Network


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

Hotspot


Dive into the research topics where Chenwei Lin is active.

Publication


Featured researches published by Chenwei Lin.


Nature | 2016

Proteogenomics connects somatic mutations to signalling in breast cancer

Philipp Mertins; D. R. Mani; Kelly V. Ruggles; Michael A. Gillette; Karl R. Clauser; Pei Wang; Xianlong Wang; Jana W. Qiao; Song Cao; Francesca Petralia; Emily Kawaler; Filip Mundt; Karsten Krug; Zhidong Tu; Jonathan T. Lei; Michael L. Gatza; Matthew D. Wilkerson; Charles M. Perou; Venkata Yellapantula; Kuan Lin Huang; Chenwei Lin; Michael D. McLellan; Ping Yan; Sherri R. Davies; R. Reid Townsend; Steven J. Skates; Jing Wang; Bing Zhang; Christopher R. Kinsinger; Mehdi Mesri

Summary Somatic mutations have been extensively characterized in breast cancer, but the effects of these genetic alterations on the proteomic landscape remain poorly understood. We describe quantitative mass spectrometry-based proteomic and phosphoproteomic analyses of 105 genomically annotated breast cancers of which 77 provided high-quality data. Integrated analyses allowed insights into the somatic cancer genome including the consequences of chromosomal loss, such as the 5q deletion characteristic of basal-like breast cancer. The 5q trans effects were interrogated against the Library of Integrated Network-based Cellular Signatures, thereby connecting CETN3 and SKP1 loss to elevated expression of EGFR, and SKP1 loss also to increased SRC. Global proteomic data confirmed a stromal-enriched group in addition to basal and luminal clusters and pathway analysis of the phosphoproteome identified a G Protein-coupled receptor cluster that was not readily identified at the mRNA level. Besides ERBB2, other amplicon-associated, highly phosphorylated kinases were identified, including CDK12, PAK1, PTK2, RIPK2 and TLK2. We demonstrate that proteogenomic analysis of breast cancer elucidates functional consequences of somatic mutations, narrows candidate nominations for driver genes within large deletions and amplified regions, and identifies therapeutic targets.


Nature Biotechnology | 2011

A targeted proteomics–based pipeline for verification of biomarkers in plasma

Jeffrey R. Whiteaker; Chenwei Lin; Jacob Kennedy; Liming Hou; Mary Trute; Izabela Sokal; Ping Yan; Regine M. Schoenherr; Lei Zhao; Uliana J. Voytovich; Karen S. Kelly-Spratt; Alexei L. Krasnoselsky; Philip R. Gafken; Jason M. Hogan; Lisa A. Jones; Pei Wang; Lynn M. Amon; Lewis A. Chodosh; Peter S. Nelson; Martin W. McIntosh; Christopher J. Kemp; Amanda G. Paulovich

High-throughput technologies can now identify hundreds of candidate protein biomarkers for any disease with relative ease. However, because there are no assays for the majority of proteins and de novo immunoassay development is prohibitively expensive, few candidate biomarkers are tested in clinical studies. We tested whether the analytical performance of a biomarker identification pipeline based on targeted mass spectrometry would be sufficient for data-dependent prioritization of candidate biomarkers, de novo development of assays and multiplexed biomarker verification. We used a data-dependent triage process to prioritize a subset of putative plasma biomarkers from >1,000 candidates previously identified using a mouse model of breast cancer. Eighty-eight novel quantitative assays based on selected reaction monitoring mass spectrometry were developed, multiplexed and evaluated in 80 plasma samples. Thirty-six proteins were verified as being elevated in the plasma of tumor-bearing animals. The analytical performance of this pipeline suggests that it should support the use of an analogous approach with human samples.


Molecular & Cellular Proteomics | 2011

Evaluation of large scale quantitative proteomic assay development using peptide affinity-based mass spectrometry

Jeffrey R. Whiteaker; Lei Zhao; Susan E. Abbatiello; Michael Burgess; Eric Kuhn; Chenwei Lin; Matthew E. Pope; Morteza Razavi; N. Leigh Anderson; Terry W. Pearson; Steven A. Carr; Amanda G. Paulovich

Stable isotope standards and capture by antipeptide antibodies (SISCAPA) couples affinity enrichment of peptides with stable isotope dilution and detection by multiple reaction monitoring mass spectrometry to provide quantitative measurement of peptides as surrogates for their respective proteins. In this report, we describe a feasibility study to determine the success rate for production of suitable antibodies for SISCAPA assays in order to inform strategies for large-scale assay development. A workflow was designed that included a multiplex immunization strategy in which up to five proteotypic peptides from a single protein target were used to immunize individual rabbits. A total of 403 proteotypic tryptic peptides representing 89 protein targets were used as immunogens. Antipeptide antibody titers were measured by ELISA and 220 antipeptide antibodies representing 89 proteins were chosen for affinity purification. These antibodies were characterized with respect to their performance in SISCAPA-multiple reaction monitoring assays using trypsin-digested human plasma matrix. More than half of the assays generated were capable of detecting the target peptide at concentrations of less than 0.5 fmol/μl in human plasma, corresponding to protein concentrations of less than 100 ng/ml. The strategy of multiplexing five peptide immunogens was successful in generating a working assay for 100% of the targeted proteins in this evaluation study. These results indicate it is feasible for a single laboratory to develop hundreds of assays per year and allow planning for cost-effective generation of SISCAPA assays.


Nature Methods | 2014

Demonstrating the feasibility of large-scale development of standardized assays to quantify human proteins

Jacob Kennedy; Susan E. Abbatiello; Kyunggon Kim; Ping Yan; Jeffrey R. Whiteaker; Chenwei Lin; Jun Seok Kim; Yuzheng Zhang; Xianlong Wang; Richard G. Ivey; Lei Zhao; Hophil Min; Youngju Lee; Myeong Hee Yu; Eun Gyeong Yang; Cheolju Lee; Pei Wang; Henry Rodriguez; Youngsoo Kim; Steven A. Carr; Amanda G. Paulovich

Multiple reaction monitoring (MRM) mass spectrometry has been successfully applied to monitor targeted proteins in biological specimens, raising the possibility that assays could be configured to measure all human proteins. We report the results of a pilot study designed to test the feasibility of a large-scale, international effort for MRM assay generation. We have configured, validated across three laboratories and made publicly available as a resource to the community 645 novel MRM assays representing 319 proteins expressed in human breast cancer. Assays were multiplexed in groups of >150 peptides and deployed to quantify endogenous analytes in a panel of breast cancer–related cell lines. The median assay precision was 5.4%, with high interlaboratory correlation (R2 > 0.96). Peptide measurements in breast cancer cell lines were able to discriminate among molecular subtypes and identify genome-driven changes in the cancer proteome. These results establish the feasibility of a large-scale effort to develop an MRM assay resource.


Molecular & Cellular Proteomics | 2012

Sequential Multiplexed Analyte Quantification Using Peptide Immunoaffinity Enrichment Coupled to Mass Spectrometry

Jeffrey R. Whiteaker; Lei Zhao; Chenwei Lin; Ping Yan; Pei Wang; Amanda G. Paulovich

Peptide immunoaffinity enrichment coupled to selected reaction monitoring (SRM) mass spectrometry (immuno-SRM) has emerged as a technology with great potential for quantitative proteomic assays. One advantage over traditional immunoassays is the tremendous potential for concurrent quantification of multiple analytes from a given sample (i.e. multiplex analysis). We sought to explore the capacity of the immuno-SRM technique for analyzing large numbers of analytes by evaluating the multiplex capabilities and demonstrating the sequential analysis of groups of peptides from a single sample. To evaluate multiplex analysis, immuno-SRM assays were arranged in groups of 10, 20, 30, 40, and 50 peptides using a common set of reagents. The multiplex immuno-SRM assays were used to measure synthetic peptides added to plasma covering several orders of magnitude concentration. Measurements made in large multiplex groups were highly correlated (r2 ≥ 0.98) and featured good agreement (bias ≤ 1%) compared with single-plex assays or a 10-plex configuration. The ability to sequentially enrich sets of analyte peptides was demonstrated by enriching groups of 10 peptides from a plasma sample in a sequential fashion. The data show good agreement (bias ≤ 1.5%) and similar reproducibility regardless of enrichment order. These significant advancements demonstrate the utility of immuno-SRM for analyzing large numbers of analytes, such as in large biomarker verification experiments or in pathway-based targeted analysis.


Proteomics | 2012

Multiplexed quantification of estrogen receptor and HER2/Neu in tissue and cell lysates by peptide immunoaffinity enrichment mass spectrometry

Regine M. Schoenherr; Jeffrey R. Whiteaker; Lei Zhao; Richard G. Ivey; Mary Trute; Jacob Kennedy; Uliana J. Voytovich; Ping Yan; Chenwei Lin; Amanda G. Paulovich

Access to a wider range of quantitative protein assays would significantly impact the number and use of tissue markers in guiding disease treatment. Quantitative mass spectrometry‐based peptide and protein assays, such as immuno‐SRM assays, have seen tremendous growth in recent years in application to protein quantification in biological fluids such as plasma or urine. Here, we extend the capability of the technique by demonstrating the application of a multiplexed immuno‐SRM assay for quantification of estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2) levels in cell line lysates and human surgical specimens. The performance of the assay was characterized using peptide response curves, with linear ranges covering approximately four orders of magnitude and limits of detection in the low fmol/mg lysate range. Reproducibility was acceptable with median coefficients of variation of approximately 10%. We applied the assay to measurements of ER and HER2 in well‐characterized cell line lysates with good discernment based on ER/HER2 status. Finally, the proteins were measured in surgically resected breast cancers, and the results showed good correlation with ER/HER2 status determined by clinical assays. This is the first implementation of the peptide‐based immuno‐SRM assay technology in cell lysates and human surgical specimens.


Molecular & Cellular Proteomics | 2015

Peptide Immunoaffinity Enrichment and Targeted Mass Spectrometry Enables Multiplex, Quantitative Pharmacodynamic Studies of Phospho-Signaling

Jeffrey R. Whiteaker; Lei Zhao; Ping Yan; Richard G. Ivey; Uliana J. Voytovich; Heather D. Moore; Chenwei Lin; Amanda G. Paulovich

In most cell signaling experiments, analytes are measured one Western blot lane at a time in a semiquantitative and often poorly specific manner, limiting our understanding of network biology and hindering the translation of novel therapeutics and diagnostics. We show the feasibility of using multiplex immuno-MRM for phospho-pharmacodynamic measurements, establishing the potential for rapid and precise quantification of cell signaling networks. A 69-plex immuno-MRM assay targeting the DNA damage response network was developed and characterized by response curves and determinations of intra- and inter-assay repeatability. The linear range was ≥3 orders of magnitude, the median limit of quantification was 2.0 fmol/mg, the median intra-assay variability was 10% CV, and the median interassay variability was 16% CV. The assay was applied in proof-of-concept studies to immortalized and primary human cells and surgically excised cancer tissues to quantify exposure–response relationships and the effects of a genomic variant (ATM kinase mutation) or pharmacologic (kinase) inhibitor. The study shows the utility of multiplex immuno-MRM for simultaneous quantification of phosphorylated and nonmodified peptides, showing feasibility for development of targeted assay panels to cell signaling networks.


Molecular & Cellular Proteomics | 2016

Immobilized Metal Affinity Chromatography Coupled to Multiple Reaction Monitoring Enables Reproducible Quantification of Phospho-signaling

Jacob Kennedy; Ping Yan; Lei Zhao; Richard G. Ivey; Uliana J. Voytovich; Heather D. Moore; Chenwei Lin; Era L. Pogosova-Agadjanyan; Derek L. Stirewalt; Kerryn W. Reding; Jeffrey R. Whiteaker; Amanda G. Paulovich

A major goal in cell signaling research is the quantification of phosphorylation pharmacodynamics following perturbations. Traditional methods of studying cellular phospho-signaling measure one analyte at a time with poor standardization, rendering them inadequate for interrogating network biology and contributing to the irreproducibility of preclinical research. In this study, we test the feasibility of circumventing these issues by coupling immobilized metal affinity chromatography (IMAC)-based enrichment of phosphopeptides with targeted, multiple reaction monitoring (MRM) mass spectrometry to achieve precise, specific, standardized, multiplex quantification of phospho-signaling responses. A multiplex immobilized metal affinity chromatography- multiple reaction monitoring assay targeting phospho-analytes responsive to DNA damage was configured, analytically characterized, and deployed to generate phospho-pharmacodynamic curves from primary and immortalized human cells experiencing genotoxic stress. The multiplexed assays demonstrated linear ranges of ≥3 orders of magnitude, median lower limit of quantification of 0.64 fmol on column, median intra-assay variability of 9.3%, median inter-assay variability of 12.7%, and median total CV of 16.0%. The multiplex immobilized metal affinity chromatography- multiple reaction monitoring assay enabled robust quantification of 107 DNA damage-responsive phosphosites from human cells following DNA damage. The assays have been made publicly available as a resource to the community. The approach is generally applicable, enabling wide interrogation of signaling networks.


Proteomics Clinical Applications | 2011

Proteome and Transcriptome Profiles of a Her2/Neu-driven Mouse Model of Breast Cancer

Regine M. Schoenherr; Karen S. Kelly-Spratt; Chenwei Lin; Jeffrey R. Whiteaker; Tao Liu; Ted Holzman; Ilsa Coleman; Li Chia Feng; Travis D. Lorentzen; Alexei L. Krasnoselsky; Pei Wang; Yan Liu; Kay E. Gurley; Lynn M. Amon; Athena A. Schepmoes; Ronald J. Moore; David G. Camp; Lewis A. Chodosh; Richard D. Smith; Peter S. Nelson; Martin W. McIntosh; Christopher J. Kemp; Amanda G. Paulovich

Purpose: We generated extensive transcriptional and proteomic profiles from a Her2‐driven mouse model of breast cancer that closely recapitulates human breast cancer. This report makes these data publicly available in raw and processed forms, as a resource to the community. Importantly, we previously made biospecimens from this same mouse model freely available through a sample repository, so researchers can obtain samples to test biological hypotheses without the need of breeding animals and collecting biospecimens.


Radiation Research | 2014

The human salivary proteome is radiation responsive

Heather D. Moore; Richard G. Ivey; Uliana J. Voytovich; Chenwei Lin; Derek L. Stirewalt; Era L. Pogosova-Agadjanyan; Amanda G. Paulovich

In the event of a nuclear incident in a heavily populated area, the surge in demand for medical evaluation will likely overwhelm our emergency care system, compromising our ability to care for victims with life-threatening injuries or exposures. Therefore, there exists a need for a rapidly deployable biological assay for radiation exposure that can be performed in the field by individuals with little to no medical training. Saliva is an attractive biofluid for this purpose, due to the relative ease of its collection and the wide array of biomolecules it contains. To determine whether the human salivary proteome is responsive to ionizing radiation exposure, we characterized the abundances of salivary proteins in humans before and after total body irradiation. Using an assay panel targeting 90 analytes (growth factors, chemokines and cytokines), we identified proteins that were significantly radiation responsive in human saliva. The responses of three proteins (monocyte chemo-attractant protein 1, interleukin 8 and intercellular adhesion molecule 1) were confirmed using independent immunoassay platforms and then verified and further characterized in 130 saliva samples from a completely independent set of 38 patients undergoing total body irradiation. The results demonstrate the potential for detecting radiation exposure based on analysis of human saliva.

Collaboration


Dive into the Chenwei Lin's collaboration.

Top Co-Authors

Avatar

Amanda G. Paulovich

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jeffrey R. Whiteaker

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Ping Yan

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Lei Zhao

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Pei Wang

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Richard G. Ivey

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jacob Kennedy

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Regine M. Schoenherr

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Uliana J. Voytovich

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Heather D. Moore

Fred Hutchinson Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge