scholarly journals Development of Humanized Diffuse Large B-Cell Lymphoma Mouse Models

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4501-4501
Author(s):  
Syed Mehdi ◽  
Ying-Zhi Xu ◽  
Leonard Shultz ◽  
Samantha L. Kendrick ◽  
Donghoon Yoon

Abstract Introduction DLBCL is a commonly diagnosed, aggressive non-Hodgkin's lymphoma with ~40% of patients experiencing refractory or relapsed disease. Development of alternative therapies that target molecular features defining these unresponsive tumors is an active area of research to significantly advance the field and improve these patient's clinical management. However, few DLBCL animal models exist to test the efficacy of newly developed treatments, and are restricted to transgenic or xenograft mice that often fail to recapitulate the heterogeneous sub-classifications of this complex disease. While transgenic mice allow for spontaneous tumor formation, these models rely on inducing expression of specific oncogenes that drive a select group of DLBCL. The xenograft model offers several advantages, such as reproducing late-stage disease and shortening the model development time, but consist of implanting the tumor cells in a localized region or subcutaneously into immune-deficient mice. Despite some benefits of the transplant approach, these models are limited by their engraftment reproducibility and interactions with host micro-environments. Here, we explored the utility of humanizing Nod-Scid-IL2Rg null (NSG) mouse strains with factors associated with enhancing myeloid and lymphoma cell growth to establish a pipeline for rapid, reliable generation of in vivo DLBCL models. Methods We transduced the well-established human DLBCL cells, U2932, with the luciferase (Luc)-EGFP gene. The Luc-expressing (U2932-Luc) tumor cells were sorted for GFP positivity (GFP +) and expanded. The U2932-Luc cells (1 x 10 6/100µl PBS) were injected IV via tail vein into 8~12-week-old mice of various humanized NSG strains (representing equal numbers of each sex). NSG mice were humanized by transgenic expression of human cytokines (either human IL6 alone or IL6 plus SCF, GM-CSF, and IL-3) with the CMV promoter. Each experiment included of U2932-Luc cell transplanted group and control groups. We assessed U2932-Luc cell engraftment and growth by weekly in vivo imaging (IVIS 200 Imager, Perkin Elmer). To evaluate the organ specific engraftment/progression, we confirmed engraftment by bioluminescence imaging at the 2 nd week, then euthanized one mouse per week. At 15 minutes before euthansia, luciferin was injected via intraperitoneal injection. Following euthanasia, the organs were excised and underwent ex vivo IVIS imaging. The spleen, lungs, and liver were then fixed with 10% formalin and embedded in paraffin. Sections were stained with hematoxylin and eosin, and an anti-CD20 antibody to evaluate the tumor morphology using a Zeiss AXIO Imager M2 microscope (Zeiss, Nashville, TN). All other mice were monitored for survival and the median survival between the IL6 and IL6/SGM3 mice were compared using the Log-rank test. Results Similar to previously reported DLBCL humanized strain (MISTRG) (Hashwah, 2019), we used the IL6/SGM3 expressing strain. However, our studies also included the IL6 only humanized strain. We found that both the IL6 and IL6/SGM3 strains were highly permissive to DLBCL growth. The IL-6 strain exhibited a heightened growth of U2932 cells relative to the IL-6/SGM3 mice. As shown in Figure 1, the IL6 mice survived longer than IL6/SGM3 mice. Significant difference between the median survival of IL6 and IL6/SGM3 mice i.e. 48 days vs 42 days was observed (p < 0.0482). The organ specific evaluation demonstrated that U2932-Luc cells were initially engrafted and grew in the lung, liver, and spleen. Subsequently, U2932 cells were found in the skeleton, ovary, and brain. Of note, we detected significantly enlargements of the kidney, spleen, and ovary at the terminal stage. Conclusions Our humanized mouse model approach of using U2932 human DLBCL cells transduced with the Luc gene in the NSG-IL6 and NSG-IL6/SGM3 mice reproduced the clinical features of an aggressive DLBCL that paralleled the original patient. This model will provide a new tool to enable expansion of patient samples while overcoming the current limitations of DLBCL xenografts and transgenic mice. The ability to maintain growth of patient-derived samples within clinically relevant locations has great potential to more accurately test patient-specific, personalized treatment strategies. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2049-2049
Author(s):  
Jiusheng Deng ◽  
Dongkyoo Park ◽  
Mengchang Wang ◽  
Qiaoya Deng ◽  
Shannon Matulis ◽  
...  

Abstract Multiple myeloma (MM) is the most common hematological malignancy in USA, characterized with excessive abnormal clonal plasma cells in the bone marrow. Despite the great improvement in MM treatment by targeting the normal plasma cell biology of the cells during the last decade, the disease still remains incurable. B-cell lymphoma-2 (Bcl2) protein has four domains: BH1 (Bcl2 homology domain 1), BH2, BH3 and BH4, and plays critical roles in promoting the survival and drug-resistant of MM cells. Unlike the BH1, BH2, and BH3 domains, the BH4 region is responsible for the anti-apoptotic function of Bcl2 protein. Current BH3-mimetic and other Bcl2 inhibitors have been trailed in clinic but showed limited therapeutic efficacy. We recently identified a small molecule BDA-366 as a Bcl2-BH4 domain antagonist with potent anti-MM effect. BDA-366 bound to the BH4 resulted in the decrease of Bcl2 phosphorylation and the exposure of the BH3 domain as detected by anti-Bcl2-BH3 antibodies. Consequently, BDA-366 treatment induced robust apoptotic death on both human MM cell lines (RPMI8226 and U266) and primary MM cells from patients. In NOD-scid/IL2Rγnull (NSG) murine xenograft model, administration of BDA-366 (20mg/kg/day, 5 doses) markedly suppressed the growth of human MM tumor cells in vivo. We propose that BDA-366 as a novel BH4-based anti-MM agent could provide a more efficient pharmacological approach to treat MM. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3287-3287
Author(s):  
Puja Sapra ◽  
Rhona Stein ◽  
Jennifer Pickett ◽  
Serengulam V. Govindan ◽  
Thomas M. Cardillo ◽  
...  

Abstract IMMU-110 is a drug immunoconjugate comprised of doxorubicin (DOX) conjugated to the humanized anti-CD74 monoclonal antibody (mAb), hLL1, at a DOX:mAb (mol/mol) ratio of 8:1. CD74 is a rapidly internalizing type-II transmembrane chaperone molecule associated with HLA-DR, and has high expression on human non-Hodgkin’s lymphoma (NHL) and multiple myeloma (MM) clinical specimens and cell lines. Here, we investigated the in vitro and in vivo efficacy of IMMU-110 in xenograft models of human NHL (Raji, Daudi) and MM (MC/CAR). In vitro cell binding of IMMU-110 with the CD74-positive cells was significantly higher than that of a non-specific isotype-matched mAb-DOX conjugate (DOX conjugated to a mAb against epithelial glycoprotein-1; DOX-hRS7), and was similar to that of naked hLL1. Both IMMU-110 and naked hLL1 bound CD74 with subnanomolar affinity. The in vitro cytotoxicity of IMMU-110 was significantly higher than non-specific antibody-DOX conjugate, DOX-hRS7, and was similar to free DOX in MC/CAR, Raji or Daudi human Burkitt’s lymphoma cells. In CD74-negative cell lines, IMMU-110 was significantly less toxic than free DOX, having similar cytotoxicity to DOX-hRS7. In vivo, IMMU-110 displayed a pharmacokinetic and biodistribution profile almost identical to that of hLL1 mAb. Both hLL1 mAb and IMMU-110 had a biphasic clearance from the circulation; the α and β half-life (t1/2) of IMMU-110 were 4.6 h and 157.9 h, respectively, and those of hLL1 were 5.4 h and 151.5 h, respectively. In biodistribution studies, no significant difference was observed between IMMU-110 and naked hLL1 with regards to normal tissue uptake. Neither IMMU-110 nor naked hLL1 mAb had a significant association with any normal body tissue. In therapy experiments, a single i.v. protein dose of 350 μg IMMU-110, injected 5 days after implantation of MC/CAR cells in SCID mice, resulted in curing 70% of the animals. Similar cure rates were observed when treatment with IMMU-110 was given 10 days after transplantation of MC/CAR cells. In the Raji xenograft model, 100% of animals were cured with a single protein dose of 120 μg IMMU-110, injected 5 days after implantation of cells. In survival studies, the efficacy of IMMU-110 was significantly better than naked hLL1, the combination of naked hLL1 and free DOX, or of a non-specific antibody-DOX conjugate, DOX-hRS7. In a tolerability study in SCID mice, no toxic effect of IMMU-110 was observed even at the highest dose tested (2.5 mg /mouse). In conclusion, treatment of B-cell lymphoma and myeloma xenograft models with single injections of IMMU-110 resulted in high levels of response and long-term survivors. IMMU-110 is being further developed as a potential therapeutic for the treatment of CD74-positive tumors.


2013 ◽  
Vol 2013 ◽  
pp. 1-12 ◽  
Author(s):  
Patrick Frost ◽  
Eileen Berlanger ◽  
Veena Mysore ◽  
Bao Hoang ◽  
YiJiang Shi ◽  
...  

We found that rapalog mTOR inhibitors induce G1 arrest in the PTEN-null HS Sultan B-cell lymphoma linein vitro, but that administration of rapalogs in a HS Sultan xenograft model resulted in significant apoptosis, and that this correlated with induction of hypoxia and inhibition of neoangiogenesis and VEGF expression. Mechanistically, rapalogs prevent cap-dependent translation, but studies have shown that cap-independent, internal ribosome entry site (IRES)-mediated translation of genes, such as c-myc and cyclin D, can provide a fail-safe mechanism that regulates tumor survival. Therefore, we tested if IRES-dependent expression of VEGF could likewise regulate sensitivity of tumor cellsin vivo. To achieve this, we developed isogenic HS Sultan cell lines that ectopically express the VEGF ORF fused to the p27 IRES, an IRES sequence that is insensitive to AKT-mediated inhibition of IRES activity and effective in PTEN-null tumors. Mice challenged with p27-VEGF transfected tumor cells were more resistant to the antiangiogenic and apoptotic effects of the rapalog, temsirolimus, and active site mTOR inhibitor, pp242. Our results confirm the critical role of VEGF expression in tumors during treatment with mTOR inhibitors and underscore the importance of IRES activity as a resistance mechanism to such targeted therapy.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2444-2444
Author(s):  
Kerstin Wenzl ◽  
Katharina Troppan ◽  
Beata Pursche ◽  
Marco Bischof ◽  
Christine Beham-Schmid ◽  
...  

Abstract Introduction and Aim: We recently described a reduced NR4A1 and NR4A3 expression chronic lymphocytic leukemia, follicular lymphoma, and diffuse large B-cell lymphoma compared to normal controls. Our survival analysis of aggressive lymphomas revealed that low NR4A1 expression was associated with poor cancer specific survival. Over-expression of NR4A1 in lymphoma cell lines led to a significantly higher proportion of lymphoma cells undergoing apoptosis and abrogated tumor growth in xenografts1. The aim of this study is to define the role of NR4A1 as a tumor suppressor in the development of lymphoid malignancies in vivo. Methods: To identify, whether the loss of NR4A1 has an impact in Myc driven lymphomagenesis we crossed EµMyc mice with NR4A1-/- mice and performed phenotypical analysis including measurement of tumor development, survival and immunophenotypic determination of the newly developed lymphomas by FACS analysis. To further investigate the impact of NR4A1 loss on the oncogenic potential of EµMyc lymphoma cells we isolated viable tumor cells (B220+ and 7AAD-) and cultured them for 72h with or without lipopolysaccharide (LPS) and determined the number of viable cells and their viability (B220 and 7AAD-staining by flow cytometry analysis) after 24h, 48h and 72h. Finally, expression levels of NR4A1, NR4A3 and Myc with or without NR4A1 loss were evaluated by using RT-qPCR. Results: EµMyc mice with NR4A1 loss (EµMyc NR4A1-/-, n=46) developed visible tumors significantly faster compared to EµMyc mice with NR4A1 (EµMyc NR4A1+/+, n=75) (median = 44 days for EµMyc NR4A1 -/- vs. 107 days for EµMyc NR4A1+/+; p<0.001). Additionally, EµMyc NR4A1-/- mice showed a significantly shorter life span (median survival = 77 days) compared to EµMyc NR4A1 +/+ mice (median survival = 156 days; p<0.001). By comparing the immunophenotype of the newly developed lymphoma between the two groups (EµMyc NR4A1+/+, n=17 and EµMyc NR4A1 /- , n=19), no significant difference was observed. Interestingly, EµMyc NR4A1-/- mice showed an increased frequency of strong CD93 expression (10 of 18, respectively, vs. 2 of 17 EµMyc NR4A1 +/+ mice, p=0.004). Since most of the EµMyc NR4A1-/- lymphoma were IgM negative (7 of 10) it might be speculated that NR4A1 loss leads to a more immature phenotype of the lymphoma. The number of viable B220+ lymphoma cells isolated from EµMyc NR4A1-/- mice was higher compared to B220+ lymphoma cells isolated from EµMyc NR4A1+/+ mice after 72h in culture with or without LPS (p=0.056; p=0,052). This was accompagnied by a higher in vitro proliferation rate as demonstrated by a higher percentage of BrdU positive cells of the EµMyc NR4A1-/- mice compared to B220+ EµMyc NR4A1+/+ cells with and without LPS stimulation (p= 0,064, p=0,038). Interestingly, we detected a 12 fold higher NR4A3 mRNA expression (p=0,038) in EµMyc NR4A1-/- tumors compared to EµMyc NR4A1+/+. Conclusion: Our data demonstrate that NR4A1 possesses tumor suppressive properties and that loss of NR4A1 accelerates Myc driven lymphomagenesis. Furthermore, this study indicates that deletion of NR4A1 confers a more aggressive behavior and increases the oncogenic potential of EµMyc driven lymphoma cells. 1. Deutsch AJ, Rinner B, Wenzl K, et al. NR4A1-mediated apoptosis suppresses lymphomagenesis and is associated with a favorable cancer specific survival in patients with aggressive B-cell lymphomas. Blood. 2014. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5155-5155
Author(s):  
Xue-Jun Zhu ◽  
Zhong-Fa Yang ◽  
Li-Xin Wang ◽  
Alan G. Rosmarin

Abstract Relapsed, refractory Hodgkin’s or non-Hodgkin’s lymphoma remains to be a challenge and lacks efficient treatment. A small number of tumor cells that escape from treatment become resistant or unresponsive to chemotherapeutic agents. These cells grow rapidly and soon regenerate into large tumors. Immune vaccination with dendritic cells (DCs) efficiently inhibits growth of small size tumor only. The progression of tumors induces accumulation of Gr-1+CD11b+ myeloid derived suppressor cells (MDSCs) in the lymphatic organs and vicinity of tumor cells. MDSCs enable tumor cells to escape from host or exogenous immune cells mediated surveillance and attack. Gemcitabine is the chemotherapeutic agent used for lymphoma treatment. Gemcitabine has been reported to selectively eliminate MDSCs in tumor bearing animals. Thus, gemcitabine not only eliminates the majority of tumor to leave fewer resistant cells, it also reduces MDSCs to improve the immune environment favorable for the subsequent tumor vaccination. In this presentation, we used A20 cells to establish a large size, murine B-cell lymphoma model. We confirmed the accumulation of MDSCs in the spleen of lymphoma-bearing mice. Gemcitabine induced increased apoptosis in isolated, cultured MDSCs. In vivo injection of gemcitabine eliminated the majority of MDSCs in the spleen of lymphoma-bearing mice. Gemcitabine treatment combined with intratumoral injection of DCs markedly reduced the size of tumor, and significantly improved the survival of the lymphoma-bearing mice. We demonstrated that intra-large-size-lymphoma injection with inactivated DCs in combination with gemcitabine chemotherapy enhanced the therapeutic efficacy of either DC-based vaccination or chemotherapeutic reagent alone. Together with other studies, our report supports the hypothesis that gemcitabine-mediated reduction in MDSCs significantly reduces the immune suppression in tumor-bearing animal environment. This study established novel experimental foundation for the combination of immune-chemotherapy to treat relapsed or refractory lymphoma in large size with enhanced efficacy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3731-3731 ◽  
Author(s):  
Ryohei Kozaki ◽  
Toshio Yoshizawa ◽  
Shuji Tohda ◽  
Tomoko Yasuhiro ◽  
Shingo Hotta ◽  
...  

Abstract Abstract 3731 Purpose: ONO-WG-307 is a small molecule inhibitor that covalently binds to Btk. Signals from B cell receptors (BCR) play a central role in signal transduction pathways regulating survival, activation, proliferation, and differentiation of B-lineage lymphoid cells. BCR signaling is implicated in the survival of malignant B cells and recent studies indicate that targeting Btk, an essential component of the BCR pathway, may be effective in the treatment of B-cell lymphoma. The activated B-cell-like (ABC) subtype of diffuse large B-cell lymphoma (DLBCL) correlates with poor prognosis and new therapies, preferably chemo-sparing therapies, or as add-on to existing treatment regimens are required to help treat patients with ABC-DLBCL. Therefore, Btk constitutes an interesting therapeutic target, thus the activity of ONO-WG-307 was evaluated in an ABC-DLBCL xenograft model. Methods: Tumor cells (TMD-8) were implanted subcutaneously into female SCID mice. Tumors were allowed to grow to a volume of 100–200 mm3 before the mice were randomized into groups based on tumor size. ONO-WG-307 was administered orally at doses up to 10 mg/kg bid. Tumors were measured two or three times weekly after initiation of treatment, and tumor volumes were determined using the formula volume (=width2xlength)/2. Animals were euthanized when the tumors reached a maximum volume of 2,000 mm3 or after a maximum period of 2 months. In parallel, an exploratory pharmacodynamic marker of Btk inhibition (Phosphorylated-Btk [P-Btk]) was also investigated in vivo. Results: Treatment with ONO-WG-307 resulted in a dose-dependent inhibition of tumor growth in a TMD-8 xenograft model. Furthermore, parallel analysis of a pharmacodynamic marker, P-Btk, supported that Btk was inhibited and the level of P-Btk inhibition was correlated with the decreased tumor volumes observed in the TMD-8 model. Conclusion: ONO-WG-307 is a highly potent and selective oral Btk inhibitor with evidence of efficacy in the ABC-DLBCL xenograft model, with Btk inhibition further supported using a PD marker. Given the need to treat and overcome disease resistance especially in ABC-DLBCL, the use of a Btk inhibitor is a novel, mechanistic approach to treating B cell malignancies. Additional work is underway, combining ONO-WG-307 with chemotherapy and other targeted agents. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4866-4866
Author(s):  
Sang-Hoon SIN ◽  
Dirk P Dittmer

Abstract Abstract 4866 MicroRNA-155 (miR-155) is overexpressed in many types of cancers including B cell lymphomas. It has also an essential function in GC development. These functions are conserved among species and miR-155 knockout (ko) mice are deficient in GC development in Peyer's patches (PP) and lymph nodes. The human tumor virus Kaposi sarcoma associated herpesvirus (KSHV) encodes an ortholog to miR-155, named miR-K12-11. MiR-155 and miR-K12-11 share 100% seed sequence homology (Nature. 2007 Dec 13;450(7172):1096-9, J Virol. 2007 Dec;81(23):12836-45). Another human herpesvirus, Epstein-Barr Virus, induces mir-155 upon infection and immortalization of naïve B cells in culture. To test the hypothesis that miR-K12-11 can complement the normal function of miR-155 in vivo, we generated transgenic mice, which express miR-K12-11, as well as the other 21 KSHV miRNAs. Using this mouse model, we show that KSHV latency locus which contains a viral ortholog of miR-155, KSHV miR-K12-11 complements B cell abnormalities associated with the lack of miR-155. Germinal center (GC) formation is rescued in spleen and PP and lowered immunoglobulin level is also augmented to normal in KSHV latency locus transgenic mice with miR-155 ko background. Furthermore, mature B cells were chronically activated, leading to hyperglobulinemia triggered by increased plasma cell frequency. Marginal zone (MZ) B cells developed hyperplasia and the mice had an augmented response to T-dependent antigen as well as the TLR4 ligand LPS in vivo. This suggests that by mimicking miR-155 (and other functions) this human tumor virus drives the expansion of infected B cells, which if left unchecked can accumulate secondary mutations leading to post-GC lymphoma and multicentric Castleman's disease. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi36-vi37
Author(s):  
Evelina Blomberg ◽  
Manuela Silginer ◽  
Michael Weller

Abstract Glioblastoma is characterized by a poor prognosis and a challenging phenotype for drug development. Although multimodal treatment, including surgery, radio- and chemotherapy is applied, the overall survival remains just above one year. Numerous clinical trials have studied targeted therapies against commonly deregulated pathways, but an efficient targeted drug is yet to be discovered. Likewise, immunotherapy has not been shown to be active. A subset of glioma tumor cells demonstrates stem-like properties; these cells are commonly referred to as glioma initiating cells (GIC). These types of cells are pluripotent and can by definition initiate and recapitulate glioma growth in experimental animals in vivo. Furthermore, these cells are often resistant to conventional therapies. Interferon β (IFN-β) is an immunomodulatory molecule with anti-cancer properties. We have previously shown that IFN-β greatly reduces sphere-formation capability of GIC. It was also confirmed that IFN-β sensitized resistant GIC to irradiation or the chemotherapeutic agent, temozolomide (TMZ). IFN-β treatment significantly prolonged survival in a xenograft model with GIC cells. In the current project, we want to use syngeneic mouse models to study the immunomodulatory effects of type I IFNs. Preliminary results indicate that abrogation of IFN signalling in tumor cells by CRISPR/Cas9 technology prolonged survival in mice only in cell lines which have substantial baseline autocrine IFN signalling. On the contrary, we did not observe a difference in survival when wild-type tumor cells were implanted in either IFNAR1 deficient or proficient hosts. Flow cytometry analysis will elucidate changes in immune cell recruitment and infiltration upon IFN signalling disruption. Moreover, we explore different treatments in combination with IFN-β as there are indications that TMZ or radiotherapy can have synergistic effects with stimulation of interferon type I signalling.


Author(s):  
Patrycja Guzik ◽  
Klaudia Siwowska ◽  
Hsin-Yu Fang ◽  
Susan Cohrs ◽  
Peter Bernhardt ◽  
...  

Abstract Purpose It was previously demonstrated that radiation effects can enhance the therapy outcome of immune checkpoint inhibitors. In this study, a syngeneic breast tumor mouse model was used to investigate the effect of [177Lu]Lu-DOTA-folate as an immune stimulus to enhance anti-CTLA-4 immunotherapy. Methods In vitro and in vivo studies were performed to characterize NF9006 breast tumor cells with regard to folate receptor (FR) expression and the possibility of tumor targeting using [177Lu]Lu-DOTA-folate. A preclinical therapy study was performed over 70 days with NF9006 tumor-bearing mice that received vehicle only (group A); [177Lu]Lu-DOTA-folate (5 MBq; 3.5 Gy absorbed tumor dose; group B); anti-CTLA-4 antibody (3 × 200 μg; group C), or both agents (group D). The mice were monitored regarding tumor growth over time and signs indicating adverse events of the treatment. Results [177Lu]Lu-DOTA-folate bound specifically to NF9006 tumor cells and tissue in vitro and accumulated in NF9006 tumors in vivo. The treatment with [177Lu]Lu-DOTA-folate or an anti-CTLA-4 antibody had only a minor effect on NF9006 tumor growth and did not substantially increase the median survival time of mice (23 day and 19 days, respectively) as compared with untreated controls (12 days). [177Lu]Lu-DOTA-folate sensitized, however, the tumors to anti-CTLA-4 immunotherapy, which became obvious by reduced tumor growth and, hence, a significantly improved median survival time of mice (> 70 days). No obvious signs of adverse effects were observed in treated mice as compared with untreated controls. Conclusion Application of [177Lu]Lu-DOTA-folate had a positive effect on the therapy outcome of anti-CTLA-4 immunotherapy. The results of this study may open new perspectives for future clinical translation of folate radioconjugates.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4217-4217
Author(s):  
Gabriela Chang ◽  
Helen M. Atkinson ◽  
Leslie R. Berry ◽  
Anthony K.C. Chan

Abstract Introduction: Unfractionated heparin (UFH) and low molecular weight heparin (LMWH) are widely used anticoagulants for thrombosis treatment. However, these anticoagulants have limitations such as increased bleeding, variable dose response, required frequent monitoring, and, in the case of LMWH, inability to inhibit thrombin. This has led to the development of a covalent complex of antithrombin and heparin (ATH), which has been shown to overcome many of these shortcomings. ATH has faster rates of inhibition of many coagulation factors, is able to inhibit clot-bound thrombin, and is a more effective inhibitor of both venous and arterial thrombosis in animal models. Moreover, in a rabbit thrombosis model, ATH has been shown to decrease clot mass and fibrin accretion, while the contrary was observed for UFH. From these observations, it was suggested that ATH may enhance fibrin breakdown and thus led to investigations into the effects of UFH and ATH on fibrinolysis. In vitro studies have shown that UFH enhances antithrombin inhibition of plasmin. In addition, ATH displays a slightly greater inhibition of plasmin generation and activity. Such studies were conducted in purified systems, in the absence of other plasmin inhibitors naturally present in plasma. Therefore, the aim of the present study was to compare the effects of UFH, LMWH, and ATH on plasmin generation in plasma. Methods: At 37°C tissue plasminogen activator (tPA) and soluble fibrin fragments (fib) were added to normal adult pooled platelet poor plasma supplemented with 0.35, 0.7, 1.4, or 2.1 U anti-Xa/ml UFH, LMWH, or ATH, to initiate plasmin generation (8.93nM tPA and 300µg/ml fib). At various time points, subsamples were mixed with excess plasminogen activator inhibitor 1 (PAI-1) (55.12nM) to stop further plasmin generation. The plasmin concentration at each time point was determined using a plasmin-specific chromogenic substrate and a standard curve produced from purified plasmin. Results: Comparisons of mean area under the curve (AUC) for plasmin generation displayed a significant decrease in plasmin generation in the presence of all three anticoagulants at all doses tested (p<0.05). Comparing the anticoagulants at similar doses, plasmin generation was significantly decreased in the presence of ATH (15384.66±1930.23nM/min) compared to LMWH (23892.28±3090.54nM/min) at 0.7 U/ml (p<0.05). At a dose of 1.4 U/ml, there was significantly less plasmin generated, over time, in the presence of UFH (20089.49±3022.1623nM/min) and ATH (19273.86±1805.7323nM/min) when compared to LMWH (24743.18±1265.1023nM/min) (p<0.05). There was no significant difference in plasmin inhibition between UFH and ATH at any of the doses tested. Conclusion: The present study supports previous findings that UFH and ATH can facilitate antithrombin inhibition of plasmin. It is also observed that LMWH catalyzes the inhibition of plasmin by antithrombin but possibly to a lesser extent. These findings suggest that ATH has a similar inhibitory effect on plasmin generation and activity in plasma compared to UFH, despite its overall superior anticoagulant properties. Therefore, previous in vivo observations displaying decrease in clot mass with administration of ATH was due to its enhanced anticoagulant abilities and not fibrinolysis enhancement. These findings add to our understanding of ATH mechanisms of action and aid in its development for clinical use. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document