Catalytic mTOR inhibitors can overcome intrinsic and acquired resistance to allosteric mTOR inhibitors

We tested the antitumor efficacy of mTOR catalytic site inhibitor MLN0128 in models with intrinsic or acquired rapamycin-resistance. Cell lines that were intrinsically rapamycin-resistant as well as those that were intrinsically rapamycinsensitive were sensitive to MLN0128 in vitro. MLN0128 inhibited both mTORC1 and mTORC2 signaling, with more robust inhibition of downstream 4E-BP1 phosphorylation and cap-dependent translation compared to rapamycin in vitro. Rapamycin-sensitive BT474 cell line acquired rapamycin resistance (BT474 RR) with prolonged rapamycin treatment in vitro. This cell line acquired an mTOR mutation (S2035F) in the FKBP12-rapamycin binding domain; mTORC1 signaling was not inhibited by rapalogs but was inhibited by MLN0128. In BT474 RR cells, MLN0128 had significantly higher growth inhibition compared to rapamycin in vitro and in vivo. Our results demonstrate that MLN0128 may be effective in tumors with intrinsic as well as acquired rapalog resistance. mTOR mutations are a mechanism of acquired resistance in vitro; the clinical relevance of this observation needs to be further evaluated.

[1]  S. Keir,et al.  Initial testing (stage 1) of the investigational mTOR kinase inhibitor MLN0128 by the pediatric preclinical testing program , 2014, Pediatric blood & cancer.

[2]  R. Nolley,et al.  Preclinical trial of a new dual mTOR inhibitor, MLN0128, using renal cell carcinoma tumorgrafts , 2014, International journal of cancer.

[3]  S. Cook,et al.  Adaptation to mTOR kinase inhibitors by amplification of eIF4E to maintain cap-dependent translation , 2014, Journal of Cell Science.

[4]  K. Shokat,et al.  Generation of a patient-derived chordoma xenograft and characterization of the phosphoproteome in a recurrent chordoma. , 2014, Journal of neurosurgery.

[5]  D. Kwiatkowski,et al.  Equivalent Benefit of Rapamycin and a Potent mTOR ATP-Competitive Inhibitor, MLN0128 (INK128), in a Mouse Model of Tuberous Sclerosis , 2013, Molecular Cancer Research.

[6]  M. Blagosklonny Rapalogs in cancer prevention , 2012, Cancer biology & therapy.

[7]  L. Donehower Rapamycin as longevity enhancer and cancer preventative agent in the context of p53 deficiency , 2012, Aging.

[8]  F. Sotgia,et al.  Caveolin-1 and accelerated host aging in the breast tumor microenvironment: chemoprevention with rapamycin, an mTOR inhibitor and anti-aging drug. , 2012, The American journal of pathology.

[9]  G. Mills,et al.  PIK3CA/PTEN Mutations and Akt Activation As Markers of Sensitivity to Allosteric mTOR Inhibitors , 2012, Clinical Cancer Research.

[10]  Christopher A. Miller,et al.  VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. , 2012, Genome research.

[11]  N. Ilić,et al.  PI3K-targeted therapy can be evaded by gene amplification along the MYC-eukaryotic translation initiation factor 4E (eIF4E) axis , 2011, Proceedings of the National Academy of Sciences.

[12]  Sarat Chandarlapaty,et al.  mTOR kinase inhibition causes feedback-dependent biphasic regulation of AKT signaling. , 2011, Cancer discovery.

[13]  A. Gonzalez-Perez,et al.  Improving the assessment of the outcome of nonsynonymous SNVs with a consensus deleteriousness score, Condel. , 2011, American journal of human genetics.

[14]  Mingming Jia,et al.  COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer , 2010, Nucleic Acids Res..

[15]  J. Manola,et al.  Combination mTOR and IGF-1R Inhibition: Phase I Trial of Everolimus and Figitumumab in Patients with Advanced Sarcomas and Other Solid Tumors , 2010, Clinical Cancer Research.

[16]  Funda Meric-Bernstam,et al.  Deciphering the role of PI3K/Akt/mTOR pathway in breast cancer biology and pathogenesis. , 2010, Clinical breast cancer.

[17]  D. Sabatini,et al.  Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress. , 2010, Molecular cell.

[18]  G. Mills,et al.  Rapamycin Regulates Stearoyl CoA Desaturase 1 Expression in Breast Cancer , 2010, Molecular Cancer Therapeutics.

[19]  H. Hakonarson,et al.  ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data , 2010, Nucleic acids research.

[20]  D. Guertin,et al.  Targeting mTOR: prospects for mTOR complex 2 inhibitors in cancer therapy , 2010, Oncogene.

[21]  Daniel Rios,et al.  Bioinformatics Applications Note Databases and Ontologies Deriving the Consequences of Genomic Variants with the Ensembl Api and Snp Effect Predictor , 2022 .

[22]  N. Sonenberg,et al.  mTORC1-Mediated Cell Proliferation, But Not Cell Growth, Controlled by the 4E-BPs , 2010, Science.

[23]  P. Bork,et al.  A method and server for predicting damaging missense mutations , 2010, Nature Methods.

[24]  K. Shokat,et al.  New inhibitors of the PI3K-Akt-mTOR pathway: insights into mTOR signaling from a new generation of Tor Kinase Domain Inhibitors (TORKinibs). , 2010, Current topics in microbiology and immunology.

[25]  G. Hortobagyi,et al.  Loss of HER2 Amplification Following Trastuzumab-Based Neoadjuvant Systemic Therapy and Survival Outcomes , 2009, Clinical Cancer Research.

[26]  S. Tsavachidis,et al.  The rapamycin-regulated gene expression signature determines prognosis for breast cancer , 2009, Molecular Cancer.

[27]  Funda Meric-Bernstam,et al.  Targeting the mTOR signaling network for cancer therapy. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[28]  C. Chresta,et al.  Ku-0063794 is a specific inhibitor of the mammalian target of rapamycin (mTOR) , 2009, The Biochemical journal.

[29]  Robbie Loewith,et al.  Active-Site Inhibitors of mTOR Target Rapamycin-Resistant Outputs of mTORC1 and mTORC2 , 2009, PLoS biology.

[30]  S. Henikoff,et al.  Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm , 2009, Nature Protocols.

[31]  Zhi Hu,et al.  An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. , 2008, Cancer research.

[32]  Gopal Singh,et al.  eIF4E knockdown decreases breast cancer cell growth without activating Akt signaling , 2008, Molecular Cancer Therapeutics.

[33]  T. Harris,et al.  Regulation of Proline-rich Akt Substrate of 40 kDa (PRAS40) Function by Mammalian Target of Rapamycin Complex 1 (mTORC1)-mediated Phosphorylation* , 2008, Journal of Biological Chemistry.

[34]  F. Meric-Bernstam,et al.  Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. , 2007, Endocrine-related cancer.

[35]  Yu Li,et al.  Identification of IRS-1 Ser-1101 as a target of S6K1 in nutrient- and obesity-induced insulin resistance , 2007, Proceedings of the National Academy of Sciences.

[36]  D. Alessi,et al.  Identification of Protor as a novel Rictor-binding component of mTOR complex-2. , 2007, The Biochemical journal.

[37]  David M Sabatini,et al.  Defining the role of mTOR in cancer. , 2007, Cancer cell.

[38]  Joon-Oh Park,et al.  MET Amplification Leads to Gefitinib Resistance in Lung Cancer by Activating ERBB3 Signaling , 2007, Science.

[39]  R. Pearson,et al.  Coordinate regulation of ribosome biogenesis and function by the ribosomal protein S6 kinase, a key mediator of mTOR function , 2007, Growth factors.

[40]  C. Proud,et al.  Methods for studying signal-dependent regulation of translation factor activity. , 2007, Methods in enzymology.

[41]  K. Inoki,et al.  Identification of Sin1 as an essential TORC2 component required for complex formation and kinase activity. , 2006, Genes & development.

[42]  J. Qin,et al.  SIN1/MIP1 Maintains rictor-mTOR Complex Integrity and Regulates Akt Phosphorylation and Substrate Specificity , 2006, Cell.

[43]  Jacob D. Jaffe,et al.  mSin1 Is Necessary for Akt/PKB Phosphorylation, and Its Isoforms Define Three Distinct mTORC2s , 2006, Current Biology.

[44]  A. Lièvre,et al.  KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. , 2006, Cancer research.

[45]  M. Hall,et al.  TOR Signaling in Growth and Metabolism , 2006, Cell.

[46]  Gordon B Mills,et al.  mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. , 2006, Cancer research.

[47]  R. Hresko,et al.  mTOR·RICTOR Is the Ser473 Kinase for Akt/Protein Kinase B in 3T3-L1 Adipocytes* , 2005, Journal of Biological Chemistry.

[48]  Huajun Yan,et al.  Mammalian target of rapamycin inhibitors activate the AKT kinase in multiple myeloma cells by up-regulating the insulin-like growth factor receptor/insulin receptor substrate-1/phosphatidylinositol 3-kinase cascade , 2005, Molecular Cancer Therapeutics.

[49]  D. McNabb,et al.  Dual Luciferase Assay System for Rapid Assessment of Gene Expression in Saccharomyces cerevisiae , 2005, Eukaryotic Cell.

[50]  Wolfgang Eiermann,et al.  Phase II study of temsirolimus (CCI-779), a novel inhibitor of mTOR, in heavily pretreated patients with locally advanced or metastatic breast cancer. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[51]  M. Murakami,et al.  Distinct Signaling Events Downstream of mTOR Cooperate To Mediate the Effects of Amino Acids and Insulin on Initiation Factor 4E-Binding Proteins , 2005, Molecular and Cellular Biology.

[52]  M. Meyerson,et al.  EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. , 2005, The New England journal of medicine.

[53]  H. Varmus,et al.  Acquired Resistance of Lung Adenocarcinomas to Gefitinib or Erlotinib Is Associated with a Second Mutation in the EGFR Kinase Domain , 2005, PLoS medicine.

[54]  D. Guertin,et al.  Phosphorylation and Regulation of Akt/PKB by the Rictor-mTOR Complex , 2005, Science.

[55]  G. Mills,et al.  Targeting Mammalian Target of Rapamycin Synergistically Enhances Chemotherapy-Induced Cytotoxicity in Breast Cancer Cells , 2004, Clinical Cancer Research.

[56]  C. Sawyers,et al.  The phosphatidylinositol 3-Kinase–AKT pathway in human cancer , 2002, Nature Reviews Cancer.

[57]  P. Houghton,et al.  4E-binding Proteins, the Suppressors of Eukaryotic Initiation Factor 4E, Are Down-regulated in Cells with Acquired or Intrinsic Resistance to Rapamycin* , 2002, The Journal of Biological Chemistry.

[58]  P. N. Rao,et al.  Clinical Resistance to STI-571 Cancer Therapy Caused by BCR-ABL Gene Mutation or Amplification , 2001, Science.

[59]  A. Gingras,et al.  Regulation of translation initiation by FRAP/mTOR. , 2001, Genes & development.

[60]  M. Kasuga,et al.  Regulation of eIF-4E BP1 Phosphorylation by mTOR* , 1997, The Journal of Biological Chemistry.

[61]  Christine C. Hudson,et al.  Phosphorylation of the translational repressor PHAS-I by the mammalian target of rapamycin. , 1997, Science.

[62]  J. Heitman,et al.  TOR Mutations Confer Rapamycin Resistance by Preventing Interaction with FKBP12-Rapamycin (*) , 1995, The Journal of Biological Chemistry.

[63]  S. Schreiber,et al.  Control of p70 S6 kinase by kinase activity of FRAP in vivo , 1995, Nature.

[64]  S. Schreiber,et al.  Identification of an 11-kDa FKBP12-rapamycin-binding domain within the 289-kDa FKBP12-rapamycin-associated protein and characterization of a critical serine residue. , 1995, Proceedings of the National Academy of Sciences of the United States of America.