Cancer Letters

Cancer Letters

Volume 502, 1 April 2021, Pages 180-188
Cancer Letters

Glutaminase inhibition with telaglenastat (CB-839) improves treatment response in combination with ionizing radiation in head and neck squamous cell carcinoma models

https://doi.org/10.1016/j.canlet.2020.12.038Get rights and content

Highlights

  • CAL-27, FaDu, and HN5 have glutamine dependent proliferation.

  • Telaglenastat inhibits HNSCC spheroid growth.

  • Telaglenastat increases IR-induced cell death.

  • Telaglenastat decreases aerobic respiration.

  • Combination of telaglenastat and IR reduces tumor size in CAL-27 xenograft mice.

Abstract

The efficacy of ionizing radiation (IR) for head and neck cancer squamous cell carcinoma (HNSCC) is limited by poorly understood mechanisms of adaptive radioresistance. Elevated glutaminase gene expression is linked to significantly reduced survival (p < 0.03). The glutaminase inhibitor, telaglenastat (CB-839), has been tested in Phase I/II cancer trials and is well tolerated by patients. This study investigated if telaglenastat enhances the cellular response to IR in HNSCC models. Using three human HNSCC cell lines and two xenograft mouse models, we examined telaglenastat's effects on radiation sensitivity. IR and telaglenastat combinatorial treatment reduced cell survival (p ≤ 0.05), spheroid size (p ≤ 0.0001) and tumor growth in CAL-27 xenograft bearing mice relative to vehicle (p ≤ 0.01), telaglenastat (p ≤ 0.05) or IR (p ≤ 0.01) monotherapy. Telaglenastat significantly reduced the Oxygen Consumption Rate/Extracellular Acidification Rate ratio in CAL-27 and HN5 cells in the presence of glucose and glutamine (p ≤ 0.0001). Telaglenastat increased oxidative stress and DNA damage in irradiated CAL-27 cells. These data suggest that combination treatment with IR and telaglenastat leads to an enhanced anti-tumor response. This pre-clinical data, combined with the established safety of telaglenastat justifies further investigation for the combination in HNSCC patients.

Introduction

Survival remains poor for head and neck cancer (HNC) patients. Although, 40% of HNC patients receive curative ionizing radiation (IR) therapy, failure within the radiation field approaches 50% with resultant morbidity and mortality [1,2]. Multimodality treatment including the use of concurrent chemotherapy with radiation is used to treat many HNCs. However, the use of systemic therapy lacks a clear molecular basis and is often administered based on empiric evidence, contributing to limited efficacy and increased toxicity [[3], [4], [5]]. Radiotherapy dose escalation is limited by the need to protect surrounding healthy tissues and current options for FDA-approved radiosensitizers are limited. Even though an increasing variety of intelligently designed, gene-targeted drugs are in or are entering clinical use, many are transient in their activity with the majority of patients recurring after a short period of benefit [6]. Therefore, there is increased need to understand and overcome mechanisms of radiation resistance to improve patient survival.

Radiation induces programmed cell survival and metabolic responses within the cell [7,8], representing potential mechanisms of adaptive resistance that reduce radiation efficacy and are consequently interesting targets for combination therapy. Preliminary Reverse Phase Protein Array analysis by our laboratory identified activation of metabolic pathways upon radiation treatment (data not shown). Based on these findings as well as increasing evidence that glutamine plays an important role in DNA damage repair, and the availability of a targeted inhibitor already in Phase I/II trials, we explore the role of elevated glutaminase as a potential mechanism of adaptive resistance to radiation therapy [[9], [10], [11]].

Glutaminase is the enzyme responsible for the conversion of glutamine to glutamate. Glutamate is subsequently converted to α-ketoglutarate, a key component of the Krebs cycle. Notably, glutaminase overexpression has been linked to increased metastasis [12]. If increased glutaminase is independently associated with worse tumor control, and radiation induces glutaminase levels or activity, then we hypothesize that inhibition of glutaminase reduces substrate availability for the Krebs cycle, decreases aerobic respiration, and potentially reduces cellular proliferation.

Inhibition of glutaminase by telaglenastat (Calithera Biosciences, Inc.) has been associated with reduced growth of lymphomas, breast, renal and pancreatic cancers in pre-clinical studies [12]. In Phase II clinical trials for renal cell and triple-negative breast cancer in combination with chemotherapy, telaglenastat exhibited increased biostability and bioavailability as compared to other glutaminase inhibitors, and was well tolerated by patients [12,13]. Notably, neither the role of glutamine in HNC pathogenesis nor the efficacy of telaglenastat in combination with radiation has been previously evaluated.

Using The Cancer Genome Atlas's (TCGA) transcriptome database, we identified that increased glutaminase gene expression was associated with reduced survival in HNSCC patients. As this association supports glutaminase as an important drug target in the treatment of HNSCC, we examined if the combination of glutaminase inhibitor, telaglenastat, and IR is more effective than monotherapy. Clonogenic assays revealed that combinatorial treatment decreased cell survival in CAL-27 and HN5 cell lines. Using 2 heterotopic HNSCC xenograft models, we identified that the combination of telaglenastat and IR reduced tumor volume relative to monotherapy. Telaglenastat also increased IR induced oxidative stress and DNA damage. In summary, our finding that the addition of telaglenastat significantly improves radiation treatment response in HNSCC provides preclinical data in support of future clinical trials.

Section snippets

Cell culture

FaDu (pharynx) cells were grown in Gibco high-glucose MEM (10% FBS, 6 mM glutamine, 1% penicillin/streptomycin, 1% Gibco MEM amino acids, 1% sodium pyruvate). HN5 (tongue) and CAL-27 (tongue) were grown in DMEM (10% FBS, 6 mM l-glutamine, 1% penicillin/streptomycin, 1% Gibco MEM amino acids and 1% sodium pyruvate). Cell lines were tested for mycoplasma every 3 months and authenticity was validated by STR.

Glutaminase activity assay

CAL-27 cells were treated with 0 or 10 Gy using a GammaCell cesium irradiator. Cell lysates

Results

To establish the role of glutaminase in HNSCC, we first queried the transcriptome database from TCGA. High glutaminase expression (upper quartile) versus low glutaminase expression (lower quartile) was associated with reduced patient survival (p = 0.03) (Fig. 1).

As HNSCC represents 90% of HNC cases, this study utilized 3 established human-derived HNSCC lines (CAL-27, HN5, and FaDu) to explore methods for reducing radiation resistance and response to combinations of radiation and glutaminase

Discussion

The primary goal of this study was to examine the role of glutaminase inhibition as a means to address adaptive resistance to IR in head and neck cancer. The combination of glutaminase inhibition with telaglenastat and IR was compared to the effects of independent treatment alone in clonogenic assays and in two xenograft models. We first demonstrate that radiation increased glutaminase activity in glutamine dependent cells within 5 min (Fig. S1). This increase could be used as a mean to exploit

CRediT authorship contribution statement

Christina A. Wicker: Conceptualization, Methodology, Software, Validation, Formal analysis, Investigation, Data curation, Writing - original draft, Writing - review & editing, Visualization. Brian G. Hunt: Investigation, Writing - review & editing. Sunil Krishnan: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Data curation, Writing - review & editing. Kathryn Aziz: Investigation, Data curation, Writing - review & editing. Shobha Parajuli: Investigation, Writing -

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

Calithera Biosciences, Inc. provided telaglenastat (CB-839) for all in vivo experiments.

We acknowledge the MD Anderson Functional Proteomics Reverse Phase Protein Array Core and Preclinical Imaging Core (PIC) at the University of Cincinnati College of Medicine for providing additional facilities and instrumentation.

We would like to thank Dr. Layne Weatherford, and McKenzie Crist for assisting with animal experiments and Dr. Eric Smith for helpful feedback on the manuscript.

References (37)

  • M. Machtay et al.

    J OURNAL OF C LINICAL O NCOLOGY factors associated with severe late toxicity after concurrent chemoradiation for locally advanced head and neck Cancer

    An RTOG Analysis

    (2020)
  • J. Watson

    Oxidants, antioxidants and the current incurability of metastatic cancers

    Open Biol

    (2013)
  • P. Dent et al.
    (2019)
  • P. Dent et al.

    MAPK pathways in radiation responses

    Oncogene

    (2003)
  • T.M. Tannir et al.

    Phase 1 study of glutaminase (GLS) inhibitor CB-839 combined with either everolimus (E) or cabozantinib (Cabo) in patients (pts) with clear cell (cc) and papillary (pap) metastatic renal cell cancer (mRCC)

  • F. Kalinsky et al.

    Meric-Bernstam, Efficacy and safety of CB-839, a small molecule inhibitor of glutaminase, in combination with paclitaxel in patients with advanced triple negative breast cancer (TNBC): initial findings from a multicenter, open-label phase 2 study

  • N.M. Motzer et al.

    ENTRATA: Randomized, double-blind, phase II study of telaglenastat(tela; CB-839) + everolimus(E) vs. placebo (pbo) + E in patients (pts) with advanced/metastatic renal cell carcinoma (mRCC)

    Eur. Soc. Med. Oncol.

    (2019)
  • M.I. Gross et al.

    Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer

    Mol. Canc. Therapeut.

    (2014)
  • Cited by (32)

    • New insights into RAS in head and neck cancer

      2023, Biochimica et Biophysica Acta - Reviews on Cancer
    • Pancreatic stellate cells exploit Wnt/β-catenin/TCF7-mediated glutamine metabolism to promote pancreatic cancer cells growth

      2023, Cancer Letters
      Citation Excerpt :

      We further explored whether altered stromal metabolism was a driver that regulates tumor proliferation and whether disruption of the metabolic crosstalk between PSCs and PCCs created metabolic vulnerability in tumor cells. Previously, researchers have focused on the role of intracellular Gln catabolism in cancer cells [40,41]. However, GLS inhibition exerted anti-cancer activity in vitro but failed in vivo [22].

    • Sex differences in brain tumor glutamine metabolism reveal sex-specific vulnerabilities to treatment

      2022, Med
      Citation Excerpt :

      Targeting glutamine metabolism continues to evolve as an attractive cancer treatment. Targeting GLS, which hydrolyzes glutamine to glutamate, was effective in pre-clinical cancer models25,26,27,28,29 and its clinical efficacy is currently being evaluated. The GLS family consists of two members, GLS1 and GLS2, which are expressed in the mitochondria and the cytosol, respectively.

    View all citing articles on Scopus
    View full text