Successful Management of Refractory Severe Thrombocytopenia Associated with Antiphospholipid Syndrome Using rituximab

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4570-4570
Author(s):  
Soad Al Jaouni

Abstract Background: Rituximab, a monoclonal antibody against the pan B-cell antigen CD20, that induces a rapid in vivo depletions of normal B lymphocytes. Although this agent was originally developed for use in patients with B-cell-NHL, its use has been successfully extended to many autoimmune disorders. Thrombocytopenia associated with antiphospholipid syndrome can be mild to severe. Severe cases can be associated with significant morbidity and mortality if refractory to the usual therapy for autoimmune thrombocytopenia. Aim: To report an effective regimen in treating a case of severe bleeding thrombocytopenia associated with antiphospholipid syndrome. Patient and Methods: A 49 year old female was diagnosed with thrombocytopenia associated with antiphospholipid syndrome and antibodies to double-stranded DNA, controlled over the last 15 years. Unfortunately, over the last two years the patient has had frequent admissions for uncontrolled nose bleed, soft palate petechiae and generalized bruising. Many treatments have been tried for this case of difficult refractory bleeding with platelets in the range 2.0–6.0 K/uL. These include systemic treatments such as corticosteroids, high pulse therapy of methylprednisolone, Imuran, i.v. immunoglobulin infusion and anti Rh-D intravenous therapy. We report an adult female patient with severe refractory bleeding thrombocytopenia associated with antiphospholipid syndrome and successful treatment with Rituximab. Her platelets count have been maintained above 200.0K/uL over a 14 months period. Unfortunately, this patient has had side effects of steroid therapy. In conclusion: Rituximab may be effective treatment on patients with refractory, severe thrombocytopenia associated with antiphospholipid syndrome and significant bleeding complications. Rituximab is a promising alternative option for the eradication of the autoantibodies and restoration of normal hemostasis while avoiding the use of high-dose steroid in refractory bleeding thrombocytopenia.

2003 ◽  
Vol 3 (12) ◽  
pp. 1667-1675 ◽  
Author(s):  
Keith A. Cockerill ◽  
Eric Smith ◽  
David S. Jones ◽  
Michael J. Branks ◽  
Merle Hayag ◽  
...  

2006 ◽  
Vol 95 (02) ◽  
pp. 354-361 ◽  
Author(s):  
Oliver Berg ◽  
Axel Heimann ◽  
Thomas Münzel ◽  
Christian-Friedrich Vahl ◽  
Oliver Kempski ◽  
...  

SummaryApplication of clopidogrel before percutaneous coronary intervention in patients with acute coronary syndrome reduces the risk of cardiac events. Clopidogrel administration before surgery increases bleeding complications after CABG. Therefore, the antithrombotic effect of the low-dose combination of clopidogrel and aspirin was investigated in an in vivo pig model of coronary artery thrombus formation with cyclic flow reductions. The platelet inhibitory effect was determined by platelet aggregation and CFR, according to the methodology described by Folts. CFR were initiated by endothelial damage and placement of a constrictor around the LAD. 30 min after CFR were established, clopidogrel (0.1 mg/kg or5 mg/kg), aspirin (1 mg/kg or 7 mg/kg) or LDC (0.1 mg/kg clopidogrel and 1 mg/kg aspirin) were administered orally. CFR-frequency was determined for further 240 min. CFR-frequency (CFR/30 min) was significantly reduced at 60 min in response to aspirin (7 mg/kg, −48%, p<0.05), and at 120 min in response to clopidogrel (5 mg/kg, −65%, p<0.05) but not at low doses of either compound. In contrast, LDC of clopidogrel (0.1 mg/kg) plus aspirin (1 mg/kg) resulted in a complete and rapid abrogation of CFR at 90 min (−70%, p<0.05). Furthermore, LDC led to reduction of platelet aggregation when CFR-frequency was already significantly decreased. In contrast, high dose groups presented a significant reduction of platelet aggregation prior to CFR-frequency decrease. Low dose combination of clopidogrel plus aspirin demonstrates a potent over additive anti-thrombotic effect in vivo with a significant reduction in thrombus formation early after drug application. The effect occurs before inhibition of platelet aggregation is detectable.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3566-3566
Author(s):  
Marco L Davila ◽  
Isabelle Riviere ◽  
Xiuyan Wang ◽  
Jae H. Park ◽  
Jolanta Stefanski ◽  
...  

Abstract Abstract 3566 B cell acute lymphoblastic leukemia (B-ALL) in adults has a dismal prognosis. Intensified, combinatorial chemotherapy yields remarkable results in pediatric ALL but less so in adults. As allogeneic hematopoietic stem cell transplantation (HSCT) is curative in only a minority of patients, there remains a dire need for effective therapies in this patient population. Building on our results obtained with genetically targeted T cells in xenogeneic models of ALL and our experience in patients with chronic lymphocytic leukemia (Brentjens, Rivière, Blood, 2011), we have recently initiated a clinical trial (NCT01044069) treating adults with relapsed or refractory B-ALL with autologous T cells expressing the 19–28z chimeric antigen receptor (CAR), a dual CD3zeta/CD28 signaling receptor specific for the B cell antigen CD19. Human peripheral blood T cells retrovirally transduced to express the 19–28z receptor specifically lyse normal and malignant B cells in vitro and eradicate established tumors in vivo in pre-clinical animal models. We report the results from the initial cohort of patients treated on this protocol. The first two patients had relapsed disease that achieved morphologic remission following re-induction chemotherapy. Following adoptive therapy with CAR-modified T cells, one patient achieved a B cell aplasia and molecular remission documented by deep sequencing, which demonstrated the disappearance of IgH rearrangements associated with the malignant clone. The other patient had achieved a molecular remission after re-induction chemotherapy but further developed a complete B cell aplasia following modified T cell infusion. While in molecular remission, both patients successfully underwent allogeneic HSCT and were therefore removed from the study. In contrast to these two patients, the third patient failed to achieve a morphologic remission after re-induction chemotherapy and had >60% blasts in the bone marrow (BM) at the time of T cell infusion. Within 12 hours of completing T cell infusion, the patient developed high-grade fevers, hypotension, and rigors. Serum analyses demonstrated a sharp rise in cytokines (IFNg, TNFa, IL6, IL2, and IL8), reflecting rapid and robust onset of T cell activation. High-dose corticosteroids initiated 5 days after T cell infusion controlled these symptoms. Post-T cell BM aspirate on day 8 demonstrated undetectable blasts. Flow cytometry could not detect blasts or B lineage cells, but readily identified 19–28z CAR+ T cells. Significantly, the patient's B-ALL tumor cells harbored a unique 9p21 cytogenetic deletion that was detected by FISH in 28% of nuclei immediately prior to T cell infusion and was reduced to 5% 8 days after T cell infusion. The next BM aspirate, performed on day 24 after T cell infusion, demonstrated a persistent morphologic remission, a recovering BM, and complete absence of any detectable 9p21 deletions. Furthermore, PCR amplification for IgH rearrangements in the BM 24 days after T cell infusion confirmed a molecular remission and a B cell aplasia. Collectively, the clinical outcomes from this initial cohort of 3 patients demonstrate for the first time the in vivo efficacy of CD19-targeted T cells to induce clinical and molecular remissions as well as B cell aplasia in adults with relapsed or refractory B-ALL. These early results strongly support further investigation of 19–28z targeted T cells to treat leukemias and suggest this is a potential salvage therapy for patients with relapsed/refractory B-ALL who are failing chemotherapy or are ineligible for allogeneic HSCT. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5612-5612
Author(s):  
Li Can ◽  
Kalyan Nadiminti ◽  
Yuqi Zhu ◽  
Yogesh Jethava ◽  
Ivana Frech ◽  
...  

Abstract Background; Major progress in the treatment of B cell tumors has been made in the past decades. Nevertheless, relapses and refractoriness to currently available chemotherapy and even to high dose therapy with stem cell transplantation still cause significant mortality. NEK2, NEver in Mitosis Gene A (NIMA)-Related Kinase 2, is a serine/threonine kinase. High expression of NEK2 increases cell survival and drug resistance, resulting in poor clinical outcome in multiple cancers including multiple myeloma and lymphoma. In this study, we used genetic mouse models to evaluate whether NEK2 is a druggable target in the treatment of B cell tumors including myeloma and diffuse large B-cell lymphoma (DLBCL). Materials and Methods: We have generated Nek2 knockout mice and crossed these with Eµ-Myc mice. RNA-sequencing was performed to determine signaling pathways related to Nek2 inhibition. Both NEK2 and USP7 (a protein interacting with NEK2) inhibitors were applied to treat myeloma and DLBCL in vitro and in vivo. Results: Mouse studies showed that Nek2 played a critical role in B cell tumor development and progression. Specifically, in genetic Eμ-MYC transgenic mice, which spontaneously develop DLBCL and Burkitt lymphoma, knockout of Nek2 prevented B cell tumor development and significantly extended mouse survival. Further, immunohistochemistry analyses showed that Nek2 was highly detected in biopsies from aggressive Burkitt lymphoma patients. Our data also indicate that both NEK2 and USP7 inhibitors significantly inhibited myeloma cell and lymphoma cell growth in vitro and in vivo models and without apparent toxicity to normal tissues. Intriguingly, the combination of USP7 inhibitor P5091 with doxorubicin blocked B cell lymphoma development and extended lymphoma mouse survival. Conclusions: Our studies demonstrate the importance of Nek2 function in tumorigenesis and progression in B cell lineage malignancies. Both NEK2 and USP7 inhibitors showed excellent efficacy in the treatment of myeloma and B-cell lymphoma. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 5 (Supplement_1) ◽  
Author(s):  
Salema Khalid ◽  
Steven Young Min

Abstract Case report - Introduction Antiphospholipid syndrome (APS) is a rare autoimmune disease that can cause venous and arterial thrombosis in virtually any organ. The spectrum of vascular events can range from superficial thrombosis to life-threatening multiple organ thromboses (catastrophic APS or CAPS). CAPS occurs in genetically susceptible individuals in response to a “trigger” such as infection, cancer, trauma, surgery, anticoagulation/immunosuppression withdrawal and SLE flares. The diagnosis of CAPS can be extremely challenging and is associated with a high morbidity and mortality. Thus, early diagnosis and treatment are critical to prevent the progression of disease and improve the prognosis. Case report - Case description We report the case of a 78-year-old gentleman who was diagnosed with systemic lupus erythematosus and antiphospholipid syndrome in 2001 after he presented with a DVT, PE, rash and arthralgia. He had positive anti-cardiolipin antibodies, Rheumatoid Factor, Ro and La antibodies, but negative anti-dsDNA. He had remained stable on warfarin, hydroxychloroquine 400mg and prednisolone 7mg for 17 years. In 2018, hydroxychloroquine was reduced to 200mg OD and steroid taper was started. Unfortunately, he presented to the Emergency Department in July 2020 with a left leg swelling. DVT was confirmed on ultrasound, despite a therapeutic INR of 2.4. He was also noted to have thrombocytopenia. Haematology advised this was in keeping with ITP and started him on 70mg of prednisolone daily. No cause for the DVT was seen on CT. However, it did show subpleural nodules within the right costophrenic angle and a repeat CT in 4 months’ time was advised. INR target was increased to 3.0—4.0 and patient was discharged. He was re-admitted 4 days later with an acute drop in haemoglobin, raised inflammatory markers and worsening kidney function. CT showed extensive retroperitoneal haematoma. It also revealed a PE as well as colonic distension with gradual tapering to normal calibre, thought to represent pseudo-obstruction. Rheumatology, haematology, general surgery and ITU were involved in the management. He was started on treatment dose clexane, given intravenous immunoglobulins and supportive blood transfusions. IVC filter was put in. Unfortunately, he dropped his GCS and an urgent CT brain showed a left posterior fossa mass with a bleed. The case was discussed with neurosurgery and neuroradiology who felt that the top differential for the intracranial lesion was an underlying metastasis – particularly a colonic met. Colonoscopy was advised. However, due to severe frailty and multiple pathologies, the patient was made palliative and was fast-tracked home. Case report - Discussion Definite CAPS is defined as thromboses in three or more organs developing in less than a week, microthrombosis in at least one organ and persistent antiphospholipid antibody (aPL) positivity. The diagnosis of probable CAPS requires three out of these four criteria. Although pathological confirmation of microthrombosis is one of the requirements for CAPS, biopsy may not be possible during an acute episode due to severe thrombocytopenia and/or unstable clinical course, as in our case. There is another category called ‘CAPS-like’ disease, where aPL-positive patients do not fulfil the definite or probable CAPS criteria. However, they still represent a significant challenge for physicians and require close monitoring and aggressive treatment. Initially, we felt that we had triggered probable CAPS or ‘CAPS-like’ disease, by reducing his hydroxychloroquine and steroids. However, he did not improve with high-dose steroids given for his thrombocytopenia. Also, autoimmune screen including anti-dsDNA and complement levels were not significant. CAPS occurs in 46% of patients with a previous diagnosis of APS, and a precipitating factor is present in half the patients. It is speculated that aPL-related clinical events respond to the two-hit theory: a second hit or trigger is needed to activate the prothrombotic properties of aPL, which is the first hit. In CAPS, the most frequently recognised trigger is infection, followed by cancer. A study showed that 9% of patients with CAPS presented with an underlying malignancy, with haematological malignancies being most common, followed by lung and colon carcinoma. Similarly, Ozguroglue et al. showed an association between high level of anticardiolipin antibody and thromboembolic events in patients with colorectal, breast, ovarian, lung, and pancreatic cancer. Recent studies also suggest an increased prevalence of certain cancers in aPL-positive patients, thereby prompting an extensive search for an occult malignancy in such cases. Case report - Key learning points Given the increased prevalence of cancers in aPL-positive patients, this case highlights the need to thoroughly investigate for an occult malignancy as a trigger for APS (classic form or CAPS) with a new episode of thrombosis, despite adequate anticoagulation. While we were focusing on tapering of the immunosuppressive medication as a possible trigger, this episode was most likely triggered by the possible metastatic malignancy – especially given the lag of almost 2 years between reduction in hydroxychloroquine and steroids and development of symptoms. It is also important to bear in mind, especially in elderly patients, that thrombotic events associated with aPL can be the first manifestation of malignancy. This emphasises the need for continuing research on the association between antiphospholipid syndrome and malignancies. While the survival rate of patients with CAPS is poor overall, the outcome of patients with CAPS is worse in the presence of malignancy. A study showed that only 40% of CAPS patients with malignancies improved. This may be due to the presence of the malignancy as well as the older age of the patients. We are looking forward to discussing CAPS at the BSR case-based conference and hope it will shed more light on diagnosis and management of this incredibly challenging condition.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4134-4134
Author(s):  
Andreas Moriaitis ◽  
Lita Freeman ◽  
Robert Shamburek ◽  
Robert Wesley ◽  
Wyndham H. Wilson ◽  
...  

Abstract BACKGROUND: Acquired severe HDL deficiency is relatively uncommon. It may occur with use of high doses of anabolic steroids or in severe liver diseases, which can lead to low LCAT activity and decreased apoA-I production. “Disappearing HDL Syndrome”*, a term first used by Goldberg and Mendez, refers to cases of severe HDL deficiency in patients that are not critically ill, sometimes long before the clinical or biochemical features of the underlying primary disease become evident. Disappearing HDL Syndrome can also result from an idiosyncratic reaction to medications, such as peroxisome proliferation-activated receptor (PPAR) agonists. Additionally, autoantibodies against LCAT in non-Hodgkin lymphoma have been described as a possible cause. Low high-density lipoprotein-cholesterol (HDL-C) is a risk factor for coronary artery disease. OBJECTIVE: To determine the cause of low HDL-C in patients with severe acquired HDL deficiency noted as an incidental finding associated with lymphoma and autoimmune lymphoproliferative syndrome (ALPS)** as well as following recombinant human IL10 therapy in psoriatric arthritis patients. Investigating mechanisms underlying acquired severe HDL deficiency in non-critically ill patients (“Disappearing HDL Syndrome”) could provide new insights into HDL metabolism and its role in lymphomagenesis. PATIENTS AND RESULTS: Patients with intravascular large B-cell lymphoma (IVLBCL, n=2), diffuse large B-cell lymphoma (DLBCL, n=1), and ALPS-FAS (n=1) presenting with markedly decreased HDL-C, low LDL-C and elevated triglycerides were identified. The abnormal lipoprotein profile returned to normal following therapy in all four cases (Figure). All of them were found to have markedly elevated serum interleukin-10 (IL-10) levels at presentation that also normalized following therapy. Besides accumulation of abnormal lymphocytes in lymph nodes and spleen, ALPS patients have elevated serum IL-10. In a cohort of ALPS patients with genetic mutations in FAS (n=93), IL-10 showed a strong inverse correlation with HDL-C (R2=0á3720, P<0á0001). A direct causal role for increased serum IL-10 in inducing the observed changes in lipoproteins was established in a randomized, placebo-controlled clinical trial of recombinant human IL-10 (rhIL-10) in psoriatic arthritis patients (n=18). Within a week of initiating subcutaneous rhIL-10 injections, HDL-C precipitously decreased to near-undetectable levels. LDL-C also decreased by over 50% (P<0.0001) and triglycerides increased by approximately 2-fold (P<0.005). All values returned to baseline after stopping IL-10 therapy. CONCLUSION: Increased IL-10 causes severe HDL-C deficiency, low LDL-C and elevated triglycerides. IL-10 is thus a potent modulator of lipoprotein levels, a potential new biomarker for B-cell disorders, and a novel cause of Disappearing HDL Syndrome. Elevated IL-10 plays a key role in linking inflammation and lipoprotein metabolism. As evidence for its causality, rhIL-10 treatment in a clinical trial of psoriasitic arthritis patients precipitously lowered HDL-C. Finding elevated IL-10 as a cause of “Disappearing HDL Syndrome” in patients with 3 different types of B-cell disorders suggests that IL-10 and low HDL-C could be useful biomarkers of disease activity in these conditions. Finally, future research focusing on strategies to alter lipoprotein levels by modulating circulating IL-10 levels or its signaling pathways may be useful for developing novel targeted therapies. *Ref: Severe acquired (secondary) high-density lipoprotein deficiency. Goldberg RB, Mendez AJ. J Clin Lipidol. 2007;1:41-56 **Ref: Natural history of autoimmune lymphoproliferative syndrome associated with FAS gene mutations. Price S, Shaw PA, Seitz A et al. Blood. 2014 Mar 27;123(13):1989-99. Figure. Concomitant drop in IL-10 associated with HDL-C recovery during treatment in patients with B-cell disorders. IL-10 and HDL-C time course before and after chemotherapy in: (A, B) 2 IVLBCL and (C) 1 DLBCL patients. The first time point (“Day 0”) represents the last lipid time point before initiating chemotherapy (arrow). D. Recovery of HDL-C and IL-10 in an ALPS patient after treatment with sirolimus over a period of 6 years. P* denotes high dose (15 mg/day) prednisone pulse therapy in this 2 year old ALPS patient. Figure. Concomitant drop in IL-10 associated with HDL-C recovery during treatment in patients with B-cell disorders. IL-10 and HDL-C time course before and after chemotherapy in: (A, B) 2 IVLBCL and (C) 1 DLBCL patients. The first time point (“Day 0”) represents the last lipid time point before initiating chemotherapy (arrow). D. Recovery of HDL-C and IL-10 in an ALPS patient after treatment with sirolimus over a period of 6 years. P* denotes high dose (15 mg/day) prednisone pulse therapy in this 2 year old ALPS patient. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1100-1100
Author(s):  
Joshua Eubanks ◽  
Wallace Hunter Baldwin ◽  
Rebecca Markovitz ◽  
Ernest T Parker ◽  
Shannon L. Meeks

Abstract Up to 30% of patients with severe hemophilia A will develop inhibitory antibodies to factor VIII (fVIII inhibitors). In addition, autoimmune antibodies to fVIII can develop in non-hemophiliacs, producing acquired hemophilia A, which frequently produces life- or limb-threatening bleeding. Patients with congenital hemophilia who develop inhibitors usually have a polyclonal antibody response directed against the A2 and C2 domains of fVIII. Patients with acquired hemophilia typically have a more limited B-cell epitope response with antibodies directed against the A2 or C2 domain but not both. We have shown that within the C2 domain of fVIII antibody epitope is more important than inhibitory titer in predicting pathogenicity in a murine in vivo bleeding model. In this project we investigated the pathogenicity of a diverse panel of anti-A2 monoclonal antibodies (Mabs) in the murine in vivo bleeding model. We have previously characterized anti-A2 antibodies into groups A, AB, B, BCD, C, D, DE, and E based on the pattern of overlap on the B-cell epitopes in a competition ELISA. Table 1 shows the characteristics of the anti-A2 Mabs. Mabs were injected retro-orbitally into Exon 16 hemophilic (E16) mice at a dose of 0.5 umg per g body weight (∼ 65nM plasma concentration). Fifteen minutes later, mice were injected with B-domain deleted human fVIII at a dose of 180U/kg (∼ 2.5nM plasma concentration). In addition, a subset of Mabs has also been tested at a high dose of 360U/kg (∼5 nM). Two hours after fVIII injection, the mice were anesthetized and a 4mm tail snip was performed. Blood was collected in a tube of normal saline over 40 minutes and measured. 4A4, 2-76, and 1D4 are all high inhibitory titer, type I Mabs that produced significant bleeding with 180 U/kg fVIII when compared to control. In addition, 2-76 and 4A4 were tested at the higher dose of fVIII and significant bleeding was again seen. In comparison, the high titer type II Mab 2-54 had a similar inhibitory titer but no significant bleeding at either dose of fVIII. B94 is a type II inhibitor with a similar inhibitory profile to 2-54, but maximum inhibition is 45% as compared to 82% for 2-54. B94 also was not pathogenic at either fVIII dose tested. Both 4C7—a non-inhibitory Mab—and B25—a very low titer Mab that would be predicted to have residual fVIII activity at the Mab concentration tested—did not produce significant bleeding. The inhibitory titer alone did not predict bleeding phenotype within a diverse panel of anti-A2 Mabs. This discrepancy combined with similar findings in the C2 domain stress the importance of inhibitor properties not detected in the standard Bethesda assay in predicting response to fVIII therapy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2232-2232 ◽  
Author(s):  
Michele Mumaw ◽  
Maria de la Fuente ◽  
Carolyn Aldana ◽  
Wei Li ◽  
Marvin T Nieman

Abstract The regulation of hemostasis and thrombus formation is a tightly controlled event that has catastrophic consequences when it is deregulated. One of the hallmarks of the thrombus is aggregated platelets. Upon platelet stimulation, adhesion molecules become activated and mediate multiple cell-cell interactions. Therapeutically, blocking platelet adhesion is a proven method for preventing pathological arterial thrombus formation. However, targeting the primary adhesion receptor, integrin αIIbβ3, results in severe bleeding complications. Therefore, identifying novel proteins or uncovering novel functions for known proteins in platelets is a necessary first step to facilitate the development of safer anti-platelet therapeutics. We have identified that the cell adhesion molecule cadherin-6 forms a functional adhesion complex with α-catenin and β-catenin in platelets. The goal of our project was to determine the mechanism of cadherin-6 mediated adhesion in platelets. Our initial experiments demonstated that cadherin-6 and β-catenin co-localize at the plasma membrane in platelets using confocal immunofluorescence microscopy. We determined that α-catenin and β-catenin co-immunoprecipitate with cadherin-6 from platelet lysates. To examine the functional role of cadherin-6 on platelet aggregation we used a cadherin-6 blocking antibody (10 μg/ml). Blocking cadherin-6 inhibited mouse platelet aggregation induced by PAR4 peptide. We next determined the role of cadherin-6 in vivo by examining carotid artery thrombosis after 7.5% FeCl3 treatment. C57Bl6 mice were injected with cadherin-6 antibody IV and labeled with rhodamine 6G by jugular vein injection. Thrombus formation was imaged in real time by fluorescent intravital microscopy. Blocking cadherin-6 prevented thrombosis for the duration of the experiment (30 min). To verify that the effects that we observed were specific to cadherin-6 expressed on platelets, we isolated platelets from donor mice and treated with cadherin-6 antibody or control IgG ex vivo. The treated platelets were perfused into recipient mice that were irradiated with 11 Gy to make the animals thrombocytopenic. The cadherin-6 antibody treated platelets formed an occlusion at 26.4 ± 3.6 min vs. 13.7 ± 2.0 min for the IgG (p=0.03). Importantly, the cadherin-6 antibody did not affect platelet counts compared to IgG controls 2.97 ± 0.40 (×108) vs. 3.02 ± 0.20 (×108). These combined studies show that caderhin-6 forms a complex with the necessary proteins required to mediate adhesion in platelets. Our results demonstrate that platelet cadherin-6 has a physiologically important role during platelet activation and thrombus formation in vivo. In summary, we have identified a novel adhesion complex in platelets that may provide a mechanism to limit platelet aggregation therapeutically. On going studies will determine the regulation of the cadherin-6/catenin complex and how cadherin-6 cooperates with other platelet adhesion molecules. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 2017 ◽  
pp. 1-8 ◽  
Author(s):  
Chen Zhang ◽  
Rui Zeng ◽  
Zhencheng Liao ◽  
Chaomei Fu ◽  
Hui Luo ◽  
...  

The human body cannot control blood loss without treatment. Available hemostatic agents are ineffective at treating cases of severe bleeding and are expensive or raise safety concerns.Bletilla striataserve as an inexpensive, natural, and promising alternative. However, no detailed studies on its hemostatic approach have been performed. The aim of this study was to examine the hemostatic effects ofB. striataMicron Particles (BSMPs) and their hemostatic mechanisms. We prepared and characterized BSMPs of different size ranges and investigated their use as hemostatic agent. BSMPs of different size ranges were characterized by scanning electron microscope. In vitro coagulation studies revealed BSMP-blood aggregate formation via stereoscope and texture analyzers. In vivo studies based on rat injury model illustrated the BSMP capabilities under conditions of hemostasis. Compared to other BSMPs of different size ranges, BSMPs of 350–250 μm are most efficient in hemostasis. As powder sizes decrease, the degree of aggregation between particles and hemostatic BSMP effects declines. The BSMP in contact with a bleeding surface locally forms a visible particle/blood aggregate as a physical barrier that facilitates hemostasis. Considering the facile preparation, low cost, and long shelf life ofB. striata, BSMPs offer great potential as mechanisms of trauma treatment.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5347-5347
Author(s):  
Yasuo Hirayama ◽  
Kyuhei Kohda ◽  
Yasuji Hirata ◽  
Hiroyuki Kuroda ◽  
Takuya Matsunaga ◽  
...  

Abstract Chemotherapy with Rituximab is widely used to treat patients with various B-cell lymphomas and auto-transplantation with Rituximab is promising strategy due to the potential for in vivo purging. However, the possibility of late onset neutropenia and immunoglobulin suppression after auto-transplantation with Rituximab has been indicated. We studied the frequency and degree of these phenomena. We performed a retrospective analysis on 26 consecutive patients at three centers during the period of January 1998 to March 2005. Thirteen patients (Follicular 8, Marzinal zone B cell lymphoma 2, Diffuse large B 3) received auto-transplantation without Rituximab (R-) compared with 13 patients (Follicular 8, MZBCL 2, DLB 3) received auto-transplantation with Rituximab (R+). In R+ patients pripheral blood stem cells were harvested after High dose AraC followed by three times of Rituximab (375mg/m2 day -2 of AraC, day7, 14). Conditioning regimen consisted of MCEC (MCNU, CBDCA, ETOP, Cy) or TBI+Cy followed by three times of Rituximab (375mg/m2 day 0, 7, 14). Mean immunoglobulin concentration one month after transplantation was 890 mg/dl for R- vs. 470 mg/dl for R+ (P=0.04). Lowest neutrophil numbers over 4 weeks after transplantation was 1.24X109/L for R- vs. 0.36X109/L for R+ (p=0.02). Late onset neutropenia (&lt;0.5X109/L) were seen in three cases of R+ group, but no case in R- group. Therapy related death was seen one case in R+ group. This case showed low immunoglobulin level after transplantation and died of Pneumocystis Carinii. These data, although preliminary, indicates that the addition of Rituximab to auto-transplantation leads to decrease in immmunoglobulin and neutrophil levels after transplantation.


Sign in / Sign up

Export Citation Format

Share Document