The Development of Novel Hemostatic Bypassing Molecules.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. SCI-20-SCI-20
Author(s):  
Rodney M. Camire ◽  
Matthew Bunce ◽  
Lacramioara Ivanciu ◽  
Raffaella Toso ◽  
Harre D. Downey ◽  
...  

Abstract Abstract SCI-20 Blood coagulation factors VIII and IX are part of the intrinsic pathway and play a critical role in maintaining normal hemostasis by activating factor X. Deficiency of either of these proteins caused by mutations in the genes encoding FVIII or FIX leads to hemophilia A or B (HA and HB), respectively. In each disease, there is impairment of the intrinsic pathway with inadequate thrombin (IIa) generation and defective hemostasis. Unfortunately, ∼20-30% of patients with FVIII deficiency and ∼3-5% of patients with FIX deficiency develop inhibitory antibodies to infused factor replacement products. This has prompted the research community to develop so-called “bypass strategies” which use other coagulation factors to provide hemostasis in these patients. In principle, infusion of FXa should bypass deficiencies in the intrinsic pathway; however it is generally thought that it has limited utility since the infused FXa could cause excessive activation of coagulation and/or more importantly, FXa is rapidly inactivated by plasma inhibitors resulting in a very short half-life (<2 min). We recently characterized variants of FXa (FXa-I16L and FXa-V17a) which have “zymogen-like” properties that could circumvent these associated problems (JBC 2008; 283: 18627). For example we have found that i) these proteins have an incompletely formed active site, making them resistant to plasma protease inhibitors; ii) in the absence of FVa, the FXa variants are, in general, refractory to active site functions and thus do not activate FVII, FV, FVIII, and prothrombin very well; and iii) the variants are thermodynamically rescued by FVa; thus at the site of injury on the activated platelet surface, where FVa is present, prothrombinase rapidly forms generating a burst of thrombin. We have begun to exploit these unique properties and evaluate whether these FXa variants could be effective and safe in bypassing the hemophilic phenotype both in vitro and in vivo. Clotting and IIa generation assays in human HA, HB and inhibitor plasma revealed that FXa-I16L could completely restore IIa generation in a FVa-dependent fashion. Furthermore, the zymogen-like conformation protects FXa-I16L in human plasma as it has a prolonged half-life (∼2 hr) versus wt-FXa (<2 min). In vivo studies using HB mice revealed that administration of FXa-I16L via tail vein almost completely corrected the prolonged aPTT. The aPTT was shortened for more than 2 hours and returned close to the starting value after 24 hr. In these experiments, infusion of the protein was well tolerated as platelet levels were unaffected over the course of the experiment with little or no change in the values for TAT, D-dimer, and fibrinogen. Next we tested whether the improvement of the clotting times was associated with in vivo hemostatic performance. Using three separate injury models (tail clip assay, FeCl2 carotid artery injury model, and cremaster muscle laser injury model) infusion FXa-I16L provided effective hemostasis. This was in contrast to the infusion of wild-type FXa which was ineffective. Taken together our data show that FXa-I16L is highly effective in correcting the hemostatic defect in human hemophilic plasma. Furthermore it improves the hemophilic phenotype in HB mice following a series of hemostatic challenges and can restore thrombus formation upon injury at both micro and macrocirculation levels. Thus zymogen-like variants of FXa have properties that indicate their ability to serve as superior therapeutic procoagulants for bypassing deficiencies upstream of the common pathway. Disclosures Camire: Wyeth: Patents & Royalties, Research Funding. Bunce:Wyeth: Research Funding. Ivanciu:Wyeth: Research Funding. Downey:Wyeth: Research Funding.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 239-239
Author(s):  
Lacramioara Ivanciu ◽  
Raffaella Toso ◽  
Alexander Schlachterman ◽  
Harre Downey ◽  
Jian-Hua Liu ◽  
...  

Abstract Hemophilia A and B are bleeding disorders caused by mutations in the genes encoding FVIII or FIX. Treatment involves protein replacement; however in some patients antibodies develop that neutralize FVIII or FIX. In these inhibitor patients, hemostasis can be achieved with activated prothrombin complex concentrates or recombinant FVIIa which bypass the intrinsic pathway generating thrombin to allow coagulation to occur. In principle, infusion of FXa should bypass deficiencies in the intrinsic pathway. However, this could trigger excessive coagulation as FXa can activate several clotting factors. Furthermore, FXa is rapidly inactivated by plasma inhibitors resulting in a short half-life (&lt;1–2 min) limiting its utility. Recently, we characterized FXa variants with unique properties that could circumvent these associated problems (JBC2008; 283: 18627). These proteins (FXa- I16L and V17A) have an incompletely formed active site and poor catalytic activity and are less sensitive to active site directed inhibitors. However, once these “zymogen-like” variants bind FVa to form prothrombinase they efficiently activate prothrombin. Based on these differential functional states, these FXa variants may act as long lived proteases in circulation that are otherwise inert in the absence of FVa but retain the ability to catalyze thrombin formation once the cofactor is made available. To evaluate their potential, we initiated a series of in vivo experiments with FXa-I16L using age-matched male HB mice on the Balb/c strain to examine the effects on systemic coagulation as well as efficacy using three injury models. Using a modified aPTT clotting assay, hemostatically normal control (wt) animals had a clot time of 27 ± 3 sec (n = 4) while in untreated HB animals it was 69 ± 4 sec (n = 10). Administration of FXa-I16L (450 μg/kg) via tail vein almost completely corrected the aPTT 10 min post infusion (n = 5; 31 ± 5 sec) whereas PBS was without effect (n = 5; 63 ± 3 sec). The aPTT was shortened for more than 2 hours (50 ± 3 sec) and returned close to the starting value after 24 hr (62 ± 3 sec). In these experiments, infusion of the protein was well tolerated as all animals survived. Furthermore, using this high dose of FXa-I16L (450 μg/kg) platelet levels only marginally decreased over the course of the experiment while there was a transient increase in TAT levels at 10 min (68 ± 33 ng/mL versus 16 ± 8 ng/mL in HB controls) which returned back to baseline after 60 min (18 ± 7 ng/mL). Next we tested whether the improvement of the clotting times was associated with in vivo hemostatic performance. We used a tail clip assay in which blood loss was measured during a 10 min period after sectioning the distal part of the tail. Infusion of FXa-I16L in HB mice (n = 7; 450 μg/kg) significantly reduced total blood loss compared to PBS (n = 7) treated animals (p &lt;0.0003). The extent of blood loss with FXa- I16L was comparable to wt mice. In another in vivo test of thrombosis, the FeCl3-carotid artery injury model, normal mice (n = 3) presented with vessel occlusion and interruption of blood flow within 9–12 min postvessel injury with 15% FeCl3 while no evidence of clot formation was found for HB controls (n = 5). After injury of HB mice with FeCl3, followed by a 10 min observation period with stable blood flow, infusion of FXa-I16L (450 μg/kg) resulted in vessel occlusion and interruption of blood flow within 5–6 min in three out of five mice. In the last in vivo injury model, thrombus formation was assessed following a laser injury to a small vessel (arteriole) in the cremaster muscle by monitoring platelet accumulation at the site of injury. Untreated HB mice (n = 13, 30 sites) failed to form clots, while all HB animals treated with FXa-I16L (n= 10, &gt;70 sites; 10, 30, or 90 μg/kg) formed a stable thrombus following injury that was comparable in size to wt mice (n = 3, 17 sites). Taken together our data show that FXa-I16L improves the hemophilic phenotype in HB mice following a series of hemostatic challenges and can restore thrombus formation upon injury at both micro and macrocirculation levels. Thus zymogen-like variants of FXa have properties that indicate their ability to serve as superior therapeutic procoagulants for bypassing deficiencies upstream of the common pathway.


1981 ◽  
Vol 46 (03) ◽  
pp. 658-661 ◽  
Author(s):  
C Korninger ◽  
J M Stassen ◽  
D Collen

SummaryThe turnover of highly purified human extrinsic plasminogen activator (EPA) (one- and two-chain form) was studied in rabbits. Following intravenous injection, EPA-activity declined rapidly. The disappearance rate of EPA from the plasma could adequately be described by a single exponential term with a t ½ of approximately 2 min for both the one-chain and two-chain forms of EPA.The clearance and organ distribution of EPA was studied by using 125I-labeled preparations. Following intravenous injection of 125I-1abeled EPA the radioactivity disappeared rapidly from the plasma also with a t ½ of approximately 2 min down to a level of 15 to 20 percent, followed by a small rise of blood radioactivity. Gel filtration of serial samples revealed that the secondary increase of the radioactivity was due to the reappearance of radioactive breakdown products in the blood. Measurement of the organ distribution of 125I at different time intervals revealed that EPA was rapidly accumulated in the liver, followed by a release of degradation products in the blood.Experimental hepatectomy markedly prolonged the half-life of EPA in the blood. Blocking the active site histidine of EPA had no effect on the half-life of EPA in blood nor on the gel filtration patterns of 125I in serial plasma samples.It is concluded that human EPA is rapidly removed from the blood of rabbits by clearance and degradation in the liver. Recognition by the liver does not require a functional active site in the enzyme. Neutralization in plasma by protease inhibitors does not represent a significant pathway of EPA inactivation in vivo.


Thorax ◽  
2021 ◽  
pp. thoraxjnl-2020-216469
Author(s):  
Alison W Ha ◽  
Tao Bai ◽  
David L Ebenezer ◽  
Tanvi Sethi ◽  
Tara Sudhadevi ◽  
...  

IntroductionNeonatal lung injury as a consequence of hyperoxia (HO) therapy and ventilator care contribute to the development of bronchopulmonary dysplasia (BPD). Increased expression and activity of lysyl oxidase (LOX), a key enzyme that cross-links collagen, was associated with increased sphingosine kinase 1 (SPHK1) in human BPD. We, therefore, examined closely the link between LOX and SPHK1 in BPD.MethodThe enzyme expression of SPHK1 and LOX were assessed in lung tissues of human BPD using immunohistochemistry and quantified (Halo). In vivo studies were based on Sphk1−/− and matched wild type (WT) neonatal mice exposed to HO while treated with PF543, an inhibitor of SPHK1. In vitro mechanistic studies used human lung microvascular endothelial cells (HLMVECs).ResultsBoth SPHK1 and LOX expressions were increased in lungs of patients with BPD. Tracheal aspirates from patients with BPD had increased LOX, correlating with sphingosine-1-phosphate (S1P) levels. HO-induced increase of LOX in lungs were attenuated in both Sphk1−/− and PF543-treated WT mice, accompanied by reduced collagen staining (sirius red). PF543 reduced LOX activity in both bronchoalveolar lavage fluid and supernatant of HLMVECs following HO. In silico analysis revealed STAT3 as a potential transcriptional regulator of LOX. In HLMVECs, following HO, ChIP assay confirmed increased STAT3 binding to LOX promoter. SPHK1 inhibition reduced phosphorylation of STAT3. Antibody to S1P and siRNA against SPNS2, S1P receptor 1 (S1P1) and STAT3 reduced LOX expression.ConclusionHO-induced SPHK1/S1P signalling axis plays a critical role in transcriptional regulation of LOX expression via SPNS2, S1P1 and STAT3 in lung endothelium.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 45-45
Author(s):  
Sushanth Gouni ◽  
Paolo Strati ◽  
Jason Westin ◽  
Loretta J. Nastoupil ◽  
Raphael E Steiner ◽  
...  

Background: Pre-clinical studies show that statins may improve the efficacy of chemoimmunotherapy in patients with DLBCL, through interference with cell membrane-initiated signaling pathways. Clinical retrospective studies, however, yield conflicting data, due to heterogeneous properties of statins, including potency and hydrophilicity. Methods: This is a retrospective analysis of patients with previously untreated, advanced stage DLBCL, non-double hit, treated with frontline R-CHOP between 01/01/2000 and 09/01/2019 (data cut-off 04/15/2020) at MD Anderson Cancer Center, and for whom data regarding statin use at time of initiation of treatment were available. Lugano 2014 response criteria were applied retrospectively for response assessment. Cellular cholesterol levels were analyzed in 6 DLBCL cell lines using an Amplex red fluorometric assay. A doxorubicin (DXR)-resistant cell line was generated exposing SUDHL4 cells to escalating doses of DXR; a DXR-resistant DLBCL patient-derived xenograft (PDX) model was established through serial transplantation and exposure to DXR. Results: 271 patients were included in the analysis, 182 (67%) were older than 60 years, 134 (49%) were male, 212 (72%) had stage IV disease, and 217 (80%) had an IPI score &gt; 3; upon pathological review, 38 (36%) cases were non-GCB type, and 18 (28%) were double-expressors; 214 (79%) were able to complete all planned 6 cycles of RCHOP. Seventy-nine (29%) patients received statins at time of initiation of chemoimmunotherapy: 15 patients received low potency statin, 51 medium and 13 high; 18 patients received hydrophilic statins and 61 lipophilic. Patients receiving statins were significantly older as compared to patients who did not (p&lt;0.001); no other significant difference in baseline characteristics was observed when comparing the 2 groups. Overall, 265 out of 271 patients were evaluable for response, as 6 stopped treatment because of toxicity before first response assessment. Among these, ORR was 95% (252/265) and CR rate was 62% (165/265). ORR rate was identical in patients who were treated with statin and those who did not (95% both, p=1). After a median follow-up of 77 months (95% CI, 70-84 months), 119 patients progressed/died, median PFS was not reached and 6-year PFS was 57%. 6-year PFS rate according to statin intensity was: 48% (low), 72% (medium), 57% (high). PFS. 6-year PFS rate was 64% for hydrophilic and 72% for lipophilic statins. Patients treated with statins had a trend for longer PFS (p=0.06), significantly longer for patients receiving medium potency statins (p=0.04). No significant difference in PFS was observed when comparing patients treated with lipophilic statins to all others (not reached vs 84 months, p=0.22). To confirm these clinical data, in-vitro and in-vivo studies were performed. Six cell lines were tested: 4 with high cholesterol content (SUDHL4, HBL1, HT, and U2932; 5.0-8.0 µg/mg protein), and 2 with low cholesterol content (DOHH2 and OCI-LY19; 1.5-2.0 µg/mg protein); the latter showed the highest sensitivity to DXR-mediated killing. The combination of lovastatin and DXR (10nM) was tested in all 4 cell lines with high cholesterol content, resulting in more cell death than either treatment alone. Lovastatin (at the nanomolar range) resensitized DXR-resistant SUDHL4 cells to DXR. Finally, in a DXR-resistant PDX model, the combination of lovastatin and DXR resulted in delayed tumor growth as compared to chemotherapy alone. Conclusions: Use of medium potency statins is associated with improved outcomes after frontline RCHOP in patients with DLBCL. This was further confirmed in functional in-vitro and in-vivo studies. Future interventional studies, aimed at improving outcomes in these patients using this novel combination, are warranted. Disclosures Westin: Amgen: Consultancy; 47: Research Funding; Kite: Consultancy, Research Funding; BMS: Consultancy, Research Funding; Morphosys: Consultancy, Research Funding; Janssen: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Genentech: Consultancy, Research Funding; Curis: Consultancy, Research Funding; Astra Zeneca: Consultancy, Research Funding. Nastoupil:Gamida Cell: Honoraria; Merck: Research Funding; TG Therapeutics: Honoraria, Research Funding; Karus Therapeutics: Research Funding; Janssen: Honoraria, Research Funding; LAM Therapeutics: Research Funding; Novartis: Honoraria, Research Funding; Bayer: Honoraria; Celgene: Honoraria, Research Funding; Genentech, Inc.: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding; Gilead/KITE: Honoraria. Neelapu:Bristol-Myers Squibb: Other: personal fees, Research Funding; Merck: Other: personal fees, Research Funding; Kite, a Gilead Company: Other: personal fees, Research Funding; Pfizer: Other: personal fees; Celgene: Other: personal fees, Research Funding; Novartis: Other: personal fees; Karus Therapeutics: Research Funding; N/A: Other; Takeda Pharmaceuticals: Patents & Royalties; Acerta: Research Funding; Cellectis: Research Funding; Poseida: Research Funding; Precision Biosciences: Other: personal fees, Research Funding; Legend Biotech: Other; Adicet Bio: Other; Allogene Therapeutics: Other: personal fees, Research Funding; Cell Medica/Kuur: Other: personal fees; Calibr: Other; Incyte: Other: personal fees; Unum Therapeutics: Other, Research Funding. Landgraf:NCI/NIH: Research Funding. Vega:NCI: Research Funding.


1977 ◽  
Author(s):  
Christine N. Vogel ◽  
Kingdon S. Henry ◽  
Roger L. Lundblad

Our intention is to study the interaction of rabbit thrombin with antithrombin III (AT-III) in vitro and in vivo. After activation of crude prothrombin with tissue thromboplastin and CaCl2, thrombin was purified and showed two species of thrombin with molecular weights of 36,000 and 39,000 daltons as determined by sodium dodecyl sulfate discontinuous gel electrophoresis. Rabbit AT-III was purified using a heparin agarose column and had a molecular weight of 55,000 daltons. The inhibition of thrombin by AT-III was followed by fibrinogen clotting assays and an AT-III-thrombin complex was observed on gel electrophoresis. For the in vivo studies both thrombin and AT-III were radiolabelled with Na125i using the solid state lactoperoxidase method and retained 99% of the pre-iodinated specific activity. Radiolabelled thrombin and a radiolabelled AT-III-thrombin complex were injected into different rabbits. The rate of removal of both was very similar with a half-life of approximately 9 hours. When radiolabelled AT-III was injected, the half-life was approximately 60 hours. Since the disappearance rate of thrombin more closely approximates that of the preformed AT-III-thrombin complex and is clearly shorter than the turnover rate of AT-III, the possibility is raised that thrombin combines in vivo with a native inhibitor such as AT-III and may in fact be removed from the circulation as a complex rather than as a native molecule.


2008 ◽  
Vol 99 (02) ◽  
pp. 271-278 ◽  
Author(s):  
Olivier D. Christophe ◽  
Beatrijs D. Oortwijn ◽  
Peter J. Lenting ◽  
Cécile V. Denis

SummaryThe life cycle of von Willebrand factor (VWF) comprises a number of distinct steps, ranging from the controlled expression of theVWF gene in endothelial cells and megakaryocytes to the removal of VWF from the circulation. The various aspects of VWF clearance have been the objects of intense research in the last few years, stimulated by observations thatVWF clearance is a relatively common component of the pathogenesis of type 1 von Willebrand disease (VWD). Moreover, improving the survival of VWF is now considered as a viable therapeutic strategy to prolong the half-life of factor VIII in order to optimise treatment of haemophilia A. The present review aims to provide an overview of recent findings with regard to the molecular basis of VWF clearance. A number of parameters have been identified that influence VWF clearance, including its glycosylation profile and a number of VWF missense mutations. In addition, in-vivo studies have been used to identify cells that contribute to the catabolism of VWF, providing a starting point for the identification of receptors that mediate the cellular uptake ofVWF.Finally, we discuss recent data describing chemically modification of VWF as an approach to prolong the half-life of the VWF/FVIII complex.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1592-1592 ◽  
Author(s):  
Lee A. Honigberg ◽  
Ashley M. Smith ◽  
Jun Chen ◽  
Patti Thiemann ◽  
Erik Verner

Abstract There is increasing evidence indicating that B-cell-receptor (BCR) signaling is required for survival of non-Hodgkin’s lymphoma (NHL) cells. Bruton’s tyrosine kinase (Btk) is required for BCR signaling and mutations that inactivate human Btk cause X-linked-agammaglobulinemia, a B-cell immunodeficiency. Although Btk functions selectively in B cells, the Btk active site is structurally similar to the active site in several Src and Abl kinases and as a result, there have been few highly selective small molecule inhibitors of Btk. We have developed a series of covalent Btk inhibitors that target Cys-481 in Btk and this approach results in increased potency and selectivity over related kinases that lack a Cys residue at this position (ChemMedChem 15:58). PCI-32765 is a Cys-481 targeting Btk inhibitor that has been optimized for potency, selectivity and pharmacokinetics. In cellular assays, PCI-32765 inhibits BCR-stimulus induced phosphorylation of Phospholipase-C-gamma, a Btk substrate, as well as downstream phosphorylation of Erk (IC50 < 100 nM). In addition, PCI-32765 induces apoptosis and inhibits proliferation in a subset of NHL cell lines including DHL-4, DHL-6, WSU-DLCL2, OCI-Ly10 and DOHH2 (IC50s = 0.6–1.6uM). We have used RNAi knockdown in DOHH2 cells as an independent method to confirm that Btk is required for lymphoma cell proliferation. In vivo, orally dosed PCI-32765 (50mg/kg) inhibits growth of DOHH2 and WSU-DLCL2 xenografts. PCI-32765 also prevents disease progression in a mouse collagen-induced arthritis model (12.5mg/kg PO), indicating that other B cell lineage diseases are sensitive to Btk inhibition. In order to further characterize the selectivity and in vivo potency of PCI-32765, we have developed PCI-33380, an active-site probe consisting of a covalent Btk inhibitor linked to the fluorophore Bodipy-FL. PCI-33380 binds to Btk and can be detected by flow cytometry or by denaturing gel electrophoresis of cell lysates. In cell lysates, the probe labels a single predominant band of the same molecular weight as Btk and this band is absent in cells from xid mice. Labeling of this band is inhibited (IC50=10nM) by a brief pre-treatment of cells with PCI-32765, indicating that the probe can be used to assess occupancy of Btk by a covalent inhibitor. We have used the probe to quantitate the inhibition of Btk by PCI-32765 in vivo. A single oral dose of PCI-32765 (10mg/kg) delivered to mice leads to rapid and complete inhibition of Btk in spleen. In addition, a single oral dose of PCI-32765 fully inhibits Btk in xenograft tumors and peripheral blood cells and this inhibition is maintained for up to 24hr. The Btk probe provides pharmacodynamic measurements that may allow optimization of dosing and schedule for in vivo studies and we are currently adapting the probe assays for use in monitoring the inhibition of Btk in human clinical trials.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3345-3345
Author(s):  
Anargyros Xenocostas ◽  
Benjamin D Hedley ◽  
Jenny E Chu ◽  
D. George Ormond ◽  
Michel Beausoleil ◽  
...  

Abstract Abstract 3345 Background: Erythropoietin (EPO) is a key regulator of erythropoiesis, and has been shown to stimulate growth, maintain viability, and promote differentiation of red blood cell precursors. The EPO receptor (EPO-R) is expressed by erythroid cells and by several non-hematopoietic cell types including various neoplastic cells. Erythropoiesis-stimulating agents (ESAs) are used clinically for the treatment of chemotherapy-induced anemia. The results of some recent randomized clinical trials have reported an increased incidence in adverse events and reduced survival in ESA-treated metastatic breast cancer patients receiving chemotherapy, potentially related to EPO-induced cancer progression. These results have raised concerns over ESA treatment in metastatic cancer patients. However, very little pre-clinical data is available regarding the impact of EPO on breast cancer metastasis. The goal of the current study was therefore to determine if EPO can influence the malignant behavior of breast cancer cells and/or influence the metastatic process. Methods: MDA-MB-468, MDA-MB-231, MDA-MB-435, and 4T-1 breast cancer cell lines were treated with recombinant human EPO (rHuEPO; 10 U/ml) or control media and screened for EPO-R mRNA expression levels by RT-PCR, and for EPO-R protein expression by Western blot and flow cytometry. MDA-MB-231 (231) and MDA-MB-435 (435) cell lines were used for functional assays in vitro and in vivo. Untreated or rHuEPO treated cells were grown in 2D and 3D in vitro systems (standard tissue culture plates and 0.6% soft agar, respectively) to determine if rHuEPO influenced growth. In vitro cell survival was also assessed in response to treatment with rHuEPO in the presence or absence of paclitaxel chemotherapy (10mg/ml), radiation (10G), or hypoxic conditions (1% O2). Following mammary fat pad injection, in vivo effects of rHuEPO (300U/kg) alone or in combination with paclitaxel treatment (10mg/kg) were assessed in mouse models of tumorigenicity and spontaneous metastasis. Results: Expression analysis of EPO-R mRNA and protein revealed a large variation in levels across different cell lines. The majority of cell lines did not express cell surface EPO-R by flow cytometry, although two cell lines (231 and 435) did show weak expression of EPO-R mRNA, with only the 231 cell line showing EPO-R expression by Western blot. In vitro, a small protective effect from rHuEPO on radiation-treated 435 cells was seen (p<0.05); however, rHuEPO treatment alone or combined with chemotherapy or hypoxia did not cause a significant increase in cell survival relative to untreated controls cells. In contrast, in vivo studies demonstrated that rHuEPO increased the incidence and burden of lung metastases in immunocompromised mice injected with 231 or 435 cells and treated with paclitaxel relative to mice treated with paclitaxel alone (p<0.05). Conclusions: The lack of an in vitro effect of rHuEPO highlights the importance of in vivo studies to delineate the effects of EPO on the metastatic process. Our novel findings demonstrate that rHuEPO can reduce the efficacy of chemotherapy in the metastatic setting in vivo, and in some cases enhance the inherent metastatic growth potential of human breast cancer cells. This work was supported by funding from the London Regional Cancer Program and Janssen Ortho Canada Disclosures: Xenocostas: Janssen Ortho: Consultancy, Honoraria, Research Funding. Allan:Janssen Ortho: Research Funding.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 808-808 ◽  
Author(s):  
Raajit K. Rampal ◽  
Suveg Pandey ◽  
Omar Abdel-Wahab ◽  
Jennifer J Tsai ◽  
Taghi Manshouri ◽  
...  

Abstract Abstract 808 A subset of patients with Philadelphia-chromosome negative myeloproliferative neoplasms (MPNs) (Polycythemia Vera (PV), Essential Thrombocytosis (ET), and Primary Myelofibrosis (PMF)) subsequently transform to acute myeloid leukemia (AML). Leukemic transformation (LT) after MPN occurs in as many as 23% of PMF patients within 10 years of diagnosis, and in 4–8% of PV and ET patients in the first 18 years after diagnosis. The development of AML after an antecedent MPN is associated with a dismal clinical outcome, and is associated with a poor response to conventional anti-leukemic therapies. Although somatic mutations in the JAK-STAT signaling pathway, including in JAK2 and MPL, occur in the majority of MPN patients, the somatic mutations that drive LT from a pre-existing MPN have not been fully delineated. Recent candidate mutational studies have identified recurrent somatic mutations in a subset of known leukemogenic disease alleles at the time of transformation from MPN to AML, including mutations in TP53, IDH1/2, TET2 and SRSF2 as well as deletions in IKZF1. However, the functional contribution of these specific genetic events to LT has not been delineated, and genetically accurate models of transformation of Philadelphia-chromosome negative MPN to AML have not been reported to date. In order to develop a genetically accurate murine model of LT, we have modeled expression of JAK2V617F mutation in combination with TP53 loss in vivo to further our understanding of progression from MPN to AML and to use this preclinical model of LT to test novel therapies. Bone marrow (BM) cells from C57/Bl6 Tp53−/− and littermate control mice were infected with JAK2V617F-IRES-GFP retrovirus, followed by transplantation of transduced cells into lethally irradiated congenic recipients. Of note, transplantation of JAK2V617F/Tp53−/− cells, but not JAK2V617F positive cells was associated with impaired survival; 50% of mice injected with JAK2V617F/Tp53−/− cells died by day 100, whereas all mice injected with JAK2V617F positive cells survived 100 days or longer (p=0.011) (figure 1). Mice injected with JAK2V617F/Tp53−/− cells presented with significant leukocytosis, with a mean WBC of 38.4 in mice engrafted with JAK2V617F/Tp53−/− cells compared with 11.4 in JAK2V617F/Tp53 wildtype mice. At the time of sacrifice, all mice engrafted with JAK2V617F/Tp53−/− cells had increased numbers of blasts in the peripheral blood and bone marrow, as assessed by morphologic evaluation and flow cytometric analysis which noted CD117 expression on leukemic blasts. BM cells from mice engrafted with JAK2V617F/Tp53−/− cells were characterized by increased serial replating (>10 platings), which was not observed in plating studies with JAK2V617F positive cells. In addition, we noted that the disease from JAK2V617F/Tp53−/− cells, but not JAK2V617F positive cells, was transplantable into secondary recipients consistent with increased self-renewal in vivo. We have begun testing the efficacy of novel therapies in this murine model, using both in vitro assays and in vivo studies in secondary transplantation studies. Treatment with the JAK kinase inhibitors INCB18424 and CYT 387 resulted in dose-dependent inhibition of colony formation in vitro. The combination of INCB18424 and Decitabine (which has demonstrated clinical efficacy in post-MPN-AML) is associated with synergistic inhibitory effects in vitro. Based on these results, we are performing in vivo studies with INCB18424, Decitabine, and INCB18424 + Decitabine, and results from these preclinical therapeutic studies will be presented in detail. Taken together, our data demonstrate that expression of JAK2V617F plus Tp53 loss, a genoptype commonly seen in patients who transform to AML after MPN, efficiently models LT in vivo. This model can now be utilized to examine the mechanisms of leukemic transformation, including assessment of the leukemic cell of origin in transformed disease. In addition this model can be utilized to test novel therapeutic strategies in a preclinical setting, which can be used to inform clinical trials in this poor-risk hematologic malignancy. Figure 1: Survival curve of mice transplanted with JAK2V617F in presence and absence of Tp53 Figure 1:. Survival curve of mice transplanted with JAK2V617F in presence and absence of Tp53 Disclosures: Verstovsek: Incyte Corporation: Research Funding; Novartis: Research Funding; AstraZeneca: Research Funding; Celgene: Research Funding; SBIO: Research Funding; Lilly Oncology: Research Funding; Bristol-Myers: Research Funding; Geron Corp.: Research Funding; Gilead: Research Funding; YM Biosciences: Research Funding; Roche: Research Funding; NS Pharma: Research Funding; Infinity Pharmaceuticals: Research Funding.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2333-2333
Author(s):  
Pamela R. Westmark ◽  
Pansakorn Tanratana ◽  
John P. Sheehan

Abstract Introduction Hemophilia B is an X-linked genetic disorder characterized by defective factor IX activity. Recombinant factor IX (rFIX) is employed as protein replacement for the treatment and prophylaxis of bleeding episodes. Antithrombin is the primary plasma inhibitor of activated factor IX (FIXa), and inhibition is enhanced by heparin/heparan sulfate. We hypothesize that selective disruption of protease interactions with heparin and antithrombin via mutations in the respective heparin- and antithrombin-binding exosites may enhance rFIX(a) efficacy by prolonging protease half-life in vivo. Aim To assess the effect of mutations in the FIX(a) heparin- and antithrombin-binding exosites on traditional coagulant activity, thrombin generation, and protease half-life in human plasma. Methods Human FIX cDNA constructs with alanine substitutions (chymotrypsinogen numbering) in the heparin exosite (K126A, K132A, K126A/K132A), antithrombin exosite (R150A), or both (K126A/R150A, K132A/R150A, K126A/K132A/R150A) were expressed in HEK293 cell lines. Recombinant zymogens were purified from conditioned media, and a portion activated to protease with human factor XIa. Zymogen and protease forms were characterized in APTT-based clotting assays, and tissue factor (TF) and FIXa-initiated thrombin generation (TG) assays in pooled human FIX-deficient plasma, respectively. Comparisons were made with human plasma-derived factor IX (pFIX) and recombinant FIX wild type (WT). Protease half-life in pooled, citrated human plasma was determined using a novel assay that detects FIXa activity by TG response. Results Zymogen coagulant activities (% WT ± S.E) were: pFIX 105.2 ± 2.8, WT 100 ± 7.1, K132A/R150A 75.8 ± 3.4, K126A 63.3 ± 2.3, R150A 62.4 ± 4.0, K132A 30.9 ± 1.0, K126A/R150A 27.0 ± 2.1, K126A/K132A 20.6 ± 9.2, and K126A/K132A/R150A 7.3 ± 3.8. Similarly, protease coagulant activities were: WT 100 ± 6.1, pFIXa 98.4 ± 11.4, K132A 91.4 ± 1.6, K132A/R150A 84.9 ± 2.8, R150A 77.1 ± 5.8, K126A 39.5 ± 2.4, K126A/R150A 25.3 ± 2.8, K126A/K132A/R150A 10.9 ± 0.6, and K126A/K132A 9.3 ± 0.6. In contrast to their relative coagulant activities, FIX K126A (1.9-fold), R150 (1.6-fold), and K132A/R150A (1.3-fold) supported increased peak thrombin concentrations during TF-triggered TG; pFIX, FIX K132A and K126A/R150A were similar to WT; and FIX K126A/K132A/R150A (0.6-fold) and K126A/K132A (0.2-fold) demonstrated marked reductions in peak thrombin relative to WT. In the FIXa-initiated TG assay, FIXa K126A/R150A and K132A/R150A (1.5-fold) demonstrated significantly increased peak thrombin concentrations; pFIXa, FIXa K132A, R150A, and K126A (0.8-1.0 fold) were similar to WT; while FIXa K126A/K132A and K126A/K132A/R150A demonstrated markedly reduced (0.2-0.3 fold) and delayed peak thrombin concentrations. In pooled, citrated FIX-deficient plasma, FIXa WT (40.9 ± 1.4 min) and K126A/K132A (37.2 ± 0.7 min) demonstrated similar half-lives, while FIXa R150A, K126A/R150A, and K132A/R150A all had half-lives > 2 hr. Conclusions Single exosite mutations resulted in mild to moderate reductions in coagulant activity, while the double mutation in the heparin exosite (K126A/K132A) markedly reduced activity, likely due to a synergistic effect on cofactor binding. Traditional coagulant activity did not accurately represent the ability of the mutant proteins to support thrombin generation. Despite variable reductions in coagulant activity, FIX K126A, K132A, R150A, K126A/R150A and K132A/R150A supported levels of plasma thrombin generation that were equal to or greater than FIX WT. The plasma half-life of FIXa WT activity was remarkably lengthy, and while mutations in the heparin exosite had negligible effects, R150A in the antithrombin exosite substantially increased protease half-life, consistent with a primary role for antithrombin in the plasma inhibition of FIXa. Thus, single exosite mutations did not significantly disrupt the procoagulant function of human FIX(a), and combined exosite mutations (K126A/R150A and K132A/R150A) maintain or enhance plasma thrombin generation while disrupting exosite-mediated regulatory mechanisms. The combination of intact procoagulant function with disruption of antithrombin- and heparin-mediated regulation of FIX(a) will potentially enhance in vivo recovery, prolong plasma half-life, and enhance the efficacy of hemophilia B replacement therapy. Disclosures: Sheehan: Novo Nordisk Access to Insight Basic Research Grant: Research Funding; Bayer Hemophilia Awards Program: Research Funding; Diagnostica Stago: reagents, reagents Other.


Sign in / Sign up

Export Citation Format

Share Document