Pathogenesis of Ponatinib Associated Vascular Disease in Chronic Myeloid Leukemia: An in Vitro Study

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2798-2798 ◽  
Author(s):  
Ayala Gover-Proaktor ◽  
Oren Pasvolsky ◽  
Pia Raanani ◽  
Arnon Nagler ◽  
Saar Shapira ◽  
...  

Abstract The introduction of several tyrosine kinase inhibitors (TKIs) into the armamentarium of chronic myeloid leukemia (CML) treatment has revolutionized the prognosis of this disease, changing it from a fatal one into a potentially curable disease. Five TKIs are now in clinical use: imatinib, nilotinib, dasatinib, bosutinib and ponatinib. This allows the clinician the privilege to personalize treatment based on the toxicity profile of the various TKIs, taking into account patients' comorbidities and lifestyle. Although side effects are common to all TKIs, each one has its unique safety profile. Consequently, one of the main challenges in the contemporary management and research of CML is the understanding and control of TKI-associated adverse events. TKI-associated vascular disease is amongst the most perturbing and poorly understood of these adverse events. Nilotinib is probably associated with an increased risk of arterial vascular events, especially peripheral artery occlusive disease (PAOD) compared with imatinib. Conversely, the notorious effect of nilotinib on PAOD, manifested in comparative clinical trials, might be attributed to a protective effect of imatinib. Similar data has started to emerge regarding ponatinib, including fatal cerebrovascular events, ranging from 20-42% of patients. These side effects of ponatinib occur after as little as 8 months of treatment. The mechanism of this increased tendency for vascular events associated with specific TKIs, remains incompletely understood, and there has been only limited research exploring the possible pathophysiological mechanisms involved. We therefore initiated a preclinical in vitro study in order to ascertain a potential molecular mechanism responsible for these adverse vascular events. Specifically, we studied the effect of nilotinib and ponatinib on human umbilical vein endothelial cells (HUVEC) in comparison to that of imatinib. When examining pharmacologically relevant concentrations (5.3µM imatinib, 4.3µM nilotinib and 0.11µM or 0.17µM ponatinib), annexin/PI staining showed that following a 24 hour exposure period, imatinib, nilotinib and ponatinib did not lead to an increase in apoptosis (DMSO (control): 8.8%±2.4; imatinib: 13.2%±1.2; nilotinib: 9.6%±0.6; ponatinib 11.1%±1.8, p-value not statistically significant). However, under these same conditions, ponatinib dramatically inhibited angiogenesis of HUVECs in a tube-formation assay. This inhibition was evident even when exposing the cells to the lower physiological concentrations of the drug (0.11µM). Relative average tube area (calculated relative to control) was as follows: DMSO (control): 1; imatinib: 0.86±0.3; nilotinib: 1±0.5; 0.11µM ponatinib (equivalent to 30mg/day): 0.4±0.1 p-value<0.05; 0.17µM ponatinib (equivalent to 45mg/day): 0.37±0.1 p-value<0.05 (Fig. 1). First signs of reduction in average tube area following ponatinib treatment were detected as early as 4 hours post exposure to the drug. This type of early effect suggests that ponatinib might inhibit initial tube formation rather than cause the tubes to dissociate after they have been formed. These results indicate that ponatinib exerts a suppressive effect on neo-angiogenesis of vascular endothelial cells. Angiogenesis is characterized by the appearance of capillarized vascular structures through enhanced expression of pro-angiogenic factors, such as VEGFR and FGFR. To the best of our knowledge, this is the first report regarding ponatinib's impact on angiogenesis in vitro. Since the suppressive effect of ponatinib on angiogenesis of HUVECs was not a result of apoptosis, it might be mediated through VEGFR or FGFR, as both are ponatinib's off-targets. An understanding of the pathophysiology of the vascular events may help guide monitoring, allowing for earlier interventions and management of vascular risk factors. In addition, it will contribute to the identification of potential targets for intervention and/or prevention in a significant minority of patients who have no other treatment options and ensure that researchers developing next generation TKIs avoid targeting the same pathways. Figure 1. Ponatinib inhibits HUVEC tube formation. HUVECs were plated on Matrigel-coated plates in presence of DMSO, ponatinib, nilotinib or imatinib. Tube formation was evaluated after 4 and 24 h by light microscope. Representative fields (ª4 magnification) are pictured. Figure 1. Ponatinib inhibits HUVEC tube formation. HUVECs were plated on Matrigel-coated plates in presence of DMSO, ponatinib, nilotinib or imatinib. Tube formation was evaluated after 4 and 24 h by light microscope. Representative fields (ª4 magnification) are pictured. Disclosures Raanani: Novartis: Consultancy, Research Funding; Pfizer: Consultancy; Ariad: Consultancy, Research Funding; BMS: Consultancy. Leader:Novartis: Research Funding.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1883-1883 ◽  
Author(s):  
Emir Hadzijusufovic ◽  
Rudolf Kirchmair ◽  
Markus Theurl ◽  
Susanne Gamperl ◽  
Daniela Lener ◽  
...  

Abstract The potent BCR-ABL1-targeting tyrosine kinase inhibitor (TKI) ponatinib is used for the treatment of patients with drug-resistant chronic myeloid leukemia (CML). However, an increased risk of development of cardiovascular events has been described in CML patients treated with ponatinib. The etiology of these adverse events is currently unknown. In an attempt to discover mechanisms underlying ponatinib-induced adverse vascular events, we have evaluated the effects of ponatinib in vitro on human vascular endothelial cells and on contraction of explanted mice aortic rings. In addition, we examined the effects of ponatinib on angiogenesis in vivo in a mouse model of hind limb ischemia. Ponatinib dose-dependently induced apoptosis in human coronary artery endothelial cells (HCAEC) in a caspase assay (relative apoptosis vs. 1% DMSO: ponatinib 50 nM: 1.79±0.38, p<0.001; ponatinib 100 nM: 2.13±0.42, p<0.001) and this drug effect could partially be blocked by addition of insulin (ponatinib 100 nM + insulin 5 µg/ml: 1.70±0.18, p<0.05). In addition, ponatinib was found to inhibit the proliferation of human umbilical vein endothelial cells (HUVEC) and the human microvascular endothelial cell line HMEC-1, with IC50 values ranging between 100 and 250 nM (p<0.05) as determined by thymidine-incorporation assay. Using a phospho-receptor tyrosine kinase assay in HCAEC, we found that ponatinib also inhibits fetal bovine serum-induced phosphorylation of the VEGF receptor KDR as well as phosphorylation of MER and insulin receptors, which play a role in angiogenesis, vascular homeostasis, and vessel protection. We also found that ponatinib (1 µM, 4 hours) increases adhesion of HUVEC to a plastic-surface compared to DMSO control (Figure). Based on clinical observations of vasoconstriction in ponatinib-treated patients, we also applied ponatinib on aortic rings harvested from C57BL/6 mice. Ponatinib (100 nM, overnight) enhanced norepinephrine-induced vasoconstriction (log EC50: control -7.76±0.06 vs. ponatinib -7.96±0.05, p<0.05, n=6) and inhibited acetylcholine-mediated vasodilatation (log IC50: control -7.45±0.05 vs. ponatinib -7.06±0.1, p<0.001) as shown by myography. These drug effects were blocked by inhibition of nitric oxide (using nitric oxide synthase inhibitor L-NNA, 100 µM) or COX (by applying diclofenac, 3 mg/l), suggesting that ponatinib promotes the generation of vasoconstricting prostanoids. Ponatinib effects were also blocked by the calcium channel blocker nifedipine (1 µM). In C57BL/6 mice, ponatinib (5 mg/kg/day for 35 days) was found to inhibit blood flow recovery in a hind limb ischemia model as shown by Laser-Doppler perfusion imaging after femoral artery ligation. The blood perfusion ratios of the ischemic limb vs. non-ischemic limb at week 5 were: control group: 0.67±0.07 vs. ponatinib: 0.56±0.1; p<0.05). Ponatinib-treated mice also developed toe and foot necrosis more frequently than control mice (necrosis score: control: 0.3 vs. ponatinib: 1.3). In summary, ponatinib affects endothelial cell growth and vasomotor function in-vitro as well as blood flow recovery in a mouse model. These findings might help explain the occurrence of vascular events in CML patients treated with ponatinib and may lead to development of therapeutic strategies for prevention and treatment of ponatinib-induced adverse events. Figure Figure. Disclosures Kirchmair: Ariad: Research Funding. Valent:Ariad: Honoraria, Research Funding; Amgen: Honoraria; Deciphera Pharmaceuticals: Research Funding; Novartis: Honoraria, Research Funding; Celgene: Honoraria, Research Funding.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3934-3934
Author(s):  
Abdel Kareem Azab ◽  
Feda Azab ◽  
Phong Quang ◽  
Patricia Maiso ◽  
Hai T Ngo ◽  
...  

Abstract Abstract 3934 INTRODUCTION: The interaction of multiple myeloma (MM) cells with the bone marrow (BM) microenvironment plays a crucial role in MM pathogenesis. The BM microenvironment in MM is characterized by an increased micro-vessel density and increased secretion of angiogenic factors. CXCR7 is a G-protein coupled receptor shown to play a major role in the adhesion, migration and angiogenesis of endothelial cells (ECs). Our interest is in the role of CXCR7 in cell trafficking of ECs and EPCs in MM. Thus we characterized ECs and EPCs from MM patients and MM animal models and examined the contribution of CXCR7 to the cell trafficking using in vitro and in vivo assays and using CXCR7-selective compound. METHODS AND RESULTS: We used flow cytometry to detect the frequency of ECs and EPCs in the BM and peripheral blood (PB) of 5 MM patients and 5 normal subjects. ECs were detected as VEGFR+ CD133- cells, while EPCs were detected as VEGFR+ CD133+ cells. MM patients had significantly higher numbers of ECs and EPCs compared to healthy donors in both the BM and the PB. These results were confirmed in a mouse model of MM in which MM cells or vehicle were injected to SCID mice and the frequency of ECs and EPCs in the BM and the PB was determined 4 weeks after injection. We found that in mice with MM significantly higher numbers of ECs and EPCs could be detected in both the BM and the PB than in control mice. CXCR7 was expressed on both ECs and EPCs isolated from MM patients, healthy donors, and control mice. The expression of CXCR7 on EPCs was higher than the expression on ECs. The expression of CXCR7 on ECs and EPCs isolated from the BM was higher than the expression on ECs and EPCs isolated from the PB, respectively. Therefore, to test the role of CXCR7 in cell-trafficking of ECs and EPCs, we injected 10mg/kg of CXCR7 inhibitor POL6926, a potent and selective CXCR7 antagonist based on the Protein Epitope Mimetics (PEM) Technology (Polyphor, Switzerland), to BALB/c mice and tested the frequency of ECs and EPC in the PB and BM of the mice at 0, 2, 4 and 24 hours after the injection. We found a 3-fold increase in ECs and 6-fold increase in EPCs in the PB; 2 hrs post the injection of the CXCR7 antagonist. The levels of EPCs in the PB returned to baseline at 4 and 24 hrs, while the level of ECs was maintained at 4hrs and went back to baseline at 24 hrs. No significant differences were found in the frequency of ECs and EPCs in the BM after the injection of the CXCR7 antagonist. To investigate the function of CXCR7 in ECs in vitro we used human umbilical vein endothelial cells (HUVECs) as a model for ECs. CXCR7 was highly expressed on HUVECs. We could demonstrate that in vitro tube formation was promoted by either co-culture of MM cells or by conditioned medium from MM cell cultures. Furthermore, migration of HUVEC cells was facilitated by conditioned medium from MM cell cultures. These data suggest that MM cells may secrete factors promoting migration of endothelial cells and pro-angiogenic factors promoting angiogenesis. In addition, we could show that in vitro tube formation is inhibited by POL6926 suggesting that CXCR7 expression on HUVECs is required for tube formation. At the test concentrations POL6926 was not cytotoxic to HUVECs since cell proliferation was unaffected. CONCLUSION: We have shown that the level of ECs and EPCs was elevated in the PB and BM of MM patients compared to normal subjects, a finding which was confirmed in a MM mouse model in which CXCR7 was highly expressed on these cells. Injection of PEM CXCR7 antagonist increased the numbers of ECs and EPCs in the PB. These results suggest that CXCR7 may play a role in the cell-trafficking and recruitment of ECs and EPCs in MM. To investigate this hypothesis, using in vitro tube formation and migration assays, we have shown that MM cells secrete factors that promote migration and angiogenesis of HUVECs and the PEM CXCR7 antagonist inhibits these processes. In subsequent studies POL6926 will be tested in vivo in animal models of MM to determine the contribution of CXCR7 in EPC trafficking and its contribution to angiogenesis progression in MM. Disclosures: Zimmermann: Polyphor: Employment. Patel:Polyphor: Employment. Romagnoli:Polyphor: Employment. Roccaro:Roche:. Ghobrial:Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Millennium: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Research Funding; Noxxon: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding.


2021 ◽  
Vol 22 (22) ◽  
pp. 12241
Author(s):  
Aleksandra Klimczak ◽  
Piotr Hinc ◽  
Agnieszka Krawczenko ◽  
Aleksandra Bielawska-Pohl ◽  
Maria Paprocka ◽  
...  

Mesenchymal stem cells (MSCs) can improve chronic wound healing; however, recent studies suggest that the therapeutic effect of MSCs is mediated mainly through the growth factors and cytokines secreted by these cells, referred to as the MSC secretome. To overcome difficulties related to the translation of cell therapy into clinical use such as efficacy, safety and cost, we propose a hydrogel loaded with a secretome from the recently established human adipose tissue mesenchymal stem cell line (HATMSC2) as a potential treatment for chronic wounds. Biocompatibility and biological activity of hydrogel-released HATMSC2 supernatant were investigated in vitro by assessing the proliferation and metabolic activity of human fibroblast, endothelial cells and keratinocytes. Hydrogel degradation was measured using hydroxyproline assay while protein released from the hydrogel was assessed by interleukin-8 (IL-8) and macrophage chemoattractant protein-1 (MCP-1) ELISAs. Pro-angiogenic activity of the developed treatment was assessed by tube formation assay while the presence of pro-angiogenic miRNAs in the HATMSC2 supernatant was investigated using real-time RT-PCR. The results demonstrated that the therapeutic effect of the HATMSC2-produced factors is maintained following incorporation into collagen hydrogel as confirmed by increased proliferation of skin-origin cells and improved angiogenic properties of endothelial cells. In addition, HATMSC2 supernatant revealed antimicrobial activity, and which therefore, in combination with the hydrogel has a potential to be used as advanced wound-healing dressing.


Cancers ◽  
2021 ◽  
Vol 13 (14) ◽  
pp. 3385
Author(s):  
Axel H. Schönthal ◽  
Steve Swenson ◽  
Radu O. Minea ◽  
Hye Na Kim ◽  
Heeyeon Cho ◽  
...  

Despite progress in the treatment of acute myeloid leukemia (AML), the clinical outcome remains suboptimal and many patients are still dying from this disease. First-line treatment consists of chemotherapy, which typically includes cytarabine (AraC), either alone or in combination with anthracyclines, but drug resistance can develop and significantly worsen prognosis. Better treatments are needed. We are developing a novel anticancer compound, NEO212, that was created by covalent conjugation of two different molecules with already established anticancer activity, the alkylating agent temozolomide (TMZ) and the natural monoterpene perillyl alcohol (POH). We investigated the anticancer activity of NEO212 in several in vitro and in vivo models of AML. Human HL60 and U937 AML cell lines, as well as different AraC-resistant AML cell lines, were treated with NEO212 and effects on cell proliferation, cell cycle, and cell death were investigated. Mice with implanted AraC-sensitive or AraC-resistant AML cells were dosed with oral NEO212, and animal survival was monitored. Our in vitro experiments show that treatment of cells with NEO212 results in growth inhibition via potent G2 arrest, which is followed by apoptotic cell death. Intriguingly, NEO212 was equally potent in highly AraC-resistant cells. In vivo, NEO212 treatment strikingly extended survival of AML mice and the majority of treated mice continued to thrive and survive without any signs of illness. At the same time, we were unable to detect toxic side effects of NEO212 treatment. All in all, the absence of side effects, combined with striking therapeutic activity even in an AraC-resistant context, suggests that NEO212 should be developed further toward clinical testing.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Zaipul I. Md Dom ◽  
Caterina Pipino ◽  
Bozena Krolewski ◽  
Kristina O’Neil ◽  
Eiichiro Satake ◽  
...  

AbstractWe recently identified a kidney risk inflammatory signature (KRIS), comprising 6 TNF receptors (including TNFR1 and TNFR2) and 11 inflammatory proteins. Elevated levels of these proteins in circulation were strongly associated with risk of the development of end-stage kidney disease (ESKD) during 10-year follow-up. It has been hypothesized that elevated levels of these proteins in circulation might reflect (be markers of) systemic exposure to TNFα. In this in vitro study, we examined intracellular and extracellular levels of these proteins in human umbilical vein endothelial cells (HUVECs) exposed to TNFα in the presence of hyperglycemia. KRIS proteins as well as 1300 other proteins were measured using the SOMAscan proteomics platform. Four KRIS proteins (including TNFR1) were down-regulated and only 1 protein (IL18R1) was up-regulated in the extracellular fraction of TNFα-stimulated HUVECs. In the intracellular fraction, one KRIS protein was down-regulated (CCL14) and 1 protein was up-regulated (IL18R1). The levels of other KRIS proteins were not affected by exposure to TNFα. HUVECs exposed to a hyperglycemic and inflammatory environment also showed significant up-regulation of a distinct set of 53 proteins (mainly in extracellular fraction). In our previous study, circulating levels of these proteins were not associated with progression to ESKD in diabetes.


2021 ◽  
Vol 18 ◽  
Author(s):  
Juxuan Ruan ◽  
Lei Wang ◽  
Jiheng Dai ◽  
Jing Li ◽  
Ning Wang ◽  
...  

Objective: Angiogenesis led by brain microvascular endothelial cells (BMECs) contributes to the remission of brain injury after brain ischemia reperfusion. In this study, we investigated the effects of hydroxysafflor yellow A(HSYA) on angiogenesis of BMECs injured by OGD/R via SIRT1-HIF-1α-VEGFA signaling pathway. Methods: The OGD/R model of BMECs was established in vitro by OGD for 2h and reoxygenation for 24h. At first, the concentrations of vascular endothelial growth factor (VEGF), Angiopoietin (ang) and platelet-derived growth factor (PDGF) in supernatant were detected by ELISA, and the proteins expression of VEGFA, Ang-2 and PDGFB in BMECs were tested by western blot; the proliferation, adhesion, migration (scratch healing and transwell) and tube formation experiment of BMECs; the expression of CD31 and CD34 were tested by immunofluorescence staining. The levels of sirtuin1(SIRT1), hypoxia-inducible factor-1α (HIF-1α), VEGFA mRNA and protein were tested. Results: HSYA up-regulated the levels of VEGF, Ang and PDGF in the supernatant of BMECs under OGD/R, and the protein expression of VEGFA, Ang-2 and PDGFB were increased; HSYA could significantly alleviate the decrease of cell proliferation, adhesion, migration and tube formation ability of BMECs during OGD/R; HSYA enhanced the fluorescence intensity of CD31 and CD34 of BMECs during OGD/R; HSYA remarkably up-regulated the expression of SIRT1, HIF-1α, VEGFA mRNA and protein after OGD/R, and these increase decreased after SIRT1 was inhibited. Conclusion: SIRT1-HIF-1α-VEGFA signaling pathway is involved in HSYA improves angiogenesis of BMECs injured by OGD/R.


2008 ◽  
Vol 100 (10) ◽  
pp. 693-698 ◽  
Author(s):  
Michael Buerke ◽  
Sebastian Schubert ◽  
Iris Reindl ◽  
Thomas Michel ◽  
Baerbel Hauroeder ◽  
...  

SummaryBivalirudin, a direct thrombin inhibitor binds specifically and reversibly to both fibrin-bound and unbound thrombin. Bivalirudin is approved for use as an anticoagulant in patients undergoing percutaneous coronary intervention. The OASIS-5 trial presented a significant increase in cardiac catheter thrombosis for the pentasaccharid fondaparinux compared to enoxaparin. Catheter thrombosis has never been reported in any trial using bivalirudin. Our study compared the development of catheter thrombosis for bivalirudin, enoxaparin, and unfractionated heparin in a controlled in-vitro environment. Ten healthy male volunteers were pretreated with aspirin 500 mg 2 hours before venesection of 50 ml of blood. The seven groups of anticoagulant combinations tested were:UFH, UFH + eptifibatide, enoxaparin, enoxaparin + eptifibatide, bivalirudin bolus, bivalirudin + eptifibatide, bivalirudin bolus + continuous infusion. The blood/anticoagulant mix continuously circulated through a cardiac guiding catheter for 60 minutes or until the catheter became blocked with thrombus. Thrombus development was assessed by weighing each catheter before and after the procedure. Electron microscopy was used to quantify the degree of erythrocyte, platelet and fibrin deposition. Following anticoagulation with bolus dose bivalirudin, the catheter was invariably occluded with thrombus after 33 minutes of circulation. However, a continuous infusion of Bivalirudin prevented the development of occlusive catheter thrombosis. In the bolus bivalirudin group the mean thrombus weight was significantly greater than in all other groups (p-value < 0.01 in all analyses). Bivalirudin given as a bolus was not sufficient to prevent cardiac catheter thrombosis in our in-vitro study. However, a continuous infusion of bivalirudin had similar anti-thrombotic efficacy compared to other treatment strategies.


Author(s):  
Teresa Al Haddad ◽  
Elie Khoury ◽  
Nada Farhat Mchayleh

Abstract Objectives The aim of the present in vitro study is to compare the remineralization brushing effect of three toothpastes and Aloe vera (AV) gel. Materials and Methods Forty sound extracted teeth were placed in a demineralizing solution for 4 days and randomly assigned to four groups: group A: 1,450-ppm fluoride toothpaste; group B: AV nonfluoridated toothpaste; group C: AV 1,000-ppm fluoridated toothpaste; and group D: AV gel. A 3-minute pH cycling was performed twice a day for each group for 12 days. Specimens were analyzed before and after by scanning electron microscope—energy dispersive X-ray. Statistical analysis The outcomes were analyzed by Kolmogorov–Smirnov’s tests, repeated-measures analyses of variance followed by univariate analyses, and Bonferroni’s multiple comparisons tests to compare the calcium-to-phosphorus (Ca:P) ratio within time among toothpaste groups. Results Following remineralization, the Ca:P ratio increased in all groups. The difference of the Ca:P ratio was not significant between groups C, D, and A. The mean ratio was significantly lower in group B (p-value = 0.026). Conclusions The AV gel demonstrated a remineralization capacity equal to that of the 1,450-ppm fluoride toothpaste. In contrast, fluoride-free AV toothpaste showed a lower remineralization efficiency. Further studies are required to understand its mechanism.


Sign in / Sign up

Export Citation Format

Share Document