Network


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

Hotspot


Dive into the research topics where Brett L. Knolhoff is active.

Publication


Featured researches published by Brett L. Knolhoff.


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 | 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.


Nature Medicine | 2016

Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy

Hong Jiang; Samarth Hegde; Brett L. Knolhoff; Yu Zhu; John M. Herndon; Melissa A Meyer; Timothy M. Nywening; William G. Hawkins; Irina M. Shapiro; David T. Weaver; Jonathan A. Pachter; Andrea Wang-Gillam; David G. DeNardo

Single-agent immunotherapy has achieved limited clinical benefit to date in patients with pancreatic ductal adenocarcinoma (PDAC). This may be a result of the presence of a uniquely immunosuppressive tumor microenvironment (TME). Critical obstacles to immunotherapy in PDAC tumors include a high number of tumor-associated immunosuppressive cells and a uniquely desmoplastic stroma that functions as a barrier to T cell infiltration. We identified hyperactivated focal adhesion kinase (FAK) activity in neoplastic PDAC cells as an important regulator of the fibrotic and immunosuppressive TME. We found that FAK activity was elevated in human PDAC tissues and correlated with high levels of fibrosis and poor CD8+ cytotoxic T cell infiltration. Single-agent FAK inhibition using the selective FAK inhibitor VS-4718 substantially limited tumor progression, resulting in a doubling of survival in the p48-Cre;LSL-KrasG12D;Trp53flox/+ (KPC) mouse model of human PDAC. This delay in tumor progression was associated with markedly reduced tumor fibrosis and decreased numbers of tumor-infiltrating immunosuppressive cells. We also found that FAK inhibition rendered the previously unresponsive KPC mouse model responsive to T cell immunotherapy and PD-1 antagonists. These data suggest that FAK inhibition increases immune surveillance by overcoming the fibrotic and immunosuppressive PDAC TME and renders tumors responsive to immunotherapy.


Nature Communications | 2016

Stromal senescence establishes an immunosuppressive microenvironment that drives tumorigenesis

Megan K. Ruhland; Andrew J. Loza; Aude-Helene Capietto; Xianmin Luo; Brett L. Knolhoff; Kevin C. Flanagan; Brian Belt; Elise Alspach; Kathleen M. Leahy; Jingqin Luo; Andras Schaffer; John R. Edwards; Gregory D. Longmore; Roberta Faccio; David G. DeNardo; Sheila A. Stewart

Age is a significant risk factor for the development of cancer. However, the mechanisms that drive age-related increases in cancer remain poorly understood. To determine if senescent stromal cells influence tumorigenesis, we develop a mouse model that mimics the aged skin microenvironment. Using this model, here we find that senescent stromal cells are sufficient to drive localized increases in suppressive myeloid cells that contributed to tumour promotion. Further, we find that the stromal-derived senescence-associated secretory phenotype factor interleukin-6 orchestrates both increases in suppressive myeloid cells and their ability to inhibit anti-tumour T-cell responses. Significantly, in aged, cancer-free individuals, we find similar increases in immune cells that also localize near senescent stromal cells. This work provides evidence that the accumulation of senescent stromal cells is sufficient to establish a tumour-permissive, chronic inflammatory microenvironment that can shelter incipient tumour cells, thus allowing them to proliferate and progress unabated by the immune system.


Immunity | 2017

Tissue-Resident Macrophages in Pancreatic Ductal Adenocarcinoma Originate from Embryonic Hematopoiesis and Promote Tumor Progression

Yu Zhu; John M. Herndon; Dorothy K. Sojka; Ki-Wook Kim; Brett L. Knolhoff; Chong Zuo; Darren R. Cullinan; Jingqin Luo; Audrey R. Bearden; Kory J. Lavine; Wayne M. Yokoyama; William G. Hawkins; Ryan C. Fields; Gwendalyn J. Randolph; David G. DeNardo

&NA; Tumor‐associated macrophages (TAMs) are essential components of the cancer microenvironment and play critical roles in the regulation of tumor progression. Optimal therapeutic intervention requires in‐depth understanding of the sources that sustain macrophages in malignant tissues. In this study, we investigated the ontogeny of TAMs in murine pancreatic ductal adenocarcinoma (PDAC) models. We identified both inflammatory monocytes and tissue‐resident macrophages as sources of TAMs. Unexpectedly, significant portions of pancreas‐resident macrophages originated from embryonic development and expanded through in situ proliferation during tumor progression. Whereas monocyte‐derived TAMs played more potent roles in antigen presentation, embryonically derived TAMs exhibited a pro‐fibrotic transcriptional profile, indicative of their role in producing and remodeling molecules in the extracellular matrix. Collectively, these findings uncover the heterogeneity of TAM origin and functions and could provide therapeutic insight for PDAC treatment. Graphical Abstract Figure. No caption available. HighlightsTAMs in PDAC are derived from both monocytes and embryonic macrophagesTissue‐resident embryonic macrophages promote PDAC progressionEmbryonically derived tissue‐resident TAMs expand in PDAC via in situ proliferationEmbryonically derived TAMs exhibit unique pro‐fibrotic activities &NA; Zhu et al. identify tissue‐resident macrophages of embryonic origin as a source of tumor‐associated macrophages in pancreatic ductal adenocarcinoma. These cells expand through in situ proliferation during tumor progression and demonstrate a unique pro‐fibrotic transcriptional profile distinct from that of their monocyte‐derived counterparts.


Cancer Research | 2016

Antagonizing Integrin β3 Increases Immunosuppression in Cancer.

Xinming Su; Alison K. Esser; Sarah R. Amend; Jingyu Xiang; Yalin Xu; Michael H. Ross; Gregory C. Fox; Takayuki Kobayashi; Veronica Steri; Kirsten Roomp; Francesca Fontana; Michelle A. Hurchla; Brett L. Knolhoff; Melissa A Meyer; Elizabeth A. Morgan; Julia C. Tomasson; Joshua S. Novack; Wei Zou; Roberta Faccio; Deborah V. Novack; Stephen Robinson; Steven L. Teitelbaum; David G. DeNardo; Jochen G. Schneider; Katherine N. Weilbaecher

Integrin β3 is critical for tumor invasion, neoangiogenesis, and inflammation, making it a promising cancer target. However, preclinical and clinical data of integrin β3 antagonists have demonstrated no benefit or worse outcomes. We hypothesized that integrin β3 could affect tumor immunity and evaluated tumors in mice with deletion of integrin β3 in macrophage lineage cells (β3KOM). β3KOM mice had increased melanoma and breast cancer growth with increased tumor-promoting M2 macrophages and decreased CD8(+) T cells. Integrin β3 antagonist, cilengitide, also enhanced tumor growth and increased M2 function. We uncovered a negative feedback loop in M2 myeloid cells, wherein integrin β3 signaling favored STAT1 activation, an M1-polarizing signal, and suppressed M2-polarizing STAT6 activation. Finally, disruption of CD8(+) T cells, macrophages, or macrophage integrin β3 signaling blocked the tumor-promoting effects of integrin β3 antagonism. These results suggest that effects of integrin β3 therapies on immune cells should be considered to improve outcomes. Cancer Res; 76(12); 3484-95. ©2016 AACR.


Clinical Cancer Research | 2017

Constitutive IRAK4 Activation Underlies Poor Prognosis and Chemoresistance in Pancreatic Ductal Adenocarcinoma.

Daoxiang Zhang; Lin Li; Hongmei Jiang; Brett L. Knolhoff; Albert C. Lockhart; Andrea Wang-Gillam; David G. DeNardo; Marianna B. Ruzinova; Kian-Huat Lim

Purpose: Aberrant activation of the NF-κB transcription factors underlies the aggressive behavior and poor outcome of pancreatic ductal adenocarcinoma (PDAC). However, clinically effective and safe NF-κB inhibitors are not yet available. Because NF-κB transcription factors can be activated by the interleukin-1 receptor-associated kinases (IRAKs) downstream of the Toll-like receptors (TLRs), but has not been explored in PDAC, we sought to investigate the role of IRAKs in the pathobiology of PDAC. Experimental Design: We examined the phosphorylation status of IRAK4 (p-IRAK4), the master regulator of TLR signaling, in PDAC cell lines, in surgical samples and commercial tissue microarray. We then performed functional studies using small-molecule IRAK1/4 inhibitor, RNA-interference, and CRISPR/Cas9n techniques to delineate the role of IRAK4 in NF-κB activity, chemoresistance, cytokine production, and growth of PDAC cells in vitro and in vivo. Results: p-IRAK4 staining was detectable in the majority of PDAC lines and about 60% of human PDAC samples. The presence of p-IRAK4 strongly correlated with phospho-NF-κB/p65 staining in PDAC samples and is predictive of postoperative relapse and poor overall survival. Inhibition of IRAK4 potently reduced NF-κB activity, anchorage-independent growth, chemoresistance, and secretion of proinflammatory cytokines from PDAC cells. Both pharmacologic suppression and genetic ablation of IRAK4 greatly abolished PDAC growth in mice and augmented the therapeutic effect of gemcitabine by promoting apoptosis, reducing tumor cell proliferation and tumor fibrosis. Conclusions: Our data established IRAK4 as a novel therapeutic target for PDAC treatment. Development of potent IRAK4 inhibitors is needed for clinical testing. Clin Cancer Res; 23(7); 1748–59. ©2016 AACR.


Nature Communications | 2018

Breast and pancreatic cancer interrupt IRF8-dependent dendritic cell development to overcome immune surveillance

Melissa A Meyer; John M Baer; Brett L. Knolhoff; Timothy M. Nywening; Roheena Z. Panni; Xinming Su; Katherine N. Weilbaecher; William G. Hawkins; Cynthia X. Ma; Ryan C. Fields; David C. Linehan; Grant A. Challen; Roberta Faccio; Rebecca Aft; David G. DeNardo

Tumors employ multiple mechanisms to evade immune surveillance. One mechanism is tumor-induced myelopoiesis, whereby the expansion of immunosuppressive myeloid cells can impair tumor immunity. As myeloid cells and conventional dendritic cells (cDCs) are derived from the same progenitors, we postulated that myelopoiesis might impact cDC development. The cDC subset, cDC1, which includes human CD141+ DCs and mouse CD103+ DCs, supports anti-tumor immunity by stimulating CD8+ T-cell responses. Here, to understand how cDC1 development changes during tumor progression, we investigated cDC bone marrow progenitors. We found localized breast and pancreatic cancers induce systemic decreases in cDC1s and their progenitors. Mechanistically, tumor-produced granulocyte-stimulating factor downregulates interferon regulatory factor-8 in cDC progenitors, and thus results in reduced cDC1 development. Tumor-induced reductions in cDC1 development impair anti-tumor CD8+ T-cell responses and correlate with poor patient outcomes. These data suggest immune surveillance can be impaired by tumor-induced alterations in cDC development.Tumors escape the immune system through many mechanisms. Here the authors show that certain tumors inhibit anti-tumor immunity by stopping the production of conventional dendritic cells (cDCs) in the bone marrow, therefore depleting the pool of cDCs available to present antigen to CD8+ T cells.


Oncogenesis | 2018

SNAIL1 action in tumor cells influences macrophage polarization and metastasis in breast cancer through altered GM-CSF secretion

Audrey Brenot; Brett L. Knolhoff; David G. DeNardo; Gregory D. Longmore

The EMT inducer SNAIL1 regulates breast cancer metastasis and its expression in human primary breast tumor predicts for poor outcomes. During tumor progression SNAIL1 has multiple effects in tumor cells that can impact metastasis. An inflammatory tumor microenvironment also impacts metastasis and recently SNAIL1 has been implicated as modulating the secretion of cytokines that can influence the tumor immune infiltrate. Using a spontaneous genetic model of breast cancer metastasis and syngeneic orthotopic transplant experiments we show that the action of SNAIL1 in primary breast tumor cells is required for breast tumor growth and metastasis. It does so, in part, by regulating production of GM-CSF, IL1α, IL-6, and TNFα by breast cancer cells. The SNAIL1-dependent tumor cell secretome modulates the primary tumor-associated macrophage (TAM) polarization. GM-CSF alone modulates TAM polarization and impacts breast cancer metastasis in vivo. This study highlights another role for breast tumor SNAIL1 in cancer progression to metastasis—modulation of the immune microenvironment of primary breast tumors.


Cancer Research | 2016

Abstract 181: Constitutive IRAK1/4 kinase activation contributes to NF-kB activity and chemoresistance in pancreatic cancer

Daoxiang Zhang; Lin Li; Hongmei Jiang; Jinsheng Yu; Brett L. Knolhoff; Richard Head; Albert C. Lockhart; David G. DeNardo; Andrea Wang-Gillam; Marianna B. Ruzinova; Kian-Huat Lim

Constitutive activation of the NF-κb transcription factor is a major molecular mechanism that contributes to the aggressive behavior and treatment resistance of pancreatic cancer. Understanding the molecular mechanisms that activate NF-κB will provide novel therapeutic opportunities to improve the dismal outcome of pancreatic cancer patients. In the present study, we showed that the Interleukin-1 Receptor-Associated Kinases 1 and 4 (IRAK1 and IRAK4) are constitutively activated in a majority of pancreatic cancer cell lines and patients samples, and are major drivers of NF-κB activity. Notably, we found that constitutive phosphorylation of IRAK4 is associated with poor patient prognosis. Suppression of IRAK1 and IRAK4 with small molecule inhibitor or RNA-interference in pancreatic cancer cells significantly reduces NF-κB activity, three-dimensional growth, invasiveness, production of inflammatory, and augment their sensitivity to chemotherapeutic agents in vitro. Notably, we showed that the NF-κB activity of pancreatic cancer cell with IRAK4 ablated using CRISPR technology can be restored with wild-type, but not kinase-dead IRAK4 mutant, supporting development of IRAK4 inhibitor as a novel therapeutic agent in pancreatic cancer. Silencing of IRAK1 or IRAK4, and more potently both, significantly abrogated the tumorigenic potential of human and murine pancreatic cancer cells as xenograft in mice. Lastly, we showed that IRAK1/4 inhibitor augments the therapeutic effect of gemcitabine in tumor-bearing mice by suppressing proliferation and increasing apoptosis of neoplastic cells, and reducing stromal fibrosis. Together, our data established IRAK4 kinase inhibitors as a promising novel class of targeted agent in pancreatic cancer. Citation Format: Daoxiang Zhang, Lin Li, Hongmei Jiang, Jinsheng Yu, Brett Knolhoff, Richard Head, Albert C. Lockhart, David G. DeNardo, Andrea Wang-Gillam, Marianna B. Ruzinova, Kian-Huat Lim. Constitutive IRAK1/4 kinase activation contributes to NF-kB activity and chemoresistance in pancreatic cancer. [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 181.

Collaboration


Dive into the Brett L. Knolhoff's collaboration.

Top Co-Authors

Avatar

David G. DeNardo

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gundumi A. Upadhya

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

William C. Chapman

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Bernard J. DuBray

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jianluo Jia

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Parvathi Balachandran

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Andrea Wang-Gillam

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

T. Mohanakumar

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Yu Zhu

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge