Network


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

Hotspot


Dive into the research topics where Karen McGovern is active.

Publication


Featured researches published by Karen McGovern.


Molecular Cancer Therapeutics | 2008

An orally bioavailable small-molecule inhibitor of Hedgehog signaling inhibits tumor initiation and metastasis in pancreatic cancer

Georg Feldmann; Volker Fendrich; Karen McGovern; Djahida Bedja; Savita Bisht; Hector Alvarez; Jan Bart M Koorstra; Nils Habbe; Collins Karikari; Michael Mullendore; Kathleen L. Gabrielson; Rajni Sharma; William Matsui; Anirban Maitra

Recent evidence suggests that blockade of aberrant Hedgehog signaling can be exploited as a therapeutic strategy for pancreatic cancer. Our previous studies using the prototype Hedgehog small-molecule antagonist cyclopamine had shown the striking inhibition of systemic metastases on Hedgehog blockade in spontaneously metastatic orthotopic xenograft models. Cyclopamine is a natural compound with suboptimal pharmacokinetics, which impedes clinical translation. In the present study, a novel, orally bioavailable small-molecule Hedgehog inhibitor, IPI-269609, was tested using in vitro and in vivo model systems. In vitro treatment of pancreatic cancer cell lines with IPI-269609 resembled effects observed using cyclopamine (i.e., Gli-responsive reporter knockdown, down-regulation of the Hedgehog target genes Gli1 and Ptch, as well as abrogation of cell migration and colony formation in soft agar). Single-agent IPI-269609 profoundly inhibited systemic metastases in orthotopic xenografts established from human pancreatic cancer cell lines, although Hedgehog blockade had minimal effect on primary tumor volume. The only discernible phenotype observed within the treated primary tumor was a significant reduction in the population of aldehyde dehydrogenase–bright cells, which we have previously identified as a clonogenic tumor-initiating population in pancreatic cancer. Selective ex vivo depletion of aldehyde dehydrogenase–bright cells with IPI-269609 was accompanied by significant reduction in tumor engraftment rates in athymic mice. Pharmacologic blockade of aberrant Hedgehog signaling might prove to be an effective therapeutic strategy for inhibition of systemic metastases in pancreatic cancer, likely through targeting subsets of cancer cells with tumor-initiating (“cancer stem cell”) properties. [Mol Cancer Ther 2008;7(9):2725–35]


Journal of Medicinal Chemistry | 2009

Discovery of a potent and orally active hedgehog pathway antagonist (IPI-926).

Martin R. Tremblay; Andre Lescarbeau; Michael J. Grogan; Eddy Tan; Grace Ruiting Lin; Brian C. Austad; Lin-Chen Yu; Mark L. Behnke; Somarajan J. Nair; Margit Hagel; Kerry White; James Conley; Joseph D. Manna; Teresa M. Alvarez-Diez; Jennifer Hoyt; Caroline N. Woodward; Jens R. Sydor; Melissa Pink; John R. Macdougall; Matthew Campbell; Jill Cushing; Jeanne Ferguson; Michael Curtis; Karen McGovern; Margaret Read; Vito J. Palombella; Julian Adams; Alfredo C. Castro

Recent evidence suggests that blocking aberrant hedgehog pathway signaling may be a promising therapeutic strategy for the treatment of several types of cancer. Cyclopamine, a plant Veratrum alkaloid, is a natural product antagonist of the hedgehog pathway. In a previous report, a seven-membered D-ring semisynthetic analogue of cyclopamine, IPI-269609 (2), was shown to have greater acid stability and better aqueous solubility compared to cyclopamine. Further modifications of the A-ring system generated three series of analogues with improved potency and/or solubility. Lead compounds from each series were characterized in vitro and evaluated in vivo for biological activity and pharmacokinetic properties. These studies led to the discovery of IPI-926 (compound 28), a novel semisynthetic cyclopamine analogue with substantially improved pharmaceutical properties and potency and a favorable pharmacokinetic profile relative to cyclopamine and compound 2. As a result, complete tumor regression was observed in a Hh-dependent medulloblastoma allograft model after daily oral administration of 40 mg/kg of compound 28.


Nature | 2016

PI3Kγ is a molecular switch that controls immune suppression

Megan M. Kaneda; Karen Messer; Natacha Ralainirina; Hongying Li; Christopher J. Leem; Sara Gorjestani; Gyunghwi Woo; Abraham V. Nguyen; Camila C. Figueiredo; Philippe Foubert; Michael C. Schmid; Melissa Pink; David G. Winkler; Matthew Rausch; Vito J. Palombella; Jeffery L. Kutok; Karen McGovern; Kelly A. Frazer; Xuefeng Wu; Michael Karin; Roman Sasik; Ezra E.W. Cohen; Judith A. Varner

Macrophages play critical, but opposite, roles in acute and chronic inflammation and cancer. In response to pathogens or injury, inflammatory macrophages express cytokines that stimulate cytotoxic T cells, whereas macrophages in neoplastic and parasitic diseases express anti-inflammatory cytokines that induce immune suppression and may promote resistance to T cell checkpoint inhibitors. Here we show that macrophage PI 3-kinase γ controls a critical switch between immune stimulation and suppression during inflammation and cancer. PI3Kγ signalling through Akt and mTor inhibits NFκB activation while stimulating C/EBPβ activation, thereby inducing a transcriptional program that promotes immune suppression during inflammation and tumour growth. By contrast, selective inactivation of macrophage PI3Kγ stimulates and prolongs NFκB activation and inhibits C/EBPβ activation, thus promoting an immunostimulatory transcriptional program that restores CD8+ T cell activation and cytotoxicity. PI3Kγ synergizes with checkpoint inhibitor therapy to promote tumour regression and increased survival in mouse models of cancer. In addition, PI3Kγ-directed, anti-inflammatory gene expression can predict survival probability in cancer patients. Our work thus demonstrates that therapeutic targeting of intracellular signalling pathways that regulate the switch between macrophage polarization states can control immune suppression in cancer and other disorders.


Nature | 2016

Overcoming resistance to checkpoint blockade therapy by targeting PI3Kγ in myeloid cells

Olivier De Henau; Matthew Rausch; David W. Winkler; Luis Felipe Campesato; Cailian Liu; Daniel Hirschhorn Cymerman; Sadna Budhu; Arnab Ghosh; Melissa Pink; Jeremy Tchaicha; Mark Douglas; Thomas T. Tibbitts; Sujata Sharma; Jennifer Proctor; Nicole Kosmider; Kerry White; Howard M. Stern; John Soglia; Julian Adams; Vito J. Palombella; Karen McGovern; Jeffery L. Kutok; Jedd D. Wolchok; Taha Merghoub

Recent clinical trials using immunotherapy have demonstrated its potential to control cancer by disinhibiting the immune system. Immune checkpoint blocking (ICB) antibodies against cytotoxic-T-lymphocyte-associated protein 4 or programmed cell death protein 1/programmed death-ligand 1 have displayed durable clinical responses in various cancers. Although these new immunotherapies have had a notable effect on cancer treatment, multiple mechanisms of immune resistance exist in tumours. Among the key mechanisms, myeloid cells have a major role in limiting effective tumour immunity. Growing evidence suggests that high infiltration of immune-suppressive myeloid cells correlates with poor prognosis and ICB resistance. These observations suggest a need for a precision medicine approach in which the design of the immunotherapeutic combination is modified on the basis of the tumour immune landscape to overcome such resistance mechanisms. Here we employ a pre-clinical mouse model system and show that resistance to ICB is directly mediated by the suppressive activity of infiltrating myeloid cells in various tumours. Furthermore, selective pharmacologic targeting of the gamma isoform of phosphoinositide 3-kinase (PI3Kγ), highly expressed in myeloid cells, restores sensitivity to ICB. We demonstrate that targeting PI3Kγ with a selective inhibitor, currently being evaluated in a phase 1 clinical trial (NCT02637531), can reshape the tumour immune microenvironment and promote cytotoxic-T-cell-mediated tumour regression without targeting cancer cells directly. Our results introduce opportunities for new combination strategies using a selective small molecule PI3Kγ inhibitor, such as IPI-549, to overcome resistance to ICB in patients with high levels of suppressive myeloid cell infiltration in tumours.


Journal of Medicinal Chemistry | 2008

Semisynthetic cyclopamine analogues as potent and orally bioavailable hedgehog pathway antagonists.

Martin R. Tremblay; Marta Nevalainen; Somarajan J. Nair; James R. Porter; Alfredo C. Castro; Mark L. Behnke; Lin-Chen Yu; Margit Hagel; Kerry White; Kerrie Faia; Louis Grenier; Matthew Campbell; Jill Cushing; Caroline N. Woodward; Jennifer Hoyt; Michael Foley; Margaret Read; Jens R. Sydor; Jeffrey K. Tong; Vito J. Palombella; Karen McGovern; Julian Adams

Herein is reported the synthesis of a novel class of hedgehog antagonists derived from cyclopamine. The acid sensitive D-ring of cyclopamine was homologated utilizing a sequence of chemoselective cyclopropanation and stereoselective acid-catalyzed rearrangement. Further modification of the A/B-ring homoallylic alcohol to the conjugated ketone led to the discovery of new cyclopamine analogues with improved pharmaceutical properties and in vitro potency (EC 50) ranging from 10 to 1000 nM.


PLOS ONE | 2010

Self-Renewal of Acute Lymphocytic Leukemia Cells Is Limited by the Hedgehog Pathway Inhibitors Cyclopamine and IPI-926

Tara L. Lin; Qiuju H. Wang; Patrick Brown; Craig D. Peacock; Akil Merchant; Sarah Brennan; Evan Jones; Karen McGovern; D. Neil Watkins; Kathleen M. Sakamoto; William Matsui

Conserved embryonic signaling pathways such as Hedgehog (Hh), Wingless and Notch have been implicated in the pathogenesis of several malignancies. Recent data suggests that Hh signaling plays a role in normal B-cell development, and we hypothesized that Hh signaling may be important in precursor B-cell acute lymphocytic leukemia (B-ALL). We found that the expression of Hh pathway components was common in human B-ALL cell lines and clinical samples. Moreover, pathway activity could be modulated by Hh ligand or several pathway inhibitors including cyclopamine and the novel SMOOTHENED (SMO) inhibitor IPI-926. The inhibition of pathway activity primarily impacted highly clonogenic B-ALL cells expressing aldehyde dehydrogenase (ALDH) by limiting their self-renewal potential both in vitro and in vivo. These data demonstrate that Hh pathway activation is common in B-ALL and represents a novel therapeutic target regulating self-renewal and persistence of the malignant clone.


Current Opinion in Chemical Biology | 2010

New developments in the discovery of small molecule Hedgehog pathway antagonists

Martin R. Tremblay; Karen McGovern; Margaret Read; Alfredo C. Castro

The Hedgehog (Hh) signaling pathway is crucial for normal embryonic development. Aberrant Hh signaling is implicated in numerous pathologic conditions including proliferative diseases such as cancer. During the past decade, academic and industrial research efforts have resulted in the discovery of a variety of Hh pathway antagonists. This review focuses on the most recent advances in this field with particular emphasis on the medicinal chemistry approaches used to discover these Hh antagonists. While most of the small molecule modulators of the Hh pathway were discovered through screening and subsequent medicinal chemistry, a number of them originated from rational design or natural products.


Cancer | 2014

Involvement and targeted intervention of dysregulated Hedgehog signaling in osteosarcoma

Winnie W. Lo; Jay S. Wunder; Brendan C. Dickson; Veronica Campbell; Karen McGovern; Benjamin A. Alman; Irene L. Andrulis

During development, the Hedgehog pathway plays important roles regulating the proliferation and differentiation of chondrocytes, providing a template for growing bone. In this study, the authors investigated the components of dysregulated Hedgehog signaling as potential therapeutic targets for osteosarcoma.


Molecular Cancer Therapeutics | 2014

Hedgehog Pathway Inhibition in Chondrosarcoma Using the Smoothened Inhibitor IPI-926 Directly Inhibits Sarcoma Cell Growth

Veronica Campbell; Puviindran Nadesan; S. Amanda Ali; Chang Ye Yale Wang; Heather Whetstone; Raymond Poon; Qingxia Wei; John Keilty; Jennifer Proctor; Lauren W. Wang; Suneel S. Apte; Karen McGovern; Benjamin A. Alman; Jay S. Wunder

Hedgehog (Hh) pathway inhibition in cancer has been evaluated in both the ligand-independent and ligand-dependent settings, where Hh signaling occurs either directly within the cancer cells or within the nonmalignant cells of the tumor microenvironment. Chondrosarcoma is a malignant tumor of cartilage in which there is ligand-dependent activation of Hh signaling. IPI-926 is a potent, orally delivered small molecule that inhibits Hh pathway signaling by binding to Smoothened (SMO). Here, the impact of Hh pathway inhibition on primary chondrosarcoma xenografts was assessed. Mice bearing primary human chondrosarcoma xenografts were treated with IPI-926. The expression levels of known Hh pathway genes, in both the tumor and stroma, and endpoint tumor volumes were measured. Gene expression profiling of tumors from IPI-926–treated mice was conducted to identify potential novel Hh target genes. Hh target genes were studied to determine their contribution to the chondrosarcoma neoplastic phenotype. IPI-926 administration results in downmodulation of the Hh pathway in primary chondrosarcoma xenografts, as demonstrated by evaluation of the Hh target genes GLI1 and PTCH1, as well as inhibition of tumor growth. Chondrosarcomas exhibited autocrine and paracrine Hh signaling, and both were affected by IPI-926. Decreased tumor growth is accompanied by histopathologic changes, including calcification and loss of tumor cells. Gene profiling studies identified genes differentially expressed in chondrosarcomas following IPI-926 treatment, one of which, ADAMTSL1, regulates chondrosarcoma cell proliferation. These studies provide further insight into the role of the Hh pathway in chondrosarcoma and provide a scientific rationale for targeting the Hh pathway in chondrosarcoma. Mol Cancer Ther; 13(5); 1259–69. ©2014 AACR.


Science Translational Medicine | 2015

Integration of Hedgehog and mutant FLT3 signaling in myeloid leukemia

Yiting Lim; Lukasz P. Gondek; Li Li; Qiuju Wang; Haley Ma; Emily Chang; David L. Huso; Sarah Foerster; Luigi Marchionni; Karen McGovern; David Neil Watkins; Craig D. Peacock; Mark Levis; B. D. Smith; Akil Merchant; Donald M. Small; William Matsui

Activation of the Hedgehog pathway drives FLT3-mutated leukemia, and dual pathway inhibition effectively inhibits tumor growth. Hedgehog to the rescue Acute myeloid leukemia is generally difficult to treat, and the presence of internal tandem duplication in a gene called FLT3 (FLT3-ITD) is associated with a particularly poor prognosis. Lim et al. discovered that patients with FLT3-ITD leukemia also have increased activity of the Hedgehog protein signaling pathway. Experiments in mouse models of FLT3-ITD confirmed the functional role of Hedgehog in the development of leukemia and showed that combined treatment targeting FLT3 and Hedgehog is effective as a therapeutic strategy in this setting. FMS-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD) mutations resulting in constitutive kinase activity are common in acute myeloid leukemia (AML) and carry a poor prognosis. Several agents targeting FLT3 have been developed, but their limited clinical activity suggests that the inhibition of other factors contributing to the malignant phenotype is required. We examined gene expression data sets as well as primary specimens and found that the expression of GLI2, a major effector of the Hedgehog (Hh) signaling pathway, was increased in FLT3-ITD compared to wild-type FLT3 AML. To examine the functional role of the Hh pathway, we studied mice in which Flt3-ITD expression results in an indolent myeloproliferative state and found that constitutive Hh signaling accelerated the development of AML by enhancing signal transducer and activator of transcription 5 (STAT5) signaling and the proliferation of bone marrow myeloid progenitors. Furthermore, combined FLT3 and Hh pathway inhibition limited leukemic growth in vitro and in vivo, and this approach may serve as a therapeutic strategy for FLT3-ITD AML.

Collaboration


Dive into the Karen McGovern's collaboration.

Top Co-Authors

Avatar

Jeffery L. Kutok

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Margaret Read

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Howard M. Stern

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge