Network


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

Hotspot


Dive into the research topics where Avery D. Posey is active.

Publication


Featured researches published by Avery D. Posey.


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).


Blood | 2014

ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells

Sonia Guedan; Xi Chen; Aviv Madar; Carmine Carpenito; Shannon E. McGettigan; Matthew J. Frigault; Jihyun Lee; Avery D. Posey; John Scholler; Nathalie Scholler; Richard Bonneau; Carl H. June

With the notable exception of B-cell malignancies, the efficacy of chimeric antigen receptor (CAR) T cells has been limited, and CAR T cells have not been shown to expand and persist in patients with nonlymphoid tumors. Here we demonstrate that redirection of primary human T cells with a CAR containing the inducible costimulator (ICOS) intracellular domain generates tumor-specific IL-17-producing effector cells that show enhanced persistence. Compared with CARs containing the CD3ζ chain alone, or in tandem with the CD28 or the 4-1BB intracellular domains, ICOS signaling increased IL-17A, IL-17F, and IL-22 following antigen recognition. In addition, T cells redirected with an ICOS-based CAR maintained a core molecular signature characteristic of TH17 cells and expressed higher levels of RORC, CD161, IL1R-1, and NCS1. Of note, ICOS signaling also induced the expression of IFN-γ and T-bet, consistent with a TH17/TH1 bipolarization. When transferred into mice with established tumors, TH17 cells that were redirected with ICOS-based CARs mediated efficient antitumor responses and showed enhanced persistence compared with CD28- or 4-1BB-based CAR T cells. Thus, redirection of TH17 cells with a CAR encoding the ICOS intracellular domain is a promising approach to augment the function and persistence of CAR T cells in hematologic malignancies.


Cancer immunology research | 2015

Identification of chimeric antigen receptors that mediate constitutive or inducible proliferation of T cells.

Matthew J. Frigault; Jihyun Lee; Maria Ciocca Basil; Carmine Carpenito; Shinichiro Motohashi; John Scholler; Omkar U. Kawalekar; Sonia Guedan; Shannon E. McGettigan; Avery D. Posey; Sonny Ang; Laurence J.N. Cooper; Jesse Platt; F. Brad Johnson; Chrystal M. Paulos; Yangbing Zhao; Michael Kalos; Michael C. Milone; Carl H. June

Frigault, Lee, and colleagues compared chimeric antigen receptors (CAR) encoding signaling domains comprising CD28, ICOS, and 4-1BB and found that some CD28 CAR-T cells have antigen-independent constitutive proliferation and cytokine secretion when highly expressed, leading to inferior antitumor effects. This study compared second-generation chimeric antigen receptors (CAR) encoding signaling domains composed of CD28, ICOS, and 4-1BB (TNFRSF9). Here, we report that certain CARs endow T cells with the ability to undergo long-term autonomous proliferation. Transduction of primary human T cells with lentiviral vectors encoding some of the CARs resulted in sustained proliferation for up to 3 months following a single stimulation through the T-cell receptor (TCR). Sustained numeric expansion was independent of cognate antigen and did not require the addition of exogenous cytokines or feeder cells after a single stimulation of the TCR and CD28. Results from gene array and functional assays linked sustained cytokine secretion and expression of T-bet (TBX21), EOMES, and GATA-3 to the effect. Sustained expression of the endogenous IL2 locus has not been reported in primary T cells. Sustained proliferation was dependent on CAR structure and high expression, the latter of which was necessary but not sufficient. The mechanism involves constitutive signaling through NF-κB, AKT, ERK, and NFAT. The propagated CAR T cells retained a diverse TCR repertoire, and cellular transformation was not observed. The CARs with a constitutive growth phenotype displayed inferior antitumor effects and engraftment in vivo. Therefore, the design of CARs that have a nonconstitutive growth phenotype may be a strategy to improve efficacy and engraftment of CAR T cells. The identification of CARs that confer constitutive or nonconstitutive growth patterns may explain observations that CAR T cells have differential survival patterns in clinical trials. Cancer Immunol Res; 3(4); 356–67. ©2015 AACR.


Current protocols in immunology | 2015

Measurement of Intracellular Ions by Flow Cytometry

Avery D. Posey; Omkar U. Kawalekar; Carl H. June

Using flow cytometry, single‐cell measurements of calcium can be made on isolated populations identified by one or more phenotypic characteristics. Most earlier techniques for measuring cellular activation parameters determined the mean value for a population of cells, which did not permit optimal resolution of the responses. The flow cytometer is particularly useful for this purpose because it can measure ion concentrations in large numbers of single cells and thereby allows ion concentration to be correlated with other parameters such as immunophenotype and cell cycle stage. A limitation of flow cytometry, however, is that it does not permit resolution of certain complex kinetic responses such as cellular oscillatory responses. This unit describes the preparation of cells, including labeling with antibodies and with calcium probes, and discusses the principles of data analysis and interpretation.


Cancer immunology research | 2017

Safety and Efficacy of Intratumoral Injections of Chimeric Antigen Receptor (CAR) T Cells in Metastatic Breast Cancer

Julia Tchou; Yangbing Zhao; Bruce L. Levine; Paul J. Zhang; Megan M. Davis; J. Joseph Melenhorst; Irina Kulikovskaya; Andrea L. Brennan; Xiaojun Liu; Simon F. Lacey; Avery D. Posey; Austin D. Williams; Alycia So; Jose R. Conejo-Garcia; Gabriela Plesa; Regina M. Young; Shannon E. McGettigan; Jean Campbell; Robert H. Pierce; J Matro; Angela DeMichele; Amy S. Clark; Laurence J.N. Cooper; Lynn M. Schuchter; Robert H. Vonderheide; Carl H. June

Transiently expressed chimeric antigen receptor T cells specific for c-Met, expressed in breast cancer, were injected into breast cancer tumors of six patients in a phase 0 clinical trial. Injections resulted in tumor necrosis and were well tolerated. Chimeric antigen receptors (CAR) are synthetic molecules that provide new specificities to T cells. Although successful in treatment of hematologic malignancies, CAR T cells are ineffective for solid tumors to date. We found that the cell-surface molecule c-Met was expressed in ∼50% of breast tumors, prompting the construction of a CAR T cell specific for c-Met, which halted tumor growth in immune-incompetent mice with tumor xenografts. We then evaluated the safety and feasibility of treating metastatic breast cancer with intratumoral administration of mRNA-transfected c-Met-CAR T cells in a phase 0 clinical trial (NCT01837602). Introducing the CAR construct via mRNA ensured safety by limiting the nontumor cell effects (on-target/off-tumor) of targeting c-Met. Patients with metastatic breast cancer with accessible cutaneous or lymph node metastases received a single intratumoral injection of 3 × 107 or 3 × 108 cells. CAR T mRNA was detectable in peripheral blood and in the injected tumor tissues after intratumoral injection in 2 and 4 patients, respectively. mRNA c-Met-CAR T cell injections were well tolerated, as none of the patients had study drug–related adverse effects greater than grade 1. Tumors treated with intratumoral injected mRNA c-Met-CAR T cells were excised and analyzed by immunohistochemistry, revealing extensive tumor necrosis at the injection site, cellular debris, loss of c-Met immunoreactivity, all surrounded by macrophages at the leading edges and within necrotic zones. We conclude that intratumoral injections of mRNA c-Met-CAR T cells are well tolerated and evoke an inflammatory response within tumors. Cancer Immunol Res; 5(12); 1152–61. ©2017 AACR.


Cancer immunology research | 2018

Improving CART-Cell Therapy of Solid Tumors with Oncolytic Virus–Driven Production of a Bispecific T-cell Engager

Anna Wing; Carlos Alberto Fajardo; Avery D. Posey; Carolyn E. Shaw; Tong Da; Regina M. Young; Ramon Alemany; Carl H. June; Sonia Guedan

The efficacy of chimeric antigen receptor T-cell therapy was improved by oncolytic-adenovirus delivery of a bispecific T-cell engager to the tumor microenvironment. T-cell activation was increased and tumor resistance overcome as a result of its dual antigen specificity. T cells expressing chimeric antigen receptors (CART) have shown significant promise in clinical trials to treat hematologic malignancies, but their efficacy in solid tumors has been limited. Oncolytic viruses have the potential to act in synergy with immunotherapies due to their immunogenic oncolytic properties and the opportunity of incorporating therapeutic transgenes in their genomes. Here, we hypothesized that an oncolytic adenovirus armed with an EGFR-targeting, bispecific T-cell engager (OAd-BiTE) would improve the outcome of CART-cell therapy in solid tumors. We report that CART cells targeting the folate receptor alpha (FR-α) successfully infiltrated preestablished xenograft tumors but failed to induce complete responses, presumably due to the presence of antigen-negative cancer cells. We demonstrated that OAd-BiTE–mediated oncolysis significantly improved CART-cell activation and proliferation, while increasing cytokine production and cytotoxicity, and showed an in vitro favorable safety profile compared with EGFR-targeting CARTs. BiTEs secreted from infected cells redirected CART cells toward EGFR in the absence of FR-α, thereby addressing tumor heterogeneity. BiTE secretion also redirected CAR-negative, nonspecific T cells found in CART-cell preparations toward tumor cells. The combinatorial approach improved antitumor efficacy and prolonged survival in mouse models of cancer when compared with the monotherapies, and this was the result of an increased BiTE-mediated T-cell activation in tumors. Overall, these results demonstrated that the combination of a BiTE-expressing oncolytic virus with adoptive CART-cell therapy overcomes key limitations of CART cells and BiTEs as monotherapies in solid tumors and encourage its further evaluation in human trials. Cancer Immunol Res; 6(5); 605–16. ©2018 AACR.


Glycobiology | 2018

Glycan-directed CAR-T cells

Catharina Steentoft; Denis Migliorini; Tiffany R King; Ulla Mandel; Carl H. June; Avery D. Posey

Cancer immunotherapy is rapidly advancing in the treatment of a variety of hematopoietic cancers, including pediatric acute lymphoblastic leukemia and diffuse large B cell lymphoma, with chimeric antigen receptor (CAR)-T cells. CARs are genetically encoded artificial T cell receptors that combine the antigen specificity of an antibody with the machinery of T cell activation. However, implementation of CAR technology in the treatment of solid tumors has been progressing much slower. Solid tumors are characterized by a number of challenges that need to be overcome, including cellular heterogeneity, immunosuppressive tumor microenvironment (TME), and, in particular, few known cancer-specific targets. Post-translational modifications that differentially occur in malignant cells generate valid cell surface, cancer-specific targets for CAR-T cells. We previously demonstrated that CAR-T cells targeting an aberrant O-glycosylation of MUC1, a common cancer marker associated with changes in cell adhesion, tumor growth and poor prognosis, could control malignant growth in mouse models. Here, we discuss the field of glycan-directed CAR-T cells and review the different classes of antibodies specific for glycan-targeting, including the generation of high affinity O-glycopeptide antibodies. Finally, we discuss historic and recently investigated glycan targets for CAR-T cells and provide our perspective on how targeting the tumor glycoproteome and/or glycome will improve CAR-T immunotherapy.


Clinical Cancer Research | 2017

CAR T-Cell Therapies in Glioblastoma: A First Look

Denis Migliorini; Pierre-Yves Dietrich; Roger Stupp; Gerald P. Linette; Avery D. Posey; Carl H. June

Glioblastoma is an aggressive malignancy with a poor prognosis. The current standard of care for newly diagnosed glioblastoma patients includes surgery to the extent, temozolomide combined with radiotherapy, and alternating electric fields therapy. After recurrence, there is no standard therapy and survival is less than 9 months. Recurrent glioblastoma offers a unique opportunity to investigate new treatment approaches in a malignancy known for remarkable genetic heterogeneity, an immunosuppressive microenvironment, and a partially permissive anatomic blood–brain barrier. Results from three first-in-man chimeric antigen receptor (CAR) T-cell trials targeting IL13Rα2, Her2/CMV, and EGFRvIII have recently been reported. Each one of these trials addresses important questions, such as T-cell trafficking to CNS, engraftment and persistence, tumor microenvironment remodeling, and monitoring of glioma response to CAR T cells. Objective radiologic responses have been reported. Here, we discuss and summarize the results of these trials and suggest opportunities for the field. Clin Cancer Res; 24(3); 535–40. ©2017 AACR.


Gene Therapy | 2018

Driving cars to the clinic for solid tumors

Mauro Castellarin; Keisuke Watanabe; Carl H. June; Christopher C. Kloss; Avery D. Posey

FDA approval of chimeric antigen receptor T cells (CART cells) is the culmination of several decades of technology development and interrogation of the properties of these gene therapies. CART cells exist as personalized “living drugs” and have demonstrated astounding anti-tumor efficacy in patients with leukemia and lymphoma. However, the future promise of CART efficacy for solid tumors, the greatest unmet burden, is met with a number of challenges that must be surmounted for effective immune responses. In this review, we discuss the next-generation developments of CARs to target solid tumors, including fine-tuned and combinational-targeting receptors. We consider the structural intricacies of the CAR molecules that influence optimal signaling and CART survival, and review pre-clinical cell-intrinsic and cell-extrinsic combinational therapy approaches.


Journal for ImmunoTherapy of Cancer | 2014

Enhancing T cell persistence of CAR-redirected T cells in solid tumors

Sonia Guedan; Shannon E. McGettigan; Avery D. Posey; Jihyun Lee; Omkar U. Kawalekar; Prachi R. Patel; Brian Keith; Carl H. June

T cell persistence is likely to promote long-term anti-tumor effects after adoptive T cell transfer. We have recently shown that incorporation of the ICOS intracellular domain into chimeric antigen receptors (CARs) significantly increased Th17 cell persistence in vivo, compared to CARs with CD28 or 4-1BB intracellular domains [1]. Here, we hypothesized that CD4+ and CD8+ T cells require distinct cytokine and costimulation signals for optimal persistence. To test this hypothesis, we compared the in vivo antitumor effects and persistence of combined CD4+ T cells (bulk or Th17-polarized) and CD8+ T cells redirected with CARs containing CD28, 4-1BB or ICOS-based costimulatory domains. Using multiple mouse tumor models, we demonstrate that the ICOS intracellular domain significantly enhanced the in vivo persistence of CAR-expressing CD4+ T cells, and that both persistence and tumor infiltration were further enhanced by culturing these cells under Th17-polarizing conditions. Importantly, Th17-polarized CD4+ T cells expressing an ICOS-based CAR significantly increased the circulatory persistence of bulk CD8+ T cells expressing either CD28- or 4-1BB-based CARs. We further demonstrate that the antitumor effect of CAR-expressing CD8+ T cells was enhanced when co-injected with ICOS-redirected Th17 cells. Collectively, our data suggest that combining Th17 CD4+ T cells redirected with an ICOS-based CAR with CD8+ CAR-T cells will enhance their persistence and antitumor efficacy.

Collaboration


Dive into the Avery D. Posey's collaboration.

Top Co-Authors

Avatar

Carl H. June

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

Laura A. Johnson

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sonia Guedan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jihyun Lee

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Prachi R. Patel

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge