1
|
Cruz-Rodriguez N, Tang H, Bateman B, Tang W, Deininger M. BCR::ABL1 Proteolysis-targeting chimeras (PROTACs): The new frontier in the treatment of Ph + leukemias? Leukemia 2024; 38:1885-1893. [PMID: 39098922 DOI: 10.1038/s41375-024-02365-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024]
Abstract
BCR::ABL1 tyrosine kinase inhibitors (TKIs) have turned chronic myeloid leukemia (CML) from a lethal condition into a chronic ailment. With optimal management, the survival of CML patients diagnosed in the chronic phase is approaching that of age-matched controls. However, only one-third of patients can discontinue TKIs and enter a state of functional cure termed treatment-free remission (TFR), while the remainder require life-long TKI therapy to avoid the recurrence of active leukemia. Approximately 10% of patients exhibit primary or acquired TKI resistance and eventually progress to the blast phase. It is thought that recurrence after attempted TFR originates from CML stem cells (LSCs) surviving despite continued suppression of BCR::ABL1 kinase. Although kinase activity is indispensable for induction of overt CML, kinase-independent scaffold functions of BCR::ABL1 are known to contribute to leukemogenesis, raising the intriguing but as yet hypothetical possibility, that degradation of BCR::ABL1 protein may accomplish what TKIs fail to achieve - eliminate residual LSCs to turn functional into real cures. The advent of BCR::ABL1 proteolysis targeting chimeras (PROTACs), heterobifunctional molecules linking a TKI-based warhead to an E3 ligase recruiter, has moved clinical protein degradation into the realm of the possible. Here we examine the molecular rationale as well as pros and cons of degrading BCR::ABL1 protein. We review reported BCR::ABL1 PROTACs, point out limitations of available data and compounds and suggest directions for future research. Ultimately, clinical testing of a potent and specific BCR::ABL1 degrader will be required to determine the efficacy and tolerability of this approach.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/metabolism
- Proteolysis
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Drug Resistance, Neoplasm
- Proto-Oncogene Proteins c-abl/metabolism
- Animals
- Proteolysis Targeting Chimera
Collapse
Affiliation(s)
| | - Hua Tang
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Weiping Tang
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael Deininger
- Versiti Blood Research Institute, Milwaukee, WI, USA.
- Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
2
|
Torres-Barrera P, Moreno-Lorenzana D, Alvarado-Moreno JA, García-Ruiz E, Lagunas C, Mayani H, Chávez-González A. Cell Contact with Endothelial Cells Favors the In Vitro Maintenance of Human Chronic Myeloid Leukemia Stem and Progenitor Cells. Int J Mol Sci 2022; 23:ijms231810326. [PMID: 36142235 PMCID: PMC9499491 DOI: 10.3390/ijms231810326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/30/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic Myeloid Leukemia (CML) originates in a leukemic stem cell that resides in the bone marrow microenvironment, where they coexist with cellular and non-cellular elements. The vascular microenvironment has been identified as an important element in CML development since an increase in the vascularization has been suggested to be related with poor prognosis; also, using murine models, it has been reported that bone marrow endothelium can regulate the quiescence and proliferation of leukemic stem and progenitor cells. This observation, however, has not been evaluated in primary human cells. In this report, we used a co-culture of primitive (progenitor and stem) CML cells with endothelial colony forming cells (ECFC) as an in vitro model to evaluate the effects of the vascular microenvironment in the leukemic hematopoiesis. Our results show that this interaction allows the in vitro maintenance of primitive CML cells through an inflammatory microenvironment able to regulate the proliferation of progenitor cells and the permanence in a quiescent state of leukemic stem cells.
Collapse
Affiliation(s)
- Patricia Torres-Barrera
- Laboratorio de Células Troncales Leucémicas, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN Siglo XXI, Instituto Mexicano del Seguro Social, CDMX 06725, Mexico
- Posgrado en Ciencias Biológicas, UNAM, CDMX 04510, Mexico
| | | | - José Antonio Alvarado-Moreno
- Unidad de Investigación Médica en Trombosis Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, CDMX 03100, Mexico
| | - Elena García-Ruiz
- Departamento de Hematología, Hospital de Especialidades, CMN La Raza, Instituto Mexicano del Seguro Social, CDMX 02990, Mexico
| | - Cesar Lagunas
- Departamento de Cirugías de Cadera, Hospital General “Villa Coapa” Instituto Mexicano del Seguro Social, CDMX 14310, Mexico
| | - Hector Mayani
- Laboratorio de Células Troncales Hematopoyéticas, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN Siglo XXI, Instituto Mexicano del Seguro Social, CDMX 06725, Mexico
| | - Antonieta Chávez-González
- Laboratorio de Células Troncales Leucémicas, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN Siglo XXI, Instituto Mexicano del Seguro Social, CDMX 06725, Mexico
- Correspondence:
| |
Collapse
|
3
|
Hodkinson KE, Bouwer N, Vaughan J. South African study of blast phase chronic myeloid leukaemia: A poor prognostic outlook. Afr J Lab Med 2022; 11:1578. [PMID: 35747555 PMCID: PMC9210180 DOI: 10.4102/ajlm.v11i1.1578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 02/22/2022] [Indexed: 11/29/2022] Open
Abstract
Background Chronic myeloid leukaemia (CML) is a haematological malignancy characterised by the translocation t(9;22)(q34;q11.2), resulting in a constitutively active tyrosine kinase. Globally, overall survival of blast crisis phase (BC) CML is one year. Newer tyrosine kinase inhibitors and allogeneic stem cell transplantation offer remission; however, refractory and relapsed disease remain the biggest challenges. Objective In South Africa, literature is lacking on BC-CML. This study aimed to determine the disease characteristics and overall survival in South Africa. Methods This retrospective, laboratory-based study reviewed all new BC-CML diagnoses via flow cytometry at Charlotte Maxeke Johannesburg Academic Hospital in Johannesburg, South Africa, between April 2016 and October 2019. BC-CML was defined as the presence of > 20% blasts with a CML history or the BCR-ABL1 fusion gene (p210/p190) in the appropriate clinical or pathological context. Survival outcomes were inferred from clinical and laboratory data. Results Twenty-two new cases of BC-CML were diagnosed (median age: 34 years). There were 20 (91%) cases with the fusion transcripts p210 and two (9%) cases with p190 BCRABL1. For blast lineage, 14 cases were myeloid (63.6%), six were lymphoid (27.3%), and two were ambiguous (9.1%). There was a 72.7% mortality (16 cases); sepsis, refractory and relapsed disease were the major causes. Patients who achieved remission had lower blast percentages, simple karyotypes, and a trend towards higher white cell and platelet counts at presentation. Conclusion Optimised management of early-stage CML, prevention and aggressive management of sepsis, with advocation for newer therapies are needed to improve the overall survival of BC-CML in South Africa.
Collapse
Affiliation(s)
- Katherine E Hodkinson
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Institution of National Health Laboratory Service, Johannesburg, South Africa
| | - Nikki Bouwer
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Institution of National Health Laboratory Service, Johannesburg, South Africa
| | - Jenifer Vaughan
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Institution of National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
4
|
Krenn PW, Montanez E, Costell M, Fässler R. Integrins, anchors and signal transducers of hematopoietic stem cells during development and in adulthood. Curr Top Dev Biol 2022; 149:203-261. [PMID: 35606057 DOI: 10.1016/bs.ctdb.2022.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hematopoietic stem cells (HSCs), the apex of the hierarchically organized blood cell production system, are generated in the yolk sac, aorta-gonad-mesonephros region and placenta of the developing embryo. To maintain life-long hematopoiesis, HSCs emigrate from their site of origin and seed in distinct microenvironments, called niches, of fetal liver and bone marrow where they receive supportive signals for self-renewal, expansion and production of hematopoietic progenitor cells (HPCs), which in turn orchestrate the production of the hematopoietic effector cells. The interactions of hematopoietic stem and progenitor cells (HSPCs) with niche components are to a large part mediated by the integrin superfamily of adhesion molecules. Here, we summarize the current knowledge regarding the functional properties of integrins and their activators, Talin-1 and Kindlin-3, for HSPC generation, function and fate decisions during development and in adulthood. In addition, we discuss integrin-mediated mechanosensing for HSC-niche interactions, ex vivo protocols aimed at expanding HSCs for therapeutic use, and recent approaches targeting the integrin-mediated adhesion in leukemia-inducing HSCs in their protecting, malignant niches.
Collapse
Affiliation(s)
- Peter W Krenn
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany; Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris-Lodron University of Salzburg, Salzburg, Austria.
| | - Eloi Montanez
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona and Bellvitge Biomedical Research Institute, L'Hospitalet del Llobregat, Barcelona, Spain
| | - Mercedes Costell
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, Burjassot, Spain; Institut Universitari de Biotecnologia i Biomedicina, Universitat de València, Burjassot, Spain
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
5
|
C-C Chemokine Receptor 7 in Cancer. Cells 2022; 11:cells11040656. [PMID: 35203305 PMCID: PMC8870371 DOI: 10.3390/cells11040656] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/01/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
C-C chemokine receptor 7 (CCR7) was one of the first two chemokine receptors that were found to be upregulated in breast cancers. Chemokine receptors promote chemotaxis of cells and tissue organization. Since under homeostatic conditions, CCR7 promotes migration of immune cells to lymph nodes, questions immediately arose regarding the ability of CCR7 to direct migration of cancer cells to lymph nodes. The literature since 2000 was examined to determine to what extent the expression of CCR7 in malignant tumors promoted migration to the lymph nodes. The data indicated that in different cancers, CCR7 plays distinct roles in directing cells to lymph nodes, the skin or to the central nervous system. In certain tumors, it may even serve a protective role. Future studies should focus on defining mechanisms that differentially regulate the unfavorable or beneficial role that CCR7 plays in cancer pathophysiology, to be able to improve outcomes in patients who harbor CCR7-positive cancers.
Collapse
|
6
|
Gurianov DS, Antonenko SV, Telegeev GD. Colocalization of BCR Protein with Clathrin, Actin, and Cortactin Suggests Its Possible Role in the Regulation of Actin Branching and Clathrin-Mediated Endocytosis. CYTOL GENET+ 2021. [DOI: 10.3103/s0095452721020055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
7
|
Future Approaches for Treating Chronic Myeloid Leukemia: CRISPR Therapy. BIOLOGY 2021; 10:biology10020118. [PMID: 33557401 PMCID: PMC7915349 DOI: 10.3390/biology10020118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022]
Abstract
Simple Summary In the last two decades, the therapeutic landscape of several tumors have changed profoundly with the introduction of drugs against proteins encoded by oncogenes. Oncogenes play an essential role in human cancer and when their encoded proteins are inhibited by specific drugs, the tumoral process can be reverted or stopped. An example of this is the case of the chronic myeloid leukemia, in which all the pathological features can be attributed by a single oncogene. Most patients with this disease now have a normal life expectancy thanks to a rationality designed inhibitor. However, the drug only blocks the protein, the oncogene continues unaffected and treatment discontinuation is only an option for a small subset of patients. With the advent of genome-editing nucleases and, especially, the CRISPR/Cas9 system, the possibilities to destroy oncogenes now is feasible. A novel therapeutic tool has been developed with unimaginable limits in cancer treatment. Recent studies support that CRISPR/Cas9 system could be a definitive therapeutic option in chronic myeloid leukemia. This work reviews the biology of chronic myeloid leukemia, the emergence of the CRISPR system, and its ability as a specific tool for this disease. Abstract The constitutively active tyrosine-kinase BCR/ABL1 oncogene plays a key role in human chronic myeloid leukemia development and disease maintenance, and determines most of the features of this leukemia. For this reason, tyrosine-kinase inhibitors are the first-line treatment, offering most patients a life expectancy like that of an equivalent healthy person. However, since the oncogene stays intact, lifelong oral medication is essential, even though this triggers adverse effects in many patients. Furthermore, leukemic stem cells remain quiescent and resistance is observed in approximately 25% of patients. Thus, new therapeutic alternatives are still needed. In this scenario, the interruption/deletion of the oncogenic sequence might be an effective therapeutic option. The emergence of CRISPR (clustered regularly interspaced short palindromic repeats) technology can offer a definitive treatment based on its capacity to induce a specific DNA double strand break. Besides, it has the advantage of providing complete and permanent oncogene knockout, while tyrosine kinase inhibitors (TKIs) only ensure that BCR-ABL1 oncoprotein is inactivated during treatment. CRISPR/Cas9 cuts DNA in a sequence-specific manner making it possible to turn oncogenes off in a way that was not previously feasible in humans. This review describes chronic myeloid leukemia (CML) disease and the main advances in the genome-editing field by which it may be treated in the future.
Collapse
|
8
|
Minciacchi VR, Kumar R, Krause DS. Chronic Myeloid Leukemia: A Model Disease of the Past, Present and Future. Cells 2021; 10:cells10010117. [PMID: 33435150 PMCID: PMC7827482 DOI: 10.3390/cells10010117] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic myeloid leukemia (CML) has been a "model disease" with a long history. Beginning with the first discovery of leukemia and the description of the Philadelphia Chromosome and ending with the current goal of achieving treatment-free remission after targeted therapies, we describe here the journey of CML, focusing on molecular pathways relating to signaling, metabolism and the bone marrow microenvironment. We highlight current strategies for combination therapies aimed at eradicating the CML stem cell; hopefully the final destination of this long voyage.
Collapse
MESH Headings
- Epigenesis, Genetic
- History, 20th Century
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/history
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Models, Biological
- Molecular Targeted Therapy
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Tumor Microenvironment/genetics
Collapse
Affiliation(s)
- Valentina R. Minciacchi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany; (V.R.M.); (R.K.)
| | - Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany; (V.R.M.); (R.K.)
| | - Daniela S. Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany; (V.R.M.); (R.K.)
- German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
- Frankfurt Cancer Institute, 60596 Frankfurt, Germany
- Faculty of Medicine, Medical Clinic II, Johann Wolfgang Goethe University, 60596 Frankfurt, Germany
- Correspondence: ; Tel.: +49-69-63395-500; Fax: +49-69-63395-519
| |
Collapse
|
9
|
Omsland M, Andresen V, Gullaksen SE, Ayuda-Durán P, Popa M, Hovland R, Brendehaug A, Enserink J, McCormack E, Gjertsen BT. Tyrosine kinase inhibitors and interferon-α increase tunneling nanotube (TNT) formation and cell adhesion in chronic myeloid leukemia (CML) cell lines. FASEB J 2020; 34:3773-3791. [PMID: 31945226 PMCID: PMC10894852 DOI: 10.1096/fj.201802061rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/21/2022]
Abstract
Chronic myeloid leukemia (CML) is a stem cell disease of the bone marrow where mechanisms of inter-leukemic communication and cell-to-cell interactions are proposed to be important for optimal therapy response. Tunneling nanotubes (TNTs) are novel intercellular communication structures transporting different cargos with potential implications in therapy resistance. Here, we have investigated TNTs in CML cells and following treatment with the highly effective CML therapeutics tyrosine kinase inhibitors (TKIs) and interferon-α (IFNα). CML cells from chronic phase CML patients as well as the blast crisis phase cell lines, Kcl-22 and K562, formed few or no TNTs. Treatment with imatinib increased TNT formation in both Kcl-22 and K562 cells, while nilotinib or IFNα increased TNTs in Kcl-22 cells only where the TNT increase was associated with adherence to fibronectin-coated surfaces, altered morphology, and reduced movement involving β1integrin. Ex vivo treated cells from chronic phase CML patients showed limited changes in TNT formation similarly to bone marrow cells from healthy individuals. Interestingly, in vivo nilotinib treatment in a Kcl-22 subcutaneous mouse model resulted in morphological changes and TNT-like structures in the tumor-derived Kcl-22 cells. Our results demonstrate that CML cells express low levels of TNTs, but CML therapeutics increase TNT formation in designated cell models indicating TNT functionality in bone marrow derived malignancies and their microenvironment.
Collapse
MESH Headings
- Animals
- Cell Adhesion/drug effects
- Cell Communication/drug effects
- Cell Line, Tumor
- Cells, Cultured
- Female
- Fluorescent Antibody Technique
- Humans
- Immunoblotting
- Integrin beta1/metabolism
- Interferon-alpha/therapeutic use
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Mice
- Microscopy, Electron, Scanning
- Protein Kinase Inhibitors/therapeutic use
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Maria Omsland
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vibeke Andresen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Stein-Erik Gullaksen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Pilar Ayuda-Durán
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Mihaela Popa
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
- KinN Therapeutics, Bergen, Norway
| | - Randi Hovland
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Atle Brendehaug
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Jorrit Enserink
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Emmet McCormack
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
10
|
Wu J, Wang A, Li X, Chen C, Qi Z, Hu C, Wang W, Wu H, Huang T, Zhao M, Wang W, Hu Z, Liu Q, Wang B, Wang L, Li L, Ge J, Ren T, Xia R, Liu J, Liu Q. Discovery and characterization of a novel highly potent and selective type II native and drug-resistant V299L mutant BCR-ABL inhibitor (CHMFL-ABL-039) for Chronic Myeloid Leukemia (CML). Cancer Biol Ther 2019; 20:877-885. [PMID: 30894066 DOI: 10.1080/15384047.2019.1579958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BCR fused ABL kinase is the critical driving oncogene for chronic myeloid leukemia (CML) and has been extensively studied as the drug discovery target in the past decade. The successful introduction of tyrosine kinase inhibitors (TKI) such as Imatinib, Dasatinib and Bosutinib has greatly improved the CML patient survival rate. However, upon the chronic treatment, a variety of TKI resistant mutants, such as the V299L mutant which has been found in more and more patients with the high-throughput sequencing technology, are observed, although the incidence is still considered rare compared to the more prevalent gatekeeper T315I mutant. However, with the progress of the precision medicine concept, the rare mutation (or the orphan drug target) has attracted more and more attention. Here we report a novel type II BCR-ABL kinase inhibitor, CHMFL-ABL-039, which not only displayed great potency (IC50: 7.9 nM) and selectivity (S score (1) = 0.02) against native ABL kinase among other kinases in the kinome, but also exhibited great potency (IC50: 27.9 nM) and selectivity against Imatinib-resistant V299L mutant among other frequently observed ABL kinase mutants. CHMFL-ABL-039 has demonstrated greater efficacies than Imatinib regarding to the anti-proliferation, inhibition of the signaling pathway, arrest of cell cycle progression, induction of apoptosis in vitro and suppression of the tumor progression in vivo in the native and V299L mutated BCR-ABL kinase-driven cells/xenograft models. It would be a useful pharmacological tool to study the TKI resistant ABL V299L mutant-mediated pathology and provide a potential precise treatment approach for this orphan CML subtype in the precision medicine era.
Collapse
Affiliation(s)
- Jiaxin Wu
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China.,b University of Science and Technology of China , Hefei , Anhui , P. R. China
| | - Aoli Wang
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China
| | - Xixiang Li
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China.,c CHMFL-HCMTC Target Therapy Joint Laboratory , Hefei , Anhui , P. R. China
| | - Cheng Chen
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China.,b University of Science and Technology of China , Hefei , Anhui , P. R. China
| | - Ziping Qi
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China.,c CHMFL-HCMTC Target Therapy Joint Laboratory , Hefei , Anhui , P. R. China
| | - Chen Hu
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China.,b University of Science and Technology of China , Hefei , Anhui , P. R. China
| | - Wenliang Wang
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China.,b University of Science and Technology of China , Hefei , Anhui , P. R. China
| | - Hong Wu
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China
| | - Tao Huang
- d Precision Targeted Therapy Discovery Center, Institute of Technology Innovation , Hefei Institutes of Physical Science, Chinese Academy of Sciences , Hefei , Anhui , P. R. China
| | - Ming Zhao
- d Precision Targeted Therapy Discovery Center, Institute of Technology Innovation , Hefei Institutes of Physical Science, Chinese Academy of Sciences , Hefei , Anhui , P. R. China
| | - Wenchao Wang
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China.,c CHMFL-HCMTC Target Therapy Joint Laboratory , Hefei , Anhui , P. R. China
| | - Zhenquan Hu
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China
| | - Qingwang Liu
- d Precision Targeted Therapy Discovery Center, Institute of Technology Innovation , Hefei Institutes of Physical Science, Chinese Academy of Sciences , Hefei , Anhui , P. R. China
| | - Beilei Wang
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China.,b University of Science and Technology of China , Hefei , Anhui , P. R. China
| | - Li Wang
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China.,b University of Science and Technology of China , Hefei , Anhui , P. R. China
| | - Lili Li
- e Department of Hematology , the First Hospital of Anhui Medical University , Hefei , Anhui , P.R. China
| | - Jian Ge
- e Department of Hematology , the First Hospital of Anhui Medical University , Hefei , Anhui , P.R. China
| | - Tao Ren
- d Precision Targeted Therapy Discovery Center, Institute of Technology Innovation , Hefei Institutes of Physical Science, Chinese Academy of Sciences , Hefei , Anhui , P. R. China
| | - Ruixiang Xia
- e Department of Hematology , the First Hospital of Anhui Medical University , Hefei , Anhui , P.R. China
| | - Jing Liu
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China.,c CHMFL-HCMTC Target Therapy Joint Laboratory , Hefei , Anhui , P. R. China
| | - Qingsong Liu
- a High Magnetic Field Laboratory , Chinese Academy of Sciences , Hefei , Anhui , P. R. China.,b University of Science and Technology of China , Hefei , Anhui , P. R. China.,c CHMFL-HCMTC Target Therapy Joint Laboratory , Hefei , Anhui , P. R. China.,d Precision Targeted Therapy Discovery Center, Institute of Technology Innovation , Hefei Institutes of Physical Science, Chinese Academy of Sciences , Hefei , Anhui , P. R. China
| |
Collapse
|
11
|
Carlo-Stella C, Mangoni L, Rizzoli V. Biology and Clinical Applications of Longterm Bone Marrow Cultures. Int J Artif Organs 2018. [DOI: 10.1177/039139889301605s14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A number of clonogenic assays for short-term bone marrow culture is now available for the quantitative analysis of the various hematopoietic progenitor cell classes. The short-term assays are not suitable to analyse either stem cell selfrenewal or interactions of hematopoietic progenitors with stromal cells, especially those requiring direct cell-to-cell or cell-to-matrix contact. The technique of longterm bone marrow culture (LTBMC) allows a sustained production of myeloid cells when marrow is placed in liquid culture at relatively high cell concentration, with appropriate supplements, temperature and feeding conditions. A peculiar feature of LTBMC is that the stromal cells promote selfrenewal as well as differentiation of the stem cells, without the need to add exogenous growth factors. The LTBMC system offers an approach able to investigate not only the proliferative and differentiative events but also sustained cell production and selfrenewal of any clonogenic cell types. In the last years, the technique of LTBMC has been increasingly used by several groups to investigate hematopoietic regulation, stromal cell function and the interactions among stromal and hematopoietic cells. In the present report, the biology of LTBMC and their possible clinical applications will be reviewed.
Collapse
Affiliation(s)
- C. Carlo-Stella
- Department of Hematology, Bone Marrow Transplantation Unit, University of Parma, Parma - Italy
| | - L. Mangoni
- Department of Hematology, Bone Marrow Transplantation Unit, University of Parma, Parma - Italy
| | - V. Rizzoli
- Department of Hematology, Bone Marrow Transplantation Unit, University of Parma, Parma - Italy
| |
Collapse
|
12
|
Cornillie SP, Bruno BJ, Lim CS, Cheatham TE. Computational Modeling of Stapled Peptides toward a Treatment Strategy for CML and Broader Implications in the Design of Lengthy Peptide Therapeutics. J Phys Chem B 2018. [DOI: 10.1021/acs.jpcb.8b01014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
13
|
Shah M, Bhatia R. Preservation of Quiescent Chronic Myelogenous Leukemia Stem Cells by the Bone Marrow Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1100:97-110. [DOI: 10.1007/978-3-319-97746-1_6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
14
|
Inhibition of SDF-1-induced migration of oncogene-driven myeloid leukemia by the L-RNA aptamer (Spiegelmer), NOX-A12, and potentiation of tyrosine kinase inhibition. Oncotarget 2017; 8:109973-109984. [PMID: 29299123 PMCID: PMC5746358 DOI: 10.18632/oncotarget.22409] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 10/25/2017] [Indexed: 01/06/2023] Open
Abstract
Resistance to targeted tyrosine kinase inhibitors (TKI) remains a challenge for the treatment of myeloid leukemias. Following treatment with TKIs, the bone marrow microenvironment has been found to harbor a small pool of surviving leukemic CD34+ progenitor cells. The long-term survival of these leukemic cells has been attributed, at least in part, to the protective effects of bone marrow stroma. We found that the NOX-A12 'Spiegelmer', an L-enantiomeric RNA oligonucleotide that inhibits SDF-1α, showed in vitro and in vivo activity against BCR-ABL- and FLT3-ITD-dependent leukemia cells. NOX-A12 was sufficient to suppress SDF-1-induced migration in vitro. The combination of NOX-A12 with TKIs reduced cell migration in the same in vitro model of SDF-1-induced chemotaxis to a greater extent than either drug alone, suggesting positive cooperativity as a result of the SDF-1 blocking function of NOX-A12 and cytotoxicity resulting from targeted oncogenic kinase inhibition. These results are consistent with our in vivo findings using a functional pre-clinical mouse model of chronic myeloid leukemia (CML), whereby we demonstrated the ability of NOX-A12, combined with the ABL kinase inhibitor, nilotinib, to reduce the leukemia burden in mice to a greater extent than either agent alone. Overall, the data support the idea of using SDF-1 inhibition in combination with targeted kinase inhibition to override drug resistance in oncogene-driven leukemia to significantly diminish or eradicate residual leukemic disease.
Collapse
|
15
|
Aggoune D, Sorel N, Bonnet ML, Goujon JM, Tarte K, Hérault O, Domenech J, Réa D, Legros L, Johnson-Ansa H, Rousselot P, Cayssials E, Guerci-Bresler A, Bennaceur-Griscelli A, Chomel JC, Turhan AG. Bone marrow mesenchymal stromal cell (MSC) gene profiling in chronic myeloid leukemia (CML) patients at diagnosis and in deep molecular response induced by tyrosine kinase inhibitors (TKIs). Leuk Res 2017; 60:94-102. [PMID: 28772207 DOI: 10.1016/j.leukres.2017.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/01/2017] [Accepted: 07/25/2017] [Indexed: 01/05/2023]
Abstract
Although it has been well-demonstrated that bone marrow mesenchymal stromal cells (MSCs) from CML patients do not belong to the Ph1-positive clone, there is growing evidence that they could play a role in the leukemogenesis process or the protection of leukemic stem cells from the effects of tyrosine kinase inhibitors (TKIs). The aim of the present study was to identify genes differentially expressed in MSCs isolated from CML patients at diagnosis (CML-MSCs) as compared to MSCs from healthy controls. Using a custom gene-profiling assay, we identified six genes over-expressed in CML-MSCs (BMP1, FOXO3, MET, MITF, NANOG, PDPN), with the two highest levels being documented for PDPN (PODOPLANIN) and NANOG. To determine whether this aberrant signature persisted in patients in deep molecular response induced by TKIs, we analyzed MSCs derived from such patients (MR-MSCs). This analysis showed that, despite the deep molecular responses, BMP1, MET, MITF, NANOG, and PDPN mRNA were upregulated in MR-MSCs. Moreover, BMP1, MITF, and NANOG mRNA expressions in MR-MSCs were found to be intermediate between control MSCs and CML-MSCs. These results suggest that CML-MSCs exhibit an abnormal gene expression pattern which might have been established during the leukemogenic process and persist in patients in deep molecular response.
Collapse
Affiliation(s)
| | - Nathalie Sorel
- INSERM, U935, F-86000 Poitiers, France; CHU de Poitiers, Service de Cancérologie Biologique, F-86021 Poitiers, France
| | | | - Jean-Michel Goujon
- CHU de Poitiers, Service d'Anatomie et cytologie pathologiques, F-86021 Poitiers, France; INSERM, U1082, F-86021 Poitiers, France
| | | | - Olivier Hérault
- CHU de Tours, Service d'Hématologie Biologique, F-37032 Tours, France; CNRS UMR 7292, équipe LNOx, Université François Rabelais, F-37032 Tours, France
| | - Jorge Domenech
- CHU de Tours, Service d'Hématologie Biologique, F-37032 Tours, France; CNRS UMR 7292, équipe LNOx, Université François Rabelais, F-37032 Tours, France
| | - Delphine Réa
- Hôpital Saint Louis, Service d'Hématologie Adulte, F-75000 Paris, France; INSERM, UMRS-1160, IUH-Université Paris Diderot-Paris 7, F-75000 Paris, France
| | - Laurence Legros
- Hôpital l'Archet, Service d'Hématologie Clinique, F-06202 Nice, France
| | | | - Philippe Rousselot
- Centre Hospitalier de Versailles, Service d'Hématologie et Oncologie, F-78150 Le Chesnay, France; EA4340, Université Versailles-Saint Quentin en Yvelines, Université Paris-Saclay, France
| | - Emilie Cayssials
- INSERM, CIC-P 0802, F-86000 Poitiers, France; CHU de Poitiers, Service d'Oncologie Hématologique et Thérapie Cellulaire, F-86000, Poitiers, France
| | | | - Annelise Bennaceur-Griscelli
- Hôpital Paul Brousse, Service d'Hématologie Biologique, F-94800 Villejuif, France; NSERM U935, F-94807 Villejuif, France; Université Paris Sud, F-94270 Le Kremlin-Bicêtre, France
| | - Jean-Claude Chomel
- INSERM, U935, F-86000 Poitiers, France; CHU de Poitiers, Service de Cancérologie Biologique, F-86021 Poitiers, France
| | - Ali G Turhan
- INSERM, U935, F-86000 Poitiers, France; Hôpital Paul Brousse, Service d'Hématologie Biologique, F-94800 Villejuif, France; NSERM U935, F-94807 Villejuif, France; Université Paris Sud, F-94270 Le Kremlin-Bicêtre, France; Hôpital Bicêtre, Service d'Hématologie Biologique, F-94270 Le Kremlin Bicêtre, France.
| |
Collapse
|
16
|
Huss R, Smith FO, Myerson DH, Deeg HJ. Homing and Immunogenicity of Murine Stromal Cells Transfected with Xenogeneic Mhc Class II Genes. Cell Transplant 2017; 4:483-91. [PMID: 8520832 DOI: 10.1177/096368979500400509] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Syngeneic (murine) and xenogeneic (canine) marrow-derived stromal cells were injected intravenously into SCID and normal mice to examine the homing pattern and persistence of these cells in vivo. By in situ hybridization, these stromal cells were detectable in the bone marrow cavity and the spleen 21 days after injection. Xenogeneic cells did not persist in normal mice but persisted in SCID mice. Conditioning of the recipients with irradiation or S-fluorouracil (5-FU) treatment did not alter these results. In addition, syngeneic murine stromal cells were transfected with the genes for canine MHC class II (DRA + DRB) and transplanted into murine recipients to investigate their homing pattern and immunogenicity. These transfected syngeneic stromal cells did also home to marrow and spleen even in normal recipients. However, these cells led to sensitization of the host towards canine antigens as shown by accelerated skin graft rejection and delayed type hypersensitivity (DTH). Thus, immunodeficient (SCID) mice allow for the homing of xenogeneic stromal cells to hemopoietic organs and for prolonged persistence. In immunocompetent (normal) mice, no xenogeneic stromal cells were identified in spleen and marrow, either because of their inability to home or more likely because of immunological rejection. In contrast, syngeneic stromal cells expressing xenogeneic MHC class II genes did home to spleen and marrow and persisted even though the recipient had become sensitized. Their survival may be due to a loss of expression of the transfected gene. Alternatively, the presentation of these xenogeneic gene products in the hemopoietic organs was such that a cytotoxic response was not induced. These results also show that stromal cells can serve as a vehicle for gene delivery, conceivably with the possibility of organ targeting.
Collapse
Affiliation(s)
- R Huss
- Programs in Transplantation Biology, Pediatric Oncology, and Pathology, Fred Hutchinson Cancer Research Center, Seattle, Washington 98104, USA
| | | | | | | |
Collapse
|
17
|
Casiopeina III-Ea, a copper-containing small molecule, inhibits the in vitro growth of primitive hematopoietic cells from chronic myeloid leukemia. Leuk Res 2016; 52:8-19. [PMID: 27855286 DOI: 10.1016/j.leukres.2016.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/01/2016] [Accepted: 11/01/2016] [Indexed: 11/21/2022]
Abstract
Several novel compounds have been developed for the treatment of different types of leukemia. In the present study, we have assessed the in vitro effects of Casiopeina III-Ea, a copper-containing small molecule, on cells from patients with Chronic Myeloid Leukemia (CML). We included primary CD34+ Lineage-negative (Lin-) cells selected from CML bone marrow, as well as the K562 and MEG01 cell lines. Bone marrow cells obtained from normal individuals - both total mononuclear cells as well as CD34+ Lin- cells- were used as controls. IC50 corresponded to 0.5μM for K562 cells, 0.63μM for MEG01 cells, 0.38μM for CML CD34+ lin- cells, and 1.0μM for normal CD34+ lin- cells. Proliferation and expansion were also inhibited to significantly higher extents in cultures of CML cells as compared to their normal counterparts. All these effects seemed to occur via a bcr-abl transcription-independent mechanism that involved a delay in cell division, an increase in cell death, generation of Reactive Oxygen Species and changes in cell cycle. Our results demonstrate that Casiopeina III-Ea possesses strong antileukemic activity in vitro, and warrant further preclinical (animal) studies to assess such effects in vivo.
Collapse
|
18
|
Opening the door to the development of novel Abl kinase inhibitors. Future Med Chem 2016; 8:2143-2165. [PMID: 27774798 DOI: 10.4155/fmc-2016-0155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The discovery of the importance of kinase activity and its relationship to the emergence and proliferation of cancer cells, due to changes in normal physiology, opened a remarkable pathway for the treatment of chronic myelogenous leukemia through intense search of drug candidates. Six Abl kinase inhibitors have received the US FDA approval as chronic myelogenous leukemia treatment, and continuous efforts in obtaining new, more effective and selective molecules are being carried out. Herein we discuss the mechanisms of Abl inhibition, structural features and ligand/protein interactions that are important for the design of new Abl kinase inhibitors. This review provides a broad overview of binding mode predictions, through molecular docking, which can be an approach to discover novel Abl kinase inhibitors.
Collapse
|
19
|
Lavanya V, Ahmed N, Khan MKA, Jamal S. Sustained mitogenic effect on K562 human chronic myelogenous leukemia cells by dietary lectin, jacalin. Glycoconj J 2016; 33:877-886. [DOI: 10.1007/s10719-016-9725-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/01/2016] [Accepted: 08/10/2016] [Indexed: 10/21/2022]
|
20
|
Montano G, Vidovic K, Palladino C, Cesaro E, Sodaro G, Quintarelli C, De Angelis B, Errichiello S, Pane F, Izzo P, Grosso M, Gullberg U, Costanzo P. WT1-mediated repression of the proapoptotic transcription factor ZNF224 is triggered by the BCR-ABL oncogene. Oncotarget 2016; 6:28223-37. [PMID: 26320177 PMCID: PMC4695056 DOI: 10.18632/oncotarget.4950] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 07/08/2015] [Indexed: 11/25/2022] Open
Abstract
The Kruppel-like protein ZNF224 is a co-factor of the Wilms’ tumor 1 protein, WT1. We have previously shown that ZNF224 exerts a specific proapoptotic role in chronic myelogenous leukemia (CML) K562 cells and contributes to cytosine arabinoside-induced apoptosis, by modulating WT1-dependent transcription of apoptotic genes. Here we demonstrate that ZNF224 gene expression is down-regulated both in BCR-ABL positive cell lines and in primary CML samples and is restored after imatinib and second generation tyrosine kinase inhibitors treatment. We also show that WT1, whose expression is positively regulated by BCR-ABL, represses transcription of the ZNF224 gene. Finally, we report that ZNF224 is significantly down-regulated in patients with BCR-ABL positive chronic phase-CML showing poor response or resistance to imatinib treatment as compared to high-responder patients. Taken as a whole, our data disclose a novel pathway activated by BCR-ABL that leads to inhibition of apoptosis through the ZNF224 repression. ZNF224 could thus represent a novel promising therapeutic target in CML.
Collapse
Affiliation(s)
- Giorgia Montano
- Department of Molecular Medicine and Medical Biotechnology University of Naples Federico II, Naples, Italy.,Department of Haematology and Transfusion Medicine, BioMedical Center, Lund University, Lund, Sweden
| | - Karina Vidovic
- Department of Haematology and Transfusion Medicine, BioMedical Center, Lund University, Lund, Sweden
| | - Chiara Palladino
- Department of Molecular Medicine and Medical Biotechnology University of Naples Federico II, Naples, Italy
| | - Elena Cesaro
- Department of Molecular Medicine and Medical Biotechnology University of Naples Federico II, Naples, Italy
| | - Gaetano Sodaro
- Department of Molecular Medicine and Medical Biotechnology University of Naples Federico II, Naples, Italy
| | - Concetta Quintarelli
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Biagio De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Santa Errichiello
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Fabrizio Pane
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Paola Izzo
- Department of Molecular Medicine and Medical Biotechnology University of Naples Federico II, Naples, Italy
| | - Michela Grosso
- Department of Molecular Medicine and Medical Biotechnology University of Naples Federico II, Naples, Italy
| | - Urban Gullberg
- Department of Haematology and Transfusion Medicine, BioMedical Center, Lund University, Lund, Sweden
| | - Paola Costanzo
- Department of Molecular Medicine and Medical Biotechnology University of Naples Federico II, Naples, Italy
| |
Collapse
|
21
|
Ravandi-Kashani F, Cortes J, Kantarjian H, Talpaz M. Chronic Myeloid Leukemia: Current Guidelines for Diagnosis and Management. Hematology 2016; 3:263-76. [DOI: 10.1080/10245332.1998.11746399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- F. Ravandi-Kashani
- Department of Leukemia, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - J. Cortes
- Department of Leukemia, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - H. Kantarjian
- Department of Leukemia, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - M. Talpaz
- Department of Bioimmuntherapy, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
22
|
Hardy CL. Specificity of Hematopoietic Stem and Progenitor Cell Homing to Bone Marrow: A Perspective. Hematology 2016; 5:391-401. [DOI: 10.1080/10245332.2000.11746535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Cheryl L. Hardy
- G.V. (Sonny) Montgomery Department of Veterans Affairs Medical Center, Department of Medicine, University of Mississippi School of Medicine, Jackson, MS
| |
Collapse
|
23
|
Schepers K, Campbell TB, Passegué E. Normal and leukemic stem cell niches: insights and therapeutic opportunities. Cell Stem Cell 2016; 16:254-67. [PMID: 25748932 DOI: 10.1016/j.stem.2015.02.014] [Citation(s) in RCA: 313] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Hematopoietic stem cells (HSCs) rely on instructive cues from the bone marrow (BM) niche to maintain their quiescence and adapt blood production to the organism's needs. Alterations in the BM niche are commonly observed in blood malignancies and directly contribute to the aberrant function of disease-initiating leukemic stem cells (LSCs). Here, we review recent insights into the cellular and molecular determinants of the normal HSC niche and describe how genetic changes in stromal cells and leukemia-induced BM niche remodeling contribute to blood malignancies. Moreover, we discuss how these findings can be applied to non-cell-autonomous therapies targeting the LSC niche.
Collapse
Affiliation(s)
- Koen Schepers
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Department of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Timothy B Campbell
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Emmanuelle Passegué
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
24
|
Manaflouyan Khajehmarjany S, Rahmani SA, Chavoshi SH, Esfahani A, Movassaghpour Akbari AA. Reliability Evaluation of Fluorescence In Situ Hybridization (FISH) and G-Banding on Bone Marrow and Peripheral Blood Cells in Chronic Myelogenous Leukemia Patients. CELL JOURNAL 2015; 17:171-80. [PMID: 25870848 PMCID: PMC4393667 DOI: 10.22074/cellj.2015.525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/15/2014] [Indexed: 01/25/2023]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease. The cytogenetic hallmark of CML is Philadelphia (Ph) chromosome. This study aimed to diagnose suspected CML patients, to monitor CML patients under therapy using cytogenetic and fluorescence in situ hybridization (FISH) techniques to analyze their bone marrow (BM) and peripheral blood (PB) samples, and finally to compare their obtained results for both specimens. This study was conducted during one-year period (2012-2013). The participants were recruited from the Hematology and Oncology Clinic of Shahid Gazi (Emam Reza) Hospital of Tabriz University of Medical Sciences, Tabriz, East Azerbaijan Province, Iran. We analyzed 90 samples from 60 suspected CML patients (30 BM and 60 PB samples). All samples were analyzed using G-banding, 5 samples using dual fusion FISH (DF-FISH) probes, as well as 30 samples using both FISH and G-banding. Among the 90 analyzed samples of 60 patients, 25 (41.66%) were Ph+ using karyotyping, whereas five cases were not analyzable, so FISH was applied and the results confirmed that only two individuals were BCR-ABL+. In the comparison between 25 BM and 25 PB samples using karyotyping, 15 (60%) and 10 (40%) were ph+, respectively. The comparison of FISH and karyotyping on 30 samples showed that 9 (30%) and 8 (26.66%) were Ph+, respectively, and only 18.18% of Ph+ patients showed atypical patterns. In the comparison between BM-cytogenetic and PB- interphase-FISH (I-FISH), BM-cytogenetic was more reliable than PB-I-FISH in detecting Ph. Our data demonstrate that FISH analysis is a rapid, reliable and sensitive technique. The comparison between BM and PB showed that PB can not be replaced by BM, even in detecting by FISH.
Collapse
Affiliation(s)
- Soheila Manaflouyan Khajehmarjany
- Department of Cellular and Molecular Biology, Islamic Azad University, Ahar Branch, East Azerbaijan, Iran ; Dr. Rahmani Medical Genetic Lab, Tabriz, East Azerbaijan, Iran
| | - Seyed Ali Rahmani
- Department of Cellular and Molecular Biology, Islamic Azad University, Ahar Branch, East Azerbaijan, Iran ; Dr. Rahmani Medical Genetic Lab, Tabriz, East Azerbaijan, Iran
| | - Seyed Hadi Chavoshi
- Department of Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan, Iran ; Emam Reza Hospital, Tabriz, East Azerbaijan, Iran
| | - Ali Esfahani
- Department of Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan, Iran ; Emam Reza Hospital, Tabriz, East Azerbaijan, Iran
| | - Ali Akbar Movassaghpour Akbari
- Department of Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan, Iran ; Emam Reza Hospital, Tabriz, East Azerbaijan, Iran
| |
Collapse
|
25
|
Influence of Bone Marrow Microenvironment on Leukemic Stem Cells: Breaking Up an Intimate Relationship. Adv Cancer Res 2015; 127:227-52. [PMID: 26093902 DOI: 10.1016/bs.acr.2015.04.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The bone marrow microenvironment (BMM) plays a critical role in hematopoietic stem cells (HSCs) maintenance and regulation. There is increasing interest in the role of the BMM in promoting leukemia stem cell (LSC) maintenance, resistance to conventional chemotherapy and targeted therapies, and ultimately disease relapse. Recent studies have enhanced our understanding of how the BMM regulates quiescence, self-renewal, and differentiation of LSC. In this comprehensive review, we discuss recent advances in our understanding of the crosstalk between the BMM and LSC, and the critical signaling pathways underlying these interactions. We also discuss potential approaches to exploit these observations to create novel strategies for targeting therapy-resistant LSC to achieve relapse-free survival in leukemic patients.
Collapse
|
26
|
Chomel JC, Aggoune D, Sorel N, Turhan AG. [Chronic myeloid leukemia stem cells: cross-talk with the niche]. Med Sci (Paris) 2014; 30:452-61. [PMID: 24801043 DOI: 10.1051/medsci/20143004022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The physiological hematopoietic niche located in bone marrow is a pluricellular structure whose components are now well identified. Within this microenvironment, hematopoietic stem cells are in direct contact with mesenchymal stromal cells, osteoblasts and sinusoidal endothelial cells. These close relationships drive specialized cellular functions (proliferation/quiescence, differentiation/self-renewal) ensuring an efficient hematopoiesis. Chronic myeloid leukemia (CML) is a major model of leukemic hematopoiesis. The BCR-ABL1 tyrosine kinase, constitutively activated in CML, plays a critical role in the pathogenesis of the disease. An intensive cross-talk between CML progenitors and the components of the hematopoietic niche has recently been demonstrated. Consequently, the occurrence of the so-called leukemic niche promotes both the proliferation of myeloid cells and the maintenance of quiescent leukemic stem cells. This bone marrow niche could also protect CML stem cells from tyrosine kinase inhibitors and probably contribute to their resistance towards targeted therapies.
Collapse
Affiliation(s)
- Jean-Claude Chomel
- Service de cancérologie biologique, CHU de Poitiers, Poitiers, France - Inserm U935, université de Poitiers, France
| | | | - Nathalie Sorel
- Service de cancérologie biologique, CHU de Poitiers, Poitiers, France - Inserm U935, université de Poitiers, France
| | - Ali G Turhan
- Inserm U935, université de Poitiers, France - hôpitaux universitaires Paris-Sud, le Kremlin Bicêtre, France - Inserm U935, université Paris-Sud 11, Paris, France
| |
Collapse
|
27
|
Ichim CV. Kinase-independent mechanisms of resistance of leukemia stem cells to tyrosine kinase inhibitors. Stem Cells Transl Med 2014; 3:405-15. [PMID: 24598782 DOI: 10.5966/sctm.2012-0159] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Tyrosine kinase inhibitors such as imatinib mesylate have changed the clinical course of chronic myeloid leukemia; however, the observation that these inhibitors do not target the leukemia stem cell implies that patients need to maintain lifelong therapy. The mechanism of this phenomenon is unclear: the question of whether tyrosine kinase inhibitors are inactive inside leukemia stem cells or whether leukemia stem cells do not require breakpoint cluster region (Bcr)-Abl signaling is currently under debate. Herein, I propose an alternative model: perhaps the leukemia stem cell requires Bcr-Abl, but is dependent on its kinase-independent functions. Kinases such as epidermal growth factor receptor and Janus kinase 2 possess kinase-independent roles in regulation of gene expression; it is worth investigating whether Bcr-Abl has similar functions. Mechanistically, Bcr-Abl is able to activate the Ras, phosphatidylinositol 3-kinase/Akt, and/or the Src-kinase Hck/Stat5 pathways in a scaffolding-dependent manner. Whereas the scaffolding activity of Bcr-Abl with Grb2 is dependent on autophosphorylation, kinases such as Hck can use Bcr-Abl as substrate, inducing phosphorylation of Y177 to enable scaffolding ability in the absence of Bcr-Abl catalytic activity. It is worth investigating whether leukemia stem cells exclusively express kinases that are able to use Bcr-Abl as substrate. A kinase-independent role for Bcr-Abl in leukemia stem cells would imply that drugs that target Bcr-Abl's scaffolding ability or its DNA-binding ability should be used in conjunction with current therapeutic regimens to increase their efficacy and eradicate the stem cells of chronic myeloid leukemia.
Collapse
MESH Headings
- Animals
- Drug Resistance, Neoplasm
- Gene Expression Regulation, Leukemic/drug effects
- Gene Expression Regulation, Leukemic/genetics
- Humans
- Leukemia
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplastic Stem Cells/enzymology
- Neoplastic Stem Cells/pathology
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Christine Victoria Ichim
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Discipline of Molecular and Cellular Biology, Sunnybrook Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Serine and proline-rich ligands enriched via phage-display technology show preferential binding to BCR/ABL expressing cells. Hematol Oncol Stem Cell Ther 2014; 7:32-40. [DOI: 10.1016/j.hemonc.2014.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/04/2014] [Indexed: 02/04/2023] Open
|
29
|
Mukhopadhyay A, Helgason GV, Karvela M, Holyoake TL. Hydroxychloroquine for chronic myeloid leukemia: complete cure on the horizon? Expert Rev Hematol 2014; 4:369-71. [DOI: 10.1586/ehm.11.34] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
30
|
Gjertsen BT, Wiig H. Investigation of therapy resistance mechanisms in myeloid leukemia by protein profiling of bone marrow extracellular fluid. Expert Rev Proteomics 2014; 9:595-8. [DOI: 10.1586/epr.12.55] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
31
|
Tyrosine kinase inhibitors and interferon. Mediterr J Hematol Infect Dis 2014; 6:e2014006. [PMID: 24455115 PMCID: PMC3894836 DOI: 10.4084/mjhid.2014.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 11/24/2013] [Indexed: 11/18/2022] Open
Abstract
The use of interferon-a (INF) in chronic myeloid leukemia, when it started in the 80s, was considered as a breakthrough in the therapy of this disease; INF administered alone or in combination with aracytin was the standard choice for treatment for Chronic Myeloid Leukemia (CML) patients unfit for bone marrow transplantation. With the appearance of the first Tyrosine Kinase Inhibitor (TKI) (imatinib) and based on the results of the pivotal IRIS trial, imatinib monotherapy was the new treatment of choice for CML, according to the ELN recommendations. The possibility of combining INF with imatinib, for obtaining better therapeutic responses in CML patients has been already tested and reported. The current challenge is the combined use of second generation TKIs with pegylated –IFN, in order to minimize failures to therapy and increase the number of CML patients with deep molecular responses, who may be able to discontinue lifelong treatment.
Collapse
|
32
|
Identification of interconnected markers for T-cell acute lymphoblastic leukemia. BIOMED RESEARCH INTERNATIONAL 2013; 2013:210253. [PMID: 23956970 PMCID: PMC3727179 DOI: 10.1155/2013/210253] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 06/04/2013] [Indexed: 12/11/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a complex disease, resulting from proliferation of differentially arrested immature T cells. The molecular mechanisms and the genes involved in the generation of T-ALL remain largely undefined. In this study, we propose a set of genes to differentiate individuals with T-ALL from the nonleukemia/healthy ones and genes that are not differential themselves but interconnected with highly differentially expressed ones. We provide new suggestions for pathways involved in the cause of T-ALL and show that network-based classification techniques produce fewer genes with more meaningful and successful results than expression-based approaches. We have identified 19 significant subnetworks, containing 102 genes. The classification/prediction accuracies of subnetworks are considerably high, as high as 98%. Subnetworks contain 6 nondifferentially expressed genes, which could potentially participate in pathogenesis of T-ALL. Although these genes are not differential, they may serve as biomarkers if their loss/gain of function contributes to generation of T-ALL via SNPs. We conclude that transcription factors, zinc-ion-binding proteins, and tyrosine kinases are the important protein families to trigger T-ALL. These potential disease-causing genes in our subnetworks may serve as biomarkers, alternative to the traditional ones used for the diagnosis of T-ALL, and help understand the pathogenesis of the disease.
Collapse
|
33
|
Schepers K, Pietras EM, Reynaud D, Flach J, Binnewies M, Garg T, Wagers AJ, Hsiao EC, Passegué E. Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. Cell Stem Cell 2013; 13:285-99. [PMID: 23850243 DOI: 10.1016/j.stem.2013.06.009] [Citation(s) in RCA: 476] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 05/07/2013] [Accepted: 06/11/2013] [Indexed: 01/16/2023]
Abstract
Multipotent stromal cells (MSCs) and their osteoblastic lineage cell (OBC) derivatives are part of the bone marrow (BM) niche and contribute to hematopoietic stem cell (HSC) maintenance. Here, we show that myeloproliferative neoplasia (MPN) progressively remodels the endosteal BM niche into a self-reinforcing leukemic niche that impairs normal hematopoiesis, favors leukemic stem cell (LSC) function, and contributes to BM fibrosis. We show that leukemic myeloid cells stimulate MSCs to overproduce functionally altered OBCs, which accumulate in the BM cavity as inflammatory myelofibrotic cells. We identify roles for thrombopoietin, CCL3, and direct cell-cell interactions in driving OBC expansion, and for changes in TGF-β, Notch, and inflammatory signaling in OBC remodeling. MPN-expanded OBCs, in turn, exhibit decreased expression of many HSC retention factors and severely compromised ability to maintain normal HSCs, but effectively support LSCs. Targeting this pathological interplay could represent a novel avenue for treatment of MPN-affected patients and prevention of myelofibrosis.
Collapse
Affiliation(s)
- Koen Schepers
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Badger-Brown KM, Gillis LC, Bailey ML, Penninger JM, Barber DL. CBL-B is required for leukemogenesis mediated by BCR-ABL through negative regulation of bone marrow homing. Leukemia 2012; 27:1146-54. [DOI: 10.1038/leu.2012.331] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
35
|
Karotki AV, Baverstock K. What mechanisms/processes underlie radiation-induced genomic instability? Cell Mol Life Sci 2012; 69:3351-60. [PMID: 22955377 PMCID: PMC11115179 DOI: 10.1007/s00018-012-1148-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 08/22/2012] [Accepted: 08/23/2012] [Indexed: 01/11/2023]
Abstract
Radiation-induced genomic instability is a modification of the cell genome found in the progeny of irradiated somatic and germ cells but that is not confined on the initial radiation-induced damage and may occur de novo many generations after irradiation. Genomic instability in the germ line does not follow Mendelian segregation and may have unpredictable outcomes in every succeeding generation. This phenomenon, for which there is extensive experimental data and some evidence in human populations exposed to ionising radiation, is not taken into account in health risk assessments. It poses an unknown morbidity/mortality burden. Based on experimental data derived over the last 20 years (up to January 2012) six mechanistic explanations for the phenomenon have been proposed in the peer-reviewed literature. This article compares these hypotheses with the empirical data to test their fitness to explain the phenomenon. As a conclusion, the most convincing explanation of radiation-induced genomic instability attributes it to an irreversible regulatory change in the dynamic interaction network of the cellular gene products, as a response to non-specific molecular damage, thus entailing the rejection of the machine metaphor for the cell in favour of one appropriate to a complex dissipative dynamic system, such as a whirlpool. It is concluded that in order to evaluate the likely morbidity/mortality associated with radiation-induced genomic instability, it will be necessary to study the damage to processes by radiation rather than damage to molecules.
Collapse
Affiliation(s)
- Andrei V. Karotki
- Radiation Group, International Agency for Research on Cancer, International Agency for Research on Cancer, 150 Cours A. Thomas, 69372 Lyon, France
| | - Keith Baverstock
- Department of Environmental Science, University of Eastern Finland, Kuopio Campus, PL 1627, 70211 Kuopio, Finland
| |
Collapse
|
36
|
Kim H, Gillis LC, Jarvis JD, Yang S, Huang K, Der S, Barber DL. Tyrosine kinase chromosomal translocations mediate distinct and overlapping gene regulation events. BMC Cancer 2011; 11:528. [PMID: 22204395 PMCID: PMC3295743 DOI: 10.1186/1471-2407-11-528] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2011] [Accepted: 12/28/2011] [Indexed: 12/19/2022] Open
Abstract
Background Leukemia is a heterogeneous disease commonly associated with recurrent chromosomal translocations that involve tyrosine kinases including BCR-ABL, TEL-PDGFRB and TEL-JAK2. Most studies on the activated tyrosine kinases have focused on proximal signaling events, but little is known about gene transcription regulated by these fusions. Methods Oligonucleotide microarray was performed to compare mRNA changes attributable to BCR-ABL, TEL-PDGFRB and TEL-JAK2 after 1 week of activation of each fusion in Ba/F3 cell lines. Imatinib was used to control the activation of BCR-ABL and TEL-PDGFRB, and TEL-JAK2-mediated gene expression was examined 1 week after Ba/F3-TEL-JAK2 cells were switched to factor-independent conditions. Results Microarray analysis revealed between 800 to 2000 genes induced or suppressed by two-fold or greater by each tyrosine kinase, with a subset of these genes commonly induced or suppressed among the three fusions. Validation by Quantitative PCR confirmed that eight genes (Dok2, Mrvi1, Isg20, Id1, gp49b, Cxcl10, Scinderin, and collagen Vα1(Col5a1)) displayed an overlapping regulation among the three tested fusion proteins. Stat1 and Gbp1 were induced uniquely by TEL-PDGFRB. Conclusions Our results suggest that BCR-ABL, TEL-PDGFRB and TEL-JAK2 regulate distinct and overlapping gene transcription profiles. Many of the genes identified are known to be involved in processes associated with leukemogenesis, including cell migration, proliferation and differentiation. This study offers the basis for further work that could lead to an understanding of the specificity of diseases caused by these three chromosomal translocations.
Collapse
Affiliation(s)
- Hani Kim
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | | | | | | | | | | | | |
Collapse
|
37
|
Exosomes released by K562 chronic myeloid leukemia cells promote angiogenesis in a Src-dependent fashion. Angiogenesis 2011; 15:33-45. [PMID: 22203239 DOI: 10.1007/s10456-011-9241-1] [Citation(s) in RCA: 251] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 11/04/2011] [Indexed: 12/11/2022]
Abstract
Exosomes, microvesicles of endocytic origin released by normal and tumor cells, play an important role in cell-to-cell communication. Angiogenesis has been shown to regulate progression of chronic myeloid leukemia (CML). The mechanism through which this happens has not been elucidated. We isolated and characterized exosomes from K562 CML cells and evaluated their effects on human umbilical endothelial cells (HUVECs). Fluorescent-labeled exosomes were internalized by HUVECs during tubular differentiation on Matrigel. Exosome localization was perinuclear early in differentiation, moving peripherally in cells undergoing elongation and connection. Exosomes move within and between nanotubular structures connecting the remodeling endothelial cells. They stimulated angiotube formation over a serum/growth factor-limited medium control, doubling total cumulative tube length (P = 0.003). Treatment of K562 cells with two clinically active tyrosine kinase inhibitors, imatinib and dasatinib, reduced their total exosome release (P < 0.009); equivalent concentrations of drug-treated exosomes induced a similar extent of tubular differentiation. However, dasatinib treatment of HUVECs markedly inhibited HUVEC response to drug control CML exosomes (P < 0.002). In an in vivo mouse Matrigel plug model angiogenesis was induced by K562 exosomes and abrogated by oral dasatinib treatment (P < 0.01). K562 exosomes induced dasatinib-sensitive Src phosphorylation and activation of downstream Src pathway proteins in HUVECs. Imatinib was minimally active against exosome stimulation of HUVEC cell differentiation and signaling. Thus, CML cell-derived exosomes induce angiogenic activity in HUVEC cells. The inhibitory effect of dasatinib on exosome production and vascular differentiation and signaling reveals a key role for Src in both the leukemia and its microenvironment.
Collapse
|
38
|
Nicolini FE, Masszi T, Shen Z, Gallagher NJ, Jootar S, Powell BL, Dorlhiac-Llacer PE, Zheng M, Szczudlo T, Turkina A. Expanding Nilotinib Access in Clinical Trials (ENACT), an open-label multicenter study of oral nilotinib in adult patients with imatinib-resistant or -intolerant chronic myeloid leukemia in accelerated phase or blast crisis. Leuk Lymphoma 2011; 53:907-14. [PMID: 22023530 DOI: 10.3109/10428194.2011.627480] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nilotinib has shown favorable safety in patients with Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML) in chronic (CML-CP) or accelerated phase (CML-AP) who failed prior imatinib, and superior efficacy over imatinib in newly diagnosed Ph+ patients with CML-CP. Reported here are the efficacy and safety data for patients in CML-AP (n = 181) or blast crisis (CML-BC) (n = 190; myeloid BC, 133; lymphoid BC, 50; unknown, seven) enrolled in an expanded access phase IIIb study. Non-hematologic adverse events were mostly mild to moderate. Drug-related myelosuppression was generally manageable with dose reductions or interruptions and infrequently led to discontinuation of nilotinib. Drug-related grade 3/4 elevations in serum bilirubin and lipase were infrequent. While an analysis of efficacy was not the primary objective of this study, significant hematologic and cytogenetic responses were observed. These results support the safety and efficacy of nilotinib in patients with advanced CML in AP and BC.
Collapse
|
39
|
Chronic myelogenous leukemia in the age of imatinib: assessing response, acceleration, and blast phase. J Hematop 2011. [DOI: 10.1007/s12308-011-0093-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
40
|
A Model of Oscillatory Blood Cell Counts in Chronic Myelogenous Leukaemia. Bull Math Biol 2011; 73:2983-3007. [DOI: 10.1007/s11538-011-9656-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 03/24/2011] [Indexed: 10/18/2022]
|
41
|
Peptide vaccine therapy for leukemia. Int J Hematol 2011; 93:274-280. [DOI: 10.1007/s12185-011-0781-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 02/01/2011] [Indexed: 10/18/2022]
|
42
|
Lee CF, Griffiths S, Rodríguez-Suárez E, Pierce A, Unwin RD, Jaworska E, Evans CA, J Gaskell S, Whetton AD. Assessment of downstream effectors of BCR/ABL protein tyrosine kinase using combined proteomic approaches. Proteomics 2011; 10:3321-42. [PMID: 20706980 DOI: 10.1002/pmic.201000176] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Leukaemic transformation is frequently associated with the aberrant activity of a protein tyrosine kinase (PTK). As such it is of clinical relevance to be able to map the effects of these leukaemogenic PTKs on haemopoietic cells at the level of phosphorylation modulation. In this paradigm study we have employed a range of proteomic approaches to analyse the effects of one such PTK, BCR/ABL. We have employed phosphoproteome enrichment techniques allied to peptide and protein quantification to identify proteins and pathways involved in cellular transformation. Amongst the proteins shown to be regulated at the post-translational level were cofilin, an actin-severing protein thus linked to altered motility and Cbl an E3 ubiquitin ligase integrally linked to the control of tyrosine kinase signalling (regulated by 5 and 6 PTKs respectively). The major class of proteins identified however were molecular chaperones. We also showed that HSP90 phosphorylation is altered by BCR/ABL action and that HSP90 plays a crucial role in oncogene stability. Further investigation with another six leukaemogenic PTKs demonstrates that this HSP90 role in oncogene stability appears to be a common phenomenon in a range of leukaemias. This opens up the potential opportunity to treat different leukaemias with HSP90 inhibitors.
Collapse
Affiliation(s)
- Chia Fang Lee
- Stem Cell and Leukaemia Proteomics Laboratory, School of Cancer, Enabling Sciences, Manchester Academic Health Science Centre, The University of Manchester, Wolfson Molecular Imaging Centre, Withington, Manchester, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Functional phosphoproteomic analysis reveals cold-shock domain protein A to be a Bcr-Abl effector-regulating proliferation and transformation in chronic myeloid leukemia. Cell Death Dis 2010; 1:e93. [PMID: 21368869 PMCID: PMC3032323 DOI: 10.1038/cddis.2010.72] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
One proposed strategy to suppress the proliferation of imatinib-resistant cells in chronic myeloid leukemia (CML) is to inhibit key proteins downstream of Bcr-Abl. The PI3K/Akt pathway is activated by Bcr-Abl and is specifically required for the growth of CML cells. To identify targets of this pathway, we undertook a proteomic screen and identified several proteins that differentially bind 14-3-3, dependent on Bcr-Abl kinase activity. An siRNA screen of candidates selected by bioinformatics analysis reveals cold-shock domain protein A (CSDA), shown previously to regulate cell cycle progression in epithelial cells, to be a positive regulator of proliferation in a CML cell line. We show that Akt can phosphorylate the serine 134 residue of CSDA but, downstream of Bcr-Abl activity, this modification is mediated through the activation of MEK/p90 ribosomal S6 kinase (RSK) signaling. Inhibition of RSK, similarly to treatment with imatinib, blocked proliferation specifically in Bcr-Abl-positive leukemia cell lines, as well as cells from CML patients. Furthermore, these primary CML cells showed an increase in CSDA phosphorylation. Expression of a CSDA phospho-deficient mutant resulted in the decrease of Bcr-Abl-dependent transformation in Rat1 cells. Our results support a model whereby phosphorylation of CSDA downstream of Bcr-Abl enhances proliferation in CML cells to drive leukemogenesis.
Collapse
|
44
|
Ciarcia R, d'Angelo D, Pacilio C, Pagnini D, Galdiero M, Fiorito F, Damiano S, Mattioli E, Lucchetti C, Florio S, Giordano A. Dysregulated calcium homeostasis and oxidative stress in chronic myeloid leukemia (CML) cells. J Cell Physiol 2010; 224:443-53. [PMID: 20432440 DOI: 10.1002/jcp.22140] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic myeloid leukemia (CML) is a hematopoietic stem cell disorder caused by the oncogenic activity of the Bcr-Abl protein, a deregulated tyrosine kinase. Calcium may act directly on cellular enzymes and in conjunction with other cellular metabolites, such as cyclic nucleotides, to regulate cell functions. Alteration in the ionized calcium concentration in the cytosol has been implicated in the initiation of secretion, contraction, and cell proliferation as well as the production of reactive oxygen species (ROS) has been correlates with normal cell proliferation through activation of growth-related signaling pathways. In this study we evaluated in peripheral blood leukocytes from CML patients the role of the balance between intracellular calcium and oxidative stress in CML disease in order to identify possible therapeutic targets in patients affected by this pathology. Our results demonstrated that peripheral blood mononuclear cells derived from CML patients displayed decreased intracellular calcium [Ca(2+)](i) fluxes both after InsP(3) as well as ATP and ionomycin (IONO) administration. CML cells showed lower levels of superoxide dismutase (SOD) activity and significantly higher malondialdehyde levels (MDA) than peripheral blood mononuclear cells derived from control patients. Finally we showed that resveratrol is able to down-regulate InsP3 and ATP effects on intracellular calcium [Ca(2+)](i) fluxes as well as the effects of ATP and IONO on oxidative stress in CML cells.
Collapse
Affiliation(s)
- Roberto Ciarcia
- Department of Structures, Functions and Biological Technologies, School of Veterinary Medicine, University of Naples Federico II, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
|
46
|
|
47
|
Deisseroth AB, Zhang W, Cha Y, Yuan T, Chen H, Sims S, Wedrychowski A, Gao PQ, Huston L, Filaccio M, Claxton D, Kornblau S, Johnson E, Zack Howard OM, Andersson B, Giglio AD, Gressot L, Kantarjian H, Talpaz M, Khouri I, Champlin R, Andreeff M, Gaozza E, Seong D, Suh SP, Ellerson D, Hu G, Chou M. New Directions in the Biology and Therapy of Chronic Myeloid Leukemia. Leuk Lymphoma 2009. [DOI: 10.3109/10428199209064884] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
48
|
Mayani H, Flores-Figueroa E, Chávez-González A. In vitro biology of human myeloid leukemia. Leuk Res 2009; 33:624-37. [DOI: 10.1016/j.leukres.2008.11.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Revised: 11/04/2008] [Accepted: 11/08/2008] [Indexed: 11/27/2022]
|
49
|
Zhelyazkova AG, Tonchev AB, Kolova P, Ivanova L, Gercheva L. Prognostic significance of hepatocyte growth factor and microvessel bone marrow density in patients with chronic myeloid leukaemia. Scand J Clin Lab Invest 2009; 68:492-500. [PMID: 18609087 DOI: 10.1080/00365510701854991] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE The aims of the study were: (1) to perform a complex angiogenic assessment in chronic myeloid leukaemia (CML) patients using multiple parameters: bone marrow microvessel density (MVD), bone marrow immunohistochemical cellular expressions of vascular endothelial growth factor (VEGF) and its receptor KDR, as well as hepatocyte growth factor (HGF) and its receptor MET, and the plasma VEGF and HGF; and (2) to determine the clinical significance of these factors for patients with CML. MATERIAL AND METHODS The VEGF and HGF plasma levels were analysed by ELISA in 38 newly diagnosed CML patients. Immunohistochemical methods were used to visualize the MVD as well as the cellular VEGF/KDR and HGF/MET expression. RESULTS We found an increased MVD, cellular VEGF/KDR and HGF/MET expression and elevated plasma VEGF and HGF in CML patients. The plasma HGF, cellular HGF and MET expression correlated with the CML phase. The plasma HGF correlated with all markers reflecting the tumour burden (leucocytes, blast percentage, splenomegaly and LDH) as well as with the phase of CML and overall survival of the patients. Cox regression analysis determined the prognostic relevance of HGF and MVD parameters, but not for the plasma VEGF and cellular VEGF and KDR. CONCLUSIONS Using a complex angiogenic assessment we determined an increased angiogenesis in CML patients. No prognostic relevance was found for VEGF plasma levels or VEGF/KDR cellular bone marrow expression. The increased cellular HGF and MET expressions could be considered high-risk factors for these patients. Plasma HGF and MVD were shown to be independent prognostic parameters for patients' survival.
Collapse
Affiliation(s)
- A G Zhelyazkova
- Department of Clinical Haematology, University Hospital St. Marina Varna, Varna University of Medicine, Varna, Bulgaria.
| | | | | | | | | |
Collapse
|
50
|
Meshkini A, Yazdanparast R. Involvement of ERK/MAPK pathway in megakaryocytic differentiation of K562 cells induced by 3-hydrogenkwadaphnin. Toxicol In Vitro 2008; 22:1503-10. [DOI: 10.1016/j.tiv.2008.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Revised: 04/09/2008] [Accepted: 05/14/2008] [Indexed: 10/22/2022]
|