Network


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

Hotspot


Dive into the research topics where Terence Purdon is active.

Publication


Featured researches published by Terence Purdon.


OncoImmunology | 2015

IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors in vivo.

Mythili Koneru; Terence Purdon; David R. Spriggs; Susmith Koneru; Renier J. Brentjens

A novel approach for the treatment of ovarian cancer includes immunotherapy with genetically engineered T cells targeted to ovarian cancer cell antigens. Using retroviral transduction, T cells can be created that express an artificial T cell receptor (TCR) termed a chimeric antigen receptor (CAR). We have generated a CAR, 4H11-28z, specific to MUC-16ecto antigen, which is the over-expressed on a majority of ovarian tumor cells and is the retained portion of MUC-16 after cleavage of CA-125. We previously demonstrated that T cells modified to express the 4H11-28z CAR eradicate orthotopic human ovarian cancer xenografts in SCID-Beige mice. However, despite the ability of CAR T cells to localize to tumors, their activation in the clinical setting can be inhibited by the tumor microenvironment, as is commonly seen for endogenous antitumor immune response. To potentially overcome this limitation, we have recently developed a construct that co-expresses both MUC16ecto CAR and IL-12 (4H11-28z/IL-12). In vitro, 4H11-28z/IL-12 CAR T cells show enhanced proliferation and robust IFNγ secretion compared to 4H11-28z CAR T cells. In SCID-Beige mice with human ovarian cancer xenografts, IL-12 secreting CAR T cells exhibit enhanced antitumor efficacy as determined by increased survival, prolonged persistence of T cells, and higher systemic IFNγ. Furthermore, in anticipation of translating these results into a phase I clinical trial which will be the first to study IL-12 secreting CAR T cells in ovarian cancer, an elimination gene has been included to allow for deletion of CAR T cells in the context of unforeseen or off-tumor on-target toxicity.


Molecular Therapy | 2015

Enhancing antitumor efficacy of chimeric antigen receptor T cells through constitutive CD40L expression.

Kevin J. Curran; Beatrijs Seinstra; Yan Nikhamin; Raymond Yeh; Yelena Usachenko; Dayenne G van Leeuwen; Terence Purdon; Hollie J. Pegram; Renier J. Brentjens

Adoptive cell therapy with genetically modified T cells expressing a chimeric antigen receptor (CAR) is a promising therapy for patients with B-cell acute lymphoblastic leukemia. However, CAR-modified T cells (CAR T cells) have mostly failed in patients with solid tumors or low-grade B-cell malignancies including chronic lymphocytic leukemia with bulky lymph node involvement. Herein, we enhance the antitumor efficacy of CAR T cells through the constitutive expression of CD40 ligand (CD40L, CD154). T cells genetically modified to constitutively express CD40L (CD40L-modified T cells) demonstrated increased proliferation and secretion of proinflammatory TH1 cytokines. Further, CD40L-modified T cells augmented the immunogenicity of CD40(+) tumor cells by the upregulated surface expression of costimulatory molecules (CD80 and CD86), adhesion molecules (CD54, CD58, and CD70), human leukocyte antigen (HLA) molecules (Class I and HLA-DR), and the Fas-death receptor (CD95). Additionally, CD40L-modified T cells induced maturation and secretion of the proinflammatory cytokine interleukin-12 by monocyte-derived dendritic cells. Finally, tumor-targeted CD19-specific CAR/CD40L T cells exhibited increased cytotoxicity against CD40(+) tumors and extended the survival of tumor-bearing mice in a xenotransplant model of CD19(+) systemic lymphoma. This preclinical data supports the clinical application of CAR T cells additionally modified to constitutively express CD40L with anticipated enhanced antitumor efficacy.


Scientific Reports | 2017

Armored CAR T cells enhance antitumor efficacy and overcome the tumor microenvironment

Oladapo Yeku; Terence Purdon; Mythili Koneru; David R. Spriggs; Renier J. Brentjens

Chimeric antigen receptor (CAR) T cell therapy has shown limited efficacy for the management of solid tumor malignancies. In ovarian cancer, this is in part due to an immunosuppressive cytokine and cellular tumor microenvironment which suppresses adoptively transferred T cells. We engineered an armored CAR T cell capable of constitutive secretion of IL-12, and delineate the mechanisms via which these CAR T cells overcome a hostile tumor microenvironment. In this report, we demonstrate enhanced proliferation, decreased apoptosis and increased cytotoxicity in the presence of immunosuppressive ascites. In vivo, we show enhanced expansion and CAR T cell antitumor efficacy, culminating in improvement in survival in a syngeneic model of ovarian peritoneal carcinomatosis. Armored CAR T cells mediated depletion of tumor associated macrophages and resisted endogenous PD-L1-induced inhibition. These findings highlight the role of the inhibitory microenvironment and how CAR T cells can be further engineered to maintain efficacy.


Nature Biotechnology | 2018

Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo

Sarwish Rafiq; Oladapo Yeku; Hollie J. Jackson; Terence Purdon; Dayenne G van Leeuwen; Dylan J Drakes; Mei Song; Matthew M. Miele; Zhuoning Li; Pei Wang; Su Yan; Jingyi Xiang; Xiaojing Ma; Venkatraman E. Seshan; Ronald C. Hendrickson; Cheng Liu; Renier J. Brentjens

The efficacy of chimeric antigen receptor (CAR) T cell therapy against poorly responding tumors can be enhanced by administering the cells in combination with immune checkpoint blockade inhibitors. Alternatively, the CAR construct has been engineered to coexpress factors that boost CAR-T cell function in the tumor microenvironment. We modified CAR-T cells to secrete PD-1-blocking single-chain variable fragments (scFv). These scFv-secreting CAR-T cells acted in both a paracrine and autocrine manner to improve the anti-tumor activity of CAR-T cells and bystander tumor-specific T cells in clinically relevant syngeneic and xenogeneic mouse models of PD-L1+ hematologic and solid tumors. The efficacy was similar to or better than that achieved by combination therapy with CAR-T cells and a checkpoint inhibitor. This approach may improve safety, as the secreted scFvs remained localized to the tumor, protecting CAR-T cells from PD-1 inhibition, which could potentially avoid toxicities associated with systemic checkpoint inhibition.


Cell Reports | 2018

Engineered Tumor-Targeted T Cells Mediate Enhanced Anti-Tumor Efficacy Both Directly and through Activation of the Endogenous Immune System

Mauro P. Avanzi; Oladapo Yeku; Xinghuo Li; Dinali Wijewarnasuriya; Dayenne G van Leeuwen; Kenneth Cheung; Hyebin Park; Terence Purdon; Anthony F. Daniyan; Matthew H. Spitzer; Renier J. Brentjens

SUMMARY Chimeric antigen receptor (CAR) T cell therapy has proven clinically beneficial against B cell acute lymphoblastic leukemia and non-Hodgkin’s lymphoma. However, suboptimal clinical outcomes have been associated with decreased expansion and persistence of adoptively transferred CAR T cells, antigen-negative relapses, and impairment by an immunosuppressive tumor microenvironment. Improvements in CAR T cell design are required to enhance clinical efficacy, as well as broaden the applicability of this technology. Here, we demonstrate that interleukin-18 (IL-18)-secreting CAR T cells exhibit enhanced in vivo expansion and persistence and significantly increase long-term survival in syngeneic mouse models of both hematological and solid malignancies. In addition, we demonstrate that IL-18-secreting CAR T cells are capable of modulating the tumor microenvironment, as well as enhancing an effective endogenous anti-tumor immune response. IL-18-secreting CART cells represent a promising strategy to enhance the clinical outcomes of adoptive T cell therapy.


Cancer Discovery | 2018

Clinical and Biological Correlates of Neurotoxicity Associated with CAR T-cell Therapy in Patients with B-cell Acute Lymphoblastic Leukemia

Bianca Santomasso; Jae H. Park; Darin Salloum; Isabelle Riviere; Jessica Flynn; Elena Mead; Elizabeth Halton; Xiuyan Wang; Brigitte Senechal; Terence Purdon; Justin R. Cross; Hui Liu; Behroze Vachha; Xi Chen; Lisa M. DeAngelis; Daniel Li; Yvette Bernal; Mithat Gonen; Hans-Guido Wendel; Michel Sadelain; Renier J. Brentjens

CD19-specific chimeric antigen receptor (CAR) T-cell therapy is highly effective against relapsed or refractory acute lymphoblastic leukemia (ALL), but is hindered by neurotoxicity. In 53 adult patients with ALL, we found a significant association of severe neurotoxicity with high pretreatment disease burden, higher peak CAR T-cell expansion, and early and higher elevations of proinflammatory cytokines in blood. Patients with severe neurotoxicity had evidence of blood-cerebrospinal fluid (CSF) barrier disruption correlating with neurotoxicity grade without association with CSF white blood cell count or CAR T-cell quantity in CSF. Proinflammatory cytokines were enriched in CSF during severe neurotoxicity with disproportionately high levels of IL6, IL8, MCP1, and IP10, suggesting central nervous system-specific production. Seizures, seizure-like activity, myoclonus, and neuroimaging characteristics suggested excitatory neurotoxicity, and we found elevated levels of endogenous excitatory agonists in CSF during neurotoxicity.Significance: We detail the neurologic symptoms and blood, CSF, and neuroimaging correlates of neurotoxicity associated with CD19 CAR T cells and identify neurotoxicity risk factors. Our findings implicate cellular components other than T cells and suggest novel links between systemic inflammation and characteristic neurotoxicity symptoms. Cancer Discov; 8(8); 958-71. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 899.


Cancer immunology research | 2017

Abstract PR08: Armored CAR T cells genetically modified to secrete IL-12 show enhanced efficacy and overcome a hostile tumor microenvironment in mouse ovarian peritoneal carcinomatosis

Oladapo Yeku; Terence Purdon; David R. Spriggs; Renier J. Brentjens

Chimeric antigen receptor T cell (CAR T) therapy for solid tumors has not replicated the success seen with hematologic malignancies such as CD19-expressing B cell acute lymphoblastic leukemia (B-ALL) and Myeloma. One hypothesis for this lack of efficacy is an immunosuppressive tumor microenvironment that renders adoptively transferred cells ineffective. Armored CAR T cells are CAR T cells that have been further engineered to express additional costimulatory ligands, soluble cytokines or secretable proteins in order to overcome a hostile tumor microenvironment. IL-12 is a proinflammatory cytokine that is only secreted by antigen presenting cells and has been shown to activate effector cells, suppress regulatory T cells and reprogram tumor associated macrophages. We hypothesize that CAR T cells that have been genetically engineered to secrete IL-12 will be more efficacious in the treatment of Muc16ecto positive ovarian cancer by overcoming an otherwise immunosuppressive ascitic tumor microenvironment. Herein we report the mechanisms of enhanced efficacy of an IL-12-armored CAR T cell (4H1128ζ-IL12) directed towards the retained portion of Muc16 (Muc16ecto). Using an immunocompetent syngeneic peritoneal carcinomatosis model of ovarian cancer (i.p ID8-muc16ecto), we show superior therapeutic efficacy of armored CAR T cells compared to second generation CARs (4H1128ζ). Tumor-bearing mice treated after 35 days with 4H1128ζ-IL12 were tumor-free for over 100 days compared to 4H1128ζ-treated mice (o.s: not reached vs 46 days, p= 0.008). Even when the tumor was allowed to establish for 42 days, mice treated with 4H1128ζ-IL12 survived longer than 4H1128ζ- treated mice (o.s: 97 vs 54 days, p= 0.0123). Investigations into the mechanism of improved survival in armored CAR T cell-treated mice showed increased in vivo expansion and modulation of the immunosuppressive ascites cytokine milieu via increased secretion of IL-12, IFN-γ, and TNF-α. Furthermore, 4H1128ζ-IL12 CAR T cells demonstrated enhanced in vitro proliferation and cytotoxic activity when cultured in ascites compared to control CAR T cells. Taken together, these results show that armored CAR T cells can be successfully engineered to overcome an otherwise immunosuppressive solid tumor microenvironment. These results represent the mechanistic companion of an ongoing phase I clinical trial of IL-12 armored CAR T cells in patients with relapsed/refractory ovarian cancer (NCT02498912). This abstract is also being presented as Poster A32. Citation Format: Oladapo Yeku, Terence Purdon, David Spriggs, Renier Brentjens. Armored CAR T cells genetically modified to secrete IL-12 show enhanced efficacy and overcome a hostile tumor microenvironment in mouse ovarian peritoneal carcinomatosis. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2016 Oct 20-23; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2017;5(3 Suppl):Abstract nr PR08.


Molecular Therapy | 2016

393. Engineering Armored T Cell Receptor-Mimic (TCRm) Chimeric Antigen Receptor (CAR) T Cells Specific for the Intracellular Protein Wilms Tumor 1 (WT1) for Treatment of Hematologic and Solid Malignancies

Sarwish Rafiq; Terence Purdon; Tao Dao; Chen Liu; David A. Scheinberg; Renier J. Brentjens

Adoptive therapy with chimeric antigen receptor (CAR) T cells specific for CD19 is clinically successful in a limited set of leukemias and most CARs studied are targeted against external antigens. Wilms Tumor Antigen 1 (WT1) protein (WT1) is an intracellular antigen overexpressed in many cancers, including leukemias and solid malignancies and is thus an appealing, broadly applicable target. We have engineered the first armored T cell receptor-mimic (TCRm) CAR against WT1. Derived from the ESK1 antibody, the second generation CAR, WT1-28z, is reactive with the RMFPNAPYL peptide of WT1 that is processed and presented on the surface of cells in the context of HLA-A*02:01. WT1-28z was further modified to secrete human IL-12 cytokine, thus creating the armored CAR WT1-28z/IL-12. T cells expressing WT1-28z or WT1-28z/IL-12 are cytotoxic against a range of both hematological and solid tumors. Importantly, both WT1-directed T cells are specific for the WT1-HLA-A*02:01 complex and are not reactive against cells that do not express both HLA-A*02:01 and WT1. In established SCID/Beige mouse models of either acute leukemia or ovarian cancer, one dose of WT1-28z T cells prolongs survival of mice over untreated or irrelevant antigen-specific CAR T cell treated mice. Furthermore, one dose of the armored WT1-28z/IL12 CAR T cells further significantly prolongs survival of mice in both models over WT1-28z CAR T cell treated mice, with a subset of mice whose disease was eradicated. The armored TCRm CAR T cells against WT1 are effective in eradicating disease in both hematologic and solid tumors and may hold great clinical potential to expand on the success of CAR T cell therapy.


Blood | 2015

Implications of Minimal Residual Disease Negative Complete Remission (MRD-CR) and Allogeneic Stem Cell Transplant on Safety and Clinical Outcome of CD19-Targeted 19-28z CAR Modified T Cells in Adult Patients with Relapsed, Refractory B-Cell ALL

Jae H. Park; Isabelle Riviere; Xiuyan Wang; Yvette Bernal; Terence Purdon; Elizabeth Halton; Yongzeng Wang; Kevin J. Curran; Craig S. Sauter; Michel Sadelain; Renier J. Brentjens


Journal of Clinical Oncology | 2015

Phase I trial of 19-28z chimeric antigen receptor modified T cells (19-28z CAR-T) post-high dose therapy and autologous stem cell transplant (HDT-ASCT) for relapsed and refractory (rel/ref) aggressive B-cell non-Hodgkin lymphoma (B-NHL).

Craig S. Sauter; Isabelle Riviere; Yvette Bernal; Xiuyan Wang; Terence Purdon; Sarah Yoo; Craig H. Moskowitz; Sergio Giralt; Matthew J. Matasar; Kevin J. Curran; Jae Hong Park; Michel Sadelain; Renier J. Brentjens

Collaboration


Dive into the Terence Purdon's collaboration.

Top Co-Authors

Avatar

Renier J. Brentjens

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Isabelle Riviere

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michel Sadelain

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Xiuyan Wang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Halton

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Oladapo Yeku

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Brigitte Senechal

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jae Hong Park

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David R. Spriggs

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jae H. Park

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge