Abstract
Cancer specific inhibitors reflective of unique metabolic needs, are rare. We describe a novel small molecule, Gboxin, that specifically inhibits primary mouse and human glioblastoma (GBM) cell growth but not mouse embryo fibroblasts or neonatal astrocytes. Gboxin rapidly and irreversibly compromises GBM oxygen consumption. Reliant on its positive charge, Gboxin associates with mitochondrial oxidative phosphorylation complexes in a proton gradient dependent manner and inhibits F0F1 ATP synthase activity. Gboxin resistant cells require a functional mitochondrial permeability transition pore that regulates pH impeding matrix accumulation. Administration of a pharmacologically stable Gboxin analog inhibits GBM allografts and patient derived xenografts. Gboxin toxicity extends to established human cancer cell lines of diverse organ origin and exposes the elevated proton gradient pH in cancer cell mitochondria as a new mode of action for antitumor reagent development.
Glioblastoma is the most aggressive and prevalent primary malignancy of the central nervous system1,2. Current treatments, dominated by radiotherapy and chemotherapy, target proliferating tumor cells and induce potent toxic side effects by harming normal proliferating cells3,4. It is possible that relatively quiescent cancer stem cells (CSCs) in tumors may evade conventional therapies3,5,6. CSCs can have metabolic characteristics that set them apart from proliferating tumor and somatic cells. While proliferative tumor cells rely on aerobic glycolysis, known as the Warburg effect, slow-cycling tumor cells may prefer mitochondrial respiration as a primary source of energy4,5,7-9.
Oxidative phosphorylation (OxPhos) plays a central role in cellular energy. Over 90 proteins encoded by both the nuclear and mitochondrial genomes comprise the OxPhos machinery. The OxPhos electron transport chain (ETC) constitutes four complexes (CI-CIV) that transfer electrons from donors generated by the TCA cycle and fatty acid oxidation to oxygen. Complexes I-IV pump protons out into the mitochondrial intermembrane space elevating pH inside this formed voltage gradient. Complex V (CV; F0F1 ATP synthase) uses the stored energy in the proton gradient to generate ATP. Reactive oxygen species (ROS), a byproduct of the ETC and ATP production, can be mitigated by several mechanisms including the mitochondrial permeability transition pore (mPTP)10,11. Several studies have examined the potential vulnerability of the ETC in cancer cells by inhibition of CI and some may hold promise upon continued validation12,14-17.
Here we describe a novel compound, Gboxin, isolated from a low passage primary culture cell-based high throughput chemical screen designed to filter out toxicity to wild type proliferating cells while limiting lethality to primary GBM stem-like cells. Cancer cells have an unusually high mitochondrial membrane potential and thus retain higher pH within the matrix18-21. Gboxin targets unique features of mitochondrial pH in GBM and other cancer cells, independent of their genetic composition, and exerts its tumor cell specific toxicity in primary culture and in vivo.
Primary GBM culture high throughput screen.
Previous cell-based “anticancer” drug screens have predominantly identified agents that interfere with mitosis, replication, or DNA damage repair22,23. To differentiate between cancer cells and normally dividing cells we considered experimental features to afford improved physiological relevance including use of primary early passage tumor cells, serum free conditions, and low oxygen tension (5%; normoxia). We used a spontaneous GBM mouse model with mutations in three GBM relevant tumor suppressors (Trp53, Pten, and NF1) under hGFAP-cre transgene mediated recombination24,25. Low passage sphere cultures from multiple tumors were pooled to establish primary “high throughput tumor sphere” (HTS) cells frozen as pooled aliquots and used for the entire screening process (see Methods). Using a 96-hour Cell-Titer-Glo® protocol, a chemical screen of 200,000 compounds was performed (Extended data Fig. 1a). To exclude nonspecific or anti-mitotic associated toxicity, a counter screen against primary low passage mouse embryonic fibroblasts (MEFs), neonatal astrocytes, and primary subventricular zone neural stem/progenitor cells (NSCs) was performed. 61 compounds emerged as specific inhibitors of HTS cells with IC50s in the nanomolar range and therapeutic windows ranging from 1 to 3 logs (data not shown). We tested the 61 compounds in S9 fraction and hepatocyte assays, which yielded 17 compounds that were further tested for chemical tractability (Extended data Fig. 1a).
Here we describe the properties of one lead compound, a benzimidazolinium (hereafter Gboxin; IC50: 150 nM; Fig. 1a, b), and its chemical derivatives that specifically inhibit HTS GBM cell growth but not cycling primary MEFs or astrocytes (Fig. 1b and Extended Data Fig. 1b) with irreversible Gboxin growth inhibition between 6 and 24 hours of exposure (Extended data Fig. 1d). Gboxin inhibits wild type neural stem/progenitor cell (NSC) growth but with a greater than tenfold higher IC50 (Extended Data Fig. 1c). Transcriptional microarray analysis after Gboxin treatment revealed specific and sustained gene expression changes in HTS cells versus MEFs and astrocytes including cell cycle and survival regulating genes such as, ATF4, and Survivin (Extended Data Fig. 1e,f and Supplementary Table 1), and Gene Ontology (GO) analysis identified multiple upregulated ATF4 stress response targets (Extended Data Fig. 1e,f; and Supplementary Table 1)26-28. Western blot analysis confirmed HTS specific elevation of ATF4 protein at 3 and 6 hours (Fig. 1c; Extended Data Fig. 1g,h). We also investigated several cancer associated signal transduction pathways following 6 hour Gboxin exposure and found that ATF4 upregulation is temporally accompanied by decreased phosphorylated-S6 levels (p-S6; Fig. 1c). Within 24 hours HTS cells underwent cell cycle arrest (G1/0:S ratio increase) followed by an apoptosis molecular signature within 3 days (Extended data Fig. 1i,j). Thus, in primary GBM (HTS) cells, Gboxin elicits rapid and specific responses leading to cell death that is not manifested in cycling primary MEFs or astrocytes.
Gboxin disrupts primary GBM cell metabolism.
The microarray data showed rapid and sustained transcriptional suppression of Txnip, a regulator of glucose uptake, consumption, and target of oxidative phosphorylation (OxPhos)29,30 (Extended Data Fig. 1e). We examined the status of mitochondrial membrane potential assayed by tetramethylrhodamine ethyl ester perchlorate (TMRE) stain following extended Gboxin treatment31 indicating reduced membrane potential in HTS cells but not in MEFs (Extended Data Fig. 1k,l). We next performed O2 consumption rate (OCR) measurements that showed a dose dependent Gboxin inhibition equally potent to well characterized OxPhos inhibitors (Fig. 2a; Extended Data Fig. 2a). Despite its unique lethality to HTS cells, Gboxin also caused acute OCR depletion in MEFs and astrocytes (Fig. 2a and Extended Data Fig. 2b). However, prolonged (30 hr.) OCR measurement showed that MEFs and astrocytes fully recover (Extended Data Fig. 2c,d). Consistent with the above data, the OxPhos inhibitors rotenone, antimycin A, and oligomycin A which both acutely and chronically impede OCR in all cells, rapidly induced ATF4 in MEFs and astrocytes (Fig. 2b; Extended Data Fig. 2e). AMPK, a rheostat for ATP availability is activated in response to OxPhos inhibition and Gboxin treated HTS cells showed rapid and robust elevation of phospho-AMPK and its target, phospho-ACC-79 (Fig. 2c)32. These data indicate a differential activity of sustained OxPhos inhibition by Gboxin on GBM cells versus wild type cells.
Gboxin interacts with respiratory chain proteins.
Through structure activity relationship studies (SAR33) we determined that the positive charge on Gboxin is essential for activity (data not shown). We further identified an active analog amenable to biotin moiety modification (B-Gboxin; IC50: 1,530 nM for HTS cells; Extended Data Fig. 3a,b). B-Gboxin conserves Gboxin properties including OCR inhibition, ATF4 induction, and phospho-S6 protein reduction (Extended Data Fig. 3c,d). Biotin mediated pull down experiments using HTS cells pre-treated with B-Gboxin followed by electrophoresis resolved multiple silver stained bands that were absent or reduced when preincubated with unbiotinylated Gboxin, indicating a Gboxin specific interaction (Fig. 3a). Mass-spec analysis of eluate from pull down samples from Gboxin, B-Gboxin, and Gboxin/B-Gboxin treated HTS cells confirmed Gboxin specific interactions and further revealed a biased presence of mitochondrial proteins (Supplementary Table 2). We next purified mitochondria from pretreated HTS cells for pull down and mass spec analysis. Of 58 detected proteins, 12 are components of the OxPhos machinery (Supplementary Table 3 and Extended Data Fig. 3e) including all ETC complexes with CV best represented (5 proteins; Extended Data Fig. 3e). We confirmed OxPhos component interactions using antibodies against components of CI, CII, CIV, and CV, respectively (Fig. 3b and Extended Data Fig. 3f) and showed that B-Gboxin/OxPhos protein interactions occur within 10 minutes (Extended Data Fig. 3g).
Co-purification interactions between B-Gboxin and OxPhos proteins in Gboxin resistant cells is considerably weakened or absent compared to those in HTS cells (Fig. 3c). Also, detection of B-Gboxin/OxPhos protein interactions requires whole cell pre-incubation and cannot be achieved with incubation of cell lysates (data not shown). Thus, the specific association with OxPhos proteins may be related to the unique environment created by the proton gradient across the mitochondrial inner membrane of cancer cells. To test this, we assayed B-Gboxin/OxPhos protein association in HTS cells in the presence of proton gradient inhibitors. Rotenone and Fccp (a hydrogen ionophore) cause rapid reduction of the mitochondrial inner membrane proton gradient and inhibited B-Gboxin interaction with OxPhos proteins. Conversely, oligomycin A acutely increases the proton gradient and augmented B-Gboxin/OxPhos protein interactions in primary MEFs and astrocytes (Extended Fig. Data 4a).
Several indicators point to CV as the functional target for Gboxin mediated cell death. OCR data demonstrate that, like CV inhibitor oligomycin A, Gboxin OCR inhibition can be bypassed by the proton ionophore, Fccp, while CI and CIII inhibitors cannot (Fig. 2a; Extended Data Fig. 2a). Furthermore, like oligomycin A, Gboxin causes an acute increase in mitochondrial membrane potential (Fig. 4a). In contrast antimycin A and rotenone cause an acute reduction of membrane potential (Fig. 4a). Finally, comparison of HTS cell gene expression following exposure to rotenone, antimycin A, oligomycin A, or Gboxin shows a related profile for oligomycin A and Gboxin (Extended Data Fig. 4b).
Blunted mPTP activity in GBM cells.
The B-Gboxin mitochondrial extract pull downs also identified the protein Adt2, a component of the mPTP (Supplementary Table 3), an unselective voltage-dependent channel to molecules of molecular mass <1.5 kDa11,34-36. Elevated reactive oxygen species (ROS) inside the mitochondrion can be expelled by transient opening of mPTP to reduce membrane potential10. mPTP activity was examined by membrane potential assays using tetramethylrhodamine methyl ester perchlorate (TMRM) fluorescence intensity, and mPTP specific blockade was achieved using the inhibitor, Cyclosporin A (CsA). Under basal conditions, MEFs respond to ROS (H2O2) by reducing membrane potential through opening of mPTP. CsA inhibition of the pore increases basal MEF membrane potential that in the presence of ROS, cannot be relieved (Fig. 4b). In contrast, HTS cells show an elevated basal membrane potential that remains unresponsive to CsA or H2O2 (Fig. 4b)19,21. CsA had no measurable effects on Gboxin function and toxicity in HTS cells (Extended Data Fig. 4c), but remarkably, in the presence of CsA, MEFs became Gboxin sensitive (Fig. 4c), accompanied by rapid ATF4 induction (Extended Data Fig. 4d) and strengthened association of OxPhos proteins with B-Gboxin (Fig. 4d). To exclude possible off-target effects of CsA, we complemented the studies with siRNAs directed against cyclophilin D (CypD; encoded by the PPIF gene), the mPTP target of CsA and achieved similar results (Extended Data Fig. 4e)37. Thus a functional mPTP is essential for Gboxin resistance.
The Gboxin SAR also yielded a functional analog amenable for live cell UV crosslink conjugation (C-Gboxin; IC 50: 350 nM) that can be probed with an Azide Fluor via click chemistry (Extended Data Fig. 5a-c)38. As demonstrated by immunofluorescence colocalization with the OxPhos CII component, SDHA, there is high accumulation of C-Gboxin in GBM cell (HTS) mitochondria (Extended Data Fig. 5d). In contrast resistant MEFs show limited mitochondrial C-Gboxin accumulation (Fig. 4e) that is reversed by CsA (Fig. 4f). These data verify the preceding biochemical data demonstrating that Gboxin specifically accumulates in cancer cell mitochondria. Cyclosporin mediated blockade of mPTP elevates mitochondrial pH (Fig. 4b), Gboxin accumulation and association with OxPhos proteins (Fig. 4d,f); and causes cellular toxicity (Fig. 4c) to previously resistant MEFs.
Gboxin toxicity extends to human tumor cells.
We next tested Gboxin activity on three independent GBM primary mouse cultures (Extended data Fig. 6a) and three GBM patient derived cultures (see Methods; Fig. 5a). All cultures exhibited Gboxin sensitivity with mouse cells having lower IC50 (~150 nM) than human GBM cells (~1 μM; Fig. 5a); indicating blunted mPTP activity in GBM cells of diverse origin39-41.
We also assessed the scope of Gboxin activity on well annotated human cancer cell lines derived from diverse tumor types (Fig. 5b-e; and data not shown). A majority of cancer cell lines were Gboxin sensitive with therapeutic windows compared to wild type resistant MEFs and astroctyes (Fig. 5b). OCR assays confirmed Gboxin coupled respiration blockade in representative cell lines (Extended Data Fig. 6b-h). Two cell lines exhibited Gboxin resistance: Daoy human medulloblastoma cells and primary murine malignant peripheral nerve sheath tumor (MPNST) cells42. These resistant tumor cells acquire Gboxin sensitivity and enhanced B-Gboxin OxPhos protein association upon CsA inhibition of the mPTP (Fig. 5e; Extended Data. Fig. 6i). Given the diverse genotypic drivers present in the sensitive human GBM and cancer cell lines, it appears that Gboxin activity is not tied to the specific driver mutations used to the generate the HTS GBM cell screen (NF1, Trp53, and Pten). Moreover, a correlation emerges that, like MEFs, the few Gboxin resistant cancer cells have a CsA responsive, functional mPTP.
Gboxin inhibits mouse and human GBM growth.
Through the SAR studies, we identified a functional analog (S-Gboxin; IC50: 470 nM; Extended Data Fig. 7a-h) that retained excellent metabolic stability, enhanced plasma stability, and PK-properties (Extended Data Fig. 7d-g) suitable for in vivo studies. Antitumor activity was first assessed by daily S-Gboxin treatment at 10 mg/kg/day beginning on day 3 or day 14 after mouse GBM (HTS cells) allograft flank implantation (Fig. 6a and Extended Data Fig. 8a). S-Gboxin treated mice showed reduced tumor volume, cellular density, proliferation, and enhanced survival in comparison to vehicle controls (Fig. 6b-d and Extended Data Fig. 8b-d). S-Gboxin treated tumors had reduced expression of the high-grade glioma makers, GFAP and Olig2 (Fig. 6b and Extended Data Fig. 8b). Primary human GBM cells were also injected into flanks of immunocompromised mice in the presence of matrigel. After visible tumors were detected (3 days) S-Gboxin was administered daily (10 mg/kg/day) resulting in significant attenuation of growth and decreased cellular density compared to controls (Extended Data Fig. 8e-h).
To overcome poor blood brain barrier penetration, we used catheter delivery to test activity in intracranial tumors. Primary mouse GBM cells were orthotopically transplanted followed by a two-week period to allow tumor seeding and surgery recovery, after which catheters were implanted at the site of injection to deliver S-Gboxin locally through subcutaneous minipumps (2.16 μg/day/mouse; see Methods for details). S-Gboxin treatment inhibited tumor growth as manifested by reduced hemorrhaging, cellular density, and decreased proliferation (Extended Data Fig. 8i-k). Histopathology analysis further showed reduced expression of the high-grade glioma makers (Extended Data Fig. 8j,l).
We next tested two independent patient derived xenografts (PDXs). Freshly engrafted patient GBM (P0) were harvested from symptomatic PDX mice and homogenized (Patient Tumor 1) or dissociated into single cells (Patient Tumor 2), and orthotopically implanted into brains of 4 mice each without culturing or additional manipulations (P1). Two weeks (Tumor 1) or 4 weeks (Tumor 2) later, minipumps were implanted to deliver vehicle or S-Gboxin to the tumor area. All vehicle treated mice first showed symptoms of morbidity (41–65 days), and were sacrificed for analysis. S-Gboxin demonstrated inhibition of GBM PDX growth as manifested by general health status, reduced cellular density, cellular proliferation, and GBM marker expression (Fig. 6e,f and Extended Data Fig. 8m). Despite daily in vivo delivery over a four-week period or more, no weight loss (Extended Data Fig. 8n), or overt signs of general health deficits were detected in treated mice (data not shown). We also examined the status of the SVZ stem cell niche following intracranial S-Gboxin treatment. Representative comparison of endogenous Nestin staining from control and S-Gboxin treated tumor bearing mice showed no abnormalities or loss of Nestin staining in the SVZ despite the obvious reduction of Nestin positive cells within the treated tumors (Extended Data Fig. 8l).
Finally, primary cultures from residual S-Gboxin treated tumors were tested for acquired Gboxin resistance. Four independent tumor primary cultures (two treated with S-Gboxin and two vehicle controls) were tested for Gboxin and S-Gboxin sensitivity. All tumor derived cultures retained full Gboxin sensitivity (Extended Data Fig. 9a,b). Thus, the failure of S-Gboxin to completely abrogate in vivo tumor growth is likely the consequence of inefficient access to the tumor over time and not to acquired resistance through secondary mechanisms, including reactivation of mPTP function.
Discussion.
Effective eradication of proliferating cells does not always result in meaningful therapeutic value. One possible explanation may lie in tumor heterogeneity and the presence of resistant quiescent stem-like cells. Here we sought to identify candidate antitumor molecules that would not display toxicity onto normal cycling cells.
Multiple mitochondrial functions are modified to sustain the cancer cell, and thus may in turn serve as cancer specific targets43-46. Many studies have investigated CI OxPhos inhibitors as cancer therapeutic targets with varying results13,14,47-49. Whether they will inhibit primary culture tumor cells, or in vivo tumors with acceptable toxicity parameters remains to be fully determined13,14. Gboxin, a novel CV inhibitor, relies on its positive charge to accumulate in GBM mitochondria with irreversible toxic effects through rapid and sustained blockade of mitochondrial respiration coupled oxidative phosphorylation. Gboxin resistance in wild type cells rested on functional activity of the mPTP. This uncovers a novel mechanism of action for targeting tumor cells dependent on the heightened proton gradient present across the mitochondrial inner membrane of GBM and other cancer cells tested (Extended Data Fig. 10)50. Elucidation of underlying causes for this apparent mPTP deficit in GBM and other Gboxin sensitive cancer cells merits careful scrutiny and may shed additional light on how metabolism alterations in cancer cells may be targeted.
Primary cultures established from residual treated tumor explants continued to exhibit Gboxin toxicity suggesting that improved delivery, or coupling with additional antitumor agents may serve to completely eradicate tumor growth. The scope of Gboxin toxicity appears to be unrelated to specific driver mutations. Its extended activity to human cancer cell lines opens the possibility that a larger subset of cancers may be susceptible to Gboxin and related targeting strategies.
Supplementary Material
Extended Data
Acknowledgements.
The authors thank Yanjiao Li, Tracey Shipman, and Samhita Bapat for technical assistance. We thank David Sabatini (Whitehead Institute) for supplying U937, NCI-H82, Cal-62 and NCI-H524 cell lines, Neal Rosen (MSKCC) for supplying Mel30, Colo205, NCI-H2030, HCT116, SK-MEL113, and A375 cell lines, Albee Messing (University of Wisconsin-Madison) for supplying primary astrocytes, and Dr. Cameron Brennan (MSKCC) for supplying ts603 IDH1 mutant human GBM cells. SKL was a recipient of the Basic Research Fellowship from American Brain Tumor Association (in memory of Theodore Sapper). L.F.P. received funding from NCI (R35: CA210100 and R01: CA131313). L.F.P. and J.K.D. received funding from CPRIT (RP100782, RP120262, and RP150242). J.K.D. acknowledges support from the Robert A. Welch Foundation (I-1422).
Footnotes
Reporting Summary. Further information on research design is available in the Nature Research Reporting Summary linked to this paper.
Data availability. All important data generated or analyzed during this study are included in this article. Additional supplementary data are available from the corresponding author upon request.
References
- 1.Stupp R et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352, 987–996, doi: 10.1056/NEJMoa043330 (2005). [DOI] [PubMed] [Google Scholar]
- 2.Wen PY & Kesari S Malignant gliomas in adults. N Engl J Med 359, 492–507, doi: 10.1056/NEJMra0708126 (2008). [DOI] [PubMed] [Google Scholar]
- 3.Chen J et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 488, 522–526, doi: 10.1038/nature11287 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Viale A & Draetta GF Metabolic Features of Cancer Treatment Resistance. Recent Results Cancer Res 207, 135–156, doi: 10.1007/978-3-319-42118-6_6 (2016). [DOI] [PubMed] [Google Scholar]
- 5.Viale A et al. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature 514, 628–632, doi: 10.1038/nature13611 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Parada LF, Dirks PB & Wechsler-Reya RJ Brain Tumor Stem Cells Remain in Play. J Clin Oncol 35, 2428–2431, doi: 10.1200/JCO.2017.73.9540 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Caro P et al. Metabolic Signatures Uncover Distinct Targets in Molecular Subsets of Diffuse Large B Cell Lymphoma. Cancer Cell 22, 547–560, doi: 10.1016/j.ccr.2012.08.014 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Cole A et al. Inhibition of the Mitochondrial Protease ClpP as a Therapeutic Strategy for Human Acute Myeloid Leukemia. Cancer Cell 27, 864–876, doi: 10.1016/j.ccell.2015.05.004 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bosc C, Selak MA & Sarry JE Resistance Is Futile: Targeting Mitochondrial Energetics and Metabolism to Overcome Drug Resistance in Cancer Treatment. Cell Metabolism 26, 705–707, doi: 10.1016/j.cmet.2017.10.013 (2017). [DOI] [PubMed] [Google Scholar]
- 10.Zorov DB, Juhaszova M & Sollott SJ Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev 94, 909–950, doi: 10.1152/physrev.00026.2013 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Biasutto L, Azzolini M, Szabo I & Zoratti M The mitochondrial permeability transition pore in AD 2016: An update. Bba-Mol Cell Res 1863, 2515–2530, doi: 10.1016/j.bbamcr.2016.02.012 (2016). [DOI] [PubMed] [Google Scholar]
- 12.Libby G et al. New Users of Metformin Are at Low Risk of Incident Cancer A cohort study among people with type 2 diabetes. Diabetes Care 32, 1620–1625, doi: 10.2337/dc08-2175 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Naguib A et al. Mitochondrial Complex I Inhibitors Expose a Vulnerability for Selective Killing of Pten-Null Cells. Cell Rep 23, 58–67, doi: 10.1016/j.celrep.2018.03.032 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Molina JR et al. An inhibitor of oxidative phosphorylation exploits cancer vulnerability. Nat Med 24, 1036–1046, doi: 10.1038/s41591-018-0052-4 (2018). [DOI] [PubMed] [Google Scholar]
- 15.Wheaton WW et al. Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis. Elife 3, e02242, doi: 10.7554/eLife.02242 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Liu X, Romero IL, Litchfield LM, Lengyel E & Locasale JW Metformin Targets Central Carbon Metabolism and Reveals Mitochondrial Requirements in Human Cancers. Cell Metab 24, 728–739, doi: 10.1016/j.cmet.2016.09.005 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lord SR et al. Integrated Pharmacodynamic Analysis Identifies Two Metabolic Adaption Pathways to Metformin in Breast Cancer. Cell Metab 28, 679–688 e674, doi: 10.1016/j.cmet.2018.08.021 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Heerdt BG, Houston MA & Augenlicht LH The intrinsic mitochondrial membrane potential of colonic carcinoma cells is linked to the probability of tumor progression. Cancer Res 65, 9861–9867, doi: 10.1158/0008-5472.Can-05-2444 (2005). [DOI] [PubMed] [Google Scholar]
- 19.Heerdt BG, Houston MA & Augenlicht LH Growth properties of colonic tumor cells are a function of the intrinsic mitochondrial membrane potential. Cancer research 66, 1591–1596, doi: 10.1158/0008-5472.CAN-05-2717 (2006). [DOI] [PubMed] [Google Scholar]
- 20.Modica-Napolitano JS & Aprille JR Delocalized lipophilic cations selectively target the mitochondria of carcinoma cells. Adv Drug Deliv Rev 49, 63–70 (2001). [DOI] [PubMed] [Google Scholar]
- 21.Bernal SD, Lampidis TJ, Summerhayes IC & Chen LB Rhodamine-123 selectively reduces clonal growth of carcinoma cells in vitro. Science 218, 1117–1119 (1982). [DOI] [PubMed] [Google Scholar]
- 22.Jekimovs C et al. Chemotherapeutic compounds targeting the DNA double-strand break repair pathways: the good, the bad, and the promising. Front Oncol 4, 86, doi: 10.3389/fonc.2014.00086 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Senese S et al. Chemical dissection of the cell cycle: probes for cell biology and anti-cancer drug development. Cell Death Dis 5, e1462, doi: 10.1038/cddis.2014.420 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kwon CH et al. Pten haploinsufficiency accelerates formation of high-grade astrocytomas. Cancer Res 68, 3286–3294, doi: 10.1158/0008-54/2.Can-07-6867 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zhu Y et al. Early inactivation of p53 tumor suppressor gene cooperating with NF1 loss induces malignant astrocytoma. Cancer cell 8, 119–130, doi: 10.1016/j.ccr.2005.07.004 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ye JB et al. The GCN2-ATF4 pathway is critical for tumour cell survival and proliferation in response to nutrient deprivation. Embo J 29, 2082–2096, doi: 10.1038/emboj.2010.81 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Milani M et al. The Role of ATF4 Stabilization and Autophagy in Resistance of Breast Cancer Cells Treated with Bortezomib. Cancer research 69, 4415–4423, doi: 10.1158/0008-5472.Can-08-2839 (2009). [DOI] [PubMed] [Google Scholar]
- 28.Quiros PM et al. Multi-omics analysis identifies ATF4 as a key regulator of the mitochondrial stress response in mammals. J Cell Biol 216, 2027–2045, doi: 10.1083/jcb.201702058 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Yu FX, Chai TF, He H, Hagen T & Luo Y Thioredoxin-interacting protein (Txnip) gene expression: sensing oxidative phosphorylation status and glycolytic rate. J Biol Chem 285, 25822–25830, doi: 10.1074/jbc.M110.108290 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Parikh H et al. TXNIP regulates peripheral glucose metabolism in humans. PLoS Med 4, e158, doi: 10.1371/journal.pmed.0040158 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Perry SW, Norman JP, Barbieri J, Brown EB & Gelbard HA Mitochondrial membrane potential probes and the proton gradient: a practical usage guide. Biotechniques 50, 98–115, doi: 10.2144/000113610 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Dasgupta B & Chhipa RR Evolving Lessons on the Complex Role of AMPK in Normal Physiology and Cancer. Trends Pharmacol Sci 37, 192–206, doi: 10.1016/j.tips.2015.11.007 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.De Brabander JK et al. Substituted Benzimidazolium, Pyrido-Imidazolium, or Pyrazino-Imidazolium Compounds as Chemotherapeutic Agents. International Application No. PCT/US2016/065751, Pub. No. WO/2017/100525 (2017). [Google Scholar]
- 34.Bonora M & Pinton P The mitochondrial permeability transition pore and cancer: molecular mechanisms involved in cell death. Front Oncol 4, 302, doi: 10.3389/fonc.2014.00302 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhou W, Marinelli F, Nief C & Faraldo-Gomez JD Atomistic simulations indicate the c-subunit ring of the F1Fo ATP synthase is not the mitochondrial permeability transition pore. Elife 6, doi: 10.7554/eLife.23781 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Baines CP & Gutierrez-Aguilar M The still uncertain identity of the channel-forming unit(s) of the mitochondrial permeability transition pore. Cell Calcium 73, 121–130, doi: 10.1016/j.ceca.2018.05.003 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Basso E et al. Properties of the permeability transition pore in mitochondria devoid of cyclophilin D. Journal of Biological Chemistry 280, 18558–18561, doi: 10.1074/jbc.C500089200 (2005). [DOI] [PubMed] [Google Scholar]
- 38.Dubinsky L, Krom BP & Meijler MM Diazirine based photoaffinity labeling. Bioorg Med Chem 20, 554–570, doi: 10.1016/j.bmc.2011.06.066 (2012). [DOI] [PubMed] [Google Scholar]
- 39.Bonora M et al. Molecular mechanisms of cell death: central implication of ATP synthase in mitochondrial permeability transition. Oncogene 34, 1608, doi: 10.1038/onc.2014.462 (2015). [DOI] [PubMed] [Google Scholar]
- 40.Brenner C & Grimm S The permeability transition pore complex in cancer cell death. Oncogene 25, 4744–4756, doi: 10.1038/sj.onc.1209609 (2006). [DOI] [PubMed] [Google Scholar]
- 41.Galluzzi L et al. Molecular mechanisms of cisplatin resistance. Oncogene 31, 1869–1883, doi: 10.1038/onc.2011.384 (2012). [DOI] [PubMed] [Google Scholar]
- 42.Mo W et al. CXCR4/CXCL12 mediate autocrine cell- cycle progression in NF1-associated malignant peripheral nerve sheath tumors. Cell 152, 1077–1090, doi: 10.1016/j.cell.2013.01.053 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Weinberg SE & Chandel NS Targeting mitochondria metabolism for cancer therapy. Nat Chem Biol 11, 9–15, doi: 10.1038/nchembio.1712 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Vander Heiden MG & DeBerardinis RJ Understanding the Intersections between Metabolism and Cancer Biology. Cell 168, doi: 10.1016/j.cell.2016.12.039 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Li F et al. Myc stimulates nuclearly encoded mitochondrial genes and mitochondrial biogenesis. Mol Cell Biol 25, 6225–6234, doi: 10.1128/MCB.25.14.6225-6234.2005 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Weinberg F et al. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc Natl Acad Sci U S A 107, 8788–8793, doi: 10.1073/pnas.1003428107 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR & Morris AD Metformin and reduced risk of cancer in diabetic patients. BMJ 330, 1304–1305, doi: 10.1136/bmj.38415.708634.F7 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Dilman VM & Anisimov VN Effect of treatment with phenformin, diphenylhydantoin or L-dopa on life span and tumour incidence in C3H/Sn mice. Gerontology 26, 241–246, doi: 10.1159/000212423 (1980). [DOI] [PubMed] [Google Scholar]
- 49.Lissanu Deribe Y et al. Mutations in the SWI/SNF complex induce a targetable dependence on oxidative phosphorylation in lung cancer. Nat Med 24, 1047–1057, doi: 10.1038/s41591-018-0019-5 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Forrest MD Why cancer cells have a more hyperpolarised mitochondrial membrane potential and emergent prospects for therapy. https://www.biorxiv.org/content/early/2015/08/21/025197 (2015).
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.