Network


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

Hotspot


Dive into the research topics where Stephen Doberstein is active.

Publication


Featured researches published by Stephen Doberstein.


Science | 2008

Discovery of a cytokine and its receptor by functional screening of the extracellular proteome.

Haishan Lin; Ernestine Lee; Kevin Hestir; Cindy Leo; Minmei Huang; Elizabeth Bosch; Robert F. Halenbeck; Ge Wu; Aileen Zhou; Dirk Behrens; Diane Hollenbaugh; Thomas Linnemann; Minmin Qin; Justin Wong; Keting Chu; Stephen Doberstein; Lewis T. Williams

To understand the system of secreted proteins and receptors involved in cell-cell signaling, we produced a comprehensive set of recombinant secreted proteins and the extracellular domains of transmembrane proteins, which constitute most of the protein components of the extracellular space. Each protein was tested in a suite of assays that measured metabolic, growth, or transcriptional responses in diverse cell types. The pattern of responses across assays was analyzed for the degree of functional selectivity of each protein. One of the highly selective proteins was a previously undescribed ligand, designated interleukin-34 (IL-34), which stimulates monocyte viability but does not affect responses in a wide spectrum of other assays. In a separate functional screen, we used a collection of extracellular domains of transmembrane proteins to discover the receptor for IL-34, which was a known cytokine receptor, colony-stimulating factor 1 (also called macrophage colony-stimulating factor) receptor. This systematic approach is thus useful for discovering new ligands and receptors and assessing the functional selectivity of extracellular regulatory proteins.


Journal of Immunology | 2007

Dominant-Negative Inhibitors of Soluble TNF Attenuate Experimental Arthritis without Suppressing Innate Immunity to Infection

Jonathan Zalevsky; Thomas Secher; Sergei A. Ezhevsky; Laure Janot; Paul Michael Steed; Christopher O'brien; Araz Eivazi; James Kung; Duc-Hanh T. Nguyen; Stephen Doberstein; François Erard; Bernhard Ryffel; David E. Szymkowski

TNF is a pleiotropic cytokine required for normal development and function of the immune system; however, TNF overexpression also induces inflammation and is associated with autoimmune diseases. TNF exists as both a soluble and a transmembrane protein. Genetic studies in mice have suggested that inflammation in disease models involves soluble TNF (solTNF) and that maintenance of innate immune function involves transmembrane TNF (tmTNF). These findings imply that selective pharmacologic inhibition of solTNF may be anti-inflammatory and yet preserve innate immunity to infection. To address this hypothesis, we now describe dominant-negative inhibitors of TNF (DN-TNFs) as a new class of biologics that selectively inhibits solTNF. DN-TNFs blocked solTNF activity in human and mouse cells, a human blood cytokine release assay, and two mouse arthritis models. In contrast, DN-TNFs neither inhibited the activity of human or mouse tmTNF nor suppressed innate immunity to Listeria infection in mice. These results establish DN-TNFs as the first selective inhibitors of solTNF, demonstrate that inflammation in mouse arthritis models is primarily driven by solTNF, and suggest that the maintenance of tmTNF activity may improve the therapeutic index of future anti-inflammatory agents.


Nature Neuroscience | 1999

Termination of phototransduction requires binding of the NINAC myosin III and the PDZ protein INAD.

Paul D. Wes; Xian Zhong Shawn Xu; Hong-Sheng Li; Fred Chien; Stephen Doberstein; Craig Montell

Many of the proteins that are critical for Drosophila phototransduction assemble into a signaling complex, signalplex, through association with the PDZ-domain protein INAD. Some of these proteins depend on INAD for proper subcellular localization to the phototransducing organelle, the rhabdomere, making it difficult to assess any physiological function of this signaling complex independent of localization. Here we demonstrated that INAD bound directly to the NINAC myosin III, yet the subcellular localization of NINAC was normal in inaD mutants. Nevertheless, the INAD binding site was sufficient to target a heterologous protein to the rhabdomeres. Disruption of the NINAC/INAD interaction delayed termination of the photoreceptor response. Thus one role of this signaling complex is in rapid deactivation of the photoresponse.


Clinical Cancer Research | 2016

NKTR-214, an Engineered Cytokine with Biased IL2 Receptor Binding, Increased Tumor Exposure, and Marked Efficacy in Mouse Tumor Models

Deborah H. Charych; Ute Hoch; John L. Langowski; Steve R. Lee; Murali Addepalli; Peter Kirk; Dawei Sheng; Xiaofeng Liu; Paul Sims; Laurie VanderVeen; Cherie F. Ali; Thomas K. H. Chang; Marina Konakova; Rhoneil Pena; Rupesh S. Kanhere; Yolanda Kirksey; Chunmei Ji; Yujun Wang; Jicai Huang; Theresa D. Sweeney; Seema S. Kantak; Stephen Doberstein

Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy for metastatic melanoma and renal cancer, with durable responses in approximately 10% of patients; however, severe side effects limit maximal dosing and thus the number of patients able to receive treatment and potential cure. NKTR-214 is a prodrug of conjugated IL2, retaining the same amino acid sequence as aldesleukin. The IL2 core is conjugated to 6 releasable polyethylene glycol (PEG) chains. In vivo, the PEG chains slowly release to generate active IL2 conjugates. Experimental Design: We evaluated the bioactivity and receptor binding of NKTR-214 and its active IL2 conjugates in vitro; the tumor immunology, tumor pharmacokinetics, and efficacy of NKTR-214 as a single agent and in combination with anti–CTLA-4 antibody in murine tumor models. Tolerability was evaluated in non-human primates. Results: In a murine melanoma tumor model, the ratio of tumor-killing CD8+ T cells to Foxp3+ regulatory T cells was greater than 400 for NKTR-214 compared with 18 for aldesleukin, supporting preferential activation of the IL2 receptor beta over IL2 receptor alpha, due to the location of PEG molecules. NKTR-214 provides a 500-fold greater exposure of the tumor to conjugated IL2 compared with aldesleukin. NKTR-214 showed efficacy as a single agent and provided durable immunity that was resistant to tumor rechallenge in combination with anti–CTLA-4 antibody. NKTR-214 was well tolerated in non-human primates. Conclusions: These data support further evaluation of NKTR-214 in humans for a variety of tumor types, adding to the repertoire of potent and potentially curative cancer immunotherapies. Clin Cancer Res; 22(3); 680–90. ©2016 AACR.


PLOS ONE | 2017

Modeling the receptor pharmacology, pharmacokinetics, and pharmacodynamics of NKTR-214, a kinetically-controlled interleukin-2 (IL2) receptor agonist for cancer immunotherapy

Deborah H. Charych; Samira Khalili; Vidula Dixit; Peter Kirk; Thomas K. H. Chang; John L. Langowski; Werner Rubas; Stephen Doberstein; Michael A. Eldon; Ute Hoch; Jonathan Zalevsky

Cytokines are potent immune modulating agents but are not ideal medicines in their natural form due to their short half-life and pleiotropic systemic effects. NKTR-214 is a clinical-stage biologic that comprises interleukin-2 (IL2) protein bound by multiple releasable polyethylene glycol (PEG) chains. In this highly PEG-bound form, the IL2 is inactive; therefore, NKTR-214 is a biologic prodrug. When administered in vivo, the PEG chains slowly release, creating a cascade of increasingly active IL2 protein conjugates bound by fewer PEG chains. The 1-PEG-IL2 and 2-PEG-IL2 species derived from NKTR-214 are the most active conjugated-IL2 species. Free-IL2 protein is undetectable in vivo as it is eliminated faster than formed. The PEG chains on NKTR-214 are located at the region of IL2 that contacts the alpha (α) subunit of the heterotrimeric IL2 receptor complex, IL2Rαβγ, reducing its ability to bind and activate the heterotrimer. The IL2Rαβγ complex is constitutively expressed on regulatory T cells (Tregs). Therefore, without the use of mutations, PEGylation reduces the affinity for IL2Rαβγ to a greater extent than for IL2Rβγ, the receptor complex predominant on CD8 T cells. NKTR-214 treatment in vivo favors activation of CD8 T cells over Tregs in the tumor microenvironment to provide anti-tumor efficacy in multiple syngeneic models. Mechanistic modeling based on in vitro and in vivo kinetic data provides insight into the mechanism of NKTR-214 pharmacology. The model reveals that conjugated-IL2 protein derived from NKTR-214 occupy IL-2Rβγ to a greater extent compared to free-IL2 protein. The model accurately describes the sustained in vivo signaling observed after a single dose of NKTR-214 and explains how the properties of NKTR-214 impart a unique kinetically-controlled immunological mechanism of action.


Journal of Pharmacology and Experimental Therapeutics | 2017

NKTR-181: A Novel Mu-Opioid Analgesic with Inherently Low Abuse Potential

Takahiro Miyazaki; Irene Y. Choi; Werner Rubas; Neel K. Anand; Cherie F. Ali; Juli Evans; Hema Gursahani; Marlene Hennessy; Grace Kim; Daniel McWeeney; Juergen W. Pfeiffer; Phi Quach; David Gauvin; Timothy A. Riley; Jennifer A. Riggs; Kathleen Gogas; Jonathan Zalevsky; Stephen Doberstein

The increasing availability of prescription opioid analgesics for the treatment of pain has been paralleled by an epidemic of opioid misuse, diversion, and overdose. The development of abuse-deterrent formulations (ADFs) of conventional opioids such as oxycodone and morphine represents an advance in the field and has had a positive but insufficient impact, as most opioids are still prescribed in highly abusable, non-ADF forms, and abusers can tamper with ADF medications to liberate the abusable opioid within. The abuse liability of mu-opioid agonists appears to be dependent on their rapid rate of entry into the central nervous system (CNS), whereas analgesic activity appears to be a function of CNS exposure alone, suggesting that a new opioid agonist with an inherently low rate of influx across the blood-brain barrier could mediate analgesia with low abuse liability, regardless of formulation or route of administration. NKTR-181 is a novel, long-acting, selective mu-opioid agonist with structural properties that reduce its rate of entry across the blood-brain barrier compared with traditional mu-opioid agonists. NKTR-181 demonstrated maximum analgesic activity comparable to that of oxycodone in hot-plate latency and acetic-acid writhing models. NKTR-181 was distinguishable from oxycodone by its reduced abuse potential in self-administration and progressive-ratio break point models, with behavioral effects similar to those of saline, as well as reduced CNS side effects as measured by the modified Irwin test. The in vitro and in vivo studies presented here demonstrate that NKTR-181 is the first selective mu-opioid agonist to combine analgesic efficacy and reduced abuse liability through the alteration of brain-entry kinetics.


Journal of Clinical Oncology | 2016

Immune memory in nonclinical models after treatment with NKTR-214, an engineered cytokine biased towards expansion of CD8+ T cells in tumor.

Deborah H. Charych; Werner Rubas; Vidula Dixit; Janet Cetz; Rhoneil Pena; John L. Langowski; Ute Hoch; Murali Addepalli; Stephen Doberstein; Jonathan Zalevsky

11545Background: NKTR-214 is an immune-stimulatory cytokine designed to provide a controlled, sustained, and CD122-biased receptor signal through the heterodimeric IL2 receptor pathway (IL2Rbg) res...


Cancer immunology research | 2016

Abstract B015: Antitumor activity of the CD122-biased immunostimulatory cytokine combined with immune checkpoint blockade requires innate and adaptive immunity

John L. Langowski; Seema S. Kantak; Rhoneil Pena; Yolanda Kirksey; Murali Addepalli; Steve S. Lee; Ute Hoch; Deborah H. Charych; Stephen Doberstein

Background: While immune checkpoint blockade is a promising therapeutic approach, combination with agents that modulate complementary pathways may improve responses. Interleukin-2 (IL-2) immunotherapy leads to long-term responses in a small percentage of cancer patients, but systemic toxicity limits its use. In addition, IL-2 expands T regulatory cells, antagonizing antitumor immunity and resulting in a poorer clinical outcome. NKTR-214 is a novel CD122-biased immunostimulatory cytokine which combines biased activation of the IL-2R beta receptor subunit, greatly favoring activation of effector over regulatory T cells, with improved pharmacokinetics and tolerability compared to Proleukin in non-human models. Here we examine the efficacy and mechanism of NKTR-214 combined with anti-CTLA-4 in murine tumor models. Methods: Mice bearing subcutaneous EMT6 mammary tumors were treated with NKTR-214 q9d, murine anti-CTLA-4 or anti-PD-1 twice-weekly, or both in combination. Immune cell profiling was assessed by flow cytometry following treatment. CD8 or NK cells were depleted in vivo by serial anti-CD8 or anti-asialo-GM1 antibody injections, respectively. Antitumor memory and specificity was assessed in complete responders by challenging with EMT6 or CT26 colon carcinoma implants with no additional treatment. Results: While NKTR-214 and anti-CTLA-4 were not as efficacious individually, their combination was synergistic and well-tolerated with 83% of test animals tumor-free. Combination treatment increased NK and memory effector CD8 cells in both tumor and spleen. Antitumor immunity by the combination was durable and specific as 70% and 100% of mice remained tumor-free after challenge with a second and third EMT6 implant, but not after a subsequent CT26 implant. NKTR-214 combined with anti-PD-1 also proved synergistic with 40% of animals remaining tumor free following treatment. In vivo depletion of either CD8 effector or NK cells abrogated efficacy suggesting both contribute to the response. Conclusions: The mechanism of NKTR-214 antitumor immunity is complementary to checkpoint inhibition. Favorable pharmacokinetics of NKTR-214 allows sustained tumor exposure and dosing schedules commensurate with other therapies. This new therapeutic combination of NKTR-214 and checkpoint inhibition may similarly enable durable responses in humans. Citation Format: John L. Langowski, Seema S. Kantak, Rhoneil Pena, Yolanda Kirksey, Murali Addepalli, Steve Lee, Ute Hoch, Deborah H. Charych, Stephen K. Doberstein. Antitumor activity of the CD122-biased immunostimulatory cytokine combined with immune checkpoint blockade requires innate and adaptive immunity. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr B015.


Cancer immunology research | 2016

Abstract B057: The CD122-biased immunostimulatory cytokine NKTR-214 combined with checkpoint blockade leads to mobilization of antitumor immunity and synergistic activity

John L. Langowski; Murali Addepalli; Laurie VanderVeen; Rhoneil Pena; Ravikumar Nutakki; Yolanda Kirksey; Ute Hoch; Jonathan Zalevsky; Stephen Doberstein; Deborah H. Charych

Background: NKTR-214 is a novel immunostimulatory cytokine which provides a sustained and biased activation signal through the heterodimeric IL-2R receptor (IL2Rbg or CD122), resulting in significant expansion of CD8+ T and NK cells relative to regulatory T cells in the tumor microenvironment. As a single agent in preclinical tumor models, NKTR-214 significantly increases the intratumoral CD8T/Treg ratio (>400). Here we examine activity and pharmacodynamic effects of NKTR-214 alone and combined with immune checkpoint blockade in both a murine tumor model and in a human in vitro co-culture system. Methods: Mice bearing subcutaneous CT26 colon tumors were treated with single agent NKTR-214 (q9d), murine anti-CTLA-4 or anti-PD-1 (twice-weekly), or combinations of these agents. Serum cytokines were assessed by multiplex ELISA (Quansys). T cell clonality and infiltration were assessed in tumors 7 days after treatment initiation, with TCR V and J beta usage determined by ImmunoSEQ (Adaptive Biotechnologies). Effects on human cytokine and chemokine expression were examined by the BioMAP Combo ELECT (DiscoverX) in vitro system utilizing human fibroblasts or endothelial cells, PBMCs and either HT29 colon adenocarcinoma or HT1299 NSCLC cell lines. 1-PEG-IL2, the most active species of the NKTR-214 prodrug, or Keytruda (anti-PD-1) was added over a range of concentrations alone or in combination, with biomarkers assessed 48 hours later. Results: In tumor-bearing mice, NKTR-214 led to increases in serum IFNg as well as increases in the chemokine MCP-1 (CCL2), a chemoattractant of CD4 T, CD8 T and NK cells. Single-agent NKTR-214 led to significant tumor growth inhibition and 10% tumor free animals, while single-agents anti-CTLA-4 or anti-PD-1 lacked efficacy. Combination of NKTR-214 with checkpoint blockade was synergistic and led to greater tumor-free animals (67% in combination with anti-CTLA-4, 90% with anti-PD-1). NKTR-214 combined with anti-PD-1 was also more efficacious than the combination of anti-CTLA-4 and anti-PD-1. TCR repertoire analysis demonstrated superior increases in TIL clonality and infiltration with NKTR-214 compared to either anti-CTLA-4 or anti-PD-1 alone. NKTR-214 combined with either mode of checkpoint inhibition led to significant increases in both parameters, with the greatest effect occurring when combined with anti-PD-1. In the human BioMAP system, NKTR-214 active species led to significant increases in granzyme B, IFNg, IL-6, IL-17A and TNFa, while these markers were only modestly induced by Keytruda alone. When NKTR-214 was combined with Keytruda, additive and synergistic increases in these factors were found. In addition, the combination significantly decreased uPA in the fibroblast-HT29 system and MDC in the endothelial-HT1299 system, both factors associated with tumor progression. Conclusions: NKTR-214 delivers sustained signaling of the IL-2 pathway. Analysis of multiple immune markers in mice and human tumor-immune cell cultures provide mechanistic rationale for the increased T and NK cell activation in the tumor microenvironment after treatment with NKTR-214. When combined with anti-PD-1, significant increases in T cell receptor clonality, tumor infiltration and immune activation biomarkers are achieved - a significant finding, given that concomitant increase in clonality and infiltration has been correlated with favorable responses in the clinic. In vitro studies with human cells demonstrate the potentiation of immune activation markers associated with anti-tumor effects in combination with anti-PD-1, further suggesting these findings may be translatable from mouse to human. NKTR-214 is currently in a Phase 1 clinical trial to evaluate the pharmacokinetics, pharmacodynamics and activity in an outpatient setting. Citation Format: John L. Langowski, Murali Addepalli, Laurie VanderVeen, Rhoneil Pena, Ravikumar Nutakki, Yolanda Kirksey, Ute Hoch, Jonathan Zalevsky, Stephen K. Doberstein, Deborah H. Charych. The CD122-biased immunostimulatory cytokine NKTR-214 combined with checkpoint blockade leads to mobilization of antitumor immunity and synergistic activity [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr B057.


Cancer Research | 2016

Abstract 558: Durable antitumor activity of the CD122-biased immunostimulatory cytokine NKTR-214 combined with immune checkpoint blockade

John L. Langowski; Rhoneil Pena; Yolanda Kirksey; Murali Addepalli; Ute Hoch; Jonathan Zalevsky; Stephen Doberstein; Deborah H. Charych

Background: While immune checkpoint blockade is a promising therapeutic approach, combination with agents that modulate complementary pathways may improve responses. Interleukin-2 (IL-2) immunotherapy leads to long-term responses in a small percentage of cancer patients, but systemic toxicity limits its use. In addition, IL-2 expands T regulatory cells, antagonizing antitumor immunity. NKTR-214 is a novel CD122-biased immunostimulatory cytokine which combines biased activation of the IL-2R beta receptor, greatly favoring activation of effector over regulatory T cells in the tumor microenvironment, with improved pharmacokinetics and tolerability compared to Proleukin® (aldesleukin) in non-clinical models. Here we examine the efficacy and mechanism of NKTR-214 combined with checkpoint blocking antibodies in murine tumor models. Methods: Mice bearing subcutaneous B16, LLC, CT26 or EMT6 tumors were treated with single agent NKTR-214 (q9d), aldesleukin (bid or qd), murine anti-CTLA-4 or anti-PD-1 (twice-weekly), or combinations of these agents. Tumor volume was measured before, during and after treatment. Immune cells were profiled by flow cytometry; CD8 or NK requirements for efficacy were assessed by in vivo depletion through serial anti-CD8 or anti-asialo-GM1 antibody injections, respectively. Antitumor memory and specificity was assessed in complete responders by challenging with EMT6 or CT26 followed by observation with no additional test article treatments. Results: In the B16 model, superior single-agent efficacy was achieved by NKTR-214 with a 10-fold lower dose than aldesleukin, yet significantly increased CD8/Treg ratio in the tumor (>400). In the EMT6 model, while both aldesleukin and NKTR-214 synergized with anti-CTLA-4, a greater percentage of complete responders were consistently observed with NKTR-214 (73% versus 44%). Antitumor immunity induced by the combination required both NK and CD8 T cell activity, and was durable and specific with mice remaining tumor-free after re-challenge with an EMT6 but not CT26 implant. Finally, NKTR-214 combined with anti-PD-1 provided superior activity in the CT26 model compared with the combination of anti-CTLA-4 and anti-PD-1 (90% versus 60% tumor-free, respectively). Conclusions: NKTR-214 enables access to the potent IL-2 pathway, providing a mechanism of action complementary to checkpoint inhibition. Favorable pharmacokinetics of NKTR-214 allows sustained tumor exposure and dosing schedules commensurate with antibody therapies. The nonclinical results support the ongoing Phase 1 trial of NKTR-214 in patients with solid tumors. Citation Format: John L. Langowski, Rhoneil Pena, Yolanda M. Kirksey, Murali K. Addepalli, Ute Hoch, Jonathan Zalevsky, Stephen K. Doberstein, Deborah H. Charych. Durable antitumor activity of the CD122-biased immunostimulatory cytokine NKTR-214 combined with immune checkpoint blockade. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 558.

Collaboration


Dive into the Stephen Doberstein's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arthur J. Chirino

California Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge