1
|
Travers RJ, Stepanian A, Jaffe I. Endothelium as a Source of Cardiovascular Toxicity From Antitumor Kinase Inhibitors. Arterioscler Thromb Vasc Biol 2024; 44:2143-2153. [PMID: 39145393 PMCID: PMC11424247 DOI: 10.1161/atvbaha.124.319864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Kinase inhibitors (KIs) targeting oncogenic molecular pathways have revolutionized cancer therapy. By directly targeting specific tumor-driving kinases, targeted therapies have fewer side effects compared with chemotherapy. Despite the enhanced specificity, cardiovascular side effects have emerged with many targeted cancer therapies that limit long-term outcomes in patients with cancer. Endothelial cells lining all blood vessels are critical to cardiovascular health and are also exposed to circulating levels of systemic anticancer therapies. Both on- and off-target perturbation of signaling pathways from KIs can cause endothelial dysfunction, resulting in cardiovascular toxicity. As such, the endothelium is a potential source, and also a therapeutic target for prevention, of cardiovascular toxicity. In this review, we examine the evidence for KI-induced endothelial cell dysfunction as a mechanism for the cardiovascular toxicities of vascular endothelial growth factor inhibitors, BCR-Abl (breakpoint cluster region-Abelson proto-oncogene) KIs, Bruton tyrosine inhibitors, and emerging information regarding endothelial toxicity of newer classes of KIs.
Collapse
Affiliation(s)
- Richard J Travers
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
- Division of Hematology and Oncology, Tufts Medical Center, Boston MA
| | - Alec Stepanian
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
| | - Iris Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
| |
Collapse
|
2
|
Gover-Proaktor A, Leshem-Lev D, Winograd-Katz S, Partouche S, Samara A, Shapira S, Nardi-Agmon I, Harari E, Younis A, Najjar A, Kornowski R, Geiger B, Raanani P, Leader A, Granot G. Dasatinib induces endothelial dysfunction leading to impaired recovery from ischaemia. Br J Haematol 2024; 205:1011-1016. [PMID: 38877865 DOI: 10.1111/bjh.19595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/04/2024] [Indexed: 10/25/2024]
Abstract
Chronic myeloid leukaemia (CML) management is complicated by treatment-emergent vascular adverse events seen with tyrosine kinase inhibitors (TKIs) such as nilotinib, dasatinib and ponatinib. Pleural effusion and pulmonary arterial hypertension (PAH) have been associated with dasatinib treatment. Endothelial dysfunction and impaired angiogenesis are hallmarks of PAH. In this study, we explored, at cellular and whole animal levels, the connection between dasatinib exposure and disruption of endothelial barrier integrity and function, leading to impaired angiogenesis. Understanding the mechanisms whereby dasatinib initiates PAH will provide opportunities for intervention and prevention of such adverse effects, and for future development of safer TKIs, thereby improving CML management.
Collapse
Affiliation(s)
| | - Dorit Leshem-Lev
- Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Sabina Winograd-Katz
- Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Shirly Partouche
- Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Aladin Samara
- Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Saar Shapira
- Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | | | - Emanuel Harari
- Cardiology Division, Assuta Ashdod University Hospital, Ben-Gurion University of the Negev, Ashdod, Israel
| | - Aseel Younis
- Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | | | - Ran Kornowski
- Department of Cardiology, Rabin Medical Center, Petah Tikva, Israel
- The Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Benjamin Geiger
- Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Pia Raanani
- The Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | - Avi Leader
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Galit Granot
- Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
- The Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
3
|
Sacha T, Krawczyk K. Ponatinib in the treatment of patients with chronic myeloid leukemia and increased cardiovascular risk: A review of management strategies. Hematol Transfus Cell Ther 2024:S2531-1379(24)00298-0. [PMID: 39448326 DOI: 10.1016/j.htct.2024.04.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/05/2024] [Accepted: 04/01/2024] [Indexed: 10/26/2024] Open
Abstract
The introduction of tyrosine kinase inhibitors has revolutionized the treatment of chronic myeloid leukemia vastly improving the prognosis and clinical outcome of most patients. It was estimated that approximately 40-50 % of patients treated with imatinib will require treatment with a second-generation or third-generation tyrosine kinase inhibitor to achieve an optimal response. The treatment duration, increased patient survival, and aging of the population receiving tyrosine kinase inhibitors raise concerns as to long-term toxicities, such as an elevated cardiovascular risk and a higher rate of comorbidities. Ponatinib is a highly potent third-generation tyrosine kinase inhibitor that was shown to be effective in patients with a wide range of ABL mutations, including T315I. The use of ponatinib is associated with significant vascular toxicity, including peripheral arterial occlusive disease, ischemic heart disease, cerebrovascular accidents, and venous thromboembolism. This review discusses the vascular toxicity of ponatinib and presents a comprehensive panel of tests for the evaluation of patients requiring ponatinib therapy. Moreover, the management of patients with cardiovascular risk factors who receive ponatinib is discussed. Finally, the strategy for establishing the optimal dose of ponatinib in patients with chronic myeloid leukemia is described.
Collapse
Affiliation(s)
- Tomasz Sacha
- Jagiellonian University Medical College, Kopernika, Kraków, Poland.
| | | |
Collapse
|
4
|
Mahapatra S, Kar P. Computational biophysical characterization of the effect of gatekeeper mutations on the binding of ponatinib to the FGFR kinase. Arch Biochem Biophys 2024; 758:110070. [PMID: 38909834 DOI: 10.1016/j.abb.2024.110070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/15/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Fibroblast Growth Factor Receptor (FGFR) is connected to numerous downstream signalling cascades regulating cellular behavior. Any dysregulation leads to a plethora of illnesses, including cancer. Therapeutics are available, but drug resistance driven by gatekeeper mutation impedes the treatment. Ponatinib is an FDA-approved drug against BCR-ABL kinase and has shown effective results against FGFR-mediated carcinogenesis. Herein, we undertake molecular dynamics simulation-based analysis on ponatinib against all the FGFR isoforms having Val to Met gatekeeper mutations. The results suggest that ponatinib is a potent and selective inhibitor for FGFR1, FGFR2, and FGFR4 gatekeeper mutations. The extensive electrostatic and van der Waals interaction network accounts for its high potency. The FGFR3_VM mutation has shown resistance towards ponatinib, which is supported by their lesser binding affinity than wild-type complexes. The disengaged molecular brake and engaged hydrophobic spine were believed to be the driving factors for weak protein-ligand interaction. Taken together, the inhibitory and structural characteristics exhibited by ponatinib may aid in thwarting resistance based on Val-to-Met gatekeeper mutations at an earlier stage of treatment and advance the design and development of other inhibitors targeted at FGFRs harboring gatekeeper mutations.
Collapse
Affiliation(s)
- Subhasmita Mahapatra
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore, 453552, Madhya Pradesh, India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore, 453552, Madhya Pradesh, India.
| |
Collapse
|
5
|
Zibrova D, Ernst T, Hochhaus A, Heller R. The BCR::ABL1 tyrosine kinase inhibitors ponatinib and nilotinib differentially affect endothelial angiogenesis and signalling. Mol Cell Biochem 2024:10.1007/s11010-024-05070-5. [PMID: 39009935 DOI: 10.1007/s11010-024-05070-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024]
Abstract
BCR::ABL1 inhibitors, the treatment of choice for the majority of patients with chronic myeloid leukaemia (CML), can cause vascular side effects that vary between agents. The exact underlying mechanisms are still poorly understood, but the vascular endothelium has been proposed as a site of origin. The present study investigates the effects of three BCR::ABL1 inhibitors, ponatinib, nilotinib and imatinib, on angiogenesis and signalling in human endothelial cells in response to vascular endothelial growth factor (VEGF). The experiments were performed in endothelial cells isolated from human umbilical veins. After exposure to imatinib, ponatinib and nilotinib, the angiogenic capacity of endothelial cells was assessed in spheroid assays. VEGF-induced signalling pathways were examined in Western blotting experiments using different specific antibodies. RNAi technology was used to downregulate proteins of interest. Intracellular cGMP levels were measured by ELISA. Imatinib had no effect on endothelial function. Ponatinib inhibited VEGF-induced sprouting, while nilotinib increased spontaneous and VEGF-stimulated angiogenesis. These effects did not involve wild-type ABL1 or ABL2, as siRNA-mediated knockdown of these kinases did not affect angiogenesis and VEGF signalling. Consistent with their effects on sprouting, ponatinib and nilotinib affected angiogenic pathways in opposite directions. While ponatinib inhibited VEGF-induced signalling and cGMP formation, nilotinib activated angiogenic signalling, in particular phosphorylation of extracellular signal-regulated kinase 1/2 (Erk1/2). The latter occurred in an epidermal growth factor receptor (EGFR)-dependent manner possibly via suppressing Fyn-related kinase (FRK), a negative regulator of EGFR signalling. Both, pharmacological inhibition of Erk1/2 or EGFR suppressed nilotinib-induced angiogenic sprouting. These results support the notion that the vascular endothelium is a site of action of BCR::ABL1 inhibitors from which side effects may arise, and that the different vascular toxicity profiles of BCR::ABL1 inhibitors may be due to their different actions at the molecular level. In addition, the as yet unknown pro-angiogenic effect of nilotinib should be considered in the treatment of patients with comorbidities associated with pathological angiogenesis, such as ocular disease, arthritis or obesity.
Collapse
Affiliation(s)
- Darya Zibrova
- Center for Molecular Biomedicine, Institute of Molecular Cell Biology, Jena University Hospital, Hans-Knöll-Straße 2, 07745, Jena, Germany
| | - Thomas Ernst
- Department of Hematology and Oncology, Jena University Hospital, Jena, Germany
| | - Andreas Hochhaus
- Department of Hematology and Oncology, Jena University Hospital, Jena, Germany
| | - Regine Heller
- Center for Molecular Biomedicine, Institute of Molecular Cell Biology, Jena University Hospital, Hans-Knöll-Straße 2, 07745, Jena, Germany.
| |
Collapse
|
6
|
Krajcsir B, Pócsi M, Fejes Z, Nagy B, Kappelmayer J, Beke Debreceni I. Ponatinib Induces a Procoagulant Phenotype in Human Coronary Endothelial Cells via Inducing Apoptosis. Pharmaceutics 2024; 16:559. [PMID: 38675220 PMCID: PMC11055157 DOI: 10.3390/pharmaceutics16040559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
BCR-ABL tyrosine kinase inhibitors (TKIs) are effective drugs in the treatment of patients with chronic myeloid leukemia. However, based on clinical studies, ponatinib was associated with the development of thrombotic complications. Since endothelial cells (ECs) regulate blood coagulation, their abnormal phenotype may play a role in the development of thrombotic events. We here aimed to investigate the effect of ponatinib on the procoagulant activity of cultured endothelial cells in vitro. Human coronary artery endothelial cells (HCAECs) were incubated with 50, 150, and 1000 nM of ponatinib. Subsequently, phosphatidylserine (PS) exposure and endothelial microvesicles (EMVs) were measured by flow cytometry. In addition, EC- and EMV-dependent thrombin generation was analyzed. To investigate pro-apoptotic effects of ponatinib, the level of Bax and Bcl-xL proteins were studied using Western blot and F3, THBD, and VCAM1 mRNAs were quantified by qPCR. Therapeutic concentrations of ponatinib significantly increased PS expression on ECs and the amount of EMVs which significantly shortened the time parameters of thrombin generation. In addition, these changes were associated with an increased ratio of Bax and Bcl-xL proteins in the presence of the decreased THBD mRNA level. Overall, ponatinib enhances the procoagulant activity of ECs via inducing apoptosis, which may contribute to thrombotic events.
Collapse
Affiliation(s)
- Bálint Krajcsir
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
- Laki Kálmán Doctoral School, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Marianna Pócsi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
| | - Zsolt Fejes
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
| | - János Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
| | - Ildikó Beke Debreceni
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
| |
Collapse
|
7
|
Nomura T, Hata A. Limb Salvage Surgery for Ponatinib-Induced Bilateral Chronic Limb-Threatening Ischemia in a Patient with Chronic Myeloid Leukemia with T315I Mutation in BCR-ABL: A Case Report. Ann Vasc Dis 2024; 17:51-54. [PMID: 38628933 PMCID: PMC11018110 DOI: 10.3400/avd.cr.23-00059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/29/2023] [Indexed: 04/19/2024] Open
Abstract
A 72-year-old woman with chronic myeloid leukemia with T315I mutation in breakpoint cluster region-abelson (BCR-ABL) was treated with ponatinib. During the course of her treatment, chronic limb-threatening ischemia developed in both lower extremities, and the left lower extremity was amputated below the knee at a previous hospital. She was referred to our department for salvage of the right lower extremity. We performed a foot bypass and multidisciplinary treatment of the wound, and achieved epithelialization in about 1 month. The rate of vascular occlusive events with ponatinib has been reported to be high, and we believe that careful monitoring is important during use.
Collapse
Affiliation(s)
- Takuo Nomura
- Department of Vascular Surgery, Juzen Memorial Hospital, Hamamatsu, Shizuoka, Japan
| | - Akito Hata
- Division of Thoracic Oncology, Kobe Minimally Invasive Cancer Center, Kobe, Hyogo, Japan
| |
Collapse
|
8
|
Aghel N, Gustafson D, Delgado D, Atenafu EG, Fish JE, Lipton JH. High sensitivity c-reactive protein and circulating biomarkers of endothelial dysfunction in patients with chronic myeloid leukemia receiving tyrosine kinase inhibitors. Leuk Lymphoma 2023; 64:2008-2017. [PMID: 37554059 DOI: 10.1080/10428194.2023.2242990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/25/2023] [Accepted: 07/05/2023] [Indexed: 08/10/2023]
Abstract
Tyrosine kinase inhibitors (TKIs) have revolutionized the management of patients with chronic myelogenous leukemia (CML); however, they may cause cardiovascular (CV) toxicities. In this cross-sectional study, we explored whether high-sensitivity C-reactive protein (hsCRP) and novel markers of vascular dysfunction were associated with exposure to specific TKIs, in 262 CML patients. Hs-CRP level was not associated with CML disease activity or treatment with a specific TKI. Body mass index (OR: 1.15, 95% CI: 1.108-1.246; p < 0.001) and CML duration (OR: 1.004, 95% CI: 1.001-1.008; p = 0.024) were independently associated with higher hs-CRP. In exploratory analyses, novel endothelial-centric markers (e.g. ET-1 and VCAM-1) were differential across the various TKIs, particularly amongst nilotinib- and ponatinib-treated patients. While Levels of hs-CRP do not appear to be correlated with specific TKIs, circulating markers of vascular dysfunction were altered in patients treated with specific TKIs and should be explored as potential markers of TKI-associated CV risk.
Collapse
Affiliation(s)
- Nazanin Aghel
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, Ted Rogers Program in Cardiotoxicity Prevention University Health Network, Toronto, Canada
- Division of Cardiology, Cardio-Oncology Program, McMaster University, Hamilton, Canada
| | - Dakota Gustafson
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Diego Delgado
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, Ted Rogers Program in Cardiotoxicity Prevention University Health Network, Toronto, Canada
| | - Eshetu G Atenafu
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network University of Toronto, Toronto, ON, Canada
| | - Jason E Fish
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, Ted Rogers Program in Cardiotoxicity Prevention University Health Network, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Jeffrey H Lipton
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Wang Y, Travers RJ, Farrell A, Lu Q, Bays JL, Stepanian A, Chen C, Jaffe IZ. Differential vascular endothelial cell toxicity of established and novel BCR-ABL tyrosine kinase inhibitors. PLoS One 2023; 18:e0294438. [PMID: 37983208 PMCID: PMC10659179 DOI: 10.1371/journal.pone.0294438] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 11/01/2023] [Indexed: 11/22/2023] Open
Abstract
BCR-ABL tyrosine kinase inhibitors (TKIs) have dramatically improved survival in Philadelphia chromosome-positive leukemias. Newer BCR-ABL TKIs provide superior cancer outcomes but with increased risk of acute arterial thrombosis, which further increases in patients with cardiovascular comorbidities and mitigates survival benefits compared to imatinib. Recent studies implicate endothelial cell (EC) damage in this toxicity by unknown mechanisms with few side-by-side comparisons of multiple TKIs and with no available data on endothelial impact of recently approved TKIs or novels TKIs being tested in clinical trials. To characterize BCR-ABL TKI induced EC dysfunction we exposed primary human umbilical vein ECs in 2D and 3D culture to clinically relevant concentrations of seven BCR-ABL TKIs and quantified their impact on EC scratch-wound healing, viability, inflammation, and permeability mechanisms. Dasatinib, ponatinib, and nilotinib, the TKIs associated with thrombosis in patients, all significantly impaired EC wound healing, survival, and proliferation compared to imatinib, but only dasatinib and ponatinib impaired cell migration and only nilotinib enhanced EC necrosis. Dasatinib and ponatinib increased leukocyte adhesion to ECs with upregulation of adhesion molecule expression in ECs (ICAM1, VCAM1, and P-selectin) and leukocytes (PSGL1). Dasatinib increased permeability and impaired cell junctional integrity in human engineered microvessels, consistent with its unique association with pleural effusions. Of the new agents, bafetinib decreased EC viability and increased microvessel permeability while asciminib and radotinib did not impact any EC function tested. In summary, the vasculotoxic TKIs (dasatinib, ponatinib, nilotinib) cause EC toxicity but with mechanistic differences, supporting the potential need for drug-specific vasculoprotective strategies. Asciminib and radotinib do not induce EC toxicity at clinically relevant concentrations suggesting a better safety profile.
Collapse
Affiliation(s)
- Yihua Wang
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America
- Tufts University, Medford, MA, United States of America
| | - Richard J. Travers
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America
- The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| | - Alanna Farrell
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America
| | - Jennifer L. Bays
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - Alec Stepanian
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America
| | - Christopher Chen
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America
| |
Collapse
|
10
|
Yan K, Zheng J, Kluth MA, Li L, Ganss C, Yard B, Magdeburg R, Frank MH, Pallavi P, Keese M. ABCB5 + mesenchymal stromal cells therapy protects from hypoxia by restoring Ca 2+ homeostasis in vitro and in vivo. Stem Cell Res Ther 2023; 14:24. [PMID: 36759868 PMCID: PMC9912525 DOI: 10.1186/s13287-022-03228-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 12/21/2022] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Hypoxia in ischemic disease impairs Ca2+ homeostasis and may promote angiogenesis. The therapeutic efficacy of mesenchymal stromal cells (MSCs) in peripheral arterial occlusive disease is well established, yet its influence on cellular Ca2+ homeostasis remains to be elucidated. We addressed the influence of ATP-binding cassette subfamily B member 5 positive mesenchymal stromal cells (ABCB5+ MSCs) on Ca2+ homeostasis in hypoxic human umbilical vein endothelial cells (HUVECs) in vitro and in vivo. METHODS Hypoxia was induced in HUVECs by Cobalt (II) chloride (CoCl2) or Deferoxamine (DFO). Dynamic changes in the cytosolic- and endoplasmic reticulum (ER) Ca2+ and changes in reactive oxygen species were assessed by appropriate fluorescence-based sensors. Metabolic activity, cell migration, and tube formation were assessed by standard assays. Acute-on-chronic ischemia in Apolipoprotein E knock-out (ApoE-/-) mice was performed by double ligation of the right femoral artery (DFLA). ABCB5+ MSC cells were injected into the ischemic limb. Functional recovery after DFLA and histology of gastrocnemius and aorta were assessed. RESULTS Hypoxia-induced impairment of cytosolic and ER Ca2+ were restored by ABCB5+ MSCs or their conditioned medium. Similar was found for changes in intracellular ROS production, metabolic activity, migratory ability and tube formation. The restoration was paralleled by an increased expression of the Ca2+ transporter Sarco-/endoplasmic reticulum ATPase 2a (SERCA2a) and the phosphorylation of Phospholamban (PLN). In acute-on-chronic ischemia, ABCB5+ MSCs treated mice showed a higher microvascular density, increased SERCA2a expression and PLN phosphorylation relative to untreated controls. CONCLUSIONS ABCB5+ MSCs therapy can restore cellular Ca2+ homeostasis, which may beneficially affect the angiogenic function of endothelial cells under hypoxia in vitro and in vivo.
Collapse
Affiliation(s)
- Kaixuan Yan
- grid.7700.00000 0001 2190 4373Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany ,grid.7700.00000 0001 2190 4373European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jiaxing Zheng
- grid.7700.00000 0001 2190 4373Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany ,grid.7700.00000 0001 2190 4373European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Lin Li
- grid.7700.00000 0001 2190 4373Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany ,grid.7700.00000 0001 2190 4373European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Ganss
- TICEBA GmbH, Heidelberg, Germany ,grid.476673.7RHEACELL GmbH & Co. KG, Heidelberg, Germany
| | - Benito Yard
- grid.7700.00000 0001 2190 4373V Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Richard Magdeburg
- grid.411778.c0000 0001 2162 1728Department of Surgery, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68161 Mannheim, Germany
| | - Markus H. Frank
- grid.38142.3c000000041936754XDepartment of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XTransplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XHarvard Stem Cell Institute, Harvard University, Cambridge, MA USA ,grid.1038.a0000 0004 0389 4302School of Medical and Health Sciences, Edith Cowan University, Perth, WA Australia
| | - Prama Pallavi
- Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,Department of Surgery, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68161, Mannheim, Germany.
| | - Michael Keese
- Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,Department for General and Visceral Surgery, Theresienkrankenhaus Mannheim, Mannheim, Germany. .,Department of Surgery, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68161, Mannheim, Germany.
| |
Collapse
|
11
|
Hypoxia damages endothelial cell angiogenic function by reducing the Ca2+ restoring ability of the endoplasmic reticulum. Biochem Biophys Res Commun 2022; 626:142-150. [DOI: 10.1016/j.bbrc.2022.07.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 07/22/2022] [Accepted: 07/27/2022] [Indexed: 11/21/2022]
|
12
|
Sacha T, Szczepanek E, Dumnicka P, Góra-Tybor J, Niesiobędzka-Krężel J, Prejzner W, Wasilewska E, Kłoczko J, Ciepłuch H, Makowska W, Patkowska E, Wasilewska J, Bober G, Kopera M, Wichary R, Kroll-Balcerzak R, Gromek T, Wach M, Rudkowska-Kazanowska A, Świniarska M, Paczkowska E, Biernat M, Joks M, Oller M, Kasza R, Kostyra A, Gil J, Grzybowska-Izydorczyk O. The Outcomes of Ponatinib Therapy in Patients With Chronic Myeloid Leukemia Resistant or Intolerant to Previous Tyrosine Kinase Inhibitors, Treated in Poland Within the Donation Program. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:405-415. [PMID: 34933827 DOI: 10.1016/j.clml.2021.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 06/14/2023]
Abstract
INTRODUCTION Tyrosine kinase inhibitors (TKIs) have greatly improved the treatment outcome for most patients with chronic myeloid leukemia (CML). Ponatinib is a new pan-inhibitor of TK active in resistant CML. This study aimed to evaluate the efficacy and safety of ponatinib in patients suffering from CML. PATIENTS AND METHODS This multicenter, non-randomized, observational, retrospective study evaluated the efficacy and safety of ponatinib administered in adult CML patients in any disease phase, including those with a detected ABL T315I mutation, which were resistant or intolerant to previous-generation TKIs. The study comprised 43 patients benefiting from the ponatinib donation program who were treated in 16 Polish centers. RESULTS For patients who started treatment with ponatinib in chronic phase (CP) (n = 23) and in accelerated phase (AP) (n = 3) the median time on ponatinib was 19.5 months (range: 1.0-35.4), and 31.7 months (range: 31.0-34.1), respectively. All these patients were in CP after 1 month of treatment and at the end of observation - none of them progressed to AP or blastic phase (BP) during the study, meaning that progression-free survival was 100% at the end of observation (35.4 months). The estimated 2-year survival in this group of patients was 84%. For all 43 patients, median survival was not reached (lower quartile 6.3 months), and estimated 2-year survival was 60%. CONCLUSION Our analysis confirmed ponatinib efficacy in a significant proportion of patients heavily pre-treated with TKIs achieving durable responses in both CP and AP/BP CML groups.
Collapse
Affiliation(s)
- Tomasz Sacha
- Department of Hematology, Jagiellonian University Medical College, Krakow, Poland.
| | - Elżbieta Szczepanek
- Department of Hematology, Jagiellonian University Medical College, Krakow, Poland
| | - Paulina Dumnicka
- Department of Medical Diagnostics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Joanna Góra-Tybor
- Department of Hematology, Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland
| | - Joanna Niesiobędzka-Krężel
- Department of Hematology, Transplantation and Internal Medicine, University Clinical Center of the Medical University of Warsaw, Poland
| | - Witold Prejzner
- Department of Hematology and Transplantology, Medical University of Gdansk, Gdansk, Poland
| | - Ewa Wasilewska
- Department of Hematology, Medical University of Bialystok, Bialystok, Poland
| | - Janusz Kłoczko
- Department of Hematology, Medical University of Bialystok, Bialystok, Poland
| | - Hanna Ciepłuch
- Department of Hematology, Copernicus Regional Oncology Centre, Gdansk, Poland
| | - Wioletta Makowska
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Elżbieta Patkowska
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Joanna Wasilewska
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Grażyna Bober
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Silesia, School of Medicine in Katowice, Katowice, Poland
| | - Małgorzata Kopera
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Silesia, School of Medicine in Katowice, Katowice, Poland
| | - Ryszard Wichary
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Silesia, School of Medicine in Katowice, Katowice, Poland
| | - Renata Kroll-Balcerzak
- Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Poznan, Poland
| | - Tomasz Gromek
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | - Małgorzata Wach
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | - Anna Rudkowska-Kazanowska
- Department of Hematology and Cancer Prevention, School of Public Health in Bytom, Medical University of Silesia in Katowice, Bytom, Poland
| | - Magdalena Świniarska
- Department of Hematology and Transplantology, Pomeranian Medical University, Szczecin, Poland
| | - Edyta Paczkowska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Monika Biernat
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Monika Joks
- Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Poznan, Poland
| | - Marta Oller
- Department of Hematology, Copernicus Regional Oncology Centre, Gdansk, Poland
| | - Renata Kasza
- Department of Hematology, Zamosc Hospital, Zamosc, Poland
| | | | - Justyna Gil
- Department of Hematooncology, Oncology Centre of the Podkarpackie Province, Brzozow, Poland
| | | |
Collapse
|
13
|
Choksey A, Timm KN. Cancer Therapy-Induced Cardiotoxicity-A Metabolic Perspective on Pathogenesis, Diagnosis and Therapy. Int J Mol Sci 2021; 23:441. [PMID: 35008867 PMCID: PMC8745714 DOI: 10.3390/ijms23010441] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/21/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022] Open
Abstract
Long-term cardiovascular complications of cancer therapy are becoming ever more prevalent due to increased numbers of cancer survivors. Cancer therapy-induced cardiotoxicity (CTIC) is an incompletely understood consequence of various chemotherapies, targeted anti-cancer agents and radiation therapy. It is typically detected clinically by a reduction in cardiac left ventricular ejection fraction, assessed by echocardiography. However, once cardiac functional decline is apparent, this indicates irreversible cardiac damage, highlighting a need for the development of diagnostics which can detect CTIC prior to the onset of functional decline. There is increasing evidence to suggest that pathological alterations to cardiac metabolism play a crucial role in the development of CTIC. This review discusses the metabolic alterations and mechanisms which occur in the development of CTIC, with a focus on doxorubicin, trastuzumab, imatinib, ponatinib, sunitinib and radiotherapy. Potential methods to diagnose and predict CTIC prior to functional cardiac decline in the clinic are evaluated, with a view to both biomarker and imaging-based approaches. Finally, the therapeutic potential of therapies which manipulate cardiac metabolism in the context of adjuvant cardioprotection against CTIC is examined. Together, an integrated view of the role of metabolism in pathogenesis, diagnosis and treatment is presented.
Collapse
Affiliation(s)
- Anurag Choksey
- Somerville College, University of Oxford, Woodstock Road, Oxford OX2 6HD, UK;
| | - Kerstin N. Timm
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
14
|
Telerman A, Granot G, Leibovitch C, Yarchovsky-Dolberg O, Shacham-Abulafia A, Partouche S, Yeshurun M, Ellis MH, Raanani P, Wolach O. Neutrophil Extracellular Traps Are Increased in Chronic Myeloid Leukemia and Are Differentially Affected by Tyrosine Kinase Inhibitors. Cancers (Basel) 2021; 14:cancers14010119. [PMID: 35008283 PMCID: PMC8750902 DOI: 10.3390/cancers14010119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/13/2021] [Accepted: 12/24/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Neutrophil extracellular traps (NETs) are a recently described form of neutrophil cellular death that has been associated with a thrombotic tendency in many diseases. We studied NET formation in neutrophils derived from patients with chronic myeloid leukemia (CML) and in CML neutrophil cell lines and demonstrated that NETs are increased in CML and that certain drugs used to treat CML (tyrosine kinase inhibitors—TKIs) increase NET formation. These findings may shed light on a novel mechanism linking CML, TKIs and vascular toxicity. Abstract Cardiovascular complications are increasingly reported with the use of certain tyrosine kinase inhibitors (TKIs) to treat chronic myeloid leukemia (CML). We studied neutrophil extracellular trap (NET) formation in CML and evaluated the effect of TKIs on NET formation. Neutrophils isolated from treatment-naïve patients with CML showed a significant increase in NET formation compared to matched controls at baseline and after stimulation with ionomycin (IO) and phorbol 12-myristate 13-acetate (PMA). Expression of citrullinated histone H3 (H3cit), peptidyl arginine deiminase 4 (PAD4) and reactive oxygen species (ROS) was significantly higher in CML samples compared to controls. Pre-treatment of neutrophils with TKIs was associated with a differential effect on NET formation, and ponatinib significantly augmented NET-associated elastase and ROS levels as compared to controls and other TKIs. BCR-ABL1 retroviral transduced HoxB8-immortalized mouse hematopoietic progenitors, which differentiate into neutrophils in-vitro, demonstrated increased H3cit & myeloperoxidase (MPO) expression consistent with excess NET formation. This was inhibited by Cl-amidine, a PAD4 inhibitor, but not by the NADPH inhibitor diphenyleneiodonium (DPI). Ponatinib pre-exposure significantly increased H3cit expression in HoxB8-BCR-ABL1 cells after stimulation with IO. In summary, CML is associated with increased NET formation, which is augmented by ponatinib, suggesting a possible role for NETs in promoting vascular toxicity in CML.
Collapse
Affiliation(s)
- Alona Telerman
- Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Hospital, Petah-Tikva 4941492, Israel; (A.T.); (G.G.); (S.P.)
| | - Galit Granot
- Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Hospital, Petah-Tikva 4941492, Israel; (A.T.); (G.G.); (S.P.)
| | - Chiya Leibovitch
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Beilinson Hospital, Petah-Tikva 4941492, Israel
| | - Osnat Yarchovsky-Dolberg
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Meir Medical Center, Hematology Institute and Blood Bank, Kfar Saba 4428164, Israel
| | - Adi Shacham-Abulafia
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Beilinson Hospital, Petah-Tikva 4941492, Israel
| | - Shirly Partouche
- Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Hospital, Petah-Tikva 4941492, Israel; (A.T.); (G.G.); (S.P.)
| | - Moshe Yeshurun
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Beilinson Hospital, Petah-Tikva 4941492, Israel
| | - Martin H. Ellis
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Meir Medical Center, Hematology Institute and Blood Bank, Kfar Saba 4428164, Israel
| | - Pia Raanani
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Beilinson Hospital, Petah-Tikva 4941492, Israel
| | - Ofir Wolach
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 39040, Israel; (C.L.); (O.Y.-D.); (A.S.-A.); (M.Y.); (M.H.E.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Beilinson Hospital, Petah-Tikva 4941492, Israel
- Correspondence: ; Tel.: +972-50-406-5590
| |
Collapse
|
15
|
Cheng S, Jin P, Li H, Pei D, Shu X. Evaluation of CML TKI Induced Cardiovascular Toxicity and Development of Potential Rescue Strategies in a Zebrafish Model. Front Pharmacol 2021; 12:740529. [PMID: 34733159 PMCID: PMC8558359 DOI: 10.3389/fphar.2021.740529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/04/2021] [Indexed: 01/20/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) to BCR-ABL1 have been successfully used to treat chronic myeloid leukemia (CML), however, multiple TKI-associated adverse events have been reported and become an emerging problem in patients. The mechanisms of TKI-induced toxicity are not fully understood and it remains challenging to predict potential cardiovascular toxicity of a compound. In this study, we established a zebrafish model to evaluate potential in vivo cardiovascular toxicity of TKIs. We treated the endothelium labeled Tg(kdrl:EGFP) transgenic zebrafish embryos with TKIs then performed confocal imaging to evaluate their vascular structure and function. We found that among FDA approved CML TKIs, ponatinib (the only approved TKI that is efficacious to T315I mutation) is the most toxic one. We then evaluated safety profiles of several clinical stage kinase inhibitors that can target T315I and found that HQP1351 treatment leads to vasculopathies similar to those induced by ponatinib while the allosteric ABL inhibitor asciminib does not induce noticeable cardiovascular defects, indicating it could be a promising therapeutic reagent for patients with T315I mutation. We then performed proof-of-principle study to rescue those TKI-induced cardiovascular toxicities and found that, among commonly used anti-hypertensive drugs, angiotensin receptor blockers such as azilsartan and valsartan are able to reduce ponatinib or HQP1351 induced cardiovascular toxicities. Together, this study establishes a zebrafish model that can be useful to evaluate cardiovascular toxicity of TKIs as well as to develop strategies to minimize TKI-induced adverse events.
Collapse
Affiliation(s)
- Shan Cheng
- School of Life Science, Westlake University, Hangzhou, China.,CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Pan Jin
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
| | - Heying Li
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Duanqing Pei
- School of Life Science, Westlake University, Hangzhou, China.,CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong, Hong Kong, SAR China
| | - Xiaodong Shu
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, China
| |
Collapse
|
16
|
Chen MT, Huang ST, Lin CW, Ko BS, Chen WJ, Huang HH, Hsiao FY. Tyrosine Kinase Inhibitors and Vascular Adverse Events in Patients with Chronic Myeloid Leukemia: A Population-Based, Propensity Score-Matched Cohort Study. Oncologist 2021; 26:974-982. [PMID: 34418220 DOI: 10.1002/onco.13944] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/09/2021] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) have shown long-term survival benefits in patients with chronic myeloid leukemia (CML). Nevertheless, significant concern has been raised regarding long-term TKI-associated vascular adverse events (VAEs). The objective of this retrospective cohort study was to investigate the incidence of VAEs in Taiwanese patients with CML treated with different TKIs (imatinib, nilotinib, and dasatinib) as well as potential risk factors. MATERIALS AND METHODS We conducted a retrospective cohort study using the Taiwan Cancer Registry Database and National Health Insurance Research Database. Adult patients diagnosed with CML from 2008 to 2016 were identified and categorized into three groups according to their first-line TKI treatment (imatinib, nilotinib, and dasatinib). Propensity score matching was performed to control for potential confounders. Cox regressions were used to estimate the hazard ratio (HR) of VAEs in different TKI groups. RESULTS In total, 1,111 patients with CML were included in our study. We found that the risk of VAEs in nilotinib users was significantly higher than that in imatinib users, with an HR of 3.13 (95% confidence interval (CI), 1.30-7.51), whereas dasatinib users also showed a nonsignificant trend for developing VAEs, with an HR of 1.71 (95% CI, 0.71-4.26). In multivariable logistic regression analysis, only nilotinib usage, older age, and history of cerebrovascular diseases were identified as significant risk factors. The annual incidence rate of VAEs was highest within the first year after the initiation of TKIs. CONCLUSION These findings can support clinicians in making treatment decisions and monitoring VAEs in patients with CML in Taiwan. IMPLICATIONS FOR PRACTICE This study found that patients with chronic myeloid leukemia (CML) treated with nilotinib and dasatinib may be exposed to a higher risk of developing vascular adverse events (VAEs) compared with those treated with imatinib. Thus, this study suggests that patients with CML who are older or have a history of cerebrovascular diseases should be under close monitoring of VAEs, particularly within the first year after the initiation of tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Mei-Tsen Chen
- Graduate Institute of Clinical Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Shih-Tsung Huang
- Graduate Institute of Clinical Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Chih-Wan Lin
- Graduate Institute of Clinical Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Bor-Sheng Ko
- Division of Hematology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Wen-Jone Chen
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Huai-Hsuan Huang
- Division of Hematology, National Taiwan University Hospital, Taipei, Taiwan
| | - Fei-Yuan Hsiao
- Graduate Institute of Clinical Pharmacy, National Taiwan University, Taipei, Taiwan.,School of Pharmacy, National Taiwan University, Taipei, Taiwan.,Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
17
|
Side-effects profile and outcomes of ponatinib in the treatment of chronic myeloid leukemia. Blood Adv 2021; 4:530-538. [PMID: 32045474 DOI: 10.1182/bloodadvances.2019000268] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 01/06/2020] [Indexed: 12/20/2022] Open
Abstract
Ponatinib is associated with cardiovascular adverse events (CAEs), and its frequency in the real world is limited. In this retrospective study, we examined the survival outcomes and associated toxicities in 78 consecutive ponatinib-treated patients with chronic myeloid leukemia (CML) at the Moffitt Cancer Center from January 2011 through December 2017. The most common non-CAE was thrombocytopenia (39.7%), occurring in a dose-dependent fashion. Eighteen patients (23.1%) experienced some form of CAE, with the most common being arrhythmia (9%) and hypertension (7.7%), whereas 3 patients experienced myocardial infarction (3.8%). Before 2014, most patients were started on ponatinib 45 mg daily. There was an inverse correlation between cardio-oncology referral and the number of CAEs (P = .0440); however, a lower ponatinib starting dose, more frequent dose reduction, and increased cardio-oncology referral all were likely to have contributed to the observed decrease in CAEs after 2014. The response rate and 5-year overall survival (OS) were higher than those observed in the Ponatinib Ph+ ALL and CML Evaluation (PACE) trial (major molecular response, 58.7% vs 40% and OS, 76% vs 73%; median follow-up of 32.5 months). Ponatinib-treated patients with chronic phase-CML did not show a significant improvement with allogeneic stem cell transplantation, whereas those with accelerated phase/blast phase-CML had a much better outcome (median OS of 32.9 months vs 9.2 months; P = .01). These results demonstrate that ponatinib is highly effective. Dose adjustments and increased awareness of the cardiotoxicities associated with ponatinib may help maximize its benefits.
Collapse
|
18
|
Anti-Angiogenic Therapy: Current Challenges and Future Perspectives. Int J Mol Sci 2021; 22:ijms22073765. [PMID: 33916438 PMCID: PMC8038573 DOI: 10.3390/ijms22073765] [Citation(s) in RCA: 157] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 02/07/2023] Open
Abstract
Anti-angiogenic therapy is an old method to fight cancer that aims to abolish the nutrient and oxygen supply to the tumor cells through the decrease of the vascular network and the avoidance of new blood vessels formation. Most of the anti-angiogenic agents approved for cancer treatment rely on targeting vascular endothelial growth factor (VEGF) actions, as VEGF signaling is considered the main angiogenesis promotor. In addition to the control of angiogenesis, these drugs can potentiate immune therapy as VEGF also exhibits immunosuppressive functions. Despite the mechanistic rational that strongly supports the benefit of drugs to stop cancer progression, they revealed to be insufficient in most cases. We hypothesize that the rehabilitation of old drugs that interfere with mechanisms of angiogenesis related to tumor microenvironment might represent a promising strategy. In this review, we deepened research on the molecular mechanisms underlying anti-angiogenic strategies and their failure and went further into the alternative mechanisms that impact angiogenesis. We concluded that the combinatory targeting of alternative effectors of angiogenic pathways might be a putative solution for anti-angiogenic therapies.
Collapse
|
19
|
Abstract
Cancer therapies can lead to a broad spectrum of cardiovascular complications. Among these, cardiotoxicities remain of prime concern, but vascular toxicities have emerged as the second most common group. The range of cancer therapies with a vascular toxicity profile and the clinical spectrum of vascular toxic effects are quite broad. Historically, venous thromboembolism has received the greatest attention but, over the past decade, the arterial toxic effects, which can present as acute vasospasm, acute thrombosis and accelerated atherosclerosis, of cancer therapies have gained greater recognition. This Review focuses on these types of cancer therapy-related arterial toxicity, including their mechanisms, and provides an update on venous thromboembolism and pulmonary hypertension associated with cancer therapies. Recommendations for the screening, treatment and prevention of vascular toxic effects of cancer therapies are outlined in the context of available evidence and society guidelines and consensus statements. The shift towards greater awareness of the vascular toxic effects of cancer therapies has further unveiled the urgent needs in this area in terms of defining best clinical practices. Well-designed and well-conducted clinical studies and registries are needed to more precisely define the incidence rates, risk factors, primary and secondary modes of prevention, and best treatment modalities for vascular toxicities related to cancer therapies. These efforts should be complemented by preclinical studies to outline the pathophysiological concepts that can be translated into the clinic and to identify drugs with vascular toxicity potential even before their widespread clinical use.
Collapse
Affiliation(s)
- Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
20
|
Yang Y, Gao H, Sun X, Sun Y, Qiu Y, Weng Q, Rao Y. Global PROTAC Toolbox for Degrading BCR-ABL Overcomes Drug-Resistant Mutants and Adverse Effects. J Med Chem 2020; 63:8567-8583. [PMID: 32657579 DOI: 10.1021/acs.jmedchem.0c00967] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The BCR-ABL fusion oncoprotein causes chronic myeloid leukemia or acute lymphoblastic leukemia in Ph+ patients because the ABL kinase is constitutively activated. However, current clinical treatment with ABL inhibitors is seriously limited by drug resistance and adverse effects. Although the emerging proteolysis-targeting chimeras (PROTACs) have been introduced to degrade BCR-ABL, most of them showed limited activity and could not overcome the common drug-resistant mutants, especially for T315I mutant. Herein, we systematically designed a set of unique PROTACs by globally targeting all the three binding sites of BCR-ABL, including dasatinib-, ponatinib-, and asciminib-based PROTACs. Our ponatinib-based PROTACs showed practical activity as dasatinib-based PROTACs, while no reported ponatinib-based PROTACs could degrade BCR-ABL before. As a proof of concept, some additional dasatinib-based PROTACs were then designed to degrade T315I mutant too. We provided a global PROTAC toolbox for degrading both wild-type and T315I-mutated BCR-ABL from each binding site. More importantly, these PROTACs showed better selectivity and less adverse effects than the inhibitors, indicating that PROTACs had great potential for overcoming clinical drug resistance and safety issues.
Collapse
Affiliation(s)
- Yiqing Yang
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.,Tsinghua University-Peking University Joint Center for Life Sciences, Beijing 100084, China
| | - Hongying Gao
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.,Tsinghua University-Peking University Joint Center for Life Sciences, Beijing 100084, China
| | - Xiuyun Sun
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.,Tsinghua University-Peking University Joint Center for Life Sciences, Beijing 100084, China
| | - Yonghui Sun
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.,Tsinghua University-Peking University Joint Center for Life Sciences, Beijing 100084, China
| | - Yueping Qiu
- Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qinjie Weng
- Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Center for Drug Safety Evaluation and Research, Zhejiang University, Hangzhou 310058, China
| | - Yu Rao
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
21
|
Haguet H, Bouvy C, Delvigne AS, Modaffari E, Wannez A, Sonveaux P, Dogné JM, Douxfils J. The Risk of Arterial Thrombosis in Patients With Chronic Myeloid Leukemia Treated With Second and Third Generation BCR-ABL Tyrosine Kinase Inhibitors May Be Explained by Their Impact on Endothelial Cells: An In-Vitro Study. Front Pharmacol 2020; 11:1007. [PMID: 32719607 PMCID: PMC7350860 DOI: 10.3389/fphar.2020.01007] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
BCR-ABL tyrosine kinase inhibitors (TKIs) revolutionized the treatment of chronic myeloid leukemia, inducing deep molecular responses, largely improving patient survival and rendering treatment-free remission possible. However, three of the five BCR-ABL TKIs, dasatinib, nilotinib, and ponatinib, increase the risk of developing arterial thrombosis. Prior investigations reported that nilotinib and ponatinib affect the endothelium, but the mechanisms by which they exert their toxic effects are still unclear. The impact of dasatinib and bosutinib on endothelial cells has been poorly investigated. Here, we aimed to provide an in vitro homogenous evaluation of the effects of BCR-ABL TKIs on the endothelium, with a special focus on the type of cell death to elucidate the mechanisms responsible for the potential cytotoxic effects of BCR-ABL TKIs nilotinib and ponatinib on endothelial cells. We tested the five BCR-ABL TKIs at three concentrations on human umbilical venous endothelial cells (HUVECs). This study highlights the endothelial toxicity of ponatinib and provides insights about the mechanisms by which it affects endothelial cell viability. Ponatinib induced apoptosis and necrosis of HUVECs after 72 h. Dasatinib affected endothelial cells in vitro by inhibiting their proliferation and decreased wound closure as soon as 24 h of treatment and even at infra-therapeutic dose (0.005 µM). Comparatively, imatinib, nilotinib, and bosutinib had little impact on endothelial cells at therapeutic concentrations. They did not induce apoptosis nor necrosis, even after 72 h of treatment but they inhibited HUVEC proliferation. Overall, this study reports various effects of BCR-ABL TKIs on endothelial cells and suggests that ponatinib and dasatinib induce arterial thrombosis through endothelial dysfunction.
Collapse
Affiliation(s)
- Hélène Haguet
- Department of Pharmacy, Namur Thrombosis and Hemostasis Center (NTHC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | | | | | | | - Adeline Wannez
- Department of Pharmacy, Namur Thrombosis and Hemostasis Center (NTHC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Jean-Michel Dogné
- Department of Pharmacy, Namur Thrombosis and Hemostasis Center (NTHC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Jonathan Douxfils
- Department of Pharmacy, Namur Thrombosis and Hemostasis Center (NTHC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
- QUALIblood s.a., Namur, Belgium
| |
Collapse
|
22
|
Vascular endothelial growth factor-activated basophils in asthmatics. Postepy Dermatol Alergol 2020; 37:584-589. [PMID: 32994783 PMCID: PMC7507161 DOI: 10.5114/ada.2020.95954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 02/07/2019] [Indexed: 11/29/2022] Open
Abstract
Introduction Vascular endothelial growth factor (VEGF) is an angiogenic cytokine and a potential stimulator of permeability and lung neovascularization in asthmatics. It also plays an important role in the development of airway remodelling and in activation of many cells, including basophils. Aim To reveal the possible role of VEGF in the activation of basophils in asthmatics – subgroups with and without irreversible bronchoconstriction. Protein CD203c on the basophils surface was used as the activation marker. To define the possible pathway of basophils VEGF-activation, the influence of a genetic factor (polymorphism del18/ins -2549 -2567 in the VEGF-promoter region) was also considered. Material and methods Our study involved 82 patients with asthma (40 patients without and 42 patients with irreversible bronchoconstriction) and a group of 40 controls. The flow cytometric methods with anti-CD203c in the samples of basophils with increasing concentrations of VEGF was used for analysis of their activity. Genotyping for VEGF-promoter region was performed by PCR-based methods. Results Patients with asthma and del/del genotype showed more significant differences in the basophils activation after stimulation with increasing concentrations of VEGF than asthmatics with ins/ins and ins/del genotype (p = 0.023) and controls with del/del genotype (p = 0.0006). Conclusions Raised basophils VEGF-activation is characteristic for examined patients with asthma and might be associated with presence of polymorphism del18/ins -2549 -2567 in the VEGF-promoter region. Furthermore, it may contribute to the development of airways remodelling – this pathway has not been considered yet.
Collapse
|
23
|
Bisceglia I, Canale ML, Lestuzzi C, Parrini I, Russo G, Colivicchi F, Gabrielli D, Gulizia MM, Iliescu CA. Acute coronary syndromes in cancer patients. J Cardiovasc Med (Hagerstown) 2020; 21:944-952. [PMID: 32520859 DOI: 10.2459/jcm.0000000000000993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
: Cardiovascular disease and cancer are responsible for the majority of deaths in the Western world. These two entities share common risk factors and their intersection will be more and more frequent in future due to general population aging and long-term cancer control. Clinical presentation, management and outcome of acute coronary syndromes (ACS) in cancer patients could differ from noncancer ones. Cancer patients were frequently excluded from clinical trials and so the paucity of data further complicates the scenario. The management of ACS in cancer patients represents a unique setting in which the risk/benefit ratio of invasive treatment should be carefully evaluated. This review focused on the available evidence of all aspects of ACS in cancer patients providing a guide to a multidisciplinary approach.
Collapse
Affiliation(s)
- Irma Bisceglia
- Servizi Cardiologici Integrati Cardiology Department, Azienda Ospedaliera San Camillo-Forlanini, Roma
| | - Maria Laura Canale
- Division of Cardiology, Azienda USL Toscana Nord Ovest, Versilia Hospital, Lido di Camaiore
| | - Chiara Lestuzzi
- Cardiology Unit, Oncology Department, CRO National Cancer Institute, Aviano
| | - Iris Parrini
- Divisione di Cardiologia, Ospedale Mauriziano, Torino
| | - Giulia Russo
- SC Centro Cardiovascolare Ospedale Maggiore, Cardiology Department, Trieste
| | - Furio Colivicchi
- Division of Cardiology, San Filippo Neri Hospital, ASL Roma 1, Rome
| | | | | | - Cezar A Iliescu
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | |
Collapse
|
24
|
Singh AP, Umbarkar P, Tousif S, Lal H. Cardiotoxicity of the BCR-ABL1 tyrosine kinase inhibitors: Emphasis on ponatinib. Int J Cardiol 2020; 316:214-221. [PMID: 32470534 DOI: 10.1016/j.ijcard.2020.05.077] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/20/2020] [Accepted: 05/24/2020] [Indexed: 12/26/2022]
Abstract
The advent of tyrosine kinase inhibitors (TKIs) targeted therapy revolutionized the treatment of chronic myeloid leukemia (CML) patients. However, cardiotoxicity associated with these targeted therapies puts the cancer survivors at higher risk. Ponatinib is a third-generation TKI for the treatment of CML patients having gatekeeper mutation T315I, which is resistant to the first and second generation of TKIs, namely, imatinib, nilotinib, dasatinib, and bosutinib. Multiple unbiased screening from our lab and others have identified ponatinib as most cardiotoxic FDA approved TKI among the entire FDA approved TKI family (total 50+). Indeed, ponatinib is the only treatment option for CML patients with T315I mutation. This review focusses on the cardiovascular risks and mechanism/s associated with CML TKIs with a particular focus on ponatinib cardiotoxicity. We have summarized our recent findings with transgenic zebrafish line harboring BNP luciferase activity to demonstrate the cardiotoxic potential of ponatinib. Additionally, we will review the recent discoveries reported by our and other laboratories that ponatinib primarily exerts its cardiotoxicity via an off-target effect on cardiomyocyte prosurvival signaling pathways, AKT and ERK. Finally, we will shed light on future directions for minimizing the adverse sequelae associated with CML-TKIs.
Collapse
Affiliation(s)
- Anand Prakash Singh
- Division of Cardiovascular Disease, UAB
- The University of Alabama at Birmingham, Birmingham, AL 35294-1913, USA.
| | - Prachi Umbarkar
- Division of Cardiovascular Disease, UAB
- The University of Alabama at Birmingham, Birmingham, AL 35294-1913, USA
| | - Sultan Tousif
- Division of Cardiovascular Disease, UAB
- The University of Alabama at Birmingham, Birmingham, AL 35294-1913, USA
| | - Hind Lal
- Division of Cardiovascular Disease, UAB
- The University of Alabama at Birmingham, Birmingham, AL 35294-1913, USA.
| |
Collapse
|
25
|
Singh AP, Glennon MS, Umbarkar P, Gupte M, Galindo CL, Zhang Q, Force T, Becker JR, Lal H. Ponatinib-induced cardiotoxicity: delineating the signalling mechanisms and potential rescue strategies. Cardiovasc Res 2020; 115:966-977. [PMID: 30629146 DOI: 10.1093/cvr/cvz006] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/06/2018] [Accepted: 01/04/2019] [Indexed: 11/13/2022] Open
Abstract
AIMS Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of chronic myelogenous leukaemia (CML). However, cardiotoxicity of these agents remains a serious concern. The underlying mechanism of these adverse cardiac effects is largely unknown. Delineation of the underlying mechanisms of TKIs associated cardiac dysfunction could guide potential prevention strategies, rescue approaches, and future drug design. This study aimed to determine the cardiotoxic potential of approved CML TKIs, define the associated signalling mechanism and identify potential alternatives. METHODS AND RESULTS In this study, we employed a zebrafish transgenic BNP reporter line that expresses luciferase under control of the nppb promoter (nppb:F-Luciferase) to assess the cardiotoxicity of all approved CML TKIs. Our in vivo screen identified ponatinib as the most cardiotoxic agent among the approved CML TKIs. Then using a combination of zebrafish and isolated neonatal rat cardiomyocytes, we delineated the signalling mechanism of ponatinib-induced cardiotoxicity by demonstrating that ponatinib inhibits cardiac prosurvival signalling pathways AKT and extra-cellular-signal-regulated kinase (ERK), and induces cardiomyocyte apoptosis. As a proof of concept, we augmented AKT and ERK signalling by administration of Neuregulin-1β (NRG-1β), and this prevented ponatinib-induced cardiomyocyte apoptosis. We also demonstrate that ponatinib-induced cardiotoxicity is not mediated by inhibition of fibroblast growth factor signalling, a well-known target of ponatinib. Finally, our comparative profiling for the cardiotoxic potential of CML approved TKIs, identified asciminib (ABL001) as a potentially much less cardiotoxic treatment option for CML patients with the T315I mutation. CONCLUSION Herein, we used a combination of in vivo and in vitro methods to systematically screen CML TKIs for cardiotoxicity, identify novel molecular mechanisms for TKI cardiotoxicity, and identify less cardiotoxic alternatives.
Collapse
Affiliation(s)
- Anand P Singh
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB#348A, Nashville, TN, USA
| | - Michael S Glennon
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB#348A, Nashville, TN, USA.,Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, 200 Lothrop, BST E1258, Pittsburgh, PA, USA
| | - Prachi Umbarkar
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB#348A, Nashville, TN, USA
| | - Manisha Gupte
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB#348A, Nashville, TN, USA
| | - Cristi L Galindo
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB#348A, Nashville, TN, USA
| | - Qinkun Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB#348A, Nashville, TN, USA
| | - Thomas Force
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB#348A, Nashville, TN, USA
| | - Jason R Becker
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB#348A, Nashville, TN, USA.,Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, 200 Lothrop, BST E1258, Pittsburgh, PA, USA
| | - Hind Lal
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB#348A, Nashville, TN, USA
| |
Collapse
|
26
|
Zito C, Manganaro R, Carerj S, Antonini-Canterin F, Benedetto F. Peripheral Artery Disease and Stroke. J Cardiovasc Echogr 2020; 30:S17-S25. [PMID: 32566462 PMCID: PMC7293872 DOI: 10.4103/jcecho.jcecho_4_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/23/2019] [Accepted: 10/03/2019] [Indexed: 12/21/2022] Open
Abstract
Peripheral artery disease (PAD) and stroke can occur as vascular complication of anticancer treatment. Although the mechanisms, monitoring, and management of cardiotoxicities have received broad attention, vascular toxicities remain often underrecognized. In addition, the development of new chemotherapeutic drugs bears the risk of vasotoxicities that are yet to be identified and may not be realized with short-term follow-up periods. The propensity to develop PAD and/or stroke reflects the complex interplay between patient's baseline risk and preexisting vascular disease, particularly hypertension and diabetes, while evidence for genetic predisposition is increasing. Chemotherapeutic agents with a prominent vascular side effect profile have been identified. Interruption of vascular endothelial growth factor (VEGF) inhibitors (VEGFIs) signaling (i.e., bevacizumab) is associated with vascular toxicity and clinical sequelae such as hypertension, stroke, and thromboembolism beyond acute coronary syndromes. Cisplatin and 5-fluorouracil are the main drugs involved in the stroke risk. In addition, circulating concentrations of VEGF are reduced by cyclophosphamide administered at continuous low doses, which might underpin some of the observed vascular toxicity, such as stroke, as seen in patients treated with VEGF inhibitors. The risk of stroke is also increased after treatment with anthracyclines that can induce endothelial dysfunction and increase arterial stiffness. Proteasome inhibitors ( bortezomib and carfilzomib) and immunomodulatory agents (thalidomide, lenalidomide, and pomalidomide), approved for use in multiple myeloma, carry a black box warning for an increased risk of stroke. Finally, head-and-neck radiotherapy is associated with a doubled risk of cerebrovascular ischemic event, especially if exposure occurs in childhood. The mechanisms involved in radiation vasculopathy are represented by endothelial dysfunction, medial necrosis, fibrosis, and accelerated atherosclerosis. However, BCR-ABL tyrosine kinase inhibitor (TKI), used for the treatment of chronic myeloid leukemia (CML), is the main antineoplastic drugs involved in the development of PAD. In particular, second- and third-generation TKIs, such as nilotinib and ponatinib, while emerging as a potent arm in contrasting CML, are associated with a higher risk of PAD development rather than traditional imatinib. Factors favoring vascular complication are the presence of traditional cardiovascular risk factors (CVRF) and predisposing genetic factors, high doses of BCR-ABL TKIs, longer time of drug exposure, and sequential use of potent TKIs. Therefore, accurate cardiovascular risk stratification is strongly recommended in patient candidate to anticancer treatment associated with higher risk of vascular complication, in order to reduce the incidence of PAD and stroke through CVRF correction and selection of appropriate tailored patient strategy of treatment. Then, a clinical follow-up, eventually associated with instrumental evaluation through vascular ultrasound, should be performed.
Collapse
Affiliation(s)
- Concetta Zito
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, Azienda Ospedaliera Universitaria "Policlinico G. Martino", Messina, Italy
| | - Roberta Manganaro
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, Azienda Ospedaliera Universitaria "Policlinico G. Martino", Messina, Italy
| | - Scipione Carerj
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, Azienda Ospedaliera Universitaria "Policlinico G. Martino", Messina, Italy
| | | | - Frank Benedetto
- Cardiology Department, Hospital 'Bianchi Melacrino Morelli' Reggio Calabria, Italy
| |
Collapse
|
27
|
Mechanisms of Cardiovascular Toxicity of BCR-ABL1 Tyrosine Kinase Inhibitors in Chronic Myelogenous Leukemia. Curr Hematol Malig Rep 2020; 15:20-30. [DOI: 10.1007/s11899-020-00560-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
28
|
Abstract
The introduction of targeted agents into modern cancer therapy pursued the goal of molecularly more specific, and thereby more effective and safer, therapies. Paradoxically, however, several toxicities were brought to greater attention, among these not only cardiac but also vascular toxicities. The latter reach far beyond venous thromboembolism and include a broad spectrum of presentations based on the vascular territories and pathomechanisms involved, including abnormal vascular reactivity, acute thrombosis, or accelerated atherosclerosis. This article provides an overview of the most common presentations and their management strategies.
Collapse
Affiliation(s)
- Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN 55902, USA.
| |
Collapse
|
29
|
Valent P, Sadovnik I, Eisenwort G, Herrmann H, Bauer K, Mueller N, Sperr WR, Wicklein D, Schumacher U. Redistribution, homing and organ-invasion of neoplastic stem cells in myeloid neoplasms. Semin Cancer Biol 2019; 60:191-201. [PMID: 31408723 DOI: 10.1016/j.semcancer.2019.07.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023]
Abstract
The development of a myeloid neoplasm is a step-wise process that originates from leukemic stem cells (LSC) and includes pre-leukemic stages, overt leukemia and a drug-resistant terminal phase. Organ-invasion may occur in any stage, but is usually associated with advanced disease and a poor prognosis. Sometimes, extra-medullary organ invasion shows a metastasis-like or even sarcoma-like destructive growth of neoplastic cells in local tissue sites. Examples are myeloid sarcoma, mast cell sarcoma and localized blast phase of chronic myeloid leukemia. So far, little is known about mechanisms underlying re-distribution and extramedullary dissemination of LSC in myeloid neoplasms. In this article, we discuss mechanisms through which LSC can mobilize out of the bone marrow niche, can transmigrate from the blood stream into extramedullary organs, can invade local tissue sites and can potentially create or support the formation of local stem cell niches. In addition, we discuss strategies to interfere with LSC expansion and organ invasion by targeted drug therapies.
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria.
| | - Irina Sadovnik
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria
| | - Gregor Eisenwort
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria
| | - Harald Herrmann
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria; Department of Radiotherapy, Medical University of Vienna, Department of Medicine III, Austria
| | - Karin Bauer
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria
| | - Niklas Mueller
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Department of Internal Medicine III, Division of Hematology and Oncology, Hospital of the Ludwig-Maximilians-University Munich, Germany
| | - Wolfgang R Sperr
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria
| | - Daniel Wicklein
- Institute of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
30
|
Hirschbuehl K, Rank A, Pfeiffer T, Schaller T, Haeckel T, Roeling J, Vlasenko D, Trepel M, Schmid C. Large Vessel Vasculitis as a Possible Mechanism of Vascular Side Effects of Ponatinib: A Case Report. J Hematol 2019; 8:83-85. [PMID: 32300450 PMCID: PMC7153681 DOI: 10.14740/jh519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/07/2019] [Indexed: 11/25/2022] Open
Abstract
Arterial occlusive events (AOEs) such as cerebrovascular, cardiovascular and peripheral arterial events are known side effects of ponatinib, assumed due to the rapid development and increase of arteriosclerosis, while the definitive pathomechanisms therefore are still unclear. We present a case of clinically apparent large vessel vasculitis and discuss this phenomenon as a possible mechanism of AOEs beside arteriosclerosis.
Collapse
Affiliation(s)
- Klaus Hirschbuehl
- Department of Hematology and Oncology, Universitaetsklinikum Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Andreas Rank
- Department of Hematology and Oncology, Universitaetsklinikum Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Tim Pfeiffer
- Department of Hematology and Oncology, Universitaetsklinikum Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Tina Schaller
- Institute of Pathology, Universitaetsklinikum Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Thomas Haeckel
- Department of Radiology, Universitaetsklinikum Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Joerg Roeling
- Internistische Gemeinschaftspraxis am Vincentinum, Franziskanergasse 14, 86152 Augsburg, Germany
| | - Dmytro Vlasenko
- Department of Visceral Surgery, Universitaetsklinikum Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Martin Trepel
- Department of Hematology and Oncology, Universitaetsklinikum Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Christoph Schmid
- Department of Hematology and Oncology, Universitaetsklinikum Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| |
Collapse
|
31
|
A phosphoproteomic signature in endothelial cells predicts vascular toxicity of tyrosine kinase inhibitors used in CML. Blood Adv 2019; 2:1680-1684. [PMID: 30021779 DOI: 10.1182/bloodadvances.2018020396] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/26/2018] [Indexed: 12/11/2022] Open
Abstract
Key Points
Newer CML kinase inhibitors increase ischemia risk and are toxic to endothelial cells where they produce a proteomic toxicity signature. This phosphoproteomic EC toxicity signature predicts bosutinib to be safe, providing a potential screening tool for safer drug development.
Collapse
|
32
|
Tan FH, Putoczki TL, Stylli SS, Luwor RB. Ponatinib: a novel multi-tyrosine kinase inhibitor against human malignancies. Onco Targets Ther 2019; 12:635-645. [PMID: 30705592 PMCID: PMC6343508 DOI: 10.2147/ott.s189391] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Human malignancies are often the result of overexpressed and constitutively active receptor and non-receptor tyrosine kinases, which ultimately lead to the mediation of key tumor-driven pathways. Several tyrosine kinases (ie, EGFR, FGFR, PDGFR, VEGFR), are aberrantly activated in most common tumors, including leukemia, glioblastoma, gastrointestinal stromal tumors, non-small-cell lung cancer, and head and neck cancers. Iclusig™ (ponatinib, previously known as AP24534) is an orally active multi-tyrosine kinase inhibitor and is currently approved by the US Food and Drug Administration for patients with chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia, specifically targeting the BCR-ABL gene mutation, T315I. Due to ponatinib's unique multi-targeted characteristics, further studies have demonstrated its ability to target other important tyrosine kinases (FGFR, PDGFR, SRC, RET, KIT, and FLT1) in other human malignancies. This review focuses on the available data of ponatinib and its molecular targets for treatment in various cancers, with a discussion on the broader potential of this agent in other cancer indications.
Collapse
Affiliation(s)
- Fiona H Tan
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia, .,Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Tracy L Putoczki
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia, .,Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC 3050, Australia
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia, .,Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Rodney B Luwor
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia,
| |
Collapse
|
33
|
Oren O, Herrmann J. Arterial events in cancer patients-the case of acute coronary thrombosis. J Thorac Dis 2018; 10:S4367-S4385. [PMID: 30701104 PMCID: PMC6328398 DOI: 10.21037/jtd.2018.12.79] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/14/2018] [Indexed: 12/21/2022]
Abstract
Patients with cancer are at high risk for both venous and arterial thrombotic complications. A variety of factors account for the greater thrombotic risk, including the underlying malignancy and numerous cancer-directed therapies. The occurrence of an acute thrombotic event in patients with cancer is associated with substantial morbidity and mortality. Acute coronary syndrome (ACS) represents a particularly important cardiovascular complication in cancer patients. With cardio-vascular risk factors becoming more prevalent in an aging cancer population that is surviving longer, questions pertaining to the appropriate management of vascular toxicity are likely to assume even greater value in the coming years. In this article, we review the current understanding of ACS in patients with cancer. The predisposition to thrombosis in a malignant host and the cancer treatments most commonly associated with vascular toxicity are reviewed. Risk prediction and management strategies are discussed, and discrepancies in the clinical evidence are highlighted.
Collapse
Affiliation(s)
- Ohad Oren
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
34
|
Paez-Mayorga J, Chen AL, Kotla S, Tao Y, Abe RJ, He ED, Danysh BP, Hofmann MCC, Le NT. Ponatinib Activates an Inflammatory Response in Endothelial Cells via ERK5 SUMOylation. Front Cardiovasc Med 2018; 5:125. [PMID: 30238007 PMCID: PMC6135907 DOI: 10.3389/fcvm.2018.00125] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/20/2018] [Indexed: 12/18/2022] Open
Abstract
Ponatinib is a multi-targeted third generation tyrosine kinase inhibitor (TKI) used in the treatment of chronic myeloid leukemia (CML) patients harboring the Abelson (Abl)-breakpoint cluster region (Bcr) T315I mutation. In spite of having superb clinical efficacy, ponatinib triggers severe vascular adverse events (VAEs) that significantly limit its therapeutic potential. On vascular endothelial cells (ECs), ponatinib promotes EC dysfunction and apoptosis, and inhibits angiogenesis. Furthermore, ponatinib-mediated anti-angiogenic effect has been suggested to play a partial role in systemic and pulmonary hypertension via inhibition of vascular endothelial growth factor receptor 2 (VEGFR2). Even though ponatinib-associated VAEs are well documented, their etiology remains largely unknown, making it difficult to efficiently counteract treatment-related adversities. Therefore, a better understanding of the mechanisms by which ponatinib mediates VAEs is critical. In cultured human aortic ECs (HAECs) treated with ponatinib, we found an increase in nuclear factor NF-kB/p65 phosphorylation and NF-kB activity, inflammatory gene expression, cell permeability, and cell apoptosis. Mechanistically, ponatinib abolished extracellular signal-regulated kinase 5 (ERK5) transcriptional activity even under activation by its upstream kinase mitogen-activated protein kinase kinase 5α (CA-MEK5α). Ponatinib also diminished expression of ERK5 responsive genes such as Krüppel-like Factor 2/4 (klf2/4) and eNOS. Because ERK5 SUMOylation counteracts its transcriptional activity, we examined the effect of ponatinib on ERK5 SUMOylation, and found that ERK5 SUMOylation is increased by ponatinib. We also found that ponatibib-mediated increased inflammatory gene expression and decreased anti-inflammatory gene expression were reversed when ERK5 SUMOylation was inhibited endogenously or exogenously. Overall, we propose a novel mechanism by which ponatinib up-regulates endothelial ERK5 SUMOylation and shifts ECs to an inflammatory phenotype, disrupting vascular homeostasis.
Collapse
Affiliation(s)
- Jesus Paez-Mayorga
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| | - Andrew L. Chen
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yunting Tao
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
| | - Rei J. Abe
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
| | - Emma D. He
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
| | - Brian P. Danysh
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marie-Claude C. Hofmann
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
| |
Collapse
|
35
|
Pouwer MG, Pieterman EJ, Verschuren L, Caspers MPM, Kluft C, Garcia RA, Aman J, Jukema JW, Princen HMG. The BCR-ABL1 Inhibitors Imatinib and Ponatinib Decrease Plasma Cholesterol and Atherosclerosis, and Nilotinib and Ponatinib Activate Coagulation in a Translational Mouse Model. Front Cardiovasc Med 2018; 5:55. [PMID: 29946549 PMCID: PMC6005845 DOI: 10.3389/fcvm.2018.00055] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/10/2018] [Indexed: 01/16/2023] Open
Abstract
Treatment with the second and third generation BCR-ABL1 tyrosine kinase inhibitors (TKIs) increases cardiovascular risk in chronic myeloid leukemia (CML) patients. We investigated the vascular adverse effects of three generations of TKIs in a translational model for atherosclerosis, the APOE*3Leiden.CETP mouse. Mice were treated for sixteen weeks with imatinib (150 mg/kg BID), nilotinib (10 and 30 mg/kg QD) or ponatinib (3 and 10 mg/kg QD), giving similar drug exposures as in CML-patients. Cardiovascular risk factors were analyzed longitudinally, and histopathological analysis of atherosclerosis and transcriptome analysis of the liver was performed. Imatinib and ponatinib decreased plasma cholesterol (imatinib, −69%, p < 0.001; ponatinib 3 mg/kg, −37%, p < 0.001; ponatinib 10 mg/kg−44%, p < 0.001) and atherosclerotic lesion area (imatinib, −78%, p < 0.001; ponatinib 3 mg/kg, −52%, p = 0.002; ponatinib 10 mg/kg, −48%, p = 0.006), which were not affected by nilotinib. In addition, imatinib increased plaque stability. Gene expression and pathway analysis demonstrated that ponatinib enhanced the mRNA expression of coagulation factors of both the contact activation (intrinsic) and tissue factor (extrinsic) pathways. In line with this, ponatinib enhanced plasma levels of FVII, whereas nilotinib increased plasma FVIIa activity. While imatinib showed a beneficial cardiovascular risk profile, nilotinib and ponatinib increased the cardiovascular risk through induction of a pro-thrombotic state.
Collapse
Affiliation(s)
- Marianne G Pouwer
- Metabolic Health Research, Gaubius Laboratory, The Netherlands Organization of Applied Scientific Research (TNO), Leiden, Netherlands.,Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Elsbet J Pieterman
- Metabolic Health Research, Gaubius Laboratory, The Netherlands Organization of Applied Scientific Research (TNO), Leiden, Netherlands
| | - Lars Verschuren
- Microbiology and Systems Biology, The Netherlands Organization of Applied Scientific Research (TNO), Zeist, Netherlands
| | - Martien P M Caspers
- Microbiology and Systems Biology, The Netherlands Organization of Applied Scientific Research (TNO), Zeist, Netherlands
| | | | - Ricardo A Garcia
- Cardiovascular Drug Discovery, Bristol-Meyers Squibb, New York, United States
| | - Jurjan Aman
- Departments of Physiology and Pulmonary Diseases, VU University Medical Center, Amsterdam, Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Hans M G Princen
- Metabolic Health Research, Gaubius Laboratory, The Netherlands Organization of Applied Scientific Research (TNO), Leiden, Netherlands
| |
Collapse
|
36
|
Gover-Proaktor A, Granot G, Pasmanik-Chor M, Pasvolsky O, Shapira S, Raz O, Raanani P, Leader A. Bosutinib, dasatinib, imatinib, nilotinib, and ponatinib differentially affect the vascular molecular pathways and functionality of human endothelial cells. Leuk Lymphoma 2018; 60:189-199. [PMID: 29741440 DOI: 10.1080/10428194.2018.1466294] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The tyrosine kinase inhibitors (TKIs), nilotinib, ponatinib, and dasatinib (but not bosutinib or imatinib), are associated with vascular adverse events (VAEs) in chronic myeloid leukemia (CML). Though the mechanism is inadequately understood, an effect on vascular cells has been suggested. We investigated the effect of imatinib, nilotinib, dasatinib, bosutinib, and ponatinib on tube formation, cell viability, and gene expression of human vascular endothelial cells (HUVECs). We found a distinct genetic profile in HUVECs treated with dasatinib, ponatinib, and nilotinib compared to bosutinib and imatinib, who resembled untreated samples. However, unique gene expression and molecular pathway alterations were detected between dasatinib, ponatinib, and nilotinib. Angiogenesis/blood vessel-related pathways and HUVEC function (tube formation/viability) were adversely affected by dasatinib, ponatinib, and nilotinib but not by imatinib or bosutinib. These results correspond to the differences in VAE profiles of these TKIs, support a direct effect on vascular cells, and provide direction for future research.
Collapse
Affiliation(s)
- Ayala Gover-Proaktor
- a Felsenstein Medical Research Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel
| | - Galit Granot
- a Felsenstein Medical Research Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel
| | - Metsada Pasmanik-Chor
- b Bioinformatics Unit, G.S.W. Faculty of Life Sciences , Tel Aviv University , Tel Aviv , Israel
| | - Oren Pasvolsky
- c Institute of Hematology, Davidoff Cancer Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel.,d Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| | - Saar Shapira
- a Felsenstein Medical Research Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel.,d Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| | - Oshrat Raz
- a Felsenstein Medical Research Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel
| | - Pia Raanani
- c Institute of Hematology, Davidoff Cancer Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel.,d Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| | - Avi Leader
- c Institute of Hematology, Davidoff Cancer Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel.,d Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
37
|
Haguet H, Douxfils J, Chatelain C, Graux C, Mullier F, Dogné JM. BCR-ABL Tyrosine Kinase Inhibitors: Which Mechanism(s) May Explain the Risk of Thrombosis? TH OPEN 2018; 2:e68-e88. [PMID: 31249931 PMCID: PMC6524858 DOI: 10.1055/s-0038-1624566] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 11/27/2017] [Indexed: 12/12/2022] Open
Abstract
Imatinib, the first-in-class BCR-ABL tyrosine kinase inhibitor (TKI), had been a revolution for the treatment of chronic myeloid leukemia (CML) and had greatly enhanced patient survival. Second- (dasatinib, nilotinib, and bosutinib) and third-generation (ponatinib) TKIs have been developed to be effective against BCR-ABL mutations making imatinib less effective. However, these treatments have been associated with arterial occlusive events. This review gathers clinical data and experiments about the pathophysiology of these arterial occlusive events with BCR-ABL TKIs. Imatinib is associated with very low rates of thrombosis, suggesting a potentially protecting cardiovascular effect of this treatment in patients with BCR-ABL CML. This protective effect might be mediated by decreased platelet secretion and activation, decreased leukocyte recruitment, and anti-inflammatory or antifibrotic effects. Clinical data have guided mechanistic studies toward alteration of platelet functions and atherosclerosis development, which might be secondary to metabolism impairment. Dasatinib, nilotinib, and ponatinib affect endothelial cells and might induce atherogenesis through increased vascular permeability. Nilotinib also impairs platelet functions and induces hyperglycemia and dyslipidemia that might contribute to atherosclerosis development. Description of the pathophysiology of arterial thrombotic events is necessary to implement risk minimization strategies.
Collapse
Affiliation(s)
- Hélène Haguet
- University of Namur, Namur Thrombosis and Hemostasis Center (NTHC), Namur Research Institute for Life Sciences (NARILIS), Department of Pharmacy, Namur, Belgium
- Université catholique de Louvain, CHU UCL Namur, Namur Thrombosis and Hemostasis Center, Hematology Laboratory, Yvoir, Belgium
| | - Jonathan Douxfils
- University of Namur, Namur Thrombosis and Hemostasis Center (NTHC), Namur Research Institute for Life Sciences (NARILIS), Department of Pharmacy, Namur, Belgium
- QUALIblood s.a., Namur, Belgium
| | - Christian Chatelain
- University of Namur, Namur Thrombosis and Hemostasis Center (NTHC), Namur Research Institute for Life Sciences (NARILIS), Department of Pharmacy, Namur, Belgium
| | - Carlos Graux
- Université catholique de Louvain, CHU UCL Namur, Namur Thrombosis and Hemostasis Center, Department of Hematology, Yvoir, Belgium
| | - François Mullier
- Université catholique de Louvain, CHU UCL Namur, Namur Thrombosis and Hemostasis Center, Hematology Laboratory, Yvoir, Belgium
| | - Jean-Michel Dogné
- University of Namur, Namur Thrombosis and Hemostasis Center (NTHC), Namur Research Institute for Life Sciences (NARILIS), Department of Pharmacy, Namur, Belgium
| |
Collapse
|
38
|
Cheng Z, Fu J, Liu G, Zhang L, Xu Q, Wang SY. Angiogenesis in JAK2 V617F positive myeloproliferative neoplasms and ruxolitinib decrease VEGF, HIF-1 enesis in JAK2 V617F positive cells. Leuk Lymphoma 2017; 59:196-203. [PMID: 28554272 DOI: 10.1080/10428194.2017.1324155] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Angiogenesis and JAK2 V617F mutation are common in BCR-ABL1 negative myeloproliferative neoplasms (MPNs). Ruxolitinib, a JAK inhibitor, is an effective treatment for some MPNs. However, the relationship between angiogenesis and JAK2 V617F and the effects of ruxolitinib on angiogenesis are still unknown. Here, we observed the correlation of JAK2 V617F mutation burden with VEGF, HIF-1a and microvascular density (MVD) in MPNs. We investigate the effect of ruxolitinib on the expression of VEGF and HIF-1α in JAK2 V617F positive cells. We found the expression levels of p-JAK2, VEGF, HIF-1a and MVD in the newly diagnosed MPNs were significantly increased and were related to the JAK2 V617F burden. Ruxolitinib can inhibit p-JAK2, VEGF, HIF-1a expression and suppress blood vessels' formation in chick embryo choriallantoic membrane. Our findings indicated that angiogenesis is related to JAK2 V617F burden and ruxolitinib could decrease VEGF and HIF-1a expression in JAK2 V617F positive cells.
Collapse
Affiliation(s)
- Zhiyong Cheng
- a Department of Hematology , The No. 1 Hospital of Baoding , Baoding , China
| | - Jianzhu Fu
- a Department of Hematology , The No. 1 Hospital of Baoding , Baoding , China
| | - Guimin Liu
- a Department of Hematology , The No. 1 Hospital of Baoding , Baoding , China
| | - Lijun Zhang
- a Department of Hematology , The No. 1 Hospital of Baoding , Baoding , China
| | - Qian Xu
- a Department of Hematology , The No. 1 Hospital of Baoding , Baoding , China
| | - Su-Yun Wang
- b Department of Hematology , Hebei General Hospital , Shijiazhuang , Hebei Province , China
| |
Collapse
|
39
|
Risk factors and mechanisms contributing to TKI-induced vascular events in patients with CML. Leuk Res 2017; 59:47-54. [PMID: 28549238 DOI: 10.1016/j.leukres.2017.05.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/18/2022]
Abstract
Vascular adverse events (VAE) are an emerging problem in patients with chronic myeloid leukemia (CML) receiving second-generation BCR-ABL1 tyrosine kinase inhibitors (TKI). Relevant VAE comprise peripheral, cerebral, and coronary artery changes in patients receiving nilotinib, venous and arterial occlusive events during ponatinib therapy, and pulmonary hypertension in patients receiving dasatinib. Although each TKI binds to a unique profile of molecular targets in leukemic cells and vascular cells, the exact etiology of drug-induced vasculopathies remains uncertain. Recent data suggest that predisposing molecular factors, pre-existing cardiovascular risk factors as well as certain comorbidities contribute to the etiology of VAE in these patients. In addition, direct effects of these TKI on vascular endothelial cells have been demonstrated and are considered to contribute essentially to VAE evolution. In the current article, we discuss mechanisms underlying the occurrence of VAE in TKI-treated patients with CML, with special emphasis on vascular and perivascular target cells and involved molecular (vascular) targets of VAE-triggering TKI. In addition, we discuss optimal patient selection and drug selection through which the risk of occurrence of cardiovascular events can hopefully be minimized while maintaining optimal anti-leukemic effects in CML, thereby following the principles of personalized medicine.
Collapse
|
40
|
Molica M, Massaro F, Breccia M. Second line small molecule therapy options for treating chronic myeloid leukemia. Expert Opin Pharmacother 2016; 18:57-65. [DOI: 10.1080/14656566.2016.1267141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Matteo Molica
- Department of Cellular Biotechnologies and Hematology, Sapienza University, Rome, Italy
| | - Fulvio Massaro
- Department of Cellular Biotechnologies and Hematology, Sapienza University, Rome, Italy
| | - Massimo Breccia
- Department of Cellular Biotechnologies and Hematology, Sapienza University, Rome, Italy
| |
Collapse
|