Network


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

Hotspot


Dive into the research topics where Nerissa Viola-Villegas is active.

Publication


Featured researches published by Nerissa Viola-Villegas.


Science Translational Medicine | 2015

PI3K inhibition results in enhanced estrogen receptor function and dependence in hormone receptor–positive breast cancer

Ana Bosch; Zhiqiang Li; Anna Bergamaschi; Haley Ellis; Eneda Toska; Aleix Prat; Jessica J. Tao; Daniel E. Spratt; Nerissa Viola-Villegas; Pau Castel; Gerard Minuesa; Natasha Morse; Jordi Rodon; Yasir H. Ibrahim; Javier Cortes; Jose Perez-Garcia; Patricia Galván; Judit Grueso; Marta Guzman; John A. Katzenellenbogen; Michaelz Kharas; Jason S. Lewis; Maura N. Dickler; Violeta Serra; Neal Rosen; Sarat Chandarlapaty; Maurizio Scaltriti; J. Baselga

Inhibition of the PI3K/AKT pathway results in induction of ER-dependent transcriptional activity and susceptibility to anti-estrogen therapy in ER-positive breast cancer. PIKing the correct therapeutic combination Mutations in a gene called PIK3CA are very common in estrogen receptor–positive breast cancers, and drugs that inhibit PI3K, the protein product of this gene, are already in clinical development. Unfortunately, these drugs are not always effective, and this study by Bosch et al. demonstrates a reason for this problem and a practical way to overcome it. By studying both mouse models and human patients’ tumors, the authors discovered that inhibition of PI3K often stimulates the activity of the estrogen receptor, which then drives tumor growth. By combining PI3K inhibitors with clinically available drugs that inhibit the estrogen receptor, the authors were able to overcome treatment resistance and effectively induce tumor regression in mouse models. Activating mutations of PIK3CA are the most frequent genomic alterations in estrogen receptor (ER)–positive breast tumors, and selective phosphatidylinositol 3-kinase α (PI3Kα) inhibitors are in clinical development. The activity of these agents, however, is not homogeneous, and only a fraction of patients bearing PIK3CA-mutant ER-positive tumors benefit from single-agent administration. Searching for mechanisms of resistance, we observed that suppression of PI3K signaling results in induction of ER-dependent transcriptional activity, as demonstrated by changes in expression of genes containing ER-binding sites and increased occupancy by the ER of promoter regions of up-regulated genes. Furthermore, expression of ESR1 mRNA and ER protein were also increased upon PI3K inhibition. These changes in gene expression were confirmed in vivo in xenografts and patient-derived models and in tumors from patients undergoing treatment with the PI3Kα inhibitor BYL719. The observed effects on transcription were enhanced by the addition of estradiol and suppressed by the anti-ER therapies fulvestrant and tamoxifen. Fulvestrant markedly sensitized ER-positive tumors to PI3Kα inhibition, resulting in major tumor regressions in vivo. We propose that increased ER transcriptional activity may be a reactive mechanism that limits the activity of PI3K inhibitors and that combined PI3K and ER inhibition is a rational approach to target these tumors.


Science Signaling | 2014

Antagonism of EGFR and HER3 Enhances the Response to Inhibitors of the PI3K-Akt Pathway in Triple-Negative Breast Cancer

Jessica J. Tao; Pau Castel; Nina Radosevic-Robin; Moshe Elkabets; Neil Auricchio; Nicola Aceto; Gregory Weitsman; Paul R. Barber; Borivoj Vojnovic; Haley Ellis; Natasha Morse; Nerissa Viola-Villegas; Ana Bosch; Dejan Juric; Saswati Hazra; Sharat Singh; Phillip Kim; Anna Bergamaschi; Shyamala Maheswaran; Tony Ng; Frédérique Penault-Llorca; Jason S. Lewis; Lisa A. Carey; Charles M. Perou; José Baselga; Maurizio Scaltriti

Predictions regarding drug resistance mechanisms and treatment strategies in triple-negative breast cancer are confirmed in tumors from patients. From Models to Breast Cancer Treatments Patients with triple-negative breast cancer (TNBC), a particularly aggressive form, have few treatment options. Targeting either the phosphatidylinositol 3-kinase to Akt (PI3K-Akt) pathway or epidermal growth factor receptor (EGFR) inhibits tumor growth in some patients, but durable responses are rare. Modeling studies using cell lines predict that the EGFR family member HER3 (human epidermal growth factor receptor 3) may confer drug resistance. Now, Tao et al. provide evidence from patient tumors to support those predictions. Treatment with PI3K-Akt pathway inhibitors increased the abundance of both total and activated HER3 in TNBC cells in culture and TNBC xenografts in mice. Residual tumors from patients treated with EGFR inhibitors had increased abundance and activation of HER3. Combining inhibitors of the PI3K-Akt pathway with a dual inhibitor of EGFR and HER3 substantially suppressed tumor growth in mice with TNBC xenografts derived from either cell lines or patients, suggesting that this combined strategy may improve therapeutic outcome in TNBC patients. Both abundant epidermal growth factor receptor (EGFR or ErbB1) and high activity of the phosphatidylinositol 3-kinase (PI3K)–Akt pathway are common and therapeutically targeted in triple-negative breast cancer (TNBC). However, activation of another EGFR family member [human epidermal growth factor receptor 3 (HER3) (or ErbB3)] may limit the antitumor effects of these drugs. We found that TNBC cell lines cultured with the EGFR or HER3 ligand EGF or heregulin, respectively, and treated with either an Akt inhibitor (GDC-0068) or a PI3K inhibitor (GDC-0941) had increased abundance and phosphorylation of HER3. The phosphorylation of HER3 and EGFR in response to these treatments was reduced by the addition of a dual EGFR and HER3 inhibitor (MEHD7945A). MEHD7945A also decreased the phosphorylation (and activation) of EGFR and HER3 and the phosphorylation of downstream targets that occurred in response to the combination of EGFR ligands and PI3K-Akt pathway inhibitors. In culture, inhibition of the PI3K-Akt pathway combined with either MEHD7945A or knockdown of HER3 decreased cell proliferation compared with inhibition of the PI3K-Akt pathway alone. Combining either GDC-0068 or GDC-0941 with MEHD7945A inhibited the growth of xenografts derived from TNBC cell lines or from TNBC patient tumors, and this combination treatment was also more effective than combining either GDC-0068 or GDC-0941 with cetuximab, an EGFR-targeted antibody. After therapy with EGFR-targeted antibodies, some patients had residual tumors with increased HER3 abundance and EGFR/HER3 dimerization (an activating interaction). Thus, we propose that concomitant blockade of EGFR, HER3, and the PI3K-Akt pathway in TNBC should be investigated in the clinical setting.


The Journal of Nuclear Medicine | 2013

Monitoring Afatinib Treatment in HER2-Positive Gastric Cancer with 18F-FDG and 89Zr-Trastuzumab PET

Yelena Y. Janjigian; Nerissa Viola-Villegas; Jason P. Holland; Vadim Divilov; Sean Carlin; Erica M. Gomes-DaGama; Gabriela Chiosis; Gregory Carbonetti; Elisa de Stanchina; Jason S. Lewis

We evaluated the ability of the PET imaging agent 89Zr-trastuzumab to delineate HER2-positive gastric cancer and to monitor the pharmacodynamic effects of the epidermal growth factor receptor (EGFR)/human epidermal growth factor receptor 2 (HER2) tyrosine kinase inhibitor afatinib. Methods: Using 89Zr-trastuzumab, 18F-FDG, or 3′-deoxy-3′-18F-fluorothymidine (18F-FLT PET), we imaged HER2-positive NCI-N87 and HER2-negative MKN74 gastric cancer xenografts in mice. Next, we examined the pharmacodynamic effects of afatinib in NCI-N87 xenografts using 89Zr-trastuzumab and 18F-FDG PET and comparing imaging results to changes in tumor size and in protein expression as monitored by Western blot and histologic studies. Results: Although 18F-FDG uptake in NCI-N87 tumors did not change, a decrease in 89Zr-trastuzumab uptake was observed in the afatinib-treated versus control groups (3.0 ± 0.0 percentage injected dose per gram (%ID/g) vs. 21.0 ± 3.4 %ID/g, respectively; P < 0.05). 89Zr-trastuzumab PET results corresponded with tumor reduction, apoptosis, and downregulation of HER2 observed on treatment with afatinib. Downregulation of total HER2, phosphorylated (p)-HER2, and p-EGFR occurred within 24 h of the first dose of afatinib, with a sustained effect over 21 d of treatment. Conclusion: Afatinib demonstrated antitumor activity in HER2-positive gastric cancer in vivo. 89Zr-trastuzumab PET specifically delineated HER2-positive gastric cancer and can be used to measure the pharmacodynamic effects of afatinib.


Bioconjugate Chemistry | 2013

18F-Labeled-Bioorthogonal Liposomes for In Vivo Targeting

Fabien Emmetiere; Christopher Irwin; Nerissa Viola-Villegas; Valerie A. Longo; Sarah M. Cheal; Pat Zanzonico; Nagavarakishore Pillarsetty; Wolfgang A. Weber; Jason S. Lewis; Thomas Reiner

Liposomes are attractive vehicles for the controlled release of drugs and cytotoxins and have a long-standing history in medical research and clinical practice. In addition to established therapeutic indications, liposomes have several favorable properties for molecular imaging, including high stability and the ability to be labeled with radioisotopes, as well as paramagnetic and fluorescent contrast agents. However, long circulation times and difficulties in creating targeted liposomes have proven challenges for imaging. In this study, we have addressed these limitations using a recently developed strategy for bioorthogonal conjugation, the reaction between tetrazines and trans-cyclooctenes. By coating radiolabeled liposomes with trans-cyclooctene and pretargeting with a tetrazine coupled to a targeted peptide, we were able to selectively enhance the retention of liposomes and bind them to tumor tissue in live animals. The rapid reaction between tetrazines and trans-cyclooctenes allowed imaging to be performed with the short-lived PET tracer (18)F, yielding signal-to-background activity ratios of 7:1. The covalent, bioorthogonally driven tumor-targeting of liposomes by in vivo click chemistry is promising and should be explored for more selective and rapid delivery of radiodiagnostics and radiotherapeutics, two classes of drugs which particularly benefit from fast clearance, low nonspecific binding, and the associated reduced toxicity to kidneys and bone marrow.


Inorganic Chemistry | 2014

Underscoring the influence of inorganic chemistry on nuclear imaging with radiometals

Brian M. Zeglis; Jacob L. Houghton; Michael J. Evans; Nerissa Viola-Villegas; Jason S. Lewis

Over the past several decades, radionuclides have matured from largely esoteric and experimental technologies to indispensible components of medical diagnostics. Driving this transition, in part, have been mutually necessary advances in biomedical engineering, nuclear medicine, and cancer biology. Somewhat unsung has been the seminal role of inorganic chemistry in fostering the development of new radiotracers. In this regard, the purpose of this Forum Article is to more visibly highlight the significant contributions of inorganic chemistry to nuclear imaging by detailing the development of five metal-based imaging agents: (64)Cu-ATSM, (68)Ga-DOTATOC, (89)Zr-transferrin, (99m)Tc-sestamibi, and (99m)Tc-colloids. In a concluding section, several unmet needs both in and out of the laboratory will be discussed to stimulate conversation between inorganic chemists and the imaging community.


Molecular Pharmaceutics | 2014

Noninvasive Imaging of PSMA in prostate tumors with (89)Zr-Labeled huJ591 engineered antibody fragments: the faster alternatives.

Nerissa Viola-Villegas; Kuntal K. Sevak; Sean Carlin; Michael G. Doran; Henry W. Evans; Derek Bartlett; Anna M. Wu; Jason S. Lewis

Engineered antibody fragments offer faster delivery with retained tumor specificity and rapid clearance from nontumor tissues. Here, we demonstrate that positron emission tomography (PET) based detection of prostate specific membrane antigen (PSMA) in prostatic tumor models using engineered bivalent antibodies built on single chain fragments (scFv) derived from the intact antibody, huJ591, offers similar tumor delineating properties but with the advantage of rapid targeting and imaging. 89Zr-radiolabeled huJ591 scFv (dimeric scFv-CH3; 89Zr-Mb) and cysteine diabodies (dimeric scFv; 89Zr-Cys-Db) demonstrated internalization and similar Kds (∼2 nM) compared to 89Zr-huJ591 in PSMA(+) cells. Tissue distribution assays established the specificities of both 89Zr-Mb and 89Zr-Cys-Db for PSMA(+) xenografts (6.2 ± 2.5% ID/g and 10.2 ± 3.4% ID/g at 12 h p.i. respectively), while minimal accumulation in PSMA(−) tumors was observed. From the PET images, 89Zr-Mb and 89Zr-Cys-Db exhibited faster blood clearance than the parent huJ591 while tumor-to-muscle ratios for all probes show comparable values across all time points. Ex vivo autoradiography and histology assessed the distribution of the probes within the tumor. Imaging PSMA-expressing prostate tumors with smaller antibody fragments offers rapid tumor accumulation and accelerated clearance; hence, shortened wait periods between tracer administration and high-contrast tumor imaging and lower dose-related toxicity are potentially realized.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Understanding the pharmacological properties of a metabolic PET tracer in prostate cancer

Nerissa Viola-Villegas; Sean Carlin; Ellen Ackerstaff; Kuntal K. Sevak; Vadim Divilov; Inna Serganova; Natalia Kruchevsky; Michael Anderson; Ronald G. Blasberg; Oleg A. Andreev; Donald M. Engelman; Jason A. Koutcher; Yana K. Reshetnyak; Jason S. Lewis

Significance Solid tumors adapt a glycolytic phenotype for their energetic requirements, leading to acidification of the extracellular environment. Targeting this global event is important to gauge the pace of tumor growth and invasiveness, as well as to provide a basis for predicting disease response to pH-dependent chemotherapies. To realize this goal, a noninvasive method is necessary to measure tumor extracellular acidification to meet clinical needs. This study explores the utility of pH (low) insertion peptide, an acidosis-targeting peptide, as a PET-based imaging probe to provide a method for quantifying extracellular pH and its correlation to known acidity markers, such as hypoxia, carbonic anhydrase IX, and lactate dehydrogenase A, within the prostate tumor tissue. Generally, solid tumors (>400 mm3) are inherently acidic, with more aggressive growth producing greater acidity. If the acidity could be targeted as a biomarker, it would provide a means to gauge the pace of tumor growth and degree of invasiveness, as well as providing a basis for predicting responses to pH-dependent chemotherapies. We have developed a 64Cu pH (low) insertion peptide (pHLIP) for targeting, imaging, and quantifying acidic tumors by PET, and our findings reveal utility in assessing prostate tumors. The new pHLIP version limits indiscriminate healthy tissue binding, and we demonstrate its targeting of extracellular acidification in three different prostate cancer models, each with different vascularization and acid-extruding protein carbonic anhydrase IX (CAIX) expression. We then describe the tumor distribution of this radiotracer ex vivo, in association with blood perfusion and known biomarkers of acidity, such as hypoxia, lactate dehydrogenase A, and CAIX. We find that the probe reveals metabolic variations between and within tumors, and discriminates between necrotic and living tumor areas.


The Journal of Nuclear Medicine | 2013

Applying PET to Broaden the Diagnostic Utility of the Clinically Validated CA19.9 Serum Biomarker for Oncology

Nerissa Viola-Villegas; Samuel L. Rice; Sean Carlin; Xiaohong Wu; Michael J. Evans; Kuntal K. Sevak; Marija Drobjnak; Govind Ragupathi; Ritsuko Sawada; Wolfgang W. Scholz; Philip O. Livingston; Jason S. Lewis

Despite their considerable advantages, many circulating biomarkers have well-documented limitations. One prominent shortcoming in oncology is a high frequency of false-positive indications for malignant disease in upfront diagnosis. Because one common cause of false positivism is biomarker production from benign disorders in unrelated host tissues, we hypothesized that probing the sites of biomarker secretion with an imaging tool could be a broadly useful strategy to deconvolute the meaning of foreboding but inconclusive circulating biomarker levels. Methods: In preparation to address this hypothesis clinically, we developed 89Zr-5B1, a fully human, antibody-based radiotracer targeting tumor-associated CA19.9 in the preclinical setting. Results: 89Zr-5B1 localized to multiple tumor models representing diseases with undetectable and supraphysiologic serum CA19.9 levels. Among these, 89Zr-5B1 detected orthotopic models of pancreatic ductal adenocarcinoma, an elusive cancer for which the serum assay is measured in humans but with limited specificity in part because of the frequency of CA19.9 secretion from benign hepatic pathologies. Conclusion: In this report, a general strategy to supplement some of the shortcomings of otherwise highly useful circulating biomarkers with immunoPET is described. To expedite the clinical validation of this model, a human monoclonal antibody to CA19.9 (a highly visible but partially flawed serum biomarker for several cancers) was radiolabeled and evaluated, and the compelling preclinical evidence suggests that the radiotracer may enhance the fidelity of diagnosis and staging of pancreatic ductal adenocarcinoma, a notoriously occult cancer.


Bioconjugate Chemistry | 2016

PET Imaging of Extracellular pH in Tumors with 64Cu- and 18F-Labeled pHLIP Peptides: A Structure–Activity Optimization Study

Dustin W. Demoin; Linden C. Wyatt; Kimberly J. Edwards; Dalya Abdel-Atti; Mirkka Sarparanta; Jacob Pourat; Valerie A. Longo; Sean Carlin; Donald M. Engelman; Oleg A. Andreev; Yana K. Reshetnyak; Nerissa Viola-Villegas; Jason S. Lewis

pH (low) insertion peptides (pHLIP peptides) target acidic extracellular environments in vivo due to pH-dependent cellular membrane insertion. Two variants (Var3 and Var7) and wild-type (WT) pHLIP peptides have shown promise for in vivo imaging of breast cancer. Two positron emitting radionuclides (64Cu and 18F) were used to label the NOTA- and NO2A-derivatized Var3, Var7, and WT peptides for in vivo biodistribution studies in 4T1 orthotopic tumor-bearing BALB/c mice. All of the constructs were radiolabeled with 64Cu or [18F]-AlF in good yield. The in vivo biodistribution of the 12 constructs in 4T1 orthotopic allografted female BALB/c mice indicated that NO2A-cysVar3, radiolabeled with either 18F (4T1 uptake; 8.9 ± 1.7%ID/g at 4 h p.i.) or 64Cu (4T1 uptake; 8.2 ± 0.9%ID/g at 4 h p.i. and 19.2 ± 1.8% ID/g at 24 h p.i.), shows the most promise for clinical translation. Additional studies to investigate other tumor models (melanoma, prostate, and brain tumor models) indicated the universality of tumor targeting of these tracers. From this study, future clinical translation will focus on 18F- or 64Cu-labeled NO2A-cysVar3.


ACS Omega | 2017

89Zr-Cobalamin PET Tracer: Synthesis, Cellular Uptake, and Use for Tumor Imaging

Akhila N. W. Kuda-Wedagedara; Jayme L. Workinger; Ebba Nexo; Robert P. Doyle; Nerissa Viola-Villegas

Vitamin B12, or cobalamin (Cbl), is an essential nutrient. Acquisition, transport, and cellular internalization of Cbl are dependent on specific binding proteins and associated receptors. The circulating transport protein transcobalamin (TC) promotes cellular uptake via binding to specific receptors such as CD320, a receptor upregulated in several cancer cell lines. In this study, we report the successful synthesis of 89Zirconium-labeled Cbl that was derivatized with desferrioxamine (89Zr-Cbl). We document the purity of the tracer and its binding to TC compared with that of unmodified cyano-Cbl (CN-Cbl). In vitro studies employing the CD320 receptor-positive breast cancer cell line MDA-MB-453 showed a 6- to 10-fold greater uptake of 89Zr-Cbl when compared with the uptake in the presence of 200-fold excess of CN-Cbl at 37 °C. We used nude mice with MDA-MB-453 tumors to study the feasibility of employing the tracer to visualize CD320 positive tumors. In vivo positron emission tomography images displayed a clear visualization of the tumor with 1.42 ± 0.48 %ID/g uptake (n = 3) at 4 h after injection (p.i.) with the tracer retained at 48 h p.i. Ex vivo biodistribution studies using 89Zr-Cbl exhibited the highest uptake in kidney and liver at 48 h p.i. Results document the feasibility of synthesizing a Cbl-based tracer suitable for both in vivo and ex vivo studies of Cbl trafficking and with the potential to visualize tumors expressing TC receptors, such as CD320.

Collaboration


Dive into the Nerissa Viola-Villegas's collaboration.

Top Co-Authors

Avatar

Jason S. Lewis

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Sean Carlin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kuntal K. Sevak

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Oleg A. Andreev

University of Rhode Island

View shared research outputs
Top Co-Authors

Avatar

Vadim Divilov

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ana Bosch

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anna M. Wu

University of California

View shared research outputs
Top Co-Authors

Avatar

Dalya Abdel-Atti

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge