Network


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

Hotspot


Dive into the research topics where Roberto Polakiewicz is active.

Publication


Featured researches published by Roberto Polakiewicz.


Clinical Cancer Research | 2009

Mutation-Specific Antibodies for the Detection of EGFR Mutations in Non–Small-Cell Lung Cancer

Jian Yu; Susan E. Kane; Jiong Wu; Elisa Benedettini; Daiqiang Li; Cynthia Reeves; Gregory Innocenti; Randy Wetzel; Katherine Crosby; Alison Becker; Michelle Ferrante; Wan Cheung Cheung; Xiqiang Hong; Lucian R. Chirieac; Lynette M. Sholl; Herbert Haack; Bradley L. Smith; Roberto Polakiewicz; Yi Tan; Ting-Lei Gu; Massimo Loda; Xinmin Zhou; Michael J. Comb

Purpose: Activating mutations within the tyrosine kinase domain of epidermal growth factor receptor (EGFR) are found in approximately 10% to 20% of non–small-cell lung cancer (NSCLC) patients and are associated with response to EGFR inhibitors. The most common NSCLC-associated EGFR mutations are deletions in exon 19 and L858R mutation in exon 21, together accounting for 90% of EGFR mutations. To develop a simple, sensitive, and reliable clinical assay for the identification of EGFR mutations in NSCLC patients, we generated mutation-specific rabbit monoclonal antibodies against each of these two most common EGFR mutations and aimed to evaluate the detection of EGFR mutations in NSCLC patients by immunohistochemistry. Experimental Design: We tested mutation-specific antibodies by Western blot, immunofluorescence, and immunohistochemistry. In addition, we stained 40 EGFR genotyped NSCLC tumor samples by immunohistochemistry with these antibodies. Finally, with a panel of four antibodies, we screened a large set of NSCLC patient samples with unknown genotype and confirmed the immunohistochemistry results by DNA sequencing. Results: These two antibodies specifically detect the corresponding mutant form of EGFR by Western blotting, immunofluorescence, and immunohistochemistry. Screening a panel of 340 paraffin-embedded NSCLC tumor samples with these antibodies showed that the sensitivity of the immunohistochemistry assay is 92%, with a specificity of 99% as compared with direct and mass spectrometry–based DNA sequencing. Conclusions: This simple assay for detection of EGFR mutations in diagnostic human tissues provides a rapid, sensitive, specific, and cost-effective method to identify lung cancer patients responsive to EGFR-based therapies.


The American Journal of Surgical Pathology | 2009

Diagnosis of NUT midline carcinoma using a NUT-specific monoclonal antibody.

Herbert Haack; Laura A. Johnson; Christopher J. Fry; Katherine Crosby; Roberto Polakiewicz; Edward B. Stelow; Seung-Mo Hong; Brian E. Schwartz; Michael J. Cameron; Mark A. Rubin; Martin C. Chang; Christopher A. French

NUT midline carcinoma (NMC) is a uniformly lethal malignancy that is defined by rearrangement of the nuclear protein in testis (NUT) gene on chromosome 15q14. NMCs are morphologically indistinguishable from other poorly differentiated carcinomas, and the diagnosis is usually made currently by fluorescence in situ hybridization (FISH). As normal NUT expression is confined to testis and ovary, we reasoned that an immunohistochemical (IHC) stain for NUT would be useful in diagnosing NMC. To this end, we raised a highly specific rabbit monoclonal antibody, C52, against a recombinant NUT polypeptide, and developed an IHC staining protocol. The sensitivity and specificity of C52 staining was evaluated in a panel of 1068 tissues, predominantly diverse types of carcinomas (n=906), including 30 NMCs. Split-apart FISH for NUT rearrangement was used as a “gold standard” diagnostic test for NMC. C52 immunoreactivity among carcinomas was confined to NMCs. IHC staining had a sensitivity of 87%, a specificity of 100%, a negative predictive value of 99%, and a positive predictive value of 100%. Two new cases of NMC containing BRD4-NUT fusions were detected by C52 IHC, but missed by conventional FISH. In both instances, these tumors contained cryptic BRD4-NUT rearrangements, as confirmed by FISH using a refined set of probes. Some germ cell tumors, including 64% of dysgerminomas, showed weak NUT immunoreactivity, consistent with the expression of NUT in normal germ cells. We conclude that IHC staining with the C52 monoclonal antibody is a highly sensitive and specific test that reliably distinguishes NMC from other forms of carcinoma. The NUT antibody is being prepared for commercial release and will be available in the near future.


Journal of Biological Chemistry | 2008

HDAC6 Is Required for Epidermal Growth Factor-induced β-Catenin Nuclear Localization

Yu Li; Xiaowu Zhang; Roberto Polakiewicz; Tso-Pang Yao; Michael J. Comb

Nuclear translocation of β-catenin is a hallmark of Wnt signaling and is associated with various cancers. In addition to the canonical Wnt pathway activated by Wnt ligands, growth factors such as epidermal growth factor (EGF) also induce β-catenin dissociation from the adherens junction complex, translocation into the nucleus, and activation of target genes such as c-myc. Here we report that EGF-induced β-catenin nuclear localization and activation of c-myc are dependent on the deacetylase HDAC6. We show that EGF induces HDAC6 translocation to the caveolae membrane and association with β-catenin. HDAC6 deacetylates β-catenin at lysine 49, a site frequently mutated in anaplastic thyroid cancer, and inhibits β-catenin phosphorylation at serine 45. HDAC6 inactivation blocks EGF-induced β-catenin nuclear localization and decreases c-Myc expression, leading to inhibition of tumor cell proliferation. These results suggest that EGF-induced nuclear localization of β-catenin is regulated by HDAC6-dependent deacetylation and provide a new mechanism by which HDAC inhibitors prevent tumor growth.


Molecular & Cellular Proteomics | 2012

PTMScan Direct: Identification and Quantification of Peptides from Critical Signaling Proteins by Immunoaffinity Enrichment Coupled with LC-MS/MS

Matthew P. Stokes; Charles Farnsworth; Albrecht Moritz; Xiaoying Jia; Kimberly Lee; Ailan Guo; Roberto Polakiewicz; Michael J. Comb

Proteomic studies of post-translational modifications by metal affinity or antibody-based methods often employ data-dependent analysis, providing rich data sets that consist of randomly sampled identified peptides because of the dynamic response of the mass spectrometer. This can complicate the primary goal of programs for drug development, mutational analysis, and kinase profiling studies, which is to monitor how multiple nodes of known, critical signaling pathways are affected by a variety of treatment conditions. Cell Signaling Technology has developed an immunoaffinity-based LC-MS/MS method called PTMScan Direct for multiplexed analysis of these important signaling proteins. PTMScan Direct enables the identification and quantification of hundreds of peptides derived from specific proteins in signaling pathways or specific protein types. Cell lines, tissues, or xenografts can be used as starting material. PTMScan Direct is compatible with both SILAC and label-free quantification. Current PTMScan Direct reagents target key nodes of many signaling pathways (PTMScan Direct: Multipathway), serine/threonine kinases, tyrosine kinases, and the Akt/PI3K pathway. Validation of each reagent includes score filtering of MS/MS assignments, filtering by identification of peptides derived from expected targets, identification of peptides homologous to expected targets, minimum signal intensity of peptide ions, and dependence upon the presence of the reagent itself compared with a negative control. The Multipathway reagent was used to study sensitivity of human cancer cell lines to receptor tyrosine kinase inhibitors and showed consistent results with previously published studies. The Ser/Thr kinase reagent was used to compare relative levels of kinase-derived phosphopeptides in mouse liver, brain, and embryo, showing tissue-specific activity of many kinases including Akt and PKC family members. PTMScan Direct will be a powerful quantitative method for elucidation of changes in signaling in a wide array of experimental systems, combining the specificity of traditional biochemical methods with the high number of data points and dynamic range of proteomic methods.


Developmental Brain Research | 1989

Postnatal expression of opioid genes in rat brain

Haim Rosen; Roberto Polakiewicz

Developmentally regulated expression of opioid genes in rat brain was studied by Northern blot hybridization, using cloned rat genes as probes. Total RNA was isolated from the rat medulla oblongata, hypothalamus and striatum at 11 stages of development from birth to postnatal day 28. The steady-state levels of opioid precursor mRNAs in the above tissues were determined by hybridization to 32P-labeled opioid DNA probes and densitometric scanning of the appropriate bands. Analysis of the relative steady-state concentrations of opioid mRNAs has shown that the tissue distribution and size of these mRNAs during postnatal development of the CNS are similar to the pattern described in the adult. However, up to 50-fold changes in the relative mRNA concentrations were observed. These changes are tissue-specific, characterized by a peak within the postnatal stage of development, and are followed by a build-up of mRNA levels characteristic of the adult. Moreover, comparison of the changes in mRNA levels of proenkephalin to those of prodynorphin at their main site of synthesis in the brain, namely the striatum, revealed striking similarities in pattern. The association of this phenomenon with a possible role of opioid genes in development is suggested.


Scientific Reports | 2016

Antibodies elicited by the first non-viral prophylactic cancer vaccine show tumor-specificity and immunotherapeutic potential.

Jason J. Lohmueller; Shuji Sato; Lana Popova; Isabel Chu; Meghan Tucker; Roberto Barberena; Gregory Innocenti; Mare Cudic; James D. Ham; Wan Cheung Cheung; Roberto Polakiewicz; Olivera J. Finn

MUC1 is a shared tumor antigen expressed on >80% of human cancers. We completed the first prophylactic cancer vaccine clinical trial based on a non-viral antigen, MUC1, in healthy individuals at-risk for colon cancer. This trial provided a unique source of potentially effective and safe immunotherapeutic drugs, fully-human antibodies affinity-matured in a healthy host to a tumor antigen. We purified, cloned, and characterized 13 IgGs specific for several tumor-associated MUC1 epitopes with a wide range of binding affinities. These antibodies bind hypoglycosylated MUC1 on human cancer cell lines and tumor tissues but show no reactivity against fully-glycosylated MUC1 on normal cells and tissues. We found that several antibodies activate complement-mediated cytotoxicity and that T cells carrying chimeric antigen receptors with the antibody variable regions kill MUC1+ target cells, express activation markers, and produce interferon gamma. Fully-human and tumor-specific, these antibodies are candidates for further testing and development as immunotherapeutic drugs.


International Journal of Molecular Sciences | 2012

Quantitative Profiling of DNA Damage and Apoptotic Pathways in UV Damaged Cells Using PTMScan Direct

Matthew P. Stokes; Xiaoying Jia; Kimberly Lee; Roberto Polakiewicz; Michael J. Comb

Traditional methods for analysis of peptides using liquid chromatography and tandem mass spectrometry (LC-MS/MS) lack the specificity to comprehensively monitor specific biological processes due to the inherent duty cycle limitations of the MS instrument and the stochastic nature of the analytical platform. PTMScan Direct is a novel, antibody-based method that allows quantitative LC-MS/MS profiling of specific peptides from proteins that reside in the same signaling pathway. New PTMScan Direct reagents have been produced that target peptides from proteins involved in DNA Damage/Cell Cycle and Apoptosis/Autophagy pathways. Together, the reagents provide access to 438 sites on 237 proteins in these signaling cascades. These reagents have been used to profile the response to UV damage of DNA in human cell lines. UV damage was shown to activate canonical DNA damage response pathways through ATM/ATR-dependent signaling, stress response pathways and induce the initiation of apoptosis, as assessed by an increase in the abundance of peptides corresponding to cleaved, activated caspases. These data demonstrate the utility of PTMScan Direct as a multiplexed assay for profiling specific cellular responses to various stimuli, such as UV damage of DNA.


Brain Research | 1989

Increase in hypothalamic pro-opiomelanocortin gene expression in response to prolonged low level lead exposure

Haim Rosen; Roberto Polakiewicz

Lead, an environmental pollutant, has been long recognized as a neurotoxic agent. Several reports have recently described behavioral disturbances caused by low level lead exposure. Lead-induced changes in neurotransmitter metabolism in the brain may help to elucidate the molecular basis for the observed behavioral alterations. In this study, the influence of low level lead exposure on the hypothalamic expression of rat opioid genes was examined. Determination of relative opioid mRNA levels revealed a dramatic increase of pro-opiomelanocortin (POMC) mRNA concentrations in response to lead. This increase was found to be specific, developmentally regulated, reversible and dependent on prolonged lead exposure. These findings may provide substantial evidence for the possible connections between early environmental inputs and the expression of neuropeptide genes in the brain.


Blood | 2006

A common phosphotyrosine signature for the Bcr-Abl kinase

Valerie Goss; Kimberly Lee; Albrecht Moritz; Julie Nardone; Erik Spek; Joan MacNeill; John Rush; Michael J. Comb; Roberto Polakiewicz


Proceedings of the National Academy of Sciences of the United States of America | 1991

Proenkephalin A in bone-derived cells.

Haim Rosen; Roberto Polakiewicz; Simone Benzakine; Zvi Bar-Shavit

Collaboration


Dive into the Roberto Polakiewicz's collaboration.

Top Co-Authors

Avatar

Kimberly Lee

Cell Signaling Technology

View shared research outputs
Top Co-Authors

Avatar

Albrecht Moritz

Cell Signaling Technology

View shared research outputs
Top Co-Authors

Avatar

John Rush

Cell Signaling Technology

View shared research outputs
Top Co-Authors

Avatar

Valerie Goss

Cell Signaling Technology

View shared research outputs
Top Co-Authors

Avatar

Ailan Guo

Cell Signaling Technology

View shared research outputs
Top Co-Authors

Avatar

Michael J. Comb

Cell Signaling Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ting-Lei Gu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Haim Rosen

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge