Network


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

Hotspot


Dive into the research topics where Catherine E. Ellis is active.

Publication


Featured researches published by Catherine E. Ellis.


Nature | 1999

Human urotensin-II is a potent vasoconstrictor and agonist for the orphan receptor GPR14

Robert S. Ames; Henry M. Sarau; Johathan K. Chambers; Robert N. Willette; Nambi Aiyar; Anne M. Romanic; Calvert Louden; James J. Foley; Charles F. Sauermelch; Robert W. Coatney; Zhaohui Ao; Jyoti Disa; Stephen Dudley Holmes; Jeffrey M. Stadel; John D. Martin; Wu-Schyong Liu; George I. Glover; Shelagh Wilson; Dean E. McNulty; Catherine E. Ellis; Nabil Elshourbagy; Usman Shabon; John J. Trill; Douglas W. P. Hay; Eliot H. Ohlstein; Derk J. Bergsma; Stephen A. Douglas

Urotensin-II (U-II) is a vasoactive ‘somatostatin-like’ cyclic peptide which was originally isolated from fish spinal cords, and which has recently been cloned from man. Here we describe the identification of an orphan human G-protein-coupled receptor homologous to rat GPR14 (refs 4, 5) and expressed predominantly in cardiovascular tissue, which functions as a U-II receptor. Goby and human U-II bind to recombinant human GPR14 with high affinity, and the binding is functionally coupled to calcium mobilization. Human U-II is found within both vascular and cardiac tissue (including coronary atheroma) and effectively constricts isolated arteries from non-human primates. The potency of vasoconstriction of U-II is an order of magnitude greater than that of endothelin-1, making human U-II the most potent mammalian vasoconstrictor identified so far. In vivo, human U-II markedly increases total peripheral resistance in anaesthetized non-human primates, a response associated with profound cardiac contractile dysfunction. Furthermore, as U-II immunoreactivity is also found within central nervous system and endocrine tissues, it may have additional activities.


Journal of Clinical Oncology | 2010

Randomized study of lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer

Kimberly L. Blackwell; Harold J. Burstein; Anna Maria Storniolo; Hope S. Rugo; George W. Sledge; Maria Koehler; Catherine E. Ellis; Michelle Casey; Svetislava J. Vukelja; Joachim Bischoff; José Baselga; Joyce O'Shaughnessy

PURPOSE Preclinical studies in ErbB2-positive cell lines demonstrated a synergistic interaction between lapatinib and trastuzumab, suggesting that dual blockade is more effective than a single agent alone. EGF104900 compared the activity of lapatinib alone or in combination with trastuzumab in patients with ErbB2-positive, trastuzumab-refractory metastatic breast cancer (MBC). PATIENTS AND METHODS Patients with ErbB2-positive MBC who experienced progression on prior trastuzumab-containing regimens were randomly assigned to receive either lapatinib alone or in combination with trastuzumab. The primary end point was progression-free survival (PFS). Secondary efficacy end points included overall response rate (ORR), clinical benefit rate (CBR; complete response, partial response, and stable disease for >/= 24 weeks), and overall survival (OS). RESULTS In the intent-to-treat population (N = 296) who received a median of three prior trastuzumab-containing regimens, the combination of lapatinib with trastuzumab was superior to lapatinib alone for PFS (hazard ratio [HR] = 0.73; 95% CI, 0.57 to 0.93; P = .008) and CBR (24.7% in the combination arm v 12.4% in the monotherapy arm; P = .01). A trend for improved OS in the combination arm was observed (HR = 0.75; 95% CI, 0.53 to 1.07; P = .106). There was no difference in ORR (10.3% in the combination arm v 6.9% in the monotherapy arm; P = .46). The most frequent adverse events were diarrhea, rash, nausea, and fatigue; diarrhea was higher in the combination arm (P = .03). The incidence of symptomatic and asymptomatic cardiac events was low (combination therapy = 2% and 3.4%; monotherapy = 0.7% and 1.4%, respectively). CONCLUSION Despite disease progression on prior trastuzumab-based therapy, lapatinib in combination with trastuzumab significantly improved PFS and CBR versus lapatinib alone, thus offering a chemotherapy-free option with an acceptable safety profile to patients with ErbB2-positive MBC.


Nature | 1999

Melanin-concentrating hormone is the cognate ligand for the orphan G-protein-coupled receptor SLC-1

Jon Chambers; Robert S. Ames; Derk J. Bergsma; Alison Muir; Laura R. Fitzgerald; Guillaume Hervieu; George M. Dytko; James J. Foley; John Martin; Wu-Schyong Liu; Janet Park; Catherine E. Ellis; Subinay Ganguly; Susan Konchar; Jane E. Cluderay; Ron A. Leslie; Shelagh Wilson; Henry M. Sarau

The underlying causes of obesity are poorly understood but probably involve complex interactions between many neurotransmitter and neuropeptide systems involved in the regulation of food intake and energy balance. Three pieces of evidence indicate that the neuropeptide melanin-concentrating hormone (MCH) is an important component of this system. First, MCH stimulates feeding when injected directly into rat brains,; second, the messenger RNA for the MCH precursor is upregulated in the hypothalamus of genetically obese mice and in fasted animals; and third, mice lacking MCH eat less and are lean. MCH antagonists might, therefore, provide a treatment for obesity. However, the development of such molecules has been hampered because the identity of the MCH receptor has been unknown until now. Here we show that the 353-amino-acid human orphan G-protein-coupled receptor SLC-1 (ref. 4) expressed in HEK293 cells binds MCH with sub-nanomolar affinity, and is stimulated by MCH to mobilize intracellular Ca2+ and reduce forskolin-elevated cyclic AMP levels. We also show that SLC-1 messenger RNA and protein is expressed in the ventromedial and dorsomedial nuclei of the hypothalamus, consistent with a role for SLC-1 in mediating the effects of MCH on feeding.


Journal of Clinical Oncology | 2012

Overall Survival Benefit With Lapatinib in Combination With Trastuzumab for Patients With Human Epidermal Growth Factor Receptor 2–Positive Metastatic Breast Cancer: Final Results From the EGF104900 Study

Kimberly L. Blackwell; Harold J. Burstein; Anna Maria Storniolo; Hope S. Rugo; George W. Sledge; Gursel Aktan; Catherine E. Ellis; Allison Florance; Svetislava J. Vukelja; Joachim Bischoff; José Baselga; Joyce O'Shaughnessy

PURPOSE Phase III EGF104900 data demonstrated that lapatinib plus trastuzumab significantly improved progression-free survival (PFS) and clinical benefit rate versus lapatinib monotherapy, offering a chemotherapy-free option for patients with heavily pretreated human epidermal growth factor receptor 2 (HER2) -positive metastatic breast cancer (MBC). Final planned overall survival (OS) analysis from EGF104900 is reported here. PATIENTS AND METHODS Patients with HER2-positive MBC whose disease progressed during prior trastuzumab-based therapies were randomly assigned to receive lapatinib monotherapy or lapatinib in combination with trastuzumab. OS and updated PFS data are presented using Kaplan-Meier curves and log-rank tests stratified for hormone receptor and visceral disease status. Subgroup analyses were conducted to identify characteristics of patients deriving the greatest clinical benefit. RESULTS In this updated final analysis of all patients randomly assigned with strata (n = 291), lapatinib plus trastuzumab continued to show superiority to lapatinib monotherapy in PFS (hazard ratio [HR], 0.74; 95% CI, 0.58 to 0.94; P = .011) and offered significant OS benefit (HR, 0.74; 95% CI, 0.57 to 0.97; P = .026). Improvements in absolute OS rates were 10% at 6 months and 15% at 12 months in the combination arm compared with the monotherapy arm. Multiple baseline factors, including Eastern Cooperative Oncology Group performance status of 0, nonvisceral disease, < three metastatic sites, and less time from initial diagnosis until random assignment, were associated with improved OS. Incidence of adverse events was consistent with previously reported rates. CONCLUSION These data demonstrated a significant 4.5-month median OS advantage with the lapatinib and trastuzumab combination and support dual HER2 blockade in patients with heavily pretreated HER2-positive MBC.


Journal of Neurochemistry | 2003

Pharmacological and immunohistochemical characterization of the APJ receptor and its endogenous ligand apelin

Andrew D. Medhurst; Carol A. Jennings; Melanie J. Robbins; Robert P. Davis; Catherine E. Ellis; Kim Winborn; Kenneth W. M. Lawrie; Guillaume Hervieu; Graham J. Riley; Jane E. Bolaky; Nicole C. Herrity; Paul R. Murdock; John G. Darker

Apelin peptides have recently been identified to be the endogenous ligands for the G protein‐coupled receptor APJ. However, little is known about the physiological roles of this ligand‐receptor pairing. In the present study we investigated the pharmacology of several apelin analogues at the human recombinant APJ receptor using radioligand binding and functional assays. This has led to the identification of key residues in the apelin peptide required for functional potency and binding affinity through structure–activity studies. In particular, we have identified that replacement of leucine in position 5, or arginine in position 2 and 4 of the C‐terminal apelin peptide, apelin‐13, resulted in significant changes in pharmacology. We also investigated the detailed localization of pre‐proapelin and APJ receptor mRNA in a wide range of human, rat and mouse tissues using quantitative RT–PCR, and carried out a detailed immunohistochemical study of the distribution of the APJ receptor in rat brain and spinal cord. Interestingly, the APJ receptor was not only co‐localized in white matter with GFAP in the spinal cord, but was also clearly localized on neurones in the brain, suggesting that this receptor and its peptide may be involved in a wide range of biological process yet to be determined.


British Journal of Pharmacology | 1998

Orphan G-protein-coupled receptors : the next generation of drug targets?

Shelagh Wilson; Derk J. Bergsma; Jon Chambers; Alison Muir; Kenneth G. M. Fantom; Catherine E. Ellis; Paul R. Murdock; Nicole C. Herrity; Jeffrey M. Stadel

The pharmaceutical industry has readily embraced genomics to provide it with new targets for drug discovery. Large scale DNA sequencing has allowed the identification of a plethora of DNA sequences distantly related to known G protein‐coupled receptors (GPCRs), a superfamily of receptors that have a proven history of being excellent therapeutic targets. In most cases the extent of sequence homology is insufficient to assign these ‘orphan’ receptors to a particular receptor subfamily. Consequently, reverse molecular pharmacological and functional genomic strategies are being employed to identify the activating ligands of the cloned receptors. Briefly, the reverse molecular pharmacological methodology includes cloning and expression of orphan GPCRs in mammalian cells and screening these cells for a functional response to cognate or surrogate agonists present in biological extract preparations, peptide libraries, and complex compound collections. The functional genomics approach involves the use of humanized yeast cells, where the yeast GPCR transduction system is engineered to permit functional expression and coupling of human GPCRs to the endogenous signalling machinery. Both systems provide an excellent platform for identifying novel receptor ligands. Once activating ligands are identified they can be used as pharmacological tools to explore receptor function and relationship to disease.


JAMA Oncology | 2015

Tumor-Infiltrating Lymphocytes and Associations With Pathological Complete Response and Event-Free Survival in HER2-Positive Early-Stage Breast Cancer Treated With Lapatinib and Trastuzumab A Secondary Analysis of the NeoALTTO Trial

Roberto Salgado; Carsten Denkert; Christine Campbell; Peter Savas; Paolo P Nucifero; Claudia Aura; Evandro de Azambuja; Holger Eidtmann; Catherine E. Ellis; José Baselga; Martine Piccart-Gebhart; Stefan Michiels; Ian Bradbury; Christos Sotiriou; Sherene Loi

IMPORTANCE The presence of tumor-infiltrating lymphocytes (TILs) is associated with improved outcomes in human epidermal growth factor receptor 2 (HER2)-positive early breast cancer treated with adjuvant trastuzumab and chemotherapy. The prognostic associations in the neoadjuvant setting of other anti-HER2 agents and combinations are unknown. OBJECTIVE To determine associations between presence of TILs, pathological complete response (pCR), and event-free survival (EFS) end points in patients with early breast cancer treated with trastuzumab, lapatinib, or the combination. DESIGN, SETTING, AND PARTICIPANTS The NeoALTTO trial (Neoadjuvant Lapatinib and/or Trastuzumab Treatment Optimization) randomly assigned 455 women with HER2-positive early-stage breast cancer between January 5, 2008, and May 27, 2010, to 1 of 3 neoadjuvant treatment arms: trastuzumab, lapatinib, or the combination for 6 weeks followed by the addition of weekly paclitaxel for 12 weeks, followed by 3 cycles of fluorouracil, epirubicin, and cyclophosphamide after surgery. The primary end point used in this study was pCR in the breast and lymph nodes, with a secondary end point of EFS. We evaluated levels of percentage of TILs using hematoxylin-eosin-stained core biopsy sections taken at diagnosis (prior to treatment) in a prospectively defined retrospective analysis. MAIN OUTCOMES AND MEASURES Levels of TILs were examined for their associations with efficacy end points adjusted for prognostic clinicopathological factors including PIK3CA genotype. RESULTS Of the 455 patients, 387 (85.1%) tumor samples were used for the present analysis. The median (interquartile range [IQR]) level of TILs was 12.5% (5.0%-30.0%), with levels lower in hormone receptor-positive (10.0% [5.0%-22.5%]) vs hormone receptor-negative (12.5% [3.0%-35.0%]) samples (P = .02). For the pCR end point, levels of TILs greater than 5% were associated with higher pCR rates independent of treatment group (adjusted odds ratio, 2.60 [95% CI, 1.26-5.39]; P = .01). With a median (IQR) follow-up time of 3.77 (3.50-4.22) years, every 1% increase in TILs was associated with a 3% decrease in the rate of an event (adjusted hazard ratio, 0.97 [95% CI, 0.95-0.99]; P = .002) across all treatment groups. CONCLUSIONS AND RELEVANCE The presence of TILs at diagnosis is an independent, positive, prognostic marker in HER2-positive early breast cancer treated with neoadjuvant anti-HER2 agents and chemotherapy for both pCR and EFS end points. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00553358.


Clinical Cancer Research | 2010

Clinical Benefit of Lapatinib-Based Therapy in Patients with Human Epidermal Growth Factor Receptor 2–Positive Breast Tumors Coexpressing the Truncated p95HER2 Receptor

Maurizio Scaltriti; Sarat Chandarlapaty; Ludmila Prudkin; Claudia Aura; José M. Jiménez; Pier Davide Angelini; Gertrudis Sánchez; Marta Guzman; Josep Lluis Parra; Catherine E. Ellis; Robert Gagnon; Maria Koehler; Henry Gomez; Charles E. Geyer; David A Cameron; J. Arribas; Neal Rosen; José Baselga

Purpose: A subgroup of human epidermal growth factor receptor 2 (HER2)–overexpressing breast tumors coexpresses p95HER2, a truncated HER2 receptor that retains a highly functional HER2 kinase domain but lacks the extracellular domain and results in intrinsic trastuzumab resistance. We hypothesized that lapatinib, a HER2 tyrosine kinase inhibitor, would be active in these tumors. We have studied the correlation between p95HER2 expression and response to lapatinib, both in preclinical models and in the clinical setting. Experimental Design: Two different p95HER2 animal models were used for preclinical studies. Expression of p95HER2 was analyzed in HER2-overexpressing breast primary tumors from a first-line lapatinib monotherapy study (EGF20009) and a second-line lapatinib in combination with capecitabine study (EGF100151). p95HER2 expression was correlated with overall response rate (complete + partial response), clinical benefit rate (complete response + partial response + stable disease ≥24 wk), and progression-free survival using logistic regression and Cox proportional hazard models. Results: Lapatinib inhibited tumor growth and the HER2 downstream signaling of p95HER2-expressing tumors. A total of 68 and 156 tumors from studies EGF20009 and EGF100151 were evaluable, respectively, for p95HER2 detection. The percentage of p95HER2-positive patients was 20.5% in the EGF20009 study and 28.5% in the EGF100151 study. In both studies, there was no statistically significant difference in progression-free survival, clinical benefit rate, and overall response rate between p95HER2-positive and p95HER2-negative tumors. Conclusions: Lapatinib as a monotherapy or in combination with capecitabine seems to be equally effective in patients with p95HER2-positive and p95HER2-negative HER2-positive breast tumors. Clin Cancer Res; 16(9); 2688–95. ©2010 AACR.


Journal of Clinical Oncology | 2015

PIK3CA Mutations Are Associated With Decreased Benefit to Neoadjuvant Human Epidermal Growth Factor Receptor 2–Targeted Therapies in Breast Cancer

Ian Majewski; Paolo Nuciforo; Lorenza Mittempergher; Astrid Bosma; Holger Eidtmann; Eileen Holmes; Christos Sotiriou; Debora Fumagalli; Jose L. Jimenez; Claudia Aura; Ludmila Prudkin; Maria Carmen Díaz-Delgado; Lorena de la Peña; Sherene Loi; Catherine E. Ellis; Nikolaus Schultz; Evandro de Azambuja; Nadia Harbeck; Martine Piccart-Gebhart; René Bernards; José Baselga

PURPOSE We investigated whether mutations in the gene encoding the phosphatidylinositol 3-kinase (PI3K) catalytic subunit (PIK3CA) correlates with response to neoadjuvant human epidermal growth factor receptor 2 (HER2) -targeted therapies in patients with breast cancer. PATIENTS AND METHODS Baseline tissue biopsies were available from patients with HER2-positive early breast cancer who were enrolled onto the Neoadjuvant Lapatinib and/or Trastuzumab Treatment Optimization trial (NeoALTTO). Activating mutations in PIK3CA were identified using mass spectrometry-based genotyping. RESULTS PIK3CA mutations were identified in 23% of HER2-positive breast tumors, and these mutations were associated with poorer outcome in all of the treatment arms. Patients treated with a combination of trastuzumab and lapatinib who had wild-type PIK3CA obtained a total pathologic complete response (pCR) rate of 53.1%, which decreased to 28.6% in patients with tumors that carried PIK3CA activating mutations (P = .012). CONCLUSION Activating mutations in PIK3CA predicted poor pCR in patients with HER2-positive breast cancer treated with neoadjuvant therapies that target HER2. Consequently, the combination of anti-HER2 agents and PI3K inhibitors is being investigated.


British Journal of Pharmacology | 1998

Functional characterisation of the human cloned 5‐HT7 receptor (long form); antagonist profile of SB‐258719

David R. Thomas; Susan A. Gittins; Lissa L. Collin; Derek N. Middlemiss; Graham J. Riley; Jim J. Hagan; Israel Simon Gloger; Catherine E. Ellis; Ian Thomson Forbes; Anthony M. Brown

1 The functional profile of the long form of the human cloned 5‐HT7 receptor (designated h5‐HT7(a)) was investigated using a number of 5‐HT receptor agonists and antagonists and compared with its binding profile. Receptor function was measured using adenylyl cyclase activity in washed membranes from HEK293 cells stably expressing the recombinant h5‐HT7(a) receptor. 2 The receptor binding profile, determined by competition with [3H]‐5‐CT, was consistent with that previously reported for the h5‐HT7(a) receptor. The selective 5‐HT7 receptor antagonist SB‐258719 ((R)‐3,N‐Dimethyl‐N‐[1‐methyl‐3‐(4‐methylpiperidin‐1‐yl)propyl]benzene sulfonamide) displayed high affinity (pKi 7.5) for the receptor. 3 In the adenylyl cyclase functional assay, 5‐CT and 8‐OH‐DPAT were both full agonists compared to 5‐HT and the rank order of potency for agonists (5‐CT>5‐HT>8‐OH‐DPAT) was the same in functional and binding studies. 4 Risperidone, methiothepin, mesulergine, clozapine, olanzapine, ketanserin and SB‐258719 antagonised surmountably 5‐CT‐stimulated adenylyl cyclase activity. Schild analysis of the antagonism by SB‐258719 gave a pA2 of 7.2±0.2 and slope not significantly different from 1, consistent with competitive antagonism. 5 The same antagonists also inhibited basal adenylyl cyclase activity with a rank order of potency in agreement with those for antagonist potency and binding affinity. Both SB‐258719 and mesulergine displayed apparent partial inverse agonist profiles compared to the other antagonists tested. These inhibitory effects of antagonists appear to be 5‐HT7 receptor‐mediated and to reflect inverse agonism. 6 It is concluded that in this expression system, the h5‐HT7(a) receptor shows the expected binding and functional profile and displays constitutive activity, revealing inverse agonist activity for a range of antagonists.

Collaboration


Dive into the Catherine E. Ellis's collaboration.

Top Co-Authors

Avatar

José Baselga

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ludmila Prudkin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maurizio Scaltriti

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael F. Press

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge