Denarda Dangaj
University of Pennsylvania
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Denarda Dangaj.
Cancer Research | 2013
Denarda Dangaj; Evripidis Lanitis; Aizhi Zhao; Shree Joshi; Yi Cheng; Raphael Sandaltzopoulos; Hyun-Jeong Ra; Gwenn Danet-Desnoyers; Daniel J. Powell; Nathalie Scholler
B7-H4 (VTCN1, B7x, B7s) is a ligand for inhibitory coreceptors on T cells implicated in antigenic tolerization. B7-H4 is expressed by tumor cells and tumor-associated macrophages (TAM), but its potential contributions to tumoral immune escape and therapeutic targeting have been less studied. To interrogate B7-H4 expression on tumor cells, we analyzed fresh primary ovarian cancer cells collected from patient ascites and solid tumors, and established cell lines before and after in vivo passaging. B7-H4 expression was detected on the surface of all fresh primary human tumors and tumor xenotransplants, but not on most established cell lines, and B7-H4 was lost rapidly by tumor xenograft cells after short-term in vitro culture. These results indicated an in vivo requirement for B7-H4 induction and defined conditions for targeting studies. To generate anti-B7-H4-targeting reagents, we isolated antibodies by differential cell screening of a yeast-display single-chain fragments variable (scFv) library derived from patients with ovarian cancer. We identified anti-B7-H4 scFv that reversed in vitro inhibition of CD3-stimulated T cells by B7-H4 protein. Notably, these reagents rescued tumor antigen-specific T-cell activation, which was otherwise inhibited by coculture with antigen-loaded B7-H4+ APCs, B7-H4+ tumor cells, or B7-H4- tumor cells mixed with B7-H4+ TAMs; peritoneal administration of anti-B7-H4 scFv delayed the growth of established tumors. Together, our findings showed that cell surface expression of B7-H4 occurs only in tumors in vivo and that antibody binding of B7-H4 could restore antitumor T-cell responses. We suggest that blocking of B7-H4/B7-H4 ligand interactions may represent a feasible therapeutic strategy for ovarian cancer.
PLOS ONE | 2012
Evripidis Lanitis; Denarda Dangaj; Ian S. Hagemann; De-Gang Song; Andrew Best; Raphael Sandaltzopoulos; George Coukos; Daniel J. Powell
The breadth of HER2 expression by primary human ovarian cancers remains controversial, which questions its suitability as a universal antigen in this malignancy. To address these issues, we performed extensive HER2 expression analysis on a wide panel of primary tumors as well as established and short-term human ovarian cancer cell lines. Conventional immunohistochemical (IHC) analysis of multiple tumor sites in 50 cases of high-grade ovarian serous carcinomas revealed HER2 overexpression in 29% of evaluated sites. However, more sensitive detection methods including flow cytometry, western blot analysis and q-PCR revealed HER2 expression in all fresh tumor cells derived from primary ascites or solid tumors as well as all established and short-term cultured cancer cell lines. Cancer cells generally expressed HER2 at higher levels than that found in normal ovarian surface epithelial (OSE) cells. Accordingly, genetically-engineered human T cells expressing an HER2-specific chimeric antigen receptor (CAR) recognized and reacted against all established or primary ovarian cancer cells tested with minimal or no reactivity against normal OSE cells. In conclusion, all human ovarian cancers express immunologically-detectable levels of HER2, indicating that IHC measurement underestimates the true frequency of HER2-expressing ovarian cancers and may limit patient access to otherwise clinically meaningful HER2-targeted therapies.
PLOS ONE | 2011
Denarda Dangaj; Karen L. Abbott; Ananda Mookerjee; Aizhi Zhao; Pamela S. Kirby; Raphael Sandaltzopoulos; Daniel J. Powell; Antonin Lamazière; Don L. Siegel; Claude Wolf; Nathalie Scholler
Tumor-infiltrating macrophages respond to microenvironmental signals by developing a tumor-associated phenotype characterized by high expression of mannose receptor (MR, CD206). Antibody cross-linking of CD206 triggers anergy in dendritic cells and CD206 engagement by tumoral mucins activates an immune suppressive phenotype in tumor-associated macrophages (TAMs). Many tumor antigens are heavily glycosylated, such as tumoral mucins, and/or attached to tumor cells by mannose residue-containing glycolipids (GPI anchors), as for example mesothelin and the family of carcinoembryonic antigen (CEA). However, the binding to mannose receptor of soluble tumor antigen GPI anchors via mannose residues has not been systematically studied. To address this question, we analyzed the binding of tumor-released mesothelin to ascites-infiltrating macrophages from ovarian cancer patients. We also modeled functional interactions between macrophages and soluble mesothelin using an in vitro system of co-culture in transwells of healthy donor macrophages with human ovarian cancer cell lines. We found that soluble mesothelin bound to human macrophages and that the binding depended on the presence of GPI anchor and of mannose receptor. We next challenged the system with antibodies directed against the mannose receptor domain 4 (CDR4-MR). We isolated three novel anti-CDR4-MR human recombinant antibodies (scFv) using a yeast-display library of human scFv. Anti-CDR4-MR scFv #G11 could block mesothelin binding to macrophages and prevent tumor-induced phenotype polarization of CD206low macrophages towards TAMs. Our findings indicate that tumor-released mesothelin is linked to GPI anchor, engages macrophage mannose receptor, and contributes to macrophage polarization towards TAMs. We propose that compounds able to block tumor antigen GPI anchor/CD206 interactions, such as our novel anti-CRD4-MR scFv, could prevent tumor-induced TAM polarization and have therapeutic potential against ovarian cancer, through polarization control of tumor-infiltrating innate immune cells.
Clinical Cancer Research | 2015
Emese Zsiros; Priyanka Duttagupta; Denarda Dangaj; Hongzhe Li; Renee Frank; Thomas Garrabrant; Ian S. Hagemann; Bruce L. Levine; Carl H. June; Lin Zhang; Ena Wang; Francesco M. Marincola; Davide Bedognetti; Daniel J. Powell; Janos L. Tanyi; Michael Feldman; Lana E. Kandalaft; George Coukos
Purpose: Chemokines are implicated in T-cell trafficking. We mapped the chemokine landscape in advanced stage ovarian cancer and characterized the expression of cognate receptors in autologous dendritic cell (DC)–vaccine primed T cells in the context of cell-based immunotherapy. Experimental Design: The expression of all known human chemokines in patients with primary ovarian cancer was analyzed on two independent microarray datasets and validated on tissue microarray. Peripheral blood T cells from five HLA-A2 patients with recurrent ovarian cancer, who previously received autologous tumor DC vaccine, underwent CD3/CD28 costimulation and expansion ex vivo. Tumor-specific T cells were identified by HER2/neu pentamer staining and were evaluated for the expression and functionality of chemokine receptors important for homing to ovarian cancer. Results: The chemokine landscape of ovarian cancer is heterogeneous with high expression of known lymphocyte-recruiting chemokines (CCL2, CCL4, and CCL5) in tumors with intraepithelial T cells, whereas CXCL10, CXCL12, and CXCL16 are expressed quasi-universally, including in tumors lacking tumor-infiltrating T cells. DC-vaccine primed T cells were found to express the cognate receptors for the above chemokines. Ex vivo CD3/CD28 costimulation and expansion of vaccine-primed Tcells upregulated CXCR3 and CXCR4, and enhanced their migration toward universally expressed chemokines in ovarian cancer. Conclusions: DC-primed tumor-specific T cells are armed with the appropriate receptors to migrate toward universal ovarian cancer chemokines, and these receptors are further upregulated by ex vivo CD3/CD28 costimulation, which render T cells more fit for migrating toward these chemokines. Clin Cancer Res; 21(12); 2840–50. ©2015 AACR.
Human Gene Therapy | 2014
Evripidis Lanitis; Jenessa B. Smith; Denarda Dangaj; Seleeke Flingai; Mathilde Poussin; Shuwen Xu; Brian J. Czerniecki; Yong F. Li; Paul F. Robbins; Daniel J. Powell
The ErbB2 protein is a member of the tyrosine kinase family of growth factor receptors that is overexpressed in cancers of the breast, ovary, stomach, kidney, colon, and lung, and therefore represents an attractive candidate antigen for targeted cancer immunotherapy. Cytotoxic T lymphocytes specific for various immunogenic ErbB2 peptides have been described, but they often exhibit both poor functional avidity and tumor reactivity. In order to generate potent CD8(+) T cells with specificity for the ErbB2(369-377) peptide, we performed one round of in vitro peptide stimulation of CD8(+) T cells isolated from an HLA-A2(+) patient who was previously vaccinated with autologous dendritic cells pulsed with HLA class I ErbB2 peptides. Using this approach, we enriched highly avid ErbB2-reactive T cells with strong ErbB2-specific, antitumor effector functions. We then stimulated these ErbB2-reactive T cells with ErbB2(+) HLA-A2(+) tumor cells in vitro and sorted tumor-activated ErbB2(369-377) peptide T cells, which allowed for the isolation of a novel T-cell receptor (TCR) with ErbB2(369-377) peptide specificity. Primary human CD8(+) T cells genetically modified to express this ErbB2-specific TCR specifically bound ErbB2(369-377) peptide containing HLA-A2 tetramers, and efficiently recognized target cells pulsed with low nanomolar concentrations of ErbB2(369-377) peptide as well as nonpulsed ErbB2(+) HLA-A2(+) tumor cell lines in vitro. In a novel xenograft model, ErbB2-redirected T cells also significantly delayed progression of ErbB2(+) HLA-A2(+) human tumor in vivo. Together, these results support the notion that redirection of normal T-cell specificity by TCR gene transfer can have potential applications in the adoptive immunotherapy of ErbB2-expressing malignancies.
Science Translational Medicine | 2018
Janos L. Tanyi; Sara Bobisse; Eran Ophir; Sandra Tuyaerts; Annalisa Roberti; Raphael Genolet; Petra Baumgartner; Brian J. Stevenson; Christian Iseli; Denarda Dangaj; Brian J. Czerniecki; Aikaterini Semilietof; Julien Racle; Alexandra Michel; Ioannis Xenarios; Cheryl Lai-Lai Chiang; Dimitri Monos; Drew A. Torigian; Harvey L. Nisenbaum; Olivier Michielin; Carl H. June; Bruce L. Levine; Daniel J. Powell; David Gfeller; Rosemarie Mick; Urania Dafni; Vincent Zoete; Alexandre Harari; George Coukos; Lana E. Kandalaft
Personalized cancer vaccines induce antitumor T cells that correlate with clinical benefit in patients with ovarian cancer. The personalized touch in cancer vaccination Transfer of autologous dendritic cells (DCs) has been investigated as a method of boosting T cell responses in therapeutic vaccines for several diseases. Tanyi et al. report the findings of a clinical study involving recurrent ovarian cancer patients. Patient DCs were pulsed with oxidized tumor lysate before transfer and given alone or in combination with immunomodulatory drugs. The DC vaccine was well tolerated and induced potent antitumor T cell responses, including to new epitopes, that correlated with better prognosis. These results suggest further testing of this vaccination regimen for inducing protective T cell immunity in cancer. We conducted a pilot clinical trial testing a personalized vaccine generated by autologous dendritic cells (DCs) pulsed with oxidized autologous whole-tumor cell lysate (OCDC), which was injected intranodally in platinum-treated, immunotherapy-naïve, recurrent ovarian cancer patients. OCDC was administered alone (cohort 1, n = 5), in combination with bevacizumab (cohort 2, n = 10), or bevacizumab plus low-dose intravenous cyclophosphamide (cohort 3, n = 10) until disease progression or vaccine exhaustion. A total of 392 vaccine doses were administered without serious adverse events. Vaccination induced T cell responses to autologous tumor antigen, which were associated with significantly prolonged survival. Vaccination also amplified T cell responses against mutated neoepitopes derived from nonsynonymous somatic tumor mutations, and this included priming of T cells against previously unrecognized neoepitopes, as well as novel T cell clones of markedly higher avidity against previously recognized neoepitopes. We conclude that the use of oxidized whole-tumor lysate DC vaccine is safe and effective in eliciting a broad antitumor immunity, including private neoantigens, and warrants further clinical testing.
Molecular Therapy | 2016
Jenessa B. Smith; Evripidis Lanitis; Denarda Dangaj; Elizabeth L. Buza; Mathilde Poussin; Caitlin Stashwick; Nathalie Scholler; Daniel J. Powell
B7-H4 protein is frequently overexpressed in ovarian cancer. Here, we engineered T cells with novel B7-H4-specific chimeric antigen receptors (CARs) that recognized both human and murine B7-H4 to test the hypothesis that B7-H4 CAR T cell therapy can be applied safely in preclinical models. B7-H4 CAR T cells specifically secreted IFN-γ and lysed B7-H4(+) targets. In vivo, B7-H4 CAR T cells displayed antitumor reactivity against B7-H4(+) human ovarian tumor xenografts. Unexpectedly, B7-H4 CAR T cell treatment reproducibly showed delayed, lethal toxicity 6-8 weeks after therapy. Comprehensive assessment of murine B7-H4 protein distribution uncovered expression in ductal and mucosal epithelial cells in normal tissues. Postmortem analysis revealed the presence of widespread histologic lesions that correlated with B7-H4(+) expression, and were inconsistent with graft versus host disease. Lastly, expression patterns of B7-H4 protein in normal human tissue were comparable to distribution in mice, advancing our understanding of B7-H4. We conclude that B7-H4 CAR therapy mediates control of cancer outgrowth. However, long-term engraftment of B7-H4 CAR T cells mediates lethal, off-tumor toxicity that is likely due to wide expression of B7-H4 in healthy mouse organs. This model system provides a unique opportunity for preclinical evaluation of safety approaches that limit CAR-mediated toxicity after tumor destruction in vivo.
BioMed Research International | 2015
Athanasios Karapetsas; Antonis Giannakakis; Denarda Dangaj; Evripidis Lanitis; Spyridon Kynigopoulos; Maria Lambropoulou; Janos L. Tanyi; Alex Galanis; Stylianos Kakolyris; Gregorios Trypsianis; George Coukos; Raphael Sandaltzopoulos
Infiltration of cytotoxic T-lymphocytes in ovarian cancer is a favorable prognostic factor. Employing a differential expression approach, we have recently identified a number of genes associated with CD8+ T-cell infiltration in early stage ovarian tumors. In the present study, we validated by qPCR the expression of two genes encoding the transmembrane proteins GPC6 and TMEM132D in a cohort of early stage ovarian cancer patients. The expression of both genes correlated positively with the mRNA levels of CD8A, a marker of T-lymphocyte infiltration [Pearson coefficient: 0.427 (p = 0.0067) and 0.861 (p < 0.0001), resp.]. GPC6 and TMEM132D expression was also documented in a variety of ovarian cancer cell lines. Importantly, Kaplan-Meier survival analysis revealed that high mRNA levels of GPC6 and/or TMEM132D correlated significantly with increased overall survival of early stage ovarian cancer patients (p = 0.032). Thus, GPC6 and TMEM132D may serve as predictors of CD8+ T-lymphocyte infiltration and as favorable prognostic markers in early stage ovarian cancer with important consequences for diagnosis, prognosis, and tumor immunobattling.
OncoImmunology | 2016
Emese Zsiros; Denarda Dangaj; Carl H. June; Lana E. Kandalaft; George Coukos
ABSTRACT Tumor barriers preventing T-cell homing and engraftment should be neutralized during cancer immunotherapy. We recently discovered that ovarian cancer expresses quasi-universal chemokines that can support T-cell homing. Furthermore, T cells elicited by whole tumor antigen dendritic-cell vaccines express cognate chemokine receptors which are upregulated by CD3/CD28 costimulation.
Molecular Therapy | 2015
Jenessa B. Smith; Evripidis Lanitis; Denarda Dangaj; Mathilde Poussin; Michael McLane; Nathalie Scholler; Daniel J. Powell
B7-H4 is a negative immune regulatory molecule frequently overexpressed on the surface of cancer cells and immunosuppressive tumor-associated macrophages (TAMs). Ligation of B7-H4 with an unknown receptor on activated T cells results in inhibited effector function via cell cycle arrest, decreased proliferation and reduced IL-2 production. B7-H4 expression levels in ovarian cancer inversely correlate with patient survival, making B7-H4 an attractive candidate for therapeutic intervention. However, trials of B7-H4 targeted therapy have not been conducted and the biodistribution of B7-H4 protein in mice or humans is not well understood.Here, we tested the hypothesis that safe and effective B7-H4 targeting can be achieved using T cells genetically redirected with chimeric antigen receptors (CARs) against B7-H4 in a preclinical model of human ovarian cancer. Four independent anti-B7-H4 CAR lentivirus constructs were generated using anti-B7H4 single chain variable fragments (scFv) with affinities ranging from low to high, which were transduced into primary human T cells. Transduced T cells efficiently expressed CARs on their cell surface, and each CAR was capable of binding both human and mouse recombinant B7-H4 protein with distinct binding patterns; two CARs (3#68 and 3#54) had high capacity for rhB7-H4 protein binding, another had low binding capacity (56), and the last had virtually none (26). In co-culture assays, high binding B7-H4 CAR T cells secreted Th1 cytokines and specifically lysed human B7-H4+ ovarian cancer cells. In an immunodeficient mouse model, transferred B7-H4 CAR T cells persisted after infusion and exerted efficient anti-tumor activity against subcutaneous B7-H4+ human ovarian tumor xenografts.Unexpectedly, B7-H4 CAR T cell treated mice reproducibly showed signs of delayed onset, lethal toxicity emerging 6-8 weeks after therapy. Postmortem analysis revealed widespread immunopathology in nearly all tissue analyzed, with marked infiltration and destruction of peripheral nerve bundles and perivasculature regions that was inconsistent with non-specific graft versus host disease (GVHD). Immunopathology was evident in both tumor-bearing and healthy mice administered B7-H-4 CAR T cells, and mice administered control CAR T cells experienced tumor regression but without concomitant toxicity, ruling out a generalizable GVHD effect.Our results indicate that CAR T cell-based targeting of B7-H4 can mediate control of B7-H4+ cancer outgrowth in vivo, however, long-term engraftment of B7-H4 CAR T cells mediates lethal on-target, off-tumor toxicity that is likely due to wide expression of B7-H4 in healthy mouse organs. Accordingly, B7-H4 CAR T cells represent an important preclinical model system for the evaluation of safety approaches that limit CAR-mediated toxicity whilst maintaining potent anti-tumor activity in vivo.