1
|
Panuzzo C, Pironi L, Maglione A, Rocco S, Stanga S, Riganti C, Kopecka J, Ali MS, Pergolizzi B, Bracco E, Cilloni D. mTORC2 Is Activated under Hypoxia and Could Support Chronic Myeloid Leukemia Stem Cells. Int J Mol Sci 2023; 24:ijms24021234. [PMID: 36674750 PMCID: PMC9865638 DOI: 10.3390/ijms24021234] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Hypoxia is a critical condition that governs survival, self-renewal, quiescence, metabolic shift and refractoriness to leukemic stem cell (LSC) therapy. The present study aims to investigate the hypoxia-driven regulation of the mammalian Target of the Rapamycin-2 (mTORC2) complex to unravel it as a novel potential target in chronic myeloid leukemia (CML) therapeutic strategies. After inducing hypoxia in a CML cell line model, we investigated the activities of mTORC1 and mTORC2. Surprisingly, we detected a significant activation of mTORC2 at the expense of mTORC1, accompanied by the nuclear localization of the main substrate phospho-Akt (Ser473). Moreover, the Gene Ontology analysis of CML patients' CD34+ cells showed enrichment in the mTORC2 signature, further strengthening our data. The deregulation of mTOR complexes highlights how hypoxia could be crucial in CML development. In conclusion, we propose a mechanism by which CML cells residing under a low-oxygen tension, i.e., in the leukemia quiescent LSCs, singularly regulate the mTORC2 and its downstream effectors.
Collapse
Affiliation(s)
- Cristina Panuzzo
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy
- Correspondence:
| | - Lucrezia Pironi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy
| | - Alessandro Maglione
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy
| | - Simone Rocco
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy
| | - Serena Stanga
- Department of Neuroscience Rita Levi Montalcini, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Turin, Italy
| | - Chiara Riganti
- Department of Oncology, University of Turin, 10043 Turin, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Turin, 10043 Turin, Italy
| | - Muhammad Shahzad Ali
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy
| | - Barbara Pergolizzi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy
| | - Enrico Bracco
- Department of Oncology, University of Turin, 10043 Turin, Italy
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy
| |
Collapse
|
2
|
Özgür Yurttaş N, Eşkazan AE. Novel therapeutic approaches in chronic myeloid leukemia. Leuk Res 2020; 91:106337. [PMID: 32200189 DOI: 10.1016/j.leukres.2020.106337] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 12/22/2022]
Abstract
The tyrosine kinase inhibitors (TKIs) have revolutionized the management of chronic myeloid leukemia (CML) and BCR-ABL1 inhibitors form the mainstay of CML treatment. Although patients with CML generally do well under TKI therapy, there is a subgroup of patients who are resistant and/or intolerant to TKIs. In these group of patients, there is the need of additional treatment strategies. In this review, we provide an update on the current knowledge of these novel treatment approaches that can be used alone and/or in combination with TKIs.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Clinical Trials as Topic
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Everolimus/therapeutic use
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/immunology
- Gene Expression
- Histone Deacetylase Inhibitors/therapeutic use
- Homoharringtonine/therapeutic use
- Humans
- Immunotherapy/methods
- Interferon-alpha/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Molecular Targeted Therapy/methods
- Niacinamide/analogs & derivatives
- Niacinamide/therapeutic use
- Piperidines/therapeutic use
- Polyethylene Glycols/therapeutic use
- Protein Kinase Inhibitors/therapeutic use
- Pyrazoles/therapeutic use
- Pyridines/therapeutic use
- Quinolones/therapeutic use
- Recombinant Proteins/therapeutic use
Collapse
Affiliation(s)
- Nurgül Özgür Yurttaş
- Division of Hematology, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Ahmet Emre Eşkazan
- Division of Hematology, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| |
Collapse
|
3
|
Massimino M, Stella S, Tirrò E, Romano C, Pennisi MS, Puma A, Manzella L, Zanghì A, Stagno F, Di Raimondo F, Vigneri P. Non ABL-directed inhibitors as alternative treatment strategies for chronic myeloid leukemia. Mol Cancer 2018; 17:56. [PMID: 29455672 PMCID: PMC5817805 DOI: 10.1186/s12943-018-0805-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/01/2018] [Indexed: 02/07/2023] Open
Abstract
The introduction of ABL Tyrosine Kinase Inhibitors (TKIs) has significantly improved the outcome of Chronic Myeloid Leukemia (CML) patients that, in large part, achieve satisfactory hematological, cytogenetic and molecular remissions. However, approximately 15-20% fail to obtain optimal responses according to the current European Leukemia Network recommendation because of drug intolerance or resistance.Moreover, a plethora of evidence suggests that Leukemic Stem Cells (LSCs) show BCR-ABL1-independent survival. Hence, they are unresponsive to TKIs, leading to disease relapse if pharmacological treatment is discontinued.All together, these biological events generate a subpopulation of CML patients in need of alternative therapeutic strategies to overcome TKI resistance or to eradicate LSCs in order to allow cure of the disease.In this review we update the role of "non ABL-directed inhibitors" targeting signaling pathways downstream of the BCR-ABL1 oncoprotein and describe immunological approaches activating specific T cell responses against CML cells.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor
- Combined Modality Therapy
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Molecular Targeted Therapy
- Signal Transduction/drug effects
- Treatment Outcome
Collapse
Affiliation(s)
- Michele Massimino
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Stefania Stella
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Elena Tirrò
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Chiara Romano
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Maria Stella Pennisi
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Adriana Puma
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Antonino Zanghì
- Department of Surgical Medical Sciences and Advanced Technologies, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Fabio Stagno
- Division of Hematology and Bone Marrow Transplant, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Francesco Di Raimondo
- Division of Hematology and Bone Marrow Transplant, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Department of Surgery, Medical and Surgical Specialties, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy.
| |
Collapse
|
4
|
Hirao T, Yamaguchi M, Kikuya M, Chibana H, Ito K, Aoki S. Altered intracellular signaling by imatinib increases the anti-cancer effects of tyrosine kinase inhibitors in chronic myelogenous leukemia cells. Cancer Sci 2017; 109:121-131. [PMID: 29121435 PMCID: PMC5765287 DOI: 10.1111/cas.13442] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 10/28/2017] [Accepted: 11/02/2017] [Indexed: 12/15/2022] Open
Abstract
Tyrosine kinase inhibitors (TKI), including imatinib (IM), improve the outcome of CML therapy. However, TKI treatment is long‐term and can induce resistance to TKI, which often leads to a poor clinical outcome in CML patients. Here, we examined the effect of continuous IM exposure on intracellular energy metabolism in K562 cells, a human Philadelphia chromosome‐positive CML cell line, and its subsequent sensitivity to anti‐cancer agents. Contrary to our expectations, we found that continuous IM exposure increased sensitivity to TKI. Cancer energy metabolism, characterized by abnormal glycolysis, is linked to cancer cell survival. Interestingly, glycolytic activity was suppressed by continuous exposure to IM, and autophagy increased to maintain cell viability by compensating for glycolytic suppression. Notably, increased sensitivity to TKI was not caused by glycolytic inhibition but by altered intracellular signaling, causing glycolytic suppression and increased autophagy, as evidenced by suppression of p70 S6 kinase 1 (S6K1) and activation of AMP‐activated protein kinase (AMPK). Using another human CML cell line (KCL22 cells) and BCR/ABL+ Ba/F3 cells (mimicking Philadelphia chromosome‐positive CML cells) confirmed that suppressing S6K1 and activating AMPK increased sensitivity to TKI. Furthermore, suppressing S6K1 and activating AMPK had a synergistic anti‐cancer effect by inhibiting autophagy in the presence of TKI. The present study provides new insight into the importance of signaling pathways that affect cellular energy metabolism, and suggests that co‐treatment with agents that disrupt energy metabolic signaling (using S6K1 suppressors and AMPK activators) plus blockade of autophagy may be strategies for TKI‐based CML therapy.
Collapse
Affiliation(s)
- Takuya Hirao
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | | | - Megumi Kikuya
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hiroji Chibana
- Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Kousei Ito
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Shigeki Aoki
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
5
|
Osoegawa A, Gills JJ, Kawabata S, Dennis PA. Rapamycin sensitizes cancer cells to growth inhibition by the PARP inhibitor olaparib. Oncotarget 2017; 8:87044-87053. [PMID: 29152062 PMCID: PMC5675614 DOI: 10.18632/oncotarget.19667] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 05/12/2017] [Indexed: 12/20/2022] Open
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPi) have been developed and tested in a context of combining it with double-stranded (ds) DNA repair defects or inhibitors, as PARP inhibitor impairs single-stranded (ss) DNA break repair, resulting in the activation of the dsDNA break repair machinery. Rapamycin has been widely prescribed for more than a decade and recent studies have revealed that it may inhibit dsDNA break repair. The combination of the PARP inhibitor olaparib and rapamycin synergistically inhibited cell proliferation in non-small cell lung cancer (NSCLC) cells, and even in triple negative breast cancer (TNBC) cells with BRCA1 mutations. Rad51, which forms a polymer on ssDNA upon dsDNA breaks, plays an essential role in homologous recombination. Olaparib induced Rad51 focus formation, while rapamycin successfully inhibited it both in vivo and in vitro, suggesting that this combination worked through the blocking of both ssDNA break repair and dsDNA break repair; hence the cells cannot go through the G2/M checkpoint. The protein level of PARP was a predictive marker for both PAR activity and Rad51 focus formation in this combination. Collectively, these data suggest that this combination could have therapeutic potential in the treatment of cancer with high PARP expression, or in combination with cytotoxic chemotherapy or radiotherapy.
Collapse
Affiliation(s)
- Atsushi Osoegawa
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joell J Gills
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shigeru Kawabata
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Phillip A Dennis
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
6
|
Inhibition of Ras-mediated signaling pathways in CML stem cells. Cell Oncol (Dordr) 2015; 38:407-18. [PMID: 26458816 DOI: 10.1007/s13402-015-0248-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Chronic myeloid leukemia (CML) is a clonal myeloproliferative disorder characterized by the presence of the BCR-ABL1 oncoprotein in cells with a hematopoietic stem cell (HSC) origin. BCR-ABL1 tyrosine kinase activity leads to constitutive activation of Ras, which in turn acts as a branch point to initiate multiple downstream signaling pathways governing proliferation, self-renewal, differentiation and apoptosis. As aberrant regulation of these cellular processes causes transformation and disease progression particularly in advanced stages of CML, investigation of these signaling pathways may uncover new therapeutic targets for the selective eradication of CML stem cells. Transcription factors play a crucial role in unbalancing the Ras signaling network and have recently been investigated as potential modulators in this regard. In this review, we first briefly summarize the Ras-associated molecular pathways that are involved in the regulation of CML stem cell properties. Next we discuss the relevance of Ras-associated transcription factors as nuclear targets in combination treatment strategies for CML. CONCLUSIONS A closer investigation of the influence of Ras-mediated signaling pathways on CML progression to blast crisis is warranted to uncover new directions for targeted therapies, particularly in cases that are resistant to current tyrosine kinase inhibitors.
Collapse
|
7
|
Garofalo F, Christoforidis D, di Summa PG, Gay B, Cherix S, Raffoul W, Demartines N, Matter M. The unresolved case of sacral chordoma: from misdiagnosis to challenging surgery and medical therapy resistance. Ann Coloproctol 2014; 30:122-31. [PMID: 24999463 PMCID: PMC4079810 DOI: 10.3393/ac.2014.30.3.122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 10/18/2013] [Indexed: 01/16/2023] Open
Abstract
PURPOSE A sacral chordoma is a rare, slow-growing, primary bone tumor, arising from embryonic notochordal remnants. Radical surgery is the only hope for cure. The aim of our present study is to analyse our experience with the challenging treatment of this rare tumor, to review current treatment modalities and to assess the outcome based on R status. METHODS Eight patients were treated in our institution between 2001 and 2011. All patients were discussed by a multidisciplinary tumor board, and an en bloc surgical resection by posterior perineal access only or by combined anterior/posterior accesses was planned based on tumor extension. RESULTS Seven patients underwent radical surgery, and one was treated by using local cryotherapy alone due to low performance status. Three misdiagnosed patients had primary surgery at another hospital with R1 margins. Reresection margins in our institution were R1 in two and R0 in one, and all three recurred. Four patients were primarily operated on at our institution and had en bloc surgery with R0 resection margins. One had local recurrence after 18 months. The overall morbidity rate was 86% (6/7 patients) and was mostly related to the perineal wound. Overall, 3 out of 7 resected patients were disease-free at a median follow-up of 2.9 years (range, 1.6-8.0 years). CONCLUSION Our experience confirms the importance of early correct diagnosis and of an R0 resection for a sacral chordoma invading pelvic structures. It is a rare disease that requires a challenging multidisciplinary treatment, which should ideally be performed in a tertiary referral center.
Collapse
Affiliation(s)
- Fabio Garofalo
- Department of Surgery, Intercantonal Hospital Broye, Payerne, Switzerland. ; Department of Visceral Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Dimitrios Christoforidis
- Department of Visceral Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. ; Department of Surgery, Hospital Civico, Lugano, Switzerland
| | - Pietro G di Summa
- Department of Plastic and Reconstructive Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Béatrice Gay
- Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Stéphane Cherix
- Department of Orthopedics, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Wassim Raffoul
- Department of Plastic and Reconstructive Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Nicolas Demartines
- Department of Visceral Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Maurice Matter
- Department of Visceral Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| |
Collapse
|
8
|
Li J, Xue L, Hao H, Li R, Luo J. Rapamycin combined with celecoxib enhanced antitumor effects of mono treatment on chronic myelogenous leukemia cells through downregulating mTOR pathway. Tumour Biol 2014; 35:6467-74. [PMID: 24682932 DOI: 10.1007/s13277-014-1820-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 03/04/2014] [Indexed: 11/26/2022] Open
Abstract
Chronic myelogenous leukemia is a neoplasm of myeloid progenitor cells. We recently found that rapamycin could induce G0/G1 phase arrest and apoptosis and inhibit proliferation of K562 cells through inhibiting mammalian target of rapamycin (mTOR) pathway. However, whether rapamycin has synergistic effects with other drugs in chronic myelogenous leukemia (CML) therapies remain unclear. Therefore, we examined the effect of rapamycin combined with celecoxib on K562 cells in vitro. The survival rates showed a significant decrease in rapamycin + celecoxib treatment group. The combination treatment also increased the G0/G1 phase cells as compared to rapamycin or celecoxib treatment alone (P < 0.05), accompanied with the decreased population of S phase cells. Meanwhile, the rate of apoptosis was 15.87 ± 2.21 % in rapamycin + celecoxib treatment group, significantly higher than that in mono treatment group (P < 0.05). Western blot and reverse transcription PCR (RT-PCR) analysis showed that the expressions of mTOR, 4E-BP1, and p70S6K were all significantly decreased in K562 cells after rapamycin + celecoxib treatment (P < 0.05). In conclusion, rapamycin combined with celecoxib could induce cell cycle arrest and apoptosis and decrease the expressions of mTOR, 4E-BP1, and p70S6K. It suggested that the combination could enhance the antitumor effects of mono treatment on CML cells through downregulating mTOR pathway.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Apoptosis/drug effects
- Celecoxib
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Phosphorylation/drug effects
- Pyrazoles/administration & dosage
- Signal Transduction/drug effects
- Sirolimus/administration & dosage
- Sulfonamides/administration & dosage
- TOR Serine-Threonine Kinases/biosynthesis
- TOR Serine-Threonine Kinases/genetics
Collapse
Affiliation(s)
- Jie Li
- Department of Hematology, Hebei General Hospital, Shijiazhuang, 050000, China
| | | | | | | | | |
Collapse
|
9
|
Yang X, He G, Gong Y, Zheng B, Shi F, Shi R, Yang X. Mammalian target of rapamycin inhibitor rapamycin enhances anti-leukemia effect of imatinib on Ph+ acute lymphoblastic leukemia cells. Eur J Haematol 2014; 92:111-20. [PMID: 24112092 DOI: 10.1111/ejh.12202] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2013] [Indexed: 02/05/2023]
Abstract
BCR-ABL fusion gene typically causes a type of acute lymphoblastic leukemia (ALL), known as Ph+ ALL. Although imatinib (IM) treatment induced high rates of complete response (CR), serious acute and late complications are frequent, whereas more vexatiously resistance to chemotherapy and clinical relapse develops. Therefore, the efficacy of treatment in Ph+ ALL is still to be determined. In this study, we focused our attention on the potential benefit of rapamycin (RAPA), an mammalian target of rapamycin (mTOR) inhibitor, in combination with IM on a Ph+ ALL cell line SUP-B15 and a primary Ph+ ALL sample in vitro. Analysis of cell proliferation showed that RAPA (50 nm) plus IM exerted good synergistic effect on Ph+ ALL cells. Notably, we found that IM treatment induced the abnormal activation of the components of mTOR signaling pathway and p-BCR-ABL, whereas RAPA potently eliminated this deleterious side effect induced by IM and might overcome the resistance to IM. The synergistic effect was also associated with the increase in autophagy, which seemed to have an opposite role with apoptosis in Ph+ ALL cells, and cell cycle arrest in G1 phase. Altogether, our results suggested that IM in combination with RAPA was more effective for Ph+ ALL cells than IM alone.
Collapse
Affiliation(s)
- Xi Yang
- Department of Hematology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | | | | | | | | | | | | |
Collapse
|
10
|
PI3K/mTOR pathway inhibitors sensitize chronic myeloid leukemia stem cells to nilotinib and restore the response of progenitors to nilotinib in the presence of stem cell factor. Cell Death Dis 2013; 4:e827. [PMID: 24091670 PMCID: PMC3824646 DOI: 10.1038/cddis.2013.309] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 06/28/2013] [Accepted: 07/15/2013] [Indexed: 02/08/2023]
Abstract
Nilotinib is a second-generation tyrosine kinase inhibitor, designed to specifically inhibit break-point cluster region (BCR)-Abelson (ABL) and developed to treat chronic myeloid leukemia (CML) in patients showing a resistance to imatinib. We previously demonstrated that nilotinib-induced apoptosis was reduced by stem cell factor (SCF) addition. Here, the SCF-activated survival pathway was investigated. BCR-ABL expression was accompanied by the activation of the SCF receptor: c-KIT. Nilotinib inhibited this activation that was restored by SCF binding. Parallel variations were observed for mammaliam target of rapamycin (mTOR) kinase and mTOR complex 1 substrate S6K. The inhibition of mTORC1 restored the response of BCR-ABL cell lines to nilotinib in the presence of SCF. PI3K inhibition restored nilotinib-induced apoptosis. On hematopoietic progenitors from CML patient's bone marrows, mTORC1 inhibition also restored nilotinib sensitivity in the presence of SCF, confirming its involvement in SCF-activated survival pathway. However, this pathway seems not to be involved in the nilotinib-induced resistance of the CML stem cell population. Conversely, PI3K inhibition sensitized both CML progenitors and stem cells to nilotinib, suggesting that, downstream PI3K, two different kinase pathways are activated in CML progenitor and stem cell populations.
Collapse
|
11
|
Stoklosa T, Glodkowska-Mrowka E, Hoser G, Kielak M, Seferynska I, Wlodarski P. Diverse mechanisms of mTOR activation in chronic and blastic phase of chronic myelogenous leukemia. Exp Hematol 2013; 41:462-9. [DOI: 10.1016/j.exphem.2013.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 12/19/2012] [Accepted: 02/01/2013] [Indexed: 12/26/2022]
|
12
|
Barrett D, Brown VI, Grupp SA, Teachey DT. Targeting the PI3K/AKT/mTOR signaling axis in children with hematologic malignancies. Paediatr Drugs 2012; 14:299-316. [PMID: 22845486 PMCID: PMC4214862 DOI: 10.2165/11594740-000000000-00000] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The phosphatidylinositiol 3-kinase (PI3K), AKT, mammalian target of rapamycin (mTOR) signaling pathway (PI3K/AKT/mTOR) is frequently dysregulated in disorders of cell growth and survival, including a number of pediatric hematologic malignancies. The pathway can be abnormally activated in childhood acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), and chronic myelogenous leukemia (CML), as well as in some pediatric lymphomas and lymphoproliferative disorders. Most commonly, this abnormal activation occurs as a consequence of constitutive activation of AKT, providing a compelling rationale to target this pathway in many of these conditions. A variety of agents, beginning with the rapamycin analogue (rapalog) sirolimus, have been used successfully to target this pathway in a number of pediatric hematologic malignancies. Rapalogs demonstrate significant preclinical activity against ALL, which has led to a number of clinical trials. Moreover, rapalogs can synergize with a number of conventional cytotoxic agents and overcome pathways of chemotherapeutic resistance for drugs commonly used in ALL treatment, including methotrexate and corticosteroids. Based on preclinical data, rapalogs are also being studied in AML, CML, and non-Hodgkin's lymphoma. Recently, significant progress has been made using rapalogs to treat pre-malignant lymphoproliferative disorders, including the autoimmune lymphoproliferative syndrome (ALPS); complete remissions in children with otherwise therapy-resistant disease have been seen. Rapalogs only block one component of the pathway (mTORC1), and newer agents are under preclinical and clinical development that can target different and often multiple protein kinases in the PI3K/AKT/mTOR pathway. Most of these agents have been tolerated in early-phase clinical trials. A number of PI3K inhibitors are under investigation. Of note, most of these also target other protein kinases. Newer agents are under development that target both mTORC1 and mTORC2, mTORC1 and PI3K, and the triad of PI3K, mTORC1, and mTORC2. Preclinical data suggest these dual- and multi-kinase inhibitors are more potent than rapalogs against many of the aforementioned hematologic malignancies. Two classes of AKT inhibitors are under development, the alkyl-lysophospholipids (APLs) and small molecule AKT inhibitors. Both classes have agents currently in clinical trials. A number of drugs are in development that target other components of the pathway, including eukaryotic translation initiation factor (eIF) 4E (eIF4E) and phosphoinositide-dependent protein kinase 1 (PDK1). Finally, a number of other key signaling pathways interact with PI3K/AKT/mTOR, including Notch, MNK, Syk, MAPK, and aurora kinase. These alternative pathways are being targeted alone and in combination with PI3K/AKT/mTOR inhibitors with promising preclinical results in pediatric hematologic malignancies. This review provides a comprehensive overview of the abnormalities in the PI3K/AKT/mTOR signaling pathway in pediatric hematologic malignancies, the agents that are used to target this pathway, and the results of preclinical and clinical trials, using those agents in childhood hematologic cancers.
Collapse
Affiliation(s)
- David Barrett
- Department of Pediatrics, Division of Oncology, Children’s
Hospital of Philadelphia, University of Pennsylvania School of Medicine,
Philadelphia, PA, USA
| | - Valerie I. Brown
- Department of Pediatrics, Division of Oncology, Children’s
Hospital of Philadelphia, University of Pennsylvania School of Medicine,
Philadelphia, PA, USA
| | - Stephan A. Grupp
- Department of Pediatrics, Division of Oncology, Children’s
Hospital of Philadelphia, University of Pennsylvania School of Medicine,
Philadelphia, PA, USA
| | - David T. Teachey
- Department of Pediatrics, Division of Oncology, Children’s
Hospital of Philadelphia, University of Pennsylvania School of Medicine,
Philadelphia, PA, USA
- Department of Pediatrics, Division of Hematology, Children’s
Hospital of Philadelphia, University of Pennsylvania School of Medicine,
Philadelphia, PA, USA
| |
Collapse
|
13
|
mTOR kinase inhibitors as a treatment strategy in hematological malignancies. Future Med Chem 2012; 4:487-504. [PMID: 22416776 DOI: 10.4155/fmc.12.14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) kinase is a key element of intracellular signal transduction, responsible for the regulation of cell growth and proliferation. Since abnormal activation of the mTOR pathway was found in several tumors, including human malignancies, it may be an attractive target for antineoplastic treatment. The first identified mTOR inhibitor was rapamycin (sirolimus). Subsequently, the most potent rapamycin analogues (rapalogues), such as everolimus, temsirolimus and deforolimus, have been developed. After encouraging preclinical experiments, several clinical trials testing the rapalogues in monotherapy or in combinations with other cytotoxic agents have been conducted in patients with hematological malignancies. Results of these studies, described in this review, indicate that inhibition of the mTOR pathway may be a very promising strategy of anti-tumor treatment in several types of lymphomas and leukemias. Recently, a second generation of more effective mTOR inhibitors has been developed. These are currently being assessed in preclinical, Phase I or I/II clinical studies.
Collapse
|
14
|
Abstract
Targeted small-molecule drugs have revolutionized treatment of chronic myeloid leukemia (CML) during the last decade. These agents interrupt a constitutively active BCR-ABL, the causative agent for CML, by interfering with adenosine 5' triphosphate-dependent ABL tyrosine kinase. Although the efficacy of tyrosine kinase inhibitors (TKIs) has resulted in overall survival of greater than 90%, TKIs are not curative. Moreover, no currently approved TKIs are effective against the T315I BCR-ABL variant. However, a new generation of TKIs with activity against T315I is on the horizon. We will highlight the clinical utility of historical CML therapeutics, those used today (first- and second-generation TKIs), and discuss treatment modalities that are under development. Recent advances have illuminated the complexity of CML, especially within the marrow microenvironment. We contend that the key to curing CML will involve strategies beyond targeting BCR-ABL because primitive human CML stem cells are not dependent on BCR-ABL. Ultimately, drug combinations or exploiting synthetic lethality may transform responses into definitive cures for CML.
Collapse
|
15
|
The PI3K/PKB signaling module as key regulator of hematopoiesis: implications for therapeutic strategies in leukemia. Blood 2012; 119:911-23. [PMID: 22065598 DOI: 10.1182/blood-2011-07-366203] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Abstract
An important mediator of cytokine signaling implicated in regulation of hematopoiesis is the PI3K/protein kinase B (PKB/c-Akt) signaling module. Constitutive activation of this signaling module has been observed in a large group of leukemias. Because activation of this signaling pathway has been demonstrated to be sufficient to induce hematologic malignancies and is thought to correlate with poor prognosis and enhanced drug resistance, it is considered to be a promising target for therapy. A high number of pharmacologic inhibitors directed against either individual or multiple components of this pathway have already been developed to improve therapy. In this review, the safety and efficacy of both single and dual-specificity inhibitors will be discussed as well as the potential of combination therapy with either inhibitors directed against other signal transduction molecules or classic chemotherapy.
Collapse
|
16
|
Abstract
The mammalian target of rapamycin (mTOR) is an intracellular serine/threonine kinase that exists as a downstream component of numerous signaling pathways. The activation of mTOR results in the production of proteins involved in cell metabolism, growth, proliferation, and angiogenesis. Aberrant activation of mTOR signaling has been identified in a number of cancers, and targeted inhibition of mTOR has been successful in achieving tumor responses, prolonging progression-free survival, and increasing overall survival in various oncologic patient populations. In particular, persistent activation of mTOR signaling has been identified in cell lines and patient samples with leukemias, Hodgkin's lymphoma (HL), non-Hodgkin's lymphoma (NHL), multiple myeloma (MM), and Waldenström's macroglobulinemia (WM). In vitro and preclinical studies using agents that inhibit mTOR signaling have demonstrated cytostatic and cytotoxic effects in these hematologic malignancies, suggesting that mTOR is a rational target for therapy in these disease states. In addition, the combination of mTOR inhibitors with traditional therapies may help to overcome the development of resistance and may improve response rates over those seen with established regimens through synergistic or additive effects. Inhibitors of mTOR signaling currently are being investigated in clinical trials of hematologic malignancies as single agents and as components of combination regimens. Thus far, promising results have been seen with the application of mTOR inhibitors as single agents in patients with relapsed or refractory leukemia, HL, NHL, MM, and WM.
Collapse
Affiliation(s)
- Anas Younes
- Department of Lymphoma/Myeloma, M.D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | |
Collapse
|
17
|
Redig AJ, Vakana E, Platanias LC. Regulation of mammalian target of rapamycin and mitogen activated protein kinase pathways by BCR-ABL. Leuk Lymphoma 2011; 52 Suppl 1:45-53. [PMID: 21299459 DOI: 10.3109/10428194.2010.546919] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A large body of evidence has established that BCR-ABL regulates engagement and activation of mammalian target of rapamycin (mTOR) and mitogen activated protein kinase (MAPK) signaling cascades. mTOR-mediated signals, as well as signals transduced by ERK, JNK, and p38 MAPK, are important components of the aberrant signaling induced by BCR-ABL. Such deregulation of mTOR or MAPK pathways contributes to BCR-ABL leukemogenesis, and their targeting with selective inhibitors provides an approach to enhance antileukemic responses and/or overcome leukemic cell resistance in chronic myeloid leukemia (CML) and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). This review explores recent advances in our understanding of mTOR and MAPK signaling in BCR-ABL-expressing leukemias and discusses the potential therapeutic targeting of these pathways in CML and Ph+ ALL.
Collapse
Affiliation(s)
- Amanda J Redig
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology/Oncology, Northwestern University Medical School and Jesse Brown VA Medical Center, Chicago, IL, USA
| | | | | |
Collapse
|
18
|
Kelly KR, Rowe JH, Padmanabhan S, Nawrocki ST, Carew JS. Mammalian target of rapamycin as a target in hematological malignancies. Target Oncol 2011; 6:53-61. [PMID: 21499765 DOI: 10.1007/s11523-011-0175-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Accepted: 03/03/2011] [Indexed: 12/12/2022]
Abstract
The mammalian target of rapamycin (mTOR) regulates protein synthesis in addition to cell growth and cell proliferation. Elucidation of the roles of the phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR pathway in the regulation of the pathogenesis of hematological neoplasms has led to the development and clinical evaluation of agents targeting this pathway for the treatment of leukemia and lymphomas. Clinical trials conducted to date have shown modest responses to mTOR inhibition in patients with various hematological malignancies. Novel agents that simultaneously target mTOR complex 2 (mTORC2) or AKT in addition to mTOR complex 1 (mTORC1) may offer an opportunity to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Kevin R Kelly
- Institute for Drug Development, Cancer Therapy & Research Center, University of Texas Health Science Center, 7979 Wurzbach Rd, San Antonio, TX 78229, USA.
| | | | | | | | | |
Collapse
|
19
|
Yamaguchi T, Miki Y, Yoshida K. The c-Abl tyrosine kinase stabilizes Pitx1 in the apoptotic response to DNA damage. Apoptosis 2010; 15:927-35. [PMID: 20563669 DOI: 10.1007/s10495-010-0488-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In the DNA damage response, c-Abl tyrosine kinase is transiently accumulated in the nucleus and induces apoptosis; however, little is known about the mechanism underlying apoptosis induction via nuclear c-Abl. Here we demonstrate that the expression of human pituitary homeobox 1 (Pitx1) transcription factor is increased after DNA damage. Notably, c-Abl controls augmentation of Pitx1 at the post-transcriptional level. Overexpression of c-Abl induces tyrosine phosphorylation of Pitx1, either directly or indirectly. We also show that, upon exposure to genotoxic stress, overexpression of Pitx1 is associated with marked induction of apoptosis that is independent of p53 status. Importantly, inhibition of c-Abl kinase activity substantially attenuates Pitx1-mediated apoptosis. These findings provide evidence that c-Abl participates in modulating Pitx1 expression in the apoptotic response to DNA damage.
Collapse
Affiliation(s)
- Tomoko Yamaguchi
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Japan
| | | | | |
Collapse
|
20
|
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
|
21
|
Targeting the translational machinery as a novel treatment strategy for hematologic malignancies. Blood 2010; 115:2127-35. [PMID: 20075156 DOI: 10.1182/blood-2009-09-220020] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The dysregulation of protein synthesis evident in the transformed phenotype has opened up a burgeoning field of research in cancer biology. Translation initiation has recently been shown to be a common downstream target of signal transduction pathways deregulated in cancer and initiated by mutated/overexpressed oncogenes and tumor suppressors. The overexpression and/or activation of proteins involved in translation initiation such as eIF4E, mTOR, and eIF4G have been shown to induce a malignant phenotype. Therefore, understanding the mechanisms that control protein synthesis is emerging as an exciting new research area with significant potential for developing innovative therapies. This review highlights molecules that are activated or dysregulated in hematologic malignancies, and promotes the transformed phenotype through the deregulation of protein synthesis. Targeting these proteins with small molecule inhibitors may constitute a novel therapeutic approach in the treatment of cancer.
Collapse
|