Network


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

Hotspot


Dive into the research topics where Melissa Smith is active.

Publication


Featured researches published by Melissa Smith.


Molecular Oncology | 2011

High tumor levels of IL6 and IL8 abrogate preclinical efficacy of the γ-secretase inhibitor, RO4929097

Wei He; Leopoldo Luistro; Daisy Carvajal; Melissa Smith; Tom Nevins; Xuefeng Yin; James Cai; Brian Higgins; Kenneth Kolinsky; Kathryn Packman; David Heimbrook; John Frederick Boylan

Interest continues to build around the early application of patient selection markers to prospectively identify patients likely to show clinical benefit from cancer therapies. Hypothesis generation and clinical strategies often begin at the preclinical stage where responder and nonresponder tumor cell lines are first identified and characterized. In the present study, we investigate the drivers of in vivo resistance to the γ‐secretase inhibitor RO4929097. Beginning at the tissue culture level, we identified apparent IL6 and IL8 expression differences that characterized tumor cell line response to RO4929097. We validated this molecular signature at the preclinical efficacy level identifying additional xenograft models resistant to the in vivo effects of RO4929097. Our data suggest that for IL6 and IL8 overexpressing tumors, RO4929097 no longer impacts angiogenesis or the infiltration of tumor associated fibroblasts. These preclinical data provide a rationale for preselecting patients possessing low levels of IL6 and IL8 prior to RO4929097 dosing. Extending this hypothesis into the clinic, we monitored patient IL6 and IL8 serum levels prior to dosing with RO4929097 during Phase I. Interestingly, the small group of patients deriving some type of clinical benefit from RO4929097 presented with low baseline levels of IL6 and IL8. Our data support the continued investigation of this patient selection marker for RO4929097 and other types of Notch inhibitors undergoing early clinical evaluation.


Molecular Therapy | 2014

Discovery of siRNA Lipid Nanoparticles to Transfect Suspension Leukemia Cells and Provide In Vivo Delivery Capability

Wei He; Michael Bennett; Leopoldo Luistro; Daisy Carvajal; Thomas D. Nevins; Melissa Smith; Gaurav Tyagi; James Cai; Xin Wei; Tai-An Lin; David Heimbrook; Kathryn Packman; John Frederick Boylan

As a powerful research tool, siRNAs therapeutic and target validation utility with leukemia cells and long-term gene knockdown is severely restricted by the lack of omnipotent, safe, stable, and convenient delivery. Here, we detail our discovery of siRNA-containing lipid nanoparticles (LNPs) able to effectively transfect several leukemia and difficult-to-transfect adherent cell lines also providing in vivo delivery to mouse spleen and bone marrow tissues through tail-vein administration. We disclose a series of novel structurally related lipids accounting for the superior transfection ability, and reveal a correlation between expression of Caveolins and successful transfection. These LNPs, bearing low toxicity and long stability of >6 months, are ideal for continuous long-term dosing. Our discovery represents the first effective siRNA-containing LNPs for leukemia cells, which not only enables high-throughput siRNA screening with leukemia cells and difficult-to-transfect adherent cells but also paves the way for the development of therapeutic siRNA for leukemia treatment.


Clinical Cancer Research | 2013

RG7212 anti-TWEAK mAb inhibits tumor growth through inhibition of tumor cell proliferation and survival signaling and by enhancing the host antitumor immune response.

Xuefeng Yin; Leopoldo Luistro; Hua Zhong; Melissa Smith; Tom Nevins; Kathleen Schostack; Holly Hilton; Tai-An Lin; Theresa Truitt; Denise Biondi; Xiaoqian Wang; Kathryn Packman; Jim Rosinski; Windy Berkofsky-Fessler; Jian-Ping Tang; Saumya Pant; David Geho; Suzana Vega-Harring; Mark DeMario; Hy Levitsky; Mary Ellen Simcox

Purpose: To explore the role of TWEAK in tumor growth and antitumor immune response and the activity and mechanism of RG7212, an antagonistic anti-TWEAK antibody, in tumor models. Experimental Design: TWEAK-induced signaling and gene expression were explored in tumor cell lines and inhibition of these effects and antitumor efficacy with RG7212 treatment was assessed in human tumor xenograft-, patient-derived xenograft, and syngeneic tumor models and phase I patients. Genetic features correlated with antitumor activity were characterized. Results: In tumor cell lines, TWEAK induces proliferation, survival, and NF-κB signaling and gene expression that promote tumor growth and suppress antitumor immune responses. TWEAK-inducible CD274, CCL2, CXCL-10 and -11 modulate T-cell and monocyte recruitment, T-cell activation, and macrophage differentiation. These factors and TWEAK-induced signaling were decreased, and tumor, blood, and spleen immune cell composition was altered with RG7212 treatment in mice. RG7212 inhibits tumor growth in vivo in models with TWEAK receptor, Fn14, expression, and markers of pathway activation. In phase I testing, signs of tumor shrinkage and stable disease were observed without dose-limiting toxicity. In a patient with advanced, Fn14-positive, malignant melanoma with evidence of tumor regression, proliferation markers were dramatically reduced, tumor T-cell infiltration increased, and tumor macrophage content decreased. Antitumor activity, a lack of toxicity in humans and animals and no evidence of antagonism with standard of care or targeted agents in mice, suggests that RG7212 is a promising agent for use in combination therapies in patients with Fn14-positive tumors. Clin Cancer Res; 19(20); 5686–98. ©2013 AACR.


Oncotarget | 2017

Combination GITR targeting/PD-1 blockade with vaccination drives robust antigen-specific antitumor immunity

Daniel O. Villarreal; Diana Chin; Melissa Smith; Leopoldo Luistro; Linda A. Snyder

Tumor progression is facilitated immunologically by mechanisms that include low antigen expression, an absence of coimmunostimulatory signals, and the presence of regulatory T cells (Tregs), all of which act to suppress and restrict effector T cells in the tumor. It may be possible to overcome these conditions by a combination of modulatory immunotherapy agents and tumor-antigen targeting to activate and drive effective antitumor T cell responses. Here, we demonstrated that co-administration of aGITR and aPD-1 monoclonal antibodies (mAb) in combination with a peptide vaccine (Vax) in mice bearing established tumors significantly delayed tumor growth and induced complete regression in 50% of the mice. This response was associated with increased expansion and functionality of potent Ag-specific polyfunctional CD8+ T cells, reduced Tregs, and the generation of memory T cells. Tumor regression correlated with the expansion of tumor-infiltrating antigen-specific CD8+ effector memory T cells, as depletion of this cell population significantly reduced the effectiveness of the triple combination Vax/aGITR/aPD-1 therapy. These findings support the concept that dual aGITR/aPD-1 combination with cancer vaccines may be a novel strategy against poorly immunogenic tumors.


Oncotarget | 2017

Targeting of CD122 enhances antitumor immunity by altering the tumor immune environment

Daniel O. Villarreal; Michael J. Allegrezza; Melissa Smith; Diana Chin; Leopoldo Luistro; Linda A. Snyder

Mounting evidence demonstrates that CD8+CD122+ T cells have suppressive properties with the capacity to inhibit T cell responses. Therefore, these cells are rational targets for cancer immunotherapy. Here, we demonstrate that CD122 monoclonal antibody (mAb; aCD122) therapy significantly suppressed tumor growth and improved long-term survival in tumor-bearing mice. This therapeutic effect correlated with enhanced polyfunctional, cytolytic intratumoral CD8+ T cells and a decrease in granulocytic myeloid-derived suppressor cells (G-MDSCs). In addition, aCD122 treatment synergized with a vaccine to augment vaccine-induced antigen (Ag)-specific CD8+ T cell responses, reject established tumors and generate memory T cells. Furthermore, aCD122 mAb synergized with an anti-GITR (aGITR) mAb to confer significant control of tumor growth. These results suggest CD122 might be a promising target for cancer immunotherapy, either as a single agent or in combination with other forms of immunotherapy.


Anti-Cancer Drugs | 2004

Antitumor activity of erlotinib (OSI-774, Tarceva) alone or in combination in human non-small cell lung cancer tumor xenograft models.

Brian Higgins; Kenneth Kolinsky; Melissa Smith; Gordon Beck; Mohammad Rashed; Violeta Adames; Michael Linn; Eric Wheeldon; Laurent Gand; Herbert Birnboeck; Gerhard Hoffmann


Journal of Medicinal Chemistry | 2006

Discovery of [4-Amino-2-(1-methanesulfonylpiperidin-4-ylamino)pyrimidin-5-yl](2,3-difluoro-6- methoxyphenyl)methanone (R547), a potent and selective cyclin-dependent kinase inhibitor with significant in vivo antitumor activity.

Xin-Jie Chu; Wanda DePinto; David Joseph Bartkovitz; Sung-Sau So; Binh Thanh Vu; Kathryn Packman; Christine Lukacs; Qingjie Ding; Nan Jiang; Ka Wang; Petra Goelzer; Xuefeng Yin; Melissa Smith; Brian Higgins; Yingsi Chen; Qing Xiang; John Anthony Moliterni; Gerald Kaplan; Bradford Graves; Allen John Lovey; Nader Fotouhi


Bioorganic & Medicinal Chemistry | 2005

RO4383596, an orally active KDR, FGFR, and PDGFR inhibitor : Synthesis and biological evaluation

Lee Apostle Mcdermott; Mary Ellen Simcox; Brian Higgins; Tom Nevins; Kenneth Kolinsky; Melissa Smith; Hong Yang; Jia K. Li; Yingsi Chen; June Ke; Navita L. Mallalieu; Tom Egan; Stan Kolis; Aruna Railkar; Louise Gerber; Kin-Chun Luk


Cancer Chemotherapy and Pharmacology | 2007

Antitumor activity of HER1/EGFR tyrosine kinase inhibitor erlotinib, alone and in combination with CPT-11 (irinotecan) in human colorectal cancer xenograft models

Jianping Chen; Melissa Smith; Kenneth Kolinsky; Violeta Adames; Nila Mehta; Luke Fritzky; Mohammad Rashed; Eric Wheeldon; Michael Linn; Brian Higgins


Bioorganic & Medicinal Chemistry Letters | 2006

Biological evaluation of a multi-targeted small molecule inhibitor of tumor-induced angiogenesis.

Lee Apostle Mcdermott; Brian Higgins; Mary Ellen Simcox; Kin-Chun Luk; Tom Nevins; Kenneth Kolinsky; Melissa Smith; Hong Yang; Jia K. Li; Yingsi Chen; June Ke; Navita L. Mallalieu; Tom Egan; Stan Kolis; Aruna Railkar; Louise Gerber; Jin-Jun Liu; Fred Konzelmann; Zhuming Zhang; Tom Flynn; Omar Morales; Yi Chen

Collaboration


Dive into the Melissa Smith'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge