Network


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

Hotspot


Dive into the research topics where Patrick Hagner is active.

Publication


Featured researches published by Patrick Hagner.


Blood | 2015

CC-122, a pleiotropic pathway modifier, mimics an interferon response and has antitumor activity in DLBCL

Patrick Hagner; Hon Wah Man; Maria Wang; Suzana Couto; Mike Breider; Chad C. Bjorklund; Courtney G. Havens; Lu G; Emily Rychak; Heather Raymon; Rama Krishna Narla; Leo Barnes; Gody Khambatta; Hsiling Chiu; Jolanta Kosek; Jian Kang; Amantangelo; Michelle Waldman; Antonia Lopez-Girona; Cai T; Michael Pourdehnad; Matthew Trotter; Tom Daniel; Peter H. Schafer; Anke Klippel; Anjan Thakurta; Rajesh Chopra; Anita Gandhi

Cereblon (CRBN), a substrate receptor of the Cullin 4 RING E3 ubiquitin ligase complex, is the target of the immunomodulatory drugs lenalidomide and pomalidomide. Recently, it was demonstrated that binding of these drugs to CRBN promotes the ubiquitination and subsequent degradation of 2 common substrates, transcription factors Aiolos and Ikaros. Here we report that CC-122, a new chemical entity termed pleiotropic pathway modifier, binds CRBN and promotes degradation of Aiolos and Ikaros in diffuse large B-cell lymphoma (DLBCL) and T cells in vitro, in vivo, and in patients, resulting in both cell autonomous as well as immunostimulatory effects. In DLBCL cell lines, CC-122-induced degradation or short hairpin RNA-mediated knockdown of Aiolos and Ikaros correlates with increased transcription of interferon (IFN)-stimulated genes independent of IFN-α, -β, and -γ production and/or secretion and results in apoptosis in both activated B-cell (ABC) and germinal center B-cell DLBCL cell lines. Our results provide mechanistic insight into the cell-of-origin independent antilymphoma activity of CC-122, in contrast to the ABC subtype selective activity of lenalidomide.


Clinical Cancer Research | 2017

A phase 2/3 multicenter, randomized, open-label study to compare the efficacy and safety of lenalidomide versus investigator's choice in patients with relapsed or refractory diffuse large B-cell lymphoma

Myron S. Czuczman; Marek Trněný; Andrew Davies; Simon Rule; Kim Linton; Nina D. Wagner-Johnston; Randy D. Gascoyne; Graham W. Slack; Pierre Brousset; David A. Eberhard; Francisco J. Hernandez-Ilizaliturri; Gilles Salles; Thomas E. Witzig; Pier Luigi Zinzani; George W. Wright; Louis M. Staudt; Yandan Yang; P. Mickey Williams; Chih Jian Lih; Jacqueline Russo; Anjan Thakurta; Patrick Hagner; Pierre Fustier; Dale Song; Ian D. Lewis

Purpose: Randomized, multicenter, open-label, phase 2/3 trial investigating lenalidomide versus investigators choice (IC) in relapsed/refractory diffuse large B-cell lymphoma (DLBCL). Experimental Design: Patients with DLBCL who received ≥2 prior therapies were stratified by DLBCL subtype [germinal center B-cell (GCB) vs. non-GCB; determined by immunohistochemistry (IHC)] and then randomized 1:1 to lenalidomide (25 mg/day, 21 days of 28-day cycle) or IC (gemcitabine, rituximab, etoposide, or oxaliplatin). Crossover to lenalidomide was permitted for IC-treated patients with radiologically confirmed progressive disease. The primary endpoint was overall response rate (ORR). Progression-free survival (PFS), overall survival, and subtype analysis [GCB vs. activated B-cell (ABC)] using gene expression profiling (GEP) were exploratory endpoints. Results: Stage 1: 102 DLBCL patients (by IHC: non-GCB, n = 54; GCB, n = 48) received ≥1 dose of lenalidomide or IC. Hematologic treatment-emergent adverse events with lenalidomide versus IC included neutropenia (42.6%; 36.4%), anemia (33.3%; 47.3%), thrombocytopenia (24.1%; 43.6%), and leukopenia (5.6%; 12.7%), respectively. Overall, lenalidomide-treated patients had an ORR of 27.5% versus 11.8% in IC (ORRs were similar regardless of IHC-defined DLBCL subtype). Median PFS was increased in patients receiving lenalidomide (13.6 weeks) versus IC (7.9 weeks; P = 0.041), with greater improvements in non-GCB patients (15.1 vs. 7.1 weeks, respectively; P = 0.021) compared with GCB (10.1 vs. 9.0 weeks, respectively; P = 0.550). Conclusions: The clinical benefit of lenalidomide monotherapy in DLBCL patients was more evident in the non-GCB subtype. Exploratory analyses suggest that this preferential benefit was more pronounced in the GEP-defined ABC population, demonstrating a need for additional studies of lenalidomide in DLBCL using GEP subtyping. Clin Cancer Res; 23(15); 4127–37. ©2017 AACR.


British Journal of Haematology | 2017

Activity of lenalidomide in mantle cell lymphoma can be explained by NK cell‐mediated cytotoxicity

Patrick Hagner; Hsiling Chiu; Maria Ortiz; Benedetta Apollonio; Maria Wang; Suzana Couto; Michelle Waldman; Erin Flynt; Alan G. Ramsay; Matthew Trotter; Anita Gandhi; Rajesh Chopra; Anjan Thakurta

Lenalidomide is an immunomodulatory agent that has demonstrated clinical benefit for patients with relapsed or refractory mantle cell lymphoma (MCL); however, despite this observed clinical activity, the mechanism of action (MOA) of lenalidomide has not been characterized in this setting. We investigated the MOA of lenalidomide in clinical samples from patients enrolled in the CC‐5013‐MCL‐002 trial (NCT00875667) comparing single‐agent lenalidomide versus investigators choice single‐agent therapy and validated our findings in pre‐clinical models of MCL. Our results revealed a significant increase in natural killer (NK) cells relative to total lymphocytes in lenalidomide responders compared to non‐responders that was associated with a trend towards prolonged progression‐free survival and overall survival. Clinical response to lenalidomide was independent of baseline tumour microenvironment expression of its molecular target, cereblon, as well as genetic mutations reported to impact clinical response to the Bruton tyrosine kinase inhibitor ibrutinib. Preclinical experiments revealed lenalidomide enhanced NK cell‐mediated cytotoxicity against MCL cells via increased lytic immunological synapse formation and secretion of granzyme B. In contrast, lenalidomide exhibited minimal direct cytotoxic effects against MCL cells. Taken together, these data provide the first insight into the clinical activity of lenalidomide against MCL, revealing a predominately immune‐mediated MOA.


Archive | 2014

METHODS FOR DETERMINING DRUG EFFICACY FOR THE TREATMENT OF DIFFUSE LARGE B-CELL LYMPHOMA, MULTIPLE MYELOMA, AND MYELOID CANCERS

Matthew William Burnell Trotter; Patrick Hagner; Courtney G. Havens; Rajesh Chopra; Anita Gandhi; Anke Klippel; Maria Yinglin Wang; Mike Breider; Suzana Couto; Yan Ren; Paul Hollenbach; Kyle J. MacBeth


Blood | 2014

CC-122 Degrades the Lymphoid Transcription Factor Aiolos (IKZF3) By Modulating Cereblon and Shows Clinical Activity in a Phase Ib Study of Subjects with Relapsed or Refractory Non-Hodgkin’s Lymphoma and Multiple Myeloma

Vincent Ribrag; Silvia Damien; Mecide Gharibo; Mercede Gironella; Armando Santoro; Drew W Rasco; William J. Edenfield; Xin Wei; Angela James; Patrick Hagner; Anita Gandhi; Rajesh Chopra; Jorge DiMartino; Michael Pourdehnad; Anne-Marie Stoppa


Archive | 2016

METHODS FOR TREATING SOLID TUMORS AND THE USE OF BIOMARKERS AS A PREDICTOR OF CLINICAL SENSITIVITY TO IMMUNOMODULATORY THERAPIES

Patrick Hagner; Anita Gandhi; Rajesh Chopra; Anke Klippel


Blood | 2016

CC-122 Is a Cereblon Modulating Agent That Is Active in Lenalidomide-Resistant and Lenalidomide/Dexamethasone-Double-Resistant Multiple Myeloma Pre-Clinical Models

Chad C. Bjorklund; Jian Kang; Ling Lu; Michael Amatangelo; Hsiling Chiu; Patrick Hagner; Anita Gandhi; Michael Pourdehnad; Anke Klippel; Anjan Thakurta


Blood | 2015

CC-122 Exhibits Potent Anti-Lymphoma Activity in Combination with Obinutuzumab through Cell Autonomous and Antibody Dependent Cell Mediated Cytotoxicity

Hsiling Chiu; Patrick Hagner; Michael Pourdehnad; Thomas O. Daniel; Rajesh Chopra; Anke Klippel; Christian Klein; Anjan Thakurta; Anita Gandhi


Blood | 2016

CC-122 Exhibits Greater Preclinical Activity in Mantle Cell Lymphoma Than Lenalidomide through a Combination of Direct Cell-Autonomous and Increased Antibody Dependent Cell-Mediated Cytotoxicity

Patrick Hagner; Hsiling Chiu; Michelle Waldman; Anke Klippel; Michael Pourdehnad; Anita Gandhi; Anjan Thakurta


Blood | 2016

A Phase 1b, Multi-Center, Open-Label Study of Novel Combinations of CC-122, CC-223, CC-292, and Rituximab in Diffuse Large B-Cell Lymphoma: CC-122-DLBCL-001

Vincent Ribrag; Julio C. Chavez; Jason Kaplan; Umberto Vitolo; Jason C. Chandler; Armando Santoro; Paolo Corradini; Philippe Cassier; Ian W. Flinn; Ranjana H. Advani; Randeep Sangha; Iris Isufi; Vaishalee P. Kenkre; Patrick Hagner; Torsten Trowe; Anita Gandhi; Xiaoling Wu; Kristen Hege; Michael Pourdehnad; John Kuruvilla

Collaboration


Dive into the Patrick Hagner's collaboration.

Researchain Logo
Decentralizing Knowledge