Dual NAMPT and BTK Targeting Leads to Synergistic Killing of Waldenström Macroglobulinemia Cells Regardless of MYD88 and CXCR4 Somatic Mutation Status

Purpose: Nicotinamide phosphoribosyltransferase (Nampt) regulates intracellular NAD+ pool and is highly expressed in a number of malignancies. FK866, a selective inhibitor of Nampt, depletes intracellular NAD+ levels, thereby blocking cellular metabolism and triggering sensitization to other drugs and cell death. Here we characterized the antitumor effects of Nampt inhibition in Waldenström macroglobulinemia. Experimental Design: We investigated Nampt role in MW cells using both mRNA and protein expression analyses. We have also used loss-of-function approaches to investigate the growth and survival effects of Nampt on MW cells and further tested the anti-MW activity of dual Nampt and BTK inhibition in vitro and in vivo. Results: We found that Waldenström macroglobulinemia cells exhibit high levels of Nampt compared with normal B cells. Loss of function studies suggested a potential oncogenic role of Nampt in Waldenström macroglobulinemia cells, and BTK-inhibitor ibrutinib and FK866 resulted in a significant and synergistic anti-Waldenström macroglobulinemia cell death, regardless of MYD88 and CXCR4 mutational status. Cell death was associated with: (i) activation of caspase-3, PARP and downregulation of Mcl-1, (ii) enhanced intracellular ATP and NAD+ depletion, (iii) inhibition of NF-κB signaling, and (iv) inhibition of multiple prosurvival signaling pathways. In a murine xenograft Waldenström macroglobulinemia model, low-dose combination FK866 and ibrutinib is well tolerated, significantly inhibits tumor growth, and prolongs host survival. Conclusions: Our results show intracellular NAD+ level as crucial for proliferation and survival of Waldenström macroglobulinemia cells, and provides the mechanistic preclinical rationale for targeting Nampt, either alone or with Ibrutinib, to overcome drug resistance and improve patient outcome in Waldenström macroglobulinemia. Clin Cancer Res; 22(24); 6099–109. ©2016 AACR.

[1]  S. Treon,et al.  MYD88 Mutations and Response to Ibrutinib in Waldenström's Macroglobulinemia. , 2015, The New England journal of medicine.

[2]  M. Davids,et al.  The BCL2 antagonist ABT‐199 triggers apoptosis, and augments ibrutinib and idelalisib mediated cytotoxicity in CXCR4Wild‐type and CXCR4WHIM mutated Waldenstrom macroglobulinaemia cells , 2015, British journal of haematology.

[3]  G. Damonte,et al.  APO866 Increases Antitumor Activity of Cyclosporin-A by Inducing Mitochondrial and Endoplasmic Reticulum Stress in Leukemia Cells , 2015, Clinical Cancer Research.

[4]  R. Advani,et al.  Ibrutinib in previously treated Waldenström's macroglobulinemia. , 2015, The New England journal of medicine.

[5]  G. Kroemer,et al.  MCL-1 dependency of cisplatin-resistant cancer cells. , 2014, Biochemical pharmacology.

[6]  A. Poeppl,et al.  On-Target Effect of FK866, a Nicotinamide Phosphoribosyl Transferase Inhibitor, by Apoptosis-Mediated Death in Chronic Lymphocytic Leukemia Cells , 2014, Clinical Cancer Research.

[7]  A. Roccaro,et al.  C1013G/CXCR4 acts as a driver mutation of tumor progression and modulator of drug resistance in lymphoplasmacytic lymphoma. , 2014, Blood.

[8]  M. Verheij,et al.  Antiapoptotic potency of Bcl-2 proteins primarily relies on their stability, not binding selectivity. , 2014, Blood.

[9]  N. Munshi,et al.  MYD88-independent growth and survival effects of Sp1 transactivation in Waldenstrom macroglobulinemia. , 2014, Blood.

[10]  S. Treon,et al.  The genomic landscape of Waldenstrom macroglobulinemia is characterized by highly recurring MYD88 and WHIM-like CXCR4 mutations, and small somatic deletions associated with B-cell lymphomagenesis. , 2014, Blood.

[11]  E. Hsi,et al.  MYD88 L265P somatic mutation: its usefulness in the differential diagnosis of bone marrow involvement by B-cell lymphoproliferative disorders. , 2013, American journal of clinical pathology.

[12]  N. Munshi,et al.  Intracellular NAD⁺ depletion enhances bortezomib-induced anti-myeloma activity. , 2013, Blood.

[13]  N. Gray,et al.  A mutation in MYD88 (L265P) supports the survival of lymphoplasmacytic cells by activation of Bruton tyrosine kinase in Waldenström macroglobulinemia. , 2013, Blood.

[14]  Susan R. Quinn,et al.  Bruton’s Tyrosine Kinase Mediates the Synergistic Signalling between TLR9 and the B Cell Receptor by Regulating Calcium and Calmodulin , 2013, PloS one.

[15]  Yunfang Li,et al.  Overexpression of Mcl-1 Confers Multidrug Resistance, Whereas Topoisomerase IIβ Downregulation Introduces Mitoxantrone-Specific Drug Resistance in Acute Myeloid Leukemia , 2013, Molecular Pharmacology.

[16]  A. Massarotti,et al.  Medicinal chemistry of nicotinamide phosphoribosyltransferase (NAMPT) inhibitors. , 2013, Journal of medicinal chemistry.

[17]  S. Treon,et al.  A new era for Waldenstrom macroglobulinemia: MYD88 L265P. , 2013, Blood.

[18]  N. Munshi,et al.  MYD88 L265P in Waldenström macroglobulinemia, immunoglobulin M monoclonal gammopathy, and other B-cell lymphoproliferative disorders using conventional and quantitative allele-specific polymerase chain reaction. , 2013, Blood.

[19]  F. Patrone,et al.  Intracellular NAD+ depletion induces autophagic death in multiple myeloma cells , 2013, Autophagy.

[20]  B. Quesnel,et al.  MYD88 L265P mutation in Waldenstrom macroglobulinemia. , 2012, Blood.

[21]  N. Munshi,et al.  Targeting NAD+ salvage pathway induces autophagy in multiple myeloma cells via mTORC1 and extracellular signal-regulated kinase (ERK1/2) inhibition. , 2012, Blood.

[22]  A. Krishnan,et al.  Waldenström's macroglobulinemia/lymphoplasmacytic lymphoma, version 2.2013. , 2012, Journal of the National Comprehensive Cancer Network : JNCCN.

[23]  M. Ziegler,et al.  The NAD metabolome — a key determinant of cancer cell biology , 2012, Nature Reviews Cancer.

[24]  N. Harris,et al.  MYD88 L265P somatic mutation in Waldenström's macroglobulinemia. , 2012, The New England journal of medicine.

[25]  D. Reinhardt,et al.  Involvement of p53 in the cytotoxic activity of the NAMPT inhibitor FK866 in myeloid leukemic cells , 2012, International journal of cancer.

[26]  Wei Zhao,et al.  Overexpression of Nampt in gastric cancer and chemopotentiating effects of the Nampt inhibitor FK866 in combination with fluorouracil. , 2011, Oncology reports.

[27]  P. Canonico,et al.  Reciprocal Potentiation of the Antitumoral Activities of FK866, an Inhibitor of Nicotinamide Phosphoribosyltransferase, and Etoposide or Cisplatin in Neuroblastoma Cells , 2011, Journal of Pharmacology and Experimental Therapeutics.

[28]  Jeffrey A Jones,et al.  Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. , 2011, Blood.

[29]  D. Hanahan,et al.  Hallmarks of Cancer: The Next Generation , 2011, Cell.

[30]  Adam R. Johnson,et al.  Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7 , 2011, Nature.

[31]  T. Mak,et al.  Regulation of cancer cell metabolism , 2011, Nature Reviews Cancer.

[32]  C. Flowers,et al.  Tipifarnib sensitizes cells to proteasome inhibition by blocking degradation of bortezomib-induced aggresomes. , 2010, Blood.

[33]  A. Ballestrero,et al.  Potent synergistic interaction between the Nampt inhibitor APO866 and the apoptosis activator TRAIL in human leukemia cells. , 2010, Experimental hematology.

[34]  J. Auwerx,et al.  The secret life of NAD+: an old metabolite controlling new metabolic signaling pathways. , 2010, Endocrine reviews.

[35]  Charles P. Lin,et al.  Dual targeting of the PI3K/Akt/mTOR pathway as an antitumor strategy in Waldenstrom macroglobulinemia. , 2010, Blood.

[36]  A. Ballestrero,et al.  Catastrophic NAD+ Depletion in Activated T Lymphocytes through Nampt Inhibition Reduces Demyelination and Disability in EAE , 2009, PloS one.

[37]  L. Cantley,et al.  Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation , 2009, Science.

[38]  P. Greaney,et al.  The NAD biosynthesis inhibitor APO866 has potent antitumor activity against hematologic malignancies. , 2009, Blood.

[39]  Dudley Lamming,et al.  Nutrient-Sensitive Mitochondrial NAD+ Levels Dictate Cell Survival , 2007, Cell.

[40]  M. Hasmann,et al.  Chemopotentiating effects of a novel NAD biosynthesis inhibitor, FK866, in combination with antineoplastic agents. , 2006, European journal of medical research.

[41]  J. Koutcher,et al.  Metabolic Signatures Associated with a NAD Synthesis Inhibitor–Induced Tumor Apoptosis Identified by 1H-Decoupled-31P Magnetic Resonance Spectroscopy , 2005, Clinical Cancer Research.

[42]  Mathias Ziegler,et al.  The new life of a centenarian: signalling functions of NAD(P). , 2004, Trends in biochemical sciences.

[43]  Max Hasmann,et al.  FK866, a highly specific noncompetitive inhibitor of nicotinamide phosphoribosyltransferase, represents a novel mechanism for induction of tumor cell apoptosis. , 2003, Cancer research.

[44]  W T McGivney,et al.  The National Comprehensive Cancer Network , 1998, Cancer.

[45]  N. Chiorazzi,et al.  mechanisms of disease Chronic Lymphocytic Leukemia , 2010 .

[46]  D. Rossi,et al.  Extracellular nicotinamide phosphoribosyltransferase (NAMPT) promotes M2 macrophage polarization in chronic lymphocytic leukemia. , 2015, Blood.

[47]  T. Chou,et al.  Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. , 1984, Advances in enzyme regulation.