Network


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

Hotspot


Dive into the research topics where Lisa M. Bixby is active.

Publication


Featured researches published by Lisa M. Bixby.


Breast Cancer Research and Treatment | 2013

Characterization of cell lines derived from breast cancers and normal mammary tissues for the study of the intrinsic molecular subtypes

Aleix Prat; Olga Karginova; Joel S. Parker; Cheng Fan; Xiaping He; Lisa M. Bixby; J. Chuck Harrell; Erick Roman; Barbara Adamo; Melissa A. Troester; Charles M. Perou

Abstract Five molecular subtypes (luminal A, luminal B, HER2-enriched, basal-like, and claudin-low) with clinical implications exist in breast cancer. Here, we evaluated the molecular and phenotypic relationships of (1) a large in vitro panel of human breast cancer cell lines (BCCLs), human mammary fibroblasts (HMFs), and human mammary epithelial cells (HMECs); (2) in vivo breast tumors; (3) normal breast cell subpopulations; (4) human embryonic stem cells (hESCs); and (5) bone marrow-derived mesenchymal stem cells (hMSC). First, by integrating genomic data of 337 breast tumor samples with 93 cell lines we were able to identify all the intrinsic tumor subtypes in the cell lines, except for luminal A. Secondly, we observed that the cell lines recapitulate the differentiation hierarchy detected in the normal mammary gland, with claudin-low BCCLs and HMFs cells showing a stromal phenotype, HMECs showing a mammary stem cell/bipotent progenitor phenotype, basal-like cells showing a luminal progenitor phenotype, and luminal B cell lines showing a mature luminal phenotype. Thirdly, we identified basal-like and highly migratory claudin-low subpopulations of cells within a subset of triple-negative BCCLs (SUM149PT, HCC1143, and HCC38). Interestingly, both subpopulations within SUM149PT were enriched for tumor-initiating cells, but the basal-like subpopulation grew tumors faster than the claudin-low subpopulation. Finally, claudin-low BCCLs resembled the phenotype of hMSCs, whereas hESCs cells showed an epithelial phenotype without basal or luminal differentiation. The results presented here help to improve our understanding of the wide range of breast cancer cell line models through the appropriate pairing of cell lines with relevant in vivo tumor and normal cell counterparts.


Clinical Cancer Research | 2017

A strong B cell response is part of the immune landscape in human high-grade serous ovarian metastases.

Anne Montfort; Oliver M. T. Pearce; Eleni Maniati; Benjamin G. Vincent; Lisa M. Bixby; Steffen Böhm; Thomas Dowe; Edmund Wilkes; Probir Chakravarty; Richard G. Thompson; Joanne Topping; Pedro R. Cutillas; Michelle Lockley; Jonathan S. Serody; Melania Capasso; Frances R. Balkwill

Purpose: In high-grade serous ovarian cancer (HGSOC), higher densities of both B cells and the CD8+ T-cell infiltrate were associated with a better prognosis. However, the precise role of B cells in the antitumor response remains unknown. As peritoneal metastases are often responsible for relapse, our aim was to characterize the role of B cells in the antitumor immune response in HGSOC metastases. Experimental Design: Unmatched pre and post-chemotherapy HGSOC metastases were studied. B-cell localization was assessed by immunostaining. Their cytokines and chemokines were measured by a multiplex assay, and their phenotype was assessed by flow cytometry. Further in vitro and in vivo assays highlighted the role of B cells and plasma cell IgGs in the development of cytotoxic responses and dendritic cell activation. Results: B cells mainly infiltrated lymphoid structures in the stroma of HGSOC metastases. There was a strong B-cell memory response directed at a restricted repertoire of antigens and production of tumor-specific IgGs by plasma cells. These responses were enhanced by chemotherapy. Interestingly, transcript levels of CD20 correlated with markers of immune cytolytic responses and immune complexes with tumor-derived IgGs stimulated the expression of the costimulatory molecule CD86 on antigen-presenting cells. A positive role for B cells in the antitumor response was also supported by B-cell depletion in a syngeneic mouse model of peritoneal metastasis. Conclusions: Our data showed that B cells infiltrating HGSOC omental metastases support the development of an antitumor response. Clin Cancer Res; 23(1); 250–62. ©2016 AACR.


Bioinformatics | 2016

Assembly-based inference of B-cell receptor repertoires from short read RNA sequencing data with V'DJer.

Lisle E. Mose; Sara R. Selitsky; Lisa M. Bixby; David Marron; Michael D. Iglesia; Jonathan S. Serody; Charles M. Perou; Benjamin G. Vincent; Joel S. Parker

Motivation: B-cell receptor (BCR) repertoire profiling is an important tool for understanding the biology of diverse immunologic processes. Current methods for analyzing adaptive immune receptor repertoires depend upon PCR amplification of VDJ rearrangements followed by long read amplicon sequencing spanning the VDJ junctions. While this approach has proven to be effective, it is frequently not feasible due to cost or limited sample material. Additionally, there are many existing datasets where short-read RNA sequencing data are available but PCR amplified BCR data are not. Results: We present here V’DJer, an assembly-based method that reconstructs adaptive immune receptor repertoires from short-read RNA sequencing data. This method captures expressed BCR loci from a standard RNA-seq assay. We applied this method to 473 Melanoma samples from The Cancer Genome Atlas and demonstrate V’DJer’s ability to accurately reconstruct BCR repertoires from short read mRNA-seq data. Availability and Implementation: V’DJer is implemented in C/C ++, freely available for academic use and can be downloaded from Github: https://github.com/mozack/vdjer Contact: [email protected] or [email protected] Supplementary information: Supplementary data are available at Bioinformatics online.


Cancer immunology research | 2015

Peptide/MHC tetramer-based sorting of CD8 T cells to a Leukemia antigen yields clonotypes drawn nonspecifically from an underlying restricted repertoire

Sally A. Hunsucker; Colleen S. McGary; Benjamin G. Vincent; Atim A. Enyenihi; Jennifer P. Waugh; Karen P. McKinnon; Lisa M. Bixby; Patricia A. Ropp; James Coghill; William A. Wood; Don A. Gabriel; Stefanie Sarantopoulos; Thomas C. Shea; Jonathan S. Serody; Gheath Alatrash; Tania Rodriguez-Cruz; Gregory Lizée; Adam Buntzman; Jeffrey A. Frelinger; Gary L. Glish; Paul M. Armistead

Hunsucker, McGary, Vincent, and colleagues report that low-frequency, antigen-specific T-cell responses may be specifically tested using tetramer-based, single-cell sorting and sequencing of the antigen-specific TCRβ clonotypes, and then mapping them onto a patients TCRβ to quantify antigen-driven clonal expansion. Testing of T cell–based cancer therapeutics often involves measuring cancer antigen–specific T-cell populations with the assumption that they arise from in vivo clonal expansion. This analysis, using peptide/MHC tetramers, is often ambiguous. From a leukemia cell line, we identified a CDK4-derived peptide epitope, UNC-CDK4-1 (ALTPVVVTL), that bound HLA-A*02:01 with high affinity and could induce CD8+ T-cell responses in vitro. We identified UNC-CDK4-1/HLA-A*02:01 tetramer+ populations in 3 of 6 patients with acute myeloid leukemia who had undergone allogeneic stem cell transplantation. Using tetramer-based, single-cell sorting and T-cell receptor β (TCRβ) sequencing, we identified recurrent UNC-CDK4-1 tetramer–associated TCRβ clonotypes in a patient with a UNC-CDK4-1 tetramer+ population, suggesting in vivo T-cell expansion to UNC-CDK4-1. In parallel, we measured the patients TCRβ repertoire and found it to be highly restricted/oligoclonal. The UNC-CDK4-1 tetramer–associated TCRβ clonotypes represented >17% of the entire TCRβ repertoire—far in excess of the UNC-CDK4-1 tetramer+ frequency—indicating that the recurrent TCRβ clonotypes identified from UNC-CDK-4-1 tetramer+ cells were likely a consequence of the extremely constrained T-cell repertoire in the patient and not in vivo UNC-CDK4-1–driven clonal T-cell expansion. Mapping recurrent TCRβ clonotype sequences onto TCRβ repertoires can help confirm or refute antigen-specific T-cell expansion in vivo. Cancer Immunol Res; 3(3); 228–35. ©2015 AACR.


Blood | 2017

T-cell expression of AhR inhibits the maintenance of pTreg cells in the gastrointestinal tract in acute GVHD

Trisha A. Dant; Kaifeng L. Lin; Danny Bruce; Stephanie A. Montgomery; Oleg V. Kolupaev; Hemamalini Bommiasamy; Lisa M. Bixby; John T. Woosley; Karen P. McKinnon; Frank J. Gonzalez; Bruce R. Blazar; Benjamin G. Vincent; James Coghill; Jonathan S. Serody

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that affects the function and development of immune cells. Here, we show that recipient mice receiving AhR-/- T cells have improved survival and decreased acute graft-versus-host disease (aGVHD) in 2 different murine allogeneic bone marrow transplant (BMT) models. We also show that CD4+ T cells lacking AhR demonstrate reduced accumulation in secondary lymphoid tissue because of low levels of proliferation 4 days after BMT. Additionally, we found a significant increase in the quantity of peripherally induced regulatory donor T (pTreg) cells in the colon of recipients transplanted with AhR-/- T cells 14 days after transplant. Blockade of AhR using a clinically available AhR antagonist greatly enhanced the in vitro generation of inducible Treg (iTreg) cells from naïve CD4+ human T cells. We have identified AhR as a novel target on donor T cells that is critical to the pathogenesis of aGVHD.


PLOS ONE | 2018

Immuno-PET imaging of tumor-infiltrating lymphocytes using zirconium-89 radiolabeled anti-CD3 antibody in immune-competent mice bearing syngeneic tumors

Denis R. Beckford Vera; Christof C. Smith; Lisa M. Bixby; Dylan M. Glatt; Stuart S. Dunn; Ryoichi Saito; William Y. Kim; Jonathan S. Serody; Benjamin G. Vincent; Matthew C. Parrott

The ability to non-invasively monitor tumor-infiltrating T cells in vivo could provide a powerful tool to visualize and quantify tumor immune infiltrates. For non-invasive evaluations in vivo, an anti-CD3 mAb was modified with desferrioxamine (DFO) and radiolabeled with zirconium-89 (Zr-89 or 89Zr). Radiolabeled 89Zr-DFO-anti-CD3 was tested for T cell detection using positron emission tomography (PET) in both healthy mice and mice bearing syngeneic bladder cancer BBN975. In vivo PET/CT and ex vivo biodistribution demonstrated preferential accumulation and visualization of tracer in the spleen, thymus, lymph nodes, and bone marrow. In tumor bearing mice, 89Zr-DFO-anti-CD3 demonstrated an 11.5-fold increase in tumor-to-blood signal compared to isotype control. Immunological profiling demonstrated no significant change to total T cell count, but observed CD4+ T cell depletion and CD8+ T cell expansion to the central and effector memory. This was very encouraging since a high CD8+ to CD4+ T cell ratio has already been associated with better patient prognosis. Ultimately, this anti-CD3 mAb allowed for in vivo imaging of homeostatic T cell distribution, and more specifically tumor-infiltrating T cells. Future applications of this radiolabeled mAb against CD3 could include prediction and monitoring of patient response to immunotherapy.


Journal of Clinical Investigation | 2018

Endogenous retroviral signatures predict immunotherapy response in clear cell renal cell carcinoma

Christof C. Smith; Kathryn E. Beckermann; Dante S. Bortone; Aguirre A. de Cubas; Lisa M. Bixby; Samuel J. Lee; Anshuman Panda; Shridar Ganesan; Gyan Bhanot; Eric Wallen; Matthew I. Milowsky; William Y. Kim; W.Kimryn Rathmell; Ronald Swanstrom; Joel S. Parker; Jonathan S. Serody; Sara R. Selitsky; Benjamin G. Vincent

Human endogenous retroviruses (hERVs) are remnants of exogenous retroviruses that have integrated into the genome throughout evolution. We developed a computational workflow, hervQuant, which identified more than 3,000 transcriptionally active hERVs within The Cancer Genome Atlas (TCGA) pan-cancer RNA-Seq database. hERV expression was associated with clinical prognosis in several tumor types, most significantly clear cell renal cell carcinoma (ccRCC). We explored two mechanisms by which hERV expression may influence the tumor immune microenvironment in ccRCC: (i) RIG-I–like signaling and (ii) retroviral antigen activation of adaptive immunity. We demonstrated the ability of hERV signatures associated with these immune mechanisms to predict patient survival in ccRCC, independent of clinical staging and molecular subtyping. We identified potential tumor-specific hERV epitopes with evidence of translational activity through the use of a ccRCC ribosome profiling (Ribo-Seq) dataset, validated their ability to bind HLA in vitro, and identified the presence of MHC tetramer–positive T cells against predicted epitopes. hERV sequences identified through this screening approach were significantly more highly expressed in ccRCC tumors responsive to treatment with programmed death receptor 1 (PD-1) inhibition. hervQuant provides insights into the role of hERVs within the tumor immune microenvironment, as well as evidence that hERV expression could serve as a biomarker for patient prognosis and response to immunotherapy.


Cancer Research | 2018

Molecular subtype-specific immunocompetent models of high-grade urothelial carcinoma reveal differential neoantigen expression and response to immunotherapy

Ryoichi Saito; Christof C. Smith; Takanobu Utsumi; Lisa M. Bixby; Jordan Kardos; Sara E. Wobker; Kyle Stewart; Shengjie Chai; Ujjawal Manocha; Kevin M. Byrd; Jeffrey S. Damrauer; Scott E. Williams; Benjamin G. Vincent; William Y. Kim

High-grade urothelial cancer contains intrinsic molecular subtypes that exhibit differences in underlying tumor biology and can be divided into luminal-like and basal-like subtypes. We describe here the first subtype-specific murine models of bladder cancer and show that Upk3a-CreERT2; Trp53L/L; PtenL/L; Rosa26LSL-Luc (UPPL, luminal-like) and BBN (basal-like) tumors are more faithful to human bladder cancer than the widely used MB49 cells. Following engraftment into immunocompetent C57BL/6 mice, BBN tumors were more responsive to PD-1 inhibition than UPPL tumors. Responding tumors within the BBN model showed differences in immune microenvironment composition, including increased ratios of CD8+:CD4+ and memory:regulatory T cells. Finally, we predicted and confirmed immunogenicity of tumor neoantigens in each model. These UPPL and BBN models will be a valuable resource for future studies examining bladder cancer biology and immunotherapy.Significance: This work establishes human-relevant mouse models of bladder cancer. Cancer Res; 78(14); 3954-68. ©2018 AACR.


Cancer Research | 2017

Abstract 1654: Development of subtype specific mouse models of bladder cancer

Christof C. Smith; Ryoichi Saito; Lisa M. Bixby; Takanobu Utsumi; Jordan Kardos; Shengjie Chai; Sara E. Wobker; Bhavani Krishnan; Jeffrey S. Damrauer; Jonathan S. Serody; David B. Darr; Benjamin G. Vincent; William Y. Kim

Introduction: High-grade, muscle-invasive bladder cancer has recently been shown to harbor intrinsic molecular subtypes with distinct biologic features. Current murine models of bladder cancer, including the prominent carcinogen induced model MB49, do not account for subtype specific characteristics, leaving a gap in available tools for understanding subtype specific differences in bladder cancer. We have developed and validated immunocompetent, subtype specific models of bladder cancer, and we have used these models to assess differential responses to immune checkpoint inhibition. Methods: Two distinct models of murine bladder cancer were developed in a C57BL/6 background. The UPPL models were generated through Pten/Trp53 conditional knockout in Uroplakin3a expressing cells. BBN models were generated through exposure of wild-type C57BL/6 mice to the carcinogen N-Butyl-N-(4-hydmoxybutyl)nitrosamine and subsequent generation of cell lines from spontaneous tumors. RNAseq was performed on several BBN and UPPL tumors and cell lines, with findings validated with flow cytometry and T/B cell receptor (TCR/BCR) amplicon sequencing of tumor infiltrating lymphocytes (TILs). Results: BBN and UPPL models reflected characteristics of human basal and luminal bladder cancers, respectively. BBN (basal) models demonstrated higher immune gene signature expression, with concordantly higher numbers of TILs compared to the UPPL (luminal) model (p Discussion: We have developed two unique classes of murine bladder cancer lines, UPPL and BBN, with gene expression and TIL profiles that closely correlate with human luminal and basal bladder cancers, respectively. The BBN and UPPL subtype specific models can serve as a tool for elucidating bladder cancer responses to immunotherapy. The mixed response of BBN963 tumors to PD-1 blockade should be an asset for assessing pathways mediating response to checkpoint blockade as well as the value of combination therapy. [C.S., R.S, B.V, W.K contributed equally to this work] Citation Format: Christof C. Smith, Ryoichi Saito, Lisa M. Bixby, Takanobu Utsumi, Jordan Kardos, Shengjie Chai, Sara E. Wobker, Bhavani Krishnan, Jeffrey S. Damrauer, Jonathan S. Serody, David Darr, Benjamin G. Vincent, William Y. Kim. Development of subtype specific mouse models of bladder cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1654. doi:10.1158/1538-7445.AM2017-1654


The Journal of Urology | 2017

MP88-20 ESTABLISHMENT OF NOVEL MOUSE BLADDER CANCER CELL LINES MIMICKING INTRINSIC SUBTYPE OF HUMAN INVASIVE BLADDER CANCER

Ryoichi Saito; Christof C. Smith; Jordan Kardos; Lisa M. Bixby; Shengjie Chai; Jeffrey S. Damrauer; Takanobu Utsumi; Sara E. Wobker; Bhavani Krishnan; Osamu Ogawa; Benjamin G. Vincent; William Y. Kim

experiments such as Western blotting analysis(WB), Hoechst33342 staining and immunostaining. The evaluation of targeting KLF4 by miR145 was performed by luciferase assay. Moreover, the effects of knockdown of KLF4 on the various phenotypes of BC cells were also examined. Furthermore, the networks involving KLF4/PTBP1/PKMs in the Warburg effect relatedmolecules were examined by WB even in clinical BC samples. Finally, we examined immunohistochemical staining to evaluate KLF4 expression. RESULTS: The expression levels of miR-145 were significantly down-regulated in clinical BC samples and BC cells compared with those in normal tissues and HUC. Luciferase assay showed that miR-145 directly targeted KLF4. Also, the Warburg related affectgenes were modulated by the transfection of miR-145 or siR-KLF4 in BC cells. Moreover, the expression levels of KLF4, PTBP1, and PKM2 is up-regulated in all clinical samples by WB. Finally, we observed KLF4-positive staining in the tumor by immunohistochemical staining. CONCLUSIONS: In this study, we indicated that miR-145 affect the Warburg effect through targeting KLF4/PTBP1/PKMs axis in BC cells, which exhibited a significant cell growth inhibition.

Collaboration


Dive into the Lisa M. Bixby's collaboration.

Top Co-Authors

Avatar

Benjamin G. Vincent

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jonathan S. Serody

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

William Y. Kim

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Christof C. Smith

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Joel S. Parker

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jordan Kardos

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles M. Perou

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jeffrey S. Damrauer

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Ryoichi Saito

Tokyo Medical and Dental University

View shared research outputs
Researchain Logo
Decentralizing Knowledge