Network


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

Hotspot


Dive into the research topics where David Geho is active.

Publication


Featured researches published by David Geho.


Lancet Oncology | 2012

Effect of the MDM2 antagonist RG7112 on the P53 pathway in patients with MDM2-amplified, well-differentiated or dedifferentiated liposarcoma: an exploratory proof-of-mechanism study

Isabelle Ray-Coquard; Jean-Yves Blay; Antoine Italiano; Axel Le Cesne; Nicolas Penel; Jianguo Zhi; Florian Heil; Ruediger Rueger; Bradford Graves; Meichun Ding; David Geho; Steven Middleton; Lyubomir T. Vassilev; Gwen Nichols; Binh Bui

BACKGROUND We report a proof-of-mechanism study of RG7112, a small-molecule MDM2 antagonist, in patients with chemotherapy-naive primary or relapsed well-differentiated or dedifferentiated MDM2-amplified liposarcoma who were eligible for resection. METHODS Patients with well-differentiated or dedifferentiated liposarcoma were enrolled at four centres in France. Patients received up to three 28-day neoadjuvant treatment cycles of RG7112 1440 mg/m(2) per day for 10 days. If a patient progressed at any point after the first cycle, the lesion was resected or, if unresectable, an end-of-study biopsy was done. The primary endpoint was to assess markers of RG7112-dependent MDM2 inhibition and P53 pathway activation (P53, P21, MDM2, Ki-67, macrophage inhibitory cytokine-1 [MIC-1], and apoptosis). All analyses were per protocol. This trial is registered with EudraCT, number 2009-015522-10. RESULTS Between June 3, and Dec 14, 2010, 20 patients were enrolled and completed pretreatment and day 8 biopsies. 18 of 20 patients had TP53 wild-type tumours and two carried missense TP53 mutations. 14 of 17 assessed patients had MDM2 gene amplification. Compared with baseline, P53 and P21 concentrations, assessed by immunohistochemistry, had increased by a median of 4·86 times (IQR 4·38-7·97; p=0·0001) and 3·48 times (2·05-4·09; p=0·0001), respectively, at day 8 (give or take 2 days). At the same timepoint, relative MDM2 mRNA expression had increased by a median of 3·03 times (1·23-4·93; p=0·003) that at baseline. The median change from baseline for Ki-67-positive tumour cells was -5·05% (IQR -12·55 to 0·05; p=0·01). Drug exposure correlated with blood concentrations of MIC-1 (p<0·0001) and haematological toxicity. One patient had a confirmed partial response and 14 had stable disease. All patients experienced at least one adverse event, mostly nausea (14 patients), vomiting (11 patients), asthenia (nine patients), diarrhoea (nine patients), and thrombocytopenia (eight patients). There were 12 serious adverse events in eight patients, the most common of which were neutropenia (six patients) and thrombocytopenia (three patients). DISCUSSION MDM2 inhibition activates the P53 pathway and decreases cell proliferation in MDM2-amplified liposarcoma. This study suggests that it is feasible to undertake neoadjuvant biopsy-driven biomarker studies in liposarcoma. FUNDING F Hoffmann-La Roche.


Clinical Cancer Research | 2015

A Phase I Monotherapy Study of RG7212, a First-in-Class Monoclonal Antibody Targeting TWEAK Signaling in Patients with Advanced Cancers

Ulrik Niels Lassen; Didier Meulendijks; Lilian L Siu; Vaios Karanikas; Morten Mau-Sorensen; Jan H. M. Schellens; Derek J. Jonker; Aaron Richard Hansen; Mary Ellen Simcox; Kathleen Schostack; Dean Bottino; Hua Zhong; Markus Roessler; Suzana Vega-Harring; Tiantom Jarutat; David Geho; Ka Wang; Mark DeMario; Glenwood D. Goss

Purpose: Tumor necrosis factor (TNF)–like weak inducer of apoptosis (TWEAK) and fibroblast growth factor-inducible molecule 14 (Fn14) are a ligand–receptor pair frequently overexpressed in solid tumors. TWEAK:Fn14 signaling regulates multiple oncogenic processes through MAPK, AKT, and NFκB pathway activation. A phase I study of RG7212, a humanized anti-TWEAK IgG1κ monoclonal antibody, was conducted in patients with advanced solid tumors expressing Fn14. Experimental Design: Dose escalations, over a 200- to 7,200-mg range, were performed with patients enrolled in weekly (QW), bi-weekly (Q2W), or every-three-week (Q3W) schedules. Primary objectives included determination of dose and safety profile. Secondary endpoints included assessments related to inhibition of TWEAK:Fn14 signaling, tumor proliferation, tumor immune cell infiltration, and pharmacokinetics. Results: In 192 treatment cycles administered to 54 patients, RG7212 was well-tolerated with no dose-limiting toxicities observed. More than 95% of related adverse events were limited to grade 1/2. Pharmacokinetics were dose proportional for all cohorts, with a t1/2 of 11 to 12 days. Pharmacodynamic changes included clearance of free and total TWEAK ligand and reductions in tumor Ki-67 and TRAF1. A patient with BRAF wild-type melanoma who received 36 weeks of RG7212 therapy had tumor regression and pharmacodynamic changes consistent with antitumor effects. Fifteen patients (28%) received 16 or more weeks of RG7212 treatment. Conclusion: RG7212 demonstrated excellent tolerability and favorable pharmacokinetics. Pharmacodynamic endpoints were consistent with reduced TWEAK:Fn14 signaling. Tumor regression was observed and prolonged stable disease was demonstrated in multiple heavily pretreated patients with solid tumors. These encouraging results support further study of RG7212. Clin Cancer Res; 21(2); 258–66. ©2014 AACR.


Clinical Cancer Research | 2013

RG7212 anti-TWEAK mAb inhibits tumor growth through inhibition of tumor cell proliferation and survival signaling and by enhancing the host antitumor immune response.

Xuefeng Yin; Leopoldo Luistro; Hua Zhong; Melissa Smith; Tom Nevins; Kathleen Schostack; Holly Hilton; Tai-An Lin; Theresa Truitt; Denise Biondi; Xiaoqian Wang; Kathryn Packman; Jim Rosinski; Windy Berkofsky-Fessler; Jian-Ping Tang; Saumya Pant; David Geho; Suzana Vega-Harring; Mark DeMario; Hy Levitsky; Mary Ellen Simcox

Purpose: To explore the role of TWEAK in tumor growth and antitumor immune response and the activity and mechanism of RG7212, an antagonistic anti-TWEAK antibody, in tumor models. Experimental Design: TWEAK-induced signaling and gene expression were explored in tumor cell lines and inhibition of these effects and antitumor efficacy with RG7212 treatment was assessed in human tumor xenograft-, patient-derived xenograft, and syngeneic tumor models and phase I patients. Genetic features correlated with antitumor activity were characterized. Results: In tumor cell lines, TWEAK induces proliferation, survival, and NF-κB signaling and gene expression that promote tumor growth and suppress antitumor immune responses. TWEAK-inducible CD274, CCL2, CXCL-10 and -11 modulate T-cell and monocyte recruitment, T-cell activation, and macrophage differentiation. These factors and TWEAK-induced signaling were decreased, and tumor, blood, and spleen immune cell composition was altered with RG7212 treatment in mice. RG7212 inhibits tumor growth in vivo in models with TWEAK receptor, Fn14, expression, and markers of pathway activation. In phase I testing, signs of tumor shrinkage and stable disease were observed without dose-limiting toxicity. In a patient with advanced, Fn14-positive, malignant melanoma with evidence of tumor regression, proliferation markers were dramatically reduced, tumor T-cell infiltration increased, and tumor macrophage content decreased. Antitumor activity, a lack of toxicity in humans and animals and no evidence of antagonism with standard of care or targeted agents in mice, suggests that RG7212 is a promising agent for use in combination therapies in patients with Fn14-positive tumors. Clin Cancer Res; 19(20); 5686–98. ©2013 AACR.


British Journal of Haematology | 2015

MDM2 antagonist clinical response association with a gene expression signature in acute myeloid leukaemia.

Hua Zhong; Gong Chen; Lori Jukofsky; David Geho; Sung Won Han; Fabian Birzele; Sabine Bader; Lucia Himmelein; James Cai; Zayed Albertyn; Mark Rothe; Laurent Essioux; Helmut Burtscher; Steven Middleton; Ruediger Rueger; Lin-Chi Chen; Markus Dangl; Gwen Nichols; William E. Pierceall

Acute myeloid leukaemia (AML) is uniquely sensitive to p53 activation 1, 2 as ≈90% of patients carry wild-type TP53 and frequent MDM2 overexpression.3 MDM2 blocks p53 transactivation and targets p53 for ubiquitin-dependent degradation.4, 5 Nutlins have been characterized as potent and selective small-molecule MDM2 antagonists.1, 6–8 RG7112 was the first such MDM2 antagonist to undergo clinical assessment in solid tumors and leukaemia trials.1, 2, 9 As not all patients with functional p53 will respond to MDM2 antagonists, diagnostic tools may identify patients likely to respond.


Clinical Cancer Research | 2016

Exposure and Tumor Fn14 expression as Determinants of Pharmacodynamics of the Anti-TWEAK Monoclonal Antibody RG7212 in Patients with Fn14-positive Solid Tumors

Didier Meulendijks; Ulrik Niels Lassen; Lillian L. Siu; Alwin D. R. Huitema; Vaios Karanikas; Morten Mau-Sorensen; Derek J. Jonker; Aaron Richard Hansen; Mary Ellen Simcox; Kathleen Schostack; Dean Bottino; Hua Zhong; Markus Roessler; Suzana Vega-Harring; Tiantom Jarutat; David Geho; Karen Wang; Mark DeMario; Glenwood D. Goss; Jan H. M. Schellens

Purpose: The TWEAK–Fn14 pathway represents a novel anticancer target that is being actively investigated. Understanding the relationship between pharmacokinetics of anti-TWEAK therapeutics and tumor pharmacodynamics is critical. We investigated exposure-response relationships of RG7212, an anti-TWEAK mAb, in patients with Fn14-expressing tumors. Experimental Design: Patients with Fn14-positive tumors (IHC≥1+) treated in a phase I first-in-human study with ascending doses of RG7212 were the basis for this analysis. Pharmacokinetics of RG7212 and dynamics of TWEAK were determined, as were changes in tumor TWEAK–Fn14 signaling in paired pre- and posttreatment tumor biopsies. The objectives of the analysis were to define exposure-response relationships and the relationship between pretreatment tumor Fn14 expression and pharmacodynamic effect. Associations between changes in TWEAK–Fn14 signaling and clinical outcome were explored. Results: Thirty-six patients were included in the analysis. RG7212 reduced plasma TWEAK to undetectable levels at all observed RG7212 exposures. In contrast, reductions in tumor Fn14 and TRAF1 protein expression were observed only at higher exposure (≥300 mg*h/mL). Significant reductions in tumor Ki-67 expression and early changes in serum concentrations of CCL-2 and MMP-9 were observed exclusively in patients with higher drug exposure who had high pretreatment tumor Fn14 expression. Pretreatment tumor Fn14 expression was not associated with outcome, but a trend toward longer time on study was observed with high versus low RG7212 exposure. Conclusions: RG7212 reduced tumor TWEAK–Fn14 signaling in a systemic exposure-dependent manner. In addition to higher exposure, relatively high Fn14 expression might be required for pharmacodynamic effect of anti-TWEAK monoclonal antibodies. Clin Cancer Res; 22(4); 858–67. ©2015 AACR.


Cancer Research | 2015

Abstract 2835: MDM2 antagonist-based therapeutic response is discriminated by a 4-gene signature in acute myeloid leukemia patients

Hua Zhong; Gong Chen; Lori Jukofsky; David Geho; Sung Won Han; Fabian Birzele; Sabine Bader; Lucia Himmelein; James Cai; Zayed Albertyn; Mark Rothe; Laurent Essioux; Helmut Burtscher; Steven Middleton; Lin-Chi Chen; Markus Dangl; William E. Pierceall; Gwen Nichols

The activity of p53, a key tumor suppressor is tightly controlled by MDM2-mediated ubiquination and degradation. Nutlins, a class of small-molecule MDM2 antagonists, have been characterized as drivers of p53 re-activation. Acute myeloid leukemia (AML) is uniquely sensitive to p53 re-activation as ∼90% of cases have wild-type TP53 and frequent MDM2 overexpression to overcome mechanisms of oncogene addiction. Personalized theranostic strategies may distinguish patients likely to clinically benefit from MDM2-antagonist therapy. Association between MDM2 antagonist (RG7112) growth inhibition (IC50s) in 287 human cancer cell lines (Cell Lines for Oncology/Chugai Accumulative Tumor Encyclopedia), and pretreatment RNAseq profiling established a classifier comprising MDM2, XPC, BBC3, and CDKN2A. This signature significantly associated with cell-line efficacy to MDM2 antagonist (odds ratio = 2.53; P RG7112 treatment was assessed in a phase 1 dose escalation trial in relapsed/refractory AML patients (NO21279). Signature scores of AML patient blood specimens at baseline significantly associated with clinical response (PD In summary, we demonstrate that a biological classifier discriminates response broadly to MDM2-antagonist therapy. The level of evidence attained by cell line efficacy modeling and response assessments in trial NO21279 (with MDM2 antagonist RG7112) and now in trial NP28679 (with MDM2 antagonist RG7388) adds substantial weight to the validity of this panel. Citation Format: Hua Zhong, Gong Chen, Lori Jukofsky, David Geho, Sung Won Han, Fabian Birzele, Sabine Bader, Lucia Himmelein, James Cai, Zayed Albertyn, Mark Rothe, Laurent Essioux, Helmut Burtscher, Steven A. Middleton, Lin-Chi Chen, Markus Dangl, William E. Pierceall, Gwen Nichols. MDM2 antagonist-based therapeutic response is discriminated by a 4-gene signature in acute myeloid leukemia patients. [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 2835. doi:10.1158/1538-7445.AM2015-2835


Blood | 2012

Results of the Phase 1 Trial of RG7112, a Small-Molecule MDM2 Antagonist, in Acute Leukemia

Michael Andreeff; Kevin R. Kelly; Karen Yee; Sarit Assouline; Roger Strair; Leslie Popplewell; David G. Bowen; Giovanni Martinelli; Mark W. Drummond; Paresh Vyas; Mark Kirschbaum; Swaminathan Padmanabhan Iyer; Kensuke Kojima; David Geho; Steven Blotner; Suzanne Cheng; Lyubomir T. Vassilev; Meichun Ding; Jianguo Zhi; Steven Middleton; Gwen Nichols


Archive | 2016

MRNA-BASED GENE EXPRESSION FOR PERSONALIZING PATIENT CANCER THERAPY WITH AN MDM2 ANTAGONIST

Hua Zhong; David Geho; Gong Chen; Gwen Nichols; Markus Dangl


Journal of Clinical Oncology | 2017

Phase Ib study of RG7112 with doxorubicin (D) in advanced soft tissue sarcoma (ASTS).

Sant P. Chawla; Jean-Yves Blay; Antoine Italiano; Martin Gutierrez; Axel Le Cesne; Carlos Gomez-Roca; Launce Gouw; Margaret von Mehren; Andrew J. Wagner; Robert G. Maki; Brian Higgins; Steven Middleton; Gwen Nichols; David Geho; Steven Blotner; Jianguo Zhi; Lin-Chi Chen


Journal of Clinical Oncology | 2017

A phase I study of MDM2 antagonist RG7112 in patients (pts) with relapsed/refractory solid tumors.

Razelle Kurzrock; Jean-Yves Blay; Binh Bui Nguyen; Andrew J. Wagner; Robert G. Maki; Gary K. Schwartz; Amita Patnaik; Lia Gore; Lin Wu; Lyubomir T. Vassilev; Michelle Ding; David Geho; Jianguo Zhi; Steven Middleton; Gwen Nichols

Collaboration


Dive into the David Geho'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

Antoine Italiano

Argonne National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge