Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Omar Elnaggar is active.

Publication


Featured researches published by Omar Elnaggar.


Cancer Immunology, Immunotherapy | 2014

Bioactive compounds or metabolites from black raspberries modulate T lymphocyte proliferation, myeloid cell differentiation and Jak/STAT signaling

Thomas A. Mace; Samantha A. King; Zeenath Ameen; Omar Elnaggar; Gregory S. Young; Kenneth M. Riedl; Steven J. Schwartz; Steven K. Clinton; Thomas J. Knobloch; Christopher M. Weghorst; Gregory B. Lesinski

Bioactive phytochemicals from natural products, such as black raspberries (BRB; Rubus occidentalis), have direct anticancer properties on malignant cells in culture and in xenograft models. BRB components inhibit cancer progression in more complex rodent carcinogenesis models. Although mechanistic targets for BRB phytochemicals in cancer cells are beginning to emerge, the potential role in modulating host immune processes impacting cancer have not been systematically examined. We hypothesized that BRB contain compounds capable of eliciting potent immunomodulatory properties that impact cellular mediators relevant to chronic inflammation and tumor progression. We studied both an ethanol extract from black raspberries (BRB-E) containing a diverse mixture of phytochemicals and two abundant phytochemical metabolites of BRB produced upon ingestion (Cyanidin-3-Rutinoside, C3R; Quercitin-3-Rutinoside, Q3R). BRB-E inhibited proliferation, and viability of CD3/CD28 activated human CD4+ and CD8+ T lymphocytes. BRB-E also limited in vitro expansion of myeloid-derived suppressor cells (MDSC) and their suppressive capacity. Pre-treatment of immune cells with BRB-E attenuated IL-6-mediated phosphorylation of signal transducer and activator of transcription-3 (STAT3) and IL-2-induced STAT5 phosphorylation. In contrast, pre-treatment of immune cells with the C3R and Q3R metabolites inhibited MDSC expansion, IL-6-mediated STAT3 signaling, but not IL-2-induced STAT5 phosphorylation and were less potent inhibitors of T cell viability. Together these data indicate that BRB extracts and their physiologically relevant metabolites contain phytochemicals that affect immune processes relevant to carcinogenesis and immunotherapy. Furthermore, specific BRB components and their metabolites may be a source of lead compounds for drug development that exhibits targeted immunological outcomes or inhibition of specific STAT-regulated signaling pathways.


Molecular Cancer Therapeutics | 2017

Dual Inhibition of MEK and PI3K/Akt Rescues Cancer Cachexia through Both Tumor Extrinsic and Intrinsic Activities.

Erin E. Talbert; Jennifer Yang; Thomas A. Mace; Matthew R. Farren; Alton B. Farris; Gregory S. Young; Omar Elnaggar; Zheng Che; Cynthia Timmers; Priyani Rajasekera; Jennifer M. Maskarinec; Mark Bloomston; Tanios Bekaii-Saab; Denis C. Guttridge; Gregory B. Lesinski

Involuntary weight loss, a part of the cachexia syndrome, is a debilitating comorbidity of cancer and currently has no treatment options. Results from a recent clinical trial at our institution showed that biliary tract cancer patients treated with a MEK inhibitor exhibited poor tumor responses but surprisingly gained weight and increased their skeletal muscle mass. This implied that MEK inhibition might be anticachectic. To test this potential effect of MEK inhibition, we utilized the established Colon-26 model of cancer cachexia and the MEK1/2 inhibitor MEK162. Results showed that MEK inhibition effectively prevented muscle wasting. Importantly, MEK162 retained its ability to spare muscle loss even in mice bearing a Colon-26 clone resistant to the MEK inhibitor, demonstrating that the effects of blocking MEK are at least in part independent of the tumor. Because single-agent MEK inhibitors have been limited as a first-line targeted therapy due to compensatory activation of other oncogenic signaling pathways, we combined MEK162 with the PI3K/Akt inhibitor buparlisib. Results showed that this combinatorial treatment significantly reduced tumor growth due to a direct activity on Colon-26 tumor cells in vitro and in vivo, while also preserving skeletal muscle mass. Together, our results suggest that as a monotherapy, MEK inhibition preserves muscle mass, but when combined with a PI3K/Akt inhibitor exhibits potent antitumor activity. Thus, combinatorial therapy might serve as a new approach for the treatment of cancer cachexia. Mol Cancer Ther; 16(2); 344–56. ©2016 AACR. See related article by Kobayashi et al., p. 357


Oncotarget | 2015

Single agent BMS-911543 Jak2 inhibitor has distinct inhibitory effects on STAT5 signaling in genetically engineered mice with pancreatic cancer

Thomas A. Mace; Reena Shakya; Omar Elnaggar; Kristin Wilson; Hannah M. Komar; Jennifer Yang; Jason R. Pitarresi; Gregory S. Young; Michael C. Ostrowski; Thomas Ludwig; Tanios Bekaii-Saab; Mark Bloomston; Gregory B. Lesinski

The Jak/STAT pathway is activated in human pancreatic ductal adenocarcinoma (PDAC) and cooperates with mutant Kras to drive initiation and progression of PDAC in murine models. We hypothesized that the small-molecule Jak2 inhibitor (BMS-911543) would elicit anti-tumor activity against PDAC and decrease immune suppressive features of the disease. We used an aggressive genetically engineered PDAC model with mutant KrasG12D, tp53R270H, and Brca1 alleles (KPC-Brca1 mice). Mice with confirmed tumor burden were treated orally with vehicle or 30 mg/kg BMS-911543 daily for 14 days. Histologic analysis of pancreata from treated mice revealed fewer foci of adenocarcinoma and significantly decreased Ki67+ cells versus controls. In vivo administration of BMS-911543 significantly reduced pSTAT5 and FoxP3 positive cells within the pancreas, but did not alter STAT3 phosphorylation. Continuous dosing of KPC-Brca1 mice with BMS-911543 resulted in a median survival of 108 days, as compared to a median survival of 87 days in vehicle treated animals, a 23% increase (p = 0.055). In vitro experiments demonstrated that PDAC cell lines were poorly sensitive to BMS-911543, requiring high micromolar concentrations to achieve targeted inhibition of Jak/STAT signaling. Similarly, BMS-911543 had little in vitro effect on the viability of both murine and human PDAC-derived stellate cell lines. However, BMS-911543 potently inhibited phosphorylation of pSTAT3 and pSTAT5 at low micromolar doses in human PBMC and reduced in vitro differentiation of Foxp3+ T regulatory cells. These results indicate that single agent Jak2i deserves further study in preclinical models of PDAC and has distinct inhibitory effects on STAT5 mediated signaling.


Molecular Cancer Therapeutics | 2017

The exportin-1 inhibitor selinexor exerts superior anti-tumor activity when combined with T cell checkpoint inhibitors.

Matthew R. Farren; Rebecca C. Hennessey; Reena Shakya; Omar Elnaggar; Gregory S. Young; Kari Kendra; Yosef Landesman; Sivan Elloul; Marsha Crochiere; Boris Klebanov; Trinayan Kashyap; Christin E. Burd; Gregory B. Lesinski

Selinexor, a selective inhibitor of nuclear export (SINE) compound targeting exportin-1, has previously been shown to inhibit melanoma cell growth in vivo. We hypothesized that combining selinexor with antibodies that block or disrupt T-cell checkpoint molecule signaling would exert superior antimelanoma activity. In vitro, selinexor increased PDCD1 and CTLA4 gene expression in leukocytes and induced CD274 gene expression in human melanoma cell lines. Mice bearing syngeneic B16F10 melanoma tumors demonstrated a significant reduction in tumor growth rate in response to the combination of selinexor and anti-PD-1 or anti-PD-L1 antibodies (P < 0.05). Similar results were obtained in B16F10-bearing mice treated with selinexor combined with anti-CTLA4 antibody. Immunophenotypic analysis of splenocytes by flow cytometry revealed that selinexor alone or in combination with anti-PD-L1 antibody significantly increased the frequency of both natural killer cells (P ≤ 0.050) and CD4+ T cells with a Th1 phenotype (P ≤ 0.050). Further experiments indicated that the antitumor effect of selinexor in combination with anti-PD-1 therapy persisted under an alternative dosing schedule but was lost when selinexor was administered daily. These data indicate that the efficacy of selinexor against melanoma may be enhanced by disrupting immune checkpoint activity. Mol Cancer Ther; 16(3); 417–27. ©2017 AACR. See related article by Tyler et al., p. 428.


Journal for ImmunoTherapy of Cancer | 2015

Selinexor, a selective inhibitor of nuclear export (SINE), shows enhanced activity in combination with PD-1/PD-L1 blockade in syngeneic murine models of colon cancer and melanoma

Matthew R. Farren; Reena Shakya; Rebecca C. Hennessey; Thomas A. Mace; Jennifer Yang; Omar Elnaggar; Gregory S. Young; Yosef Landesman; Robert W. Carlson; Sivan Elloul; Marsha Crochiere; Christin E. Burd; Gregory B. Lesinski

Exportin-1 (XPO1) is a nuclear export protein with >220 cargo proteins, including tumor suppressors and cell cycle modulators. Selinexor is a SINE (Selective Inhibitor of Nuclear Export) compound that has been administered to >900 cancer patients in Phase I and II trials to date, with evidence of efficacy and tolerability. Selinexor blocks nuclear export of NFAT1c, STAT1 and STAT3, which are implicated in regulating the inhibitory T cell receptor PD-1 and its ligand, PD-L1. We hypothesized that selinexor would upregulate T cell checkpoint molecule expression, and that combination treatment with anti-PD-1 or anti-PD-L1 would thereby enhance the ability of selinexor to elicit antitumor activity. Selinexor increased PD-1 gene expression by ~2-fold in normal lymphocytes and induced PD-L1 gene expression in tumor cell lines. Mice bearing syngeneic colon tumors (colon26) treated with selinexor and anti-PD-1 for 2 weeks demonstrated a significant reduction in tumor growth rate (P < 0.05), while monotherapy with either agent had no significant effect on tumor growth. Similar results were obtained in mice bearing syngeneic B16F10 melanoma tumors, whereby combined treatment with selinexor + anti-PD-1 was superior to either single agent alone (p < 0.034). Combined therapy of mice bearing B16F10 tumors with selinexor and anti-PD-L1 was similarly effective, with significantly smaller tumors at the study endpoint (p < 0.001). No weight loss or signs of toxicity were evident in any in vivo study. Immunophenotypic analysis by flow cytometry revealed that selinexor alone or in combination with anti-PD-1/anti-PD-L1 significantly increased the percentage of splenic NK cells (p≤0.050), while selinexor ± anti-PD-L1 significantly increased the percentage of splenic Th1 T cells (p≤0.011), all compared to vehicle treated mice. Interestingly, combining selinexor with anti-PD-L1 significantly decreased the percentage of splenocytes that expressed PD-L1 (p < 0.001). These data indicate that the efficacy of selinexor may be enhanced by disrupting the pre-existing PD-1/PD-L1 signaling in effector cells (T and NK cells). Altogether, these data suggest that the efficacy of selinexor in combination with anti-PD-1 or anti PD L1 in mouse syngeneic tumor models may be due to both disrupting immunosuppressive PD-1/PD-L1 signaling and increasing the frequency of potentially tumor reactive NK cells and Th1 T cells. This provides a rational basis for this treatment combination as a novel therapeutic approach for advanced cancer.


Cancer Research | 2016

Abstract 2319: Selinexor, a selective inhibitor of nuclear export (SINE), enhances the in vivo efficacy of checkpoint blockade with antibodies targeting CTLA4 or PD-1/PD-L1 in melanoma

Matthew R. Farren; Reena Shakya; Rebecca C. Hennessey; Thomas A. Mace; Jennifer Yang; Omar Elnaggar; Gregory S. Young; Yosef Landesman; Robert W. Carlson; Sivan Elloul; Marsha Crochiere; Christin E. Burd; Gregory B. Lesinski

Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA Selinexor is a SINE (Selective Inhibitor of Nuclear Export) compound that has been administered to >1000 cancer patients in Phase I and II trials to date, with evidence of efficacy and tolerability. This small molecule targets exportin-1 (XPO1), a key nuclear export protein with >200 cargo proteins which include both tumor suppressors and cell cycle modulators. As a result, selinexor blocks nuclear export of proteins including IκB, NFAT1c, STAT1 and STAT3, which regulate expression of the inhibitory T cell receptors CTLA4, PD1 and its ligand, PD-L1. We hypothesized that selinexor would upregulate T cell checkpoint molecule expression, and thereby enhance the anti-tumor activity of antibodies targeting PD-1/PD-L1 or CTLA4. Human (A375, CHL-1) and murine (B16F10) melanoma cell lines expressed high levels of PD-L1 protein at baseline, and PD-L1 expression was induced following selinexor treatment in numerous other tumor cell lines (including HCT-116, MDA-MB-468, MV-4-11, OVCAR-8, and PC-3). Examination of lymphocytes revealed that selinexor also increased expression of PD-1 and CTLA4 by ∼2-fold. Mice bearing syngeneic B16F10 melanoma tumors treated with selinexor (15 mg/kg 2 x weekly) and anti-CTLA4 (250 μg, 2 x weekly) demonstrated a significant reduction in tumor growth rate (p = 0.0065) while monotherapy had no significant effect on tumor growth. Similar results were obtained in mice bearing B16F10 melanoma treated with the combination of selinexor + anti-PD-1 (200 μg, 2 x weekly, p < 0.034) or selinexor + anti-PD-L1 (100-200 μg, 2 x weekly, p < 0.001). Importantly, no weight loss or signs of toxicity were evident in any in vivo study. Further immunophenotypic analyses have been completed in animals receiving selinexor alone or in combination with anti-PD-L1. In combination treated mice, we observed a significantly increased percentage of splenic NK cells (p ≤ 0.050), and a significantly increased percentage of splenic Th1 T cells (p≤0.011), all compared to vehicle treated mice. Interestingly, combining selinexor with anti-PD-L1 significantly decreased the percentage of splenocytes that expressed PD-L1 (p<0.001). These changes are indicative of increased anti-tumor immune activity; however, they were accompanied by significantly increased percentages of myeloid cell subsets in combination treated mice (p ≤ 0.050). The immunologic significance of this myeloid cell expansion is currently under investigation. These data indicate that the efficacy of selinexor may be enhanced by disrupting immune checkpoints in effector cells (T and NK cells). This provides data in support of novel, evidenced-based combinations involving immunotherapy with XPO1 inhibition that deserve further investigation for advanced cancer. Citation Format: Matthew R. Farren, Reena Shakya, Rebecca Hennessey, Thomas Mace, Jennifer Yang, Omar Elnaggar, Gregory Young, Yosef Landesman, Robert Carlson, Sivan Elloul, Marsha Crochiere, Christin Burd, Gregory B. Lesinski. Selinexor, a selective inhibitor of nuclear export (SINE), enhances the in vivo efficacy of checkpoint blockade with antibodies targeting CTLA4 or PD-1/PD-L1 in melanoma. [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 2319.


Cancer Research | 2015

Abstract 2656: Combined inhibition of MEK and PI3K elicits anti-tumor activity in human cholangiocarcinoma

Jennifer Yang; Omar Elnaggar; Thomas A. Mace; Matthew R. Farren; Gregory S. Young; Patrice Lee; Kaitlin Keenan; Zheng Che; Jacob M. Kaufman; Denis C. Guttridge; David P. Carbone; Cynthia Timmers; Tanios Bekaii-Saab; Gregory B. Lesinski

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Cholangiocarcinoma (CC) responds poorly to chemo- and immunotherapy and is nearly always fatal within one year. In recent years we have gained important insight into the signaling pathways that drive this cancer. These cancers are characterized by deregulated mitogen-activated protein kinase (MAPK), PI3 kinase/Akt and a dependence on the IL-6 axis of signal transduction. Importantly, we previously reported clinical responses for CC patients receiving single agent MEK inhibitors, indicating these agents have activity in this disease. We hypothesized that dual targeting of these pathways in CC using MEK162 and buparlisib would lead to potent antitumor and immunomodulatory activity, possibly circumventing resistance to single agent MEK162. In a panel of n = 7 human CC cell lines with diverse genetic profiles, constitutive phosphorylation of ERK (7/7) and Akt (2/7) was observed. Human CC cell lines displayed variable sensitivity to the growth inhibitory and pro-apoptotic effects of single agent MEK162 or buparlisib. CC cell lines with basal AKT phosphorylation (WITT, Mz-Cha-1) showed greater sensitivity to growth inhibition by buparlisib (IC50 10-20 μM), as compared to CC lines lacking pAKT (HuCCT1, HuH28, IC50 > 20μM). Immunoblot analysis confirmed decreased phosphorylated ERK (pERK) in the HuCCT1 and SNU-478 CC cell lines following treatment with MEK162. Culture supernatants from four separate human CC cell lines displayed significant reductions in IL-6, VEGF, and GM CSF in a concentration-dependent manner after treatment with MEK162. Immunomodulatory effects of MEK162 were also evident, independent of its tumor-intrinsic effects upon CC cell lines. Namely, it significantly reduced IL 6/GM-CSF driven MDSC differentiation (HLA-DRlo CD11b+ CD33+) from healthy normal donor PBMC in vitro. These observations were not due to cytotoxic activity as treatment with MEK162 for 72 hours did not reduce viability of bulk human PBMCs. Finally, to evaluate the effect of this treatment combination, in vivo studies were conducted in athymic mice bearing Mz-Cha1 or SNU-478 xenografts. Tumor bearing mice received daily oral administration of MEK162 (30 mg/kg), buparlisib (25 mg/kg), or both agents combined. Vehicle treated animals served as negative controls. Inhibition of tumor growth was observed following administration of single agent MEK162 or buparlisib as compared to control animals. This effect was further enhanced in the combination treated animals. Body weight of animals indicated this regimen was well-tolerated. Together, these data suggest that dual PI3K and MEK inhibition can target CC with varying genotypes and represents a promising therapeutic regimen with potential for direct antitumor activity and immunomodulation in CC. Citation Format: Jennifer Yang, Omar Elnaggar, Thomas Mace, Matthew Farren, Gregory Young, Patrice Lee, Kaitlin Keenan, Zheng Che, Jacob Kaufman, Denis Guttridge, David Carbone, Cynthia Timmers, Tanios Bekaii-Saab, Gregory Lesinski. Combined inhibition of MEK and PI3K elicits anti-tumor activity in human cholangiocarcinoma. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2656. doi:10.1158/1538-7445.AM2015-2656


Cancer Research | 2015

Abstract 4695: Dual targeting of MEK and PI3K pathways can act via tumor-intrinsic mechanisms to overcome resistance and tumor-extrinsic mechanisms to modulate immunity and limit cancer cachexia

Jennifer Yang; Erin E. Talbert; Omar Elnaggar; Priyani Rajasekera; Thomas A. Mace; Matthew R. Farren; Zheng Che; Benjamin Swanson; Gregory S. Young; Ericka Haverick; Cynthia Timmers; Mark Bloomston; Tanios Bekaii-Saab; Denis C. Guttridge; Gregory B. Lesinski

Prior studies by our group have shown that single-agent MEK inhibitors have clinical activity in cholangiocarcinoma (CC), including objective responses. Yet no relationship was found between clinical benefit, and oncogenic driver mutations such as KRAS or BRAF. Strategies to expand upon MEKi have focused on combinations with agents targeting resistance, such as the PI3K/Akt pathway. CC patients treated with MEKi also had reduced pro-inflammatory cytokines and weight gain with restoration of muscle mass. These findings are clinically significant, and led us to hypothesize that MEKi act via tumor-extrinsic mechanisms to modulate immunity and limit cancer cachexia, while overcoming tumor-intrinsic resistance. We used the colon-26 adenocarcinoma model to evaluate the effect of single and combined treatment with MEK162 (30mg/kg) and buparlisib (25mg/kg) on muscle wasting, tumor growth, and immune modulation. This murine model depends on interleukin-6 (IL-6) and recapitulates the cancer cachexia syndrome. Single agent MEK162 inhibited growth initially, but after 14 days, tumor volume was comparable between MEKi and vehicle treated mice, possibly from acquired MEK162 resistance. Despite these data, reduced serum IL-6 and splenic Gr1+CD11b+ cells were evident compared to control mice (mean = 358 pg/mL to 43 pg/mL, and 13.8% to 8.4%, respectively), while body weight from MEK162 treated mice was spared (mean = 24.0g to 20.5g in control mice). In addition, markers of muscle catabolism, including the E3 ubiquitin ligases Atrogin-1 (from 1 to 0.043 in combo) and MuRF1 (from 1 to 0.03 in combo), and the autophagy gene, Bnip3, were all reduced in tibialis anterior muscles from tumor-bearing mice treated with MEK162. Similar to our clinical data, these results suggest MEK162 modulates immune biomarkers, and acts as an anti-cachexia agent. Since cross talk between MAPK and PI3K/AKT pathways promotes resistance to monotherapy, we next investigated the effects of MEK162 combined with buparlisib in the colon-26 model. As before, single agent MEK162 had a modest growth inhibitory effect, yet rescued body weight. Consistent with in vitro studies, single agent buparlisib was cytostatic, but not as effective as MEK162 in rescuing weight loss. However, dual treatment with MEK162 + buparlisib significantly inhibited tumor growth. This combination also significantly reduced splenic MDSC (mean = 37% to 9% in control mice) and increased CD4+ and CD8+ (mean = 24% to 6%, and 7.7% to 2%, respectively) T cells. These results suggest that dual inhibition of MEK and PI3K has efficacy, while MEKi may have under-appreciated tumor-extrinsic mechanisms of activity that can be leveraged to benefit patients with advanced malignancy. This treatment combination will be evaluated in CC patients in the setting of a Phase II clinical trial. Citation Format: Jennifer Yang, Erin Talbert, Omar Elnaggar, Priyani Rajasekera, Thomas Mace, Matthew Farren, Zheng Che, Benjamin Swanson, Gregory Young, Ericka Haverick, Cynthia Timmers, Mark Bloomston, Tanios Bekaii-Saab, Denis Guttridge, Gregory Lesinski. Dual targeting of MEK and PI3K pathways can act via tumor-intrinsic mechanisms to overcome resistance and tumor-extrinsic mechanisms to modulate immunity and limit cancer cachexia. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4695. doi:10.1158/1538-7445.AM2015-4695


Journal for ImmunoTherapy of Cancer | 2014

BMS-911543 inhibits viability of tumor and stromal cells and limits disease progression in genetically engineered mice with pancreatic cancer.

Thomas A. Mace; Reena Shakya; Benjamin Swanson; Thomas Ludwig; Omar Elnaggar; Hannah M. Komar; Jennifer Yang; Gregory S. Young; Wendy L. Frankel; Peter Muscarella; Tanios Bekaii-Saab; Mark Bloomston; Gregory B. Lesinski

Pancreatic cancer (PCa) is resistant to cytotoxic therapies, and the profound immune suppressive nature of this disease renders patients non-responsive to immunologic therapies. Signaling downstream of IL-6 is important in the genesis and progression of PCa. The IL-6/Jak2/STAT3 axis also mediates expansion of myeloid-derived suppressor cells (MDSC). Indeed, the Jak2/STAT3 pathway is activated in human PCa specimens and cooperates with activated Kras to drive initiation and progression of PCa in murine models. We hypothesized that the Jak2 specific inhibitor (BMS-911543; Bristol-Myers Squibb) would elicit anti-tumor activity against PCa and decrease immune suppressive features of the disease. Treatment of Kras wild type (BxPC-3) and mutant (MIA Paca-2, Panc-1) human PCa cell lines in vitro with BMS-911543 decreased viability in a concentration-dependent manner. Similar inhibitory effects of BMS-911543 were also observed upon stromal-derived pancreatic stellate cells in vitro. These stromal cells have been previously shown by our group to rely on STAT3 signaling for survival, and secrete cytokines involved in MDSC generation. BMS-911543 also had immunomodulatory effects, as it significantly reduced IL-6/GM-CSF driven MDSC differentiation (HLA-DRlo CD11b+ CD33+) from healthy normal donor PBMC in vitro. To evaluate in vivo activity of BMS-911543, we used a genetically engineered PCa model with conditional expression of mutant KrasG12D, tp53R270H, and Brca1 alleles in pancreatic cancer cells (Brca1-KPC mice). This model accurately recapitulates many histologic and systemic manifestations of PCa observed in patients, but at an accelerated rate, which is advantageous for therapeutic studies. By 5 weeks of age, Brca1-KPC mice have adenocarcinoma with 100% penetrance, pSTAT3 in the tumor and stroma, high levels of MDSC, and increased plasma IL-6. At 5-6 weeks of age, mice (n = 5/group) were imaged by bioluminescent imaging (BLI) to confirm tumor burden and were then treated with vehicle or BMS-911543 by oral gavage daily for 14 days. Histologic analysis of pancreata from treated mice showed fewer foci of adenocarcinoma when compared to vehicle controls. Analysis of immune biomarkers will focus on Jak2/STAT3 driven processes such as the percentage of MDSC, T cell subset analysis and inflammatory cytokines both systemically and in the tumor microenvironment. These results provide rationale for the design of future clinical trials that target Jak/STAT signaling in patients with PCa.


Cancer Research | 2014

Abstract 2585: Bioactive compounds or metabolites from black raspberries modulate T lymphocyte function, myeloid cell differentiation and Jak/STATsignaling

Thomas A. Mace; Samantha J. King; Zeenath Ameen; Omar Elnaggar; Gregory S. Young; Kenneth M. Riedl; Steven J. Schwartz; Steven K. Clinton; Thomas J. Knobloch; Christopher M. Weghorst; Gregory B. Lesinski

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Bioactive phyotochemicals from natural products, such as black raspberries have direct anti-cancer properties on malignant cells in culture and in xenograft models. In addition, black raspberry components inhibit cancer progression in more complex rodent carcinogenesis models. Although mechanistic targets for black raspberry phytochemicals in cancer cells are beginning to emerge, the potential role in modulating host immune processes impacting cancer have not been systematically examined. We hypothesized that black raspberries contain compounds capable of eliciting potent immunomodulatory properties that impact cellular mediators relevant to chronic inflammation and tumor progression. In the present report, we studied both an ethanol extract from black raspberries (BRB-E) containing a diverse mixture of phytochemicals and two abundant phytochemical metabolites of black raspberries produced upon ingestion (Cyanidin-3-Rutinoside, C3R; Quercitin-3-rutinoside, Q3R). BRB-E inhibited proliferation of CD3/CD28 activated human CD4+ and CD8+ T lymphocytes. BRB-E also limited the viability of CD3+ T cells cultured in IL-2, and were potent inhibitors of IL-2-induced STAT5 phosphorylation. Conversely, BRB-E significantly enhanced the monokine-induced by gamma (MIG)-mediated chemotaxis of CD8+ T lymphocytes in transwell assays. This enhanced chemotaxis was associated with upregulated expression of CXCR3 on T lymphocytes. BRB-E also inhibited the in vitro expansion of myeloid-derived suppressor cells (MDSC). Consistent with the reduction in MDSC, pre-treatment of immune cells with BRB E attenuated IL-6-mediated signal transducer and activator of transcription-3 (STAT3) phosphorylation. Finally, the C3R and Q3R metabolites were also found to be immunomodulatory, as C3R inhibited proliferation of CD3/CD28 activated T lymphocytes and STAT5 phosphorylation, while Q3R inhibited MDSC expansion and STAT3 signaling but not STAT5. Together these data indicate that black raspberries or their physiologically-relevant metabolites contain phytochemicals that affect immune processes relevant to carcinogenesis. Furthermore, specific components and their metabolites may be a source of lead compounds for drug development that exhibit targeted immunological outcomes or inhibition of specific STAT-regulated signaling pathways. Citation Format: Thomas A. Mace, Samantha King, Zeenath Ameen, Omar Elnaggar, Gregory Young, Kenneth Riedl, Steven Schwartz, Steven Clinton, Thomas Knobloch, Christopher Weghorst, Gregory B. Lesinski. Bioactive compounds or metabolites from black raspberries modulate T lymphocyte function, myeloid cell differentiation and Jak/STATsignaling. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2585. doi:10.1158/1538-7445.AM2014-2585

Collaboration


Dive into the Omar Elnaggar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge