Network


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

Hotspot


Dive into the research topics where Gwendolyn Binder-Scholl is active.

Publication


Featured researches published by Gwendolyn Binder-Scholl.


Blood | 2013

Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma

Gerald P. Linette; Edward A. Stadtmauer; Marcela V. Maus; Aaron P. Rapoport; Bruce L. Levine; Lyndsey Emery; Leslie A. Litzky; Adam Bagg; Beatriz M. Carreno; Patrick J. Cimino; Gwendolyn Binder-Scholl; Dominic P. Smethurst; Andrew B. Gerry; Nick Pumphrey; Alan D. Bennett; Joanna E. Brewer; Joseph Dukes; Jane Harper; Helen K. Tayton-Martin; Bent K. Jakobsen; Namir J. Hassan; Michael Kalos; Carl H. June

An obstacle to cancer immunotherapy has been that the affinity of T-cell receptors (TCRs) for antigens expressed in tumors is generally low. We initiated clinical testing of engineered T cells expressing an affinity-enhanced TCR against HLA-A*01-restricted MAGE-A3. Open-label protocols to test the TCRs for patients with myeloma and melanoma were initiated. The first two treated patients developed cardiogenic shock and died within a few days of T-cell infusion, events not predicted by preclinical studies of the high-affinity TCRs. Gross findings at autopsy revealed severe myocardial damage, and histopathological analysis revealed T-cell infiltration. No MAGE-A3 expression was detected in heart autopsy tissues. Robust proliferation of the engineered T cells in vivo was documented in both patients. A beating cardiomyocyte culture generated from induced pluripotent stem cells triggered T-cell killing, which was due to recognition of an unrelated peptide derived from the striated muscle-specific protein titin. These patients demonstrate that TCR-engineered T cells can have serious and not readily predictable off-target and organ-specific toxicities and highlight the need for improved methods to define the specificity of engineered TCRs.


Science Translational Medicine | 2012

Decade-Long Safety and Function of Retroviral-Modified Chimeric Antigen Receptor T Cells

John Scholler; Troy Brady; Gwendolyn Binder-Scholl; Wei-Ting Hwang; Gabriela Plesa; Kristen Hege; Ashley N. Vogel; Michael Kalos; James L. Riley; Steven G. Deeks; Ronald T. Mitsuyasu; Wendy B. Bernstein; Naomi Aronson; Bruce L. Levine; Frederic D. Bushman; Carl H. June

Adoptively transferred chimeric antigen receptor T cells have stable stem cell–like persistence for at least a decade and more than 500 years of patient safety. Standing the Test of Time Retroviral vectors were once the mainstay of gene transfer because they could stably integrate into the host genome. However, some patients in early trials developed leukemia because of insertional mutagenesis. Now, Scholler et al. report that retroviral vector–mediated gene transfer in T cells may not have the same safety concerns, and that these cells may persist over a decade in patients. The authors followed patients from three clinical trials who received T cells transduced with gammaretroviruses carrying a chimeric antigen receptor. They found that these cells were present in recipients over a decade after infusion at levels higher than those induced by standard vaccines. These cells were still functional, had stable levels of engraftment, and did not require host immunosuppression before transplant. Moreover, the authors found no evidence of integration-induced immortalization, with no observable enrichment of integration sites near genes involved in growth control or transformation. Thus, the safety of retroviral vectors may be cell type–specific, opening up engineered T cells as a delivery platform for therapeutics. The success of adoptive T cell gene transfer for treatment of cancer and HIV is predicated on generating a response that is both durable and safe. We report long-term results from three clinical trials to evaluate gammaretroviral vector–engineered T cells for HIV. The vector encoded a chimeric antigen receptor (CAR) composed of CD4 linked to the CD3ζ signaling chain (CD4ζ). CAR T cells were detected in 98% of samples tested for at least 11 years after infusion at frequencies that exceeded average T cell levels after most vaccine approaches. The CD4ζ transgene retained expression and function. There was no evidence of vector-induced immortalization of cells; integration site distributions showed no evidence of persistent clonal expansion or enrichment for integration sites near genes implicated in growth control or transformation. The CD4ζ T cells had stable levels of engraftment, with decay half-lives that exceeded 16 years, in marked contrast to previous trials testing engineered T cells. These findings indicate that host immunosuppression before T cell transfer is not required to achieve long-term persistence of gene-modified T cells. Further, our results emphasize the safety of T cells modified by retroviral gene transfer in clinical application, as measured in >500 patient-years of follow-up. Thus, previous safety issues with integrating viral vectors are hematopoietic stem cell or transgene intrinsic, and not a general feature of retroviral vectors. Engineered T cells are a promising form of synthetic biology for long-term delivery of protein-based therapeutics. These results provide a framework to guide the therapy of a wide spectrum of human diseases.


Science Translational Medicine | 2013

Identification of a Titin-Derived HLA-A1–Presented Peptide as a Cross-Reactive Target for Engineered MAGE A3–Directed T Cells

Brian J. Cameron; Andrew B. Gerry; Joseph Dukes; Jane Harper; Vivekanandan Kannan; Frayne Bianchi; Francis Grand; Joanna E. Brewer; Minnal Gupta; Gabriela Plesa; Giovanna Bossi; Annelise Vuidepot; Alex Powlesland; Alison Legg; Katherine J. Adams; Alan D. Bennett; Nicholas J. Pumphrey; Daniel Williams; Gwendolyn Binder-Scholl; Irina Kulikovskaya; Bruce L. Levine; James L. Riley; Angel Varela-Rohena; Edward A. Stadtmauer; Aaron P. Rapoport; Gerald P. Linette; Carl H. June; Namir J. Hassan; Michael Kalos; Bent K. Jakobsen

T cells engineered to express affinity-enhanced TCRs directed to a MAGE A3 peptide cross-react with a similar, but unrelated, self-peptide. Cross-Reactive Adoptive Therapy Engineering T cells with enhanced affinity to cancer targets is a promising therapy. However, one key bottleneck in this strategy is the identification of targets that are expressed on cancer cells but not on normal healthy tissue. One way to identify these antigens is by looking at the family of cancer-testis antigens, which have restricted expression in normal tissue but are frequently up-regulated in tumors. Cameron et al. now report that a T cell engineered to target one such antigen—MAGE A3—cross-reacts with a peptide from a muscle protein, Titin. The authors developed a T cell that targeted a MAGE A3 antigen for use in adoptive immunotherapy. Although extensive preclinical investigations demonstrated no off-target antigen recognition, patients who received these T cells had serious adverse events, including fatal cardiac toxicity. The authors then used amino acid scanning to search for potential cross-reactivity of these T cells with an off-target peptide and identified a peptide derived from the muscle protein Titin. Because affinity-enhanced T cells are highly potent, this cross-reactivity was likely the cause of the off-target toxicity. This study highlights methods that may be used to prevent cross-reactivity in future trials of adoptive immunotherapy. MAGE A3, which belongs to the family of cancer-testis antigens, is an attractive target for adoptive therapy given its reactivation in various tumors and limited expression in normal tissues. We developed an affinity-enhanced T cell receptor (TCR) directed to a human leukocyte antigen (HLA)–A*01–restricted MAGE A3 antigen (EVDPIGHLY) for use in adoptive therapy. Extensive preclinical investigations revealed no off-target antigen recognition concerns; nonetheless, administration to patients of T cells expressing the affinity-enhanced MAGE A3 TCR resulted in a serious adverse event (SAE) and fatal toxicity against cardiac tissue. We present a description of the preclinical in vitro functional analysis of the MAGE A3 TCR, which failed to reveal any evidence of off-target activity, and a full analysis of the post-SAE in vitro investigations, which reveal cross-recognition of an off-target peptide. Using an amino acid scanning approach, a peptide from the muscle protein Titin (ESDPIVAQY) was identified as an alternative target for the MAGE A3 TCR and the most likely cause of in vivo toxicity. These results demonstrate that affinity-enhanced TCRs have considerable effector functions in vivo and highlight the potential safety concerns for TCR-engineered T cells. Strategies such as peptide scanning and the use of more complex cell cultures are recommended in preclinical studies to mitigate the risk of off-target toxicity in future clinical investigations.


Nature Medicine | 2015

NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma

Aaron P. Rapoport; Edward A. Stadtmauer; Gwendolyn Binder-Scholl; Olga Goloubeva; Dan T. Vogl; Simon F. Lacey; Ashraf Badros; Alfred L. Garfall; Brendan M. Weiss; Jeffrey Finklestein; Irina Kulikovskaya; Sanjoy K. Sinha; Shari Kronsberg; Minnal Gupta; Sarah Bond; Luca Melchiori; Joanna E. Brewer; Alan D. Bennett; Andrew B. Gerry; Nicholas J. Pumphrey; Daniel Williams; Helen K. Tayton-Martin; Lilliam Ribeiro; Tom Holdich; Saul Yanovich; Nancy M. Hardy; Jean Yared; Naseem Kerr; Sunita Philip; Sandra Westphal

Despite recent therapeutic advances, multiple myeloma (MM) remains largely incurable. Here we report results of a phase I/II trial to evaluate the safety and activity of autologous T cells engineered to express an affinity-enhanced T cell receptor (TCR) recognizing a naturally processed peptide shared by the cancer-testis antigens NY-ESO-1 and LAGE-1. Twenty patients with antigen-positive MM received an average 2.4 × 109 engineered T cells 2 d after autologous stem cell transplant. Infusions were well tolerated without clinically apparent cytokine-release syndrome, despite high IL-6 levels. Engineered T cells expanded, persisted, trafficked to marrow and exhibited a cytotoxic phenotype. Persistence of engineered T cells in blood was inversely associated with NY-ESO-1 levels in the marrow. Disease progression was associated with loss of T cell persistence or antigen escape, in accordance with the expected mechanism of action of the transferred T cells. Encouraging clinical responses were observed in 16 of 20 patients (80%) with advanced disease, with a median progression-free survival of 19.1 months. NY-ESO-1–LAGE-1 TCR–engineered T cells were safe, trafficked to marrow and showed extended persistence that correlated with clinical activity against antigen-positive myeloma.


Human Gene Therapy | 2013

Efficient clinical scale gene modification via zinc finger nuclease-targeted disruption of the HIV co-receptor CCR5

Dawn A. Maier; Andrea L. Brennan; Shuguang Jiang; Gwendolyn Binder-Scholl; Gary Lee; Gabriela Plesa; Zhaohui Zheng; Julio Cotte; Carmine Carpenito; Travis Wood; S. Kaye Spratt; Dale Ando; Philip D. Gregory; Michael C. Holmes; Elena E. Perez; James L. Riley; Richard G. Carroll; Carl H. June; Bruce L. Levine

Since HIV requires CD4 and a co-receptor, most commonly C-C chemokine receptor 5 (CCR5), for cellular entry, targeting CCR5 expression is an attractive approach for therapy of HIV infection. Treatment of CD4(+) T cells with zinc-finger protein nucleases (ZFNs) specifically disrupting chemokine receptor CCR5 coding sequences induces resistance to HIV infection in vitro and in vivo. A chimeric Ad5/F35 adenoviral vector encoding CCR5-ZFNs permitted efficient delivery and transient expression following anti-CD3/anti-CD28 costimulation of T lymphocytes. We present data showing CD3/CD28 costimulation substantially improved transduction efficiency over reported methods for Ad5/F35 transduction of T lymphocytes. Modifications to the laboratory scale process, incorporating clinically compatible reagents and methods, resulted in a robust ex vivo manufacturing process capable of generating >10(10) CCR5 gene-edited CD4+ T cells from healthy and HIV+ donors. CD4+ T-cell phenotype, cytokine production, and repertoire were comparable between ZFN-modified and control cells. Following consultation with regulatory authorities, we conducted in vivo toxicity studies that showed no detectable ZFN-specific toxicity or T-cell transformation. Based on these findings, we initiated a clinical trial testing the safety and feasibility of CCR5 gene-edited CD4+ T-cell transfer in study subjects with HIV-1 infection.


Blood | 2013

Antiviral effects of autologous CD4 T cells genetically modified with a conditionally replicating lentiviral vector expressing long antisense to HIV.

Pablo Tebas; David Stein; Gwendolyn Binder-Scholl; Rithun Mukherjee; Troy Brady; Tessio Rebello; Laurent Humeau; Michael Kalos; Emmanouil Papasavvas; Luis J. Montaner; Daniel Schullery; Farida Shaheen; Andrea L. Brennan; Zhaohui Zheng; Julio Cotte; Vladimir Slepushkin; Elizabeth Veloso; Adonna Mackley; Wei-Ting Hwang; Faten Aberra; Jenny Zhan; Jean D. Boyer; Ronald G. Collman; Frederic D. Bushman; Bruce L. Levine; Carl H. June

We report the safety and tolerability of 87 infusions of lentiviral vector–modified autologous CD4 T cells (VRX496-T; trade name, Lexgenleucel-T) in 17 HIV patients with well-controlled viremia. Antiviral effects were studied during analytic treatment interruption in a subset of 13 patients. VRX496-T was associated with a decrease in viral load set points in 6 of 8 subjects (P = .08). In addition, A → G transitions were enriched in HIV sequences after infusion, which is consistent with a model in which transduced CD4 T cells exert antisense-mediated genetic pressure on HIV during infection. Engraftment of vector-modified CD4 T cells was measured in gut-associated lymphoid tissue and was correlated with engraftment in blood. The engraftment half-life in the blood was approximately 5 weeks, with stable persistence in some patients for up to 5 years. Conditional replication of VRX496 was detected periodically through 1 year after infusion. No evidence of clonal selection of lentiviral vector–transduced T cells or integration enrichment near oncogenes was detected. This is the first demonstration that gene-modified cells can exert genetic pressure on HIV. We conclude that gene-modified T cells have the potential to decrease the fitness of HIV-1 and conditionally replicative lentiviral vectors have a promising safety profile in T cells.


Molecular Therapy | 2012

Replication-Competent Retroviruses in Gene-Modified T Cells Used in Clinical Trials: Is It Time to Revise the Testing Requirements?

Adham S. Bear; Richard A. Morgan; Kenneth Cornetta; Carl H. June; Gwendolyn Binder-Scholl; Mark E. Dudley; Steven A. Feldman; Steven A. Rosenberg; Sheila A Shurtleff; Cliona M. Rooney; Helen E. Heslop; Gianpietro Dotti

Adoptive T-cell transfer is recognized as an innovative treatment strategy for various malignant diseases.1,2 To improve the efficacy and sometimes the safety of this approach, T cells can be genetically manipulated to modify their antigen specificity, to enhance their in vivo survival and trafficking to specific tissues, or to be eliminated in the event of undesired toxic effects.3 γ-retroviral vectors are frequently used to obtain robust and stable genetic modification of human T lymphocytes because these vectors can efficiently integrate within the genome and ensure that the inserted transgene is passed to the progeny of infected cells.


Journal for ImmunoTherapy of Cancer | 2015

Preclinical safety testing of an affinity-optimised MAGE-A10 T cell receptor for adoptive T cell therapy

Andrew B. Gerry; Joseph P. Sanderson; Manoj Saini; Barbara Tavano; Roslin Y Docta; Nicholas J. Pumphrey; Miguel Maroto; Ellen Border; Alan D. Bennett; Gwendolyn Binder-Scholl; Tom Holdich; Lini Pandite; R. G. Amado; Bent K. Jakobsen

Background Preclinical safety testing for adoptive immunotherapies utilising T cell receptor-(TCR) engineered T cells relies on stringent target validation and a robust pre-clinical specificity testing strategy. Specific safety concerns include ontarget off-tumour toxicity and off-target toxicity. Avoidance of these toxicities requires that the expression profile of the target antigen be shown to be specific to tumour tissues, and that the TCR is shown to be specific.


Cancer Research | 2015

Abstract 4701: NY-ESO T cells administered post ASCT for MM exhibit extended functionality without exhaustion in a natural pattern of effector and memory programming

Aaron P. Rapoport; Edward A. Stadtmauer; Luca Melchiori; Ryan Wong; Eduardo Davila; Gwendolyn Binder-Scholl; Tom Holdich; Dan T. Vogl; Brendan M. Weiss; Jeffrey Finkelstein; Simon F. Lacey; Sarah Bond; Marylene Fortin; Yoav Peretz; Joanna E. Brewer; Alan D. Bennett; Andrew B. Gerry; Nick Pumphrey; Helen K. Tayton-Martin; Lilliam Ribeiro; Ashraf Badros; Saul Yanovich; Nancy M. Hardy; Jean Yared; Naseem Kerr; Sunita Philip; Sandra Wesphal; Bruce L. Levine; Carl H. June; Michael Kalos

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Adoptive immunotherapy for cancer has been limited by a lack of antigen specificity, low levels of target expression, and failure to break self-tolerance. We hypothesized that infusion of genetically modified tumor-specific T cells following autologous stem cell transplant (ASCT) may overcome these barriers for multiple myeloma (MM). To test this, we conducted a phase I/II clinical trial ([NCT01352286][1]) in which T cells engineered with an HLA-A*0201 restricted, affinity-enhanced TCR recognizing NY-ESO-1 / LAGE-1 peptides (NY-ESOc259-T), were infused in the setting of profound lymphodepletion that accompanies high-dose chemotherapy given with ASCT. HLA-A*0201 MM patients eligible for ASCT, with antigen positive tumor were enrolled. NY-ESOc259-T was manufactured in a 10 day process using anti-CD3/CD28 microbeads and lentiviral vector, and was administered two days following ASCT. IMWG criteria were used to assess response at day 100 with the addition of a near complete response category (nCR) due to the common occurrence of oligoclonal banding observed following rapid post-ASCT immune reconstitution. Blood and marrow samples were taken at multiple timepoints for serum cytokine analysis, NY-ESOc259-T persistence and trafficking, multiparameter flow analysis to examine the phenotype and function of NY-ESOc259-T, and tumor biomarker analysis. 25 of 29 enrolled patients were infused. A mean of 2.8 × 109 engineered cells were administered (range 8.3 × 108-4.2 × 109), and the average transduction efficiency was 33% (range 30%-45%). Patients tended to have advanced disease (64% chromosomal abnormalities, and 24% prior ASCT). At 3 months, 67% (16/24) and 58% (14/24) of patients were in VGPR and nCR or better, respectively. Infusions were well-tolerated and no cytokine release syndrome was reported. NY-ESOc259-T persisted at 6 months in all but one patient, and in a subset of patients at 2 years; marrow infiltration was consistently observed from day 7 through day 180. NY-ESOc259-T initially displayed a dominant activated effector phenotype which converted towards a dominant effector memory phenotype by 1 year post infusion, in a pattern that mirrored clinical responses. Persisting cells demonstrated a polyfunctional response (IFN-γ and TNF-α) with a cytotoxic (CD107a and granzyme B) signature without overexpression of exhaustion markers (PD-1, LAG-3, and TIM-3). Tumor biomarker analysis is ongoing. MM relapse occurred in 13/25 patients. This data show that NY-ESOc259-T cells exhibit robust trafficking and expansion, durable persistence without exhaustion, and follow a natural immune expansion and contraction pattern consistent with an antigen-driven mechanism of action. Relapse correlated with a loss of persistence or tumor antigen escape, suggesting that targeting multiple antigens and maintenance infusions may increase durable remissions. Citation Format: Aaron Rapoport, Edward Stadtmauer, Luca Melchiori, Ryan Wong, Eduardo Davila, Gwendolyn Binder-Scholl, Tom Holdich, Dan Vogl, Brendan Weiss, Jeffrey Finkelstein, Simon Lacey, Sarah Bond, Marylene Fortin, Yoav Peretz, Joanna Brewer, Alan Bennett, Andrew Gerry, Nick Pumphrey, Helen Tayton-Martin, Lilliam Ribeiro, Ashraf Badros, Saul Yanovich, Nancy Hardy, Jean Yared, Naseem Kerr, Sunita Philip, Sandra Wesphal, Bruce L. Levine, Carl June, Michael Kalos, Bent Jakobsen. NY-ESO T cells administered post ASCT for MM exhibit extended functionality without exhaustion in a natural pattern of effector and memory programming. [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 4701. doi:10.1158/1538-7445.AM2015-4701 [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT01352286&atom=%2Fcanres%2F75%2F15_Supplement%2F4701.atom


Journal for ImmunoTherapy of Cancer | 2013

Adoptive immunotherapy with engineered T cells expressing and HLA-A2 restricted affinity-enhanced TCR for LAGE-1 and NY-ESO-1 in patients with multiple myeloma following auto-SCT

Aaron P. Rapoport; Edward A. Stadtmauer; Dan T. Vogl; Brendan M. Weiss; Gwendolyn Binder-Scholl; Dominic P. Smethurst; Jeffrey Finklestein; Irina Kulikovskaya; Minnal Gupta; Joanna E. Brewer; Alan D. Bennett; Andrew B. Gerry; Nick Pumphrey; Helen K. Tayton-Martin; Lilliam Ribeiro; Ashraf Badros; Saul Yanovich; Görgün Akpek; Naseem Kerr; Sunita Philip; Sandra Westphal; Levine L Bruce; Bent K. Jakobsen; Carl H. June; Michael Kalos

Meeting abstracts We report on a 26 patient phase I/II clinical trial ([NCT01352286][1]) to investigate the safety, feasibility and anti-tumor activity of T cells engineered to express an affinity-enhanced TCR that recognizes the NY-ESO-1/LAGE-1 peptide complex HLA-A*0201-SLLMWITQC. Patients with

Collaboration


Dive into the Gwendolyn Binder-Scholl's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carl H. June

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bruce L. Levine

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dan T. Vogl

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Brendan M. Weiss

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Minnal Gupta

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge