Network


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

Hotspot


Dive into the research topics where Kate McNamara is active.

Publication


Featured researches published by Kate McNamara.


Nature Medicine | 2008

Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers.

Jeffrey A. Engelman; Liang Chen; Xiaohong Tan; Katherine Crosby; Alexander R. Guimaraes; Rabi Upadhyay; S. Michel Maira; Kate McNamara; Samanthi A. Perera; Youngchul Song; Lucian R. Chirieac; Ramneet Kaur; Angela Lightbown; Jessica Simendinger; Timothy Q. Li; Robert F. Padera; Carlos Garcia-Echeverria; Ralph Weissleder; Umar Mahmood; Lewis C. Cantley; Kwok-Kin Wong

Somatic mutations that activate phosphoinositide 3-kinase (PI3K) have been identified in the p110-α catalytic subunit (encoded by PIK3CA). They are most frequently observed in two hotspots: the helical domain (E545K and E542K) and the kinase domain (H1047R). Although the p110-α mutants are transforming in vitro, their oncogenic potential has not been assessed in genetically engineered mouse models. Furthermore, clinical trials with PI3K inhibitors have recently been initiated, and it is unknown if their efficacy will be restricted to specific, genetically defined malignancies. In this study, we engineered a mouse model of lung adenocarcinomas initiated and maintained by expression of p110-α H1047R. Treatment of these tumors with NVP-BEZ235, a dual pan–PI3K and mammalian target of rapamycin (mTOR) inhibitor in clinical development, led to marked tumor regression as shown by positron emission tomography–computed tomography, magnetic resonance imaging and microscopic examination. In contrast, mouse lung cancers driven by mutant Kras did not substantially respond to single-agent NVP-BEZ235. However, when NVP-BEZ235 was combined with a mitogen-activated protein kinase kinase (MEK) inhibitor, ARRY-142886, there was marked synergy in shrinking these Kras-mutant cancers. These in vivo studies suggest that inhibitors of the PI3K-mTOR pathway may be active in cancers with PIK3CA mutations and, when combined with MEK inhibitors, may effectively treat KRAS mutated lung cancers.


Nature | 2007

LKB1 modulates lung cancer differentiation and metastasis.

Hongbin Ji; Matthew R. Ramsey; D. Neil Hayes; Cheng Fan; Kate McNamara; Piotr Kozlowski; Chad Torrice; Michael C. Wu; Takeshi Shimamura; Samanthi A. Perera; Mei Chih Liang; Dongpo Cai; George N. Naumov; Lei Bao; Cristina M. Contreras; Danan Li; Liang Chen; Janakiraman Krishnamurthy; Jussi Koivunen; Lucian R. Chirieac; Robert F. Padera; Roderick T. Bronson; Neal I. Lindeman; David C. Christiani; Xihong Lin; Geoffrey I. Shapiro; Pasi A. Jänne; Bruce E. Johnson; Matthew Meyerson; David J. Kwiatkowski

Germline mutation in serine/threonine kinase 11 (STK11, also called LKB1) results in Peutz–Jeghers syndrome, characterized by intestinal hamartomas and increased incidence of epithelial cancers. Although uncommon in most sporadic cancers, inactivating somatic mutations of LKB1 have been reported in primary human lung adenocarcinomas and derivative cell lines. Here we used a somatically activatable mutant Kras-driven model of mouse lung cancer to compare the role of Lkb1 to other tumour suppressors in lung cancer. Although Kras mutation cooperated with loss of p53 or Ink4a/Arf (also known as Cdkn2a) in this system, the strongest cooperation was seen with homozygous inactivation of Lkb1. Lkb1-deficient tumours demonstrated shorter latency, an expanded histological spectrum (adeno-, squamous and large-cell carcinoma) and more frequent metastasis compared to tumours lacking p53 or Ink4a/Arf. Pulmonary tumorigenesis was also accelerated by hemizygous inactivation of Lkb1. Consistent with these findings, inactivation of LKB1 was found in 34% and 19% of 144 analysed human lung adenocarcinomas and squamous cell carcinomas, respectively. Expression profiling in human lung cancer cell lines and mouse lung tumours identified a variety of metastasis-promoting genes, such as NEDD9, VEGFC and CD24, as targets of LKB1 repression in lung cancer. These studies establish LKB1 as a critical barrier to pulmonary tumorigenesis, controlling initiation, differentiation and metastasis.


Biological Psychiatry | 2006

Telomere Shortening and Mood Disorders: Preliminary Support for a Chronic Stress Model of Accelerated Aging

Naomi M. Simon; Jordan W. Smoller; Kate McNamara; Richard S. Maser; Alyson K. Zalta; Mark H. Pollack; Andrew A. Nierenberg; Maurizio Fava; Kwok-Kin Wong

BACKGROUND Little is known about the biological mechanisms underlying the excess medical morbidity and mortality associated with mood disorders. Substantial evidence supports abnormalities in stress-related biological systems in depression. Accelerated telomere shortening may reflect stress-related oxidative damage to cells and accelerated aging, and severe psychosocial stress has been linked to telomere shortening. We propose that chronic stress associated with mood disorders may contribute to excess vulnerability for diseases of aging such as cardiovascular disease and possibly some cancers through accelerated organismal aging. METHODS Telomere length was measured by Southern Analysis in 44 individuals with chronic mood disorders and 44 nonpsychiatrically ill age-matched control subjects. RESULTS Telomere length was significantly shorter in those with mood disorders, representing as much as 10 years of accelerated aging. CONCLUSIONS These results provide preliminary evidence that mood disorders are associated with accelerated aging and may suggest a novel mechanism for mood disorder-associated morbidity and mortality.


Nature | 2007

Chromosomally unstable mouse tumours have genomic alterations similar to diverse human cancers

Richard S. Maser; Bhudipa Choudhury; Peter J. Campbell; Bin Feng; Kwok-Kin Wong; Alexei Protopopov; Jennifer O'Neil; Alejandro Gutierrez; Elena Ivanova; Ilana Perna; Eric Lin; Vidya Mani; Shan Jiang; Kate McNamara; Sara Zaghlul; Sarah Edkins; Claire Stevens; Cameron Brennan; Eric Martin; Ruprecht Wiedemeyer; Omar Kabbarah; Cristina Nogueira; Gavin Histen; Marc R. Mansour; Veronique Duke; Letizia Foroni; Adele K. Fielding; Anthony H. Goldstone; Jacob M. Rowe; Yaoqi A. Wang

Highly rearranged and mutated cancer genomes present major challenges in the identification of pathogenetic events driving the neoplastic transformation process. Here we engineered lymphoma-prone mice with chromosomal instability to assess the usefulness of mouse models in cancer gene discovery and the extent of cross-species overlap in cancer-associated copy number aberrations. Along with targeted re-sequencing, our comparative oncogenomic studies identified FBXW7 and PTEN to be commonly deleted both in murine lymphomas and in human T-cell acute lymphoblastic leukaemia/lymphoma (T-ALL). The murine cancers acquire widespread recurrent amplifications and deletions targeting loci syntenic to those not only in human T-ALL but also in diverse human haematopoietic, mesenchymal and epithelial tumours. These results indicate that murine and human tumours experience common biological processes driven by orthologous genetic events in their malignant evolution. The highly concordant nature of genomic events encourages the use of genomically unstable murine cancer models in the discovery of biological driver events in the human oncogenome.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Epidermal growth factor receptor variant III mutations in lung tumorigenesis and sensitivity to tyrosine kinase inhibitors.

Hongbin Ji; Xiaojun Zhao; Yuki Yuza; Takeshi Shimamura; Danan Li; Alexei Protopopov; Boonim L. Jung; Kate McNamara; Huili Xia; Karen A. Glatt; Roman K. Thomas; Hidefumi Sasaki; James W. Horner; Michael J. Eck; Albert Mitchell; Yangping Sun; Ruqayyah Al-Hashem; Roderick T. Bronson; Sridhar K. Rabindran; Carolyn M. Discafani; Elizabeth A. Maher; Geoffrey I. Shapiro; Matthew Meyerson; Kwok-Kin Wong

The tyrosine kinase inhibitors gefitinib (Iressa) and erlotinib (Tarceva) have shown anti-tumor activity in the treatment of non-small cell lung cancer (NSCLC). Dramatic and durable responses have occurred in NSCLC tumors with mutations in the tyrosine kinase domain of the epidermal growth factor receptor (EGFR). In contrast, these inhibitors have shown limited efficacy in glioblastoma, where a distinct EGFR mutation, the variant III (vIII) in-frame deletion of exons 2–7, is commonly found. In this study, we determined that EGFRvIII mutation was present in 5% (3/56) of analyzed human lung squamous cell carcinoma (SCC) but was not present in human lung adenocarcinoma (0/123). We analyzed the role of the EGFRvIII mutation in lung tumorigenesis and its response to tyrosine kinase inhibition. Tissue-specific expression of EGFRvIII in the murine lung led to the development of NSCLC. Most importantly, these lung tumors depend on EGFRvIII expression for maintenance. Treatment with an irreversible EGFR inhibitor, HKI-272, dramatically reduced the size of these EGFRvIII-driven murine tumors in 1 week. Similarly, Ba/F3 cells transformed with the EGFRvIII mutant were relatively resistant to gefitinib and erlotinib in vitro but proved sensitive to HKI-272. These findings suggest a therapeutic strategy for cancers harboring the EGFRvIII mutation.


Cancer Cell | 2010

Integrative genomic and proteomic analyses identify targets for Lkb1-deficient metastatic lung tumors.

Julian Carretero; Takeshi Shimamura; Klarisa Rikova; Autumn L. Jackson; Matthew D. Wilkerson; Christa L. Borgman; Matthew S. Buttarazzi; Benjamin Sanofsky; Kate McNamara; Kathleyn A. Brandstetter; Zandra E. Walton; Ting Lei Gu; Katherine Crosby; Geoffrey I. Shapiro; Sauveur Michel Maira; Hongbin Ji; Diego H. Castrillon; Carla F. Kim; Carlos Garcia-Echeverria; Nabeel Bardeesy; Norman E. Sharpless; Neil Hayes; William Y. Kim; Jeffrey A. Engelman; Kwok-Kin Wong

In mice, Lkb1 deletion and activation of Kras(G12D) results in lung tumors with a high penetrance of lymph node and distant metastases. We analyzed these primary and metastatic de novo lung cancers with integrated genomic and proteomic profiles, and have identified gene and phosphoprotein signatures associated with Lkb1 loss and progression to invasive and metastatic lung tumors. These studies revealed that SRC is activated in Lkb1-deficient primary and metastatic lung tumors, and that the combined inhibition of SRC, PI3K, and MEK1/2 resulted in synergistic tumor regression. These studies demonstrate that integrated genomic and proteomic analyses can be used to identify signaling pathways that may be targeted for treatment.


European Neuropsychopharmacology | 2008

A Detailed Examination of Cytokine Abnormalities in Major Depressive Disorder

Naomi M. Simon; Kate McNamara; Candice W. Chow; Richard S. Maser; George I. Papakostas; Mark H. Pollack; Andrew A. Nierenberg; Maurizio Fava; Kwok-Kin Wong

Recent technological advances offer an opportunity to further elucidate the complex cytokine network in Major Depressive Disorder (MDD). Twenty cytokines were simultaneously assessed in 49 individuals with MDD and 49 age and gender matched controls. Multiple pro-inflammatory and two anti-inflammatory cytokines were significantly elevated in the MDD sample, including an antidepressant naïve subset. These data support a generalized chronic inflammatory state in MDD, and implicate additional cytokines and chemokines previously linked to cardiovascular disease.


Oncogene | 2006

K-ras activation generates an inflammatory response in lung tumors

Hongbin Ji; Houghton Am; Thomas J. Mariani; Samanthi A. Perera; Kim Cb; Robert F. Padera; Giovanni Tonon; Kate McNamara; Marconcini La; El-Bardeesy N; Roderick T. Bronson; David J. Sugarbaker; Richard S. Maser; Steven D. Shapiro; Kwok-Kin Wong

Activating mutations in K-ras are one of the most common genetic alterations in human lung cancer. To dissect the role of K-ras activation in bronchial epithelial cells during lung tumorigenesis, we created a model of lung adenocarcinoma by generating a conditional mutant mouse with both Clara cell secretory protein (CC10)-Cre recombinase and the Lox-Stop-Lox K-rasG12D alleles. The activation of K-ras mutant allele in CC10 positive cells resulted in a progressive phenotype characterized by cellular atypia, adenoma and ultimately adenocarcinoma. Surprisingly, K-ras activation in the bronchiolar epithelium is associated with a robust inflammatory response characterized by an abundant infiltration of alveolar macrophages and neutrophils. These mice displayed early mortality in the setting of this pulmonary inflammatory response with a median survival of 8 weeks. Bronchoalveolar lavage fluid from these mutant mice contained the MIP-2, KC, MCP-1 and LIX chemokines that increased significantly with age. Cell lines derived from these tumors directly produced MIP-2, LIX and KC. This model demonstrates that K-ras activation in the lung induces the elaboration of inflammatory chemokines and provides an excellent means to further study the complex interactions between inflammatory cells, chemokines and tumor progression.


Cancer Research | 2007

Mutations in BRAF and KRAS Converge on Activation of the Mitogen-Activated Protein Kinase Pathway in Lung Cancer Mouse Models

Hongbin Ji; Zhenxiong Wang; Samanthi A. Perera; Danan Li; Mei Chih Liang; Sara Zaghlul; Kate McNamara; Liang Chen; Mitchell Albert; Yanping Sun; Ruqayyah Al-Hashem; Lucian R. Chirieac; Robert F. Padera; Roderick T. Bronson; Roman K. Thomas; Levi A. Garraway; Pasi A. Jänne; Bruce E. Johnson; Lynda Chin; Kwok-Kin Wong

Mutations in the BRAF and KRAS genes occur in approximately 1% to 2% and 20% to 30% of non-small-cell lung cancer patients, respectively, suggesting that the mitogen-activated protein kinase (MAPK) pathway is preferentially activated in lung cancers. Here, we show that lung-specific expression of the BRAF V600E mutant induces the activation of extracellular signal-regulated kinase (ERK)-1/2 (MAPK) pathway and the development of lung adenocarcinoma with bronchioloalveolar carcinoma features in vivo. Deinduction of transgene expression led to dramatic tumor regression, paralleled by dramatic dephosphorylation of ERK1/2, implying a dependency of BRAF-mutant lung tumors on the MAPK pathway. Accordingly, in vivo pharmacologic inhibition of MAPK/ERK kinase (MEK; MAPKK) using a specific MEK inhibitor, CI-1040, induced tumor regression associated with inhibition of cell proliferation and induction of apoptosis in these de novo lung tumors. CI-1040 treatment also led to dramatic tumor shrinkage in murine lung tumors driven by a mutant KRas allele. Thus, somatic mutations in different signaling intermediates of the same pathway induce exquisite dependency on a shared downstream effector. These results unveil a potential common vulnerability of BRAF and KRas mutant lung tumors that potentially affects rational deployment of MEK targeted therapies to non-small-cell lung cancer patients.


Cancer Research | 2008

Hsp90 Inhibition Suppresses Mutant EGFR-T790M Signaling and Overcomes Kinase Inhibitor Resistance

Takeshi Shimamura; Danan Li; Hongbin Ji; Henry J. Haringsma; Elizabeth Liniker; Christa L. Borgman; April M. Lowell; Yuko Minami; Kate McNamara; Samanthi A. Perera; Sara Zaghlul; Roman K. Thomas; Heidi Greulich; Susumu Kobayashi; Lucian R. Chirieac; Robert F. Padera; Shigeto Kubo; Masaya Takahashi; Daniel G. Tenen; Matthew Meyerson; Kwok-Kin Wong; Geoffrey I. Shapiro

The epidermal growth factor receptor (EGFR) secondary kinase domain T790M non-small cell lung cancer (NSCLC) mutation enhances receptor catalytic activity and confers resistance to the reversible tyrosine kinase inhibitors gefitinib and erlotinib. Currently, irreversible inhibitors represent the primary approach in clinical use to circumvent resistance. We show that higher concentrations of the irreversible EGFR inhibitor CL-387,785 are required to inhibit EGFR phosphorylation in T790M-expressing cells compared with EGFR mutant NSCLC cells without T790M. Additionally, CL-387,785 does not fully suppress phosphorylation of other activated receptor tyrosine kinases (RTK) in T790M-expressing cells. These deficiencies result in residual Akt and mammalian target of rapamycin (mTOR) activities. Full suppression of EGFR-mediated signaling in T790M-expressing cells requires the combination of CL-387,785 and rapamycin. In contrast, Hsp90 inhibition overcomes these limitations in vitro and depletes cells of EGFR, other RTKs, and phospho-Akt and inhibits mTOR signaling whether or not T790M is present. EGFR-T790M-expressing cells rendered resistant to CL-387,785 by a kinase switch mechanism retain sensitivity to Hsp90 inhibition. Finally, Hsp90 inhibition causes regression in murine lung adenocarcinomas driven by mutant EGFR (L858R) with or without T790M. However, efficacy in the L858R-T790M model requires a more intense treatment schedule and responses were transient. Nonetheless, these findings suggest that Hsp90 inhibitors may be effective in T790M-expressing cells and offer an alternative therapeutic strategy for this subset of lung cancers.

Collaboration


Dive into the Kate McNamara's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert F. Padera

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Hongbin Ji

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lucian R. Chirieac

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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge