Network


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

Hotspot


Dive into the research topics where James Sutton is active.

Publication


Featured researches published by James Sutton.


Nature Medicine | 2013

CSF-1R inhibition alters macrophage polarization and blocks glioma progression

Stephanie M. Pyonteck; Leila Akkari; Alberto J. Schuhmacher; Robert L. Bowman; Lisa Sevenich; Daniela F. Quail; Oakley C. Olson; Marsha L. Quick; Jason T. Huse; Virginia Teijeiro; Manu Setty; Christina S. Leslie; Yoko Oei; Alicia Pedraza; Jianan Zhang; Cameron Brennan; James Sutton; Eric C. Holland; Dylan Daniel; Johanna A. Joyce

Glioblastoma multiforme (GBM) comprises several molecular subtypes, including proneural GBM. Most therapeutic approaches targeting glioma cells have failed. An alternative strategy is to target cells in the glioma microenvironment, such as tumor-associated macrophages and microglia (TAMs). Macrophages depend on colony stimulating factor-1 (CSF-1) for differentiation and survival. We used an inhibitor of the CSF-1 receptor (CSF-1R) to target TAMs in a mouse proneural GBM model, which significantly increased survival and regressed established tumors. CSF-1R blockade additionally slowed intracranial growth of patient-derived glioma xenografts. Surprisingly, TAMs were not depleted in treated mice. Instead, glioma-secreted factors, including granulocyte-macrophage CSF (GM-CSF) and interferon-γ (IFN-γ), facilitated TAM survival in the context of CSF-1R inhibition. Expression of alternatively activated M2 markers decreased in surviving TAMs, which is consistent with impaired tumor-promoting functions. These gene signatures were associated with enhanced survival in patients with proneural GBM. Our results identify TAMs as a promising therapeutic target for proneural gliomas and establish the translational potential of CSF-1R inhibition for GBM.


Science | 2016

The tumor microenvironment underlies acquired resistance to CSF-1R inhibition in gliomas

Daniela F. Quail; Robert L. Bowman; Leila Akkari; Marsha L. Quick; Alberto J. Schuhmacher; Jason T. Huse; Eric C. Holland; James Sutton; Johanna A. Joyce

Another pathway to cancer resistance Therapies targeting the tumor microenvironment show promise for treating cancer. For example, antibodies targeting colony-stimulating factor-1 receptor (CSF-1R) inhibit protumorigenic macrophages and regress tumors in mouse models of glioblastoma multiforme (GBM), a deadly form of brain cancer. Quail et al. found that although CSR-1R blockade prolonged survival in mouse models of GBM, more than 50% of tumors eventually recurred. Recurrence was correlated with elevated PI3-K activity in tumors, driven by macrophage-secreted IGF-1. Blocking PI3-K and IGF-1 signaling in rebounding tumors prolonged survival. Thus, tumors can acquire resistance to therapy through intrinsic changes and through changes in their microenvironment. Science, this issue p. 10.1126/science.aad3018 Brain tumors can acquire resistance to therapy through changes to their microenvironment. INTRODUCTION Therapies targeted against the tumor microenvironment (TME) represent a promising approach for treating cancer. This appeal arises in part from the decreased likelihood of acquired resistance through mutations in target TME cells, as is frequently observed with cancer cell–targeted therapies. Although classical mechanisms of tumor cell–intrinsic resistance to cytotoxic and targeted agents have been well-defined—including aberrant drug metabolism and transport, drug target mutation, and activation of alternative survival pathways—it still remains unclear whether resistance to TME-directed therapies follows similar principles. Given that TME-targeted agents are increasingly being evaluated in the clinic, it is becoming critical to mechanistically define how resistance may evolve in response to these therapies in order to provide long-term disease management for patients. RATIONALE Macrophages and microglia are of the most abundant noncancerous cell types in glioblastoma multiforme (GBM), in some cases accounting for up to 30% of the total tumor composition. Macrophages accumulate with GBM progression and can be acutely targeted via inhibition of colony-stimulating factor–1 receptor (CSF-1R) to regress high-grade gliomas in animal models. However, it is currently unknown whether and how resistance emerges in response to sustained CSF-1R blockade in GBM. Despite this, multiple clinical trials are currently underway testing the efficacy of CSF-1R inhibition in glioma patients. Therefore, determining whether long-term CSF-1R inhibition can stably regress GBM by using animal models is an important and timely question to address. RESULTS Using genetic mouse models of GBM, we show that although overall survival is significantly prolonged in response to CSF-1R inhibition, tumors recur eventually in >50% of mice. Upon isolation and transplantation of recurrent tumor cells into naïve animals, gliomas reestablish sensitivity to CSF-1R inhibition, indicating that resistance is microenvironment-driven. Through RNA-sequencing of glioma cells and macrophages purified from treated tumors and ex vivo cell culture assays, we found elevated phosphatidylinositol 3-kinase (PI3K) pathway activity in recurrent GBM after CSF-1R inhibition, driven by macrophage-derived insulin-like growth factor–1 (IGF-1) and tumor cell IGF-1 receptor (IGF-1R). Consequently, combining IGF-1R or PI3K blockade with continuous CSF-1R inhibition in recurrent tumors significantly prolonged overall survival. In contrast, monotherapy with IGF-1R or PI3K inhibitors in rebound or treatment-naïve tumors was less effective, indicating the necessity of combination therapy to expose PI3K signaling dependency in recurrent disease. Mechanistically, we found that activation of macrophages in recurrent tumors by IL4 led to elevated Stat6 and nuclear factor of activated T cells (NFAT) signaling upstream of Igf1, and inhibition of either of these pathways in vivo was sufficient to significantly extend survival. CONCLUSION We have identified a mechanism of drug resistance that can circumvent therapeutic response to a TME-targeted therapy and promote disease recurrence in the absence of tumor cell–intrinsic alterations. Specifically, we have uncovered a heterotypic paracrine signaling interaction that is initiated by the TME and drives resistance to CSF-1R inhibition through IGF-1R/PI3K signaling. Given that PI3K signaling is aberrantly activated in a substantial proportion of GBM patients, and that recent clinical trial results show limited efficacy in recurrent (albeit very advanced) GBM, it is possible that this pathway could similarly contribute to intrinsic resistance to CSF-1R inhibition. Our findings underscore the importance of bidirectional feedback between cancer cells and their microenvironment and support the notion that although stromal cells are less susceptible to genetic mutation than are cancer cells, a tumor can nonetheless acquire a resistant phenotype by exploiting its extracellular environment. Resistance to CSF-1R inhibition in glioma. (A) Macrophages contribute to GBM progression by creating a protumorigenic niche associated with M2-like gene expression. CSF-1R is a critical receptor for macrophage biology and is under clinical evaluation as a therapeutic target in glioma . (B) Targeting CSF-1R early in gliomagenesis significantly prolongs survival in mouse models. CSF-1R inhibition reprograms macrophages to become antitumorigenic by down-regulating M2-like genes and enhancing phagocytosis. Tumor-derived survival factors sustain macrophage viability despite CSF-1R blockade


eLife | 2015

Compensatory induction of MYC expression by sustained CDK9 inhibition via a BRD4-dependent mechanism

Huasong Lu; Yuahua Xue; Guoying K Yu; Carolina Arias; Julie Lin; Susan Fong; Michel Faure; Ben Weisburd; Xiaodan Ji; Alexandre Mercier; James Sutton; Kunxin Luo; Zhenhai Gao; Qiang Zhou

CDK9 is the kinase subunit of positive transcription elongation factor b (P-TEFb) that enables RNA polymerase (Pol) IIs transition from promoter-proximal pausing to productive elongation. Although considerable interest exists in CDK9 as a therapeutic target, little progress has been made due to lack of highly selective inhibitors. Here, we describe the development of i-CDK9 as such an inhibitor that potently suppresses CDK9 phosphorylation of substrates and causes genome-wide Pol II pausing. While most genes experience reduced expression, MYC and other primary response genes increase expression upon sustained i-CDK9 treatment. Essential for this increase, the bromodomain protein BRD4 captures P-TEFb from 7SK snRNP to deliver to target genes and also enhances CDK9s activity and resistance to inhibition. Because the i-CDK9-induced MYC expression and binding to P-TEFb compensate for P-TEFbs loss of activity, only simultaneously inhibiting CDK9 and MYC/BRD4 can efficiently induce growth arrest and apoptosis of cancer cells, suggesting the potential of a combinatorial treatment strategy. DOI: http://dx.doi.org/10.7554/eLife.06535.001


International Journal of Toxicology | 2011

Investigation of Correlation Among Safety Biomarkers in Serum, Histopathological Examination, and Toxicogenomics:

Tao Wang; Maria Papoutsi; Marion Wiesmann; Marc DeCristofaro; M. Craig Keselica; Elizabeth Skuba; Robert H. Spaet; Judit Markovits; Armin Wolf; Pierre Moulin; Francois Pognan; Paul Vancutsem; Lew Petryk; James Sutton; Salah-Dine Chibout; William Kluwe

This article addresses the issue of miscorrelation between hepatic injury biomarkers and histopathological findings in the drug development context. Our studies indicate that the use of toxicogenomics can aid in the drug development decision-making process associated with such miscorrelated data. BLZ945 was developed as a Colony-Stimulating Factor 1 Receptor (CSF-1R) inhibitor. Treatment of BLZ945 in rats and monkeys increased serum alanine aminotransferase (ALT) and aspartate transaminase (AST). However, liver hypertrophy was the only histopathological liver finding in rats, and there was no change in the livers of monkeys. Longer treatment of BLZ945 in rats for 6 weeks caused up to 6-fold elevation of ALT, yet hepatocyte necrosis was not detected microscopically. Toxicogenomic profiling of liver samples demonstrated that the genes associated with early response to liver injury, apoptosis/necrosis, inflammation, oxidative stress, and metabolic enzymes were upregulated. Studies are ongoing to evaluate the mechanisms underlying BL945-induced ALT and AST elevations.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of nonbenzamidine factor VIIa inhibitors using a biaryl acid scaffold

Scott A. Bolton; James Sutton; Rushith Anumula; Gregory S. Bisacchi; Bruce L. Jacobson; William A. Slusarchyk; Uwe D. Treuner; Shung C. Wu; Guohua Zhao; Zulan Pi; Steven Sheriff; Rebecca A. Smirk; Sharon N. Bisaha; Daniel L. Cheney; Anzhi Wei; William A. Schumacher; Karen S. Hartl; Eddie C.-K. Liu; Robert Zahler; Steven M. Seiler

In this Letter, we describe the synthesis of several nonamidine analogs of biaryl acid factor VIIa inhibitor 1 containing weakly basic or nonbasic P1 groups. 2-Aminoisoquinoline was found to be an excellent surrogate for the benzamidine group (compound 2) wherein potent inhibition of factor VIIa is maintained relative to most other related serine proteases. In an unanticipated result, the m-benzamide P1 (compounds 21a and 21b) proved to be a viable benzamidine replacement, albeit with a 20-40 fold loss in potency against factor VIIa.


Journal of Biological Chemistry | 2016

Molecular basis of mRNA cap recognition by Influenza B polymerase PB2 subunit

Lili Xie; Charles Wartchow; Steven Shia; Kyoko Uehara; Micah Steffek; Robert Warne; James Sutton; Gladys T. Muiru; Vincent H. J. Leonard; Dirksen E. Bussiere; Xiaolei Ma

Influenza virus polymerase catalyzes the transcription of viral mRNAs by a process known as “cap-snatching,” where the 5′-cap of cellular pre-mRNA is recognized by the PB2 subunit and cleaved 10–13 nucleotides downstream of the cap by the endonuclease PA subunit. Although this mechanism is common to both influenza A (FluA) and influenza B (FluB) viruses, FluB PB2 recognizes a wider range of cap structures including m7GpppGm-, m7GpppG-, and GpppG-RNA, whereas FluA PB2 utilizes methylated G-capped RNA specifically. Biophysical studies with isolated PB2 cap-binding domain (PB2cap) confirm that FluB PB2 has expanded mRNA cap recognition capability, although the affinities toward m7GTP are significantly reduced when compared with FluA PB2. The x-ray co-structures of the FluB PB2cap with bound cap analogs m7GTP and GTP reveal an inverted GTP binding mode that is distinct from the cognate m7GTP binding mode shared between FluA and FluB PB2. These results delineate the commonalities and differences in the cap-binding site between FluA and FluB PB2 and will aid structure-guided drug design efforts to identify dual inhibitors of both FluA and FluB PB2.


Xenobiotica | 2015

Phenotypic and metabolic investigation of a CSF-1R kinase receptor inhibitor (BLZ945) and its pharmacologically active metabolite

Joel Krauser; Yi Jin; Markus Walles; Ulrike Pfaar; James Sutton; Marion Wiesmann; Daniel Graf; Veronique Pflimlin-Fritschy; Thierry Wolf; Gian Camenisch; Piet Swart

Abstract 1.u20024-[2((1R,2R)-2-Hydroxycyclohexylamino)-benzothiazol-6-yloxyl]-pyridine-2-carboxylic acid methylamide (BLZ945) is a small molecule inhibitor of CSF-1R kinase activity within osteoclasts designed to prevent skeletal related events in metastatic disease. Key metabolites were enzymatically and structurally characterized to understand the metabolic fate of BLZ945 and pharmacological implications. The relative intrinsic clearances for metabolites were derived from in vitro studies using human hepatocytes, microsomes and phenotyped with recombinant P450 enzymes. 2.u2002Formation of a pharmacologically active metabolite (M9) was observed in human hepatocytes. The M9 metabolite is a structural isomer (diastereomer) of BLZ945 and is about 4-fold less potent. This isomer was enzymatically formed via P450 oxidation of the BLZ945 hydroxyl group, followed by aldo–keto reduction to the alcohol (M9). 3.u2002Two reaction phenotyping approaches based on fractional clearances were applied to BLZ945 using hepatocytes and liver microsomes. The fraction metabolized (fm) or contribution ratio was determined for each metabolic reaction type (oxidation, glucuronidation or isomerization) as well as for each metabolite. The results quantitatively illustrate contribution ratios of the involved enzymes and pathways, e.g. the isomerization to metabolite M9 accounted for 24% intrinsic clearance in human hepatocytes. In summary, contribution ratios for the Phase I and Phase II pathways can be determined in hepatocytes.


Cancer Research | 2010

Abstract 3629: BLZ945, a selective c-fms (CSF-1R) kinase inhibitor for the suppression of tumor-induced osteolytic lesions in bone

Marion Wiesmann; Dylan Daniel; Nancy Pryer; James Sutton; Victoria Sung; Tao Wang; Ursula Jeffry; Yoko Oei; Susan Kaufman; William P. Lenahan; Isabelle Lee; Kay Huh; Janet Sim

Aberrant activation of osteoclasts due to bone metastasis causes osteolysis, skeletal-related events and severe pain in cancer patients. Macrophage-Colony-stimulating Factor (M-CSF) signaling through its receptor c-fms / Colony-Stimulating Factor-1 Receptor (CSF-1R) in the monocytic lineage is essential for osteoclastogenesis, providing an opportunity to inhibit this pathway and suppress tumor-induced osteolysis. BLZ945 is an orally active, potent and selective CSF-1R inhibitor. BLZ945 inhibits CSF-1R activity with an IC50 of 1nM and is more than 1000-fold selective against its closest receptor tyrosine kinase homologs c-KIT and Platelet-derived Growth Factor Receptor beta (PDGFRb) as well as more than 200 additional kinases, confirming the selectivity of the compound. BLZ945 potently inhibited proliferation of the M-CSF-dependent murine leukemia cell line MNFS60 (EC50 67 nM) consistent with the inhibition of the CSF-1R kinase activity. Tyrosine phosphorylation in HEK293 cells overexpressing human CSF-1R was inhibited with an EC50 of 58 nM. Functional activity of BLZ945 was shown by inhibition of osteoclastogenesis using human osteoclast precursors. The MNFS60 allograft model was used to evaluate the pharmacodynamics of BLZ945 by monitoring dose and time dependent changes in tyrosine phoshorylation of CSF-1R to select doses and regimens for in vivo efficacy studies. A single dose of BLZ945 at 200 mg/kg maximally suppressed CSF-1R phosphorylation of >50% for more than 16 hours in this model and was selected for further evaluation in mouse models of breast (MDA-MB-231Luc) and prostate (PC-3MLuc) tumor-induced osteolysis (TIO). In these models human tumor cells are injected directly into the tibia of nude mice and bone destruction is measured by x-ray analysis and serum levels of tartrate-resistant acid phosphatase 5b (TRAP5b), a marker of osteoclast activity. Daily dosing of BLZ945 at 200 mg/kg resulted in a significant reduction in the progression of osteolysis, with > 50% reduction in osteolytic lesion severity in both TIO models, as compared to vehicle-treated animals. Reductions in serum TRAP5b were also observed at the end of the studies. BLZ945 showed enhanced activity in combination with zoledronate in the PC-3MLuc model, with the combination reducing osteolytic severity scores by 90%. In addition anti-osteolytic activity was observed in a nude rat model of osteolysis induced by intratibial injection of MDA-MB-231 cells. In naive cynomolgus monkeys, treatment with BLZ945 resulted in dose-dependent reductions of serum TRAP5b and cross-linked collagen (CTX) in both serum and urine, consistent with the expected effect of inhibiting bone resorption. These data support the testing of CSF-1R inhibitors in advanced cancer patients with bone metastases and skeletal related events. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3629.


Cell Reports | 2018

Type I Interferon Receptor Signaling of Neurons and Astrocytes Regulates Microglia Activation during Viral Encephalitis

Chintan Chhatbar; Claudia N. Detje; Elena Grabski; Katharina Borst; Julia Spanier; Luca Ghita; David A. Elliott; Marta Joana Costa Jordão; Nora Mueller; James Sutton; Chittappen K. Prajeeth; Viktoria Gudi; Michael A. Klein; Marco Prinz; Frank Bradke; Martin Stangel; Ulrich Kalinke

n Summaryn n In sterile neuroinflammation, a pathological role is proposed for microglia, whereas in viral encephalitis, their function is not entirely clear. Many viruses exploit the odorant system and enter the CNS via the olfactory bulb (OB). Upon intranasal vesicular stomatitis virus instillation, we show an accumulation ofxa0activated microglia and monocytes in the OB. Depletion of microglia during encephalitis results in enhanced virus spread and increased lethality. Activation, proliferation, and accumulation of microglia are regulated by type I IFN receptor signaling of neurons and astrocytes, but not of microglia. Morphological analysis of myeloid cells shows that type I IFN receptor signaling of neurons has a stronger impact on the activation of myeloid cells than of astrocytes. Thus, in the infected CNS, the cross talk among neurons, astrocytes, and microglia is critical for full microglia activation and protection from lethal encephalitis.n n


ACS Medicinal Chemistry Letters | 2018

Optimization of 3-Pyrimidin-4-yl-oxazolidin-2-ones as Orally Bioavailable and Brain Penetrant Mutant IDH1 Inhibitors

Qian Zhao; James R. Manning; James Sutton; Abran Costales; Martin Sendzik; Cynthia Shafer; Julian Levell; Gang Liu; Thomas Caferro; Young Shin Cho; Mark G. Palermo; Gregg Chenail; Julia Dooley; Brian Villalba; Ali Farsidjani; Jinyun Chen; Stephanie Dodd; Ty Gould; Guiqing Liang; Kelly Slocum; Minying Pu; Brant Firestone; Joseph D. Growney; Tycho Heimbach; Raymond Pagliarini

Mutant isocitrate dehydrogenase 1 (IDH1) is an attractive therapeutic target for the treatment of various cancers such as AML, glioma, and glioblastoma. We have evaluated 3-pyrimidin-4-yl-oxazolidin-2-ones as mutant IDH1 inhibitors that bind to an allosteric, induced pocket of IDH1R132H. This Letter describes SAR exploration focused on improving both the in vitro and in vivo metabolic stability of the compounds, leading to the identification of 19 as a potent and selective mutant IDH1 inhibitor that has demonstrated brain penetration and excellent oral bioavailability in rodents. In a preclinical patient-derived IDH1 mutant xenograft tumor model study, 19 efficiently inhibited the production of the biomarker 2-HG.

Collaboration


Dive into the James Sutton's collaboration.

Researchain Logo
Decentralizing Knowledge