Network


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

Hotspot


Dive into the research topics where Tyler J. Curiel is active.

Publication


Featured researches published by Tyler J. Curiel.


Nature Medicine | 2004

Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival

Tyler J. Curiel; George Coukos; Linhua Zou; Xavier Alvarez; Pui Cheng; Peter Mottram; Melina Evdemon-Hogan; Jose R. Conejo-Garcia; Lin Zhang; Matthew E. Burow; Yun Zhu; Shuang Wei; Ilona Kryczek; Ben Daniel; Alan N. Gordon; Leann Myers; Andrew A. Lackner; Mary L. Disis; Keith L. Knutson; Lieping Chen; Weiping Zou

Regulatory T (Treg) cells mediate homeostatic peripheral tolerance by suppressing autoreactive T cells. Failure of host antitumor immunity may be caused by exaggerated suppression of tumor-associated antigen–reactive lymphocytes mediated by Treg cells; however, definitive evidence that Treg cells have an immunopathological role in human cancer is lacking. Here we show, in detailed studies of CD4+CD25+FOXP3+ Treg cells in 104 individuals affected with ovarian carcinoma, that human tumor Treg cells suppress tumor-specific T cell immunity and contribute to growth of human tumors in vivo. We also show that tumor Treg cells are associated with a high death hazard and reduced survival. Human Treg cells preferentially move to and accumulate in tumors and ascites, but rarely enter draining lymph nodes in later cancer stages. Tumor cells and microenvironmental macrophages produce the chemokine CCL22, which mediates trafficking of Treg cells to the tumor. This specific recruitment of Treg cells represents a mechanism by which tumors may foster immune privilege. Thus, blocking Treg cell migration or function may help to defeat human cancer.


Nature Medicine | 2003

Blockade of B7-H1 improves myeloid dendritic cell–mediated antitumor immunity

Tyler J. Curiel; Shuang Wei; Haidong Dong; Xavier Alvarez; Pui Cheng; Peter Mottram; Roman Krzysiek; Keith L. Knutson; Ben Daniel; Maria Zimmermann; Odile David; Matthew E. Burow; Alan N. Gordon; Nina Dhurandhar; Leann Myers; Ruth E. Berggren; Akseli Hemminki; Ronald D. Alvarez; Dominique Emilie; David T. Curiel; Lieping Chen; Weiping Zou

Suppression of dendritic cell function in cancer patients is thought to contribute to the inhibition of immune responses and disease progression. Molecular mechanisms of this suppression remain elusive, however. Here, we show that a fraction of blood monocyte-derived myeloid dendritic cells (MDCs) express B7-H1, a member of the B7 family, on the cell surface. B7-H1 could be further upregulated by tumor environmental factors. Consistent with this finding, virtually all MDCs isolated from the tissues or draining lymph nodes of ovarian carcinomas express B7-H1. Blockade of B7-H1 enhanced MDC-mediated T-cell activation and was accompanied by downregulation of T-cell interleukin (IL)-10 and upregulation of IL-2 and interferon (IFN)-γ. T cells conditioned with the B7-H1–blocked MDCs had a more potent ability to inhibit autologous human ovarian carcinoma growth in non-obese diabetic–severe combined immunodeficient (NOD-SCID) mice. Therefore, upregulation of B7-H1 on MDCs in the tumor microenvironment downregulates T-cell immunity. Blockade of B7-H1 represents one approach for cancer immunotherapy.


Nature Medicine | 2001

Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells

Weiping Zou; Véronique Machelon; Aurore Coulomb-L'Hermin; Jozef Borvak; Françoise Nomé; Tatyana Isaeva; Shuang Wei; Roman Krzysiek; Ingrid Durand-Gasselin; Alan N. Gordon; Terri Pustilnik; David T. Curiel; Pierre Galanaud; Frédérique Capron; Dominique Emilie; Tyler J. Curiel

Dendritic-cell (DC) trafficking and function in tumors is poorly characterized, with studies confined to myeloid DCs (DC1s). Tumors inhibit DC1 migration and function, likely hindering specific immunity. The role of plasmacytoid DCs (DC2s) in tumor immunity is unknown. We show here that malignant human ovarian epithelial tumor cells express very high levels of stromal-derived factor-1, which induces DC2 precursor (preDC2) chemotaxis and adhesion/transmigration, upregulates preDC2 very late antigen (VLA)-5, and protects preDC2s from tumor macrophage interleukin-10–induced apoptosis, all through CXC chemokine receptor-4. The VLA-5 ligand vascular-cell adhesion molecule-1 mediated preDC2 adhesion/transmigration. Tumor preDC2s induced significant T-cell interleukin-10 unrelated to preDC2 differentiation or activation state, and this contributed to poor T-cell activation. Myeloid precursor DCs (preDC1s) were not detected. Tumors may weaken immunity by attracting preDC2s and protecting them from the harsh microenvironment, and by altering preDC1 distribution.


Journal of Experimental Medicine | 2006

B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma

Ilona Kryczek; Linhua Zou; Paulo C. Rodriguez; Gefeng Zhu; Shuang Wei; Peter Mottram; Michael J. Brumlik; Pui Cheng; Tyler J. Curiel; Leann Myers; Andrew A. Lackner; Xavier Alvarez; Augusto C. Ochoa; Lieping Chen; Weiping Zou

Tumor-associated macrophages are a prominent component of ovarian cancer stroma and contribute to tumor progression. B7-H4 is a recently identified B7 family molecule. We show that primary ovarian tumor cells express intracellular B7-H4, whereas a fraction of tumor macrophages expresses surface B7-H4. B7-H4+ tumor macrophages, but not primary ovarian tumor cells, suppress tumor-associated antigen-specific T cell immunity. Blocking B7-H4-, but not arginase-, inducible nitric oxide synthase or B7-H1 restored the T cell stimulating capacity of the macrophages and contributes to tumor regression in vivo. Interleukin (IL)-6 and IL-10 are found in high concentrations in the tumor microenvironment. These cytokines stimulate macrophage B7-H4 expression. In contrast, granulocyte/macrophage colony-stimulating factor and IL-4, which are limited in the tumor microenvironment, inhibit B7-H4 expression. Ectopic expression of B7-H4 makes normal macrophages suppressive. Thus, B7-H4+ tumor macrophages constitute a novel suppressor cell population in ovarian cancer. B7-H4 expression represents a critical checkpoint in determining host responses to dysfunctional cytokines in ovarian cancer. Blocking B7-H4 or depleting B7-H4+ tumor macrophages may represent novel strategies to enhance T cell tumor immunity in cancer.


Journal of Clinical Investigation | 2007

Tregs and rethinking cancer immunotherapy

Tyler J. Curiel

Tumors express antigens that should induce immune-mediated rejection, but spontaneous rejection of established tumors is rare. Recent work demonstrates that one reason for the lack of tumor rejection is that tumors actively defeat host immunity. This concept forces us to rethink current approaches to harnessing potent, specific host immunity to battle cancer, most of which are based on the paradigm that inducing more antitumor immune cells alone is therapeutic. However, as I discuss in this Personal Perspective, a newer paradigm predicts that reducing tumor-driven immune suppression will be clinically beneficial. CD4+CD25+ Tregs are one mechanism of tumor-driven immune evasion that provide prototypical targets for testing novel anticancer treatment strategies within the newer paradigm.


Cancer Research | 2005

Plasmacytoid dendritic cells induce CD8+ regulatory T cells in human ovarian carcinoma.

Shuang Wei; Ilona Kryczek; Linhua Zou; Ben Daniel; Pui Cheng; Peter Mottram; Tyler J. Curiel; Andrzej Lange; Weiping Zou

To directly dissect the role of each immune component in human tumor immunopathogenesis, we have studied the interaction between dendritic cells and T cells in the tumor environment of patients with ovarian carcinoma. We previously reported that functional plasmacytoid dendritic cells, but not functionally mature myeloid dendritic cells, accumulated in tumor microenvironments. We now show that tumor ascites macrophage-derived dendritic cells induced tumor-associated antigen-specific CD8+ T cells with effector functions. Strikingly, tumor ascites plasmacytoid dendritic cells induced interleukin-10+ CCR7+ CD45RO+ CD8+ regulatory T cells. Four characteristics have been identified in tumor plasmacytoid dendritic cell-induced CD8+ regulatory T cells: (a) induction of CD8+ regulatory T cells is independent of CD4+ CD25+ T cells; (b) CD8+ regulatory T cells significantly suppress myeloid dendritic cell-mediated tumor-associated antigen-specific T cell effector functions through interleukin-10; (c) repetitive myeloid dendritic cell stimulation can recover CD8+ regulatory T cell-mediated poor T cell proliferation, but not T cell effector function; (d) CD8+ regulatory T cells express functional CCR7, and efficiently migrate with lymphoid homing chemokine MIP-3beta. Primary suppressive CCR7+ CD45RO+ CD8+ T cells are found in the tumor environment of patients with ovarian cancers. Thus, tumor-associated plasmacytoid dendritic cells contribute to the tumor environmental immunosuppressive network. Collectively, tumors manipulate tumor microenvironmental dendritic cell subset distribution and function to subvert tumor immunity. The data are relevant to understanding tumor immunopathology as well as reevaluating tumor immunotherapeutic strategies.


Cancer Research | 2004

Bone Marrow Is a Reservoir for CD4+CD25+ Regulatory T Cells that Traffic through CXCL12/CXCR4 Signals

Linhua Zou; Brian G. Barnett; Hana Safah; Vincent F. LaRussa; Melina Evdemon-Hogan; Peter Mottram; Shuang Wei; Odile David; Tyler J. Curiel; Weiping Zou

CD4+CD25+ regulatory T cells (Tregs) mediate peripheral T-cell homeostasis and contribute to self-tolerance. Their homeostatic and pathologic trafficking is poorly understood. Under homeostatic conditions, we show a relatively high prevalence of functional Tregs in human bone marrow. Bone marrow strongly expresses functional stromal-derived factor (CXCL12), the ligand for CXCR4. Human Tregs traffic to and are retained in bone marrow through CXCR4/CXCL12 signals as shown in chimeric nonobese diabetic/severe combined immunodeficient mice. Granulocyte colony-stimulating factor (G-CSF) reduces human bone marrow CXCL12 expression in vivo, associated with mobilization of marrow Tregs to peripheral blood in human volunteers. These findings show a mechanism for homeostatic Treg trafficking and indicate that bone marrow is a significant reservoir for Tregs. These data also suggest a novel mechanism explaining reduced acute graft-versus-host disease and improvement in autoimmune diseases following G-CSF treatment.


Journal of Immunology | 2003

Cross-Talk in the Innate Immune System: Neutrophils Instruct Recruitment and Activation of Dendritic Cells during Microbial Infection

Soumaya Bennouna; Susan K. Bliss; Tyler J. Curiel; Eric Y. Denkers

Type I inflammatory cytokines are essential for immunity to many microbial pathogens, including Toxoplasma gondii. Dendritic cells (DC) are key to initiating type 1 immunity, but neutrophils are also a source of chemokines and cytokines involved in Th1 response ignition. We found that T. gondii triggered neutrophil synthesis of CC chemokine ligand (CCL)3, CCL4, CCL5, and CCL20, chemokines that were strongly chemotactic for immature DC. Moreover, supernatants obtained from parasite-stimulated polymorphonuclear leukocytes induced DC IL-12(p40) and TNF-α production. Parasite-triggered neutrophils also released factors that induced DC CD40 and CD86 up-regulation, and this response was dependent upon parasite-triggered neutrophil TNF-α production. In vivo evidence that polymorphonuclear leukocytes exert an important influence on DC activation was obtained by examining splenic DC cytokine production following infection of neutrophil-depleted mice. These animals displayed severely curtailed splenic DC IL-12 and TNF-α production, as revealed by ex vivo flow cytometric analysis and in vitro culture assay. Our results reveal a previously unrecognized regulatory role for neutrophils in DC function during microbial infection, and suggest that cross-talk between these cell populations is an important component of the innate immune response to infection.


Journal of Clinical Oncology | 2016

Safety and Efficacy of Durvalumab (MEDI4736), an Anti-Programmed Cell Death Ligand-1 Immune Checkpoint Inhibitor, in Patients With Advanced Urothelial Bladder Cancer

Christophe Massard; Michael S. Gordon; Sunil Sharma; Saeed Rafii; Zev A. Wainberg; Jason J. Luke; Tyler J. Curiel; Gerardo Colon-Otero; Omid Hamid; Rachel E. Sanborn; Peter H. O'Donnell; Alexandra Drakaki; Winston Tan; John Kurland; Marlon Rebelatto; Xiaoping Jin; John A. Blake-Haskins; Ashok Kumar Gupta; Neil Howard Segal

PURPOSE To investigate the safety and efficacy of durvalumab, a human monoclonal antibody that binds programmed cell death ligand-1 (PD-L1), and the role of PD-L1 expression on clinical response in patients with advanced urothelial bladder cancer (UBC). METHODS A phase 1/2 multicenter, open-label study is being conducted in patients with inoperable or metastatic solid tumors. We report here the results from the UBC expansion cohort. Durvalumab (MEDI4736, 10 mg/kg every 2 weeks) was administered intravenously for up to 12 months. The primary end point was safety, and objective response rate (ORR, confirmed) was a key secondary end point. An exploratory analysis of pretreatment tumor biopsies led to defining PD-L1-positive as ≥ 25% of tumor cells or tumor-infiltrating immune cells expressing membrane PD-L1. RESULTS A total of 61 patients (40 PD-L1-positive, 21 PD-L1-negative), 93.4% of whom received one or more prior therapies for advanced disease, were treated (median duration of follow-up, 4.3 months). The most common treatment-related adverse events (AEs) of any grade were fatigue (13.1%), diarrhea (9.8%), and decreased appetite (8.2%). Grade 3 treatment-related AEs occurred in three patients (4.9%); there were no treatment-related grade 4 or 5 AEs. One treatment-related AE (acute kidney injury) resulted in treatment discontinuation. The ORR was 31.0% (95% CI, 17.6 to 47.1) in 42 response-evaluable patients, 46.4% (95% CI, 27.5 to 66.1) in the PD-L1-positive subgroup, and 0% (95% CI, 0.0 to 23.2) in the PD-L1-negative subgroup. Responses are ongoing in 12 of 13 responding patients, with median duration of response not yet reached (range, 4.1+ to 49.3+ weeks). CONCLUSION Durvalumab demonstrated a manageable safety profile and evidence of meaningful clinical activity in PD-L1-positive patients with UBC, many of whom were heavily pretreated.


American Journal of Reproductive Immunology | 2005

Regulatory T cells in ovarian cancer: biology and therapeutic potential.

Brian G. Barnett; Ilona Kryczek; Pui Cheng; Weiping Zou; Tyler J. Curiel

Tumors express tumor‐associated antigens (TAA) and thus should be the object of immune attack. Nonetheless, spontaneous clearance of established tumors is rare. 1 Much work has demonstrated that tumors have numerous strategies either to prevent presentation of TAA, or to prevent TAA presentation in the context of T‐cell co‐signaling molecules. 1,2 Thus, it was thought that lack of TAA‐specific immunity was largely a passive process: tumors simply did not present enough TAA, or antigen‐presenting cells did not have sufficient stimulatory capacity. On this basis, attempts were made to bolster TAA‐specific immunity by using optimal antigen‐presenting cells or by growing TAA‐specific effector T cells ex vivo followed by adoptive transfer. 3 These approaches met with some success in mouse models of human tumors, and showed some early clinical efficacy in human trials, although long‐term efficacy remains to be established, and logistical problems are considerable. 4 These studies established the concept that experimentally induced TAA‐specific immunity is a rational and potentially efficacious means to treat cancer, including ovarian cancer. Nonetheless, recent work demonstrates that lack of naturally induced TAA‐specific immunity is not simply a passive process. 5–12 We discuss recent data clearly demonstrating that ‘tumors actively prevent induction of TAA‐specific immunity through induction of TAA‐specific tolerance’. 13 This tolerance is mediated in part by regulatory T cells (Tregs). Means to revert these tolerizing conditions represent a novel anticancer therapeutic stratagem. We discuss Tregs in this regard in human ovarian cancer and present evidence that depleting Treg in human cancer, including ovarian cancer, using denileukin diftitox (Ontak), improves immunity and may be therapeutic.

Collaboration


Dive into the Tyler J. Curiel's collaboration.

Top Co-Authors

Avatar

Vincent Hurez

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Weiping Zou

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Michael J. Brumlik

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Aijie Liu

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Benjamin J. Daniel

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Srilakshmi Pandeswara

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Tahiro Shin

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Bin Zhang

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Lishi Sun

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Álvaro Padrón

University of Texas Health Science Center at San Antonio

View shared research outputs
Researchain Logo
Decentralizing Knowledge