Network


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

Hotspot


Dive into the research topics where Jennifer Brogdon is active.

Publication


Featured researches published by Jennifer Brogdon.


Journal of Experimental Medicine | 2013

Activating Fc γ receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies

Yannick Bulliard; Rose Jolicoeur; Maurice Windman; Sarah Rue; Seth Ettenberg; Deborah Knee; Nicholas Wilson; Glenn Dranoff; Jennifer Brogdon

Antibodies that coengage activating FcγRs expressed by tumor-associated leukocytes facilitate the selective elimination of intratumoral T cells.


Science Translational Medicine | 2015

Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma.

Laura A. Johnson; John Scholler; Takayuki Ohkuri; Akemi Kosaka; Prachi R. Patel; Shannon E. McGettigan; Arben Nace; Tzvete Dentchev; Pramod Thekkat; Andreas Loew; Alina C. Boesteanu; Alexandria P. Cogdill; Taylor Chen; Joseph A. Fraietta; Christopher C. Kloss; Avery D. Posey; Boris Engels; Reshma Singh; Tucker Ezell; Neeraja Idamakanti; Melissa Ramones; Na Li; Li Zhou; Gabriela Plesa; John T. Seykora; Hideho Okada; Carl H. June; Jennifer Brogdon; Marcela V. Maus

A chimeric antigen receptor redirects T cells to treat glioblastoma. CAR T cells drive glioblastoma therapy Immunotherapy with chimeric antigen receptor (CAR) T cells can successfully treat B cell malignancies, but expansion into solid tumors has been limited by the lack of availability of tumor-specific antigens. Now, Johnson et al. target CAR T cells to a variant III mutation of the epidermal growth factor receptor (EGFRvIII), which is thought to be enriched in glioblastoma stem cells. They found that a low-affinity single-chain variable fragment was specific for EGFRvIII over wild-type EGFR and that CAR T cells transduced with this fragment were able to target antigen-expressing cells in vitro and in vivo in multiple mouse xenograft models of human glioblastoma. These cells are currently being moved into the clinic in a phase 1 clinical trial. Chimeric antigen receptors (CARs) are synthetic molecules designed to redirect T cells to specific antigens. CAR-modified T cells can mediate long-term durable remissions in B cell malignancies, but expanding this platform to solid tumors requires the discovery of surface targets with limited expression in normal tissues. The variant III mutation of the epidermal growth factor receptor (EGFRvIII) results from an in-frame deletion of a portion of the extracellular domain, creating a neoepitope. We chose a vector backbone encoding a second-generation CAR based on efficacy of a murine scFv–based CAR in a xenograft model of glioblastoma. Next, we generated a panel of humanized scFvs and tested their specificity and function as soluble proteins and in the form of CAR-transduced T cells; a low-affinity scFv was selected on the basis of its specificity for EGFRvIII over wild-type EGFR. The lead candidate scFv was tested in vitro for its ability to direct CAR-transduced T cells to specifically lyse, proliferate, and secrete cytokines in response to antigen-bearing targets. We further evaluated the specificity of the lead CAR candidate in vitro against EGFR-expressing keratinocytes and in vivo in a model of mice grafted with normal human skin. EGFRvIII-directed CAR T cells were also able to control tumor growth in xenogeneic subcutaneous and orthotopic models of human EGFRvIII+ glioblastoma. On the basis of these results, we have designed a phase 1 clinical study of CAR T cells transduced with humanized scFv directed to EGFRvIII in patients with either residual or recurrent glioblastoma (NCT02209376).


Immunology and Cell Biology | 2014

OX40 engagement depletes intratumoral Tregs via activating FcγRs, leading to antitumor efficacy

Yannick Bulliard; Rose Jolicoeur; Jimin Zhang; Glenn Dranoff; Nicholas S. Wilson; Jennifer Brogdon

Antibodies targeting checkpoint inhibitors or co‐stimulatory receptors on T cells have shown significant antitumor efficacy in preclinical and clinical studies. In mouse tumor models, engagement of activating Fcγ receptor (FcγR)‐expressing immune cells was recently shown to be required for the tumoricidal activity of antibodies recognizing the tumor necrosis factor superfamily receptor (TNFR) GITR (CD357) and CTLA‐4 (CD152). In particular, activating FcγRs facilitated the selective elimination of intratumoral T‐cell populations. However, it remains unclear whether FcγRs contribute to the antitumor efficacy of other immunomodulatory antibodies. Here, we explored the mechanism of antitumor activity mediated by an agonistic antibody (clone OX86) to the co‐stimulatory TNFR OX40 (CD134). OX40 was highly expressed by intratumoral T cells, particularly those of the FoxP3+ regulatory T‐cell (Treg) lineage. OX86 administration resulted in the depletion of intratumoral regulatory T cells in an activating FcγR‐dependent manner, which correlated with tumor regression. Together with previous data from our group and others, these findings support a mechanism whereby antibodies targeting antigens highly expressed by intratumoral T cells can mediate their elimination by FcγR‐expressing immune cells, and facilitate subsequent antitumor immunity.


Science Translational Medicine | 2017

A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma

Donald M. O’Rourke; MacLean P. Nasrallah; Arati Desai; J. Joseph Melenhorst; Keith Mansfield; Jennifer J.D. Morrissette; Maria Martinez-Lage; Steven Brem; Eileen Maloney; Angela Shen; Randi Isaacs; Suyash Mohan; Gabriela Plesa; Simon F. Lacey; Jean-Marc Navenot; Zhaohui Zheng; Bruce L. Levine; Hideho Okada; Carl H. June; Jennifer Brogdon; Marcela V. Maus

A trial of autologous T cells redirected to a specific mutation in glioblastoma patients illustrates mechanisms of resistance. Speeding toward CAR T cell therapy for glioblastoma Chimeric antigen receptor (CAR) T cells have been successfully implemented for treating leukemia and are now being investigated for solid tumors. O’Rourke et al. conducted a phase 1 safety study of autologous CAR T cells targeted to EGFR variant III in glioblastoma patients. Treatment seemed to be well tolerated, which is critical because other CAR T cell products have been implicated in devastating central nervous system complications. Of the 10 patients enrolled, 7 had surgical intervention, allowing for some analysis of the tumors and T cells in patients’ brains. The results of this trial indicate that CAR T cell therapy is a viable option for treating glioblastoma. We conducted a first-in-human study of intravenous delivery of a single dose of autologous T cells redirected to the epidermal growth factor receptor variant III (EGFRvIII) mutation by a chimeric antigen receptor (CAR). We report our findings on the first 10 recurrent glioblastoma (GBM) patients treated. We found that manufacturing and infusion of CAR-modified T cell (CART)–EGFRvIII cells are feasible and safe, without evidence of off-tumor toxicity or cytokine release syndrome. One patient has had residual stable disease for over 18 months of follow-up. All patients demonstrated detectable transient expansion of CART-EGFRvIII cells in peripheral blood. Seven patients had post–CART-EGFRvIII surgical intervention, which allowed for tissue-specific analysis of CART-EGFRvIII trafficking to the tumor, phenotyping of tumor-infiltrating T cells and the tumor microenvironment in situ, and analysis of post-therapy EGFRvIII target antigen expression. Imaging findings after CART immunotherapy were complex to interpret, further reinforcing the need for pathologic sampling in infused patients. We found trafficking of CART-EGFRvIII cells to regions of active GBM, with antigen decrease in five of these seven patients. In situ evaluation of the tumor environment demonstrated increased and robust expression of inhibitory molecules and infiltration by regulatory T cells after CART-EGFRvIII infusion, compared to pre–CART-EGFRvIII infusion tumor specimens. Our initial experience with CAR T cells in recurrent GBM suggests that although intravenous infusion results in on-target activity in the brain, overcoming the adaptive changes in the local tumor microenvironment and addressing the antigen heterogeneity may improve the efficacy of EGFRvIII-directed strategies in GBM.


Nature | 2014

In vivo discovery of immunotherapy targets in the tumour microenvironment

Penghui Zhou; Donald R. Shaffer; Diana A. Alvarez Arias; Yukoh Nakazaki; Wouter Pos; Alexis J. Torres; Viviana Cremasco; Stephanie K. Dougan; Glenn S. Cowley; Kutlu G. Elpek; Jennifer Brogdon; John Lamb; Shannon J. Turley; Hidde L. Ploegh; David E. Root; J. Christopher Love; Glenn Dranoff; Nir Hacohen; Harvey Cantor; Kai W. Wucherpfennig

Recent clinical trials showed that targeting of inhibitory receptors on T cells induces durable responses in a subset of cancer patients, despite advanced disease. However, the regulatory switches controlling T-cell function in immunosuppressive tumours are not well understood. Here we show that such inhibitory mechanisms can be systematically discovered in the tumour microenvironment. We devised an in vivo pooled short hairpin RNA (shRNA) screen in which shRNAs targeting negative regulators became highly enriched in murine tumours by releasing a block on T-cell proliferation upon tumour antigen recognition. Such shRNAs were identified by deep sequencing of the shRNA cassette from T cells infiltrating tumour or control tissues. One of the target genes was Ppp2r2d, a regulatory subunit of the PP2A phosphatase family. In tumours, Ppp2r2d knockdown inhibited T-cell apoptosis and enhanced T-cell proliferation as well as cytokine production. Key regulators of immune function can therefore be discovered in relevant tissue microenvironments.


The New England Journal of Medicine | 2017

Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas

Stephen J. Schuster; Jakub Svoboda; Elise A. Chong; Sunita D. Nasta; Anthony R. Mato; Özlem Anak; Jennifer Brogdon; Iulian Pruteanu-Malinici; Vijay Bhoj; Daniel J. Landsburg; Mariusz A. Wasik; Bruce L. Levine; Simon F. Lacey; J. Joseph Melenhorst; David L. Porter; Carl H. June

Background Patients with diffuse large B‐cell lymphoma or follicular lymphoma that is refractory to or that relapses after immunochemotherapy and transplantation have a poor prognosis. High response rates have been reported with the use of T cells modified by chimeric antigen receptor (CAR) that target CD19 in B‐cell cancers, although data regarding B‐cell lymphomas are limited. Methods We used autologous T cells that express a CD19‐directed CAR (CTL019) to treat patients with diffuse large B‐cell lymphoma or follicular lymphoma that had relapsed or was refractory to previous treatments. Patients were monitored for response to treatment, toxic effects, the expansion and persistence of CTL019 cells in vivo, and immune recovery. Results A total of 28 adult patients with lymphoma received CTL019 cells, and 18 of 28 had a response (64%; 95% confidence interval [CI], 44 to 81). Complete remission occurred in 6 of 14 patients with diffuse large B‐cell lymphoma (43%; 95% CI, 18 to 71) and 10 of 14 patients with follicular lymphoma (71%; 95% CI, 42 to 92). CTL019 cells proliferated in vivo and were detectable in the blood and bone marrow of patients who had a response and patients who did not have a response. Sustained remissions were achieved, and at a median follow‐up of 28.6 months, 86% of patients with diffuse large B‐cell lymphoma who had a response (95% CI, 33 to 98) and 89% of patients with follicular lymphoma who had a response (95% CI, 43 to 98) had maintained the response. Severe cytokine‐release syndrome occurred in 5 patients (18%). Serious encephalopathy occurred in 3 patients (11%); 2 cases were self‐limiting and 1 case was fatal. All patients in complete remission by 6 months remained in remission at 7.7 to 37.9 months (median, 29.3 months) after induction, with a sustained reappearance of B cells in 8 of 16 patients and with improvement in levels of IgG in 4 of 10 patients and of IgM in 6 of 10 patients at 6 months or later and in levels of IgA in 3 of 10 patients at 18 months or later. Conclusions CTL019 cells can be effective in the treatment of relapsed or refractory diffuse large B‐cell lymphoma and follicular lymphoma. High rates of durable remission were observed, with recovery of B cells and immunoglobulins in some patients. Transient encephalopathy developed in approximately one in three patients and severe cytokine‐release syndrome developed in one in five patients. (Funded by Novartis and others; ClinicalTrials.gov number, NCT02030834.)


European Journal of Cancer | 2016

Rationale for anti-GITR cancer immunotherapy

Deborah A. Knee; Becker Hewes; Jennifer Brogdon

Over the past decade, our understanding of cancer immunotherapy has evolved from assessing peripheral responses in the blood to monitoring changes in the tumour microenvironment. Both preclinical and clinical experience has taught us that modulation of the tumour microenvironment has significant implications to generating robust antitumour immunity. Clinical benefit has been well documented to correlate with a tumour microenvironment that contains a dense infiltration of CD8+CD45RO+ T effectors and a high ratio of CD8+ T cells to FoxP3+ regulatory T cells (Tregs). In preclinical tumour models, modulation of the Glucocorticoid induced TNF receptor (GITR)/GITR ligand (GITRL) axis suggests this pathway may provide the desired biological outcome of inhibiting Treg function while activating CD8+ T effector cells. This review will focus on the scientific rationale and considerations for the therapeutic targeting of GITR for cancer immunotherapy and will discuss possible combination strategies to enhance clinical benefit.


Nature Medicine | 2018

Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia

Joseph A. Fraietta; Simon F. Lacey; Elena Orlando; Iulian Pruteanu-Malinici; Mercy Gohil; Stefan Lundh; Alina C. Boesteanu; Yan Wang; Roddy S. O’Connor; Wei-Ting Hwang; Edward Pequignot; David E Ambrose; Changfeng Zhang; Nicholas Wilcox; Felipe Bedoya; Corin Dorfmeier; Fang Chen; Lifeng Tian; Harit Parakandi; Minnal Gupta; Regina M. Young; F. Brad Johnson; Irina Kulikovskaya; Li Liu; Jun Xu; Sadik Kassim; Megan M. Davis; Bruce L. Levine; Noelle V. Frey; Don L. Siegel

Tolerance to self-antigens prevents the elimination of cancer by the immune system1,2. We used synthetic chimeric antigen receptors (CARs) to overcome immunological tolerance and mediate tumor rejection in patients with chronic lymphocytic leukemia (CLL). Remission was induced in a subset of subjects, but most did not respond. Comprehensive assessment of patient-derived CAR T cells to identify mechanisms of therapeutic success and failure has not been explored. We performed genomic, phenotypic and functional evaluations to identify determinants of response. Transcriptomic profiling revealed that CAR T cells from complete-responding patients with CLL were enriched in memory-related genes, including IL-6/STAT3 signatures, whereas T cells from nonresponders upregulated programs involved in effector differentiation, glycolysis, exhaustion and apoptosis. Sustained remission was associated with an elevated frequency of CD27+CD45RO–CD8+ T cells before CAR T cell generation, and these lymphocytes possessed memory-like characteristics. Highly functional CAR T cells from patients produced STAT3-related cytokines, and serum IL-6 correlated with CAR T cell expansion. IL-6/STAT3 blockade diminished CAR T cell proliferation. Furthermore, a mechanistically relevant population of CD27+PD-1–CD8+ CAR T cells expressing high levels of the IL-6 receptor predicts therapeutic response and is responsible for tumor control. These findings uncover new features of CAR T cell biology and underscore the potential of using pretreatment biomarkers of response to advance immunotherapies.An IL-6/STAT3 signature and memory CD8 T cell subset in preinfusion chimeric antigen receptor–expressing T cells associate with response in patients with high-risk chronic lymphocytic leukemia.


Nature Medicine | 2018

Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia

Elena Orlando; Xia Han; Catherine Tribouley; Patricia A. Wood; Rebecca J. Leary; Markus Riester; John E. Levine; Muna Qayed; Stephan A. Grupp; Michael Boyer; Barbara De Moerloose; Eneida R. Nemecek; Henrique Bittencourt; Hidefumi Hiramatsu; Jochen Buechner; Stella M. Davies; Michael R. Verneris; Kevin Nguyen; Jennifer Brogdon; Hans Bitter; Michael Morrissey; Piotr Pierog; Serafino Pantano; Jeffrey A. Engelman; Wendy Winckler

We identified genetic mutations in CD19 and loss of heterozygosity at the time of CD19– relapse to chimeric antigen receptor (CAR) therapy. The mutations are present in the vast majority of resistant tumor cells and are predicted to lead to a truncated protein with a nonfunctional or absent transmembrane domain and consequently to a loss of surface antigen. This irreversible loss of CD19 advocates for an alternative targeting or combination CAR approach.Mutations in the CD19 gene suggesting irreversible loss of its surface expression are identified in the majority of analyzed cases of CD19– relapse in two clinical trials of pediatric ALL CD19 CAR T therapy, offering considerations for the rational choice of follow-up therapies.


Journal for ImmunoTherapy of Cancer | 2014

Pre-clinical validation of a humanized anti-EGFR variant III chimeric antigen receptor and phase I trial of CART-EGFRvIII in glioblastoma

Laura A. Johnson; John Scholler; Takayuki Ohkuri; Akemi Kosaka; Prachi R. Patel; Shannon E. McGettigan; Arben Nace; Pramod Thekkat; Andreas Loew; Taylor J. Chen; Joseph A. Fraietta; Avery D. Posey; Alina C. Boesteanu; Alexandria P. Cogdill; Boris Engels; Reshma Singh; Tucker Ezell; Neeraja Idamakanti; Gabriela Plesa; John T. Seykora; Hideho Okada; Carl H. June; Jennifer Brogdon; Marcela V. Maus

Chimeric antigen receptors are synthetic molecules designed to re-direct T cells to specific surface antigens; CAR-modified T cells can mediate long-term durable remissions in B cell malignancies, but expanding this platform to solid tumors requires the discovery of novel surface targets with limited expression. The variant III mutation of the epidermal growth factor receptor (EGFR variant III) is the most common variant of the EGF receptor observed in human tumors, and results from an in-frame deletion of a portion of the extracellular domain. In glioblastoma, the EGFRvIII mutation is oncogenic, portends a poor prognosis, and is thought to be enriched in glioblastoma stem cells. However, because the neoepitope of EGFR variant III is based on a small peptide sequence, an antibody or single-chain variable fragment (scFv) directed to this epitope must be rigorously tested to confirm lack of cross-reactivity to the ubiquitously expressed normal EGFR. Having selected a candidate murine scFv directed to EGFRvIII and a vector backbone encoding a second generation CAR, we generated a panel of humanized scFvs and tested their specificity and function as soluble proteins and in the form of CAR-transduced T cells. The lead candidate scFv was tested in vitro for its ability to direct CAR-transduced T cells to kill antigen-bearing targets effectively, and proliferate and secrete cytokines specifically in response to antigen. We further evaluated the specificity of the lead candidate CAR by comparing it to a cetuximab-based CAR which does not discriminate between EGFR and EGFR variant III; the two CARs, along with negative controls, were tested in vitro against primary cells derived from a panel of normal tissues, and in vivo in immunodeficient mice grafted with normal human skin, which naturally expresses EGFR. CAR-T cells were also able to control tumor growth in xenogeneic subcutaneous and orthotopic models of human EGFR variant III+ glioblastoma. We have designed a Phase I clinical study of CAR T cells transduced with humanized scFv directed to EGFR variant III in patients with glioblastoma.

Collaboration


Dive into the Jennifer Brogdon's collaboration.

Top Co-Authors

Avatar

Carl H. June

National Marrow Donor Program

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Glass

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

John Scholler

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Simon F. Lacey

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Bruce L. Levine

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

David L. Porter

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge