NSG Mouse

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 1953 Experts worldwide ranked by ideXlab platform

Zhirui Wang - One of the best experts on this subject based on the ideXlab platform.

  • diphtheria toxin based anti human cd19 immunotoxin for targeting human cd19 tumors
    Molecular Oncology, 2017
    Co-Authors: Qian Zheng, Qi Huang, Zhaohui Wang, Huiping Zhang, Christene A. Huang, Joren C. Madsen, David H. Sachs, Zhirui Wang
    Abstract:

    CD19 is expressed on normal and neoplastic B cells and is a promising target for immunotherapy. However, there is still an unmet need to further develop novel therapeutic drugs for the treatment of the refractory/relapsing human CD19+ tumors. We have developed a diphtheria toxin-based anti-human CD19 immunotoxin for targeting human CD19+ tumors. We have constructed three isoforms of the CD19 immunotoxin: monovalent, bivalent, and foldback diabody. In vitro binding affinity and efficacy analysis demonstrated that the bivalent isoform had the highest binding affinity and in vitro efficacy. The in vivo efficacy of the CD19 immunotoxins was assessed using human CD19+ JeKo-1 tumor-bearing NOD/SCID IL-2 receptor γ-/- (NSG) Mouse model. In these animals, CD19 immunotoxins significantly prolonged the median survival from 31 days in controls to 34, 36, and 40 days in animals receiving the monovalent isoform, foldback diabody isoform, and bivalent isoform, respectively. The bivalent CD19 immunotoxin is a promising therapeutic drug candidate for targeting relapsing/refractory human CD19+ tumors.

Sanghoon Lee - One of the best experts on this subject based on the ideXlab platform.

  • obinutuzumab ga101 vs rituximab significantly enhances cell death antibody dependent cytotoxicity and improves overall survival against cd20 primary mediastinal b cell lymphoma pmbl in a xenograft nod scid il2rgnull NSG Mouse model a potential target
    Oncotarget, 2020
    Co-Authors: Yaya Chu, Aradhana Awasthi, Sanghoon Lee, Dina Edani, Changhong Yin, Jessica Hochberg, Tishi Shah, Taehoon Chung, Janet Ayello, Carmella Van De Ven
    Abstract:

    Primary mediastinal large B-cell lymphoma (PMBL), a distinct mature B-cell lymphoma, expresses CD20 and has recently been successfully treated with the combination of a type I anti-CD20 monoclonal antibody, rituximab, with multiple combination chemotherapy regimens. Obinutuzumab is a glycoengineered type II anti-CD20 monoclonal antibody (mAb), recognizing a unique CD20 extracellular membrane epitope with enhanced antibody dependent cellular cytotoxicity (ADCC) vs rituximab. We hypothesize that obinutuzumab vs rituximab will significantly enhance in-vitro and in-vivo cytotoxicity against PMBL. PMBL cells were treated with equal dose of obinutuzumab and rituximab for 24 hours (1-100 μg/ml). ADCC were performed with ex-vivo expanded natural killer cells at 10:1 E: T ratio. Mice were xenografted with intravenous injections of luciferase expressing Karpas1106P cells and treated every 7 days for 8 weeks. Tumor burden was monitored by IVIS spectrum system. Compared with rituximab, obinutuzumab significantly inhibited PMBL cell proliferation (p = 0.01), promoted apoptosis (p = 0.05) and enhanced ADCC (p = 0.0002) against PMBL. Similarly, in PMBL xenografted NOD scid gamma mice, obinutuzumab significantly enhanced survival than rituximab when treated with equal doses (p = 0.05). Taken together our results suggest that obinutuzumab significantly enhanced natural killer cytotoxicity, reduced PMBL proliferation and prolonged the overall survival in humanized PMBL xenografted NOD scid gamma mice.

  • abstract 3838 upregulation of dleu1 significantly prolongs survival in a rituximab treated dleu1 knockout human burkitt lymphoma bl xenograft NSG Mouse model dleu1 may act as a tumor suppressor gene in pediatric bl
    Cancer Research, 2016
    Co-Authors: Sanghoon Lee, Changhong Yin, Tishi Shah, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    BACKGROUND: Burkitt lymphoma (BL) is the most common histological subtype of non-Hodgkin lymphoma (NHL) in children and adolescents (Cairo et al., Blood 2007; Miles/Cairo., BJHaem 2012). We identified that pediatric BL patients with chromosome 13q deletion, particularly 13q14.3, had significantly poorer outcome and inferior overall survival despite aggressive short, intensive multi-agent chemotherapy (Poirel/Cairo et al., Leuk 2009; Nelson/Cairo/Sanger et al., BJH 2009). Deleted in lymphocytic leukemia 1 (DLEU1) is a BL classifier gene in the 13q14.3 region (Dave et al., NEJM 2006) and interacts with C-MYC, TUBB2C and UBR1 (Stelzl et al., Cell 2005). We have previously reported a regulated DLEU1 expression significantly associated with changes in BL proliferation in vitro (Lee/Cairo et al., AACR 2015). We hypothesize that DLEU1 may have function as a tumor suppressor gene; however, functions and mechanisms underlying DLEU1 expression in the BL are poorly understood. OBJECTIVES: We hypothesize that DLEU1 may act as a tumor suppressor gene in BL and therefore examined whether up-regulation of DLEU1 results in changes in survival following targeted immunotherapy. METHODS: DLEU1 stably overexpressing Raji BL cells (Lee/Cairo et al., ASH 2013) were transfected with a firefly luciferase expression plasmid, kindly provided by Laurence Cooper MD, PhD. Four- to six- week-old female NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice from The Jackson Lab. Tumor burden and tumor progression were monitored by bioluminescence imaging system. Mice were treated with either rituximab (30 mg/kg) or cyclophosphamide (25 mg/kg) by intraperitoneal injection at 7 day intervals. Survival rates were analyzed by the Kaplan-Meier method using the Prism Version 6.0 software. RESULTS: We observed that DLEU1 stably overexpressing Raji BL cells xenografted (GFP-DLEU1) mice had significantly extended survival compared to GFP control mice following treatment rituximab (59 vs 51 days, p CONCLUSIONS: We demonstrated that the upregulation of DLEU1 expression significantly improved the survival in DLEU1 overexpressing BL cells xenografted NSG mice following rituximab, cyclophosphamide and in combination treatment. Therefore, the up-regulation of DLEU1 expression in BL may in part result in sensitizing to chemoimmunotherapy induced survival and may result in a consideration of potential therapeutic strategies Citation Format: Sanghoon Lee, Changhong Yin, Tishi Shah, Janet Ayello, Erin Morris, Carmella van de Ven, Mitchell S. Cairo. Upregulation of DLEU1 significantly prolongs survival in a rituximab-treated DLEU1 knockout human Burkitt lymphoma (BL) xenograft NSG Mouse model: DLEU1 may act as a tumor suppressor gene in pediatric BL. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3838.

  • ibrutinib significantly prolonged survival in a human burkitt lymphoma bl xenograft NSG Mouse model ibrutinib may be a potential adjuvant agent in the treatment of bl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Timmy Oconnell, Lauren Harrison, Matthew J Barth, Rodney R Miles, Paul J Galardy
    Abstract:

    BACKGROUND : Burkitt Lymphoma (BL) represents approximately 40% of all childhood and adolescent non-Hodgkin lymphoma (Miles/Cairo, BJH 2012 ). Children with relapsed or progressive BL develop chemoimmunotherapy-resistant disease and can rarely be cured with salvage therapy (Cairo et al, JCO 2012). Bruton's tyrosine kinase (BTK) is a regulator of normal B-cell development and is activated upon B-cell receptor (BCR) stimulation. Chronic active BCR signaling through BTK activation can be inhibited by the selective and covalent BTK inhibitor, ibrutinib (Young/Staudt et al, Nat Rev Drug Dicov 2013). Preclinical studies of ibrutinib in chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL) suggest that its inhibitory effect on cell proliferation is in the range of 1.0uM to 25.0uM (Herman et al, Blood 2011; Cinar et al, Leuk Res 2013), which is higher than the typical plasma concentration range in patients at the recommended dose of 560mg (<0.6uM) (Advani et al, JCO 2013). Despite these relatively high IC50 values in vitro , ibrutinib has been highly effective in the treatment of patients with refractory CLL and MCL (Byrd et al, NEJM 2013 and Wang et al, NEJM 2013). Ibrutinib was initially approved by the FDA (IMBRUVICA, USPI) for patients with MCL in November 2013 and CLL in February 2014, who have received at least one prior therapy. BL, however, is associated with tonic vs chronic active BCR signaling; while, both CLL and MCL have chronic active BCR signaling. These findings suggest that the antitumor activity of ibrutinib in BL, which is associated with tonic BCR signaling, may be less pronounced than in Activated B cell-like diffuse large B-cell lymphoma (ABC-DLBCL), CLL or MCL. OBJECTIVES: We hypothesize that ibrutinib may be a potential adjuvant agent in the treatment of BL. Therefore, we investigated the in vivo anti-tumor activity of ibrutinib in BL xenografted NSG mice. METHODS : Raji Burkitt lymphoma (BL) cells (ATCC) were exposed to ibrutinib (0-10uM, generously provided by Janssen R&D LLC) alone and in combination with dexamethasone (1uM) for 5 days and then MTS cell proliferation assay (Promega) was performed. The IC50 values were determined with CompuSynTM software (Chou and Martin, ComboSyn, 2005) based on data derived from MTS assay. Raji BL cells were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. NSG mice (NOD.Cg-Prkdcscid Il2rgtm1Wjl /SzJ, 6-8 weeks old) from Jackson laboratory were irradiated (2.5 Gy) and then mice were subcutaneously injected with one million ffluc-zeo tumor cells. Tumor progression and tumor burden were monitored by Bioluminescent Imaging system using the Xenogen IVIS-200 (Caliper Life Sciences). Mice were orally gavaged with either vehicle or ibrutinib (12.5mg/kg) for 10 days (once a day) at post-tumor cells injection. Analysis of survival rates were determined by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS : Ibrutinib compared to control induced a significant dose-dependent decrease in cell proliferation in Raji BL cells (5uM, 0.561 ± 0.170, p<0.03, IC50=5.20uM) following 120 hours (5 days) of ibrutinib treatment. Interestingly, we observed a significant decrease in cell proliferation in Raji BL cells (5uM, 0.213 ± 0.078, p<0.002, IC=0.50 ± 0.374uM) following ibrutinib and 1uM dexamethasone combination treatment following 120 hours (5 days) of treatment. We observed a significant decrease of tumor progression by tumor luminescence signal intensity following ibrutinib treated Raji BL xenografted NSG mice at day 20 (12.5mg/kg, p<0.001 and 25mg/kg, p<0.05) and day 25 (12.5mg/kg, p<0.05 and 25mg/kg, p<0.05) compared to control (Figure 1A).Ibrutinib (12.5 mg/kg) treated mice (n=39) significantly prolonged survival compared to vehicle controls (n=28) (p<0.0001) (Figure 1B). CONCLUSIONS : Ibrutinib (12.5mg/kg) significantly decreased tumor progression and significantly increased survival in Raji BL xenografted NSG mice, suggesting that ibrutinib may be a potential adjuvant agent for therapy in the treatment of BL. Future studies will investigate the efficacy of combination therapy with ibrutinib on survival in BL xenografted NOD/SCID mice. ![Figure][1] Disclosures Galardy: Mission Therapeutics: Research Funding. [1]: pending:yes

  • downregulation of dleu1 significantly shortened survival in a rituximab treated dleu1 knockout human burkitt lymphoma bl xenograft NSG Mouse model dleu1 may act as a tumor suppressor gene in pediatric bl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    BACKGROUND : Burkitt lymphoma (BL) is the most common histological subtype of non-Hodgkin lymphoma (NHL) in children and adolescents (Cairo et al., Blood, 2007; Miles/Cairo, BJH, 2012). We have previously identified secondary chromosomal aberrations in 70% of pediatric BL patients (PBL) with a C-MYC gene rearrangement (Poirel/Cairo et al., Leukemia, 2009). Specifically, we identified significantly inferior event free survival (EFS) and overall survival in children and adolescents with a specific loss of the 13q14.3 locus (Poirel/Cairo et al., Leukemia, 2009; Nelson/Cairo/Sanger et al., Br. J. Haematol., 2009). In a multivariate analysis controlling for stage, lactate dehydrogenase levels, country of treatment, and group classification, children with BL who had a 13q deletion had significantly poorer EFS compared to the remainder of patients treated with the same French-American-British (FAB) chemotherapy regimen (Poirel/Cairo et al., Leukemia, 2009). Deleted in lymphocytic leukemia 1 (DLEU1) is a BL classifier gene in the 13q14.3 region (Dave et al., NEJM, 2006) and interacts with C-MYC , Tubulin beta-2C chain (TUBB2C), E3 ubiquitin-protein ligase (UBR1), cellular tumor antigen p53, and Ras association (RalGDS/AF-6) domain family member 1 (RASSF1) (Stelzl et al., Cell, 2005). The sequence-specific Transcription Activator-Like Effector Nucleases (TALENs) have been developed for targeted genome editing in in vitro and in vivo experiments with high efficiency as a new therapeutic tool (Sander et al., Nat. Biotechnol., 2011). We have previously reported a down-regulated DLEU1 mRNA expression significantly associated with an increase in BL proliferation in vitro (Lee/Cairo et al., AACR, 2015). We hypothesize that DLEU1 may have function as a tumor suppressor gene; however, the role of DLEU1 regulating programmed cell death in BL are poorly understood. OBJECTIVES : We hypothesize that DLEU1 may act as a tumor suppressor gene in BL and whether down-regulation of DLEU1 expression results in changes in BL survival following targeted immunotherapy. METHODS : TALENs induced DLEU1 knockout Raji cells were previously generated (Lee/Cairo et al., ASH, 2013) and DLEU1 knockout cells were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. Four- to six- week-old female NSG (NOD.Cg-Prkdc scid Il2rg tm1Wjl /SzJ) mice from The Jackson Laboratory were irradiated (2.5 Gy), and then mice were subcutaneously injected with 1x10 6 ffluc-zeo tumor cells. Tumor burden and tumor progression were monitored by bioluminescence imaging system using the Xenogen IVIS-200 (Caliper Life Sciences). Mice were treated with either PBS, IgG isotype control, rituximab (30 mg/kg) or cyclophosphamide (25 mg/kg) and in combination by intraperitoneal (i.p) injection at 7 day intervals. Survival rates were analyzed by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS: There were significant increases of luciferase signal intensity in DLEU1 knockout mice (DLEU1-KO) compared to that in wild type (WT) mice on day 31 of rituximab (p vs 48 days, p CONCLUSIONS : The down-regulation of DLEU1 expression significantly decreased the survival rate in DLEU1-KO xenografted NSG mice following rituximab and in combination with cyclophosphamide treatment. Therefore, the down-regulation of DLEU1 expression in BL may in part result in immunotherapy resistance and may result in a consideration of alternative therapeutic strategies. Disclosures No relevant conflicts of interest to declare.

  • ruxolitinib significantly prolongs survival in both a primary mediastinal b cell lymphoma pmbl and hodgkin lymphoma hl xenograft NSG Mouse model ruxolitinib may be a potential adjuvant agent in the treatment of pmbl and of hl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    Abstract BACKGROUND: Primary Mediastinal large B-cell lymphoma (PMBL) and Hodgkin lymphoma (HL) are two of the most common malignancies among adolescents and young adults (AYA). PMBL and HL share similar molecular features by gene expression profiling and pathogenesis (Rosenwald et al., J Exp Med., 2003). HL represents only approximately 4-5% of all cancers in children younger than 15 years of age, but is the most common cancer in the 15-35 yrs AYA group. While the prognosis is excellent with AYA HL, there are significant late effects secondary to chemoradiotherapy and therefore new targeted agents are needed to avoid these morbid late effects in HL (Hochberg/Cairo et al., Br. J. Haem., 2009). Frequent gains of chromosome 9p exhibit higher Janus Kinase 2 (JAK2) transcript levels with increased JAK2 activity (Bentz et al., Genes Chromosomes Cancer, 2001), suggesting aberrant activity of JAK2 and Signal Transducer and Activator of Transcription (STAT) pathways, which may in part play an important role in the pathogenesis of HL and PMBL. Ruxolitinib is a potent and selective JAK1/JAK2 inhibitor against myeloproliferative neoplasms (MPNs) that consistently exhibits dysregulation of the JAK1/JAK2 pathway, including those MPNs with a JAK2V617F mutation. Ruxolitinb also inhibits JAK2/STAT5 signaling in vitro and in a murine model of MPN (Quintas-Cardama et al., Blood, 2010). OBJECTIVES: We hypothesize that ruxolitinib may potentially function as targeted therapeutic agent for both PMBL and HL and therefore, investigated the efficacy of ruxolitinib in PMBL and HL cells xenografted into NSG mice. METHODS: Cell proliferation and apoptosis analysis were assessed using MTS and Caspase-3/7 assay (Promega), respectively. The expression of protein was examined by immunoblotting and statistical significance was determined by Student t-test. Karpas-1106P PMBL cell line and L-428 HL cell line were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. The six to eight weeks old Female NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice were from Jackson laboratory. The ffluc-zeo NSG mice were irradiated (2.5 Gy) and then were subcutaneously injected with 1x106 ffluc-zeo Karpas-1106P or L-428 tumor cells. Tumor burden and tumor progression were monitored by bioluminescence imaging (BLI) using the Xenogen IVIS-200 (Caliper Life Sciences) for up to 60 days. Mice were orally gavaged with either vehicle or ruxolitinib (45.0mg/kg or 90.0mg/kg) (generously provided by Incyte Corporation, Wilmington, DE, USA) for 21 days. Survival rates were analyzed by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS: We observed that ruxolitinib significantly decreased the phosphorylation of STAT3, -5 and -6 in Karpas-1106P PMBL and L-428 HL cells. The reduction in cell proliferation by 10uM ruxolitinib are 41% in Karpas-1106P (p CONCLUSIONS: Ruxolitinib significantly inhibited JAK2 enzymatic activity by the abrogation of the phosphorylation of STAT3, STAT5 and STAT6, and ruxolitinib showed significant anti-proliferative effects. Ruxolitinib (45mg/kg) significantly inhibited tumor progression and significantly prolonged survival in PMBL and HL xenografted NSG mice compared to control mice. Ruxolitinib may be a potential adjuvant agent in the treatment of PMBL and HL. Future studies will focus on whether the addition of ruxolitinib to chemoimmunotherapy improves the survival in a NSG PMBL and HL xenograft Mouse models. Download : Download high-res image (68KB) Download : Download full-size image Disclosures No relevant conflicts of interest to declare.

Carmella Van De Ven - One of the best experts on this subject based on the ideXlab platform.

  • obinutuzumab ga101 vs rituximab significantly enhances cell death antibody dependent cytotoxicity and improves overall survival against cd20 primary mediastinal b cell lymphoma pmbl in a xenograft nod scid il2rgnull NSG Mouse model a potential target
    Oncotarget, 2020
    Co-Authors: Yaya Chu, Aradhana Awasthi, Sanghoon Lee, Dina Edani, Changhong Yin, Jessica Hochberg, Tishi Shah, Taehoon Chung, Janet Ayello, Carmella Van De Ven
    Abstract:

    Primary mediastinal large B-cell lymphoma (PMBL), a distinct mature B-cell lymphoma, expresses CD20 and has recently been successfully treated with the combination of a type I anti-CD20 monoclonal antibody, rituximab, with multiple combination chemotherapy regimens. Obinutuzumab is a glycoengineered type II anti-CD20 monoclonal antibody (mAb), recognizing a unique CD20 extracellular membrane epitope with enhanced antibody dependent cellular cytotoxicity (ADCC) vs rituximab. We hypothesize that obinutuzumab vs rituximab will significantly enhance in-vitro and in-vivo cytotoxicity against PMBL. PMBL cells were treated with equal dose of obinutuzumab and rituximab for 24 hours (1-100 μg/ml). ADCC were performed with ex-vivo expanded natural killer cells at 10:1 E: T ratio. Mice were xenografted with intravenous injections of luciferase expressing Karpas1106P cells and treated every 7 days for 8 weeks. Tumor burden was monitored by IVIS spectrum system. Compared with rituximab, obinutuzumab significantly inhibited PMBL cell proliferation (p = 0.01), promoted apoptosis (p = 0.05) and enhanced ADCC (p = 0.0002) against PMBL. Similarly, in PMBL xenografted NOD scid gamma mice, obinutuzumab significantly enhanced survival than rituximab when treated with equal doses (p = 0.05). Taken together our results suggest that obinutuzumab significantly enhanced natural killer cytotoxicity, reduced PMBL proliferation and prolonged the overall survival in humanized PMBL xenografted NOD scid gamma mice.

  • abstract 3838 upregulation of dleu1 significantly prolongs survival in a rituximab treated dleu1 knockout human burkitt lymphoma bl xenograft NSG Mouse model dleu1 may act as a tumor suppressor gene in pediatric bl
    Cancer Research, 2016
    Co-Authors: Sanghoon Lee, Changhong Yin, Tishi Shah, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    BACKGROUND: Burkitt lymphoma (BL) is the most common histological subtype of non-Hodgkin lymphoma (NHL) in children and adolescents (Cairo et al., Blood 2007; Miles/Cairo., BJHaem 2012). We identified that pediatric BL patients with chromosome 13q deletion, particularly 13q14.3, had significantly poorer outcome and inferior overall survival despite aggressive short, intensive multi-agent chemotherapy (Poirel/Cairo et al., Leuk 2009; Nelson/Cairo/Sanger et al., BJH 2009). Deleted in lymphocytic leukemia 1 (DLEU1) is a BL classifier gene in the 13q14.3 region (Dave et al., NEJM 2006) and interacts with C-MYC, TUBB2C and UBR1 (Stelzl et al., Cell 2005). We have previously reported a regulated DLEU1 expression significantly associated with changes in BL proliferation in vitro (Lee/Cairo et al., AACR 2015). We hypothesize that DLEU1 may have function as a tumor suppressor gene; however, functions and mechanisms underlying DLEU1 expression in the BL are poorly understood. OBJECTIVES: We hypothesize that DLEU1 may act as a tumor suppressor gene in BL and therefore examined whether up-regulation of DLEU1 results in changes in survival following targeted immunotherapy. METHODS: DLEU1 stably overexpressing Raji BL cells (Lee/Cairo et al., ASH 2013) were transfected with a firefly luciferase expression plasmid, kindly provided by Laurence Cooper MD, PhD. Four- to six- week-old female NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice from The Jackson Lab. Tumor burden and tumor progression were monitored by bioluminescence imaging system. Mice were treated with either rituximab (30 mg/kg) or cyclophosphamide (25 mg/kg) by intraperitoneal injection at 7 day intervals. Survival rates were analyzed by the Kaplan-Meier method using the Prism Version 6.0 software. RESULTS: We observed that DLEU1 stably overexpressing Raji BL cells xenografted (GFP-DLEU1) mice had significantly extended survival compared to GFP control mice following treatment rituximab (59 vs 51 days, p CONCLUSIONS: We demonstrated that the upregulation of DLEU1 expression significantly improved the survival in DLEU1 overexpressing BL cells xenografted NSG mice following rituximab, cyclophosphamide and in combination treatment. Therefore, the up-regulation of DLEU1 expression in BL may in part result in sensitizing to chemoimmunotherapy induced survival and may result in a consideration of potential therapeutic strategies Citation Format: Sanghoon Lee, Changhong Yin, Tishi Shah, Janet Ayello, Erin Morris, Carmella van de Ven, Mitchell S. Cairo. Upregulation of DLEU1 significantly prolongs survival in a rituximab-treated DLEU1 knockout human Burkitt lymphoma (BL) xenograft NSG Mouse model: DLEU1 may act as a tumor suppressor gene in pediatric BL. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3838.

  • ibrutinib significantly prolonged survival in a human burkitt lymphoma bl xenograft NSG Mouse model ibrutinib may be a potential adjuvant agent in the treatment of bl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Timmy Oconnell, Lauren Harrison, Matthew J Barth, Rodney R Miles, Paul J Galardy
    Abstract:

    BACKGROUND : Burkitt Lymphoma (BL) represents approximately 40% of all childhood and adolescent non-Hodgkin lymphoma (Miles/Cairo, BJH 2012 ). Children with relapsed or progressive BL develop chemoimmunotherapy-resistant disease and can rarely be cured with salvage therapy (Cairo et al, JCO 2012). Bruton's tyrosine kinase (BTK) is a regulator of normal B-cell development and is activated upon B-cell receptor (BCR) stimulation. Chronic active BCR signaling through BTK activation can be inhibited by the selective and covalent BTK inhibitor, ibrutinib (Young/Staudt et al, Nat Rev Drug Dicov 2013). Preclinical studies of ibrutinib in chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL) suggest that its inhibitory effect on cell proliferation is in the range of 1.0uM to 25.0uM (Herman et al, Blood 2011; Cinar et al, Leuk Res 2013), which is higher than the typical plasma concentration range in patients at the recommended dose of 560mg (<0.6uM) (Advani et al, JCO 2013). Despite these relatively high IC50 values in vitro , ibrutinib has been highly effective in the treatment of patients with refractory CLL and MCL (Byrd et al, NEJM 2013 and Wang et al, NEJM 2013). Ibrutinib was initially approved by the FDA (IMBRUVICA, USPI) for patients with MCL in November 2013 and CLL in February 2014, who have received at least one prior therapy. BL, however, is associated with tonic vs chronic active BCR signaling; while, both CLL and MCL have chronic active BCR signaling. These findings suggest that the antitumor activity of ibrutinib in BL, which is associated with tonic BCR signaling, may be less pronounced than in Activated B cell-like diffuse large B-cell lymphoma (ABC-DLBCL), CLL or MCL. OBJECTIVES: We hypothesize that ibrutinib may be a potential adjuvant agent in the treatment of BL. Therefore, we investigated the in vivo anti-tumor activity of ibrutinib in BL xenografted NSG mice. METHODS : Raji Burkitt lymphoma (BL) cells (ATCC) were exposed to ibrutinib (0-10uM, generously provided by Janssen R&D LLC) alone and in combination with dexamethasone (1uM) for 5 days and then MTS cell proliferation assay (Promega) was performed. The IC50 values were determined with CompuSynTM software (Chou and Martin, ComboSyn, 2005) based on data derived from MTS assay. Raji BL cells were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. NSG mice (NOD.Cg-Prkdcscid Il2rgtm1Wjl /SzJ, 6-8 weeks old) from Jackson laboratory were irradiated (2.5 Gy) and then mice were subcutaneously injected with one million ffluc-zeo tumor cells. Tumor progression and tumor burden were monitored by Bioluminescent Imaging system using the Xenogen IVIS-200 (Caliper Life Sciences). Mice were orally gavaged with either vehicle or ibrutinib (12.5mg/kg) for 10 days (once a day) at post-tumor cells injection. Analysis of survival rates were determined by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS : Ibrutinib compared to control induced a significant dose-dependent decrease in cell proliferation in Raji BL cells (5uM, 0.561 ± 0.170, p<0.03, IC50=5.20uM) following 120 hours (5 days) of ibrutinib treatment. Interestingly, we observed a significant decrease in cell proliferation in Raji BL cells (5uM, 0.213 ± 0.078, p<0.002, IC=0.50 ± 0.374uM) following ibrutinib and 1uM dexamethasone combination treatment following 120 hours (5 days) of treatment. We observed a significant decrease of tumor progression by tumor luminescence signal intensity following ibrutinib treated Raji BL xenografted NSG mice at day 20 (12.5mg/kg, p<0.001 and 25mg/kg, p<0.05) and day 25 (12.5mg/kg, p<0.05 and 25mg/kg, p<0.05) compared to control (Figure 1A).Ibrutinib (12.5 mg/kg) treated mice (n=39) significantly prolonged survival compared to vehicle controls (n=28) (p<0.0001) (Figure 1B). CONCLUSIONS : Ibrutinib (12.5mg/kg) significantly decreased tumor progression and significantly increased survival in Raji BL xenografted NSG mice, suggesting that ibrutinib may be a potential adjuvant agent for therapy in the treatment of BL. Future studies will investigate the efficacy of combination therapy with ibrutinib on survival in BL xenografted NOD/SCID mice. ![Figure][1] Disclosures Galardy: Mission Therapeutics: Research Funding. [1]: pending:yes

  • downregulation of dleu1 significantly shortened survival in a rituximab treated dleu1 knockout human burkitt lymphoma bl xenograft NSG Mouse model dleu1 may act as a tumor suppressor gene in pediatric bl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    BACKGROUND : Burkitt lymphoma (BL) is the most common histological subtype of non-Hodgkin lymphoma (NHL) in children and adolescents (Cairo et al., Blood, 2007; Miles/Cairo, BJH, 2012). We have previously identified secondary chromosomal aberrations in 70% of pediatric BL patients (PBL) with a C-MYC gene rearrangement (Poirel/Cairo et al., Leukemia, 2009). Specifically, we identified significantly inferior event free survival (EFS) and overall survival in children and adolescents with a specific loss of the 13q14.3 locus (Poirel/Cairo et al., Leukemia, 2009; Nelson/Cairo/Sanger et al., Br. J. Haematol., 2009). In a multivariate analysis controlling for stage, lactate dehydrogenase levels, country of treatment, and group classification, children with BL who had a 13q deletion had significantly poorer EFS compared to the remainder of patients treated with the same French-American-British (FAB) chemotherapy regimen (Poirel/Cairo et al., Leukemia, 2009). Deleted in lymphocytic leukemia 1 (DLEU1) is a BL classifier gene in the 13q14.3 region (Dave et al., NEJM, 2006) and interacts with C-MYC , Tubulin beta-2C chain (TUBB2C), E3 ubiquitin-protein ligase (UBR1), cellular tumor antigen p53, and Ras association (RalGDS/AF-6) domain family member 1 (RASSF1) (Stelzl et al., Cell, 2005). The sequence-specific Transcription Activator-Like Effector Nucleases (TALENs) have been developed for targeted genome editing in in vitro and in vivo experiments with high efficiency as a new therapeutic tool (Sander et al., Nat. Biotechnol., 2011). We have previously reported a down-regulated DLEU1 mRNA expression significantly associated with an increase in BL proliferation in vitro (Lee/Cairo et al., AACR, 2015). We hypothesize that DLEU1 may have function as a tumor suppressor gene; however, the role of DLEU1 regulating programmed cell death in BL are poorly understood. OBJECTIVES : We hypothesize that DLEU1 may act as a tumor suppressor gene in BL and whether down-regulation of DLEU1 expression results in changes in BL survival following targeted immunotherapy. METHODS : TALENs induced DLEU1 knockout Raji cells were previously generated (Lee/Cairo et al., ASH, 2013) and DLEU1 knockout cells were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. Four- to six- week-old female NSG (NOD.Cg-Prkdc scid Il2rg tm1Wjl /SzJ) mice from The Jackson Laboratory were irradiated (2.5 Gy), and then mice were subcutaneously injected with 1x10 6 ffluc-zeo tumor cells. Tumor burden and tumor progression were monitored by bioluminescence imaging system using the Xenogen IVIS-200 (Caliper Life Sciences). Mice were treated with either PBS, IgG isotype control, rituximab (30 mg/kg) or cyclophosphamide (25 mg/kg) and in combination by intraperitoneal (i.p) injection at 7 day intervals. Survival rates were analyzed by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS: There were significant increases of luciferase signal intensity in DLEU1 knockout mice (DLEU1-KO) compared to that in wild type (WT) mice on day 31 of rituximab (p vs 48 days, p CONCLUSIONS : The down-regulation of DLEU1 expression significantly decreased the survival rate in DLEU1-KO xenografted NSG mice following rituximab and in combination with cyclophosphamide treatment. Therefore, the down-regulation of DLEU1 expression in BL may in part result in immunotherapy resistance and may result in a consideration of alternative therapeutic strategies. Disclosures No relevant conflicts of interest to declare.

  • ruxolitinib significantly prolongs survival in both a primary mediastinal b cell lymphoma pmbl and hodgkin lymphoma hl xenograft NSG Mouse model ruxolitinib may be a potential adjuvant agent in the treatment of pmbl and of hl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    Abstract BACKGROUND: Primary Mediastinal large B-cell lymphoma (PMBL) and Hodgkin lymphoma (HL) are two of the most common malignancies among adolescents and young adults (AYA). PMBL and HL share similar molecular features by gene expression profiling and pathogenesis (Rosenwald et al., J Exp Med., 2003). HL represents only approximately 4-5% of all cancers in children younger than 15 years of age, but is the most common cancer in the 15-35 yrs AYA group. While the prognosis is excellent with AYA HL, there are significant late effects secondary to chemoradiotherapy and therefore new targeted agents are needed to avoid these morbid late effects in HL (Hochberg/Cairo et al., Br. J. Haem., 2009). Frequent gains of chromosome 9p exhibit higher Janus Kinase 2 (JAK2) transcript levels with increased JAK2 activity (Bentz et al., Genes Chromosomes Cancer, 2001), suggesting aberrant activity of JAK2 and Signal Transducer and Activator of Transcription (STAT) pathways, which may in part play an important role in the pathogenesis of HL and PMBL. Ruxolitinib is a potent and selective JAK1/JAK2 inhibitor against myeloproliferative neoplasms (MPNs) that consistently exhibits dysregulation of the JAK1/JAK2 pathway, including those MPNs with a JAK2V617F mutation. Ruxolitinb also inhibits JAK2/STAT5 signaling in vitro and in a murine model of MPN (Quintas-Cardama et al., Blood, 2010). OBJECTIVES: We hypothesize that ruxolitinib may potentially function as targeted therapeutic agent for both PMBL and HL and therefore, investigated the efficacy of ruxolitinib in PMBL and HL cells xenografted into NSG mice. METHODS: Cell proliferation and apoptosis analysis were assessed using MTS and Caspase-3/7 assay (Promega), respectively. The expression of protein was examined by immunoblotting and statistical significance was determined by Student t-test. Karpas-1106P PMBL cell line and L-428 HL cell line were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. The six to eight weeks old Female NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice were from Jackson laboratory. The ffluc-zeo NSG mice were irradiated (2.5 Gy) and then were subcutaneously injected with 1x106 ffluc-zeo Karpas-1106P or L-428 tumor cells. Tumor burden and tumor progression were monitored by bioluminescence imaging (BLI) using the Xenogen IVIS-200 (Caliper Life Sciences) for up to 60 days. Mice were orally gavaged with either vehicle or ruxolitinib (45.0mg/kg or 90.0mg/kg) (generously provided by Incyte Corporation, Wilmington, DE, USA) for 21 days. Survival rates were analyzed by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS: We observed that ruxolitinib significantly decreased the phosphorylation of STAT3, -5 and -6 in Karpas-1106P PMBL and L-428 HL cells. The reduction in cell proliferation by 10uM ruxolitinib are 41% in Karpas-1106P (p CONCLUSIONS: Ruxolitinib significantly inhibited JAK2 enzymatic activity by the abrogation of the phosphorylation of STAT3, STAT5 and STAT6, and ruxolitinib showed significant anti-proliferative effects. Ruxolitinib (45mg/kg) significantly inhibited tumor progression and significantly prolonged survival in PMBL and HL xenografted NSG mice compared to control mice. Ruxolitinib may be a potential adjuvant agent in the treatment of PMBL and HL. Future studies will focus on whether the addition of ruxolitinib to chemoimmunotherapy improves the survival in a NSG PMBL and HL xenograft Mouse models. Download : Download high-res image (68KB) Download : Download full-size image Disclosures No relevant conflicts of interest to declare.

Changhong Yin - One of the best experts on this subject based on the ideXlab platform.

  • obinutuzumab ga101 vs rituximab significantly enhances cell death antibody dependent cytotoxicity and improves overall survival against cd20 primary mediastinal b cell lymphoma pmbl in a xenograft nod scid il2rgnull NSG Mouse model a potential target
    Oncotarget, 2020
    Co-Authors: Yaya Chu, Aradhana Awasthi, Sanghoon Lee, Dina Edani, Changhong Yin, Jessica Hochberg, Tishi Shah, Taehoon Chung, Janet Ayello, Carmella Van De Ven
    Abstract:

    Primary mediastinal large B-cell lymphoma (PMBL), a distinct mature B-cell lymphoma, expresses CD20 and has recently been successfully treated with the combination of a type I anti-CD20 monoclonal antibody, rituximab, with multiple combination chemotherapy regimens. Obinutuzumab is a glycoengineered type II anti-CD20 monoclonal antibody (mAb), recognizing a unique CD20 extracellular membrane epitope with enhanced antibody dependent cellular cytotoxicity (ADCC) vs rituximab. We hypothesize that obinutuzumab vs rituximab will significantly enhance in-vitro and in-vivo cytotoxicity against PMBL. PMBL cells were treated with equal dose of obinutuzumab and rituximab for 24 hours (1-100 μg/ml). ADCC were performed with ex-vivo expanded natural killer cells at 10:1 E: T ratio. Mice were xenografted with intravenous injections of luciferase expressing Karpas1106P cells and treated every 7 days for 8 weeks. Tumor burden was monitored by IVIS spectrum system. Compared with rituximab, obinutuzumab significantly inhibited PMBL cell proliferation (p = 0.01), promoted apoptosis (p = 0.05) and enhanced ADCC (p = 0.0002) against PMBL. Similarly, in PMBL xenografted NOD scid gamma mice, obinutuzumab significantly enhanced survival than rituximab when treated with equal doses (p = 0.05). Taken together our results suggest that obinutuzumab significantly enhanced natural killer cytotoxicity, reduced PMBL proliferation and prolonged the overall survival in humanized PMBL xenografted NOD scid gamma mice.

  • abstract 3838 upregulation of dleu1 significantly prolongs survival in a rituximab treated dleu1 knockout human burkitt lymphoma bl xenograft NSG Mouse model dleu1 may act as a tumor suppressor gene in pediatric bl
    Cancer Research, 2016
    Co-Authors: Sanghoon Lee, Changhong Yin, Tishi Shah, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    BACKGROUND: Burkitt lymphoma (BL) is the most common histological subtype of non-Hodgkin lymphoma (NHL) in children and adolescents (Cairo et al., Blood 2007; Miles/Cairo., BJHaem 2012). We identified that pediatric BL patients with chromosome 13q deletion, particularly 13q14.3, had significantly poorer outcome and inferior overall survival despite aggressive short, intensive multi-agent chemotherapy (Poirel/Cairo et al., Leuk 2009; Nelson/Cairo/Sanger et al., BJH 2009). Deleted in lymphocytic leukemia 1 (DLEU1) is a BL classifier gene in the 13q14.3 region (Dave et al., NEJM 2006) and interacts with C-MYC, TUBB2C and UBR1 (Stelzl et al., Cell 2005). We have previously reported a regulated DLEU1 expression significantly associated with changes in BL proliferation in vitro (Lee/Cairo et al., AACR 2015). We hypothesize that DLEU1 may have function as a tumor suppressor gene; however, functions and mechanisms underlying DLEU1 expression in the BL are poorly understood. OBJECTIVES: We hypothesize that DLEU1 may act as a tumor suppressor gene in BL and therefore examined whether up-regulation of DLEU1 results in changes in survival following targeted immunotherapy. METHODS: DLEU1 stably overexpressing Raji BL cells (Lee/Cairo et al., ASH 2013) were transfected with a firefly luciferase expression plasmid, kindly provided by Laurence Cooper MD, PhD. Four- to six- week-old female NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice from The Jackson Lab. Tumor burden and tumor progression were monitored by bioluminescence imaging system. Mice were treated with either rituximab (30 mg/kg) or cyclophosphamide (25 mg/kg) by intraperitoneal injection at 7 day intervals. Survival rates were analyzed by the Kaplan-Meier method using the Prism Version 6.0 software. RESULTS: We observed that DLEU1 stably overexpressing Raji BL cells xenografted (GFP-DLEU1) mice had significantly extended survival compared to GFP control mice following treatment rituximab (59 vs 51 days, p CONCLUSIONS: We demonstrated that the upregulation of DLEU1 expression significantly improved the survival in DLEU1 overexpressing BL cells xenografted NSG mice following rituximab, cyclophosphamide and in combination treatment. Therefore, the up-regulation of DLEU1 expression in BL may in part result in sensitizing to chemoimmunotherapy induced survival and may result in a consideration of potential therapeutic strategies Citation Format: Sanghoon Lee, Changhong Yin, Tishi Shah, Janet Ayello, Erin Morris, Carmella van de Ven, Mitchell S. Cairo. Upregulation of DLEU1 significantly prolongs survival in a rituximab-treated DLEU1 knockout human Burkitt lymphoma (BL) xenograft NSG Mouse model: DLEU1 may act as a tumor suppressor gene in pediatric BL. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3838.

  • ibrutinib significantly prolonged survival in a human burkitt lymphoma bl xenograft NSG Mouse model ibrutinib may be a potential adjuvant agent in the treatment of bl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Timmy Oconnell, Lauren Harrison, Matthew J Barth, Rodney R Miles, Paul J Galardy
    Abstract:

    BACKGROUND : Burkitt Lymphoma (BL) represents approximately 40% of all childhood and adolescent non-Hodgkin lymphoma (Miles/Cairo, BJH 2012 ). Children with relapsed or progressive BL develop chemoimmunotherapy-resistant disease and can rarely be cured with salvage therapy (Cairo et al, JCO 2012). Bruton's tyrosine kinase (BTK) is a regulator of normal B-cell development and is activated upon B-cell receptor (BCR) stimulation. Chronic active BCR signaling through BTK activation can be inhibited by the selective and covalent BTK inhibitor, ibrutinib (Young/Staudt et al, Nat Rev Drug Dicov 2013). Preclinical studies of ibrutinib in chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL) suggest that its inhibitory effect on cell proliferation is in the range of 1.0uM to 25.0uM (Herman et al, Blood 2011; Cinar et al, Leuk Res 2013), which is higher than the typical plasma concentration range in patients at the recommended dose of 560mg (<0.6uM) (Advani et al, JCO 2013). Despite these relatively high IC50 values in vitro , ibrutinib has been highly effective in the treatment of patients with refractory CLL and MCL (Byrd et al, NEJM 2013 and Wang et al, NEJM 2013). Ibrutinib was initially approved by the FDA (IMBRUVICA, USPI) for patients with MCL in November 2013 and CLL in February 2014, who have received at least one prior therapy. BL, however, is associated with tonic vs chronic active BCR signaling; while, both CLL and MCL have chronic active BCR signaling. These findings suggest that the antitumor activity of ibrutinib in BL, which is associated with tonic BCR signaling, may be less pronounced than in Activated B cell-like diffuse large B-cell lymphoma (ABC-DLBCL), CLL or MCL. OBJECTIVES: We hypothesize that ibrutinib may be a potential adjuvant agent in the treatment of BL. Therefore, we investigated the in vivo anti-tumor activity of ibrutinib in BL xenografted NSG mice. METHODS : Raji Burkitt lymphoma (BL) cells (ATCC) were exposed to ibrutinib (0-10uM, generously provided by Janssen R&D LLC) alone and in combination with dexamethasone (1uM) for 5 days and then MTS cell proliferation assay (Promega) was performed. The IC50 values were determined with CompuSynTM software (Chou and Martin, ComboSyn, 2005) based on data derived from MTS assay. Raji BL cells were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. NSG mice (NOD.Cg-Prkdcscid Il2rgtm1Wjl /SzJ, 6-8 weeks old) from Jackson laboratory were irradiated (2.5 Gy) and then mice were subcutaneously injected with one million ffluc-zeo tumor cells. Tumor progression and tumor burden were monitored by Bioluminescent Imaging system using the Xenogen IVIS-200 (Caliper Life Sciences). Mice were orally gavaged with either vehicle or ibrutinib (12.5mg/kg) for 10 days (once a day) at post-tumor cells injection. Analysis of survival rates were determined by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS : Ibrutinib compared to control induced a significant dose-dependent decrease in cell proliferation in Raji BL cells (5uM, 0.561 ± 0.170, p<0.03, IC50=5.20uM) following 120 hours (5 days) of ibrutinib treatment. Interestingly, we observed a significant decrease in cell proliferation in Raji BL cells (5uM, 0.213 ± 0.078, p<0.002, IC=0.50 ± 0.374uM) following ibrutinib and 1uM dexamethasone combination treatment following 120 hours (5 days) of treatment. We observed a significant decrease of tumor progression by tumor luminescence signal intensity following ibrutinib treated Raji BL xenografted NSG mice at day 20 (12.5mg/kg, p<0.001 and 25mg/kg, p<0.05) and day 25 (12.5mg/kg, p<0.05 and 25mg/kg, p<0.05) compared to control (Figure 1A).Ibrutinib (12.5 mg/kg) treated mice (n=39) significantly prolonged survival compared to vehicle controls (n=28) (p<0.0001) (Figure 1B). CONCLUSIONS : Ibrutinib (12.5mg/kg) significantly decreased tumor progression and significantly increased survival in Raji BL xenografted NSG mice, suggesting that ibrutinib may be a potential adjuvant agent for therapy in the treatment of BL. Future studies will investigate the efficacy of combination therapy with ibrutinib on survival in BL xenografted NOD/SCID mice. ![Figure][1] Disclosures Galardy: Mission Therapeutics: Research Funding. [1]: pending:yes

  • downregulation of dleu1 significantly shortened survival in a rituximab treated dleu1 knockout human burkitt lymphoma bl xenograft NSG Mouse model dleu1 may act as a tumor suppressor gene in pediatric bl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    BACKGROUND : Burkitt lymphoma (BL) is the most common histological subtype of non-Hodgkin lymphoma (NHL) in children and adolescents (Cairo et al., Blood, 2007; Miles/Cairo, BJH, 2012). We have previously identified secondary chromosomal aberrations in 70% of pediatric BL patients (PBL) with a C-MYC gene rearrangement (Poirel/Cairo et al., Leukemia, 2009). Specifically, we identified significantly inferior event free survival (EFS) and overall survival in children and adolescents with a specific loss of the 13q14.3 locus (Poirel/Cairo et al., Leukemia, 2009; Nelson/Cairo/Sanger et al., Br. J. Haematol., 2009). In a multivariate analysis controlling for stage, lactate dehydrogenase levels, country of treatment, and group classification, children with BL who had a 13q deletion had significantly poorer EFS compared to the remainder of patients treated with the same French-American-British (FAB) chemotherapy regimen (Poirel/Cairo et al., Leukemia, 2009). Deleted in lymphocytic leukemia 1 (DLEU1) is a BL classifier gene in the 13q14.3 region (Dave et al., NEJM, 2006) and interacts with C-MYC , Tubulin beta-2C chain (TUBB2C), E3 ubiquitin-protein ligase (UBR1), cellular tumor antigen p53, and Ras association (RalGDS/AF-6) domain family member 1 (RASSF1) (Stelzl et al., Cell, 2005). The sequence-specific Transcription Activator-Like Effector Nucleases (TALENs) have been developed for targeted genome editing in in vitro and in vivo experiments with high efficiency as a new therapeutic tool (Sander et al., Nat. Biotechnol., 2011). We have previously reported a down-regulated DLEU1 mRNA expression significantly associated with an increase in BL proliferation in vitro (Lee/Cairo et al., AACR, 2015). We hypothesize that DLEU1 may have function as a tumor suppressor gene; however, the role of DLEU1 regulating programmed cell death in BL are poorly understood. OBJECTIVES : We hypothesize that DLEU1 may act as a tumor suppressor gene in BL and whether down-regulation of DLEU1 expression results in changes in BL survival following targeted immunotherapy. METHODS : TALENs induced DLEU1 knockout Raji cells were previously generated (Lee/Cairo et al., ASH, 2013) and DLEU1 knockout cells were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. Four- to six- week-old female NSG (NOD.Cg-Prkdc scid Il2rg tm1Wjl /SzJ) mice from The Jackson Laboratory were irradiated (2.5 Gy), and then mice were subcutaneously injected with 1x10 6 ffluc-zeo tumor cells. Tumor burden and tumor progression were monitored by bioluminescence imaging system using the Xenogen IVIS-200 (Caliper Life Sciences). Mice were treated with either PBS, IgG isotype control, rituximab (30 mg/kg) or cyclophosphamide (25 mg/kg) and in combination by intraperitoneal (i.p) injection at 7 day intervals. Survival rates were analyzed by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS: There were significant increases of luciferase signal intensity in DLEU1 knockout mice (DLEU1-KO) compared to that in wild type (WT) mice on day 31 of rituximab (p vs 48 days, p CONCLUSIONS : The down-regulation of DLEU1 expression significantly decreased the survival rate in DLEU1-KO xenografted NSG mice following rituximab and in combination with cyclophosphamide treatment. Therefore, the down-regulation of DLEU1 expression in BL may in part result in immunotherapy resistance and may result in a consideration of alternative therapeutic strategies. Disclosures No relevant conflicts of interest to declare.

  • ruxolitinib significantly prolongs survival in both a primary mediastinal b cell lymphoma pmbl and hodgkin lymphoma hl xenograft NSG Mouse model ruxolitinib may be a potential adjuvant agent in the treatment of pmbl and of hl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    Abstract BACKGROUND: Primary Mediastinal large B-cell lymphoma (PMBL) and Hodgkin lymphoma (HL) are two of the most common malignancies among adolescents and young adults (AYA). PMBL and HL share similar molecular features by gene expression profiling and pathogenesis (Rosenwald et al., J Exp Med., 2003). HL represents only approximately 4-5% of all cancers in children younger than 15 years of age, but is the most common cancer in the 15-35 yrs AYA group. While the prognosis is excellent with AYA HL, there are significant late effects secondary to chemoradiotherapy and therefore new targeted agents are needed to avoid these morbid late effects in HL (Hochberg/Cairo et al., Br. J. Haem., 2009). Frequent gains of chromosome 9p exhibit higher Janus Kinase 2 (JAK2) transcript levels with increased JAK2 activity (Bentz et al., Genes Chromosomes Cancer, 2001), suggesting aberrant activity of JAK2 and Signal Transducer and Activator of Transcription (STAT) pathways, which may in part play an important role in the pathogenesis of HL and PMBL. Ruxolitinib is a potent and selective JAK1/JAK2 inhibitor against myeloproliferative neoplasms (MPNs) that consistently exhibits dysregulation of the JAK1/JAK2 pathway, including those MPNs with a JAK2V617F mutation. Ruxolitinb also inhibits JAK2/STAT5 signaling in vitro and in a murine model of MPN (Quintas-Cardama et al., Blood, 2010). OBJECTIVES: We hypothesize that ruxolitinib may potentially function as targeted therapeutic agent for both PMBL and HL and therefore, investigated the efficacy of ruxolitinib in PMBL and HL cells xenografted into NSG mice. METHODS: Cell proliferation and apoptosis analysis were assessed using MTS and Caspase-3/7 assay (Promega), respectively. The expression of protein was examined by immunoblotting and statistical significance was determined by Student t-test. Karpas-1106P PMBL cell line and L-428 HL cell line were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. The six to eight weeks old Female NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice were from Jackson laboratory. The ffluc-zeo NSG mice were irradiated (2.5 Gy) and then were subcutaneously injected with 1x106 ffluc-zeo Karpas-1106P or L-428 tumor cells. Tumor burden and tumor progression were monitored by bioluminescence imaging (BLI) using the Xenogen IVIS-200 (Caliper Life Sciences) for up to 60 days. Mice were orally gavaged with either vehicle or ruxolitinib (45.0mg/kg or 90.0mg/kg) (generously provided by Incyte Corporation, Wilmington, DE, USA) for 21 days. Survival rates were analyzed by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS: We observed that ruxolitinib significantly decreased the phosphorylation of STAT3, -5 and -6 in Karpas-1106P PMBL and L-428 HL cells. The reduction in cell proliferation by 10uM ruxolitinib are 41% in Karpas-1106P (p CONCLUSIONS: Ruxolitinib significantly inhibited JAK2 enzymatic activity by the abrogation of the phosphorylation of STAT3, STAT5 and STAT6, and ruxolitinib showed significant anti-proliferative effects. Ruxolitinib (45mg/kg) significantly inhibited tumor progression and significantly prolonged survival in PMBL and HL xenografted NSG mice compared to control mice. Ruxolitinib may be a potential adjuvant agent in the treatment of PMBL and HL. Future studies will focus on whether the addition of ruxolitinib to chemoimmunotherapy improves the survival in a NSG PMBL and HL xenograft Mouse models. Download : Download high-res image (68KB) Download : Download full-size image Disclosures No relevant conflicts of interest to declare.

Janet Ayello - One of the best experts on this subject based on the ideXlab platform.

  • obinutuzumab ga101 vs rituximab significantly enhances cell death antibody dependent cytotoxicity and improves overall survival against cd20 primary mediastinal b cell lymphoma pmbl in a xenograft nod scid il2rgnull NSG Mouse model a potential target
    Oncotarget, 2020
    Co-Authors: Yaya Chu, Aradhana Awasthi, Sanghoon Lee, Dina Edani, Changhong Yin, Jessica Hochberg, Tishi Shah, Taehoon Chung, Janet Ayello, Carmella Van De Ven
    Abstract:

    Primary mediastinal large B-cell lymphoma (PMBL), a distinct mature B-cell lymphoma, expresses CD20 and has recently been successfully treated with the combination of a type I anti-CD20 monoclonal antibody, rituximab, with multiple combination chemotherapy regimens. Obinutuzumab is a glycoengineered type II anti-CD20 monoclonal antibody (mAb), recognizing a unique CD20 extracellular membrane epitope with enhanced antibody dependent cellular cytotoxicity (ADCC) vs rituximab. We hypothesize that obinutuzumab vs rituximab will significantly enhance in-vitro and in-vivo cytotoxicity against PMBL. PMBL cells were treated with equal dose of obinutuzumab and rituximab for 24 hours (1-100 μg/ml). ADCC were performed with ex-vivo expanded natural killer cells at 10:1 E: T ratio. Mice were xenografted with intravenous injections of luciferase expressing Karpas1106P cells and treated every 7 days for 8 weeks. Tumor burden was monitored by IVIS spectrum system. Compared with rituximab, obinutuzumab significantly inhibited PMBL cell proliferation (p = 0.01), promoted apoptosis (p = 0.05) and enhanced ADCC (p = 0.0002) against PMBL. Similarly, in PMBL xenografted NOD scid gamma mice, obinutuzumab significantly enhanced survival than rituximab when treated with equal doses (p = 0.05). Taken together our results suggest that obinutuzumab significantly enhanced natural killer cytotoxicity, reduced PMBL proliferation and prolonged the overall survival in humanized PMBL xenografted NOD scid gamma mice.

  • abstract 3838 upregulation of dleu1 significantly prolongs survival in a rituximab treated dleu1 knockout human burkitt lymphoma bl xenograft NSG Mouse model dleu1 may act as a tumor suppressor gene in pediatric bl
    Cancer Research, 2016
    Co-Authors: Sanghoon Lee, Changhong Yin, Tishi Shah, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    BACKGROUND: Burkitt lymphoma (BL) is the most common histological subtype of non-Hodgkin lymphoma (NHL) in children and adolescents (Cairo et al., Blood 2007; Miles/Cairo., BJHaem 2012). We identified that pediatric BL patients with chromosome 13q deletion, particularly 13q14.3, had significantly poorer outcome and inferior overall survival despite aggressive short, intensive multi-agent chemotherapy (Poirel/Cairo et al., Leuk 2009; Nelson/Cairo/Sanger et al., BJH 2009). Deleted in lymphocytic leukemia 1 (DLEU1) is a BL classifier gene in the 13q14.3 region (Dave et al., NEJM 2006) and interacts with C-MYC, TUBB2C and UBR1 (Stelzl et al., Cell 2005). We have previously reported a regulated DLEU1 expression significantly associated with changes in BL proliferation in vitro (Lee/Cairo et al., AACR 2015). We hypothesize that DLEU1 may have function as a tumor suppressor gene; however, functions and mechanisms underlying DLEU1 expression in the BL are poorly understood. OBJECTIVES: We hypothesize that DLEU1 may act as a tumor suppressor gene in BL and therefore examined whether up-regulation of DLEU1 results in changes in survival following targeted immunotherapy. METHODS: DLEU1 stably overexpressing Raji BL cells (Lee/Cairo et al., ASH 2013) were transfected with a firefly luciferase expression plasmid, kindly provided by Laurence Cooper MD, PhD. Four- to six- week-old female NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice from The Jackson Lab. Tumor burden and tumor progression were monitored by bioluminescence imaging system. Mice were treated with either rituximab (30 mg/kg) or cyclophosphamide (25 mg/kg) by intraperitoneal injection at 7 day intervals. Survival rates were analyzed by the Kaplan-Meier method using the Prism Version 6.0 software. RESULTS: We observed that DLEU1 stably overexpressing Raji BL cells xenografted (GFP-DLEU1) mice had significantly extended survival compared to GFP control mice following treatment rituximab (59 vs 51 days, p CONCLUSIONS: We demonstrated that the upregulation of DLEU1 expression significantly improved the survival in DLEU1 overexpressing BL cells xenografted NSG mice following rituximab, cyclophosphamide and in combination treatment. Therefore, the up-regulation of DLEU1 expression in BL may in part result in sensitizing to chemoimmunotherapy induced survival and may result in a consideration of potential therapeutic strategies Citation Format: Sanghoon Lee, Changhong Yin, Tishi Shah, Janet Ayello, Erin Morris, Carmella van de Ven, Mitchell S. Cairo. Upregulation of DLEU1 significantly prolongs survival in a rituximab-treated DLEU1 knockout human Burkitt lymphoma (BL) xenograft NSG Mouse model: DLEU1 may act as a tumor suppressor gene in pediatric BL. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3838.

  • ibrutinib significantly prolonged survival in a human burkitt lymphoma bl xenograft NSG Mouse model ibrutinib may be a potential adjuvant agent in the treatment of bl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Timmy Oconnell, Lauren Harrison, Matthew J Barth, Rodney R Miles, Paul J Galardy
    Abstract:

    BACKGROUND : Burkitt Lymphoma (BL) represents approximately 40% of all childhood and adolescent non-Hodgkin lymphoma (Miles/Cairo, BJH 2012 ). Children with relapsed or progressive BL develop chemoimmunotherapy-resistant disease and can rarely be cured with salvage therapy (Cairo et al, JCO 2012). Bruton's tyrosine kinase (BTK) is a regulator of normal B-cell development and is activated upon B-cell receptor (BCR) stimulation. Chronic active BCR signaling through BTK activation can be inhibited by the selective and covalent BTK inhibitor, ibrutinib (Young/Staudt et al, Nat Rev Drug Dicov 2013). Preclinical studies of ibrutinib in chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL) suggest that its inhibitory effect on cell proliferation is in the range of 1.0uM to 25.0uM (Herman et al, Blood 2011; Cinar et al, Leuk Res 2013), which is higher than the typical plasma concentration range in patients at the recommended dose of 560mg (<0.6uM) (Advani et al, JCO 2013). Despite these relatively high IC50 values in vitro , ibrutinib has been highly effective in the treatment of patients with refractory CLL and MCL (Byrd et al, NEJM 2013 and Wang et al, NEJM 2013). Ibrutinib was initially approved by the FDA (IMBRUVICA, USPI) for patients with MCL in November 2013 and CLL in February 2014, who have received at least one prior therapy. BL, however, is associated with tonic vs chronic active BCR signaling; while, both CLL and MCL have chronic active BCR signaling. These findings suggest that the antitumor activity of ibrutinib in BL, which is associated with tonic BCR signaling, may be less pronounced than in Activated B cell-like diffuse large B-cell lymphoma (ABC-DLBCL), CLL or MCL. OBJECTIVES: We hypothesize that ibrutinib may be a potential adjuvant agent in the treatment of BL. Therefore, we investigated the in vivo anti-tumor activity of ibrutinib in BL xenografted NSG mice. METHODS : Raji Burkitt lymphoma (BL) cells (ATCC) were exposed to ibrutinib (0-10uM, generously provided by Janssen R&D LLC) alone and in combination with dexamethasone (1uM) for 5 days and then MTS cell proliferation assay (Promega) was performed. The IC50 values were determined with CompuSynTM software (Chou and Martin, ComboSyn, 2005) based on data derived from MTS assay. Raji BL cells were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. NSG mice (NOD.Cg-Prkdcscid Il2rgtm1Wjl /SzJ, 6-8 weeks old) from Jackson laboratory were irradiated (2.5 Gy) and then mice were subcutaneously injected with one million ffluc-zeo tumor cells. Tumor progression and tumor burden were monitored by Bioluminescent Imaging system using the Xenogen IVIS-200 (Caliper Life Sciences). Mice were orally gavaged with either vehicle or ibrutinib (12.5mg/kg) for 10 days (once a day) at post-tumor cells injection. Analysis of survival rates were determined by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS : Ibrutinib compared to control induced a significant dose-dependent decrease in cell proliferation in Raji BL cells (5uM, 0.561 ± 0.170, p<0.03, IC50=5.20uM) following 120 hours (5 days) of ibrutinib treatment. Interestingly, we observed a significant decrease in cell proliferation in Raji BL cells (5uM, 0.213 ± 0.078, p<0.002, IC=0.50 ± 0.374uM) following ibrutinib and 1uM dexamethasone combination treatment following 120 hours (5 days) of treatment. We observed a significant decrease of tumor progression by tumor luminescence signal intensity following ibrutinib treated Raji BL xenografted NSG mice at day 20 (12.5mg/kg, p<0.001 and 25mg/kg, p<0.05) and day 25 (12.5mg/kg, p<0.05 and 25mg/kg, p<0.05) compared to control (Figure 1A).Ibrutinib (12.5 mg/kg) treated mice (n=39) significantly prolonged survival compared to vehicle controls (n=28) (p<0.0001) (Figure 1B). CONCLUSIONS : Ibrutinib (12.5mg/kg) significantly decreased tumor progression and significantly increased survival in Raji BL xenografted NSG mice, suggesting that ibrutinib may be a potential adjuvant agent for therapy in the treatment of BL. Future studies will investigate the efficacy of combination therapy with ibrutinib on survival in BL xenografted NOD/SCID mice. ![Figure][1] Disclosures Galardy: Mission Therapeutics: Research Funding. [1]: pending:yes

  • downregulation of dleu1 significantly shortened survival in a rituximab treated dleu1 knockout human burkitt lymphoma bl xenograft NSG Mouse model dleu1 may act as a tumor suppressor gene in pediatric bl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    BACKGROUND : Burkitt lymphoma (BL) is the most common histological subtype of non-Hodgkin lymphoma (NHL) in children and adolescents (Cairo et al., Blood, 2007; Miles/Cairo, BJH, 2012). We have previously identified secondary chromosomal aberrations in 70% of pediatric BL patients (PBL) with a C-MYC gene rearrangement (Poirel/Cairo et al., Leukemia, 2009). Specifically, we identified significantly inferior event free survival (EFS) and overall survival in children and adolescents with a specific loss of the 13q14.3 locus (Poirel/Cairo et al., Leukemia, 2009; Nelson/Cairo/Sanger et al., Br. J. Haematol., 2009). In a multivariate analysis controlling for stage, lactate dehydrogenase levels, country of treatment, and group classification, children with BL who had a 13q deletion had significantly poorer EFS compared to the remainder of patients treated with the same French-American-British (FAB) chemotherapy regimen (Poirel/Cairo et al., Leukemia, 2009). Deleted in lymphocytic leukemia 1 (DLEU1) is a BL classifier gene in the 13q14.3 region (Dave et al., NEJM, 2006) and interacts with C-MYC , Tubulin beta-2C chain (TUBB2C), E3 ubiquitin-protein ligase (UBR1), cellular tumor antigen p53, and Ras association (RalGDS/AF-6) domain family member 1 (RASSF1) (Stelzl et al., Cell, 2005). The sequence-specific Transcription Activator-Like Effector Nucleases (TALENs) have been developed for targeted genome editing in in vitro and in vivo experiments with high efficiency as a new therapeutic tool (Sander et al., Nat. Biotechnol., 2011). We have previously reported a down-regulated DLEU1 mRNA expression significantly associated with an increase in BL proliferation in vitro (Lee/Cairo et al., AACR, 2015). We hypothesize that DLEU1 may have function as a tumor suppressor gene; however, the role of DLEU1 regulating programmed cell death in BL are poorly understood. OBJECTIVES : We hypothesize that DLEU1 may act as a tumor suppressor gene in BL and whether down-regulation of DLEU1 expression results in changes in BL survival following targeted immunotherapy. METHODS : TALENs induced DLEU1 knockout Raji cells were previously generated (Lee/Cairo et al., ASH, 2013) and DLEU1 knockout cells were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. Four- to six- week-old female NSG (NOD.Cg-Prkdc scid Il2rg tm1Wjl /SzJ) mice from The Jackson Laboratory were irradiated (2.5 Gy), and then mice were subcutaneously injected with 1x10 6 ffluc-zeo tumor cells. Tumor burden and tumor progression were monitored by bioluminescence imaging system using the Xenogen IVIS-200 (Caliper Life Sciences). Mice were treated with either PBS, IgG isotype control, rituximab (30 mg/kg) or cyclophosphamide (25 mg/kg) and in combination by intraperitoneal (i.p) injection at 7 day intervals. Survival rates were analyzed by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS: There were significant increases of luciferase signal intensity in DLEU1 knockout mice (DLEU1-KO) compared to that in wild type (WT) mice on day 31 of rituximab (p vs 48 days, p CONCLUSIONS : The down-regulation of DLEU1 expression significantly decreased the survival rate in DLEU1-KO xenografted NSG mice following rituximab and in combination with cyclophosphamide treatment. Therefore, the down-regulation of DLEU1 expression in BL may in part result in immunotherapy resistance and may result in a consideration of alternative therapeutic strategies. Disclosures No relevant conflicts of interest to declare.

  • ruxolitinib significantly prolongs survival in both a primary mediastinal b cell lymphoma pmbl and hodgkin lymphoma hl xenograft NSG Mouse model ruxolitinib may be a potential adjuvant agent in the treatment of pmbl and of hl
    Blood, 2015
    Co-Authors: Sanghoon Lee, Changhong Yin, Janet Ayello, Carmella Van De Ven, Erin Morris, Mitchell S Cairo
    Abstract:

    Abstract BACKGROUND: Primary Mediastinal large B-cell lymphoma (PMBL) and Hodgkin lymphoma (HL) are two of the most common malignancies among adolescents and young adults (AYA). PMBL and HL share similar molecular features by gene expression profiling and pathogenesis (Rosenwald et al., J Exp Med., 2003). HL represents only approximately 4-5% of all cancers in children younger than 15 years of age, but is the most common cancer in the 15-35 yrs AYA group. While the prognosis is excellent with AYA HL, there are significant late effects secondary to chemoradiotherapy and therefore new targeted agents are needed to avoid these morbid late effects in HL (Hochberg/Cairo et al., Br. J. Haem., 2009). Frequent gains of chromosome 9p exhibit higher Janus Kinase 2 (JAK2) transcript levels with increased JAK2 activity (Bentz et al., Genes Chromosomes Cancer, 2001), suggesting aberrant activity of JAK2 and Signal Transducer and Activator of Transcription (STAT) pathways, which may in part play an important role in the pathogenesis of HL and PMBL. Ruxolitinib is a potent and selective JAK1/JAK2 inhibitor against myeloproliferative neoplasms (MPNs) that consistently exhibits dysregulation of the JAK1/JAK2 pathway, including those MPNs with a JAK2V617F mutation. Ruxolitinb also inhibits JAK2/STAT5 signaling in vitro and in a murine model of MPN (Quintas-Cardama et al., Blood, 2010). OBJECTIVES: We hypothesize that ruxolitinib may potentially function as targeted therapeutic agent for both PMBL and HL and therefore, investigated the efficacy of ruxolitinib in PMBL and HL cells xenografted into NSG mice. METHODS: Cell proliferation and apoptosis analysis were assessed using MTS and Caspase-3/7 assay (Promega), respectively. The expression of protein was examined by immunoblotting and statistical significance was determined by Student t-test. Karpas-1106P PMBL cell line and L-428 HL cell line were stably transfected with a firefly luciferase expression plasmid (ffluc-zeo), kindly provided by Laurence Cooper MD, PhD. The six to eight weeks old Female NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice were from Jackson laboratory. The ffluc-zeo NSG mice were irradiated (2.5 Gy) and then were subcutaneously injected with 1x106 ffluc-zeo Karpas-1106P or L-428 tumor cells. Tumor burden and tumor progression were monitored by bioluminescence imaging (BLI) using the Xenogen IVIS-200 (Caliper Life Sciences) for up to 60 days. Mice were orally gavaged with either vehicle or ruxolitinib (45.0mg/kg or 90.0mg/kg) (generously provided by Incyte Corporation, Wilmington, DE, USA) for 21 days. Survival rates were analyzed by the Kaplan-Meier method and differences evaluated by log-rank test using the Prism Version 6.0 software. RESULTS: We observed that ruxolitinib significantly decreased the phosphorylation of STAT3, -5 and -6 in Karpas-1106P PMBL and L-428 HL cells. The reduction in cell proliferation by 10uM ruxolitinib are 41% in Karpas-1106P (p CONCLUSIONS: Ruxolitinib significantly inhibited JAK2 enzymatic activity by the abrogation of the phosphorylation of STAT3, STAT5 and STAT6, and ruxolitinib showed significant anti-proliferative effects. Ruxolitinib (45mg/kg) significantly inhibited tumor progression and significantly prolonged survival in PMBL and HL xenografted NSG mice compared to control mice. Ruxolitinib may be a potential adjuvant agent in the treatment of PMBL and HL. Future studies will focus on whether the addition of ruxolitinib to chemoimmunotherapy improves the survival in a NSG PMBL and HL xenograft Mouse models. Download : Download high-res image (68KB) Download : Download full-size image Disclosures No relevant conflicts of interest to declare.