Network


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

Hotspot


Dive into the research topics where David G. DeNardo is active.

Publication


Featured researches published by David G. DeNardo.


Cancer Discovery | 2011

Leukocyte Complexity Predicts Breast Cancer Survival and Functionally Regulates Response to Chemotherapy

David G. DeNardo; Donal J. Brennan; Elton Rexhepaj; Brian Ruffell; Stephen L. Shiao; Stephen F. Madden; William M. Gallagher; Nikhil Wadhwani; Scott D. Keil; Sharfaa A. Junaid; Hope S. Rugo; E. Shelley Hwang; Karin Jirström; Brian L. West; Lisa M. Coussens

UNLABELLED Immune-regulated pathways influence multiple aspects of cancer development. In this article we demonstrate that both macrophage abundance and T-cell abundance in breast cancer represent prognostic indicators for recurrence-free and overall survival. We provide evidence that response to chemotherapy is in part regulated by these leukocytes; cytotoxic therapies induce mammary epithelial cells to produce monocyte/macrophage recruitment factors, including colony stimulating factor 1 (CSF1) and interleukin-34, which together enhance CSF1 receptor (CSF1R)-dependent macrophage infiltration. Blockade of macrophage recruitment with CSF1R-signaling antagonists, in combination with paclitaxel, improved survival of mammary tumor-bearing mice by slowing primary tumor development and reducing pulmonary metastasis. These improved aspects of mammary carcinogenesis were accompanied by decreased vessel density and appearance of antitumor immune programs fostering tumor suppression in a CD8+ T-cell-dependent manner. These data provide a rationale for targeting macrophage recruitment/response pathways, notably CSF1R, in combination with cytotoxic therapy, and identification of a breast cancer population likely to benefit from this novel therapeutic approach. SIGNIFICANCE These findings reveal that response to chemotherapy is in part regulated by the tumor immune microenvironment and that common cytotoxic drugs induce neoplastic cells to produce monocyte/macrophage recruitment factors, which in turn enhance macrophage infiltration into mammary adenocarcinomas. Blockade of pathways mediating macrophage recruitment, in combination with chemotherapy, significantly decreases primary tumor progression, reduces metastasis, and improves survival by CD8+ T-cell-dependent mechanisms, thus indicating that the immune microenvironment of tumors can be reprogrammed to instead foster antitumor immunity and improve response to cytotoxic therapy.


Cancer Cell | 2009

CD4+ T Cells Regulate Pulmonary Metastasis of Mammary Carcinomas by Enhancing Protumor Properties of Macrophages

David G. DeNardo; Jairo B. Barreto; Pauline Andreu; Lesley Vasquez; David Tawfik; Nikita Kolhatkar; Lisa M. Coussens

During breast cancer development, increased presence of leukocytes in neoplastic stroma parallels disease progression; however, the functional significance of leukocytes in regulating protumor versus antitumor immunity in the breast remains poorly understood. Utilizing the MMTV-PyMT model of mammary carcinogenesis, we demonstrate that IL-4-expressing CD4(+) T lymphocytes indirectly promote invasion and subsequent metastasis of mammary adenocarcinomas by directly regulating the phenotype and effector function of tumor-associated CD11b(+)Gr1(-)F4/80(+) macrophages that in turn enhance metastasis through activation of epidermal growth factor receptor signaling in malignant mammary epithelial cells. Together, these data indicate that antitumor acquired immune programs can be usurped in protumor microenvironments and instead promote malignancy by engaging cellular components of the innate immune system functionally involved in regulating epithelial cell behavior.


Breast Cancer Research | 2007

Inflammation and breast cancer. Balancing immune response: crosstalk between adaptive and innate immune cells during breast cancer progression

David G. DeNardo; Lisa M. Coussens

Recent insights into the molecular and cellular mechanisms underlying cancer development have revealed that immune cells functionally regulate epithelial cancer development and progression. Moreover, accumulated clinical and experimental data indicate that the outcome of an immune response toward an evolving breast neoplasm is largely determined by the type of immune response elicited. Acute tumor-directed immune responses involving cytolytic T lymphocytes appear to protect against tumor development, whereas immune responses involving chronic activation of humoral immunity, infiltration by Th2 cells, and protumor-polarized innate inflammatory cells result in the promotion of tumor development and disease progression. Herein we review this body of literature and summarize important new findings revealing the paradoxical role of innate and adaptive leukocytes as regulators of breast carcinogenesis.


Cancer Cell | 2010

FcRγ Activation Regulates Inflammation-Associated Squamous Carcinogenesis

Pauline Andreu; Magnus Johansson; Nesrine I. Affara; Ferdinando Pucci; Tingting Tan; Simon Junankar; Lidiya Korets; Julia Lam; David Tawfik; David G. DeNardo; Luigi Naldini; Karin E. de Visser; Michele De Palma; Lisa M. Coussens

Chronically activated leukocytes recruited to premalignant tissues functionally contribute to cancer development; however, mechanisms underlying pro- versus anti-tumor programming of neoplastic tissues by immune cells remain obscure. Using the K14-HPV16 mouse model of squamous carcinogenesis, we report that B cells and humoral immunity foster cancer development by activating Fcgamma receptors (FcgammaRs) on resident and recruited myeloid cells. Stromal accumulation of autoantibodies in premalignant skin, through their interaction with activating FcgammaRs, regulate recruitment, composition, and bioeffector functions of leukocytes in neoplastic tissue, which in turn promote neoplastic progression and subsequent carcinoma development. These findings support a model in which B cells, humoral immunity, and activating FcgammaRs are required for establishing chronic inflammatory programs that promote de novo carcinogenesis.


Cancer Research | 2013

Targeting Tumor-Infiltrating Macrophages Decreases Tumor-Initiating Cells, Relieves Immunosuppression, and Improves Chemotherapeutic Responses

Jonathan B. Mitchem; Donal J. Brennan; Brett L. Knolhoff; Brian Belt; Yu Zhu; Dominic E. Sanford; Larisa Belaygorod; Danielle Carpenter; Lynne Collins; David Piwnica-Worms; Stephen M. Hewitt; Girish Mallya Udupi; William M. Gallagher; Craig D. Wegner; Brian L. West; Andrea Wang-Gillam; Peter S. Goedegebuure; David C. Linehan; David G. DeNardo

Tumor-infiltrating immune cells can promote chemoresistance and metastatic spread in aggressive tumors. Consequently, the type and quality of immune responses present in the neoplastic stroma are highly predictive of patient outcome in several cancer types. In addition to host immune responses, intrinsic tumor cell activities that mimic stem cell properties have been linked to chemoresistance, metastatic dissemination, and the induction of immune suppression. Cancer stem cells are far from a static cell population; rather, their presence seems to be controlled by highly dynamic processes that are dependent on cues from the tumor stroma. However, the impact immune responses have on tumor stem cell differentiation or expansion is not well understood. In this study, we show that targeting tumor-infiltrating macrophages (TAM) and inflammatory monocytes by inhibiting either the myeloid cell receptors colony-stimulating factor-1 receptor (CSF1R) or chemokine (C-C motif) receptor 2 (CCR2) decreases the number of tumor-initiating cells (TIC) in pancreatic tumors. Targeting CCR2 or CSF1R improves chemotherapeutic efficacy, inhibits metastasis, and increases antitumor T-cell responses. Tumor-educated macrophages also directly enhanced the tumor-initiating capacity of pancreatic tumor cells by activating the transcription factor STAT3, thereby facilitating macrophage-mediated suppression of CD8(+) T lymphocytes. Together, our findings show how targeting TAMs can effectively overcome therapeutic resistance mediated by TICs.


Cancer Research | 2010

Macrophage Expression of Hypoxia-Inducible Factor-1α Suppresses T-Cell Function and Promotes Tumor Progression

Andrew Doedens; Christian Stockmann; Mark P. Rubinstein; Debbie Liao; Na Zhang; David G. DeNardo; Lisa M. Coussens; Michael Karin; Ananda W. Goldrath; Randall S. Johnson

T cells can inhibit tumor growth, but their function in the tumor microenvironment is often suppressed. Many solid tumors exhibit abundant macrophage infiltration and low oxygen tension, yet how hypoxic conditions may affect innate immune cells and their role in tumor progression is poorly understood. Targeted deletion of the hypoxia-responsive transcription factor hypoxia-inducible factor-1α (HIF-1α) in macrophages in a progressive murine model of breast cancer resulted in reduced tumor growth, although vascular endothelial growth factor-A levels and vascularization were unchanged. Tumor-associated macrophages can suppress tumor-infiltrating T cells by several mechanisms, and we found that hypoxia powerfully augmented macrophage-mediated T-cell suppression in vitro in a manner dependent on macrophage expression of HIF-1α. Our findings link the innate immune hypoxic response to tumor progression through induction of T-cell suppression in the tumor microenvironment.


Cancer Research | 2014

CSF1/CSF1R Blockade Reprograms Tumor-Infiltrating Macrophages and Improves Response to T Cell Checkpoint Immunotherapy in Pancreatic Cancer Models.

Yu Zhu; Brett L. Knolhoff; Melissa A Meyer; Timothy M. Nywening; Brian L. West; Jianyang Luo; Andrea Wang-Gillam; Simon Peter Goedegebuure; David C. Linehan; David G. DeNardo

Cancer immunotherapy generally offers limited clinical benefit without coordinated strategies to mitigate the immunosuppressive nature of the tumor microenvironment. Critical drivers of immune escape in the tumor microenvironment include tumor-associated macrophages and myeloid-derived suppressor cells, which not only mediate immune suppression, but also promote metastatic dissemination and impart resistance to cytotoxic therapies. Thus, strategies to ablate the effects of these myeloid cell populations may offer great therapeutic potential. In this report, we demonstrate in a mouse model of pancreatic ductal adenocarcinoma (PDAC) that inhibiting signaling by the myeloid growth factor receptor CSF1R can functionally reprogram macrophage responses that enhance antigen presentation and productive antitumor T-cell responses. Investigations of this response revealed that CSF1R blockade also upregulated T-cell checkpoint molecules, including PDL1 and CTLA4, thereby restraining beneficial therapeutic effects. We found that PD1 and CTLA4 antagonists showed limited efficacy as single agents to restrain PDAC growth, but that combining these agents with CSF1R blockade potently elicited tumor regressions, even in larger established tumors. Taken together, our findings provide a rationale to reprogram immunosuppressive myeloid cell populations in the tumor microenvironment under conditions that can significantly empower the therapeutic effects of checkpoint-based immunotherapeutics.


Cancer and Metastasis Reviews | 2010

Interactions between lymphocytes and myeloid cells regulate pro- versus anti-tumor immunity

David G. DeNardo; Pauline Andreu; Lisa M. Coussens

Tumor-associated myeloid cells have been implicated in regulating many of the “hallmarks of cancer” and thus fostering solid tumor development and metastasis. However, the same innate leukocytes also participate in anti-tumor immunity and restraint of malignant disease. While many factors regulate the propensity of myeloid cells to promote or repress cancerous growths, polarized adaptive immune responses by B and T lymphocytes have been identified as regulators of many aspects of myeloid cell biology by specifically regulating their functional capabilities. Here, we detail the diversity of heterogeneous B and T lymphocyte populations and their impacts on solid tumor development through their abilities to regulate myeloid cell function in solid tumors.


Cancer and Metastasis Reviews | 2008

Immune cells as mediators of solid tumor metastasis

David G. DeNardo; Magnus Johansson; Lisa M. Coussens

Outgrowths of disseminated metastases remain the primary cause of mortality in cancer patients; however, molecular and cellular mechanisms regulating metastatic spread remain largely elusive. Recent insights into these mechanisms have refined the seed and soil hypothesis and it is now recognized that metastasis of solid tumors requires collaborative interactions between malignant cells and a diverse assortment of “activated” stromal cells at both primary and secondary tumor locations. Specifically, persistent pro-tumor immune responses (inflammation), now generally accepted as potentiating primary tumor development, are also being recognized as mediators of cancer metastasis. Thus, novel anti-cancer therapeutic strategies targeting molecular and/or cellular mechanisms regulating these collaborative interactions may provide efficacious relief for metastatic disease. This review focuses on recent literature revealing new mechanisms whereby immune cells regulate metastatic progression, with a primary focus on breast cancer.


Immunological Reviews | 2008

Polarized immune responses differentially regulate cancer development

Magnus Johansson; David G. DeNardo; Lisa M. Coussens

Summary: Tumor‐associated immune responses assert varied responses toward developing neoplasms that can either act to eradicate malignant cells via engagement of potent cytotoxic programs or alternatively enhance tumor growth through release of multifunctional pro‐tumor mediators. Seemingly paradoxical, these disparate activities reflect a continuum of polarization (or activation) states possible for distinct leukocyte subsets that demonstrate tissue, organ, and tumor selectivity. Herein, we review clinical and experimental studies investigating cellular and molecular mechanisms utilized by neoplastic tissues to alternatively polarize immune responses that favor either pro‐ or anti‐tumor immunity.

Collaboration


Dive into the David G. DeNardo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrea Wang-Gillam

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

David C. Linehan

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Brett L. Knolhoff

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

William G. Hawkins

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Roheena Z. Panni

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Brian Belt

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Dominic E. Sanford

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Timothy M. Nywening

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Ryan C. Fields

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge