Network


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

Hotspot


Dive into the research topics where Warren Denning is active.

Publication


Featured researches published by Warren Denning.


Clinical Cancer Research | 2016

Epithelial-Mesenchymal Transition Is Associated with a Distinct Tumor Microenvironment Including Elevation of Inflammatory Signals and Multiple Immune Checkpoints in Lung Adenocarcinoma.

Yanyan Lou; Lixia Diao; Edwin Roger Parra Cuentas; Warren Denning; Limo Chen; You Hong Fan; Lauren Averett Byers; Jing Wang; Vassiliki Papadimitrakopoulou; Carmen Behrens; Jaime Rodriguez; Patrick Hwu; Ignacio I. Wistuba; John V. Heymach; Don L. Gibbons

Purpose: Promising results in the treatment of non–small cell lung cancer (NSCLC) have been seen with agents targeting immune checkpoints, such as programmed cell death 1 (PD-1) or programmed death ligand-1 (PD-L1). However, only a select group of patients respond to these interventions. The identification of biomarkers that predict clinical benefit to immune checkpoint blockade is critical to successful clinical translation of these agents. Methods: We conducted an integrated analysis of three independent large datasets, including The Cancer Genome Atlas of lung adenocarcinoma and two datasets from MD Anderson Cancer Center (Houston, TX), Profiling of Resistance Patterns and Oncogenic Signaling Pathways in Evaluation of Cancers of the Thorax (named PROSPECT) and Biomarker-Integrated Approaches of Targeted Therapy for Lung Cancer Elimination (named BATTLE-1). Comprehensive analysis of mRNA gene expression, reverse-phase protein array, IHC, and correlation with clinical data were performed. Results: Epithelial–mesenchymal transition (EMT) is highly associated with an inflammatory tumor microenvironment in lung adenocarcinoma, independent of tumor mutational burden. We found immune activation coexistent with elevation of multiple targetable immune checkpoint molecules, including PD-L1, PD-L2, PD-1, TIM-3, B7-H3, BTLA, and CTLA-4, along with increases in tumor infiltration by CD4+Foxp3+ regulatory T cells in lung adenocarcinomas that displayed an EMT phenotype. Furthermore, we identify B7-H3 as a prognostic marker for NSCLC. Conclusions: The strong association between EMT status and an inflammatory tumor microenvironment with elevation of multiple targetable immune checkpoint molecules warrants further investigation of using EMT as a predictive biomarker for immune checkpoint blockade agents and other immunotherapies in NSCLC and possibly a broad range of other cancers. Clin Cancer Res; 22(14); 3630–42. ©2016 AACR. See related commentary by Datar and Schalper, p. 3422


Cancer Research | 2016

STK11/LKB1 Deficiency Promotes Neutrophil Recruitment and Proinflammatory Cytokine Production to Suppress T-cell Activity in the Lung Tumor Microenvironment

Shohei Koyama; Esra A. Akbay; Yvonne Y. Li; Amir R. Aref; Ferdinandos Skoulidis; Grit S. Herter-Sprie; Kevin A. Buczkowski; Yan Liu; Mark M. Awad; Warren Denning; Lixia Diao; Jing Wang; Edwin R. Parra-Cuentas; Ignacio I. Wistuba; Margaret Soucheray; Tran C. Thai; Hajime Asahina; Shunsuke Kitajima; Abigail Altabef; Jillian D. Cavanaugh; Kevin Rhee; Peng Gao; Haikuo Zhang; Peter E. Fecci; Takeshi Shimamura; Matthew D. Hellmann; John V. Heymach; F. Stephen Hodi; Gordon J. Freeman; David A. Barbie

STK11/LKB1 is among the most commonly inactivated tumor suppressors in non-small cell lung cancer (NSCLC), especially in tumors harboring KRAS mutations. Many oncogenes promote immune escape, undermining the effectiveness of immunotherapies, but it is unclear whether the inactivation of tumor suppressor genes, such as STK11/LKB1, exerts similar effects. In this study, we investigated the consequences of STK11/LKB1 loss on the immune microenvironment in a mouse model of KRAS-driven NSCLC. Genetic ablation of STK11/LKB1 resulted in accumulation of neutrophils with T-cell-suppressive effects, along with a corresponding increase in the expression of T-cell exhaustion markers and tumor-promoting cytokines. The number of tumor-infiltrating lymphocytes was also reduced in LKB1-deficient mouse and human tumors. Furthermore, STK11/LKB1-inactivating mutations were associated with reduced expression of PD-1 ligand PD-L1 in mouse and patient tumors as well as in tumor-derived cell lines. Consistent with these results, PD-1-targeting antibodies were ineffective against Lkb1-deficient tumors. In contrast, treating Lkb1-deficient mice with an IL6-neutralizing antibody or a neutrophil-depleting antibody yielded therapeutic benefits associated with reduced neutrophil accumulation and proinflammatory cytokine expression. Our findings illustrate how tumor suppressor mutations can modulate the immune milieu of the tumor microenvironment, and they offer specific implications for addressing STK11/LKB1-mutated tumors with PD-1-targeting antibody therapies.


Cancer Discovery | 2018

STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma

Ferdinandos Skoulidis; Michael E. Goldberg; Danielle Greenawalt; Matthew D. Hellmann; Mark M. Awad; Justin F. Gainor; Alexa B. Schrock; Ryan J. Hartmaier; Sally E. Trabucco; Siraj M. Ali; Julia A. Elvin; Gaurav Singal; Jeffrey S. Ross; David Fabrizio; Peter Szabo; Han Chang; Ariella Sasson; Sujaya Srinivasan; Stefan Kirov; Joseph D. Szustakowski; Patrik Vitazka; Robin Edwards; Jose A. Bufill; Neelesh Sharma; Sai-Hong Ignatius Ou; Nir Peled; David R. Spigel; Hira Rizvi; Elizabeth Jimenez Aguilar; Brett W. Carter

KRAS is the most common oncogenic driver in lung adenocarcinoma (LUAC). We previously reported that STK11/LKB1 (KL) or TP53 (KP) comutations define distinct subgroups of KRAS-mutant LUAC. Here, we examine the efficacy of PD-1 inhibitors in these subgroups. Objective response rates to PD-1 blockade differed significantly among KL (7.4%), KP (35.7%), and K-only (28.6%) subgroups (P < 0.001) in the Stand Up To Cancer (SU2C) cohort (174 patients) with KRAS-mutant LUAC and in patients treated with nivolumab in the CheckMate-057 phase III trial (0% vs. 57.1% vs. 18.2%; P = 0.047). In the SU2C cohort, KL LUAC exhibited shorter progression-free (P < 0.001) and overall (P = 0.0015) survival compared with KRASMUT;STK11/LKB1WT LUAC. Among 924 LUACs, STK11/LKB1 alterations were the only marker significantly associated with PD-L1 negativity in TMBIntermediate/High LUAC. The impact of STK11/LKB1 alterations on clinical outcomes with PD-1/PD-L1 inhibitors extended to PD-L1-positive non-small cell lung cancer. In Kras-mutant murine LUAC models, Stk11/Lkb1 loss promoted PD-1/PD-L1 inhibitor resistance, suggesting a causal role. Our results identify STK11/LKB1 alterations as a major driver of primary resistance to PD-1 blockade in KRAS-mutant LUAC.Significance: This work identifies STK11/LKB1 alterations as the most prevalent genomic driver of primary resistance to PD-1 axis inhibitors in KRAS-mutant lung adenocarcinoma. Genomic profiling may enhance the predictive utility of PD-L1 expression and tumor mutation burden and facilitate establishment of personalized combination immunotherapy approaches for genomically defined LUAC subsets. Cancer Discov; 8(7); 822-35. ©2018 AACR.See related commentary by Etxeberria et al., p. 794This article is highlighted in the In This Issue feature, p. 781.


Cancer Research | 2016

Abstract 4137: Loss of LKB1 mediates an immune inert phenotype in human lung adenocarcinoma

Warren Denning; Ferdinandos Skoulidis; Li Shen; Vassiliki Papadimitrakopoulou; Lixia Diao; Yanyan Lou; Lauren Averett Byers; Jing Wang; Jaime Rodriguez Canales; Ignacio I. Wistuba; John N. Weinstein; Don L. Gibbons; John V. Heymach

Background: We recently have reported that co-mutations within KRAS-mutated lung adenocarcinoma (LUAD) define a distinct biological phenotype and may offer insights into therapeutic targeting using this classification system. The analysis revealed striking differences in checkpoint ligand expression and T cell infiltration between TP53 and STK11 (LKB1) co-mutated KRAS LUAD. We, therefore, hypothesized that loss of LKB1promotes an immune-suppressed phenotype in KRAS LUAD. To test this, we analyzed 100 immune-related genes from The Cancer Genome Atlas datasets, as well as three other sets of resected tumors from NSLC patients from MD Anderson Cancer Center. Results: Our results revealed that tumors lacking LKB1 (LKB1-loss), as defined by mutation or gene-expression signature, resulted in a down regulation of: tumor associated immunosuppressive ligands, PD-L1, PD-L2, B7-H3, B7-H4, HVEM; T cell associated immunosuppressive ligands PD-1, CTLA-4, BTLA; co-stimulation ligands such as CD80, -86, -40L, and -40. We further examined if this loss of immunosuppression and co-stimulation resulted in changes in markers of T cell phenotypes. Indeed, tumors lacking LKB1-loss tumors demonstrated a distinct lack absence of several T cell markers including CD3, -4, & -8, but also RORC, T-bet, GATA-3, and FoxP3. To determine if myeloid cell populations, known mediators of immune suppression, were also diminished in LKB1-loss tumors, we analyzed expression of CD11c, -103, -11b, -68, and CD33. Consistent with T cell makers and suppressive ligand markers, myeloid markers were down regulated in LKB1-loss tumors. Because common immune suppression markers were lost, we then examined the expression of antigen presentation (HLA-I, HLA-II) and amino acid transporters as possible alternative methods for immune suppression. In LKB1-loss tumors, both HLA-I and HLA-II markers were down regulated, compared to tumors with intact LKB1 expression. Furthermore, genes associated with the antigen presentation complex, LMP2, TAP1&2, and B2M were also down regulated. In contrast to the immune markers described above, cationic transporters (SLC7A2), anionic transporters (SLC7A11), and neutral transporters (SLC3A2) were universally upregulated in LKB1-loss tumors. Conclusions: LKB1 loss is associated with an inert immune phenotype. Our data suggests that reduced antigen presentation and amino acid depletion, but not checkpoint factors or MDSCs, are likely mediator of this phenotype. Citation Format: Warren L. Denning, Ferdinandos Skoulidis, Li Shen, Vassiliki Papadimitrakopoulou, Lixia Diao, Yanyan Lou, Lauren A. Byers, Jing Wang, Jaime R. Canales, Ignacio I. Wistuba, John Weinstein, Don L. Gibbons, John V. Heymach. Loss of LKB1 mediates an immune inert phenotype in human lung adenocarcinoma. [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 4137.


Cancer Research | 2015

Abstract 3364: Epithelial-mesenchymal transition is associated with a profound inflammatory tumor microenvironment in lung adenocarcinoma

Yanyan Lou; Lixia Diao; Parra Cuentas Edwin Roger; Warren Denning; Limo Chen; Youhong Fan; Jaime Rodriguez; Lauren Averett Byers; Jing Wang; Vassiliki Papadimitrakopoulou; Behrens Carmen; Ignacio I. Wistuba; Patrick Hwu; John V. Heymach; Don L. Gibbons

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Promising results in the treatment of non-small cell lung cancer (NSCLC) have been seen with immunomodulatory agents targeting immune checkpoints, such as programmed cell death 1 (PD-1) or programmed cell death 1 ligand (PD-L1). However, only a select group of patients respond to these interventions. The identification of biomarkers that predict clinical benefit to immune-based approaches, such as immune checkpoint blockade, is critical to successful translation of these agents. By using integrated gene array analysis in three large independent NSCLC patient datasets along with immunohistochemistry study (IHC), reverse phase protein array (RPPA) and in vivo animal study, we demonstrated that epithelial-mesenchymal transition (EMT) is highly associated with a profound inflammatory tumor microenvironment in lung adenocarcinoma. Our data revealed immune activation and simultaneous development of multiple immune suppressive mechanisms, including elevation of immune checkpoints such as PD-L1, PD-L2, PD-1, TIM-3, BTLA and CTLA-4, as well as an increase in tumor infiltrating CD4+Foxp3+ regulatory T cells, IL-6 and indoleamine 2,3-dioxygenase (IDO) in lung adenocarcinomas with a mesenchymal phenotype. Our data suggested that EMT might represent a potential biomarker to select the patients who will benefit from immune checkpoint blockade agents and other immunotherapies in NSCLC and possibly other cancers. Note: This abstract was not presented at the meeting. Citation Format: Yanyan Lou, Lixia Diao, Parra Cuentas Edwin Roger, Warren L. Denning, Limo Chen, Youhong Fan, Jaime Rodriguez, Lauren Byers, Jing Wang, Vassiliki Papadimitrakopoulou, Behrens Carmen, Ignacio I. Wistuba, Patrick Hwu, John V. Heymach, Don L. Gibbons. Epithelial-mesenchymal transition is associated with a profound inflammatory tumor microenvironment in lung adenocarcinoma. [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 3364. doi:10.1158/1538-7445.AM2015-3364


Journal of Clinical Oncology | 2017

STK11/LKB1 co-mutations to predict for de novo resistance to PD-1/PD-L1 axis blockade in KRAS-mutant lung adenocarcinoma.

Ferdinandos Skoulidis; Matthew D. Hellmann; Mark M. Awad; Hira Rizvi; Brett W. Carter; Warren Denning; Yasir Elamin; Jianjun Zhang; Giulia Costanza Leonardi; Darragh Halpenny; Andrew J. Plodkowski; Niamh Long; Jeremy J. Erasmus; Vassiliki Papadimitrakopoulou; Kwok-Kin Wong; Ignacio I. Wistuba; Pasi A. Jänne; Charles M. Rudin; John V. Heymach


Journal of Clinical Oncology | 2016

The CDK4/6 inhibitor G1T28 to protect immune cells and fibroblasts from chemotherapy and radiation-induced toxicity in vitro and to maintain efficacy of chemotherapy in SCLC Rb-deficient cells.

Irene Guijarro; Warren Denning; Renata Ferrarotto; Alissa Poteete; Uma Giri; John E. Bisi; Jessica A. Sorrentino; Patrick J. Roberts; Jay C. Strum; Rajesh Malik; Emily Roarty; John V. Heymach


publisher | None

title

author


Journal of Thoracic Oncology | 2017

MA 05.02 STK11/LKB1 Loss of Function Genomic Alterations Predict Primary Resistance to PD-1/PD-L1 Axis Blockade in KRAS-Mutant NSCLC

Ferdinandos Skoulidis; L. Albacker; Matthew D. Hellmann; Mark M. Awad; Justin F. Gainor; M. Goldberg; Alexa B. Schrock; Julia A. Elvin; Jeffrey S. Ross; H. Rizvi; Brett W. Carter; Jeremy J. Erasmus; Darragh Halpenny; Andrew J. Plodkowski; Niamh Long; M. Nishino-Habatu; Warren Denning; Jaime Rodriguez-Canales; Pamela Villalobos; E. Parra Cuentas; Lynette M. Sholl; Jennifer Sauter; Yasir Elamin; Jiexin Zhang; Giulia Costanza Leonardi; Kwok-Kin Wong; P.J. Stephens; Vassiliki Papadimitrakopoulou; Ignacio I. Wistuba; J. Wolchok


Journal of Thoracic Oncology | 2017

P2.03a-048 The CDK4/6 Inhibitor G1T28 Protects Immune Cells from Cisplatin-Induced Toxicity in vivo and Inhibits SCLC Tumor Growth: Topic: Clinical Trials

Irene Guijarro; Alissa Poteete; Renata Ferrarotto; Warren Denning; Haifa Hamdi; Patrick J. Roberts; Rajesh Malik; John E. Bisi; Jessica A. Sorrentino; Jay C. Strum; Emily Roarty; John V. Heymach

Collaboration


Dive into the Warren Denning's collaboration.

Top Co-Authors

Avatar

John V. Heymach

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ignacio I. Wistuba

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ferdinandos Skoulidis

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Vassiliki Papadimitrakopoulou

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jing Wang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lixia Diao

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lauren Averett Byers

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Don L. Gibbons

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matthew D. Hellmann

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge