1
|
Metabolic alterations mediated by STAT3 promotes drug persistence in CML. Leukemia 2021; 35:3371-3382. [PMID: 34120146 PMCID: PMC8632690 DOI: 10.1038/s41375-021-01315-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 05/16/2021] [Accepted: 05/28/2021] [Indexed: 01/07/2023]
Abstract
Leukemic stem cells (LSCs) can acquire non-mutational resistance following drug treatment leading to therapeutic failure and relapse. However, oncogene-independent mechanisms of drug persistence in LSCs are incompletely understood, which is the primary focus of this study. We integrated proteomics, transcriptomics, and metabolomics to determine the contribution of STAT3 in promoting metabolic changes in tyrosine kinase inhibitor (TKI) persistent chronic myeloid leukemia (CML) cells. Proteomic and transcriptional differences in TKI persistent CML cells revealed BCR-ABL-independent STAT3 activation in these cells. While knockout of STAT3 inhibited the CML cells from developing drug-persistence, inhibition of STAT3 using a small molecule inhibitor sensitized the persistent CML cells to TKI treatment. Interestingly, given the role of phosphorylated STAT3 as a transcription factor, it localized uniquely to genes regulating metabolic pathways in the TKI-persistent CML stem and progenitor cells. Subsequently, we observed that STAT3 dysregulated mitochondrial metabolism forcing the TKI-persistent CML cells to depend on glycolysis, unlike TKI-sensitive CML cells, which are more reliant on oxidative phosphorylation. Finally, targeting pyruvate kinase M2, a rate-limiting glycolytic enzyme, specifically eradicated the TKI-persistent CML cells. By exploring the role of STAT3 in altering metabolism, we provide critical insight into identifying potential therapeutic targets for eliminating TKI-persistent LSCs.
Collapse
|
2
|
Desplat V, Vincenzi M, Lucas R, Moreau S, Savrimoutou S, Rubio S, Pinaud N, Bigat D, Enriquez E, Marchivie M, Routier S, Sonnet P, Rossi F, Ronga L, Guillon J. Synthesis and Antiproliferative Effect of Ethyl 4-[4-(4-Substituted Piperidin-1-yl)]benzylpyrrolo[1,2-a
]quinoxalinecarboxylate Derivatives on Human Leukemia Cells. ChemMedChem 2017; 12:940-953. [DOI: 10.1002/cmdc.201700049] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 02/17/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Vanessa Desplat
- UFR des Sciences Pharmaceutiques; Univ. Bordeaux; 33076 Bordeaux cedex France
- INSERM U1035, Cellules souches hématopoïétiques normales et leucémiques; 33000 Bordeaux France
| | - Marian Vincenzi
- UFR des Sciences Pharmaceutiques, Univ. Bordeaux; ARNA Laboratory; 33076 Bordeaux cedex France
- INSERM U1212, UMR CNRS 5320; ARNA Laboratory; 33000 Bordeaux France
- Department of Pharmacy and CIRPeB; University of Naples “Federico II”; Via Mezzocannone 16 80134 Naples Italy
| | - Romain Lucas
- UFR des Sciences Pharmaceutiques, Univ. Bordeaux; ARNA Laboratory; 33076 Bordeaux cedex France
- INSERM U1212, UMR CNRS 5320; ARNA Laboratory; 33000 Bordeaux France
| | - Stéphane Moreau
- UFR des Sciences Pharmaceutiques, Univ. Bordeaux; ARNA Laboratory; 33076 Bordeaux cedex France
- INSERM U1212, UMR CNRS 5320; ARNA Laboratory; 33000 Bordeaux France
| | - Solène Savrimoutou
- UFR des Sciences Pharmaceutiques, Univ. Bordeaux; ARNA Laboratory; 33076 Bordeaux cedex France
- INSERM U1212, UMR CNRS 5320; ARNA Laboratory; 33000 Bordeaux France
| | - Sandra Rubio
- UFR des Sciences Pharmaceutiques, Univ. Bordeaux; ARNA Laboratory; 33076 Bordeaux cedex France
- INSERM U1212, UMR CNRS 5320; ARNA Laboratory; 33000 Bordeaux France
| | - Noël Pinaud
- ISM-CNRS UMR 5255; Univ. Bordeaux; 351 cours de la Libération 33405 Talence cedex France
| | - David Bigat
- UFR des Sciences Pharmaceutiques, Univ. Bordeaux; ARNA Laboratory; 33076 Bordeaux cedex France
- INSERM U1212, UMR CNRS 5320; ARNA Laboratory; 33000 Bordeaux France
| | - Elodie Enriquez
- UFR des Sciences Pharmaceutiques, Univ. Bordeaux; ARNA Laboratory; 33076 Bordeaux cedex France
- INSERM U1212, UMR CNRS 5320; ARNA Laboratory; 33000 Bordeaux France
| | - Mathieu Marchivie
- ICMCB CNRS-UPR 9048; Univ. Bordeaux; 87 Avenue du Docteur Schweitzer 33608 Pessac cedex France
| | - Sylvain Routier
- Institut de Chimie Organique et analytique; Univ. Orleans, CNRS UMR 7311, ICOA; BP 6759, rue de Chartres 45067 Orléans cedex 2 France
| | - Pascal Sonnet
- Laboratoire de Glycochimie, des Antimicrobiens et des Agroressouces, UMR CNRS 7378, UFR de Pharmacie; Université de Picardie Jules Verne; 1 rue des Louvels 80037 Amiens cedex 01 France
| | - Filomena Rossi
- Department of Pharmacy and CIRPeB; University of Naples “Federico II”; Via Mezzocannone 16 80134 Naples Italy
| | - Luisa Ronga
- UFR des Sciences Pharmaceutiques, Univ. Bordeaux; ARNA Laboratory; 33076 Bordeaux cedex France
- INSERM U1212, UMR CNRS 5320; ARNA Laboratory; 33000 Bordeaux France
| | - Jean Guillon
- UFR des Sciences Pharmaceutiques, Univ. Bordeaux; ARNA Laboratory; 33076 Bordeaux cedex France
- INSERM U1212, UMR CNRS 5320; ARNA Laboratory; 33000 Bordeaux France
| |
Collapse
|
3
|
Desplat V, Vincenzi M, Lucas R, Moreau S, Savrimoutou S, Pinaud N, Lesbordes J, Peyrilles E, Marchivie M, Routier S, Sonnet P, Rossi F, Ronga L, Guillon J. Synthesis and evaluation of the cytotoxic activity of novel ethyl 4-[4-(4-substitutedpiperidin-1-yl)]benzyl-phenylpyrrolo[1,2-a]quinoxaline-carboxylate derivatives in myeloid and lymphoid leukemia cell lines. Eur J Med Chem 2016; 113:214-27. [DOI: 10.1016/j.ejmech.2016.02.047] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 11/17/2022]
|
4
|
Guiro K, Patel SA, Greco SJ, Rameshwar P, Arinzeh TL. Investigating breast cancer cell behavior using tissue engineering scaffolds. PLoS One 2015; 10:e0118724. [PMID: 25837691 PMCID: PMC4383476 DOI: 10.1371/journal.pone.0118724] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 01/13/2015] [Indexed: 12/16/2022] Open
Abstract
Despite early detection through the use of mammograms and aggressive intervention, breast cancer (BC) remains a clinical dilemma. BC can resurge after >10 years of remission. Studies indicate that BC cells (BCCs) with self-renewal and chemoresistance could be involved in dormancy. The majority of studies use in vitro, two-dimensional (2-D) monolayer cultures, which do not recapitulate the in vivo microenvironment. Thus, to determine the effect of three-dimensional (3-D) microenvironment on BCCs, this study fabricated tissue engineering scaffolds made of poly (ε-caprolactone) (PCL) having aligned or random fibers. Random and aligned fibers mimic, respectively, the random and highly organized collagen fibers found in the tumor extracellular matrix. Chemoresistant BCCs were obtained by treating with carboplatin. Western blot analysis of carboplatin resistant (treated) MDA-MB-231 (highly invasive, basal-like) and T47D (low-invasive, luminal) BCCs showed an increase in Bcl-2, Oct-4 and Sox-2, suggesting protection from apoptosis and increase in stem-like markers. Further studies with MDA-MB-231 BCCs seeded on the scaffolds showed little to no change in cell number over time for non-treated BCCs whereas on tissue culture polystyrene (TCP), non-treated BCCs displayed a significant increase in cell number at days 4 and 7 as compared to day 1 (p<0.05). Treated BCCs did not proliferate on TCP and the fibrous scaffolds. Little to no cyclin D1 was expressed for non-treated BCCs on TCP. On fibrous scaffolds, non-treated BCCs stained for cyclin D1 during the 7-day culture period. Treated BCCs expressed cyclin D1 on TCP and fibrous scaffolds during the 7-day culture period. Proliferation, viability and cell cycle analysis indicated that this 3-D culture prompted the aggressive BCCs to adopt a dormant phenotype, while the treated BCCs retained their phenotype. The findings indicate that random and aligned fibrous PCL scaffolds may provide a useful system to study how the 3-D microenvironment affects the behavior of BCCs.
Collapse
Affiliation(s)
- Khadidiatou Guiro
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, United States of America
| | - Shyam A. Patel
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Steven J. Greco
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Pranela Rameshwar
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Treena L. Arinzeh
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
5
|
Striving to achieve safe, permanent treatment discontinuation in chronic myeloid leukemia. Leuk Res 2013; 37:1395-403. [DOI: 10.1016/j.leukres.2013.07.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 07/19/2013] [Indexed: 12/11/2022]
|
6
|
Novel Combination Treatments Targeting Chronic Myeloid Leukemia Stem Cells. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2012; 12:94-105. [DOI: 10.1016/j.clml.2011.10.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/18/2011] [Accepted: 10/27/2011] [Indexed: 11/23/2022]
|
7
|
Roy S, Jørgensen HG, Roy P, Abed El Baky M, Melo JV, Strathdee G, Holyoake TL, Bartholomew C. BCR-ABL1 tyrosine kinase sustained MECOM expression in chronic myeloid leukaemia. Br J Haematol 2012; 157:446-56. [PMID: 22372463 DOI: 10.1111/j.1365-2141.2012.09078.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 02/01/2012] [Indexed: 01/21/2023]
Abstract
MECOM oncogene expression correlates with chronic myeloid leukaemia (CML) progression. Here we show that the knockdown of MECOM (E) and MECOM (ME) isoforms reduces cell division at low cell density, inhibits colony-forming cells by 34% and moderately reduces BCR-ABL1 mRNA and protein expression but not tyrosine kinase catalytic activity in K562 cells. We also show that both E and ME are expressed in CD34(+) selected cells of both CML chronic phase (CML-CP), and non-CML (normal) origin. Furthermore, MECOM mRNA and protein expression were repressed by imatinib mesylate treatment of CML-CP CD34(+) cells, K562 and KY01 cell lines whereas imatinib had no effect in non-CML BCR-ABL1 -ve CD34(+) cells. Together these results suggest that BCR-ABL1 tyrosine kinase catalytic activity regulates MECOM gene expression in CML-CP progenitor cells and that the BCR-ABL1 oncoprotein partially mediates its biological activity through MECOM. MECOM gene expression in CML-CP progenitor cells would provide an in vivo selective advantage, contributing to CML pathogenesis.
Collapse
Affiliation(s)
- Swagata Roy
- Department of Life Sciences, City Campus, Glasgow Caledonian University, Glasgow, UK
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Gallipoli P, Abraham SA, Holyoake TL. Hurdles toward a cure for CML: the CML stem cell. Hematol Oncol Clin North Am 2011; 25:951-66, v. [PMID: 22054728 DOI: 10.1016/j.hoc.2011.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Chronic myeloid leukemia (CML) is the first cancer in which a genetic alteration was proven to be of pathogenic significance and is considered a disease model for oncogene addiction, targeted therapy, and cancer stem cells (CSCs). The introduction of tyrosine kinase inhibitors (TKIs) resulted in dramatic improvement in response and survival for patients with CML in chronic phase (CP); however, CSCs are spared by TKIs. In this article, we review the role of CSCs in CML in CP, their persistence following TKI treatment, and current approaches to target this population in an attempt to achieve disease cure.
Collapse
Affiliation(s)
- Paolo Gallipoli
- Section of Experimental Haematology, Cancer Division, Faculty of Medicine, University of Glasgow, Paul O'Gorman Leukaemia Research Centre, Gartnavel General Hospital, 1053 Great Western Road, Glasgow, G12 0YN, UK
| | | | | |
Collapse
|
9
|
Pellicano F, Simara P, Sinclair A, Helgason GV, Copland M, Grant S, Holyoake TL. The MEK inhibitor PD184352 enhances BMS-214662-induced apoptosis in CD34+ CML stem/progenitor cells. Leukemia 2011; 25:1159-67. [PMID: 21483442 PMCID: PMC3643208 DOI: 10.1038/leu.2011.67] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 01/25/2011] [Accepted: 03/03/2011] [Indexed: 11/09/2022]
Abstract
The cytotoxic farnesyl transferase inhibitor BMS-214662 has been shown to potently induce mitochondrial apoptosis in primitive CD34+ chronic myeloid leukaemia (CML) stem/progenitor cells. Here, to enhance the BMS-214662 apoptotic effect, we further targeted the extracellular signal-regulated kinase (ERK) pathway, downstream of BCR-ABL, by treating CD34+ CML stem/progenitor cells with a highly selective adenosine triphosphate (ATP) non-competitive MEK inhibitor, PD184352. PD184352 increased the apoptotic effect of BMS-214662 in a CML blast crisis cell line, K562, and in primary chronic phase CD34+ CML cells. Compared with BMS-214662, after combination treatment we observed inhibition of ERK phosphorylation, increased Annexin-V levels, caspase-3, -8 and -9 activation and potentiated mitochondrial damage, associated with decreased levels of anti-apoptotic BCL-2 family protein MCL-1. Inhibition of K-RAS function by a dominant-negative mutant resulted in CML cell death and this process was further enhanced by the addition of BMS-214662 and PD184352. Together, these findings suggest that the addition of a MEK inhibitor improves the ability of BMS-214662 to selectively target CML stem/progenitor cells, notoriously insensitive to tyrosine kinase inhibitor treatment and presumed to be responsible for the persistence and relapse of the disease.
Collapse
MESH Headings
- Antigens, CD34/analysis
- Apoptosis/drug effects
- Benzamides/pharmacology
- Benzodiazepines/pharmacology
- Blast Crisis/enzymology
- Blast Crisis/pathology
- Drug Screening Assays, Antitumor
- Drug Synergism
- Enzyme Inhibitors/pharmacology
- Farnesyltranstransferase/antagonists & inhibitors
- Genes, Dominant
- Genes, ras
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/enzymology
- Humans
- Imidazoles/pharmacology
- K562 Cells/drug effects
- K562 Cells/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myeloid, Chronic-Phase/enzymology
- Leukemia, Myeloid, Chronic-Phase/pathology
- MAP Kinase Kinase 1/genetics
- MAP Kinase Kinase Kinases/antagonists & inhibitors
- Neoplasm Proteins/antagonists & inhibitors
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/enzymology
- Oncogene Protein p21(ras)/genetics
- Recombinant Fusion Proteins/genetics
- Tumor Cells, Cultured/drug effects
- Tumor Cells, Cultured/enzymology
Collapse
Affiliation(s)
- F Pellicano
- Paul O'Gorman Leukaemia Research Centre, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | | | | | | | | | |
Collapse
|
10
|
Burke AC, Swords RT, Kelly K, Giles FJ. Current status of agents active against the T315I chronic myeloid leukemia phenotype. Expert Opin Emerg Drugs 2011; 16:85-103. [DOI: 10.1517/14728214.2011.531698] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
11
|
Desplat V, Moreau S, Belisle-Fabre S, Thiolat D, Uranga J, Lucas R, Moor LD, Massip S, Jarry C, Mossalayi DM, Sonnet P, Déléris G, Guillon J. Synthesis and evaluation of the antiproliferative activity of novel isoindolo[2,1-a]quinoxaline and indolo[1,2-a]quinoxaline derivatives. J Enzyme Inhib Med Chem 2011; 26:657-67. [DOI: 10.3109/14756366.2010.548326] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Vanessa Desplat
- Université Bordeaux Segalen, CNRS FRE 3396, Pharmacochimie, Bordeaux
| | - Stéphane Moreau
- Université Bordeaux Segalen, CNRS FRE 3396, Pharmacochimie, Bordeaux
| | | | - Denis Thiolat
- Université Bordeaux Segalen, PPF Médicaments-Parasitologie, Bordeaux
| | - Juliette Uranga
- Université Bordeaux Segalen, CNRS FRE 3396, Pharmacochimie, Bordeaux
| | - Romain Lucas
- Université Bordeaux Segalen, CNRS FRE 3396, Pharmacochimie, Bordeaux
| | - Laure de Moor
- Université Bordeaux Segalen, CNRS FRE 3396, Pharmacochimie, Bordeaux
| | - Stéphane Massip
- Université Bordeaux Segalen, CNRS FRE 3396, Pharmacochimie, Bordeaux
| | - Christian Jarry
- Université Bordeaux Segalen, CNRS FRE 3396, Pharmacochimie, Bordeaux
| | | | - Pascal Sonnet
- Université de Picardie Jules Verne, UMR-CNRS 6219, Laboratoire des Glucides, Faculté de Pharmacie, Amiens, France
| | - Gérard Déléris
- Université Bordeaux Segalen, CNRS FRE 3396, Pharmacochimie, Bordeaux
| | - Jean Guillon
- Université Bordeaux Segalen, CNRS FRE 3396, Pharmacochimie, Bordeaux
| |
Collapse
|
12
|
Zhou P, Hatziieremia S, Elliott MA, Scobie L, Crossan C, Michie AM, Holyoake TL, Halbert GW, Jørgensen HG. Uptake of synthetic Low Density Lipoprotein by leukemic stem cells--a potential stem cell targeted drug delivery strategy. J Control Release 2010; 148:380-7. [PMID: 20869412 DOI: 10.1016/j.jconrel.2010.09.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 09/14/2010] [Accepted: 09/15/2010] [Indexed: 01/14/2023]
Abstract
Chronic Myeloid Leukemia (CML) stem/progenitor cells, which over-express Bcr-Abl, respond to imatinib by a reversible block in proliferation without significant apoptosis. As a result, patients are unlikely to be cured owing to the persistence of leukemic quiescent stem cells (QSC) capable of initiating relapse. Previously, we have reported that intracellular levels of imatinib in primary primitive CML cells (CD34+38(lo/⁻)), are significantly lower than in CML progenitor cells (total CD34+) and leukemic cell lines. The aim of this study was to determine if potentially sub-therapeutic intracellular drug concentrations in persistent leukemic QSC may be overcome by targeted drug delivery using synthetic Low Density Lipoprotein (sLDL) particles. As a first step towards this goal, however, the extent of uptake of sLDL by leukemic cell lines and CML patient stem/progenitor cells was investigated. Results with non-drug loaded particles have shown an increased and preferential uptake of sLDL by Bcr-Abl positive cell lines in comparison to Bcr-Abl negative. Furthermore, CML CD34+ and primitive CD34+38(lo/⁻) cells accumulated significantly higher levels of sLDL when compared with non-CML CD34+ cells. Thus, drug-loading the sLDL nanoparticles could potentially enhance intracellular drug concentrations in primitive CML cells and thus aid their eradication.
Collapse
MESH Headings
- Antigens, CD34/immunology
- Antineoplastic Agents/administration & dosage
- Cell Line, Tumor
- Cell Membrane Permeability
- Cells, Cultured
- Drug Delivery Systems/methods
- Hematopoietic Stem Cells/metabolism
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukocytes, Mononuclear/metabolism
- Lipoproteins, LDL/chemical synthesis
- Lipoproteins, LDL/chemistry
- Lipoproteins, LDL/pharmacokinetics
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
Collapse
Affiliation(s)
- Peixun Zhou
- Paul O'Gorman Leukemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Gartnavel General Hospital, 1053 Great Western Road, Glasgow G120NY, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
Chronic myeloid leukemia (CML) arises as a consequence of a chromosomal translocation giving rise to the Philadelphia chromosome and Bcr-Abl oncogene. CML is a clonal disease of stem cell origin and an excellent example of a malignancy in which tumor-initiating cells may hold the key to disease eradication. The known molecular basis of CML has enabled the development of Abl-specific tyrosine kinase inhibitors, such as imatinib mesylate. However, the success of tyrosine kinase inhibitors, as rationally designed first-line therapies, has been tempered by problems of disease persistence and resistance. Residual disease has been shown to be enriched within the stem cell compartment and to persist at stable levels for up to 5 years of complete cytogenetic response. This finding has led to further searches for novel strategies aimed at eliminating these cells; such strategies may be essential in achieving cure. The most significant recent findings are discussed in this review.
Collapse
|
14
|
A diphtheria toxin interleukin-3 fusion protein synergizes with tyrosine kinase inhibitors in killing leukemic progenitors from BCR/ABL positive acute leukemia. Leuk Res 2010; 34:1035-42. [DOI: 10.1016/j.leukres.2009.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Revised: 11/04/2009] [Accepted: 12/16/2009] [Indexed: 11/22/2022]
|
15
|
Traer E, Deininger MW. How Much and How Long: Tyrosine Kinase Inhibitor Therapy in Chronic Myeloid Leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2010; 10 Suppl 1:S20-6. [DOI: 10.3816/clml.2010.s.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
16
|
Zhang B, Strauss AC, Chu S, Li M, Ho Y, Shiang KD, Snyder DS, Huettner CS, Shultz L, Holyoake T, Bhatia R. Effective targeting of quiescent chronic myelogenous leukemia stem cells by histone deacetylase inhibitors in combination with imatinib mesylate. Cancer Cell 2010; 17:427-42. [PMID: 20478526 PMCID: PMC2873971 DOI: 10.1016/j.ccr.2010.03.011] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2009] [Revised: 12/18/2009] [Accepted: 03/19/2010] [Indexed: 11/15/2022]
Abstract
Imatinib mesylate (IM) induces remission in chronic myelogenous leukemia (CML) patients but does not eliminate leukemia stem cells (LSCs), which remain a potential source of relapse. Here we investigated the ability of HDAC inhibitors (HDACis) to target CML stem cells. Treatment with HDACis combined with IM effectively induced apoptosis in quiescent CML progenitors resistant to elimination by IM alone, and eliminated CML stem cells capable of engrafting immunodeficient mice. In vivo administration of HDACis with IM markedly diminished LSCs in a transgenic mouse model of CML. The interaction of IM and HDACis inhibited genes regulating hematopoietic stem cell maintenance and survival. HDACi treatment represents an effective strategy to target LSCs in CML patients receiving tyrosine kinase inhibitors.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis
- Benzamides
- Cell Proliferation
- Fusion Proteins, bcr-abl/metabolism
- Histone Deacetylase Inhibitors/pharmacology
- Histone Deacetylase Inhibitors/therapeutic use
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Mice, Transgenic
- Neoplastic Stem Cells/drug effects
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
Collapse
Affiliation(s)
- Bin Zhang
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Adam C. Strauss
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Su Chu
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Min Li
- Department of Information Sciences, City of Hope National Medical Center, Duarte, CA
| | - Yinwei Ho
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Keh-Dong Shiang
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA
- Department of Information Sciences, City of Hope National Medical Center, Duarte, CA
| | - David S Snyder
- Department of Hematology and HCT, City of Hope National Medical Center, Duarte, CA
| | - Claudia S. Huettner
- Beffer Institute of Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA
| | | | - Tessa Holyoake
- Section of Experimental Haematology, Cancer Division, University of Glasgow, Scotland, UK
| | - Ravi Bhatia
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
17
|
Hiwase DK, White DL, Powell JA, Saunders VA, Zrim SA, Frede AK, Guthridge MA, Lopez AF, D'Andrea RJ, To LB, Melo JV, Kumar S, Hughes TP. Blocking cytokine signaling along with intense Bcr-Abl kinase inhibition induces apoptosis in primary CML progenitors. Leukemia 2010; 24:771-8. [PMID: 20130598 DOI: 10.1038/leu.2009.299] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In chronic myeloid leukemia (CML) cell lines, brief exposure to pharmacologically relevant dasatinib concentrations results in apoptosis. In this study, we assess the impact of intensity and duration of Bcr-Abl kinase inhibition on primary CD34(+) progenitors of chronic phase CML patients. As CML cells exposed to dasatinib in vivo are in a cytokine-rich environment, we also assessed the effect of cytokines (six growth factors cocktail or granulocyte-macrophage colony-stimulating factor (CSF) or granulocyte-CSF) in combination with dasatinib. In the presence of cytokines, short-term intense Bcr-Abl kinase inhibition (>or=90% p-Crkl inhibition) with 100 nM dasatinib did not reduce CD34(+) colony-forming cells (CFCs). In contrast, without cytokines, short-term exposure to dasatinib reduced CML-CD34(+) CFCs by 70-80%. When cytokines were added immediately after short-term exposure to dasatinib, CML-CD34(+) cells remained viable, suggesting that oncogene dependence of these cells can be overcome by concomitant or subsequent exposure to cytokines. Additional inhibition of Janus tyrosine kinase (Jak) activity re-established the sensitivity of CML progenitors to intense Bcr-Abl kinase inhibition despite the presence of cytokines. These findings support the contention that therapeutic strategies combining intense Bcr-Abl kinase inhibition and blockade of cytokine signaling pathways can be effective for eradication of CML progenitors.
Collapse
Affiliation(s)
- D K Hiwase
- Division of Haematology, SA Pathology, Adelaide, South Australia, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Imatinib mesylate has transformed the treatment for chronic myeloid leukemia (CML). The vast majority of patients obtain hematologic remission, with a low probability of progression of disease. Yet imatinib rarely cures CML, and current recommendations dictate lifelong treatment with imatinib. In this review we analyze the biology behind the failure of imatinib to fully eradicate CML. We review evidence that indicates that the leukemic stem cell for CML is inherently resistant to imatinib, and that imatinib treatment itself may enhance this resistance.
Collapse
Affiliation(s)
- Robert L Redner
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
19
|
Optimising chronic myeloid leukaemia therapy in the face of resistance to tyrosine kinase inhibitors – A synthesis of clinical and laboratory data. Blood Rev 2010; 24:1-9. [DOI: 10.1016/j.blre.2009.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
20
|
Sang L, Roberts JM, Coller HA. Hijacking HES1: how tumors co-opt the anti-differentiation strategies of quiescent cells. Trends Mol Med 2009; 16:17-26. [PMID: 20022559 DOI: 10.1016/j.molmed.2009.11.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 11/03/2009] [Accepted: 11/17/2009] [Indexed: 12/19/2022]
Abstract
Quiescent and tumor cells share the ability to evade irreversible cell fates. Recent studies have shown that the transcriptional regulator Hairy and Enhancer of Split 1 (HES1) protects quiescent fibroblasts from differentiation or senescence. HES1 is highly expressed in rhabdomyosarcomas, and the inhibition of HES1 restores differentiation in these cells. Pathways that lead to elevated HES1 levels, such as the Notch and Hedgehog pathways, are frequently upregulated in tumors. Compounds that inhibit these pathways induce differentiation and apoptosis in cancer cells and several are in clinical trials. HES1 might repress gene expression in part by recruiting histone deacetylases (HDACs). HDACs inhibit differentiation, whereas histone deacetylase inhibitors (HDACis) induce differentiation or apoptosis in tumors and are also showing promise as therapeutics. Small molecules that directly target HES1 itself were recently identified. Here, we discuss the importance of HES1 function in quiescent and tumor cells. Elucidating the pathways that control quiescence could provide valuable information not only for treating cancer but also other diseases.
Collapse
Affiliation(s)
- Liyun Sang
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | |
Collapse
|
21
|
Quintás-Cardama A, Cortés JE. The next generation of therapies for chronic myeloid leukemia. CLINICAL LYMPHOMA & MYELOMA 2009; 9 Suppl 4:S395-403. [PMID: 20007109 DOI: 10.3816/clm.2009.s.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Therapy with the tyrosine kinase inhibitor (TKI) represents the current standard first-line therapy for the management of patients with chronic myeloid leukemia (CML). Although most patients respond satisfactorily to imatinib, a subset of patients develops resistance mainly because of the acquisition of mutations within the kinase domain of BCR-ABL1 that impair the ability of TKIs to block the activity of the enzyme. Moreover, BCR-ABL1 transcripts can be detected in most patients by molecular techniques, underscoring the limitations of imatinib to eradicate minimal residual disease. Although the resistance conferred by most BCR-ABL1 mutations can be overcome with the use of second-generation TKIs such as nilotinib, dasastinib, bosutinib, or bafetinib, the T315I mutation, which represents a common resistance pathway in CML, remains unassailable to TKI therapy. We herein discuss current research efforts in 2 areas of vital importance in CML research, the management of patients with imatinib-resistant mutations, with particular emphasis on those carrying T315I, and the eradication of residual disease.
Collapse
Affiliation(s)
- Alfonso Quintás-Cardama
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, 77030, USA
| | | |
Collapse
|
22
|
BMS-214662 induces mitochondrial apoptosis in chronic myeloid leukemia (CML) stem/progenitor cells, including CD34+38- cells, through activation of protein kinase Cbeta. Blood 2009; 114:4186-96. [PMID: 19738029 DOI: 10.1182/blood-2009-05-219550] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a hematopoietic stem cell disorder maintained by cancer stem cells. To target this population, we investigated the mechanism of action of BMS-214662, developed as a farnesyl transferase inhibitor (FTI) and unique in inducing apoptosis in these cells. By contrast, a related congener and equally effective FTI, BMS-225975 does not induce apoptosis, indicating a novel mechanism of action. BMS-214662 significantly and selectively induced apoptosis in primitive CD34(+)38(-) CML compared with normal cells. Apoptosis proceeded via the intrinsic pathway: Bax conformational changes, loss of mitochondrial membrane potential, generation of reactive oxygen species, release of cytochrome c, and caspase-9/3 activation were noted. Up-regulation of protein kinase Cbeta (PKCbeta), down-regulation of E2F1, and phosphorylation of cyclin A-associated cyclin-dependent kinase 2 preceded these changes. Cotreatment of CML CD34(+) and CD34(+)38(-) cells with PKC modulators, bryostatin-1, or hispidin markedly decreased these early events and the subsequent apoptosis. None of these events was elicited by BMS-214662 in normal CD34(+) cells or by BMS-225975 in CML CD34(+) cells. These data suggest that BMS-214662 selectively elicits a latent apoptotic pathway in CML stem cells that is initiated by up-regulation of PKCbeta and mediated by Bax activation, providing a molecular framework for development of novel therapeutics.
Collapse
|
23
|
Volpe G, Panuzzo C, Ulisciani S, Cilloni D. Imatinib resistance in CML. Cancer Lett 2009; 274:1-9. [DOI: 10.1016/j.canlet.2008.06.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Revised: 03/20/2008] [Accepted: 06/02/2008] [Indexed: 12/25/2022]
|
24
|
Abstract
The development of cancer vaccines directed against myeloid leukaemias has been a research area of intense interest in the past decade. Both human studies in vitro and mouse models in vivo have demonstrated that leukaemia-associated antigens (LAAs), such as the fusion protein BCR-ABL, Wilms' tumour protein and proteinase 3, may serve as effective targets for cellular immunotherapy. Peptide-based vaccines are able to induce cytotoxic T-lymphocyte responses that kill leukaemia cells. Based on these results, pilot clinical trials have been initiated in chronic and acute myeloid leukaemia and other haematological malignancies, which include vaccination of patients with synthetic peptides derived from these LAAs. Results from these trials show that peptide vaccines are able to induce immune responses that are sometimes associated with clinical benefit. These early clinical results are promising and provide valuable information for future improvement of the vaccines. This chapter will focus mainly on discussing the preclinical studies of peptide vaccines in human systems, the results from clinical trials and the future prospects for vaccine therapy for myeloid leukaemia.
Collapse
Affiliation(s)
- Tao Dao
- Molecular Pharmacology & Chemistry Program, Leukemia Service, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | |
Collapse
|
25
|
Quintás-Cardama A, Cortes J. Nilotinib: a phenylamino-pyrimidine derivative with activity against BCR-ABL, KIT and PDGFR kinases. Future Oncol 2008; 4:611-21. [PMID: 18922118 DOI: 10.2217/14796694.4.5.611] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The BCR-ABL kinase inhibitor imatinib mesylate is currently the standard therapy for patients with chronic myeloid leukemia (CML). However, mutations within the ABL kinase domain interfering with drug binding have been identified as the main mechanism of resistance to imatinib. Multiple distinct BCR-ABL kinase mutant isoforms conferring varying degrees of resistance to tyrosine kinase inhibitors have been reported. Nilotinib is a tyrosine kinase inhibitor 30-fold more potent than imatinib against BCR-ABL kinase. Nilotinib is active against a wide range of imatinib-resistant BCR-ABL mutant isoforms, except for T315I. Results from Phase II studies of nilotinib for patients with CML after failure or intolerance to imatinib therapy have shown a favorable toxicity profile and confirmed the high efficacy of nilotinib in this setting. Studies addressing the activity of nilotinib in newly-diagnosed patients with CML are underway. Furthermore, nilotinib is a potent inhibitor of KIT and PDGFR kinases. Here, we review the preclinical development of nilotinib and the activity of this agent in patients with CML and in tumors driven by KIT and/or PDGFR mutant kinases, such as gastrointestinal stromal tumors and some forms of clonal hypereosinophilia.
Collapse
|
26
|
Padmanabhan S, Ravella S, Curiel T, Giles F. Current status of therapy for chronic myeloid leukemia: a review of drug development. Future Oncol 2008; 4:359-77. [PMID: 18518762 DOI: 10.2217/14796694.4.3.359] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic myeloid leukemia (CML) has led the way for developing rational drug development in cancer. Most cases of CML diagnosed and treated in chronic phase are extremely well controlled with imatinib monotherapy, and primary resistance is very uncommon. Even though the treatment failure rate is low, the emergence of drug resistance and the lack of eradication of the hematopoietic stem cell clone has prompted a wave of drugs to address one or both these problems. Several clinical trials (Phase I and II) of dasatinib or nilotinib in the treatment of imatinib-resistant or -intolerant Ph chromosome-positive leukemia have already reported a remarkable rate of hematologic response greater than 90% for chronic-phase patients. These drugs minimize the risk of acquired drug resistance that is particularly seen within the first 24-36 months of therapy, and can prevent early failure in these patients, Furthermore, rational, noncross-resistant combinations that include a T315I inhibitor and drugs that can eradicate the hematopoietic stem cell clone may extend the coverage to virtually all patients with bcr-abl. Here we review the 6-year impact of the 'magic pill', Gleevec, (Glivec), including the emerging problems with its treatment, the efficacy data of dasatinib and nilotinib and the very promising data of the newer generation of drugs for CML.
Collapse
Affiliation(s)
- Swami Padmanabhan
- Department of Hematology/Oncology, Institute for Drug Development, Cancer Therapy and Research Center, San Antonio, TX, USA
| | | | | | | |
Collapse
|
27
|
McCloskey SM, McMullin MF, Walker B, Irvine AE. The therapeutic potential of the proteasome in leukaemia. Hematol Oncol 2008; 26:73-81. [PMID: 18324639 DOI: 10.1002/hon.848] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Many cellular processes converge on the proteasome, and its key regulatory role is increasingly being recognized. Proteasome inhibition allows the manipulation of many cellular pathways including apoptotic and cell cycle mechanisms. The proteasome inhibitor bortezomib has enhanced responses in newly diagnosed patients with myeloma and provides a new line of therapy in relapsed and refractory patients. Malignant cells are more sensitive to proteasome inhibition than normal haematopoietic cells. Proteasome inhibition enhances many conventional therapies and its role in leukaemia is promising.
Collapse
|
28
|
Abstract
Tyrosine kinase inhibitor (TKI) therapy for chronic myeloid leukaemia (CML) is the consummate success story for targeted therapy, yet relapse is a nearly inevitable consequence of cessation or interruption of therapy. Primitive TKI-refractory CML stem cells are the likely source of these relapses, as they provide sanctuary for the Philadelphia chromosome. In advanced disease, their progressively anaplastic progeny ultimately maintain CML independently of the CML haematopoietic stem cell (HSC). Interestingly, there are at least two distinct cell types capable of self-renewal in different phases of CML: first, a primitive HSC with BCR-ABL mutation, which maintains the more indolent chronic-phase disease and, second, a coexisting mutated progenitor cell which acquires stem cell characteristics responsible for rapid cell expansion in advanced disease.
Collapse
MESH Headings
- Benzamides
- Drug Resistance, Neoplasm/genetics
- Epigenesis, Genetic
- Fusion Proteins, bcr-abl/genetics
- Hematopoietic Stem Cells/enzymology
- Hematopoietic Stem Cells/pathology
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Neoplastic Stem Cells/enzymology
- Neoplastic Stem Cells/pathology
- Piperazines/therapeutic use
- Protein Kinase Inhibitors/therapeutic use
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Pyrimidines/therapeutic use
- Tumor Suppressor Proteins/genetics
Collapse
Affiliation(s)
- Michael Savona
- University of Michigan, Internal Medicine-Hematology Oncology, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109-45936, USA.
| | | |
Collapse
|
29
|
Snead JL, O'Hare T, Eide CA, Deininger MW. New Strategies for the First-Line Treatment of Chronic Myeloid Leukemia: Can Resistance Be Avoided? ACTA ACUST UNITED AC 2008; 8 Suppl 3:S107-17. [DOI: 10.3816/clm.2008.s.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
30
|
Copland M, Pellicano F, Richmond L, Allan EK, Hamilton A, Lee FY, Weinmann R, Holyoake TL. BMS-214662 potently induces apoptosis of chronic myeloid leukemia stem and progenitor cells and synergizes with tyrosine kinase inhibitors. Blood 2008; 111:2843-53. [PMID: 18156496 DOI: 10.1182/blood-2007-09-112573] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chronic myeloid leukemia (CML), a hematopoietic stem-cell disorder, cannot be eradicated by conventional chemotherapy or the tyrosine kinase inhibitor imatinib mesylate (IM). To target CML stem/progenitor cells, we investigated BMS-214662, a cytotoxic farnesyltransferase inhibitor, previously reported to kill nonproliferating tumor cells. IM or dasatinib alone reversibly arrested proliferation of CML stem/progenitor cells without inducing apoptosis. In contrast, BMS-214662, alone or in combination with IM or dasatinib, potently induced apoptosis of both proliferating and quiescent CML stem/progenitor cells with less than 1% recovery of Philadelphia-positive long-term culture-initiating cells. Normal stem/progenitor cells were relatively spared by BMS-214662, suggesting selectivity for leukemic stem/progenitor cells. The ability to induce selective apoptosis of leukemic stem/progenitor cells was unique to BMS-214662 and not seen with a structurally similar agent BMS-225975. BMS-214662 was cytotoxic against CML blast crisis stem/progenitor cells, particularly in combination with a tyrosine kinase inhibitor and equally effective in cell lines harboring wild-type vs mutant BCR-ABL, including the T315I mutation. This is the first report of an agent with activity in resistant and blast crisis CML that selectively kills CML stem/progenitor cells through apoptosis and offers potential for eradication of chronic phase CML.
Collapse
MESH Headings
- Antigens, CD34/metabolism
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Benzamides
- Benzodiazepines/pharmacology
- Blast Crisis/pathology
- Caspase 3/metabolism
- Cell Death/drug effects
- Cell Survival/drug effects
- Dasatinib
- Drug Screening Assays, Antitumor
- Drug Synergism
- Farnesyltranstransferase/antagonists & inhibitors
- Fusion Proteins, bcr-abl/chemistry
- Fusion Proteins, bcr-abl/metabolism
- Humans
- Imatinib Mesylate
- Imidazoles/pharmacology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mutation/genetics
- Neoplastic Stem Cells/pathology
- Philadelphia Chromosome
- Piperazines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Protein Structure, Tertiary
- Pyrimidines/pharmacology
- Thiazoles/pharmacology
Collapse
Affiliation(s)
- Mhairi Copland
- Section of Experimental Haematology and Haemopoietic Stem Cells, Division of Cancer Sciences and Molecular Pathology, University of Glasgow, Glasgow, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
CML (chronic myeloid leukaemia) is a myeloproliferative disease that originates in an HSC (haemopoietic stem cell) as a result of the t(9;22) translocation, giving rise to the Ph (Philadelphia chromosome) and bcr-abl oncoprotein. The disease starts in CP (chronic phase), but as a result of genomic instability, it progresses over time to accelerated phase and then to BC (blast crisis), becoming increasingly resistant to therapy. bcr-abl is a constitutively active tyrosine kinase that has been targeted by TKIs (tyrosine kinase inhibitors), including IM (imatinib mesylate), nilotinib and dasatinib. We have developed various flow cytometry techniques to enable us to isolate candidate CML stem cells from CP patients at diagnosis that efflux Hoechst dye, express CD34, lack CD38 and are cytokine-non-responsive in culture over periods of up to 12 days in growth factors. These stem cells have been shown to regenerate bcr-abl-positive haemopoiesis in immunocompromised mice upon transplantation. We previously demonstrated that IM was antiproliferative for CML stem cells but did not induce apoptosis. Clinical experience now confirms that IM may not target CML stem cells in vivo with few patients achieving complete molecular remission and relapse occurring rapidly upon drug withdrawal. Our recent efforts have focused on understanding why CML stem cells are resistant to IM and on trying to find novel ways to induce apoptosis of this population. We have shown that CML stem cells express very high levels of functional wild-type bcr-abl; no kinase domain mutations have been detected in the stem cell population. Dasatinib, a more potent multitargeted TKI than IM, inhibits bcr-abl activity more efficiently than IM but still does not induce apoptosis of the stem cell population. Most recently, we have tested a number of novel drug combinations and found that FTIs (farnesyl transferase inhibitors) have activity against CML. BMS-214662 is the most effective of these and induces apoptosis of phenotypically and functionally defined CML stem cells in vitro, as a single agent and in combination with IM or dasatinib. The effect against CML stem cells is selective with little effect on normal stem cells. The drug is also effective against BC CML stem cells and equally effective against wild-type and mutant bcr-abl, including the most resistant mutant T315I. In association with apoptosis, there is activation of caspase 8 and caspase 3, inhibition of the MAPK pathway, IAP-1 (inhibitor of apoptosis protein-1), NF-kappaB (nuclear factor kappaB) and iNOS (inducible nitric oxide synthase). Furthermore, BMS-214662 synergizes with MEK1/2 [MAPK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) kinase 1/2] inhibitors, suggesting a second mechanism other that RAS inhibition for induction of apoptosis. Our intentions are now to explore the activity of BMS-214662 in other cancer stem cell disorders and to move this preclinical work to a clinical trial combining dasatinib with BMS-214662 in CML.
Collapse
|
32
|
Giles FJ, DeAngelo DJ, Baccarani M, Deininger M, Guilhot F, Hughes T, Mauro M, Radich J, Ottmann O, Cortes J. Optimizing Outcomes for Patients With Advanced Disease in Chronic Myelogenous Leukemia. Semin Oncol 2008; 35:S1-17; quiz S18-20. [DOI: 10.1053/j.seminoncol.2007.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
33
|
Jagani Z, Singh A, Khosravi-Far R. FoxO tumor suppressors and BCR-ABL-induced leukemia: a matter of evasion of apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1785:63-84. [PMID: 17980712 PMCID: PMC2180393 DOI: 10.1016/j.bbcan.2007.10.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Revised: 10/04/2007] [Accepted: 10/06/2007] [Indexed: 12/14/2022]
Abstract
Numerous studies have revealed that the BCR-ABL oncoprotein abnormally engages a multitude of signaling pathways, some of which may be important for its leukemogenic properties. Central to this has been the determination that the tyrosine kinase function of BCR-ABL is mainly responsible for its transforming potential, and can be targeted with small molecule inhibitors, such as imatinib mesylate (Gleevec, STI-571). Despite this apparent success, the development of clinical resistance to imatinib therapy, and the inability of imatinib to eradicate BCR-ABL-positive malignant hematopoietic progenitors demand detailed investigations of additional effector pathways that can be targeted for CML treatment. The promotion of cellular survival via the suppression of apoptotic pathways is a fundamental characteristic of tumor cells that enables resistance to anti-cancer therapies. As substrates of survival kinases such as Akt, the FoxO family of transcription factors, particularly FoxO3a, has emerged as playing an important role in the cell cycle arrest and apoptosis of hematopoietic cells. This review will discuss our current understanding of BCR-ABL signaling with a focus on apoptotic suppressive mechanisms and alternative approaches to CML therapy, as well as the potential for FoxO transcription factors as novel therapeutic targets.
Collapse
Affiliation(s)
- Zainab Jagani
- Department of Pathology, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston MA 02115
- Biological and Biomedical Sciences (BBS) Program at Harvard Medical School
| | - Amrik Singh
- Department of Pathology, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston MA 02115
| | - Roya Khosravi-Far
- Department of Pathology, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston MA 02115
- Biological and Biomedical Sciences (BBS) Program at Harvard Medical School
| |
Collapse
|
34
|
Melo JV, Chuah C. Novel agents in CML therapy: tyrosine kinase inhibitors and beyond. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2008; 2008:427-435. [PMID: 19074121 DOI: 10.1182/asheducation-2008.1.427] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The emergence of resistance to imatinib has become a significant problem despite the remarkable clinical results achieved with this tyrosine kinase inhibitor in the treatment of chronic myeloid leukaemia. The most common cause of imatinib resistance is the selection of leukemic clones with point mutations in the Abl kinase domain. These mutations lead to amino acid substitutions and prevent the appropriate binding of imatinib. Genomic amplification of BCR-ABL, modulation of drug efflux or influx transporters, and Bcr-Abl-independent mechanisms also play important roles in the development of resistance. Persistent disease is another therapeutic challenge and may in part, be due to the inability of imatinib to eradicate primitive stem cell progenitors. A multitude of novel agents have been developed and have shown in vitro and in vivo efficacy in overcoming imatinib resistance. In this review, we will discuss the current status of the ATP-competitive and non-ATP-competitive Bcr-Abl tyrosine kinase inhibitors. We will also describe inhibitors acting on targets found in signaling pathways downstream of Bcr-Abl, such as the Ras-Raf-mitogen-activated protein kinase and phosphatidylinositol-3 kinase-Akt-mammalian target of rapamycin pathways, and targets without established links with Bcr-Abl.
Collapse
Affiliation(s)
- Junia V Melo
- Division of Haematology, Institute of Medical & Veterinary Science, Adelaide SA, Australia.
| | | |
Collapse
|
35
|
Graham SM, Vass JK, Holyoake TL, Graham GJ. Transcriptional Analysis of Quiescent and Proliferating CD34+ Human Hemopoietic Cells from Normal and Chronic Myeloid Leukemia Sources. Stem Cells 2007; 25:3111-20. [PMID: 17717066 DOI: 10.1634/stemcells.2007-0250] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Quiescent and dividing hemopoietic stem cells (HSC) display marked differences in their ability to move between the peripheral circulation and the bone marrow. Specifically, long-term engraftment potential predominantly resides in the quiescent HSC subfraction, and G-CSF mobilization results in the preferential accumulation of quiescent HSC in the periphery. In contrast, stem cells from chronic myeloid leukemia (CML) patients display a constitutive presence in the circulation. To understand the molecular basis for this, we have used microarray technology to analyze the transcriptional differences between dividing and quiescent, normal, and CML-derived CD34+ cells. Our data show a remarkable transcriptional similarity between normal and CML dividing cells, suggesting that the effects of BCR-ABL on the CD34+ cell transcriptome are more limited than previously thought. In addition, we show that quiescent CML cells are more similar to their dividing counterparts than quiescent normal cells are to theirs. We also show these transcriptional differences to be reflected in the altered proliferative activity of normal and CML CD34+ cells. Of the most interest is that the major class of genes that is more abundant in the quiescent cells compared with the dividing cells encodes members of the chemokine family. We propose a role for chemokines expressed by quiescent HSC in the orchestration of CD34+ cell mobilization. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
MESH Headings
- Antigens, CD34/biosynthesis
- Antigens, CD34/genetics
- Cell Cycle/genetics
- Cell Division/genetics
- Cell Proliferation
- Cells, Cultured
- Female
- Gene Expression Profiling/methods
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Male
- Neoplastic Cells, Circulating/metabolism
- Neoplastic Cells, Circulating/pathology
- Oligonucleotide Array Sequence Analysis/methods
- Resting Phase, Cell Cycle/genetics
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Susan M Graham
- Experimental Haematology, Division of Cancer Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | | | | | | |
Collapse
|
36
|
Krause DS, Van Etten RA. Right on target: eradicating leukemic stem cells. Trends Mol Med 2007; 13:470-81. [PMID: 17981087 PMCID: PMC4344828 DOI: 10.1016/j.molmed.2007.09.003] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 09/04/2007] [Accepted: 09/07/2007] [Indexed: 12/18/2022]
Abstract
Less than a third of adults with acute myeloid leukemia (AML) are cured by current treatments, emphasizing the need for new approaches to therapy. The discovery over a decade ago that myeloid leukemias originate from rare stem-like cells that can transfer the disease to immunodeficient mice suggested that these 'leukemia stem cells' (LSCs) are responsible for relapse of leukemia following conventional or targeted cancer therapy and that eradication of LSCs might be necessary to cure the disease permanently. Several recent studies have provided insight into the signaling pathways underlying the LSC phenotype and have also described approaches to eliminate LSCs with antibodies. Here, we review recent advances in LSC research and discuss novel therapeutic strategies to specifically target LSCs.
Collapse
Affiliation(s)
- Daniela S. Krause
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Richard A. Van Etten
- Molecular Oncology Research Institute and Division of Hematology/Oncology, Tufts-New England Medical Center, Boston, MA 02111, USA
| |
Collapse
|
37
|
Singh T, Casson C. Strategies for Overcoming Imatinib Mesylate Resistance in Chronic Myelogenous Leukemia. Am J Ther 2007; 14:484-7. [PMID: 17890939 DOI: 10.1097/01.mjt.0000212892.89962.0b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Imatinib mesylate was approved for the treatment of chronic myelogenous leukemia more than 5 years ago. This drug enabled us to put a very high percentage of patients into hematologic, cytogenetic, and molecular remission. Some patients were resistant to Imatinib from the onset of the treatment whereas others became resistant after showing an initial response to Imatinib mesylate. Various strategies have been tried to overcome the resistance to this drug including using newer combinations, higher dosage of Imatinib and development of newer compounds. This article will discuss these strategies in detail.
Collapse
Affiliation(s)
- Tejvir Singh
- Heartland Oncology and Hematology, Council Bluffs, IA, USA.
| | | |
Collapse
|
38
|
Konstantinopoulos PA, Karamouzis MV, Papavassiliou AG. Post-translational modifications and regulation of the RAS superfamily of GTPases as anticancer targets. Nat Rev Drug Discov 2007; 6:541-55. [PMID: 17585331 DOI: 10.1038/nrd2221] [Citation(s) in RCA: 357] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The involvement of the RAS superfamily of monomeric GTPases in carcinogenesis is increasingly being appreciated. A complex array of post-translational modifications and a highly sophisticated protein network regulate the spatio-temporal activation of these GTPases. Previous attempts to pharmacologically target this family have focused on the development of farnesyltransferase inhibitors, but the performance of such agents in cancer clinical trials has not been as good as hoped. Here, we review emerging druggable targets and novel therapeutic approaches targeting prenylation and post-prenylation modifications and the functional regulation of GDP/GTP exchange as exciting alternatives for anticancer therapy.
Collapse
|
39
|
Abstract
The RAS gene product is normally a membrane-localized G protein (N-Ras, K-Ras and H-Ras) of 21 kDa classically described as a molecular off/on switch. It is inactive when bound to guanosine diphosphate and active when bound to GTP. When mutated, the gene produces an abnormal protein resistant to GTP hydrolysis by GTPase, resulting in a constitutively active GTP-bound protein that stimulates a critical network of signal transduction pathways that lead to cellular proliferation, survival and differentiation. At least three downstream effector pathways have been described, including Raf/MEK/ERK, PI3K/AKT and RalGDS, but they are not completely understood. Ras pathways are also important downstream effectors of several receptor tyrosine kinases localized in the cell membrane, most notably the BCR-ABL fusion protein seen in patients with Philadelphia chromosome positive chronic myelogenous leukemia. An important consideration in designing strategies to block Ras stimulatory effect is that Ras proteins are synthesized in the cytosol, but require post-translational modifications and attachment to anchor proteins or membrane binding sites in the cell membrane to be biologically active. Farnesyl transferase inhibitors (FTIs) are probably the best-studied class of Ras inhibitors in hematologic malignancies. They block the enzyme farnesyl-transferase (FTase), which is essential for post-translational modification. However, it has been observed that the Ras proteins also can be geranylgeranylated in the presence of FTIs, thus allowing membrane localization and activation, which limits their effectiveness. It is now hypothesized that their mechanism of action may be through FTase inhibition involving other signal transduction pathways. S-trans, trans-farnesylthiosalicylic acid, which was first designed as a prenylated protein methyltransferase inhibitor, has shown in vitro activity against all activated Ras proteins by dislodging them from their membrane-anchoring sites. Here, Ras biology, its signaling pathways and its implications as a therapeutic target in hematologic malignancies are reviewed.
Collapse
Affiliation(s)
- Yesid Alvarado
- University of Texas MD Anderson Cancer Center, Department of Leukemia, Box 428, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | | |
Collapse
|
40
|
Abstract
Chronic myeloid leukaemia (CML) is a clonal disorder of the haemopoietic stem cell arising as a consequence of the formation of the bcr-abl oncogene. The particular molecular basis of this condition has enabled the development of therapies that selectively target diseased cells. The success of the rationally designed first-line therapy imatinib mesylate (IM) is tempered by the problems of disease persistence and resistance. Novel strategies have been identified to take forward therapy in CML and these will be discussed in this review. This work is generated from a review of published literature and contains particular insight into the work performed by our group in this field.
Collapse
Affiliation(s)
- Nicholas B Heaney
- ATMU: Cancer Division, Section of Experimental Haematology & Haemopoietic Stem Cells, University of Glasgow, UK
| | | |
Collapse
|
41
|
Jabbour E, Cortes JE, Giles FJ, O'Brien S, Kantarjian HM. Current and emerging treatment options in chronic myeloid leukemia. Cancer 2007; 109:2171-81. [PMID: 17431887 DOI: 10.1002/cncr.22661] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Treatments for chronic myeloid leukemia (CML) represent a success story in molecular medicine. The development of imatinib, a tyrosine kinase inhibitor (TKI) targeted against the causative Bcr-Abl oncoprotein in CML, has resulted in hematologic and cytogenetic remissions in all phases of CML. A significant proportion of patients are resistant to imatinib or develop resistance during treatment. This is often a result of mutated forms of the Bcr-Abl oncoprotein to which imatinib is unable to bind. Several strategies have been developed to overcome the problem of imatinib resistance, including high-dose imatinib, novel targeted agents, and combination treatments. Novel agents include dasatinib, a potent TKI that inhibits several critical oncogenic proteins and which has recently been approved for patients with CML who are resistant or intolerant to imatinib; and nilotinib, a potent selective Bcr-Abl kinase inhibitor currently in clinical development. Other agents in development include SKI-606 and INNO-406. Stem cell transplantation remains a useful option, although it is not generally used as first-line treatment. Overall, there are an increasing number of treatment options available for patients with CML.
Collapse
Affiliation(s)
- Elias Jabbour
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
42
|
Mancini M, Brusa G, Zuffa E, Corrado P, Martinelli G, Grafone T, Barbieri E, Santucci MA. Persistent Cdk2 inactivation drives growth arrest of BCR-ABL-expressing cells in response to dual inhibitor of SRC and ABL kinases SKI606. Leuk Res 2007; 31:979-87. [PMID: 17129604 DOI: 10.1016/j.leukres.2006.09.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Revised: 07/27/2006] [Accepted: 09/27/2006] [Indexed: 12/22/2022]
Abstract
Complementary inhibition of tyrosine and SRC kinases implement dual SRC/ABL inhibitor effects in chronic myeloid leukemia (CML). Here, we show that one such inhibitor, SKI-606, induces persistent Cdk2 inactivation leading to growth arrest of BCR-ABL-expressing cells either IM-sensitive or driven to IM-resistance by other events than gene overexpression and point mutations. Inhibition of Akt serine/threonine kinase, a phosphatidylinositol 3 kinase (PI-3k) target that integrates p210 TK signaling with membrane-associated SRC kinases, is a central component of restored expression and subcellular redistribution of Cdk2 regulatory signals (p21 and p27 and Cdc25A phosphatase) in response to SKI-606. The putative roles of growth factor (namely IL-3) autocrine loop in BCR-ABL-expressing progenitor progression towards a drug-resistant phenotype are discussed.
Collapse
Affiliation(s)
- Manuela Mancini
- Istituto di Ematologia e Oncologia Medica "Lorenzo e Ariosto Seràgnoli", University of Bologna-Medical School, Via Massarenti 9, 40138-Bologna, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Raz T, Nardi V, Azam M, Cortes J, Daley GQ. Farnesyl transferase inhibitor resistance probed by target mutagenesis. Blood 2007; 110:2102-9. [PMID: 17536018 PMCID: PMC1976354 DOI: 10.1182/blood-2006-12-064907] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Mutation in the target oncoprotein is a common mechanism of resistance to tyrosine kinase inhibitors, as exemplified by the many BCR/ABL mutations that thwart imatinib activity in patients with chronic myelogenous leukemia. It remains unclear whether normal cellular protein targets of chemotherapeutics will evolve drug resistance via mutation to a similar extent. We conducted an in vitro screen for resistance to lonafarnib, a farnesyl protein transferase inhibitor that blocks prenylation of a number of proteins important in cell proliferation, and identified 9 mutations clustering around the lonafarnib binding site. In patients treated with a combination of imatinib and lonafarnib, we identified farnesyl protein transferase mutations in residues identified in our screen. Substitutions at Y361 were found in patients prior to treatment initiation, suggesting that these mutants might confer a proliferative advantage to leukemia cells, which we were able to confirm in cell culture. In vitro mutagenesis of normal cellular enzymes can be exploited to identify mutations that confer chemotherapy resistance to novel agents.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Apoptosis
- Benzamides
- Cell Proliferation/drug effects
- Drug Resistance, Neoplasm
- Enzyme Inhibitors/pharmacology
- Farnesyltranstransferase/antagonists & inhibitors
- Farnesyltranstransferase/genetics
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Mutagenesis
- Mutagenesis, Site-Directed
- Mutation/genetics
- Pilot Projects
- Piperazines/administration & dosage
- Piperidines/administration & dosage
- Protein Conformation
- Protein Prenylation
- Pyridines/administration & dosage
- Pyrimidines/administration & dosage
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Tal Raz
- Division of Hematology/Oncology, Children's Hospital Boston, and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
44
|
Jiang X, Smith C, Eaves A, Eaves C. The challenges of targeting chronic myeloid leukemia stem cells. ACTA ACUST UNITED AC 2007; 7 Suppl 2:S71-80. [PMID: 17382016 DOI: 10.3816/clm.2007.s.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Chronic myeloid leukemia (CML) is sustained by a clonally amplified population of Bcr Abl-positive pluripotent stem cells. Persistence of a large, functionally intact yet suppressed residual normal hematopoietic stem cell population in most patients with CML has made it possible to aim at the development of curative therapies. However, achieving this goal requires the identification of agents that will eradicate the leukemic stem cell population. Several potent Bcr-Abl-targeted drugs have now been introduced into clinical practice with remarkable effects. Nevertheless, accumulating data indicate that the leukemic CML stem cells in patients with chronic phase CML are less responsive to these agents than the bulk of the neoplastic cells. In this article, we review emerging evidence that CML stem cells have a number of unusual properties that underlie their relative insensitivity to treatment, including those that specifically target the Bcr-Abl oncoprotein. The biology of the neoplastic stem cells in patients with CML is clearly important to the future attainment of cures and might also prove a paradigm relevant to other types of malignancies that are sustained by transformed stem cell populations.
Collapse
MESH Headings
- ATP-Binding Cassette Transporters/analysis
- Benzamides
- Cell Differentiation
- Fusion Proteins, bcr-abl/analysis
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Genomic Instability
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Neoplastic Stem Cells/drug effects
- Philadelphia Chromosome
- Piperazines/therapeutic use
- Pyrimidines/therapeutic use
Collapse
Affiliation(s)
- Xiaoyan Jiang
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | | | | | | |
Collapse
|
45
|
Zhang B, Groffen J, Heisterkamp N. Increased resistance to a farnesyltransferase inhibitor by N-cadherin expression in Bcr/Abl-P190 lymphoblastic leukemia cells. Leukemia 2007; 21:1189-97. [PMID: 17392819 DOI: 10.1038/sj.leu.2404667] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In leukemia patients, resistance to drug treatment develops while the malignant cells can interact with and derive support from their microenvironment, such as bone marrow stroma. To model this process, lymphoblastic leukemia cells from BCR/ABL transgenic mice were treated with the farnesyltransferase inhibitor (FTI) SCH66336 while in coculture with primary mouse embryonic fibroblasts. Coculture with fibroblasts allowed the outgrowth of a subpopulation of drug-resistant lymphoblasts that expressed N-cadherin, a cell-cell adhesion protein that normally is only expressed on specific cell types, including hematopoietic stem cells and fibroblasts. N-cadherin expression promoted increased adhesion of the lymphoblasts to the fibroblasts. Importantly, de novo expression of N-cadherin in parental nonexpressing lymphoblasts using lentiviral transduction increased the ability of the cells to survive FTI treatment. We conclude that FTI drug treatment of Bcr/Abl-positive lymphoblastic leukemia cells that are in contact with a defined microenvironment induces the selective survival of a more primitive subpopulation of leukemia cells that expresses N-cadherin. Experimental drug treatment of cancer cells in model systems that include a microenvironment may reveal novel molecules that contribute to drug resistance and may aid in the design of specific therapies to eradicate more primitive cells.
Collapse
Affiliation(s)
- B Zhang
- Division of Hematology, Section of Molecular Carcinogenesis, Saban Research Institute, Childrens Hospital Los Angeles and the Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | | | | |
Collapse
|
46
|
Jørgensen HG, Allan EK, Jordanides NE, Mountford JC, Holyoake TL. Nilotinib exerts equipotent antiproliferative effects to imatinib and does not induce apoptosis in CD34+ CML cells. Blood 2007; 109:4016-9. [PMID: 17213283 DOI: 10.1182/blood-2006-11-057521] [Citation(s) in RCA: 237] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chronic myeloid leukemia (CML) stem and progenitor cells overexpress BcrAbl and are insensitive to imatinib mesylate (IM). We therefore investigated whether these cells were efficiently targeted by nilotinib. In K562, the inhibitory concentration (IC50) of nilotinib was 30 nM versus 600 nM for IM, consistent with its reported 20-fold-higher potency. However, in primary CD34(+) CML cells, nilotinib and IM were equipotent for inhibition of BcrAbl activity, producing equivalent but incomplete reduction in CrkL phosphorylation at 5 microM. CML CD34(+) cells were still able to expand over 72 hours with 5 microM of either drug, although there was a concentration-dependent restriction of amplification. As for IM, the most primitive cells (CFSE(max)) persisted and accumulated over 72 hours with nilotinib and remained caspase-3 negative. Furthermore, nilotinib with IM led to further accumulation of this population, suggesting at least additive antiproliferative effects. These results confirmed that, like IM, the predominant effect of nilotinib is antiproliferative rather than proapoptotic.
Collapse
MESH Headings
- Antigens, CD34
- Apoptosis/drug effects
- Benzamides
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm/drug effects
- Drug Synergism
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/metabolism
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Neoplastic Stem Cells/enzymology
- Piperazines/agonists
- Piperazines/pharmacology
- Protein Kinase Inhibitors/agonists
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines/agonists
- Pyrimidines/pharmacology
- Time Factors
- Tumor Stem Cell Assay
Collapse
Affiliation(s)
- Heather G Jørgensen
- Section of Experimental Haematology, Division of Cancer Sciences and Molecular Pathology, University of Glasgow, 10 Alexandra Parade, Glasgow, UK
| | | | | | | | | |
Collapse
|
47
|
Cortes J, Jabbour E, Daley GQ, O'Brien S, Verstovsek S, Ferrajoli A, Koller C, Zhu Y, Statkevich P, Kantarjian H. Phase 1 study of lonafarnib (SCH 66336) and imatinib mesylate in patients with chronic myeloid leukemia who have failed prior single-agent therapy with imatinib. Cancer 2007; 110:1295-302. [PMID: 17623836 DOI: 10.1002/cncr.22901] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Lonafarnib is an orally bioavailable nonpetidomimetic farnesyl transferase inhibitor with significant activity against BCR-ABL-positive cell lines and primary human chronic myeloid leukemia (CML) cells. Lonafarnib can inhibit the proliferation of imatinib-resistant cells and increases imatinib-induced apoptosis in vitro in cells from imatinib-resistant patients. METHODS The authors conducted a phase 1 study of lonafarnib in combination with imatinib in patients with CML who failed imatinib therapy. The starting dose level for patients with chronic phase (CP) disease was imatinib, 400 mg/day, plus lonafarnib at a dose of 100 mg twice daily. The starting dose levels for accelerated phase (AP) and blast phase (BP) disease were 600 mg/day and 100 mg twice daily, respectively. RESULTS A total of 23 patients were treated (9 with CP, 11 with AP, and 3 with BP) for a median of 25 weeks (range, 4-102 weeks). Of those with CP disease, 2 patients had grade 3 (according to the National Cancer Institute Common Toxicity Criteria [version 2.0]) dose-limiting toxicities (DLTs) at the 400 + 125-mg dose, including diarrhea (2 patients), vomiting (1 patient), and fatigue (1 patient). In patients with AP/BP disease, DLTs were observed at the 600 + 125-mg dose and was comprised of diarrhea (1 patient) and hypokalemia (1 patient). Eight patients (35%) responded; 3 with CP disease achieved a complete hematologic response (CHR) (2 patients) and a complete cytogenetic response (1 patient). Three patients with AP disease responded (2 CHR, 1 partial cytogenetic response), and 2 patients with BP disease demonstrated hematologic improvement. Pharmacokinetics data suggest no apparent increase in exposure or changes in the pharmacokinetics of either lonafarnib or imatinib when they are coadministered. CONCLUSIONS The results of the current study indicate that the combination of lonafarnib and imatinib is well tolerated and the maximum tolerated dose of lonafarnib is 100 mg twice daily when combined with imatinib at a dose of either 400 mg or 600 mg daily.
Collapse
Affiliation(s)
- Jorge Cortes
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Cortes J, Quintás-Cardama A, Garcia-Manero G, O'Brien S, Jones D, Faderl S, Ebarb T, Giles F, Thomas D, Kantarjian H. Phase 1 study of tipifarnib in combination with imatinib for patients with chronic myelogenous leukemia in chronic phase after imatinib failure. Cancer 2007; 110:2000-6. [PMID: 17849425 DOI: 10.1002/cncr.23006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The tolerability and efficacy of the combination of tipifarnib, an orally bioavailable nonpeptidomimetic farnesyl transferase inhibitor, and imatinib was investigated in patients with chronic myelogenous leukemia in chronic phase who had failed imatinib. METHODS Twenty-six patients (13 [50%] with Abl kinase domain mutations) were treated. The initial dose level was tipifarnib at a dose of 300 mg twice daily and imatinib at a dose of 300 mg daily. Therapy was escalated following a '3 + 3' phase 1 design and the maximum tolerated dose was defined as tipifarnib at a dose of 400 mg twice daily and imatinib at a dose of 400 mg daily. Therapy was administered for a median of 26 weeks (range, 3-150 weeks). RESULTS Adverse events included diarrhea in 21 patients (81%) and nausea in 18 patients (69%), but were generally grade 2 or less (using the revised National Cancer Institute Common Toxicity Criteria). Grade 3-4 neutropenia and thrombocytopenia occurred in 11 patients (42%) and 8 patients (31%), respectively. Sixteen patients discontinued therapy (5 due to toxicity and 11 due to lack of response or disease progression). Hematologic responses were attained by 17 (68%) of 25 assessable patients. Nine patients (36%) also achieved a cytogenetic response (3 complete responses, 4 partial responses, and 2 minimal responses), including 4 patients harboring mutant Bcr-Abl tyrosine kinases. One patient bearing the highly imatinib-resistant T315I mutant achieved a partial cytogenetic response. The median response duration was 3 months (range, 2-30+ months). CONCLUSIONS The combination of tipifarnib and imatinib is well tolerated and has activity against several Abl kinase domain mutants. Combinations of tipifarnib with more potent tyrosine kinase inhibitors warrant further investigation.
Collapse
Affiliation(s)
- Jorge Cortes
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The advent of the Bcr-Abl selective tyrosine kinase inhibitor imatinib mesylate (Glivec, Gleevec, Novartis, East Hanover, NJ) has substantially changed the treatment landscape for chronic myelogenous leukemia (CML). However, some patients, primarily those with advanced disease, are either initially refractory to imatinib or eventually develop imatinib resistance. Imatinib resistance or intolerance frequently depends on the re-emergence of Bcr-Abl kinase activity, but can also indicate Bcr-Abl-independent disease progression. Results from phase II/III trials suggest rates of resistance and relapse correlate with stage of disease and with the monitoring parameters: hematologic, cytogenetic, and molecular responses. To date, more than 40 different point mutations that code for distinct single amino acid substitutions in the Bcr-Abl kinase domain have been isolated from imatinib-resistant patients. These mutations affect amino acids involved in imatinib binding or in regulatory regions of the Bcr-Abl kinase domain, resulting in decreased sensitivity to imatinib while retaining aberrant kinase activity. Early mutation detection may aid in risk stratification and molecular-based treatment decisions. To overcome imatinib-resistant disease, novel tyrosine kinase inhibitors with activity against imatinib-resistant mutations and/or with inhibition of alternative pathways, such as Src activation, have recently been developed. Additional strategies include imatinib dose escalation, combination therapy, and treatment interruption to stop clonal selection of resistant cells.
Collapse
MESH Headings
- Benzamides
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/physiology
- Fusion Proteins, bcr-abl/drug effects
- Fusion Proteins, bcr-abl/genetics
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology
- Mutation/drug effects
- Piperazines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines/pharmacology
- Randomized Controlled Trials as Topic
Collapse
Affiliation(s)
- Andreas Hochhaus
- III. Medizinische Klinik, Medizinische Fakultät Mannheim, University of Heidelberg, Mannheim, Germany.
| | | | | | | |
Collapse
|
50
|
Melo JV, Chuah C. Resistance to imatinib mesylate in chronic myeloid leukaemia. Cancer Lett 2006; 249:121-32. [PMID: 16949736 DOI: 10.1016/j.canlet.2006.07.010] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2006] [Revised: 07/27/2006] [Accepted: 07/28/2006] [Indexed: 02/04/2023]
Abstract
Despite the remarkable results achieved with imatinib for the treatment of chronic myeloid leukaemia, the emergence of resistance to this tyrosine kinase inhibitor has become a significant problem. Much progress has been recently made in elucidating the mechanisms which underlie imatinib resistance. The most common cause of such drug resistance is the selection of leukaemic clones with point mutations in the Abl kinase domain leading to amino acid substitutions which prevent the appropriate binding of the drug. Other mechanisms include genomic amplification of BCR-ABL and modulation of drug efflux or influx transporters. There is a pressing need, therefore, to develop and test novel drugs and strategies. Two such compounds are now being explored in clinical trials. This review will describe the molecular basis of imatinib-resistance and strategies to overcome resistance.
Collapse
Affiliation(s)
- Junia V Melo
- Department of Haematology, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom.
| | | |
Collapse
|