Circulating endothelial cells in patients with acute myeloid leukemia

2005 ◽  
Vol 75 (6) ◽  
pp. 492-497 ◽  
Author(s):  
Agnieszka Wierzbowska ◽  
Tadeusz Robak ◽  
Anna Krawczynska ◽  
Agata Wrzesien-Kus ◽  
Agnieszka Pluta ◽  
...  
Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2776-2776
Author(s):  
Agnieszka Wierzbowska ◽  
Tadeusz Robak ◽  
Anna Krawczynska ◽  
Agata Wrzesien-Kus ◽  
Agnieszka Pluta ◽  
...  

Abstract Introduction. The circulating endothelial cells (CEC) are proposed to be a noninvasive marker of angiogenesis. The number of CEC in peripheral blood of patients (pts) with acute myeloid leukemia (AML) has not been investigated so far. Patients and Methods. We evaluated the count of resting (rCEC) and activated (aCEC) CEC and circulating endothelial progenitor cells (CEPC) as well as apoptotic CEC (CECAnnV+) in 62 AML pts at the time of diagnosis and 30 healthy controls. Additionally in 26 pts measurements were performed at the time of response evaluation and in 15 pts also 24 h after the first and last dose of chemotherapy. The levels of CEC were correlated with known prognostic factors and response to treatment. CEC were evaluated by the four colour flow cytometry using a panel of previously described monoclonal antibodies and an appropriate analysis gate. CEPC were defined as negative for hematopoietic marker CD45 and positive for endothelial cells markers CD34, CD31 and the endothelial progenitor marker CD133. Resting CEC were defined as CD45−, CD133−, CD31+, CD34+, CD146+ and negative for activation markers (CD105, CD106). CD105 or CD106 positive mature endothelial cells were classified as activated CEC. Apoptotic CEC were CD146 and Annexin V positive. Results. In untreated AML pts we observed 10-fold higher CEC level (median 29,3/μL) than in the control group (2,95/μL) p<0,0001. The numbers of aCEC (12,7/μL), rCEC (12,3/μL) and CEPC (1,7/μL) were significantly higher in AML pts at diagnosis when compared to healthy controls (aCEC 0,9/μL, rCEC 1,6/μL and 0,1/μL; p<0,0001). CECAnnV+ count was also 10-fold higher in AML (1,5/μL) than in controls (0,15/μL; p<0,0001). Both CEC and CECAnnV+ counts did not correlate with WBC, hemoglobin and platelets count as well as percentage of blasts in bone marrow and absolute blast count. The positive correlations between CEC number and CEPC count (r=0,435; p<0,001), CECAnnV+ count (r=0,502; p<0,01) as well as LDH activity (r=0,328; p<0,02) were found. The significant decrease of aCEC and rCEC numbers 24 hours after the first dose of chemotherapy was noted in patients who achieved complete remission (CR)(p<0,04) but not in pts refractory to treatment. Moreover aCEC, rCEC, CEPC and CECAnnV+ counts determined at the time of response’s evaluation were significantly lower then at the time of diagnosis in pts who achieved CR (p<0,01) and did not differ in refractory AML. There was no difference between levels of both viable and apoptotic CEC in AML pts in CR and in the control group (p>0,05). Conclusions. The CEC and CECAnnV+ levels are significantly higher in AML patients than in healthy subjects and correlate with response to treatment. Further investigation should be undertaken to better determine their prognostic and therapeutic value.


2016 ◽  
Vol 12 (3) ◽  
pp. 1965-1970 ◽  
Author(s):  
Asmaa Mohammed Zahran ◽  
Sanaa Shaker Aly ◽  
Hanan Ahmed Altayeb ◽  
Arwa Mohammed Ali

2016 ◽  
Vol 44 (5) ◽  
pp. 378-389 ◽  
Author(s):  
Russell J. Pizzo ◽  
Mitra Azadniv ◽  
Naxin Guo ◽  
Joshua Acklin ◽  
Kimberly Lacagnina ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2546-2546 ◽  
Author(s):  
Leylah Drusbosky ◽  
Amy Meacham ◽  
Elizabeth Wise ◽  
Edward W Scott ◽  
Christopher R Cogle

Abstract Causes of refractory acute myeloid leukemia (AML) are unknown, but may be related to the bone marrow (BM) vascular network given the close relationship between hematopoiesis and the vasculature. We hypothesized that endothelial cells (ECs) provide a protective advantage to AML cells. To test this hypothesis, we first cultured human AML cells with and without primary bone marrow endothelial cells (BMECs). Co-cultures of AML cells and BMECs were then exposed to increasing doses of cytarabine chemotherapy. There was a 2-fold decrease in leukemia cell death of AML cells when adhered to BMECs compared to non-adhered (30% vs. 60%, P < 0.0001). Even irradiated BMECs protected AML cells from cytarabine chemotherapy. To identify adhesion molecules mediating this protective effect, we analyzed cell membranes and supernatants of the cytarabine-treated co-cultures using protein microarrays. After cytarabine exposure, the Lutheran blood group glycoprotein basal cell adhesion molecule (BCAM) was upregulated in both human AML cells and BMECs. Prior work has shown BCAM as a receptor for Laminin and VLA5. As AML cells are known to express VLA5, we hypothesized that blocking BCAM may represent a novel therapeutic strategy. Blocking BCAM with neutralizing antibodies resulted in a 75% increase in non-adherent AML cells. Together, these in vitro results support the concept that ECs may be a protective reservoir for AML cells, at a minimum by means of adhesion molecules. BCAM represents a viable target. Because of the complex nature of the leukemia microenvironment, we sought to test this concept in vivo. Prior intravital efforts have focused on calvaria bone, which may over-represent the endosteal niche and under-represent the vascular niche due to the very close approximation of bone surfaces. Therefore, we created an intravital animal model of human AML to track single AML cells in the bone marrow of mouse long bones. In brief, we irradiated NOD/scid/IL2Rγnull (NSG) mice, drilled a window on the tibia surface, xenotransplanted fluorescently tagged human AML cells via IV injection, and then analyzed the tibias by fluorescent microscopy for the presence of AML cells at various time points after transplant. Initially the AML cells homed to endosteal surfaces of the bone marrow as early as one day after transplant. Over time, the AML cells lining the endosteum remained as single cells or very small clusters of a few cells. However, in the central marrow region the AML cells proliferated into massive clusters around blood vessels. Ongoing experiments are being performed to determine the disruption and resurgence of AML cells after chemotherapy and blockade of adhesion molecules. In sum, ECs protect human AML cells from chemotherapy by direct contact and the BCAM/Laminin/VLA5 axis may be a therapeutic target. Using our unique intravital imaging model of bone marrow in long bones, the endosteal niche appears to be the first site of homing and engraftment while the vascular niche appears to be the site for leukemia proliferation/progression. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1057-1057 ◽  
Author(s):  
Eric Gars ◽  
Christie S McGee ◽  
Raphael C Bosse ◽  
Elizabeth Wise ◽  
Amy Meacham ◽  
...  

Abstract Abstract 1057 Acute myeloid leukemia (AML) cells depend on endothelial cells for survival and proliferation. By targeting endothelial cells with a novel vascular disrupting agent, we recently demonstrated regression of AML (Madlambayan, et al., Blood 2010). In an effort to discover a more selective, anti-vascular therapy for leukemia, we hypothesized that targeting endothelial cell-derived paracrine and leukemia cell-derived autocrine growth factors would result in regression of disease. In particular, our strategy focused on vascular endothelial growth factor (VEGF), platelet derived growth factor (PDGF) and stem cell factor (SCF), which are known to promote leukemia cell proliferation. Receptors for each of these growth factors are potently inhibited by pazopanib, which is an orally available tyrosine kinase inhibitor. The aim of our study was to determine the dependence of leukemia cell survival and proliferation on the combined receptor signaling of VEGF, PDGF and SCF. Leukemia cell lines (KG-1, HL60 and K562) were incubated at various durations with varying concentrations of pazopanib. Leukemia cell proliferation was quantified using XTT assay. Apoptosis induction was analysed by flow cytometry (Annexin V and PI staining). Pazopanib effectively impaired proliferation in leukemia cells in vitro in a dose and time dependent fashion. During a 16-hour incubation of leukemia cells, pazopanib showed a 50% lethal concentration (LC50) of 22.57 μM (r = .986), 41.6 μM (r = .991), and 81.97 μM (r = .996) for HL60, K562, and KG-1 cells, respectively. Staining with Annexin V and PI identified apoptosis as the main cause of cell death after exposure to pazopanib. The IC50 of apoptosis for HL60 cells was 132.5 ± 2.7 μM (p = 0.0002). These results indicate that leukemia cells depend on the combined signaling of VEGF, PDGF, and SCF, and suggest that selective inhibition by pazopanib may be a promising therapeutic for AML. Furthermore, pazopanib is orally available and associated with minimal side effects, thus representing an attractive candidate for further testing in AML. Disclosures: No relevant conflicts of interest to declare.


2009 ◽  
Vol 18 (8) ◽  
pp. 1235-1242 ◽  
Author(s):  
Roxana Bistrian ◽  
Annette Dorn ◽  
Dietrich C.C. Möbest ◽  
Brigitte Rüster ◽  
Ralf Ludwig ◽  
...  

Cells ◽  
2020 ◽  
Vol 9 (10) ◽  
pp. 2310
Author(s):  
Jan Philipp Bewersdorf ◽  
Amer M. Zeidan

Up to 18% of patients with acute myeloid leukemia (AML) present with a white blood cell (WBC) count of greater than 100,000/µL, a condition that is frequently referred to as hyperleukocytosis. Hyperleukocytosis has been associated with an adverse prognosis and a higher incidence of life-threatening complications such as leukostasis, disseminated intravascular coagulation (DIC), and tumor lysis syndrome (TLS). The molecular processes underlying hyperleukocytosis have not been fully elucidated yet. However, the interactions between leukemic blasts and endothelial cells leading to leukostasis and DIC as well as the processes in the bone marrow microenvironment leading to the massive entry of leukemic blasts into the peripheral blood are becoming increasingly understood. Leukemic blasts interact with endothelial cells via cell adhesion molecules such as various members of the selectin family which are upregulated via inflammatory cytokines released by leukemic blasts. Besides their role in the development of leukostasis, cell adhesion molecules have also been implicated in leukemic stem cell survival and chemotherapy resistance and can be therapeutically targeted with specific inhibitors such as plerixafor or GMI-1271 (uproleselan). However, in the absence of approved targeted therapies supportive treatment with the uric acid lowering agents allopurinol and rasburicase as well as aggressive intravenous fluid hydration for the treatment and prophylaxis of TLS, transfusion of blood products for the management of DIC, and cytoreduction with intensive chemotherapy, leukapheresis, or hydroxyurea remain the mainstay of therapy for AML patients with hyperleukocytosis.


2014 ◽  
Vol 2014 ◽  
pp. 1-12 ◽  
Author(s):  
Hilde Kvestad ◽  
Lasse Evensen ◽  
James B. Lorens ◽  
Øystein Bruserud ◽  
Kimberley J. Hatfield

The combined use of the histone deacetylase inhibitor valproic acid (VPA), the retinoic acid receptor-α agonist all-trans retinoic acid (ATRA), and the deoxyribonucleic acid polymerase-α inhibitor cytarabine (Ara-C) is now considered for disease-stabilizing treatment of acute myeloid leukemia (AML). Leukemogenesis and leukemia cell chemoresistance seem to be supported by neighbouring stromal cells in the bone marrow, and we have therefore investigated the effects of these drugs on primary human endothelial cells and the osteoblastic Cal72 cell line. The results show that VPA and Ara-C have antiproliferative effects, and the antiproliferative/cytotoxic effect of Ara-C was seen at low concentrations corresponding to serum levels found during low-dose in vivo treatment. Furthermore, in functional assays of endothelial migration and tube formation VPA elicited an antiangiogenic effect, whereas ATRA elicited a proangiogenic effect. Finally, VPA and ATRA altered the endothelial cell release of angiogenic mediators; ATRA increased levels of CXCL8, PDGF-AA, and VEGF-D, while VPA decreased VEGF-D and PDGF-AA/BB levels and both drugs reduced MMP-2 levels. Several of these mediators can enhance AML cell proliferation and/or are involved in AML-induced bone marrow angiogenesis, and direct pharmacological effects on stromal cells may thus indirectly contribute to the overall antileukemic activity of this triple drug combination.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2662-2662 ◽  
Author(s):  
Diana Passaro ◽  
Alessandro Di Tullio ◽  
Ander Abarrategi ◽  
Kevin Rouault-Pierre ◽  
Katie Foster ◽  
...  

Abstract The biological and clinical behavior of hematological malignancies are not only determined by the properties of the leukemic cells themselves, but are also highly affected by interaction with the microenvironment, pointing to the existence of an active crosstalk between the two compartments. Previous studies showed that acute myeloid leukemia (AML) cells actively modify endothelial cells ex vivovia several pathways, mainly mediated by VEGF. However, as anti-VEGF therapies haven't produced successful results in clinical trials, an extensive study of the crosstalk between AML and the vascular niche in the bone marrow (BM) is required to provide new therapeutic strategies. In the present study we combined the use of mouse models of AML, human AML patient-derived xenografts (PDX) and direct analysis on patient-derived BM biopsies to provide a global, reliable picture of the bone marrow vasculature in AML disease. We found several abnormalities in the vascular architecture and function in PDX, such as increased number of endothelial cells, increased microvascular density (MVD), decreased vascular mean diameter and increased hypoxia. Furthermore, using two-photon confocal intravital imaging we witnessed increased vascular permeability upon AML engraftment, observed homogeneously among different PDX. Interestingly, induction chemotherapy failed to normalize the vascular permeability in the BM, despite significant reduction in AML engraftment. We identified increased nitric oxide (NO) as a major mediator of the AML-induced vascular leakiness in the BM. Increased levels of NO and activated NOS3 were found in PDX and in an independent cohort of patient-derived BM biopsies. Strikingly, inhibition of NO production using genetic and pharmacological approaches reduced the vascular permeability, potentiated the normal HSC function and significantly improved treatment response in PDX. These results strongly support the notion of a primary function of the vascular permeability in AML progression, drug response and in affecting normal stem cell function, and they call for clinical trials incorporating NOS inhibitors during the remission phase to target the abnormal vascular niche and improve the treatment response. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document