Network


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

Hotspot


Dive into the research topics where Victoria Smith is active.

Publication


Featured researches published by Victoria Smith.


Nature Medicine | 2010

Allosteric inhibition of lysyl oxidase-like-2 impedes the development of a pathologic microenvironment

Vivian Barry-Hamilton; Rhyannon Spangler; Derek Marshall; Scott McCauley; Hector Rodriguez; Miho Oyasu; Amanda Mikels; Maria Vaysberg; Haben Ghermazien; Carol Wai; Carlos Aurelio Garcia; Arleene C. Velayo; Brett Jorgensen; Donna Hiroko Tokuoka Biermann; Daniel Tsai; Jennifer M. Green; Shelly Zaffryar-Eilot; Alison Kay Holzer; Scott Ogg; Dung Thai; Gera Neufeld; Peter Van Vlasselaer; Victoria Smith

We have identified a new role for the matrix enzyme lysyl oxidase–like-2 (LOXL2) in the creation and maintenance of the pathologic microenvironment of cancer and fibrotic disease. Our analysis of biopsies from human tumors and fibrotic lung and liver tissues revealed an increase in LOXL2 in disease-associated stroma and limited expression in healthy tissues. Targeting LOXL2 with an inhibitory monoclonal antibody (AB0023) was efficacious in both primary and metastatic xenograft models of cancer, as well as in liver and lung fibrosis models. Inhibition of LOXL2 resulted in a marked reduction in activated fibroblasts, desmoplasia and endothelial cells, decreased production of growth factors and cytokines and decreased transforming growth factor-β (TGF-β) pathway signaling. AB0023 outperformed the small-molecule lysyl oxidase inhibitor β-aminoproprionitrile. The efficacy and safety of LOXL2-specific AB0023 represents a new therapeutic approach with broad applicability in oncologic and fibrotic diseases.


Assay and Drug Development Technologies | 2013

Extracellular Matrix Remodeling: The Common Denominator in Connective Tissue DiseasesPossibilities for Evaluation and Current Understanding of the Matrix as More Than a Passive Architecture, but a Key Player in Tissue Failure

Morten A. Karsdal; M.J. Nielsen; Jannie Marie Bülow Sand; Kim Henriksen; Federica Genovese; A.-C. Bay-Jensen; Victoria Smith; Joanne I. Adamkewicz; Claus Christiansen; Diana Julie Leeming

Increased attention is paid to the structural components of tissues. These components are mostly collagens and various proteoglycans. Emerging evidence suggests that altered components and noncoded modifications of the matrix may be both initiators and drivers of disease, exemplified by excessive tissue remodeling leading to tissue stiffness, as well as by changes in the signaling potential of both intact matrix and fragments thereof. Although tissue structure until recently was viewed as a simple architecture anchoring cells and proteins, this complex grid may contain essential information enabling the maintenance of the structure and normal functioning of tissue. The aims of this review are to (1) discuss the structural components of the matrix and the relevance of their mutations to the pathology of diseases such as fibrosis and cancer, (2) introduce the possibility that post-translational modifications (PTMs), such as protease cleavage, citrullination, cross-linking, nitrosylation, glycosylation, and isomerization, generated during pathology, may be unique, disease-specific biochemical markers, (3) list and review the range of simple enzyme-linked immunosorbent assays (ELISAs) that have been developed for assessing the extracellular matrix (ECM) and detecting abnormal ECM remodeling, and (4) discuss whether some PTMs are the cause or consequence of disease. New evidence clearly suggests that the ECM at some point in the pathogenesis becomes a driver of disease. These pathological modified ECM proteins may allow insights into complicated pathologies in which the end stage is excessive tissue remodeling, and provide unique and more pathology-specific biochemical markers.


European Respiratory Journal | 2014

Serum lysyl oxidase-like 2 levels and idiopathic pulmonary fibrosis disease progression

Jason W. Chien; Thomas J. Richards; Kevin F. Gibson; Yingze Zhang; Kathleen O. Lindell; Lixin Shao; Susan K. Lyman; Joanne I. Adamkewicz; Victoria Smith; Naftali Kaminski; Thomas O’Riordan

We evaluated whether lysyl oxidase-like 2 (LOXL2), which promotes cross-linking of collagen in pathological stroma, was detectable in serum from idiopathic pulmonary fibrosis (IPF) patients, and assessed its relationship with IPF disease progression. Patients from the ARTEMIS-IPF (n=69) and the Genomic and Proteomic Analysis of Disease Progression in IPF (GAP) (n=104) studies were analysed. Baseline serum LOXL2 (sLOXL2) levels were compared with baseline clinical and physiological surrogates of disease severity, and the association with IPF disease progression was assessed using a classification and regression tree (CART) method. sLOXL2 correlated weakly with forced vital capacity and carbon monoxide diffusion capacity (r -0.24–0.05) in both cohorts. CART-determined thresholds were similar: ARTEMIS-IPF 800 pg·mL−1 and GAP 700 pg·mL−1. In ARTEMIS-IPF, higher sLOXL2 (>800 pg·mL−1) was associated with increased risk for disease progression (hazard ratio (HR) 5.41, 95% CI 1.65–17.73). Among GAP subjects with baseline spirometric data (n=70), higher sLOXL2 levels (>700 pg·mL−1) were associated with more disease progression events (HR 1.78, 95% CI 1.01–3.11). Among all GAP subjects, higher sLOXL2 levels were associated with increased risk for mortality (HR 2.28, 95% CI 1.18–4.38). These results suggest that higher sLOXL2 levels are associated with increased risk for IPF disease progression. However, due to multiple limitations, these results require validation. Higher baseline sLOXL2 levels: no correlation with IPF severity, but association with disease progression http://ow.ly/sEbGW


The Lancet Respiratory Medicine | 2017

Efficacy of simtuzumab versus placebo in patients with idiopathic pulmonary fibrosis: a randomised, double-blind, controlled, phase 2 trial

Ganesh Raghu; Kevin K. Brown; Harold R. Collard; Vincent Cottin; Kevin F. Gibson; Robert J. Kaner; David J. Lederer; Fernando J. Martinez; Paul W. Noble; Jin Woo Song; Athol U. Wells; Timothy Whelan; Wim Wuyts; Emmanuel Moreau; Scott Patterson; Victoria Smith; Selina Bayly; Jason W. Chien; Qi Gong; Jenny Zhang; Thomas G. O'Riordan

BACKGROUND Lysyl oxidase-like 2 (LOXL2) catalyses collagen cross-linking and is implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF). The aim of this study was to investigate the efficacy and safety of simtuzumab, a monoclonal antibody against LOXL2, in patients with IPF. METHODS In this randomised, double-blind, phase 2 trial, we recruited patients aged 45-85 years with definite IPF diagnosed prior to 3 years of screening from 183 hospitals and respiratory clinics in 14 countries. Eligible patients, stratified by baseline forced vital capacity (FVC), serum LOXL2 (sLOXL2) concentrations, and pirfenidone and nintedanib use, were randomly assigned (1:1) to inject 125 mg/mL simtuzumab or placebo subcutaneously once a week. The primary endpoints were progression-free survival, defined as time to all-cause death or a categorical decrease from baseline in FVC % predicted, in the intention-to-treat population, in patients with sLOXL2 concentrations in the 50th percentile or higher, and in patients with sLOXL2 concentrations in the 75th percentile or higher. Treatment duration was event-driven, and interim analyses were planned and conducted after approximately 120 and 200 progression-free survival events, respectively, occurred. We compared treatment groups with the stratified log-rank test. This study is registered with ClinicalTrials.gov, number NCT01769196. FINDINGS Patients with IPF were recruited between Jan 31, 2013, and June 1, 2015. The intention-to-treat population included 544 randomly assigned patients (272 patients in both groups), and the safety population included 543 randomly assigned patients who received at least one dose of study medication. The study was terminated when the second interim analysis met the prespecified futility stopping criteria in the intention-to-treat population. We noted no difference in progression-free survival between simtuzumab and placebo in the intention-to-treat population (median progression free survival times of 12·6 months and 15·4 months for simtuzumab and placebo, respectively; stratified HR 1·13, 95% CI 0·88-1·45; p=0·329) and in patients with baseline sLOXL2 in the 50th percentile or higher (median progression-free survival 11·7 months and 14·3 months for simtuzumab and placebo, respectively; stratified HR 1·03, 95% CI 0·74-1·43; p=0·851), or in the 75th percentile or higher (median progression-free survival 11·6 months and 16·9 months for simtuzumab and placebo, respectively; stratified HR 1·20, 95% CI 0·72-2·00; p=0·475). The incidence of adverse events and serious adverse events was similar between treatment groups. The most common adverse events in both the simtuzumab and placebo groups were dyspnoea, cough, upper respiratory tract infection, and worsening of IPF; and the most common grade 3 or 4 adverse events were worsening of IPF, dyspnoea, and pneumonia. INTERPRETATION Simtuzumab did not improve progression-free survival in a well-defined population of patients with IPF. Our data do not support the use of simtuzumab for patients with IPF. FUNDING Gilead Sciences Inc.


Gut | 2017

Selective targeting of lysyl oxidase-like 2 (LOXL2) suppresses hepatic fibrosis progression and accelerates its reversal

Naoki Ikenaga; Zhen-Wei Peng; Kahini A. Vaid; Susan B. Liu; Shuhei Yoshida; Deanna Y. Sverdlov; Amanda Mikels-Vigdal; Victoria Smith; Detlef Schuppan; Yury Popov

Background/Aims We studied the role of lysyl oxidase-like 2 (LOXL2) in collagen crosslinking and hepatic progenitor cell (HPC) differentiation, and the therapeutic efficacy of a LOXL2-blocking monoclonal antibody on liver fibrosis progression/reversal in mice. Methods Anti-LOXL2 antibody, control antilysyl oxidase antibody or placebo was administered during thioacetamide (TAA)-induced fibrosis progression or during recovery. Therapeutic efficacy in biliary fibrosis was tested in BALB/c.Mdr2−/− and 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-fed mice. Collagen crosslinking, fibrosis progression and reversal were assessed histologically and biochemically. HPC differentiation was studied in primary EpCAM(+) liver cells in vitro. Results LOXL2 was virtually absent from healthy but strongly induced in fibrotic liver, with predominant localisation within fibrotic septa. Delayed anti-LOXL2 treatment of active TAA fibrosis significantly reduced collagen crosslinking and histological signs of bridging fibrosis, with a 53% reduction in morphometric collagen deposition. In established TAA fibrosis, LOXL2 inhibition promoted fibrosis reversal, with enhanced splitting and thinning of fibrotic septa, and a 45% decrease in collagen area at 4 weeks of recovery. In the Mdr2−/− and DDC-induced models of biliary fibrosis, anti-LOXL2 antibody similarly achieved significant antifibrotic efficacy and suppressed the ductular reaction, while hepatocyte replication increased. Blocking LOXL2 had a profound direct effect on primary EpCAM(+) HPC behaviour in vitro, promoting their differentiation towards hepatocytes, while inhibiting ductal cell lineage commitment. Conclusions LOXL2 mediates collagen crosslinking and fibrotic matrix stabilisation during liver fibrosis, and independently promotes fibrogenic HPC differentiation. By blocking these two convergent profibrotic pathways, therapeutic LOXL2 inhibition attenuates both parenchymal and biliary fibrosis and promotes fibrosis reversal.


Nature Communications | 2016

Targeting LOXL2 for cardiac interstitial fibrosis and heart failure treatment

Jin Yang; Konstantinos Savvatis; Jong Seok Kang; Peidong Fan; Hongyan Zhong; Karen Schwartz; Vivian E. Barry; Amanda Mikels-Vigdal; Serge Karpinski; Dmytro Kornyeyev; Joanne I. Adamkewicz; Xuhui Feng; Qiong Zhou; Ching Shang; Praveen Kumar; Dillon Phan; Mario Kasner; Begoña López; Javier Díez; Keith C. Wright; Roxanne L. Kovacs; Peng Sheng Chen; Thomas Quertermous; Victoria Smith; Lina Yao; Carsten Tschöpe; Ching Pin Chang

Interstitial fibrosis plays a key role in the development and progression of heart failure. Here, we show that an enzyme that crosslinks collagen—Lysyl oxidase-like 2 (Loxl2)—is essential for interstitial fibrosis and mechanical dysfunction of pathologically stressed hearts. In mice, cardiac stress activates fibroblasts to express and secrete Loxl2 into the interstitium, triggering fibrosis, systolic and diastolic dysfunction of stressed hearts. Antibody-mediated inhibition or genetic disruption of Loxl2 greatly reduces stress-induced cardiac fibrosis and chamber dilatation, improving systolic and diastolic functions. Loxl2 stimulates cardiac fibroblasts through PI3K/AKT to produce TGF-β2, promoting fibroblast-to-myofibroblast transformation; Loxl2 also acts downstream of TGF-β2 to stimulate myofibroblast migration. In diseased human hearts, LOXL2 is upregulated in cardiac interstitium; its levels correlate with collagen crosslinking and cardiac dysfunction. LOXL2 is also elevated in the serum of heart failure (HF) patients, correlating with other HF biomarkers, suggesting a conserved LOXL2-mediated mechanism of human HF.


The FASEB Journal | 2016

Lysyl oxidase activity contributes to collagen stabilization during liver fibrosis progression and limits spontaneous fibrosis reversal in mice

Susan B. Liu; Naoki Ikenaga; Zhen-Wei Peng; Deanna Y. Sverdlov; Andrew Greenstein; Victoria Smith; Detlef Schuppan; Yury Popov

Collagen stabilization through irreversible cross‐linking is thought to promote hepatic fibrosis progression and limit its reversibility. However, the mechanism of this process remains poorly defined. We studied the functional contribution of lysyl oxidase (LOX) to collagen stabilization and hepatic fibrosis progression/reversal in vivo using chronic administration of irreversible LOX inhibitor β‐aminopropionitrile (BAPN, or vehicle as control) in C57Bl/6J mice with carbon tetrachloride (CCl4)‐induced fibrosis. Fibrotic matrix stability was directly assessed using a stepwise collagen extraction assay and fibrotic septae morphometry. Liver cells and fibrosis were studied by histologic, biochemical methods and quantitative real‐time reverse‐transcription PCR. During fibrosis progression, BAPN administration suppressed accumulation of cross‐linked collagens, and fibrotic septae showed widening and collagen fibrils splitting, reminiscent of remodeling signs observed during fibrosis reversal. LOX inhibition attenuated hepatic stellate cell activation markers and promoted F4/80‐positive scar‐associated macrophage infiltration without an increase in liver injury. In reversal experiments, BAPN‐treated fibrotic mice demonstrated accelerated fibrosis reversal after CCl4 withdrawal. Our findings demonstrate for the first time that LOX contributes significantly to collagen stabilization in liver fibrosis, promotes fibrogenic activation of attenuated hepatic stellate cells, and limits fibrosis reversal. Our data support the concept of pharmacologic targeting of LOX pathway to inhibit liver fibrosis and promote its resolution.—Liu, S. B., Ikenaga, N., Peng, Z.‐W., Sverdlov, D. Y., Greenstein, A., Smith, V., Schuppan, D., Popov, Y. Lysyl oxidase activity contributes to collagen stabilization during liver fibrosis progression and limits spontaneous fibrosis reversal in mice. FASEB J. 30, 1599–1609 (2016). www.fasebj.org


The FASEB Journal | 2011

Receptor activity modifying protein-3 mediates the protumorigenic activity of lysyl oxidase-like protein-2

Vera Brekhman; Jennie Lugassie; Shelly Zaffryar-Eilot; Edmond Sabo; Ofra Kessler; Victoria Smith; Hana Golding; Gera Neufeld

Lysyl oxidase‐like protein‐2 (LOXL2) induces epithelial to mesenchymal transition and promotes invasiveness. To understand the mechanisms involved, we examined the effect of LOXL2 overexpression in MCF‐7 cells on gene expression. We found that LOXL2 up‐regulated the expression of receptor activity modifying protein‐3 (RAMP3). Expression of RAMP3 in MDA‐MB‐231 cells in which LOXL2 expression was inhibited restored vimentin expression, invasiveness, and tumor development. Inhibition of RAMP3 expression in MDA‐MB‐231 cells mimicked the effects produced by inhibition of LOXL2expression and was accompanied by inhibition of p38 phosphorylation. LOXL2 overexpression in these cells did not restore invasiveness, suggesting that RAMP3 functions downstream to LOXL2. LOXL2 and RAMP3 are strongly coexpressed in human colon, breast, and gastric carcinomas but not in normal colon or gastric epithelial cells. RAMP3 associates with several G‐protein‐coupled receptors forming receptors for peptides, such as adrenomedullin and amylin. We hypothesized that RAMP3 could function as a transducer of autocrine signals induced by such peptides. However, the proinvasive effects of RAMP3 could not be abrogated following inhibition of the expression or activity of these peptides. Our experiments suggest that the protumorigenic effects of LOXL2 are partially mediated by RAMP3 and that RAMP3 inhibitors may function as antitumorigenic agents.—Brekhman, V., Lugassie, J., Zaffryar‐Eilot, S., Sabo, E., Kessler, O., Smith, V.,Golding, H., Neufeld, G. Receptor activity modifying protein‐3 mediates the protu‐morigenic activity of lysyl oxidase‐like protein‐2. FASEBJ. 25, 55–65 (2011). www.fasebj.org


BMC Cancer | 2013

Extracellular matrix specific protein fingerprints measured in serum can separate pancreatic cancer patients from healthy controls

Nicholas Willumsen; Cecilie L. Bager; Diana Julie Leeming; Victoria Smith; Morten A. Karsdal; David Dornan; A.-C. Bay-Jensen

BackgroundPancreatic cancer (PC) is an aggressive disease with an urgent need for biomarkers. Hallmarks of PC include increased collagen deposition (desmoplasia) and increased matrix metalloproteinase (MMP) activity. The aim of this study was to investigate whether protein fingerprints of specific MMP-generated collagen fragments differentiate PC patients from healthy controls when measured in serum.MethodsThe levels of biomarkers reflecting MMP-mediated degradation of type I (C1M), type III (C3M) and type IV (C4M, C4M12a1) collagen were assessed in serum samples from PC patients (n = 15) and healthy controls (n = 33) using well-characterized and validated competitive ELISAs.ResultsThe MMP-generated collagen fragments were significantly elevated in serum from PC patients as compared to controls. The diagnostic power of C1M, C3M, C4M and C4M12 were ≥83% (p < 0.001) and when combining all biomarkers 99% (p < 0.0001).ConclusionsA panel of serum biomarkers reflecting altered MMP-mediated collagen turnover is able to differentiate PC patients from healthy controls. These markers may increase the understanding of mode of action of the disease and, if validated in larger clinical studies, provide an improved and additional tool in the PC setting.


Chest | 2015

Development of Novel Agents for Idiopathic Pulmonary Fibrosis: Progress in Target Selection and Clinical Trial Design

Thomas G. O'Riordan; Victoria Smith; Ganesh Raghu

Idiopathic pulmonary fibrosis (IPF) is a progressive, fatal disease. Until recently, the standard therapy for this disease has been essentially supportive, with the exception of a minority of patients who were eligible for lung transplantation. The development pathway for novel medications for IPF has been complicated. There have been several challenges, including an incomplete understanding of the pathogenesis, unpredictable clinical course, lack of validated biomarkers, the low clinical predictive value of animal models of lung injury, and the need to commit to large clinical trials of long duration to obtain initial evidence of clinical efficacy. Despite these challenges, the combination of recent advances in translational medicine and the unprecedented increase in clinical data accumulated from recent large clinical trials has stimulated an increase in the number of clinical development programs for IPF. Clinical programs are increasingly characterized by rational target selection, preclinical optimization of therapeutic molecules, and an emphasis on efficient clinical trial design. A lower rate of functional decline in patients treated with pirfenidone and nintedanib was demonstrated in large clinical trials. In October 2014, these two drugs became the first agents to be approved by the US Food and Drug Administration for the treatment of IPF. (Pirfenidone had already been approved in several countries outside the United States.) In November 2014, the European Medicines Agency approved the use of nintedanib for IPF. The landscape for management of IPF has markedly changed with the advent of approved therapeutic options for IPF. In this article, we review the strategies that are being used to increase the likelihood of success in clinical development programs of novel disease-modifying agents in IPF.

Collaboration


Dive into the Victoria Smith's collaboration.

Top Co-Authors

Avatar

Jason W. Chien

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Lixin Shao

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Derek Marshall

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yury Popov

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xiaoming Li

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Carolin Lackner

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge