Network


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

Hotspot


Dive into the research topics where Elizabeth Luis is active.

Publication


Featured researches published by Elizabeth Luis.


Nature Immunology | 2011

IL-17C regulates the innate immune function of epithelial cells in an autocrine manner

Vladimir Ramirez-Carrozzi; Arivazhagan Sambandam; Elizabeth Luis; Zhongua Lin; Surinder Jeet; Justin Lesch; Jason A. Hackney; Janice Kim; Meijuan Zhou; Joyce Lai; Zora Modrusan; Tao Sai; Wyne P. Lee; Min Xu; Patrick Caplazi; Lauri Diehl; Jason de Voss; Mercedesz Balazs; Lino C. Gonzalez; Harinder Singh; Wenjun Ouyang; Rajita Pappu

Interleukin 17C (IL-17C) is a member of the IL-17 family that is selectively induced in epithelia by bacterial challenge and inflammatory stimuli. Here we show that IL-17C functioned in a unique autocrine manner, binding to a receptor complex consisting of the receptors IL-17RA and IL-17RE, which was preferentially expressed on tissue epithelial cells. IL-17C stimulated epithelial inflammatory responses, including the expression of proinflammatory cytokines, chemokines and antimicrobial peptides, which were similar to those induced by IL-17A and IL-17F. However, IL-17C was produced by distinct cellular sources, such as epithelial cells, in contrast to IL-17A, which was produced mainly by leukocytes, especially those of the TH17 subset of helper T cells. Whereas IL-17C promoted inflammation in an imiquimod-induced skin-inflammation model, it exerted protective functions in dextran sodium sulfate–induced colitis. Thus, IL-17C is an essential autocrine cytokine that regulates innate epithelial immune responses.


Nature | 2015

Novel antibody–antibiotic conjugate eliminates intracellular S. aureus

Sophie M. Lehar; Thomas H. Pillow; Min Xu; Leanna Staben; Kimberly Kajihara; Richard Vandlen; Laura DePalatis; Helga Raab; Wouter L. W. Hazenbos; J. Hiroshi Morisaki; Janice Kim; Summer Park; Martine Darwish; Byoung-Chul Lee; Hilda Hernandez; Kelly M. Loyet; Patrick Lupardus; Rina Fong; Donghong Yan; Cecile Chalouni; Elizabeth Luis; Yana Khalfin; Emile Plise; Jonathan Cheong; Joseph P. Lyssikatos; Magnus Strandh; Klaus Koefoed; Peter S. Andersen; John A. Flygare; Man Wah Tan

Staphylococcus aureus is considered to be an extracellular pathogen. However, survival of S. aureus within host cells may provide a reservoir relatively protected from antibiotics, thus enabling long-term colonization of the host and explaining clinical failures and relapses after antibiotic therapy. Here we confirm that intracellular reservoirs of S. aureus in mice comprise a virulent subset of bacteria that can establish infection even in the presence of vancomycin, and we introduce a novel therapeutic that effectively kills intracellular S. aureus. This antibody–antibiotic conjugate consists of an anti-S. aureus antibody conjugated to a highly efficacious antibiotic that is activated only after it is released in the proteolytic environment of the phagolysosome. The antibody–antibiotic conjugate is superior to vancomycin for treatment of bacteraemia and provides direct evidence that intracellular S. aureus represents an important component of invasive infections.


Cancer Research | 2004

EphB2 as a Therapeutic Antibody Drug Target for the Treatment of Colorectal Cancer

Weiguang Mao; Elizabeth Luis; Sarajane Ross; Johnny Silva; Christine Tan; Craig Crowley; Clarissa J. Chui; Gretchen Franz; Peter D. Senter; Hartmut Koeppen; Paul Polakis

Analysis of human colorectal cancer specimens revealed overexpression of the EphB2 receptor tyrosine kinase. Monoclonal antibodies (MAbs) to extracellular sequence of EphB2 were raised and tested for activity against colorectal cancer cells. One of the MAbs, 2H9, effectively blocked the interaction of ephB2 with ephrin ligands and inhibited the resulting autophosphorylation of the receptor. However, this antibody did not affect the proliferation of cancer cells expressing ephB2. Immunocytochemical analysis revealed rapid internalization of the MAb 2H9 on binding ephB2, suggesting that target-dependent cell killing could be achieved with an antibody-drug conjugate. When MAb 2H9 was conjugated to monomethylauristatin E through a cathepsin B-cleavable linker, it specifically killed ephB2-expressing cancer cells in vitro and in vivo. Our results suggest that ephB2 is an attractive target for immunoconjugate cancer therapy.


Science Translational Medicine | 2011

Amelioration of Type 2 Diabetes by Antibody-Mediated Activation of Fibroblast Growth Factor Receptor 1

Ai-Luen Wu; Ganesh Kolumam; Scott Stawicki; Yongmei Chen; Jun Li; Jose Zavala-Solorio; Khanhky Phamluong; Bo Feng; Li Li; Scot A. Marsters; Lance Kates; Nicholas van Bruggen; Maya Leabman; Anne Wong; David West; Howard M. Stern; Elizabeth Luis; Hok Seon Kim; Daniel G. Yansura; Andrew S. Peterson; Ellen Filvaroff; Yan Wu; Junichiro Sonoda

Antibody-mediated activation of fibroblast growth factor receptor 1 reverses the diabetic phenotype in mice, likely by affecting brown adipose tissues. Getting at Brown Fat It’s fun to indulge in holiday cheer, if only a holiday miracle allowed one to avoid the often-linked weight gain. At the molecular level, obesity and type 2 diabetes can be linked by the fibroblast growth factor (FGF) family of proteins and their receptors (FGFRs), with some factors showing disease-reversing capabilities. For instance, overweight, diabetic mice treated with FGF21 regain normal metabolism and lose weight, even without spending hours on a treadmill. However, attempts to use this fat-burning factor in humans have not been successful, owing to poor pharmacokinetics as well as concerns over negative effects of modified FGF21 proteins. In this issue, Wu and colleagues describe an antibody-based FGF21 mimic that circumvents these limitations to overcome metabolic disease in mice. The authors reasoned that robust drugs that closely mimic FGF21 function would similarly exert antidiabetic effects. Using phage display technology, Wu et al. identified monoclonal antibodies (R1MAbs) that were specifically targeted tissues that play key roles in diabetes and obesity, including adipose (fat) tissue. In contrast to FGF21, which binds several forms of the FGFR throughout the body, the phage-derived R1MAbs bound only to FGFR1—a receptor present in the pancreas and in brown and white adipose tissues. Diabetic mice with high blood sugar (hyperglycemia) were injected once with either R1MAbs or a control antibody. Within 1 week, blood glucose concentrations in the R1MAb-treated mice were normalized and remained at lower levels compared to placebo-treated mice for more than 1 month without reaching dangerously low blood glucose concentrations (hypoglycemia). The R1MAbs also helped the diabetic mice to lose weight, indicating that this antibody agonist of FGFR1 is a dual-action drug for both diabetes and obesity. Wu et al. also shed light on the mechanism of action of their R1MAbs, showing that they work via FGFR homodimerization in brown adipose tissue. With improved pharmacokinetics over FGF21, in addition to a specific receptor-targeting mechanism, these R1MAbs could enter human clinical trials for diabetes and other obesity-related diseases in the near future. Unfortunately, a miracle drug won’t be available in time for the holidays, so perhaps, this year, opt for the sugar-free egg nog. Clinical use of recombinant fibroblast growth factor 21 (FGF21) for the treatment of type 2 diabetes and other disorders linked to obesity has been proposed; however, its clinical development has been challenging owing to its poor pharmacokinetics. Here, we describe an alternative antidiabetic strategy using agonistic anti-FGFR1 (FGF receptor 1) antibodies (R1MAbs) that mimic the metabolic effects of FGF21. A single injection of R1MAb into obese diabetic mice induced acute and sustained amelioration of hyperglycemia, along with marked improvement in hyperinsulinemia, hyperlipidemia, and hepatosteatosis. R1MAb activated the mitogen-activated protein kinase pathway in adipose tissues, but not in liver, and neither FGF21 nor R1MAb improved glucose clearance in lipoatrophic mice, which suggests that adipose tissues played a central role in the observed metabolic effects. In brown adipose tissues, both FGF21 and R1MAb induced phosphorylation of CREB (cyclic adenosine 5′-monophosphate response element–binding protein), and mRNA expression of PGC-1α (peroxisome proliferator–activated receptor-γ coactivator 1α) and the downstream genes associated with oxidative metabolism. Collectively, we propose FGFR1 in adipose tissues as a major functional receptor for FGF21, as an upstream regulator of PGC-1α, and as a compelling target for antibody-based therapy for type 2 diabetes and other obesity-associated disorders.


Developmental Cell | 2011

Robo4 Maintains Vessel Integrity and Inhibits Angiogenesis by Interacting with UNC5B

Alexander W. Koch; Thomas Mathivet; Bruno Larrivée; Raymond K. Tong; Joe Kowalski; Laurence Pibouin-Fragner; Karine Bouvrée; Scott Stawicki; Katrina Nicholes; Nisha Rathore; Suzie J. Scales; Elizabeth Luis; Raquel del Toro; Catarina Freitas; Christiane Bréant; Annie Michaud; Pierre Corvol; Jean-Léon Thomas; Yan Wu; Franklin Peale; Ryan J. Watts; Marc Tessier-Lavigne; Anil Bagri; Anne Eichmann

Robo4 is an endothelial cell-specific member of the Roundabout axon guidance receptor family. To identify Robo4 binding partners, we performed a protein-protein interaction screen with the Robo4 extracellular domain. We find that Robo4 specifically binds to UNC5B, a vascular Netrin receptor, revealing unexpected interactions between two endothelial guidance receptors. We show that Robo4 maintains vessel integrity by activating UNC5B, which inhibits signaling downstream of vascular endothelial growth factor (VEGF). Function-blocking monoclonal antibodies against Robo4 and UNC5B increase angiogenesis and disrupt vessel integrity. Soluble Robo4 protein inhibits VEGF-induced vessel permeability and rescues barrier defects in Robo4(-/-) mice, but not in mice treated with anti-UNC5B. Thus, Robo4-UNC5B signaling maintains vascular integrity by counteracting VEGF signaling in endothelial cells, identifying a novel function of guidance receptor interactions in the vasculature.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Molecular basis for negative regulation of the glucagon receptor

Christopher M. Koth; Jeremy Murray; Susmith Mukund; Azadeh Madjidi; Alexandra Minn; Holly J. Clarke; Terence Wong; Vicki Chiang; Elizabeth Luis; Alberto Estevez; Jesus Rondon; Yingnan Zhang; Isidro Hotzel; Bernard B. Allan

Members of the class B family of G protein-coupled receptors (GPCRs) bind peptide hormones and have causal roles in many diseases, ranging from diabetes and osteoporosis to anxiety. Although peptide, small-molecule, and antibody inhibitors of these GPCRs have been identified, structure-based descriptions of receptor antagonism are scarce. Here we report the mechanisms of glucagon receptor inhibition by blocking antibodies targeting the receptors extracellular domain (ECD). These studies uncovered a role for the ECD as an intrinsic negative regulator of receptor activity. The crystal structure of the ECD in complex with the Fab fragment of one antibody, mAb1, reveals that this antibody inhibits glucagon receptor by occluding a surface extending across the entire hormone-binding cleft. A second antibody, mAb23, blocks glucagon binding and inhibits basal receptor activity, indicating that it is an inverse agonist and that the ECD can negatively regulate receptor activity independent of ligand binding. Biochemical analyses of receptor mutants in the context of a high-resolution ECD structure show that this previously unrecognized inhibitory activity of the ECD involves an interaction with the third extracellular loop of the receptor and suggest that glucagon-mediated structural changes in the ECD accompany receptor activation. These studies have implications for the design of drugs to treat class B GPCR-related diseases, including the potential for developing novel allosteric regulators that target the ECDs of these receptors.


Molecular Cancer Therapeutics | 2009

Molecular predictors of response to a humanized anti–insulin-like growth factor-I receptor monoclonal antibody in breast and colorectal cancer

Jiping Zha; Carol O'Brien; Heidi Savage; Ling-Yuh Huw; Fiona Zhong; Leanne Berry; Gail Lewis Phillips; Elizabeth Luis; Guy Cavet; Xiaolan Hu; Lukas C. Amler; Mark R. Lackner

The insulin-like growth factor-I receptor (IGF-IR) pathway is required for the maintenance of the transformed phenotype in neoplastic cells and hence has been the subject of intensive drug discovery efforts. A key aspect of successful clinical development of targeted therapies directed against IGF-IR will be identification of responsive patient populations. Toward that end, we have endeavored to identify predictive biomarkers of response to an anti-IGF-IR-targeting monoclonal antibody in preclinical models of breast and colorectal cancer. We find that levels of the IGF-IR itself may have predictive value in these tumor types and identify other gene expression predictors of in vitro response. Studies in breast cancer models suggest that IGF-IR expression is both correlated and functionally linked with estrogen receptor signaling and provide a basis for both patient stratification and rational combination therapy with antiestrogen-targeting agents. In addition, we find that levels of other components of the signaling pathway such as the adaptor proteins IRS1 and IRS2, as well as the ligand IGF-II, have predictive value and report on the development of a pathway-focused panel of diagnostic biomarkers that could be used to test these hypotheses during clinical development of IGF-IR-targeting therapies. [Mol Cancer Ther 2009;8(8):2110–21]


Nuclear Medicine and Biology | 2010

Site-specifically 89Zr-labeled monoclonal antibodies for ImmunoPET

Jeff N. Tinianow; Herman S. Gill; Annie Ogasawara; Judith E. Flores; Alexander N. Vanderbilt; Elizabeth Luis; Richard Vandlen; Martine Darwish; Jagath R. Junutula; Simon-P. Williams; Jan Marik

UNLABELLED Three thiol reactive reagents were developed for the chemoselective conjugation of desferrioxamine (Df) to a monoclonal antibody via engineered cysteine residues (thio-trastuzumab). The in vitro stability and in vivo imaging properties of site-specifically radiolabeled (89)Zr-Df-thio-trastuzumab conjugates were investigated. METHODS The amino group of desferrioxamine B was acylated by bromoacetyl bromide, N-hydroxysuccinimidyl iodoacetate, or N-hydroxysuccinimidyl 4-[N-maleimidomethyl]cyclohexane-1-carboxylate to obtain thiol reactive reagents bromoacetyl-desferrioxamine (Df-Bac), iodoacetyl-desferrioxamine (Df-Iac) and maleimidocyclohexyl-desferrioxamine (Df-Chx-Mal), respectively. Df-Bac and Df-Iac alkylated the free thiol groups of thio-trastuzumab by nucleophilic substitution forming Df-Ac-thio-trastuzumab, while the maleimide reagent Df-Chx-Mal reacted via Michael addition to provide Df-Chx-Mal-thio-trastuzumab. The conjugates were radiolabeled with (89)Zr and evaluated for serum stability, and their positron emission tomography (PET) imaging properties were investigated in a BT474M1 (HER2-positive) breast tumor mouse model. RESULTS The chemoselective reagents were obtained in 14% (Df-Bac), 53% (Df-Iac) and 45% (Df-Chx-Mal) yields. Site-specific conjugation of Df-Chx-Mal to thio-trastuzumab was complete within 1 h at pH 7.5, while Df-Iac and Df-Bac respectively required 2 and 5 h at pH 9. Each Df modified thio-trastuzumab was chelated with (89)Zr in yields exceeding 75%. (89)Zr-Df-Ac-thio-trastuzumab and (89)Zr-Df-Chx-Mal-thio-trastuzumab were stable in mouse serum and exhibited comparable PET imaging capabilities in a BT474M1 (HER2-positive) breast cancer model reaching 20-25 %ID/g of tumor uptake and a tumor to blood ratio of 6.1-7.1. CONCLUSIONS The new reagents demonstrated good reactivity with engineered thiol groups of trastuzumab and very good chelation properties with (89)Zr. The site-specifically (89)Zr-labeled thio-antibodies were stable in serum and showed PET imaging properties comparable to lysine conjugates.


Cancer Research | 2014

Antitumor Efficacy of a Bispecific Antibody That Targets HER2 and Activates T Cells

Teemu T. Junttila; Ji Li; Jennifer Johnston; Maria Hristopoulos; Robyn Clark; Diego Ellerman; Bu-Er Wang; Yijin Li; Mary Mathieu; Guangmin Li; Judy Young; Elizabeth Luis; Gail Lewis Phillips; Eric Stefanich; Christoph Spiess; Andrew G. Polson; Bryan Irving; Justin Scheer; Melissa R. Junttila; Mark S. Dennis; Robert F. Kelley; Klara Totpal; Allen Ebens

Clinical results from the latest strategies for T-cell activation in cancer have fired interest in combination immunotherapies that can fully engage T-cell immunity. In this study, we describe a trastuzumab-based bispecific antibody, HER2-TDB, which targets HER2 and conditionally activates T cells. HER2-TDB specifically killed HER2-expressing cancer cells at low picomolar concentrations. Because of its unique mechanism of action, which is independent of HER2 signaling or chemotherapeutic sensitivity, HER2-TDB eliminated cells refractory to currently approved HER2 therapies. HER2-TDB exhibited potent antitumor activity in four preclinical model systems, including MMTV-huHER2 and huCD3 transgenic mice. PD-L1 expression in tumors limited HER2-TDB activity, but this resistance could be reversed by anti-PD-L1 treatment. Thus, combining HER2-TDB with anti-PD-L1 yielded a combination immunotherapy that enhanced tumor growth inhibition, increasing the rates and durability of therapeutic response.


EBioMedicine | 2015

Sustained Brown Fat Stimulation and Insulin Sensitization by a Humanized Bispecific Antibody Agonist for Fibroblast Growth Factor Receptor 1/βKlotho Complex

Ganesh Kolumam; Mark Z. Chen; Raymond K. Tong; Jose Zavala-Solorio; Lance Kates; Nicholas van Bruggen; Jed Ross; Shelby K. Wyatt; Vineela D. Gandham; Richard A. D. Carano; Diana Ronai Dunshee; Ai-Luen Wu; Benjamin Haley; Keith R. Anderson; Søren Warming; Xin Y. Rairdan; Nicholas Lewin-Koh; Yingnan Zhang; Johnny Gutierrez; Amos Baruch; Thomas Gelzleichter; Dale Stevens; Sharmila Rajan; Travis W. Bainbridge; Jean-Michel Vernes; Y. Gloria Meng; James Ziai; Robert Soriano; Matthew J. Brauer; Yongmei Chen

Dissipating excess calories as heat through therapeutic stimulation of brown adipose tissues (BAT) has been proposed as a potential treatment for obesity-linked disorders. Here, we describe the generation of a humanized effector-less bispecific antibody that activates fibroblast growth factor receptor (FGFR) 1/βKlotho complex, a common receptor for FGF21 and FGF19. Using this molecule, we show that antibody-mediated activation of FGFR1/βKlotho complex in mice induces sustained energy expenditure in BAT, browning of white adipose tissue, weight loss, and improvements in obesity-associated metabolic derangements including insulin resistance, hyperglycemia, dyslipidemia and hepatosteatosis. In mice and cynomolgus monkeys, FGFR1/βKlotho activation increased serum high-molecular-weight adiponectin, which appears to contribute over time by enhancing the amplitude of the metabolic benefits. At the same time, insulin sensitization by FGFR1/βKlotho activation occurs even before the onset of weight loss in a manner that is independent of adiponectin. Together, selective activation of FGFR1/βKlotho complex with a long acting therapeutic antibody represents an attractive approach for the treatment of type 2 diabetes and other obesity-linked disorders through enhanced energy expenditure, insulin sensitization and induction of high-molecular-weight adiponectin.

Collaboration


Dive into the Elizabeth Luis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge