Skip to main content
Top
Published in: Cellular Oncology 1/2017

01-02-2017 | Review

Novel strategies for targeting leukemia stem cells: sounding the death knell for blood cancer

Authors: Antonieta Chavez-Gonzalez, Babak Bakhshinejad, Katayoon Pakravan, Monica L. Guzman, Sadegh Babashah

Published in: Cellular Oncology | Issue 1/2017

Login to get access

Abstract

Background

Cancer stem cells (CSCs), also known as tumor-initiating cells (TICs), are characterized by high self-renewal and multi-lineage differentiation capacities. CSCs are thought to play indispensable roles in the initiation, progression and metastasis of many types of cancer. Leukemias are thought to be initiated and maintained by a specific sub-type of CSC, the leukemia stem cell (LSC). An important feature of LSCs is their resistance to standard therapy, which may lead to relapse. Increasing efforts are aimed at developing novel therapeutic strategies that selectively target LSCs, while sparing their normal counterparts and, thus, minimizing adverse treatment-associated side-effects. These LSC targeting therapies aim to eradicate LSCs through affecting mechanisms that control their survival, self-renewal, differentiation, proliferation and cell cycle progression. Some LSC targeting therapies have already been proven successful in pre-clinical studies and they are now being tested in clinical studies, mainly in combination with conventional treatment regimens.

Conclusions

A growing body of evidence indicates that the selective targeting of LSCs represents a promising approach to improve disease outcome. Beyond doubt, the CSC hypothesis has added a new dimension to the area of anticancer research, thereby paving the way for shaping a new trend in cancer therapy.
Appendix
Available only for authorised users
Literature
3.
go back to reference D.C. Taussig, F. Miraki-Moud, F. Anjos-Afonso, D.J. Pearce, K. Allen, C. Ridler, D. Lillington, H. Oakervee, J. Cavenagh, S.G. Agrawal, T.A. Lister, J.G. Gribben, D. Bonnet, Anti-CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initiating cells. Blood 112, 568–575 (2008). doi:10.1182/blood-2007-10-118331 CrossRefPubMed D.C. Taussig, F. Miraki-Moud, F. Anjos-Afonso, D.J. Pearce, K. Allen, C. Ridler, D. Lillington, H. Oakervee, J. Cavenagh, S.G. Agrawal, T.A. Lister, J.G. Gribben, D. Bonnet, Anti-CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initiating cells. Blood 112, 568–575 (2008). doi:10.​1182/​blood-2007-10-118331 CrossRefPubMed
4.
go back to reference D.C. Taussig, J. Vargaftig, F. Miraki-Moud, E. Griessinger, K. Sharrock, T. Luke, D. Lillington, H. Oakervee, J. Cavenagh, S.G. Agrawal, T.A. Lister, J.G. Gribben, D. Bonnet, Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34(-) fraction. Blood 115, 1976–1984 (2010). doi:10.1182/blood-2009-02-206565 CrossRefPubMedPubMedCentral D.C. Taussig, J. Vargaftig, F. Miraki-Moud, E. Griessinger, K. Sharrock, T. Luke, D. Lillington, H. Oakervee, J. Cavenagh, S.G. Agrawal, T.A. Lister, J.G. Gribben, D. Bonnet, Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34(-) fraction. Blood 115, 1976–1984 (2010). doi:10.​1182/​blood-2009-02-206565 CrossRefPubMedPubMedCentral
5.
go back to reference S. Aref, O. Salama, Y. Al-Tonbary, M. Fouda, A. Menessy, M. El-Sherbiny, L and E selectins in acute myeloid leukemia: expression, clinical relevance and relation to patient outcome. Hematology 7, 83–87 (2002). doi:10.1080/10245330290028579 CrossRefPubMed S. Aref, O. Salama, Y. Al-Tonbary, M. Fouda, A. Menessy, M. El-Sherbiny, L and E selectins in acute myeloid leukemia: expression, clinical relevance and relation to patient outcome. Hematology 7, 83–87 (2002). doi:10.​1080/​1024533029002857​9 CrossRefPubMed
7.
go back to reference F. Ishikawa, S. Yoshida, Y. Saito, A. Hijikata, H. Kitamura, S. Tanaka, R. Nakamura, T. Tanaka, H. Tomiyama, N. Saito, M. Fukata, T. Miyamoto, B. Lyons, K. Ohshima, N. Uchida, S. Taniguchi, O. Ohara, K. Akashi, M. Harada, L.D. Shultz, Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat. Biotechnol. 25, 1315–1321 (2007). doi:10.1038/nbt1350 CrossRefPubMed F. Ishikawa, S. Yoshida, Y. Saito, A. Hijikata, H. Kitamura, S. Tanaka, R. Nakamura, T. Tanaka, H. Tomiyama, N. Saito, M. Fukata, T. Miyamoto, B. Lyons, K. Ohshima, N. Uchida, S. Taniguchi, O. Ohara, K. Akashi, M. Harada, L.D. Shultz, Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat. Biotechnol. 25, 1315–1321 (2007). doi:10.​1038/​nbt1350 CrossRefPubMed
8.
go back to reference T. Holyoake, X. Jiang, C. Eaves, A. Eaves, Isolation of a highly quiescent subpopulation of primitive leukemic cells in chronic myeloid leukemia. Blood 94, 2056–2064 (1999)PubMed T. Holyoake, X. Jiang, C. Eaves, A. Eaves, Isolation of a highly quiescent subpopulation of primitive leukemic cells in chronic myeloid leukemia. Blood 94, 2056–2064 (1999)PubMed
9.
go back to reference C. Zhao, A. Chen, C.H. Jamieson, M. Fereshteh, A. Abrahamsson, J. Blum, H.Y. Kwon, J. Kim, J.P. Chute, D. Rizzieri, M. Munchhof, T. VanArsdale, P.A. Beachy, T. Reya, Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature 458, 776–779 (2009). doi:10.1038/nature07737 CrossRefPubMedPubMedCentral C. Zhao, A. Chen, C.H. Jamieson, M. Fereshteh, A. Abrahamsson, J. Blum, H.Y. Kwon, J. Kim, J.P. Chute, D. Rizzieri, M. Munchhof, T. VanArsdale, P.A. Beachy, T. Reya, Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature 458, 776–779 (2009). doi:10.​1038/​nature07737 CrossRefPubMedPubMedCentral
11.
go back to reference C.T. Jordan, D. Upchurch, S.J. Szilvassy, M.L. Guzman, D.S. Howard, A.L. Pettigrew, T. Meyerrose, R. Rossi, B. Grimes, D.A. Rizzieri, S.M. Luger, G.L. Phillips, The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia 14, 1777–1784 (2000)CrossRefPubMed C.T. Jordan, D. Upchurch, S.J. Szilvassy, M.L. Guzman, D.S. Howard, A.L. Pettigrew, T. Meyerrose, R. Rossi, B. Grimes, D.A. Rizzieri, S.M. Luger, G.L. Phillips, The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia 14, 1777–1784 (2000)CrossRefPubMed
14.
go back to reference Y. Kikushige, T. Shima, S. Takayanagi, S. Urata, T. Miyamoto, H. Iwasaki, K. Takenaka, T. Teshima, T. Tanaka, Y. Inagaki, K. Akashi, TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell 7, 708–717 (2010). doi:10.1016/j.stem.2010.11.014 CrossRefPubMed Y. Kikushige, T. Shima, S. Takayanagi, S. Urata, T. Miyamoto, H. Iwasaki, K. Takenaka, T. Teshima, T. Tanaka, Y. Inagaki, K. Akashi, TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell 7, 708–717 (2010). doi:10.​1016/​j.​stem.​2010.​11.​014 CrossRefPubMed
15.
go back to reference M.P. Scolnik, R. Morilla, M.M. de Bracco, D. Catovsky, E. Matutes, CD34 and CD117 are overexpressed in AML and may be valuable to detect minimal residual disease. Leuk. Res. 26, 615–619 (2002)CrossRefPubMed M.P. Scolnik, R. Morilla, M.M. de Bracco, D. Catovsky, E. Matutes, CD34 and CD117 are overexpressed in AML and may be valuable to detect minimal residual disease. Leuk. Res. 26, 615–619 (2002)CrossRefPubMed
16.
go back to reference A. Chavez-Gonzalez, E. Dorantes-Acosta, D. Moreno-Lorenzana, A. Alvarado-Moreno, L. Arriaga-Pizano, H. Mayani, Expression of CD90, CD96, CD117, and CD123 on different hematopoietic cell populations from pediatric patients with acute myeloid leukemia. Arch. Med. Res. 45, 343–350 (2014). doi:10.1016/j.arcmed.2014.04.001 CrossRefPubMed A. Chavez-Gonzalez, E. Dorantes-Acosta, D. Moreno-Lorenzana, A. Alvarado-Moreno, L. Arriaga-Pizano, H. Mayani, Expression of CD90, CD96, CD117, and CD123 on different hematopoietic cell populations from pediatric patients with acute myeloid leukemia. Arch. Med. Res. 45, 343–350 (2014). doi:10.​1016/​j.​arcmed.​2014.​04.​001 CrossRefPubMed
17.
go back to reference J.M. Gerber, B.D. Smith, B. Ngwang, H. Zhang, M.S. Vala, L. Morsberger, S. Galkin, M.I. Collector, B. Perkins, M.J. Levis, C.A. Griffin, S.J. Sharkis, M.J. Borowitz, J.E. Karp, R.J. Jones, A clinically relevant population of leukemic CD34(+)CD38(-) cells in acute myeloid leukemia. Blood 119, 3571–3577 (2012). doi:10.1182/blood-2011-06-364182 CrossRefPubMedPubMedCentral J.M. Gerber, B.D. Smith, B. Ngwang, H. Zhang, M.S. Vala, L. Morsberger, S. Galkin, M.I. Collector, B. Perkins, M.J. Levis, C.A. Griffin, S.J. Sharkis, M.J. Borowitz, J.E. Karp, R.J. Jones, A clinically relevant population of leukemic CD34(+)CD38(-) cells in acute myeloid leukemia. Blood 119, 3571–3577 (2012). doi:10.​1182/​blood-2011-06-364182 CrossRefPubMedPubMedCentral
18.
go back to reference M. Jaras, P. Johnels, N. Hansen, H. Agerstam, P. Tsapogas, M. Rissler, C. Lassen, T. Olofsson, O.W. Bjerrum, J. Richter, T. Fioretos, Isolation and killing of candidate chronic myeloid leukemia stem cells by antibody targeting of IL-1 receptor accessory protein. Proc. Natl. Acad. Sci. U. S. A. 107, 16280–16285 (2010). doi:10.1073/pnas.1004408107 CrossRefPubMedPubMedCentral M. Jaras, P. Johnels, N. Hansen, H. Agerstam, P. Tsapogas, M. Rissler, C. Lassen, T. Olofsson, O.W. Bjerrum, J. Richter, T. Fioretos, Isolation and killing of candidate chronic myeloid leukemia stem cells by antibody targeting of IL-1 receptor accessory protein. Proc. Natl. Acad. Sci. U. S. A. 107, 16280–16285 (2010). doi:10.​1073/​pnas.​1004408107 CrossRefPubMedPubMedCentral
19.
go back to reference H. Herrmann, I. Sadovnik, S. Cerny-Reiterer, T. Rulicke, G. Stefanzl, M. Willmann, G. Hoermann, M. Bilban, K. Blatt, S. Herndlhofer, M. Mayerhofer, B. Streubel, W.R. Sperr, T.L. Holyoake, C. Mannhalter, P. Valent, Dipeptidylpeptidase IV (CD26) defines leukemic stem cells (LSC) in chronic myeloid leukemia. Blood 123, 3951–3962 (2014). doi:10.1182/blood-2013-10-536078 CrossRefPubMed H. Herrmann, I. Sadovnik, S. Cerny-Reiterer, T. Rulicke, G. Stefanzl, M. Willmann, G. Hoermann, M. Bilban, K. Blatt, S. Herndlhofer, M. Mayerhofer, B. Streubel, W.R. Sperr, T.L. Holyoake, C. Mannhalter, P. Valent, Dipeptidylpeptidase IV (CD26) defines leukemic stem cells (LSC) in chronic myeloid leukemia. Blood 123, 3951–3962 (2014). doi:10.​1182/​blood-2013-10-536078 CrossRefPubMed
20.
go back to reference M.E. Chamuleau, G.J. Ossenkoppele, A. van Rhenen, L. van Dreunen, S.M. Jirka, A. Zevenbergen, G.J. Schuurhuis, A.A. van de Loosdrecht, High TRAIL-R3 expression on leukemic blasts is associated with poor outcome and induces apoptosis-resistance which can be overcome by targeting TRAIL-R2. Leuk. Res. 35, 741–749 (2011). doi:10.1016/j.leukres.2010.12.032 CrossRefPubMed M.E. Chamuleau, G.J. Ossenkoppele, A. van Rhenen, L. van Dreunen, S.M. Jirka, A. Zevenbergen, G.J. Schuurhuis, A.A. van de Loosdrecht, High TRAIL-R3 expression on leukemic blasts is associated with poor outcome and induces apoptosis-resistance which can be overcome by targeting TRAIL-R2. Leuk. Res. 35, 741–749 (2011). doi:10.​1016/​j.​leukres.​2010.​12.​032 CrossRefPubMed
21.
go back to reference M. Terwijn, N. Feller, A. van Rhenen, A. Kelder, G. Westra, S. Zweegman, G. Ossenkoppele, G.J. Schuurhuis, Interleukin-2 receptor alpha-chain (CD25) expression on leukaemic blasts is predictive for outcome and level of residual disease in AML. Eur. J. Cancer 45, 1692–1699 (2009). doi:10.1016/j.ejca.2009.02.021 CrossRefPubMed M. Terwijn, N. Feller, A. van Rhenen, A. Kelder, G. Westra, S. Zweegman, G. Ossenkoppele, G.J. Schuurhuis, Interleukin-2 receptor alpha-chain (CD25) expression on leukaemic blasts is predictive for outcome and level of residual disease in AML. Eur. J. Cancer 45, 1692–1699 (2009). doi:10.​1016/​j.​ejca.​2009.​02.​021 CrossRefPubMed
22.
go back to reference M. Terwijn, W. Zeijlemaker, A. Kelder, A.P. Rutten, A.N. Snel, W.J. Scholten, T. Pabst, G. Verhoef, B. Lowenberg, S. Zweegman, G.J. Ossenkoppele, G.J. Schuurhuis, Leukemic stem cell frequency: a strong biomarker for clinical outcome in acute myeloid leukemia. PLoS One 9, e107587 (2014). doi:10.1371/journal.pone.0107587 CrossRefPubMedPubMedCentral M. Terwijn, W. Zeijlemaker, A. Kelder, A.P. Rutten, A.N. Snel, W.J. Scholten, T. Pabst, G. Verhoef, B. Lowenberg, S. Zweegman, G.J. Ossenkoppele, G.J. Schuurhuis, Leukemic stem cell frequency: a strong biomarker for clinical outcome in acute myeloid leukemia. PLoS One 9, e107587 (2014). doi:10.​1371/​journal.​pone.​0107587 CrossRefPubMedPubMedCentral
23.
go back to reference A.V. Krivtsov, D. Twomey, Z. Feng, M.C. Stubbs, Y. Wang, J. Faber, J.E. Levine, J. Wang, W.C. Hahn, D.G. Gilliland, T.R. Golub, S.A. Armstrong, Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 442, 818–822 (2006). doi:10.1038/nature04980 CrossRefPubMed A.V. Krivtsov, D. Twomey, Z. Feng, M.C. Stubbs, Y. Wang, J. Faber, J.E. Levine, J. Wang, W.C. Hahn, D.G. Gilliland, T.R. Golub, S.A. Armstrong, Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 442, 818–822 (2006). doi:10.​1038/​nature04980 CrossRefPubMed
24.
go back to reference L.I. Shlush, S. Zandi, A. Mitchell, W.C. Chen, J.M. Brandwein, V. Gupta, J.A. Kennedy, A.D. Schimmer, A.C. Schuh, K.W. Yee, J.L. McLeod, M. Doedens, J.J. Medeiros, R. Marke, H.J. Kim, K. Lee, J.D. McPherson, T.J. Hudson, A.M. Brown, F. Yousif, Q.M. Trinh, L.D. Stein, M.D. Minden, J.C. Wang, J.E. Dick, Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 506, 328–333 (2014). doi:10.1038/nature13038 CrossRefPubMedPubMedCentral L.I. Shlush, S. Zandi, A. Mitchell, W.C. Chen, J.M. Brandwein, V. Gupta, J.A. Kennedy, A.D. Schimmer, A.C. Schuh, K.W. Yee, J.L. McLeod, M. Doedens, J.J. Medeiros, R. Marke, H.J. Kim, K. Lee, J.D. McPherson, T.J. Hudson, A.M. Brown, F. Yousif, Q.M. Trinh, L.D. Stein, M.D. Minden, J.C. Wang, J.E. Dick, Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 506, 328–333 (2014). doi:10.​1038/​nature13038 CrossRefPubMedPubMedCentral
26.
go back to reference S. Babashah, Cancer stem cells: emerging concepts and future perspectives in translational oncology (Springer International Publishing, Switzerland, 2015)CrossRef S. Babashah, Cancer stem cells: emerging concepts and future perspectives in translational oncology (Springer International Publishing, Switzerland, 2015)CrossRef
27.
go back to reference P.R. Hamann, L.M. Hinman, I. Hollander, C.F. Beyer, D. Lindh, R. Holcomb, W. Hallett, H.R. Tsou, J. Upeslacis, D. Shochat, A. Mountain, D.A. Flowers, I. Bernstein, Gemtuzumab ozogamicin, a potent and selective anti-CD33 antibody-calicheamicin conjugate for treatment of acute myeloid leukemia. Bioconjug. Chem. 13, 47–58 (2002)CrossRefPubMed P.R. Hamann, L.M. Hinman, I. Hollander, C.F. Beyer, D. Lindh, R. Holcomb, W. Hallett, H.R. Tsou, J. Upeslacis, D. Shochat, A. Mountain, D.A. Flowers, I. Bernstein, Gemtuzumab ozogamicin, a potent and selective anti-CD33 antibody-calicheamicin conjugate for treatment of acute myeloid leukemia. Bioconjug. Chem. 13, 47–58 (2002)CrossRefPubMed
29.
go back to reference A. Dutour, V. Marin, I. Pizzitola, S. Valsesia-Wittmann, D. Lee, E. Yvon, H. Finney, A. Lawson, M. Brenner, A. Biondi, E. Biagi, R. Rousseau, In vitro and in vivo antitumor effect of anti-CD33 chimeric receptor-expressing EBV-CTL against CD33 acute myeloid leukemia. Adv. Hematol. 2012, 683065 (2012). doi:10.1155/2012/683065 CrossRefPubMedPubMedCentral A. Dutour, V. Marin, I. Pizzitola, S. Valsesia-Wittmann, D. Lee, E. Yvon, H. Finney, A. Lawson, M. Brenner, A. Biondi, E. Biagi, R. Rousseau, In vitro and in vivo antitumor effect of anti-CD33 chimeric receptor-expressing EBV-CTL against CD33 acute myeloid leukemia. Adv. Hematol. 2012, 683065 (2012). doi:10.​1155/​2012/​683065 CrossRefPubMedPubMedCentral
30.
go back to reference I. Pizzitola, V. Agostoni, E. Cribioli, M. Pule, R. Rousseau, H. Finney, A. Lawson, A. Biondi, E. Biagi, V. Marin, In vitro comparison of three different chimeric receptor-modified effector T-cell populations for leukemia cell therapy. J. Immunother. 34, 469–479 (2011). doi:10.1097/CJI.0b013e31821e763b CrossRefPubMed I. Pizzitola, V. Agostoni, E. Cribioli, M. Pule, R. Rousseau, H. Finney, A. Lawson, A. Biondi, E. Biagi, V. Marin, In vitro comparison of three different chimeric receptor-modified effector T-cell populations for leukemia cell therapy. J. Immunother. 34, 469–479 (2011). doi:10.​1097/​CJI.​0b013e31821e763b​ CrossRefPubMed
31.
go back to reference M. Feuring-Buske, A.E. Frankel, R.L. Alexander, B. Gerhard, D.E. Hogge, A diphtheria toxin-interleukin 3 fusion protein is cytotoxic to primitive acute myeloid leukemia progenitors but spares normal progenitors. Cancer Res. 62, 1730–1736 (2002)PubMed M. Feuring-Buske, A.E. Frankel, R.L. Alexander, B. Gerhard, D.E. Hogge, A diphtheria toxin-interleukin 3 fusion protein is cytotoxic to primitive acute myeloid leukemia progenitors but spares normal progenitors. Cancer Res. 62, 1730–1736 (2002)PubMed
33.
go back to reference L. Jin, E.M. Lee, H.S. Ramshaw, S.J. Busfield, A.G. Peoppl, L. Wilkinson, M.A. Guthridge, D. Thomas, E.F. Barry, A. Boyd, D.P. Gearing, G. Vairo, A.F. Lopez, J.E. Dick, R.B. Lock, Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Cell Stem Cell 5, 31–42 (2009). doi:10.1016/j.stem.2009.04.018 CrossRefPubMed L. Jin, E.M. Lee, H.S. Ramshaw, S.J. Busfield, A.G. Peoppl, L. Wilkinson, M.A. Guthridge, D. Thomas, E.F. Barry, A. Boyd, D.P. Gearing, G. Vairo, A.F. Lopez, J.E. Dick, R.B. Lock, Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Cell Stem Cell 5, 31–42 (2009). doi:10.​1016/​j.​stem.​2009.​04.​018 CrossRefPubMed
34.
go back to reference S.J. Busfield, M. Biondo, M. Wong, H.S. Ramshaw, E.M. Lee, S. Ghosh, H. Braley, C. Panousis, A.W. Roberts, S.Z. He, D. Thomas, L. Fabri, G. Vairo, R.B. Lock, A.F. Lopez, A.D. Nash, Targeting of acute myeloid leukemia in vitro and in vivo with an anti-CD123 mAb engineered for optimal ADCC. Leukemia 28, 2213–2221 (2014). doi:10.1038/leu.2014.128 CrossRefPubMed S.J. Busfield, M. Biondo, M. Wong, H.S. Ramshaw, E.M. Lee, S. Ghosh, H. Braley, C. Panousis, A.W. Roberts, S.Z. He, D. Thomas, L. Fabri, G. Vairo, R.B. Lock, A.F. Lopez, A.D. Nash, Targeting of acute myeloid leukemia in vitro and in vivo with an anti-CD123 mAb engineered for optimal ADCC. Leukemia 28, 2213–2221 (2014). doi:10.​1038/​leu.​2014.​128 CrossRefPubMed
35.
go back to reference I. Pizzitola, F. Anjos-Afonso, K. Rouault-Pierre, F. Lassailly, S. Tettamanti, O. Spinelli, A. Biondi, E. Biagi, D. Bonnet, Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo. Leukemia 28, 1596–1605 (2014). doi:10.1038/leu.2014.62 CrossRefPubMed I. Pizzitola, F. Anjos-Afonso, K. Rouault-Pierre, F. Lassailly, S. Tettamanti, O. Spinelli, A. Biondi, E. Biagi, D. Bonnet, Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo. Leukemia 28, 1596–1605 (2014). doi:10.​1038/​leu.​2014.​62 CrossRefPubMed
36.
go back to reference S. Gill, S.K. Tasian, M. Ruella, O. Shestova, Y. Li, D.L. Porter, M. Carroll, G. Danet-Desnoyers, J. Scholler, S.A. Grupp, C.H. June, M. Kalos, Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells. Blood 123, 2343–2354 (2014). doi:10.1182/blood-2013-09-529537 CrossRefPubMedPubMedCentral S. Gill, S.K. Tasian, M. Ruella, O. Shestova, Y. Li, D.L. Porter, M. Carroll, G. Danet-Desnoyers, J. Scholler, S.A. Grupp, C.H. June, M. Kalos, Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells. Blood 123, 2343–2354 (2014). doi:10.​1182/​blood-2013-09-529537 CrossRefPubMedPubMedCentral
37.
go back to reference Z. Gadhoum, J. Delaunay, E. Maquarre, L. Durand, V. Lancereaux, J. Qi, J. Robert-Lezenes, C. Chomienne, F. Smadja-Joffe, The effect of anti-CD44 monoclonal antibodies on differentiation and proliferation of human acute myeloid leukemia cells. Leuk. Lymphoma 45, 1501–1510 (2004). doi:10.1080/1042819042000206687 CrossRefPubMed Z. Gadhoum, J. Delaunay, E. Maquarre, L. Durand, V. Lancereaux, J. Qi, J. Robert-Lezenes, C. Chomienne, F. Smadja-Joffe, The effect of anti-CD44 monoclonal antibodies on differentiation and proliferation of human acute myeloid leukemia cells. Leuk. Lymphoma 45, 1501–1510 (2004). doi:10.​1080/​1042819042000206​687 CrossRefPubMed
39.
40.
go back to reference J.L. Berkowitz, J.E. Janik, D.M. Stewart, E.S. Jaffe, M. Stetler-Stevenson, J.H. Shih, T.A. Fleisher, M. Turner, N.E. Urquhart, G.H. Wharfe, W.D. Figg, C.J. Peer, C.K. Goldman, T.A. Waldmann, J.C. Morris, Safety, efficacy, and pharmacokinetics/pharmacodynamics of daclizumab (anti-CD25) in patients with adult T-cell leukemia/lymphoma. Clin. Immunol. 155, 176–187 (2014). doi:10.1016/j.clim.2014.09.012 CrossRefPubMedPubMedCentral J.L. Berkowitz, J.E. Janik, D.M. Stewart, E.S. Jaffe, M. Stetler-Stevenson, J.H. Shih, T.A. Fleisher, M. Turner, N.E. Urquhart, G.H. Wharfe, W.D. Figg, C.J. Peer, C.K. Goldman, T.A. Waldmann, J.C. Morris, Safety, efficacy, and pharmacokinetics/pharmacodynamics of daclizumab (anti-CD25) in patients with adult T-cell leukemia/lymphoma. Clin. Immunol. 155, 176–187 (2014). doi:10.​1016/​j.​clim.​2014.​09.​012 CrossRefPubMedPubMedCentral
41.
go back to reference J. Cerny, H. Yu, M. Ramanathan, G.D. Raffel, W.V. Walsh, N. Fortier, L. Shanahan, E. O’Rourke, J. Bednarik, B. Barton, A. Kroll-Desrosiers, S. Hao, B. Woda, L. Hutchinson, A.M. Evens, A.G. Rosmarin, R. Nath, Expression of CD25 independently predicts early treatment failure of acute myeloid leukaemia (AML). Br. J. Haematol. 160, 262–266 (2013). doi:10.1111/bjh.12109 CrossRefPubMed J. Cerny, H. Yu, M. Ramanathan, G.D. Raffel, W.V. Walsh, N. Fortier, L. Shanahan, E. O’Rourke, J. Bednarik, B. Barton, A. Kroll-Desrosiers, S. Hao, B. Woda, L. Hutchinson, A.M. Evens, A.G. Rosmarin, R. Nath, Expression of CD25 independently predicts early treatment failure of acute myeloid leukaemia (AML). Br. J. Haematol. 160, 262–266 (2013). doi:10.​1111/​bjh.​12109 CrossRefPubMed
42.
go back to reference G.J.R. J. N. Allan, E. J. Feldman, J. M. Scandura, E. K. Ritchie, L. Lam, W. Xie, H-T Hsu, D. C. Hassane, M. L. Guzman, In 56th ASH Annual Meeting and Exposition, (San Francisco, CA) (2014) G.J.R. J. N. Allan, E. J. Feldman, J. M. Scandura, E. K. Ritchie, L. Lam, W. Xie, H-T Hsu, D. C. Hassane, M. L. Guzman, In 56th ASH Annual Meeting and Exposition, (San Francisco, CA) (2014)
43.
go back to reference Z. Guo, A. Wang, W. Zhang, M. Levit, Q. Gao, C. Barberis, M. Tabart, J. Zhang, D. Hoffmann, D. Wiederschain, J. Rocnik, F. Sun, J. Murtie, C. Lengauer, S. Gross, B. Zhang, H. Cheng, V. Patel, L. Schio, F. Adrian, M. Dorsch, C. Garcia-Echeverria, S.M. Huang, PIM inhibitors target CD25-positive AML cells through concomitant suppression of STAT5 activation and degradation of MYC oncogene. Blood 124, 1777–1789 (2014). doi:10.1182/blood-2014-01-551234 CrossRefPubMed Z. Guo, A. Wang, W. Zhang, M. Levit, Q. Gao, C. Barberis, M. Tabart, J. Zhang, D. Hoffmann, D. Wiederschain, J. Rocnik, F. Sun, J. Murtie, C. Lengauer, S. Gross, B. Zhang, H. Cheng, V. Patel, L. Schio, F. Adrian, M. Dorsch, C. Garcia-Echeverria, S.M. Huang, PIM inhibitors target CD25-positive AML cells through concomitant suppression of STAT5 activation and degradation of MYC oncogene. Blood 124, 1777–1789 (2014). doi:10.​1182/​blood-2014-01-551234 CrossRefPubMed
45.
go back to reference H. Lu, Q. Zhou, V. Deshmukh, H. Phull, J. Ma, V. Tardif, R.R. Naik, C. Bouvard, Y. Zhang, S. Choi, B.R. Lawson, S. Zhu, C.H. Kim, P.G. Schultz, Targeting human C-type lectin-like molecule-1 (CLL1) with a bispecific antibody for immunotherapy of acute myeloid leukemia. Angew. Chem. Int. Ed. Engl. 53, 9841–9845 (2014). doi:10.1002/anie.201405353 CrossRefPubMedPubMedCentral H. Lu, Q. Zhou, V. Deshmukh, H. Phull, J. Ma, V. Tardif, R.R. Naik, C. Bouvard, Y. Zhang, S. Choi, B.R. Lawson, S. Zhu, C.H. Kim, P.G. Schultz, Targeting human C-type lectin-like molecule-1 (CLL1) with a bispecific antibody for immunotherapy of acute myeloid leukemia. Angew. Chem. Int. Ed. Engl. 53, 9841–9845 (2014). doi:10.​1002/​anie.​201405353 CrossRefPubMedPubMedCentral
46.
go back to reference M. Askmyr, H. Agerstam, N. Hansen, S. Gordon, A. Arvanitakis, M. Rissler, G. Juliusson, J. Richter, M. Jaras, T. Fioretos, Selective killing of candidate AML stem cells by antibody targeting of IL1RAP. Blood 121, 3709–3713 (2013). doi:10.1182/blood-2012-09-458935 CrossRefPubMed M. Askmyr, H. Agerstam, N. Hansen, S. Gordon, A. Arvanitakis, M. Rissler, G. Juliusson, J. Richter, M. Jaras, T. Fioretos, Selective killing of candidate AML stem cells by antibody targeting of IL1RAP. Blood 121, 3709–3713 (2013). doi:10.​1182/​blood-2012-09-458935 CrossRefPubMed
47.
go back to reference D.E. Berardi, C. Flumian, P.B. Campodonico, A.J. Urtreger, M.I. Diaz Bessone, A.N. Motter, E.D. Bal de Kier Joffe, E.F. Farias, L.B. Todaro, Myoepithelial and luminal breast cancer cells exhibit different responses to all-trans retinoic acid. Cell. Oncol. 38, 289–305 (2015). doi:10.1007/s13402-015-0230-z CrossRef D.E. Berardi, C. Flumian, P.B. Campodonico, A.J. Urtreger, M.I. Diaz Bessone, A.N. Motter, E.D. Bal de Kier Joffe, E.F. Farias, L.B. Todaro, Myoepithelial and luminal breast cancer cells exhibit different responses to all-trans retinoic acid. Cell. Oncol. 38, 289–305 (2015). doi:10.​1007/​s13402-015-0230-z CrossRef
48.
go back to reference A. Kakizuka, W.H. Miller Jr., K. Umesono, R.P. Warrell Jr., S.R. Frankel, V.V. Murty, E. Dmitrovsky, R.M. Evans, Chromosomal translocation t(15;17) in human acute promyelocytic leukemia fuses RAR alpha with a novel putative transcription factor, PML. Cell 66, 663–674 (1991)CrossRefPubMed A. Kakizuka, W.H. Miller Jr., K. Umesono, R.P. Warrell Jr., S.R. Frankel, V.V. Murty, E. Dmitrovsky, R.M. Evans, Chromosomal translocation t(15;17) in human acute promyelocytic leukemia fuses RAR alpha with a novel putative transcription factor, PML. Cell 66, 663–674 (1991)CrossRefPubMed
49.
go back to reference P.V. Sanchez, S.T. Glantz, S. Scotland, M.T. Kasner, M. Carroll, Induced differentiation of acute myeloid leukemia cells by activation of retinoid X and liver X receptors. Leukemia 28, 749–760 (2014). doi:10.1038/leu.2013.202 CrossRefPubMed P.V. Sanchez, S.T. Glantz, S. Scotland, M.T. Kasner, M. Carroll, Induced differentiation of acute myeloid leukemia cells by activation of retinoid X and liver X receptors. Leukemia 28, 749–760 (2014). doi:10.​1038/​leu.​2013.​202 CrossRefPubMed
51.
go back to reference H. El Hajj, Z. Dassouki, C. Berthier, E. Raffoux, L. Ades, O. Legrand, R. Hleihel, U. Sahin, N. Tawil, A. Salameh, K. Zibara, N. Darwiche, M. Mohty, H. Dombret, P. Fenaux, H. de The, A. Bazarbachi, Retinoic acid and arsenic trioxide trigger degradation of mutated NPM1, resulting in apoptosis of AML cells. Blood 125, 3447–3454 (2015). doi:10.1182/blood-2014-11-612416 CrossRefPubMed H. El Hajj, Z. Dassouki, C. Berthier, E. Raffoux, L. Ades, O. Legrand, R. Hleihel, U. Sahin, N. Tawil, A. Salameh, K. Zibara, N. Darwiche, M. Mohty, H. Dombret, P. Fenaux, H. de The, A. Bazarbachi, Retinoic acid and arsenic trioxide trigger degradation of mutated NPM1, resulting in apoptosis of AML cells. Blood 125, 3447–3454 (2015). doi:10.​1182/​blood-2014-11-612416 CrossRefPubMed
52.
go back to reference X. Zheng, A. Seshire, B. Ruster, G. Bug, T. Beissert, E. Puccetti, D. Hoelzer, R. Henschler, M. Ruthardt, Arsenic but not all-trans retinoic acid overcomes the aberrant stem cell capacity of PML/RARalpha-positive leukemic stem cells. Haematologica 92, 323–331 (2007)CrossRefPubMed X. Zheng, A. Seshire, B. Ruster, G. Bug, T. Beissert, E. Puccetti, D. Hoelzer, R. Henschler, M. Ruthardt, Arsenic but not all-trans retinoic acid overcomes the aberrant stem cell capacity of PML/RARalpha-positive leukemic stem cells. Haematologica 92, 323–331 (2007)CrossRefPubMed
53.
go back to reference Y. Jiang, Z.H. Xue, W.Z. Shen, K.M. Du, H. Yan, Y. Yu, Z.G. Peng, M.G. Song, J.H. Tong, Z. Chen, Y. Huang, M. Lubbert, G.Q. Chen, Desferrioxamine induces leukemic cell differentiation potentially by hypoxia-inducible factor-1 alpha that augments transcriptional activity of CCAAT/enhancer-binding protein-alpha. Leukemia 19, 1239–1247 (2005). doi:10.1038/sj.leu.2403734 CrossRefPubMed Y. Jiang, Z.H. Xue, W.Z. Shen, K.M. Du, H. Yan, Y. Yu, Z.G. Peng, M.G. Song, J.H. Tong, Z. Chen, Y. Huang, M. Lubbert, G.Q. Chen, Desferrioxamine induces leukemic cell differentiation potentially by hypoxia-inducible factor-1 alpha that augments transcriptional activity of CCAAT/enhancer-binding protein-alpha. Leukemia 19, 1239–1247 (2005). doi:10.​1038/​sj.​leu.​2403734 CrossRefPubMed
54.
go back to reference M. Roth, B. Will, G. Simkin, S. Narayanagari, L. Barreyro, B. Bartholdy, R. Tamari, C.S. Mitsiades, A. Verma, U. Steidl, Eltrombopag inhibits the proliferation of leukemia cells via reduction of intracellular iron and induction of differentiation. Blood 120, 386–394 (2012). doi:10.1182/blood-2011-12-399667 CrossRefPubMedPubMedCentral M. Roth, B. Will, G. Simkin, S. Narayanagari, L. Barreyro, B. Bartholdy, R. Tamari, C.S. Mitsiades, A. Verma, U. Steidl, Eltrombopag inhibits the proliferation of leukemia cells via reduction of intracellular iron and induction of differentiation. Blood 120, 386–394 (2012). doi:10.​1182/​blood-2011-12-399667 CrossRefPubMedPubMedCentral
55.
go back to reference M. Szwed, A. Laroche-Clary, J. Robert, Z. Jozwiak, Efficacy of doxorubicin-transferrin conjugate in apoptosis induction in human leukemia cells through reactive oxygen species generation. Cell. Oncol. 39, 107–118 (2016). doi:10.1007/s13402-015-0256-2 CrossRef M. Szwed, A. Laroche-Clary, J. Robert, Z. Jozwiak, Efficacy of doxorubicin-transferrin conjugate in apoptosis induction in human leukemia cells through reactive oxygen species generation. Cell. Oncol. 39, 107–118 (2016). doi:10.​1007/​s13402-015-0256-2 CrossRef
56.
go back to reference J. Liu, X. Wei, Y. Wu, Y. Wang, Y. Qiu, J. Shi, H. Zhou, Z. Lu, M. Shao, L. Yu, L. Tong, Giganteaside D induces ROS-mediated apoptosis in human hepatocellular carcinoma cells through the MAPK pathway. Cell. Oncol. 39, 333–342 (2016). doi:10.1007/s13402-016-0273-9 CrossRef J. Liu, X. Wei, Y. Wu, Y. Wang, Y. Qiu, J. Shi, H. Zhou, Z. Lu, M. Shao, L. Yu, L. Tong, Giganteaside D induces ROS-mediated apoptosis in human hepatocellular carcinoma cells through the MAPK pathway. Cell. Oncol. 39, 333–342 (2016). doi:10.​1007/​s13402-016-0273-9 CrossRef
58.
go back to reference D.J. Goff, A. Court Recart, A. Sadarangani, H.J. Chun, C.L. Barrett, M. Krajewska, H. Leu, J. Low-Marchelli, W. Ma, A.Y. Shih, J. Wei, D. Zhai, I. Geron, M. Pu, L. Bao, R. Chuang, L. Balaian, J. Gotlib, M. Minden, G. Martinelli, J. Rusert, K.H. Dao, K. Shazand, P. Wentworth, K.M. Smith, C.A. Jamieson, S.R. Morris, K. Messer, L.S. Goldstein, T.J. Hudson, M. Marra, K.A. Frazer, M. Pellecchia, J.C. Reed, C.H. Jamieson, A pan-BCL2 inhibitor renders bone-marrow-resident human leukemia stem cells sensitive to tyrosine kinase inhibition. Cell Stem Cell 12, 316–328 (2013). doi:10.1016/j.stem.2012.12.011 CrossRefPubMedPubMedCentral D.J. Goff, A. Court Recart, A. Sadarangani, H.J. Chun, C.L. Barrett, M. Krajewska, H. Leu, J. Low-Marchelli, W. Ma, A.Y. Shih, J. Wei, D. Zhai, I. Geron, M. Pu, L. Bao, R. Chuang, L. Balaian, J. Gotlib, M. Minden, G. Martinelli, J. Rusert, K.H. Dao, K. Shazand, P. Wentworth, K.M. Smith, C.A. Jamieson, S.R. Morris, K. Messer, L.S. Goldstein, T.J. Hudson, M. Marra, K.A. Frazer, M. Pellecchia, J.C. Reed, C.H. Jamieson, A pan-BCL2 inhibitor renders bone-marrow-resident human leukemia stem cells sensitive to tyrosine kinase inhibition. Cell Stem Cell 12, 316–328 (2013). doi:10.​1016/​j.​stem.​2012.​12.​011 CrossRefPubMedPubMedCentral
59.
go back to reference A.S. Corbin, A. Agarwal, M. Loriaux, J. Cortes, M.W. Deininger, B.J. Druker, Human chronic myeloid leukemia stem cells are insensitive to imatinib despite inhibition of BCR-ABL activity. J. Clin. Invest. 121, 396–409 (2011). doi:10.1172/JCI35721 CrossRefPubMed A.S. Corbin, A. Agarwal, M. Loriaux, J. Cortes, M.W. Deininger, B.J. Druker, Human chronic myeloid leukemia stem cells are insensitive to imatinib despite inhibition of BCR-ABL activity. J. Clin. Invest. 121, 396–409 (2011). doi:10.​1172/​JCI35721 CrossRefPubMed
60.
go back to reference M. Konopleva, M. Milella, P. Ruvolo, J.C. Watts, M.R. Ricciardi, B. Korchin, T. McQueen, W. Bornmann, T. Tsao, P. Bergamo, D.H. Mak, W. Chen, J. McCubrey, A. Tafuri, M. Andreeff, MEK inhibition enhances ABT-737-induced leukemia cell apoptosis via prevention of ERK-activated MCL-1 induction and modulation of MCL-1/BIM complex. Leukemia 26, 778–787 (2012). doi:10.1038/leu.2011.287 CrossRefPubMed M. Konopleva, M. Milella, P. Ruvolo, J.C. Watts, M.R. Ricciardi, B. Korchin, T. McQueen, W. Bornmann, T. Tsao, P. Bergamo, D.H. Mak, W. Chen, J. McCubrey, A. Tafuri, M. Andreeff, MEK inhibition enhances ABT-737-induced leukemia cell apoptosis via prevention of ERK-activated MCL-1 induction and modulation of MCL-1/BIM complex. Leukemia 26, 778–787 (2012). doi:10.​1038/​leu.​2011.​287 CrossRefPubMed
61.
go back to reference M. Rahmani, M.M. Aust, E. Attkisson, D.C. Williams Jr., A. Ferreira-Gonzalez, S. Grant, Inhibition of Bcl-2 antiapoptotic members by obatoclax potently enhances sorafenib-induced apoptosis in human myeloid leukemia cells through a Bim-dependent process. Blood 119, 6089–6098 (2012). doi:10.1182/blood-2011-09-378141 CrossRefPubMedPubMedCentral M. Rahmani, M.M. Aust, E. Attkisson, D.C. Williams Jr., A. Ferreira-Gonzalez, S. Grant, Inhibition of Bcl-2 antiapoptotic members by obatoclax potently enhances sorafenib-induced apoptosis in human myeloid leukemia cells through a Bim-dependent process. Blood 119, 6089–6098 (2012). doi:10.​1182/​blood-2011-09-378141 CrossRefPubMedPubMedCentral
62.
go back to reference M. Skrtic, S. Sriskanthadevan, B. Jhas, M. Gebbia, X. Wang, Z. Wang, R. Hurren, Y. Jitkova, M. Gronda, N. Maclean, C.K. Lai, Y. Eberhard, J. Bartoszko, P. Spagnuolo, A.C. Rutledge, A. Datti, T. Ketela, J. Moffat, B.H. Robinson, J.H. Cameron, J. Wrana, C.J. Eaves, M.D. Minden, J.C. Wang, J.E. Dick, K. Humphries, C. Nislow, G. Giaever, A.D. Schimmer, Inhibition of mitochondrial translation as a therapeutic strategy for human acute myeloid leukemia. Cancer Cell 20, 674–688 (2011). doi:10.1016/j.ccr.2011.10.015 CrossRefPubMedPubMedCentral M. Skrtic, S. Sriskanthadevan, B. Jhas, M. Gebbia, X. Wang, Z. Wang, R. Hurren, Y. Jitkova, M. Gronda, N. Maclean, C.K. Lai, Y. Eberhard, J. Bartoszko, P. Spagnuolo, A.C. Rutledge, A. Datti, T. Ketela, J. Moffat, B.H. Robinson, J.H. Cameron, J. Wrana, C.J. Eaves, M.D. Minden, J.C. Wang, J.E. Dick, K. Humphries, C. Nislow, G. Giaever, A.D. Schimmer, Inhibition of mitochondrial translation as a therapeutic strategy for human acute myeloid leukemia. Cancer Cell 20, 674–688 (2011). doi:10.​1016/​j.​ccr.​2011.​10.​015 CrossRefPubMedPubMedCentral
64.
go back to reference K. Goetze, C.G. Fabian, A. Siebers, L. Binz, D. Faber, S. Indraccolo, G. Nardo, U.G. Sattler, W. Mueller-Klieser, Manipulation of tumor metabolism for therapeutic approaches: ovarian cancer-derived cell lines as a model system. Cell. Oncol. (Dordr) 38, 377–385 (2015). doi:10.1007/s13402-015-0237-5 CrossRef K. Goetze, C.G. Fabian, A. Siebers, L. Binz, D. Faber, S. Indraccolo, G. Nardo, U.G. Sattler, W. Mueller-Klieser, Manipulation of tumor metabolism for therapeutic approaches: ovarian cancer-derived cell lines as a model system. Cell. Oncol. (Dordr) 38, 377–385 (2015). doi:10.​1007/​s13402-015-0237-5 CrossRef
66.
go back to reference D. Anastasiou, Y. Yu, W.J. Israelsen, J.K. Jiang, M.B. Boxer, B.S. Hong, W. Tempel, S. Dimov, M. Shen, A. Jha, H. Yang, K.R. Mattaini, C.M. Metallo, B.P. Fiske, K.D. Courtney, S. Malstrom, T.M. Khan, C. Kung, A.P. Skoumbourdis, H. Veith, N. Southall, M.J. Walsh, K.R. Brimacombe, W. Leister, S.Y. Lunt, Z.R. Johnson, K.E. Yen, K. Kunii, S.M. Davidson, H.R. Christofk, C.P. Austin, J. Inglese, M.H. Harris, J.M. Asara, G. Stephanopoulos, F.G. Salituro, S. Jin, L. Dang, D.S. Auld, H.W. Park, L.C. Cantley, C.J. Thomas, M.G. Vander Heiden, Pyruvate kinase M2 activators promote tetramer formation and suppress tumorigenesis. Nat. Chem. Biol. 8, 839–847 (2012). doi:10.1038/nchembio.1060 CrossRefPubMedPubMedCentral D. Anastasiou, Y. Yu, W.J. Israelsen, J.K. Jiang, M.B. Boxer, B.S. Hong, W. Tempel, S. Dimov, M. Shen, A. Jha, H. Yang, K.R. Mattaini, C.M. Metallo, B.P. Fiske, K.D. Courtney, S. Malstrom, T.M. Khan, C. Kung, A.P. Skoumbourdis, H. Veith, N. Southall, M.J. Walsh, K.R. Brimacombe, W. Leister, S.Y. Lunt, Z.R. Johnson, K.E. Yen, K. Kunii, S.M. Davidson, H.R. Christofk, C.P. Austin, J. Inglese, M.H. Harris, J.M. Asara, G. Stephanopoulos, F.G. Salituro, S. Jin, L. Dang, D.S. Auld, H.W. Park, L.C. Cantley, C.J. Thomas, M.G. Vander Heiden, Pyruvate kinase M2 activators promote tetramer formation and suppress tumorigenesis. Nat. Chem. Biol. 8, 839–847 (2012). doi:10.​1038/​nchembio.​1060 CrossRefPubMedPubMedCentral
67.
go back to reference K.M. Parnell, J.M. Foulks, R.N. Nix, A. Clifford, J. Bullough, B. Luo, A. Senina, D. Vollmer, J. Liu, V. McCarthy, Y. Xu, M. Saunders, X.H. Liu, S. Pearce, K. Wright, M. O’Reilly, M.V. McCullar, K.K. Ho, S.B. Kanner, Pharmacologic activation of PKM2 slows lung tumor xenograft growth. Mol. Cancer Ther. 12, 1453–1460 (2013). doi:10.1158/1535-7163.MCT-13-0026 CrossRefPubMed K.M. Parnell, J.M. Foulks, R.N. Nix, A. Clifford, J. Bullough, B. Luo, A. Senina, D. Vollmer, J. Liu, V. McCarthy, Y. Xu, M. Saunders, X.H. Liu, S. Pearce, K. Wright, M. O’Reilly, M.V. McCullar, K.K. Ho, S.B. Kanner, Pharmacologic activation of PKM2 slows lung tumor xenograft growth. Mol. Cancer Ther. 12, 1453–1460 (2013). doi:10.​1158/​1535-7163.​MCT-13-0026 CrossRefPubMed
68.
go back to reference T. Hitosugi, S. Kang, M.G. Vander Heiden, T.W. Chung, S. Elf, K. Lythgoe, S. Dong, S. Lonial, X. Wang, G.Z. Chen, J. Xie, T.L. Gu, R.D. Polakiewicz, J.L. Roesel, T.J. Boggon, F.R. Khuri, D.G. Gilliland, L.C. Cantley, J. Kaufman, J. Chen, Tyrosine phosphorylation inhibits PKM2 to promote the Warburg effect and tumor growth. Sci. Signal. 2, ra73 (2009). doi:10.1126/scisignal.2000431 CrossRefPubMedPubMedCentral T. Hitosugi, S. Kang, M.G. Vander Heiden, T.W. Chung, S. Elf, K. Lythgoe, S. Dong, S. Lonial, X. Wang, G.Z. Chen, J. Xie, T.L. Gu, R.D. Polakiewicz, J.L. Roesel, T.J. Boggon, F.R. Khuri, D.G. Gilliland, L.C. Cantley, J. Kaufman, J. Chen, Tyrosine phosphorylation inhibits PKM2 to promote the Warburg effect and tumor growth. Sci. Signal. 2, ra73 (2009). doi:10.​1126/​scisignal.​2000431 CrossRefPubMedPubMedCentral
69.
go back to reference E.M. Sturgill, M.L. Guzman, In Proceedings of the 105th Annual Meeting of the American Association for Cancer Research, (San Diego, CA) (2014) E.M. Sturgill, M.L. Guzman, In Proceedings of the 105th Annual Meeting of the American Association for Cancer Research, (San Diego, CA) (2014)
72.
go back to reference E.D. Lagadinou, A. Sach, K. Callahan, R.M. Rossi, S.J. Neering, M. Minhajuddin, J.M. Ashton, S. Pei, V. Grose, K.M. O’Dwyer, J.L. Liesveld, P.S. Brookes, M.W. Becker, C.T. Jordan, BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell 12, 329–341 (2013). doi:10.1016/j.stem.2012.12.013 CrossRefPubMedPubMedCentral E.D. Lagadinou, A. Sach, K. Callahan, R.M. Rossi, S.J. Neering, M. Minhajuddin, J.M. Ashton, S. Pei, V. Grose, K.M. O’Dwyer, J.L. Liesveld, P.S. Brookes, M.W. Becker, C.T. Jordan, BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell 12, 329–341 (2013). doi:10.​1016/​j.​stem.​2012.​12.​013 CrossRefPubMedPubMedCentral
73.
go back to reference C. Craddock, L. Quek, N. Goardon, S. Freeman, S. Siddique, M. Raghavan, A. Aztberger, A. Schuh, D. Grimwade, A. Ivey, P. Virgo, R. Hills, T. McSkeane, J. Arrazi, S. Knapper, C. Brookes, B. Davies, A. Price, K. Wall, M. Griffiths, J. Cavenagh, R. Majeti, I. Weissman, A. Burnett, P. Vyas, Azacitidine fails to eradicate leukemic stem/progenitor cell populations in patients with acute myeloid leukemia and myelodysplasia. Leukemia 27, 1028–1036 (2013). doi:10.1038/leu.2012.312 CrossRefPubMed C. Craddock, L. Quek, N. Goardon, S. Freeman, S. Siddique, M. Raghavan, A. Aztberger, A. Schuh, D. Grimwade, A. Ivey, P. Virgo, R. Hills, T. McSkeane, J. Arrazi, S. Knapper, C. Brookes, B. Davies, A. Price, K. Wall, M. Griffiths, J. Cavenagh, R. Majeti, I. Weissman, A. Burnett, P. Vyas, Azacitidine fails to eradicate leukemic stem/progenitor cell populations in patients with acute myeloid leukemia and myelodysplasia. Leukemia 27, 1028–1036 (2013). doi:10.​1038/​leu.​2012.​312 CrossRefPubMed
75.
go back to reference P. Jo, K. Jung, M. Grade, L.C. Conradi, H.A. Wolff, J. Kitz, H. Becker, J. Ruschoff, A. Hartmann, T. Beissbarth, A. Muller-Dornieden, M. Ghadimi, R. Schneider-Stock, J. Gaedcke, CpG island methylator phenotype infers a poor disease-free survival in locally advanced rectal cancer. Surgery 151, 564–570 (2012). doi:10.1016/j.surg.2011.08.013 CrossRefPubMed P. Jo, K. Jung, M. Grade, L.C. Conradi, H.A. Wolff, J. Kitz, H. Becker, J. Ruschoff, A. Hartmann, T. Beissbarth, A. Muller-Dornieden, M. Ghadimi, R. Schneider-Stock, J. Gaedcke, CpG island methylator phenotype infers a poor disease-free survival in locally advanced rectal cancer. Surgery 151, 564–570 (2012). doi:10.​1016/​j.​surg.​2011.​08.​013 CrossRefPubMed
76.
77.
go back to reference T. Schenk, W.C. Chen, S. Gollner, L. Howell, L. Jin, K. Hebestreit, H.U. Klein, A.C. Popescu, A. Burnett, K. Mills, R.A. Casero Jr., L. Marton, P. Woster, M.D. Minden, M. Dugas, J.C. Wang, J.E. Dick, C. Muller-Tidow, K. Petrie, A. Zelent, Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat. Med. 18, 605–611 (2012). doi:10.1038/nm.2661 CrossRefPubMedPubMedCentral T. Schenk, W.C. Chen, S. Gollner, L. Howell, L. Jin, K. Hebestreit, H.U. Klein, A.C. Popescu, A. Burnett, K. Mills, R.A. Casero Jr., L. Marton, P. Woster, M.D. Minden, M. Dugas, J.C. Wang, J.E. Dick, C. Muller-Tidow, K. Petrie, A. Zelent, Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat. Med. 18, 605–611 (2012). doi:10.​1038/​nm.​2661 CrossRefPubMedPubMedCentral
78.
go back to reference S.R. Daigle, E.J. Olhava, C.A. Therkelsen, C.R. Majer, C.J. Sneeringer, J. Song, L.D. Johnston, M.P. Scott, J.J. Smith, Y. Xiao, L. Jin, K.W. Kuntz, R. Chesworth, M.P. Moyer, K.M. Bernt, J.C. Tseng, A.L. Kung, S.A. Armstrong, R.A. Copeland, V.M. Richon, R.M. Pollock, Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20, 53–65 (2011). doi:10.1016/j.ccr.2011.06.009 CrossRefPubMedPubMedCentral S.R. Daigle, E.J. Olhava, C.A. Therkelsen, C.R. Majer, C.J. Sneeringer, J. Song, L.D. Johnston, M.P. Scott, J.J. Smith, Y. Xiao, L. Jin, K.W. Kuntz, R. Chesworth, M.P. Moyer, K.M. Bernt, J.C. Tseng, A.L. Kung, S.A. Armstrong, R.A. Copeland, V.M. Richon, R.M. Pollock, Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20, 53–65 (2011). doi:10.​1016/​j.​ccr.​2011.​06.​009 CrossRefPubMedPubMedCentral
80.
go back to reference D.P. Mould, A.E. McGonagle, D.H. Wiseman, E.L. Williams, A.M. Jordan, Reversible inhibitors of LSD1 as therapeutic agents in acute myeloid leukemia: clinical significance and progress to date. Med. Res. Rev. 35, 586–618 (2014). doi:10.1002/med.21334 CrossRefPubMed D.P. Mould, A.E. McGonagle, D.H. Wiseman, E.L. Williams, A.M. Jordan, Reversible inhibitors of LSD1 as therapeutic agents in acute myeloid leukemia: clinical significance and progress to date. Med. Res. Rev. 35, 586–618 (2014). doi:10.​1002/​med.​21334 CrossRefPubMed
81.
go back to reference W. Fiskus, Y. Wang, A. Sreekumar, K.M. Buckley, H. Shi, A. Jillella, C. Ustun, R. Rao, P. Fernandez, J. Chen, R. Balusu, S. Koul, P. Atadja, V.E. Marquez, K.N. Bhalla, Combined epigenetic therapy with the histone methyltransferase EZH2 inhibitor 3-deazaneplanocin A and the histone deacetylase inhibitor panobinostat against human AML cells. Blood 114, 2733–2743 (2009). doi:10.1182/blood-2009-03-213496 CrossRefPubMedPubMedCentral W. Fiskus, Y. Wang, A. Sreekumar, K.M. Buckley, H. Shi, A. Jillella, C. Ustun, R. Rao, P. Fernandez, J. Chen, R. Balusu, S. Koul, P. Atadja, V.E. Marquez, K.N. Bhalla, Combined epigenetic therapy with the histone methyltransferase EZH2 inhibitor 3-deazaneplanocin A and the histone deacetylase inhibitor panobinostat against human AML cells. Blood 114, 2733–2743 (2009). doi:10.​1182/​blood-2009-03-213496 CrossRefPubMedPubMedCentral
82.
go back to reference J. Zhou, C. Bi, L.L. Cheong, S. Mahara, S.C. Liu, K.G. Tay, T.L. Koh, Q. Yu, W.J. Chng, The histone methyltransferase inhibitor, DZNep, up-regulates TXNIP, increases ROS production, and targets leukemia cells in AML. Blood 118, 2830–2839 (2011). doi:10.1182/blood-2010-07-294827 CrossRefPubMed J. Zhou, C. Bi, L.L. Cheong, S. Mahara, S.C. Liu, K.G. Tay, T.L. Koh, Q. Yu, W.J. Chng, The histone methyltransferase inhibitor, DZNep, up-regulates TXNIP, increases ROS production, and targets leukemia cells in AML. Blood 118, 2830–2839 (2011). doi:10.​1182/​blood-2010-07-294827 CrossRefPubMed
83.
go back to reference A. Chaturvedi, M.M. Araujo Cruz, N. Jyotsana, A. Sharma, H. Yun, K. Gorlich, M. Wichmann, A. Schwarzer, M. Preller, F. Thol, J. Meyer, R. Haemmerle, E.A. Struys, E.E. Jansen, U. Modlich, Z. Li, L.M. Sly, R. Geffers, R. Lindner, D.J. Manstein, U. Lehmann, J. Krauter, A. Ganser, M. Heuser, Mutant IDH1 promotes leukemogenesis in vivo and can be specifically targeted in human AML. Blood 122, 2877–2887 (2013). doi:10.1182/blood-2013-03-491571 CrossRefPubMed A. Chaturvedi, M.M. Araujo Cruz, N. Jyotsana, A. Sharma, H. Yun, K. Gorlich, M. Wichmann, A. Schwarzer, M. Preller, F. Thol, J. Meyer, R. Haemmerle, E.A. Struys, E.E. Jansen, U. Modlich, Z. Li, L.M. Sly, R. Geffers, R. Lindner, D.J. Manstein, U. Lehmann, J. Krauter, A. Ganser, M. Heuser, Mutant IDH1 promotes leukemogenesis in vivo and can be specifically targeted in human AML. Blood 122, 2877–2887 (2013). doi:10.​1182/​blood-2013-03-491571 CrossRefPubMed
84.
go back to reference M.E. Figueroa, S. Lugthart, Y. Li, C. Erpelinck-Verschueren, X. Deng, P.J. Christos, E. Schifano, J. Booth, W. van Putten, L. Skrabanek, F. Campagne, M. Mazumdar, J.M. Greally, P.J. Valk, B. Lowenberg, R. Delwel, A. Melnick, DNA methylation signatures identify biologically distinct subtypes in acute myeloid leukemia. Cancer Cell 17, 13–27 (2010). doi:10.1016/j.ccr.2009.11.020 CrossRefPubMedPubMedCentral M.E. Figueroa, S. Lugthart, Y. Li, C. Erpelinck-Verschueren, X. Deng, P.J. Christos, E. Schifano, J. Booth, W. van Putten, L. Skrabanek, F. Campagne, M. Mazumdar, J.M. Greally, P.J. Valk, B. Lowenberg, R. Delwel, A. Melnick, DNA methylation signatures identify biologically distinct subtypes in acute myeloid leukemia. Cancer Cell 17, 13–27 (2010). doi:10.​1016/​j.​ccr.​2009.​11.​020 CrossRefPubMedPubMedCentral
86.
go back to reference C. Frelin, V. Imbert, E. Griessinger, A.C. Peyron, N. Rochet, P. Philip, C. Dageville, A. Sirvent, M. Hummelsberger, E. Berard, M. Dreano, N. Sirvent, J.F. Peyron, Targeting NF-kappaB activation via pharmacologic inhibition of IKK2-induced apoptosis of human acute myeloid leukemia cells. Blood 105, 804–811 (2005). doi:10.1182/blood-2004-04-1463 CrossRefPubMed C. Frelin, V. Imbert, E. Griessinger, A.C. Peyron, N. Rochet, P. Philip, C. Dageville, A. Sirvent, M. Hummelsberger, E. Berard, M. Dreano, N. Sirvent, J.F. Peyron, Targeting NF-kappaB activation via pharmacologic inhibition of IKK2-induced apoptosis of human acute myeloid leukemia cells. Blood 105, 804–811 (2005). doi:10.​1182/​blood-2004-04-1463 CrossRefPubMed
87.
go back to reference K. Vazquez-Santillan, J. Melendez-Zajgla, L. Jimenez-Hernandez, G. Martinez-Ruiz, V. Maldonado, NF-kappaB signaling in cancer stem cells: a promising therapeutic target? Cell. Oncol. 38, 327–339 (2015). doi:10.1007/s13402-015-0236-6 CrossRef K. Vazquez-Santillan, J. Melendez-Zajgla, L. Jimenez-Hernandez, G. Martinez-Ruiz, V. Maldonado, NF-kappaB signaling in cancer stem cells: a promising therapeutic target? Cell. Oncol. 38, 327–339 (2015). doi:10.​1007/​s13402-015-0236-6 CrossRef
88.
go back to reference M.L. Guzman, S.J. Neering, D. Upchurch, B. Grimes, D.S. Howard, D.A. Rizzieri, S.M. Luger, C.T. Jordan, Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood 98, 2301–2307 (2001)CrossRefPubMed M.L. Guzman, S.J. Neering, D. Upchurch, B. Grimes, D.S. Howard, D.A. Rizzieri, S.M. Luger, C.T. Jordan, Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood 98, 2301–2307 (2001)CrossRefPubMed
89.
go back to reference Y. Kagoya, A. Yoshimi, K. Kataoka, M. Nakagawa, K. Kumano, S. Arai, H. Kobayashi, T. Saito, Y. Iwakura, M. Kurokawa, Positive feedback between NF-kappaB and TNF-alpha promotes leukemia-initiating cell capacity. J. Clin. Invest. 124, 528–542 (2014). doi:10.1172/JCI68101 CrossRefPubMedPubMedCentral Y. Kagoya, A. Yoshimi, K. Kataoka, M. Nakagawa, K. Kumano, S. Arai, H. Kobayashi, T. Saito, Y. Iwakura, M. Kurokawa, Positive feedback between NF-kappaB and TNF-alpha promotes leukemia-initiating cell capacity. J. Clin. Invest. 124, 528–542 (2014). doi:10.​1172/​JCI68101 CrossRefPubMedPubMedCentral
91.
go back to reference P. Diamanti, C.V. Cox, J.P. Moppett, A. Blair, Parthenolide eliminates leukemia-initiating cell populations and improves survival in xenografts of childhood acute lymphoblastic leukemia. Blood 121, 1384–1393 (2013). doi:10.1182/blood-2012-08-448852 CrossRefPubMed P. Diamanti, C.V. Cox, J.P. Moppett, A. Blair, Parthenolide eliminates leukemia-initiating cell populations and improves survival in xenografts of childhood acute lymphoblastic leukemia. Blood 121, 1384–1393 (2013). doi:10.​1182/​blood-2012-08-448852 CrossRefPubMed
92.
go back to reference M.L. Guzman, R.M. Rossi, S. Neelakantan, X. Li, C.A. Corbett, D.C. Hassane, M.W. Becker, J.M. Bennett, E. Sullivan, J.L. Lachowicz, A. Vaughan, C.J. Sweeney, W. Matthews, M. Carroll, J.L. Liesveld, P.A. Crooks, C.T. Jordan, An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood 110, 4427–4435 (2007). doi:10.1182/blood-2007-05-090621 CrossRefPubMedPubMedCentral M.L. Guzman, R.M. Rossi, S. Neelakantan, X. Li, C.A. Corbett, D.C. Hassane, M.W. Becker, J.M. Bennett, E. Sullivan, J.L. Lachowicz, A. Vaughan, C.J. Sweeney, W. Matthews, M. Carroll, J.L. Liesveld, P.A. Crooks, C.T. Jordan, An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood 110, 4427–4435 (2007). doi:10.​1182/​blood-2007-05-090621 CrossRefPubMedPubMedCentral
94.
go back to reference M.L. Guzman, N. Yang, K.K. Sharma, M. Balys, C.A. Corbett, C.T. Jordan, M.W. Becker, U. Steidl, O. Abdel-Wahab, R.L. Levine, G. Marcucci, G.J. Roboz, D.C. Hassane, Selective activity of the histone deacetylase inhibitor AR-42 against leukemia stem cells: a novel potential strategy in acute myelogenous leukemia. Mol. Cancer Ther. 13, 1979–1990 (2014). doi:10.1158/1535-7163.MCT-13-0963 CrossRefPubMedPubMedCentral M.L. Guzman, N. Yang, K.K. Sharma, M. Balys, C.A. Corbett, C.T. Jordan, M.W. Becker, U. Steidl, O. Abdel-Wahab, R.L. Levine, G. Marcucci, G.J. Roboz, D.C. Hassane, Selective activity of the histone deacetylase inhibitor AR-42 against leukemia stem cells: a novel potential strategy in acute myelogenous leukemia. Mol. Cancer Ther. 13, 1979–1990 (2014). doi:10.​1158/​1535-7163.​MCT-13-0963 CrossRefPubMedPubMedCentral
95.
go back to reference N. Lounnas, C. Frelin, N. Gonthier, P. Colosetti, A. Sirvent, J.P. Cassuto, F. Berthier, N. Sirvent, P. Rousselot, M. Dreano, J.F. Peyron, V. Imbert, NF-kappaB inhibition triggers death of imatinib-sensitive and imatinib-resistant chronic myeloid leukemia cells including T315I Bcr-Abl mutants. Int. J. Cancer 125, 308–317 (2009). doi:10.1002/ijc.24294 CrossRefPubMed N. Lounnas, C. Frelin, N. Gonthier, P. Colosetti, A. Sirvent, J.P. Cassuto, F. Berthier, N. Sirvent, P. Rousselot, M. Dreano, J.F. Peyron, V. Imbert, NF-kappaB inhibition triggers death of imatinib-sensitive and imatinib-resistant chronic myeloid leukemia cells including T315I Bcr-Abl mutants. Int. J. Cancer 125, 308–317 (2009). doi:10.​1002/​ijc.​24294 CrossRefPubMed
97.
go back to reference T. Skorski, P. Kanakaraj, M. Nieborowska-Skorska, M.Z. Ratajczak, S.C. Wen, G. Zon, A.M. Gewirtz, B. Perussia, B. Calabretta, Phosphatidylinositol-3 kinase activity is regulated by BCR/ABL and is required for the growth of Philadelphia chromosome-positive cells. Blood 86, 726–736 (1995)PubMed T. Skorski, P. Kanakaraj, M. Nieborowska-Skorska, M.Z. Ratajczak, S.C. Wen, G. Zon, A.M. Gewirtz, B. Perussia, B. Calabretta, Phosphatidylinositol-3 kinase activity is regulated by BCR/ABL and is required for the growth of Philadelphia chromosome-positive cells. Blood 86, 726–736 (1995)PubMed
98.
go back to reference C. Billottet, V.L. Grandage, R.E. Gale, A. Quattropani, C. Rommel, B. Vanhaesebroeck, A. Khwaja, A selective inhibitor of the p110delta isoform of PI 3-kinase inhibits AML cell proliferation and survival and increases the cytotoxic effects of VP16. Oncogene 25, 6648–6659 (2006). doi:10.1038/sj.onc.1209670 CrossRefPubMed C. Billottet, V.L. Grandage, R.E. Gale, A. Quattropani, C. Rommel, B. Vanhaesebroeck, A. Khwaja, A selective inhibitor of the p110delta isoform of PI 3-kinase inhibits AML cell proliferation and survival and increases the cytotoxic effects of VP16. Oncogene 25, 6648–6659 (2006). doi:10.​1038/​sj.​onc.​1209670 CrossRefPubMed
99.
go back to reference A.M. Martelli, C. Evangelisti, F. Chiarini, J.A. McCubrey, The phosphatidylinositol 3-kinase/Akt/mTOR signaling network as a therapeutic target in acute myelogenous leukemia patients. Oncotarget 1, 89–103 (2010)CrossRefPubMedPubMedCentral A.M. Martelli, C. Evangelisti, F. Chiarini, J.A. McCubrey, The phosphatidylinositol 3-kinase/Akt/mTOR signaling network as a therapeutic target in acute myelogenous leukemia patients. Oncotarget 1, 89–103 (2010)CrossRefPubMedPubMedCentral
100.
go back to reference P.L. Tazzari, A. Cappellini, F. Ricci, C. Evangelisti, V. Papa, T. Grafone, G. Martinelli, R. Conte, L. Cocco, J.A. McCubrey, A.M. Martelli, Multidrug resistance-associated protein 1 expression is under the control of the phosphoinositide 3 kinase/Akt signal transduction network in human acute myelogenous leukemia blasts. Leukemia 21, 427–438 (2007). doi:10.1038/sj.leu.2404523 CrossRefPubMed P.L. Tazzari, A. Cappellini, F. Ricci, C. Evangelisti, V. Papa, T. Grafone, G. Martinelli, R. Conte, L. Cocco, J.A. McCubrey, A.M. Martelli, Multidrug resistance-associated protein 1 expression is under the control of the phosphoinositide 3 kinase/Akt signal transduction network in human acute myelogenous leukemia blasts. Leukemia 21, 427–438 (2007). doi:10.​1038/​sj.​leu.​2404523 CrossRefPubMed
102.
go back to reference V. Papa, P.L. Tazzari, F. Chiarini, A. Cappellini, F. Ricci, A.M. Billi, C. Evangelisti, E. Ottaviani, G. Martinelli, N. Testoni, J.A. McCubrey, A.M. Martelli, Proapoptotic activity and chemosensitizing effect of the novel Akt inhibitor perifosine in acute myelogenous leukemia cells. Leukemia 22, 147–160 (2008). doi:10.1038/sj.leu.2404980 CrossRefPubMed V. Papa, P.L. Tazzari, F. Chiarini, A. Cappellini, F. Ricci, A.M. Billi, C. Evangelisti, E. Ottaviani, G. Martinelli, N. Testoni, J.A. McCubrey, A.M. Martelli, Proapoptotic activity and chemosensitizing effect of the novel Akt inhibitor perifosine in acute myelogenous leukemia cells. Leukemia 22, 147–160 (2008). doi:10.​1038/​sj.​leu.​2404980 CrossRefPubMed
104.
go back to reference C. Recher, O. Beyne-Rauzy, C. Demur, G. Chicanne, C. Dos Santos, V.M. Mas, D. Benzaquen, G. Laurent, F. Huguet, B. Payrastre, Antileukemic activity of rapamycin in acute myeloid leukemia. Blood 105, 2527–2534 (2005). doi:10.1182/blood-2004-06-2494 CrossRefPubMed C. Recher, O. Beyne-Rauzy, C. Demur, G. Chicanne, C. Dos Santos, V.M. Mas, D. Benzaquen, G. Laurent, F. Huguet, B. Payrastre, Antileukemic activity of rapamycin in acute myeloid leukemia. Blood 105, 2527–2534 (2005). doi:10.​1182/​blood-2004-06-2494 CrossRefPubMed
106.
go back to reference F. Chiarini, A. Lonetti, G. Teti, E. Orsini, D. Bressanin, A. Cappellini, F. Ricci, P.L. Tazzari, A. Ognibene, M. Falconi, P. Pagliaro, I. Iacobucci, G. Martinelli, S. Amadori, J.A. McCubrey, A.M. Martelli, A combination of temsirolimus, an allosteric mTOR inhibitor, with clofarabine as a new therapeutic option for patients with acute myeloid leukemia. Oncotarget 3, 1615–1628 (2012)CrossRefPubMedPubMedCentral F. Chiarini, A. Lonetti, G. Teti, E. Orsini, D. Bressanin, A. Cappellini, F. Ricci, P.L. Tazzari, A. Ognibene, M. Falconi, P. Pagliaro, I. Iacobucci, G. Martinelli, S. Amadori, J.A. McCubrey, A.M. Martelli, A combination of temsirolimus, an allosteric mTOR inhibitor, with clofarabine as a new therapeutic option for patients with acute myeloid leukemia. Oncotarget 3, 1615–1628 (2012)CrossRefPubMedPubMedCentral
107.
go back to reference D.C. Hassane, S. Sen, M. Minhajuddin, R.M. Rossi, C.A. Corbett, M. Balys, L. Wei, P.A. Crooks, M.L. Guzman, C.T. Jordan, Chemical genomic screening reveals synergism between parthenolide and inhibitors of the PI-3 kinase and mTOR pathways. Blood 116, 5983–5990 (2010). doi:10.1182/blood-2010-04-278044 CrossRefPubMed D.C. Hassane, S. Sen, M. Minhajuddin, R.M. Rossi, C.A. Corbett, M. Balys, L. Wei, P.A. Crooks, M.L. Guzman, C.T. Jordan, Chemical genomic screening reveals synergism between parthenolide and inhibitors of the PI-3 kinase and mTOR pathways. Blood 116, 5983–5990 (2010). doi:10.​1182/​blood-2010-04-278044 CrossRefPubMed
108.
go back to reference K. Meja, C. Stengel, R. Sellar, D. Huszar, B.R. Davies, R.E. Gale, D.C. Linch, A. Khwaja, PIM and AKT kinase inhibitors show synergistic cytotoxicity in acute myeloid leukaemia that is associated with convergence on mTOR and MCL1 pathways. Br. J. Haematol. 167, 69–79 (2014). doi:10.1111/bjh.13013 CrossRefPubMed K. Meja, C. Stengel, R. Sellar, D. Huszar, B.R. Davies, R.E. Gale, D.C. Linch, A. Khwaja, PIM and AKT kinase inhibitors show synergistic cytotoxicity in acute myeloid leukaemia that is associated with convergence on mTOR and MCL1 pathways. Br. J. Haematol. 167, 69–79 (2014). doi:10.​1111/​bjh.​13013 CrossRefPubMed
109.
go back to reference F. Pellicano, M.T. Scott, G.V. Helgason, L.E. Hopcroft, E.K. Allan, M. Aspinall-O’Dea, M. Copland, A. Pierce, B.J. Huntly, A.D. Whetton, T.L. Holyoake, The antiproliferative activity of kinase inhibitors in chronic myeloid leukemia cells is mediated by FOXO transcription factors. Stem Cells 32, 2324–2337 (2014). doi:10.1002/stem.1748 CrossRefPubMedPubMedCentral F. Pellicano, M.T. Scott, G.V. Helgason, L.E. Hopcroft, E.K. Allan, M. Aspinall-O’Dea, M. Copland, A. Pierce, B.J. Huntly, A.D. Whetton, T.L. Holyoake, The antiproliferative activity of kinase inhibitors in chronic myeloid leukemia cells is mediated by FOXO transcription factors. Stem Cells 32, 2324–2337 (2014). doi:10.​1002/​stem.​1748 CrossRefPubMedPubMedCentral
110.
go back to reference N. Chapuis, J. Tamburini, A.S. Green, C. Vignon, V. Bardet, A. Neyret, M. Pannetier, L. Willems, S. Park, A. Macone, S.M. Maira, N. Ifrah, F. Dreyfus, O. Herault, C. Lacombe, P. Mayeux, D. Bouscary, Dual inhibition of PI3K and mTORC1/2 signaling by NVP-BEZ235 as a new therapeutic strategy for acute myeloid leukemia. Clin. Cancer Res. 16, 5424–5435 (2010). doi:10.1158/1078-0432.CCR-10-1102 CrossRefPubMed N. Chapuis, J. Tamburini, A.S. Green, C. Vignon, V. Bardet, A. Neyret, M. Pannetier, L. Willems, S. Park, A. Macone, S.M. Maira, N. Ifrah, F. Dreyfus, O. Herault, C. Lacombe, P. Mayeux, D. Bouscary, Dual inhibition of PI3K and mTORC1/2 signaling by NVP-BEZ235 as a new therapeutic strategy for acute myeloid leukemia. Clin. Cancer Res. 16, 5424–5435 (2010). doi:10.​1158/​1078-0432.​CCR-10-1102 CrossRefPubMed
111.
go back to reference X. Ding, F. Li, J. McKnight, C. Schmidt, K. Strooisma, H. Shimizu, K. Faber, J.A. Ware, B. Dean, A supported liquid extraction-LC-MS/MS method for determination of GDC-0980 (Apitolisib), a dual small-molecule inhibitor of class 1A phosphoinositide 3-kinase and mammalian target of rapamycin, in human plasma. J. Pharm. Biomed. Anal. 100, 150–156 (2014). doi:10.1016/j.jpba.2014.08.001 CrossRefPubMed X. Ding, F. Li, J. McKnight, C. Schmidt, K. Strooisma, H. Shimizu, K. Faber, J.A. Ware, B. Dean, A supported liquid extraction-LC-MS/MS method for determination of GDC-0980 (Apitolisib), a dual small-molecule inhibitor of class 1A phosphoinositide 3-kinase and mammalian target of rapamycin, in human plasma. J. Pharm. Biomed. Anal. 100, 150–156 (2014). doi:10.​1016/​j.​jpba.​2014.​08.​001 CrossRefPubMed
112.
go back to reference P. Yu, A.D. Laird, X. Du, J. Wu, K.A. Won, K. Yamaguchi, P.P. Hsu, F. Qian, C.T. Jaeger, W. Zhang, C.A. Buhr, P. Shen, W. Abulafia, J. Chen, J. Young, A. Plonowski, F.M. Yakes, F. Chu, M. Lee, F. Bentzien, S.T. Lam, S. Dale, D.J. Matthews, P. Lamb, P. Foster, Characterization of the activity of the PI3K/mTOR inhibitor XL765 (SAR245409) in tumor models with diverse genetic alterations affecting the PI3K pathway. Mol. Cancer Ther. 13, 1078–1091 (2014). doi:10.1158/1535-7163.MCT-13-0709 CrossRefPubMed P. Yu, A.D. Laird, X. Du, J. Wu, K.A. Won, K. Yamaguchi, P.P. Hsu, F. Qian, C.T. Jaeger, W. Zhang, C.A. Buhr, P. Shen, W. Abulafia, J. Chen, J. Young, A. Plonowski, F.M. Yakes, F. Chu, M. Lee, F. Bentzien, S.T. Lam, S. Dale, D.J. Matthews, P. Lamb, P. Foster, Characterization of the activity of the PI3K/mTOR inhibitor XL765 (SAR245409) in tumor models with diverse genetic alterations affecting the PI3K pathway. Mol. Cancer Ther. 13, 1078–1091 (2014). doi:10.​1158/​1535-7163.​MCT-13-0709 CrossRefPubMed
114.
go back to reference K. Ito, A. Hirao, F. Arai, K. Takubo, S. Matsuoka, K. Miyamoto, M. Ohmura, K. Naka, K. Hosokawa, Y. Ikeda, T. Suda, Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat. Med. 12, 446–451 (2006). doi:10.1038/nm1388 CrossRefPubMed K. Ito, A. Hirao, F. Arai, K. Takubo, S. Matsuoka, K. Miyamoto, M. Ohmura, K. Naka, K. Hosokawa, Y. Ikeda, T. Suda, Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat. Med. 12, 446–451 (2006). doi:10.​1038/​nm1388 CrossRefPubMed
116.
go back to reference M. Nieborowska-Skorska, P.K. Kopinski, R. Ray, G. Hoser, D. Ngaba, S. Flis, K. Cramer, M.M. Reddy, M. Koptyra, T. Penserga, E. Glodkowska-Mrowka, E. Bolton, T.L. Holyoake, C.J. Eaves, S. Cerny-Reiterer, P. Valent, A. Hochhaus, T.P. Hughes, H. van der Kuip, M. Sattler, W. Wiktor-Jedrzejczak, C. Richardson, A. Dorrance, T. Stoklosa, D.A. Williams, T. Skorski, Rac2-MRC-cIII-generated ROS cause genomic instability in chronic myeloid leukemia stem cells and primitive progenitors. Blood 119, 4253–4263 (2012). doi:10.1182/blood-2011-10-385658 CrossRefPubMedPubMedCentral M. Nieborowska-Skorska, P.K. Kopinski, R. Ray, G. Hoser, D. Ngaba, S. Flis, K. Cramer, M.M. Reddy, M. Koptyra, T. Penserga, E. Glodkowska-Mrowka, E. Bolton, T.L. Holyoake, C.J. Eaves, S. Cerny-Reiterer, P. Valent, A. Hochhaus, T.P. Hughes, H. van der Kuip, M. Sattler, W. Wiktor-Jedrzejczak, C. Richardson, A. Dorrance, T. Stoklosa, D.A. Williams, T. Skorski, Rac2-MRC-cIII-generated ROS cause genomic instability in chronic myeloid leukemia stem cells and primitive progenitors. Blood 119, 4253–4263 (2012). doi:10.​1182/​blood-2011-10-385658 CrossRefPubMedPubMedCentral
117.
go back to reference S. Pei, M. Minhajuddin, K.P. Callahan, M. Balys, J.M. Ashton, S.J. Neering, E.D. Lagadinou, C. Corbett, H. Ye, J.L. Liesveld, K.M. O’Dwyer, Z. Li, L. Shi, P. Greninger, J. Settleman, C. Benes, F.K. Hagen, J. Munger, P.A. Crooks, M.W. Becker, C.T. Jordan, Targeting aberrant glutathione metabolism to eradicate human acute myelogenous leukemia cells. J. Biol. Chem. 288, 33542–33558 (2013). doi:10.1074/jbc.M113.511170 CrossRefPubMedPubMedCentral S. Pei, M. Minhajuddin, K.P. Callahan, M. Balys, J.M. Ashton, S.J. Neering, E.D. Lagadinou, C. Corbett, H. Ye, J.L. Liesveld, K.M. O’Dwyer, Z. Li, L. Shi, P. Greninger, J. Settleman, C. Benes, F.K. Hagen, J. Munger, P.A. Crooks, M.W. Becker, C.T. Jordan, Targeting aberrant glutathione metabolism to eradicate human acute myelogenous leukemia cells. J. Biol. Chem. 288, 33542–33558 (2013). doi:10.​1074/​jbc.​M113.​511170 CrossRefPubMedPubMedCentral
118.
go back to reference X. Chen, X. Shi, C. Zhao, X. Li, X. Lan, S. Liu, H. Huang, N. Liu, S. Liao, D. Zang, W. Song, Q. Liu, B.Z. Carter, Q.P. Dou, X. Wang, J. Liu, Anti-rheumatic agent auranofin induced apoptosis in chronic myeloid leukemia cells resistant to imatinib through both Bcr/Abl-dependent and -independent mechanisms. Oncotarget 5, 9118–9132 (2014)CrossRefPubMedPubMedCentral X. Chen, X. Shi, C. Zhao, X. Li, X. Lan, S. Liu, H. Huang, N. Liu, S. Liao, D. Zang, W. Song, Q. Liu, B.Z. Carter, Q.P. Dou, X. Wang, J. Liu, Anti-rheumatic agent auranofin induced apoptosis in chronic myeloid leukemia cells resistant to imatinib through both Bcr/Abl-dependent and -independent mechanisms. Oncotarget 5, 9118–9132 (2014)CrossRefPubMedPubMedCentral
120.
go back to reference K. Moulick, J.H. Ahn, H. Zong, A. Rodina, L. Cerchietti, E.M. Gomes DaGama, E. Caldas-Lopes, K. Beebe, F. Perna, K. Hatzi, L.P. Vu, X. Zhao, D. Zatorska, T. Taldone, P. Smith-Jones, M. Alpaugh, S.S. Gross, N. Pillarsetty, T. Ku, J.S. Lewis, S.M. Larson, R. Levine, H. Erdjument-Bromage, M.L. Guzman, S.D. Nimer, A. Melnick, L. Neckers, G. Chiosis, Affinity-based proteomics reveal cancer-specific networks coordinated by Hsp90. Nat. Chem. Biol. 7, 818–826 (2011). doi:10.1038/nchembio.670 CrossRefPubMedPubMedCentral K. Moulick, J.H. Ahn, H. Zong, A. Rodina, L. Cerchietti, E.M. Gomes DaGama, E. Caldas-Lopes, K. Beebe, F. Perna, K. Hatzi, L.P. Vu, X. Zhao, D. Zatorska, T. Taldone, P. Smith-Jones, M. Alpaugh, S.S. Gross, N. Pillarsetty, T. Ku, J.S. Lewis, S.M. Larson, R. Levine, H. Erdjument-Bromage, M.L. Guzman, S.D. Nimer, A. Melnick, L. Neckers, G. Chiosis, Affinity-based proteomics reveal cancer-specific networks coordinated by Hsp90. Nat. Chem. Biol. 7, 818–826 (2011). doi:10.​1038/​nchembio.​670 CrossRefPubMedPubMedCentral
122.
go back to reference H. Reikvam, K.J. Hatfield, E. Ersvaer, R. Hovland, J. Skavland, B.T. Gjertsen, K. Petersen, O. Bruserud, Expression profile of heat shock proteins in acute myeloid leukaemia patients reveals a distinct signature strongly associated with FLT3 mutation status--consequences and potentials for pharmacological intervention. Br. J. Haematol. 156, 468–480 (2012). doi:10.1111/j.1365-2141.2011.08960.x CrossRefPubMed H. Reikvam, K.J. Hatfield, E. Ersvaer, R. Hovland, J. Skavland, B.T. Gjertsen, K. Petersen, O. Bruserud, Expression profile of heat shock proteins in acute myeloid leukaemia patients reveals a distinct signature strongly associated with FLT3 mutation status--consequences and potentials for pharmacological intervention. Br. J. Haematol. 156, 468–480 (2012). doi:10.​1111/​j.​1365-2141.​2011.​08960.​x CrossRefPubMed
124.
go back to reference W. Yu, Q. Rao, M. Wang, Z. Tian, D. Lin, X. Liu, J. Wang, The Hsp90 inhibitor 17-allylamide-17-demethoxygeldanamycin induces apoptosis and differentiation of Kasumi-1 harboring the Asn822Lys KIT mutation and down-regulates KIT protein level. Leuk. Res. 30, 575–582 (2006). doi:10.1016/j.leukres.2005.08.028 W. Yu, Q. Rao, M. Wang, Z. Tian, D. Lin, X. Liu, J. Wang, The Hsp90 inhibitor 17-allylamide-17-demethoxygeldanamycin induces apoptosis and differentiation of Kasumi-1 harboring the Asn822Lys KIT mutation and down-regulates KIT protein level. Leuk. Res. 30, 575–582 (2006). doi:10.​1016/​j.​leukres.​2005.​08.​028
125.
go back to reference J.E. Lancet, I. Gojo, M. Burton, M. Quinn, S.M. Tighe, K. Kersey, Z. Zhong, M.X. Albitar, K. Bhalla, A.L. Hannah, M.R. Baer, Phase I study of the heat shock protein 90 inhibitor alvespimycin (KOS-1022, 17-DMAG) administered intravenously twice weekly to patients with acute myeloid leukemia. Leukemia 24, 699–705 (2010). doi:10.1038/leu.2009.292 CrossRefPubMed J.E. Lancet, I. Gojo, M. Burton, M. Quinn, S.M. Tighe, K. Kersey, Z. Zhong, M.X. Albitar, K. Bhalla, A.L. Hannah, M.R. Baer, Phase I study of the heat shock protein 90 inhibitor alvespimycin (KOS-1022, 17-DMAG) administered intravenously twice weekly to patients with acute myeloid leukemia. Leukemia 24, 699–705 (2010). doi:10.​1038/​leu.​2009.​292 CrossRefPubMed
126.
go back to reference C. Gallerne, A. Prola, C. Lemaire, Hsp90 inhibition by PU-H71 induces apoptosis through endoplasmic reticulum stress and mitochondrial pathway in cancer cells and overcomes the resistance conferred by Bcl-2. Biochim. Biophys. Acta 1833, 1356–1366 (2013). doi:10.1016/j.bbamcr.2013.02.014 CrossRefPubMed C. Gallerne, A. Prola, C. Lemaire, Hsp90 inhibition by PU-H71 induces apoptosis through endoplasmic reticulum stress and mitochondrial pathway in cancer cells and overcomes the resistance conferred by Bcl-2. Biochim. Biophys. Acta 1833, 1356–1366 (2013). doi:10.​1016/​j.​bbamcr.​2013.​02.​014 CrossRefPubMed
127.
go back to reference S.R. Ambati, E.C. Lopes, K. Kosugi, U. Mony, A. Zehir, S.K. Shah, T. Taldone, A.L. Moreira, P.A. Meyers, G. Chiosis, M.A. Moore, Pre-clinical efficacy of PU-H71, a novel HSP90 inhibitor, alone and in combination with bortezomib in Ewing sarcoma. Mol. Oncol. 8, 323–336 (2014). doi:10.1016/j.molonc.2013.12.005 CrossRefPubMed S.R. Ambati, E.C. Lopes, K. Kosugi, U. Mony, A. Zehir, S.K. Shah, T. Taldone, A.L. Moreira, P.A. Meyers, G. Chiosis, M.A. Moore, Pre-clinical efficacy of PU-H71, a novel HSP90 inhibitor, alone and in combination with bortezomib in Ewing sarcoma. Mol. Oncol. 8, 323–336 (2014). doi:10.​1016/​j.​molonc.​2013.​12.​005 CrossRefPubMed
128.
go back to reference H. Zong, A. Gozman, E. Caldas-Lopes, T. Taldone, E. Sturgill, S. Brennan, S.O. Ochiana, E.M. Gomes-DaGama, S. Sen, A. Rodina, J. Koren 3rd, M.W. Becker, C.M. Rudin, A. Melnick, R.L. Levine, G.J. Roboz, S.D. Nimer, G. Chiosis, M.L. Guzman, A hyperactive signalosome in acute myeloid leukemia drives addiction to a tumor-specific Hsp90 species. Cell Rep. 13, 2159–2173 (2015). doi:10.1016/j.celrep.2015.10.073 CrossRefPubMedPubMedCentral H. Zong, A. Gozman, E. Caldas-Lopes, T. Taldone, E. Sturgill, S. Brennan, S.O. Ochiana, E.M. Gomes-DaGama, S. Sen, A. Rodina, J. Koren 3rd, M.W. Becker, C.M. Rudin, A. Melnick, R.L. Levine, G.J. Roboz, S.D. Nimer, G. Chiosis, M.L. Guzman, A hyperactive signalosome in acute myeloid leukemia drives addiction to a tumor-specific Hsp90 species. Cell Rep. 13, 2159–2173 (2015). doi:10.​1016/​j.​celrep.​2015.​10.​073 CrossRefPubMedPubMedCentral
129.
go back to reference C. Hurtz, K. Hatzi, L. Cerchietti, M. Braig, E. Park, Y.M. Kim, S. Herzog, P. Ramezani-Rad, H. Jumaa, M.C. Muller, W.K. Hofmann, A. Hochhaus, B.H. Ye, A. Agarwal, B.J. Druker, N.P. Shah, A.M. Melnick, M. Muschen, BCL6-mediated repression of p53 is critical for leukemia stem cell survival in chronic myeloid leukemia. J. Exp. Med. 208, 2163–2174 (2011). doi:10.1084/jem.20110304 CrossRefPubMedPubMedCentral C. Hurtz, K. Hatzi, L. Cerchietti, M. Braig, E. Park, Y.M. Kim, S. Herzog, P. Ramezani-Rad, H. Jumaa, M.C. Muller, W.K. Hofmann, A. Hochhaus, B.H. Ye, A. Agarwal, B.J. Druker, N.P. Shah, A.M. Melnick, M. Muschen, BCL6-mediated repression of p53 is critical for leukemia stem cell survival in chronic myeloid leukemia. J. Exp. Med. 208, 2163–2174 (2011). doi:10.​1084/​jem.​20110304 CrossRefPubMedPubMedCentral
130.
go back to reference J.M. Polo, T. Dell’Oso, S.M. Ranuncolo, L. Cerchietti, D. Beck, G.F. Da Silva, G.G. Prive, J.D. Licht, A. Melnick, Specific peptide interference reveals BCL6 transcriptional and oncogenic mechanisms in B-cell lymphoma cells. Nat. Med. 10, 1329–1335 (2004). doi:10.1038/nm1134 CrossRefPubMed J.M. Polo, T. Dell’Oso, S.M. Ranuncolo, L. Cerchietti, D. Beck, G.F. Da Silva, G.G. Prive, J.D. Licht, A. Melnick, Specific peptide interference reveals BCL6 transcriptional and oncogenic mechanisms in B-cell lymphoma cells. Nat. Med. 10, 1329–1335 (2004). doi:10.​1038/​nm1134 CrossRefPubMed
131.
go back to reference C. Duy, J.J. Yu, R. Nahar, S. Swaminathan, S.M. Kweon, J.M. Polo, E. Valls, L. Klemm, S. Shojaee, L. Cerchietti, W. Schuh, H.M. Jack, C. Hurtz, P. Ramezani-Rad, S. Herzog, H. Jumaa, H.P. Koeffler, I.M. de Alboran, A.M. Melnick, B.H. Ye, M. Muschen, BCL6 is critical for the development of a diverse primary B cell repertoire. J. Exp. Med. 207, 1209–1221 (2010). doi:10.1084/jem.20091299 CrossRefPubMedPubMedCentral C. Duy, J.J. Yu, R. Nahar, S. Swaminathan, S.M. Kweon, J.M. Polo, E. Valls, L. Klemm, S. Shojaee, L. Cerchietti, W. Schuh, H.M. Jack, C. Hurtz, P. Ramezani-Rad, S. Herzog, H. Jumaa, H.P. Koeffler, I.M. de Alboran, A.M. Melnick, B.H. Ye, M. Muschen, BCL6 is critical for the development of a diverse primary B cell repertoire. J. Exp. Med. 207, 1209–1221 (2010). doi:10.​1084/​jem.​20091299 CrossRefPubMedPubMedCentral
133.
go back to reference A.E. Abrahamsson, I. Geron, J. Gotlib, K.H. Dao, C.F. Barroga, I.G. Newton, F.J. Giles, J. Durocher, R.S. Creusot, M. Karimi, C. Jones, J.L. Zehnder, A. Keating, R.S. Negrin, I.L. Weissman, C.H. Jamieson, Glycogen synthase kinase 3beta missplicing contributes to leukemia stem cell generation. Proc. Natl. Acad. Sci. U. S. A. 106, 3925–3929 (2009). doi:10.1073/pnas.0900189106 CrossRefPubMedPubMedCentral A.E. Abrahamsson, I. Geron, J. Gotlib, K.H. Dao, C.F. Barroga, I.G. Newton, F.J. Giles, J. Durocher, R.S. Creusot, M. Karimi, C. Jones, J.L. Zehnder, A. Keating, R.S. Negrin, I.L. Weissman, C.H. Jamieson, Glycogen synthase kinase 3beta missplicing contributes to leukemia stem cell generation. Proc. Natl. Acad. Sci. U. S. A. 106, 3925–3929 (2009). doi:10.​1073/​pnas.​0900189106 CrossRefPubMedPubMedCentral
135.
go back to reference J.P. Radich, H. Dai, M. Mao, V. Oehler, J. Schelter, B. Druker, C. Sawyers, N. Shah, W. Stock, C.L. Willman, S. Friend, P.S. Linsley, Gene expression changes associated with progression and response in chronic myeloid leukemia. Proc. Natl. Acad. Sci. U. S. A. 103, 2794–2799 (2006). doi:10.1073/pnas.0510423103 CrossRefPubMedPubMedCentral J.P. Radich, H. Dai, M. Mao, V. Oehler, J. Schelter, B. Druker, C. Sawyers, N. Shah, W. Stock, C.L. Willman, S. Friend, P.S. Linsley, Gene expression changes associated with progression and response in chronic myeloid leukemia. Proc. Natl. Acad. Sci. U. S. A. 103, 2794–2799 (2006). doi:10.​1073/​pnas.​0510423103 CrossRefPubMedPubMedCentral
136.
140.
go back to reference S. Song, T. Christova, S. Perusini, S. Alizadeh, R.Y. Bao, B.W. Miller, R. Hurren, Y. Jitkova, M. Gronda, M. Isaac, B. Joseph, R. Subramaniam, A. Aman, A. Chau, D.E. Hogge, S.J. Weir, J. Kasper, A.D. Schimmer, R. Al-awar, J.L. Wrana, L. Attisano, Wnt inhibitor screen reveals iron dependence of beta-catenin signaling in cancers. Cancer Res. 71, 7628–7639 (2011). doi:10.1158/0008-5472.CAN-11-2745 CrossRefPubMed S. Song, T. Christova, S. Perusini, S. Alizadeh, R.Y. Bao, B.W. Miller, R. Hurren, Y. Jitkova, M. Gronda, M. Isaac, B. Joseph, R. Subramaniam, A. Aman, A. Chau, D.E. Hogge, S.J. Weir, J. Kasper, A.D. Schimmer, R. Al-awar, J.L. Wrana, L. Attisano, Wnt inhibitor screen reveals iron dependence of beta-catenin signaling in cancers. Cancer Res. 71, 7628–7639 (2011). doi:10.​1158/​0008-5472.​CAN-11-2745 CrossRefPubMed
142.
143.
go back to reference F. Damm, B. Markus, F. Thol, M. Morgan, G. Gohring, B. Schlegelberger, J. Krauter, M. Heuser, O.A. Bernard, A. Ganser, TET2 mutations in cytogenetically normal acute myeloid leukemia: clinical implications and evolutionary patterns. Genes Chromosomes Cancer 53, 824–832 (2014). doi:10.1002/gcc.22191 CrossRefPubMed F. Damm, B. Markus, F. Thol, M. Morgan, G. Gohring, B. Schlegelberger, J. Krauter, M. Heuser, O.A. Bernard, A. Ganser, TET2 mutations in cytogenetically normal acute myeloid leukemia: clinical implications and evolutionary patterns. Genes Chromosomes Cancer 53, 824–832 (2014). doi:10.​1002/​gcc.​22191 CrossRefPubMed
144.
go back to reference C. Lobry, P. Ntziachristos, D. Ndiaye-Lobry, P. Oh, L. Cimmino, N. Zhu, E. Araldi, W. Hu, J. Freund, O. Abdel-Wahab, S. Ibrahim, D. Skokos, S.A. Armstrong, R.L. Levine, C.Y. Park, I. Aifantis, Notch pathway activation targets AML-initiating cell homeostasis and differentiation. J. Exp. Med. 210, 301–319 (2013). doi:10.1084/jem.20121484 CrossRefPubMedPubMedCentral C. Lobry, P. Ntziachristos, D. Ndiaye-Lobry, P. Oh, L. Cimmino, N. Zhu, E. Araldi, W. Hu, J. Freund, O. Abdel-Wahab, S. Ibrahim, D. Skokos, S.A. Armstrong, R.L. Levine, C.Y. Park, I. Aifantis, Notch pathway activation targets AML-initiating cell homeostasis and differentiation. J. Exp. Med. 210, 301–319 (2013). doi:10.​1084/​jem.​20121484 CrossRefPubMedPubMedCentral
145.
go back to reference C. Dierks, R. Beigi, G.R. Guo, K. Zirlik, M.R. Stegert, P. Manley, C. Trussell, A. Schmitt-Graeff, K. Landwerlin, H. Veelken, M. Warmuth, Expansion of Bcr-Abl-positive leukemic stem cells is dependent on Hedgehog pathway activation. Cancer Cell 14, 238–249 (2008). doi:10.1016/j.ccr.2008.08.003 CrossRefPubMed C. Dierks, R. Beigi, G.R. Guo, K. Zirlik, M.R. Stegert, P. Manley, C. Trussell, A. Schmitt-Graeff, K. Landwerlin, H. Veelken, M. Warmuth, Expansion of Bcr-Abl-positive leukemic stem cells is dependent on Hedgehog pathway activation. Cancer Cell 14, 238–249 (2008). doi:10.​1016/​j.​ccr.​2008.​08.​003 CrossRefPubMed
147.
go back to reference L. Jin, Y. Tabe, S. Konoplev, Y. Xu, C.E. Leysath, H. Lu, S. Kimura, A. Ohsaka, M.B. Rios, L. Calvert, H. Kantarjian, M. Andreeff, M. Konopleva, CXCR4 up-regulation by imatinib induces chronic myelogenous leukemia (CML) cell migration to bone marrow stroma and promotes survival of quiescent CML cells. Mol. Cancer Ther. 7, 48–58 (2008). doi:10.1158/1535-7163.MCT-07-0042 CrossRefPubMed L. Jin, Y. Tabe, S. Konoplev, Y. Xu, C.E. Leysath, H. Lu, S. Kimura, A. Ohsaka, M.B. Rios, L. Calvert, H. Kantarjian, M. Andreeff, M. Konopleva, CXCR4 up-regulation by imatinib induces chronic myelogenous leukemia (CML) cell migration to bone marrow stroma and promotes survival of quiescent CML cells. Mol. Cancer Ther. 7, 48–58 (2008). doi:10.​1158/​1535-7163.​MCT-07-0042 CrossRefPubMed
148.
go back to reference L. Brault, A. Rovo, S. Decker, C. Dierks, A. Tzankov, J. Schwaller, CXCR4-SERINE339 regulates cellular adhesion, retention and mobilization, and is a marker for poor prognosis in acute myeloid leukemia. Leukemia 28, 566–576 (2014). doi:10.1038/leu.2013.201 CrossRefPubMed L. Brault, A. Rovo, S. Decker, C. Dierks, A. Tzankov, J. Schwaller, CXCR4-SERINE339 regulates cellular adhesion, retention and mobilization, and is a marker for poor prognosis in acute myeloid leukemia. Leukemia 28, 566–576 (2014). doi:10.​1038/​leu.​2013.​201 CrossRefPubMed
149.
go back to reference E. Weisberg, A.K. Azab, P.W. Manley, A.L. Kung, A.L. Christie, R. Bronson, I.M. Ghobrial, J.D. Griffin, Inhibition of CXCR4 in CML cells disrupts their interaction with the bone marrow microenvironment and sensitizes them to nilotinib. Leukemia 26, 985–990 (2012). doi:10.1038/leu.2011.360 CrossRefPubMed E. Weisberg, A.K. Azab, P.W. Manley, A.L. Kung, A.L. Christie, R. Bronson, I.M. Ghobrial, J.D. Griffin, Inhibition of CXCR4 in CML cells disrupts their interaction with the bone marrow microenvironment and sensitizes them to nilotinib. Leukemia 26, 985–990 (2012). doi:10.​1038/​leu.​2011.​360 CrossRefPubMed
150.
go back to reference S. Tavor, I. Petit, S. Porozov, A. Avigdor, A. Dar, L. Leider-Trejo, N. Shemtov, V. Deutsch, E. Naparstek, A. Nagler, T. Lapidot, CXCR4 regulates migration and development of human acute myelogenous leukemia stem cells in transplanted NOD/SCID mice. Cancer Res. 64, 2817–2824 (2004)CrossRefPubMed S. Tavor, I. Petit, S. Porozov, A. Avigdor, A. Dar, L. Leider-Trejo, N. Shemtov, V. Deutsch, E. Naparstek, A. Nagler, T. Lapidot, CXCR4 regulates migration and development of human acute myelogenous leukemia stem cells in transplanted NOD/SCID mice. Cancer Res. 64, 2817–2824 (2004)CrossRefPubMed
152.
go back to reference M. Shahjahani, J. Mohammadiasl, F. Noroozi, M. Seghatoleslami, S. Shahrabi, F. Saba, N. Saki, Molecular basis of chronic lymphocytic leukemia diagnosis and prognosis. Cell. Oncol. 38, 93–109 (2015). doi:10.1007/s13402-014-0215-3 CrossRef M. Shahjahani, J. Mohammadiasl, F. Noroozi, M. Seghatoleslami, S. Shahrabi, F. Saba, N. Saki, Molecular basis of chronic lymphocytic leukemia diagnosis and prognosis. Cell. Oncol. 38, 93–109 (2015). doi:10.​1007/​s13402-014-0215-3 CrossRef
153.
go back to reference A. Chávez-González, S. Avilés-Vázquez, D. Moreno-Lorenzana, H. Mayani, in Stem cell biology in normal life and diseases, ed. by K. Alimoghaddam (InTech, 2013) A. Chávez-González, S. Avilés-Vázquez, D. Moreno-Lorenzana, H. Mayani, in Stem cell biology in normal life and diseases, ed. by K. Alimoghaddam (InTech, 2013)
155.
go back to reference R. Bhatia, M. Holtz, N. Niu, R. Gray, D.S. Snyder, C.L. Sawyers, D.A. Arber, M.L. Slovak, S.J. Forman, Persistence of malignant hematopoietic progenitors in chronic myelogenous leukemia patients in complete cytogenetic remission following imatinib mesylate treatment. Blood 101, 4701–4707 (2003). doi:10.1182/blood-2002-09-2780 CrossRefPubMed R. Bhatia, M. Holtz, N. Niu, R. Gray, D.S. Snyder, C.L. Sawyers, D.A. Arber, M.L. Slovak, S.J. Forman, Persistence of malignant hematopoietic progenitors in chronic myelogenous leukemia patients in complete cytogenetic remission following imatinib mesylate treatment. Blood 101, 4701–4707 (2003). doi:10.​1182/​blood-2002-09-2780 CrossRefPubMed
156.
go back to reference M. Copland, A. Hamilton, L.J. Elrick, J.W. Baird, E.K. Allan, N. Jordanides, M. Barow, J.C. Mountford, T.L. Holyoake, Dasatinib (BMS-354825) targets an earlier progenitor population than imatinib in primary CML but does not eliminate the quiescent fraction. Blood 107, 4532–4539 (2006). doi:10.1182/blood-2005-07-2947 CrossRefPubMed M. Copland, A. Hamilton, L.J. Elrick, J.W. Baird, E.K. Allan, N. Jordanides, M. Barow, J.C. Mountford, T.L. Holyoake, Dasatinib (BMS-354825) targets an earlier progenitor population than imatinib in primary CML but does not eliminate the quiescent fraction. Blood 107, 4532–4539 (2006). doi:10.​1182/​blood-2005-07-2947 CrossRefPubMed
157.
go back to reference H.G. Jorgensen, E.K. Allan, N.E. Jordanides, J.C. Mountford, T.L. Holyoake, Nilotinib exerts equipotent antiproliferative effects to imatinib and does not induce apoptosis in CD34+ CML cells. Blood 109, 4016–4019 (2007). doi:10.1182/blood-2006-11-057521 CrossRefPubMed H.G. Jorgensen, E.K. Allan, N.E. Jordanides, J.C. Mountford, T.L. Holyoake, Nilotinib exerts equipotent antiproliferative effects to imatinib and does not induce apoptosis in CD34+ CML cells. Blood 109, 4016–4019 (2007). doi:10.​1182/​blood-2006-11-057521 CrossRefPubMed
158.
go back to reference A. Hamilton, G.V. Helgason, M. Schemionek, B. Zhang, S. Myssina, E.K. Allan, F.E. Nicolini, C. Muller-Tidow, R. Bhatia, V.G. Brunton, S. Koschmieder, T.L. Holyoake, Chronic myeloid leukemia stem cells are not dependent on Bcr-Abl kinase activity for their survival. Blood 119, 1501–1510 (2012). doi:10.1182/blood-2010-12-326843 CrossRefPubMedPubMedCentral A. Hamilton, G.V. Helgason, M. Schemionek, B. Zhang, S. Myssina, E.K. Allan, F.E. Nicolini, C. Muller-Tidow, R. Bhatia, V.G. Brunton, S. Koschmieder, T.L. Holyoake, Chronic myeloid leukemia stem cells are not dependent on Bcr-Abl kinase activity for their survival. Blood 119, 1501–1510 (2012). doi:10.​1182/​blood-2010-12-326843 CrossRefPubMedPubMedCentral
159.
160.
go back to reference J.H. Black, J.A. McCubrey, M.C. Willingham, J. Ramage, D.E. Hogge, A.E. Frankel, Diphtheria toxin-interleukin-3 fusion protein (DT(388)IL3) prolongs disease-free survival of leukemic immunocompromised mice. Leukemia 17, 155–159 (2003). doi:10.1038/sj.leu.2402744 CrossRefPubMed J.H. Black, J.A. McCubrey, M.C. Willingham, J. Ramage, D.E. Hogge, A.E. Frankel, Diphtheria toxin-interleukin-3 fusion protein (DT(388)IL3) prolongs disease-free survival of leukemic immunocompromised mice. Leukemia 17, 155–159 (2003). doi:10.​1038/​sj.​leu.​2402744 CrossRefPubMed
Metadata
Title
Novel strategies for targeting leukemia stem cells: sounding the death knell for blood cancer
Authors
Antonieta Chavez-Gonzalez
Babak Bakhshinejad
Katayoon Pakravan
Monica L. Guzman
Sadegh Babashah
Publication date
01-02-2017
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 1/2017
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-016-0297-1

Other articles of this Issue 1/2017

Cellular Oncology 1/2017 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