et al. CB-839, a glutaminase inhibitor, in combination with cabozantinib in patients with clear cell and papillary metastatic renal cell cancer (mRCC): results of a phase I study. J Clin Oncol. 2019;37:549.CrossRef Meric-Bernstam F, Lee RJ, Carthon BC, Iliopoulos O, Mier JW, Patel MR, et al. CB-839, a glutaminase inhibitor, in combination with cabozantinib in patients with clear cell and papillary metastatic renal cell cancer (mRCC): results of a phase I study. J Clin Oncol. 2019;37:549.CrossRef
Metadata
Title
Blockade of glutamine-dependent cell survival augments antitumor efficacy of CPI-613 in head and neck cancer
Authors
Liwei Lang
Fang Wang
Zhichun Ding
Xiangdong Zhao
Reid Loveless
Jin Xie
Chloe Shay
Peng Qiu
Yonggang Ke
Nabil F. Saba
Yong Teng
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-02207-y

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine