18 results on '"Johanna M. Schafer"'
Search Results
2. Sex-biased adaptive immune regulation in cancer development and therapy
- Author
-
Johanna M. Schafer, Tong Xiao, Hyunwoo Kwon, Katharine Collier, Yuzhou Chang, Hany Abdel-Hafiz, Chelsea Bolyard, Dongjun Chung, Yuanquan Yang, Debasish Sundi, Qin Ma, Dan Theodorescu, Xue Li, and Zihai Li
- Subjects
Physiology ,Immunology ,Cancer ,Science - Abstract
Summary: The cancer research field is finally starting to unravel the mystery behind why males have a higher incidence and mortality rate than females for nearly all cancer types of the non-reproductive systems. Here, we explain how sex – specifically sex chromosomes and sex hormones – drives differential adaptive immunity across immune-related disease states including cancer, and why males are consequently more predisposed to tumor development. We highlight emerging data on the roles of cell-intrinsic androgen receptors in driving CD8+ T cell dysfunction or exhaustion in the tumor microenvironment and summarize ongoing clinical efforts to determine the impact of androgen blockade on cancer immunotherapy. Finally, we outline a framework for future research in cancer biology and immuno-oncology, underscoring the importance of a holistic research approach to understanding the mechanisms of sex dimorphisms in cancer, so sex will be considered as an imperative factor for guiding treatment decisions in the future.
- Published
- 2022
- Full Text
- View/download PDF
3. Redefining the catalytic HECT domain boundaries for the HECT E3 ubiquitin ligase family
- Author
-
Emma I. Kane, Steven A. Beasley, Johanna M. Schafer, Justine E. Bohl, Young Sun Lee, Kayla J. Rich, Elizabeth F. Bosia, and Donald E. Spratt
- Subjects
Catalytic Domain ,Ubiquitin-Protein Ligases ,Ubiquitination ,Biophysics ,Humans ,Cell Biology ,Ubiquitins ,Molecular Biology ,Biochemistry - Abstract
There are 28 unique human members of the homologous to E6AP C-terminus (HECT) E3 ubiquitin ligase family. Each member of the HECT E3 ubiquitin ligases contains a conserved bilobal HECT domain of approximately 350 residues found near their C-termini that is responsible for their respective ubiquitylation activities. Recent studies have begun to elucidate specific roles that each HECT E3 ubiquitin ligase has in various cancers, age-induced neurodegeneration, and neurological disorders. New structural models have been recently released for some of the HECT E3 ubiquitin ligases, but many HECT domain structures have yet to be examined due to chronic insolubility and/or protein folding issues. Building on these recently published structural studies coupled with our in-house experiments discussed in the present study, we suggest that the addition of ∼50 conserved residues preceding the N-terminal to the current UniProt defined boundaries of the HECT domain are required for isolating soluble, stable, and active HECT domains. We show using in silico bioinformatic analyses coupled with secondary structural prediction software that this predicted N-terminal α-helix found in all 28 human HECT E3 ubiquitin ligases forms an obligate amphipathic α-helix that binds to a hydrophobic pocket found within the HECT N-terminal lobe. The present study brings forth the proposal to redefine the residue boundaries of the HECT domain to include this N-terminal extension that will likely be critical for future biochemical, structural, and therapeutic studies on the HECT E3 ubiquitin ligase family.
- Published
- 2022
4. Editorial Comment
- Author
-
Johanna M. Schafer and Debasish Sundi
- Subjects
Urology - Published
- 2022
5. Androgen conspires with the CD8 + T cell exhaustion program and contributes to sex bias in cancer
- Author
-
Hyunwoo Kwon, Johanna M. Schafer, No-Joon Song, Satoshi Kaneko, Anqi Li, Tong Xiao, Anjun Ma, Carter Allen, Komal Das, Lei Zhou, Brian Riesenberg, Yuzhou Chang, Payton Weltge, Maria Velegraki, David Y. Oh, Lawrence Fong, Qin Ma, Debasish Sundi, Dongjun Chung, Xue Li, and Zihai Li
- Subjects
Immunology ,General Medicine - Abstract
Sex bias exists in the development and progression of nonreproductive organ cancers, but the underlying mechanisms are enigmatic. Studies so far have focused largely on sexual dimorphisms in cancer biology and socioeconomic factors. Here, we establish a role for CD8 + T cell–dependent antitumor immunity in mediating sex differences in tumor aggressiveness, which is driven by the gonadal androgen but not sex chromosomes. A male bias exists in the frequency of intratumoral antigen-experienced Tcf7 /TCF1 + progenitor exhausted CD8 + T cells that are devoid of effector activity as a consequence of intrinsic androgen receptor (AR) function. Mechanistically, we identify a novel sex-specific regulon in progenitor exhausted CD8 + T cells and a pertinent contribution from AR as a direct transcriptional transactivator of Tcf7 /TCF1. The T cell–intrinsic function of AR in promoting CD8 + T cell exhaustion in vivo was established using multiple approaches including loss-of-function studies with CD8-specific Ar knockout mice. Moreover, ablation of the androgen-AR axis rewires the tumor microenvironment to favor effector T cell differentiation and potentiates the efficacy of anti–PD-1 immune checkpoint blockade. Collectively, our findings highlight androgen-mediated promotion of CD8 + T cell dysfunction in cancer and imply broader opportunities for therapeutic development from understanding sex disparities in health and disease.
- Published
- 2022
6. Systematic literature review and testing of HER2 status in urothelial carcinoma (UC)
- Author
-
Vadim S Koshkin, Christine Boyiddle, Naomi Schwartz, Judy Yu, Kristina S. Yu, Ashley Kang, Lisa Bloudek, Qijun Fang, Johanna M. Schafer, Amanda F. Baker, Farzaneh H. Sayedian, and Emilie Scherrer
- Subjects
Cancer Research ,Oncology - Abstract
556 Background: Recent clinical trials suggest an emerging role for HER2-targeted therapy in locally advanced and metastatic UC (LA/mUC). The prevalence of HER2 expression and gene amplification (encoded by ERBB2) in LA/mUC has not been well defined, as testing for HER2 expression in LA/mUC is not part of current routine practice and is not standardized. We report (1) findings of a systematic literature review (SLR) of HER2 status in LA/mUC and (2) preliminary results of an ongoing evaluation of HER2 status in UC assaying HER2 protein expression by immunohistochemistry (IHC) and gene amplification by in situ hybridization (ISH). Methods: (1) The SLR used databases PubMed and EMBASE to identify English-language studies of LA/mUC HER2 status published Jan2000 – Oct2021. We used the following definitions: HER2-positive (HER2+) was defined as IHC 3+, or IHC 2+ with HER2 gene amplification (Amp+). HER2-low was defined as IHC 2+/Amp–, or as IHC 1+. HER2-zero was defined as IHC 0. Weighted averages were calculated to estimate population prevalence. (2) Commercially sourced, formalin-fixed paraffin-embedded surgical resections of primary UC were evaluated by trained readers for HER2 protein expression using the VENTANA HER2/neu (4B5) Rabbit Monoclonal Primary Antibody IHC assay and for HER2 gene amplification using the VENTANA HER2 Dual ISH DNA Probe Cocktail that detects both ERBB2 and its residing chromosome, chromosome 17 (Chr17), using a two-color chromogenic stain. HER2 IHC staining was scored based on an established scoring algorithm for gastric cancer. HER2 gene amplification was defined by a HER2/Chr17 ratio ≥2.0. Results: (1) Of 744 records screened for the SLR, 45 studies reported HER2 status, including 10,602 patients (pts) with LA/mUC. A variety of assays and scoring guidelines were used. In the 4 studies (862 pts) reporting data applicable to our predefined criteria for HER2 status, the percentage of HER2+ ranged from 6.7% to 37.5% (weighted average, 13.1%; 95% CI, 7.3%–18.8%). (2) Of 252 UC samples evaluated, 38 were HER2+ (15.1%; 95% CI, 11.2%–20.0%), 74 were HER2-low (29.4%; 95% CI, 24.1%–35.3%), and 140 were HER2-zero (55.5%; 95% CI: 49.4%–61.6%; Table). The HER2 gene was amplified in 31 (12.3%), among them 24 (77.4%) at stage III or IV muscle-invasive UC (MIUC). Conclusions: The SLR revealed wide variability of HER2 status in LA/mUC, highlighting a lack of standardized methods for assessing and defining HER2 status. In our large study using standardized laboratory methods, 44% of UC samples were HER2+ or HER2-low, and HER2 status distribution was consistent with that reported for pts with LA/mUC. Results suggest a potentially important role for HER2-targeted therapy for UC. [Table: see text]
- Published
- 2023
7. Acquisition of aneuploidy drives mutant p53-associated gain-of-function phenotypes
- Author
-
Brian D. Lehmann, Clayton B. Marshall, Lindsay N. Redman-Rivera, Rachel Hongo, Ferrin C. Wheeler, Johanna M. Schafer, Kathryn E. Beckermann, Quanhu Sheng, Timothy M. Shaver, Hailing Jin, and Jennifer A. Pietenpol
- Subjects
Science ,Mutant ,Mutation, Missense ,General Physics and Astronomy ,Aneuploidy ,Biology ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,Loss of Function Mutation ,Neoplasms ,Genotype ,Genetic variation ,medicine ,Humans ,Missense mutation ,Tumour-suppressor proteins ,Genetics ,Regulation of gene expression ,Mutation ,Multidisciplinary ,Oncogenes ,General Chemistry ,medicine.disease ,Phenotype ,Gene Expression Regulation, Neoplastic ,Gain of Function Mutation ,Mutant Proteins ,Tumor Suppressor Protein p53 - Abstract
p53 is mutated in over half of human cancers. In addition to losing wild-type (WT) tumor-suppressive function, mutant p53 proteins are proposed to acquire gain-of-function (GOF) activity, leading to novel oncogenic phenotypes. To study mutant p53 GOF mechanisms and phenotypes, we genetically engineered non-transformed and tumor-derived WT p53 cell line models to express endogenous missense mutant p53 (R175H and R273H) or to be deficient for p53 protein (null). Characterization of the models, which initially differed only by TP53 genotype, revealed that aneuploidy frequently occurred in mutant p53-expressing cells. GOF phenotypes occurred clonally in vitro and in vivo, were independent of p53 alteration and correlated with increased aneuploidy. Further, analysis of outcome data revealed that individuals with aneuploid-high tumors displayed unfavorable prognoses, regardless of the TP53 genotype. Our results indicate that genetic variation resulting from aneuploidy accounts for the diversity of previously reported mutant p53 GOF phenotypes., Previous studies report that mutant p53 proteins have gain-of-function activities and cause oncogenic phenotypes. Herein, the authors engineered two isogenic epithelial cell lines to express wild-type or missense mutant p53 or be deficient for p53 protein and show that aneuploidy drives several of the GOF phenotypes previously ascribed to mutant p53.
- Published
- 2021
8. Tyramide Signal-Amplified Immunofluorescence of MYCN and MYC in Human Tissue Specimens and Cell Line Cultures
- Author
-
Johanna M. Schafer and Jennifer A. Pietenpol
- Subjects
Medulloblastoma ,Gene isoform ,medicine.diagnostic_test ,Cell growth ,Strategy and Management ,Mechanical Engineering ,Metals and Alloys ,Biology ,medicine.disease ,Immunofluorescence ,Industrial and Manufacturing Engineering ,Prostate cancer ,Cell culture ,Neuroblastoma ,medicine ,Cancer research ,Methods Article ,Transcription factor ,neoplasms - Abstract
MYC family members, MYC, MYCN, and MYCL, are oncogenic transcription factors that regulate the expression of genes involved in normal development, cell growth, proliferation, metabolism, and survival. While MYC is amplified and/or overexpressed across a variety of tissue types, MYCN is often overexpressed in tumors of the nervous system (neuroblastoma and medulloblastoma) or with neuroendocrine features (neuroendocrine prostate cancer). Given recent reports that MYCN expression is also deregulated in a variety of non-neuronal tissue types, we investigated whether MYCN was also deregulated in triple-negative breast cancer (TNBC). In contrast to previous individual immuno-fluorescence (IF) stains against higher expressing MYC family isoform protein, we developed an IF stain to simultaneously detect both MYCN- and MYC-expressing cells within the same tumor cell population. Our methodology allows for the detection of low level MYCN and MYC expression and can be multiplexed with additional protein probes. Herein, using tyramide signal amplification (TSA), we present two protocols for the IF detection of MYCN and MYC on formalin-fixed paraffin embedded (FFPE) tumor sections and in cell lines fixed in situ after growth as adherent cultures on chambered microscope slides.
- Published
- 2020
9. Targeting MYCN-expressing triple-negative breast cancer with BET and MEK inhibitors
- Author
-
Johanna M. Schafer, Melinda E. Sanders, Bapsi Chakravarthy, Peggy Scherle, Matthew C. Stubbs, Jennifer A. Pietenpol, Clayton B. Marshall, Hailing Jin, Yu Shyr, Violeta Sanchez, Brian D. Lehmann, Phillip Liu, Lindsay N. Redman, Scott W. Hiebert, Justin M. Balko, Paula I. Gonzalez-Ericsson, J. Scott Beeler, Kimberly N. Johnson, Quanhu Sheng, Bret C. Mobley, Joshua A. Bauer, and Joseph T. Roland
- Subjects
Mitogen-Activated Protein Kinase Kinases ,N-Myc Proto-Oncogene Protein ,Chemotherapy ,Oncogene ,business.industry ,medicine.medical_treatment ,Proteins ,Triple Negative Breast Neoplasms ,General Medicine ,medicine.disease ,Xenograft Model Antitumor Assays ,Article ,In vitro ,Bromodomain ,Breast cancer ,In vivo ,Cell culture ,Cell Line, Tumor ,Cancer research ,medicine ,Animals ,Humans ,Neoplasm Recurrence, Local ,business ,Triple-negative breast cancer - Abstract
Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer that does not respond to endocrine therapy or human epidermal growth factor receptor 2 (HER2)-targeted therapies. Individuals with TNBC experience higher rates of relapse and shorter overall survival compared to patients with receptor-positive breast cancer subtypes. Preclinical discoveries are needed to identify, develop, and advance new drug targets to improve outcomes for patients with TNBC. Herein, we report that MYCN, an oncogene typically overexpressed in tumors of the nervous system or with neuroendocrine features, is heterogeneously expressed within a substantial fraction of primary and recurrent TNBC and is expressed in an even higher fraction of TNBCs that do not display a pathological complete response after neoadjuvant chemotherapy. We performed high-throughput chemical screens on TNBC cell lines with varying amounts of MYCN expression and determined that cells with higher expression of MYCN were more sensitive to bromodomain and extra-terminal motif (BET) inhibitors. Combined BET and MEK inhibition resulted in a synergistic decrease in viability, both in vitro and in vivo, using cell lines and patient-derived xenograft (PDX) models. Our preclinical data provide a rationale to advance a combination of BET and MEK inhibitors to clinical investigation for patients with advanced MYCN-expressing TNBC.
- Published
- 2020
10. Abstract P2-09-24: Combination treatment with bromodomain and extra-terminal motif inhibitors in triple-negative breast cancer
- Author
-
Phillip Liu, Matthew C. Stubbs, Peggy Scherle, LN Redman, Brian D. Lehmann, Johanna M. Schafer, Bruce Ruggeri, and Jennifer A. Pietenpol
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Disease ,medicine.disease ,Bromodomain ,Combined treatment ,Breast cancer ,Apoptosis ,Internal medicine ,medicine ,Signal transduction ,business ,Triple-negative breast cancer - Abstract
Triple-negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer. TNBC affects younger women and is characterized by earlier rates of relapse, higher frequency of visceral metastases, and shorter survival outcomes when compared to ER+ or HER2+ disease. Although the disease only represents ˜15% of all breast cancer cases, it accounts for 25% of all breast cancer deaths – with treatment options currently limited to chemotherapy. Development of targeted therapies for TNBC is challenging due to molecular heterogeneity and lack of high-frequency “driver” alterations amenable to therapeutic intervention. Recent studies have demonstrated increased sensitivity of TNBC to the anti-proliferative effects of Bromodomain and Extra-Terminal motif inhibitor (BETi) compared to the other breast cancer subtypes. To determine mechanisms of sensitivity to BETi, we analyzed the effect of a BETi across a panel of TNBC cell line models and identified cell lines that were both sensitive and insensitive to BETi. With the intent of identifying biomarkers of sensitivity, we performed RNA-seq and precision nuclear run-on and sequencing (PRO-seq) on both sensitive and insensitive cell line models and data generated identified significant differences in key growth regulatory and apoptotic signaling pathways, including notable differences in Myc-dependent signaling. Our data suggest potential biomarkers of BETi-sensitivity that may be of value in further pre-clinical studies. Further, our results provide mechanistic rationale for combinations of BETi with select, targeted therapies in a disease that is in need of new therapeutic intervention. Citation Format: Schafer JM, Lehmann BD, Redman LN, Liu P, Stubbs M, Ruggeri B, Scherle P, Pietenpol JA. Combination treatment with bromodomain and extra-terminal motif inhibitors in triple-negative breast cancer [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P2-09-24.
- Published
- 2018
11. Abstract 2489: A functional genomics approach to determine mutant p53 gain-of-function mechanisms and phenotypes in tumorigenesis
- Author
-
Hailing Jin, Johanna M. Schafer, Kathryn E. Beckermann, Ferrin C. Wheeler, Lindsay N. Redman-Rivera, Brian D. Lehmann, Timothy M. Shaver, Rachel Hongo, Jennifer A. Pietenpol, and Quanhu Sheng
- Subjects
Cancer Research ,Gain of function ,Oncology ,Mutant ,medicine ,Computational biology ,Biology ,Carcinogenesis ,medicine.disease_cause ,Phenotype ,Functional genomics - Abstract
Two of the most common events in human tumors are mutation of the tumor suppressor gene TP53 and development of aneuploidy. In addition to losing their wild-type (WT) tumor-suppressive function, mutant p53 proteins are proposed to acquire gain-of-function (GOF) activity, leading to novel oncogenic phenotypes. Mechanistic understanding of mutant p53 GOF activities is complicated by the diversity and context-specific nature of reported GOF phenotypes. The study of mutant p53 GOF activities is especially challenging because mutations in p53 are positively correlated with the development of aneuploidy, which can increase heterogeneity through diverse chromosomal alterations and itself contributes to tumorigenesis. To study mutant p53 GOF mechanisms and phenotypes, we used CRISPR/Cas9-mediated genome editing and developed two isogenic epithelial cell line models (one non-transformed and one tumor-derived) that express the most frequently occurring p53 missense mutations (R175H and R273H), are deficient for functional p53 protein (null), or retain the wild-type (WT) protein. In these engineered models, endogenous p53 expression is regulated by the native p53 promoter, thus providing a controlled system for rigorous functional experimentation across different p53 states. Additionally, the use of clonally-derived cell lines originating from the same near diploid parental genetic background allows for assessment of the genomic alterations and resulting molecular heterogeneity following mutation of TP53. Through genomic, transcriptomic, and cellular based assays we have validated our cell line models and found that missense mutant and p53 null cells display loss of p53 function. Through functional genomics analyses comparing isogenic epithelial cells, which initially differed only by the TP53 genotype, we have evaluated the relationship between mutant p53 and aneuploidy and assessed whether our clonal cell lines display several previously reported mutant p53 GOF phenotypes such as altered gene expression, proliferation, metabolism, drug sensitivity, and migration. Further, using lentiviral mediated knockdown of p53 protein we evaluated the dependency of these phenotypes on expression of mutant p53 protein. Finally, data from The Cancer Genome Atlas (TCGA) was used for the analysis of manifestations of clinical mutant p53 GOF phenotypes. The dissection of mutant p53 GOF phenotypes will improve the current understanding of the role of mutant p53 in tumorigenesis. The results generated from these studies have the potential for clinical translation in major types of human cancer that have high-frequency p53 mutation. Citation Format: Lindsay N. Redman-Rivera, Timothy M. Shaver, Hailing Jin, Johanna M. Schafer, Quanhu Sheng, Rachel A. Hongo, Kathryn E. Beckermann, Brian D. Lehmann, Ferrin C. Wheeler, Jennifer A. Pietenpol. A functional genomics approach to determine mutant p53 gain-of-function mechanisms and phenotypes in tumorigenesis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2489.
- Published
- 2021
12. The Amyloid Precursor Protein Has a Flexible Transmembrane Domain and Binds Cholesterol
- Author
-
Wade D. Van Horn, Johanna M. Schafer, Yuanli Song, Paul J. Barrett, Andrew J. Beel, Eric J. Hustedt, Charles R. Sanders, and Arina Hadziselimovic
- Subjects
Amino Acid Motifs ,Molecular Sequence Data ,Plasma protein binding ,Protein Structure, Secondary ,Article ,Amyloid beta-Protein Precursor ,Amyloid precursor protein ,Humans ,Amino Acid Sequence ,Binding site ,Nuclear Magnetic Resonance, Biomolecular ,Peptide sequence ,Micelles ,chemistry.chemical_classification ,Binding Sites ,Multidisciplinary ,biology ,Cholesterol binding ,Electron Spin Resonance Spectroscopy ,Peptide Fragments ,Transmembrane protein ,Protein Structure, Tertiary ,Amino acid ,Transmembrane domain ,Cholesterol ,Biochemistry ,chemistry ,Mutation ,biology.protein ,Protein Binding - Abstract
Insights into Amyloidogenesis The amyloid-β (Aβ) peptides associated with Alzheimer's disease are generated by cleavage of the transmembrane C-terminal domain (C99) of the amyloid precursor protein by the enzyme γ-secretase. Barrett et al. (p. 1168 ) used nuclear magnetic resonance (NMR) and electron paramagnetic resonance spectroscopy to show that C99 contains surface-associated N- and C-terminal helices and a flexibly curved transmembrane helix that is well suited to processive cleavage by γ-secretase. Elevated cholesterol levels have been found to increase Aβ generation. NMR titration together with mutagenesis revealed a binding site for cholesterol within C99 that included a motif previously implicated in protein oligomerization.
- Published
- 2012
13. Abstract 1518: Mechanisms of bromodomain and extra-terminal motif inhibitor (BETi) sensitivity in triple-negative breast cancer (TNBC)
- Author
-
Peggy Scherle, Phillip Liu, Matthew C. Stubbs, Johanna M. Schafer, Jennifer A. Pietenpol, and Brian D. Lehmann
- Subjects
Cancer Research ,business.industry ,Estrogen receptor ,Disease ,Bioinformatics ,medicine.disease ,Bromodomain ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Oncology ,Apoptosis ,030220 oncology & carcinogenesis ,Progesterone receptor ,Cancer research ,Medicine ,030212 general & internal medicine ,Signal transduction ,business ,Triple-negative breast cancer - Abstract
Triple-negative breast cancers (TNBCs) lack expression of estrogen receptor (ER), progesterone receptor (PR), and amplifications in the human epidermal growth factor receptor 2 (HER2). With currently no FDA-approved targeted therapies for TNBC, patients with TNBC have a higher risk of local and distant recurrence, and an overall increased rate of mortality. Recent studies have demonstrated increased sensitivity of TNBC to the anti-proliferative effects of BETi compared to the other breast cancer subtypes. To determine mechanisms of sensitivity to BET inhibition, we analyzed the effect of two BET inhibitors, INCB054329 and OTX-015, across a panel of TNBC cell line models and identified cell lines that were both sensitive and insensitive to BETi. With the intent of identifying biomarkers of sensitivity, we performed RNA-seq and precision nuclear run-on and sequencing (PRO-seq) on both sensitive and resistant cell line models and data generated identified significant differences in key growth regulatory and apoptotic signaling pathways, including notable differences in Myc-dependent signaling. Our data suggest potential biomarkers of BETi sensitivity that may be of value in further pre-clinical studies. Further, our results provide mechanistic rationale for combinations of BETi with select, targeted therapies in a disease that is in need of new therapeutic intervention. Citation Format: Johanna Schafer, Brian Lehmann, Phillip Liu, Matthew Stubbs, Peggy Scherle, Jennifer Pietenpol. Mechanisms of bromodomain and extra-terminal motif inhibitor (BETi) sensitivity in triple-negative breast cancer (TNBC) [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 1518. doi:10.1158/1538-7445.AM2017-1518
- Published
- 2017
14. Abstract 531: Activity of the selective FGFR 1, 2 and 3 inhibitor INCB054828 in genetically-defined models of triple-negative breast cancer
- Author
-
Darlise DiMatteo, Jennifer A. Pietenpol, Melody Diamond, Bruce Ruggeri, Liangxing Wu, Luping Lin, Reid Huber, Alla Volgina, Peggy Scherle, Timothy Burn, Gregory Hollis, Jin Lu, Brian D. Lehmann, Sang Hyun Lee, Phillip C.C. Liu, and Johanna M. Schafer
- Subjects
0301 basic medicine ,Cancer Research ,Kinase ,Fibroblast growth factor receptor 1 ,Cancer ,Biology ,medicine.disease ,Fibroblast growth factor ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Oncology ,chemistry ,Fibroblast growth factor receptor ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Growth inhibition ,Triple-negative breast cancer - Abstract
Activation of the Fibroblast Growth Factor (FGF)-FGF Receptor (FGFR) signaling axis occurs in many human cancers. In preclinical models, cell lines with genetic aberrations in FGF/FGFR genes are preferentially inhibited by compounds that selectively target the FGFR kinase. INCB54828 is a potent, selective, and reversible inhibitor of FGFR1, 2 and 3 that is currently in Phase 2 clinical trials for advanced malignancies characterized by FGF-FGFR alterations. In this study, we investigated the efficacy of INCB054828 in models of triple-negative breast cancer (TNBC). FGFR1 and FGFR2 are amplified in approximately 4% and 5% of TNBC, respectively, and oncogenic fusion proteins including FGFR3-TACC3 have also been identified in some TNBC specimens. To profile the activity of INCB054828, we screened a panel of diverse TNBC cell lines that are representative of each of the four subtypes of TNBC. Three human TNBC lines MFM223, SUM185 and SUM52PE were highly sensitive to INCB054828 in viability assays. Each of these responsive cell lines has a known alteration in FGFR, whereas TNBC lines lacking any aberrations in FGF/FGFR genes were refractory to growth inhibition. Inhibition of cell viability was associated with suppression of growth promoting pathways including Ras-MAPK. To confirm this association in vivo, four PDX models of TNBC were tested: two chemo-refractory models with FGFR1 amplification (CNV = 4 and 6) and two without any known FGF/FGFR alterations. Both of the models with FGFR1 copy number gain showed a response to INCB054828 as monotherapy with 36 and 78% tumor growth inhibition that was statistically significant vs vehicle control (P Citation Format: Phillip C.C. Liu, Brian D. Lehmann, Bruce Ruggeri, Darlise DiMatteo, Johanna M. Schafer, Jin Lu, Sang Hyun Lee, Luping Lin, Timothy C. Burn, Melody Diamond, Alla Volgina, Liangxing Wu, Gregory Hollis, Reid Huber, Jennifer A. Pietenpol, Peggy Scherle. Activity of the selective FGFR 1, 2 and 3 inhibitor INCB054828 in genetically-defined models of triple-negative breast 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 531. doi:10.1158/1538-7445.AM2017-531
- Published
- 2017
15. PIK3CA mutations in androgen receptor-positive triple negative breast cancer confer sensitivity to the combination of PI3K and androgen receptor inhibitors
- Author
-
Melinda E. Sanders, Carlos L. Arteaga, Brian D. Lehmann, Jennifer A. Pietenpol, Henry L. Gomez, Xi Chen, Luojia Tang, Christopher S Pendleton, Justin M. Balko, Kimberly C. Johnson, Joshua A. Bauer, Johanna M. Schafer, and Gordon B. Mills
- Subjects
Bicalutamide ,Cell Survival ,Class I Phosphatidylinositol 3-Kinases ,Androgen Receptor Positive ,Antineoplastic Agents ,Triple Negative Breast Neoplasms ,Pharmacology ,Biology ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,medicine ,Androgen Receptor Antagonists ,Animals ,Cluster Analysis ,Humans ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Triple-negative breast cancer ,Phosphoinositide-3 Kinase Inhibitors ,Medicine(all) ,Gene Expression Profiling ,TOR Serine-Threonine Kinases ,Dihydrotestosterone ,Xenograft Model Antitumor Assays ,Androgen receptor ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,Drug Resistance, Neoplasm ,Receptors, Androgen ,Mutation ,Female ,medicine.drug ,Signal Transduction ,Research Article - Abstract
Introduction Triple negative breast cancer (TNBC) is a heterogeneous collection of biologically diverse cancers, which contributes to variable clinical outcomes. Previously, we identified a TNBC subtype that has a luminal phenotype and expresses the androgen receptor (AR+). TNBC cells derived from these luminal AR + tumors have high frequency phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) mutations. The purpose of this study was to determine if targeting phosphoinositide 3-kinase (PI3K) alone or in combination with an AR antagonist is effective in AR + TNBC. Methods We determined the frequency of activating PIK3CA mutations in AR + and AR- TNBC clinical cases. Using AR + TNBC cell line and xenograft models we evaluated the effectiveness of PI3K inhibitors, used alone or in combination with an AR antagonist, on tumor cell growth and viability. Results PIK3CA kinase mutations were highly clonal, more frequent in AR + vs. AR- TNBC (40% vs. 4%), and often associated with concurrent amplification of the PIK3CA locus. PI3K/mTOR inhibitors had an additive growth inhibitory effect when combined with genetic or pharmacological AR targeting in AR + TNBC cells. We also analyzed the combination of bicalutamide +/- the pan-PI3K inhibitor GDC-0941 or the dual PI3K/mTOR inhibitor GDC-0980 in xenograft tumor studies and observed additive effects. Conclusions While approximately one third of TNBC patients respond to neoadjuvant/adjuvant chemotherapy, recent studies have shown that patients with AR + TNBC are far less likely to benefit from the current standard of care chemotherapy regimens and novel targeted approaches need to be investigated. In this study, we show that activating PIK3CA mutations are enriched in AR + TNBC; and, we show that the growth and viability of AR + TNBC cell line models is significantly reduced after treatment with PI3K inhibitors used in combination with an AR antagonist. These results provide rationale for pre-selection of TNBC patients with a biomarker (AR expression) to investigate the use of AR antagonists in combination with PI3K/mTOR inhibitors. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0406-x) contains supplementary material, which is available to authorized users.
- Published
- 2014
16. The E3 ligase PARC mediates the degradation of cytosolic cytochrome c to promote survival in neurons and cancer cells
- Author
-
Johanna M. Schafer, Brian T. Golitz, Vijay Swahari, Allyson Evans, Yolanda Y. Huang, Anna R. Cliffe, Mohanish Deshmukh, Adam J. Kole, Vivian Gama, Yue Xiong, Xin Hai Pei, and Noah Sciaky
- Subjects
biology ,Cytochrome c ,HEK 293 cells ,Cell Biology ,Mitochondrion ,Biochemistry ,Molecular biology ,Cell biology ,Ubiquitin ligase ,Cytosol ,Apoptosis ,Cancer cell ,biology.protein ,APAF1 ,Molecular Biology - Abstract
The ability to withstand mitochondrial damage is especially critical for the survival of postmitotic cells, such as neurons. Likewise, cancer cells can also survive mitochondrial stress. We found that cytochrome c (Cyt c), which induces apoptosis upon its release from damaged mitochondria, is targeted for proteasome-mediated degradation in mouse neurons, cardiomyocytes, and myotubes and in human glioma and neuroblastoma cells, but not in proliferating human fibroblasts. In mouse neurons, apoptotic protease-activating factor 1 (Apaf-1) prevented the proteasome-dependent degradation of Cyt c in response to induced mitochondrial stress. An RNA interference screen in U-87 MG glioma cells identified p53-associated Parkin-like cytoplasmic protein (PARC, also known as CUL9) as an E3 ligase that targets Cyt c for degradation. The abundance of PARC positively correlated with differentiation in mouse neurons, and overexpression of PARC reduced the abundance of mitochondrially-released cytosolic Cyt c in various cancer cell lines and in mouse embryonic fibroblasts. Conversely, neurons from Parc-deficient mice had increased sensitivity to mitochondrial damage, and neuroblastoma or glioma cells in which PARC or ubiquitin was knocked down had increased abundance of mitochondrially-released cytosolic Cyt c and decreased viability in response to stress. These findings suggest that PARC-mediated ubiquitination and degradation of Cyt c is a strategy engaged by both neurons and cancer cells to prevent apoptosis during conditions of mitochondrial stress.
- Published
- 2014
- Full Text
- View/download PDF
17. Abstract P3-04-03: PIK3CA mutations in androgen receptor-positive triple negative breast cancer confer sensitivity to the combination of PI3K and androgen receptor inhibitors
- Author
-
Jennifer A. Pietenpol, Johanna M. Schafer, Melinda E. Sanders, Brian D. Lehmann, Luojia Tang, Carlos L. Arteaga, Xi Chen, Christopher S Pendleton, Gordon B. Mills, Joshua A. Bauer, Henry L. Gomez, Justin M. Balko, and Kimberly C. Johnson
- Subjects
Cancer Research ,Bicalutamide ,business.industry ,Kinase ,medicine.drug_class ,Androgen Receptor Positive ,Pharmacology ,medicine.disease ,Androgen ,Androgen receptor ,Breast cancer ,Oncology ,Cancer research ,Medicine ,business ,PI3K/AKT/mTOR pathway ,Triple-negative breast cancer ,medicine.drug - Abstract
Triple negative breast cancer (TNBC) is a heterogeneous collection of biologically diverse cancers, which contributes to variable clinical outcomes. Previously, we identified a TNBC subtype that has a luminal phenotype and expresses androgen receptor (AR+). TNBC cells derived from these luminal AR+ tumors have high frequency PIK3CA mutations. The purpose of this study was to determine if targeting PI3K alone or in combination with an AR antagonist is effective in AR+ TNBC. Methods: We determined the frequency of activating PIK3CA mutations in AR+ and AR- TNBC clinical cases. Using AR+ TNBC cell lines and xenograft models we evaluated the effectiveness of PI3K inhibitors, used alone or in combination with an AR antagonist, on tumor cell growth and viability. Results: PIK3CA kinase mutations were highly clonal, more frequent in AR+ vs. AR- TNBC (40% vs. 4%), and often associated with concurrent amplification of the PIK3CA locus. PI3K/mTOR inhibitors had an additive growth inhibitory effect when combined with genetic or pharmacological AR targeting in AR+ TNBC cells. We also analyzed the combination of bicalutamide +/- the pan-PI3K inhibitor GDC-0941 or the dual PI3K/mTOR inhibitor GDC-0980 in xenograft tumor studies and observed additive effects. Conclusions: While approximately one third of TNBC patients respond to neoadjuvant/adjuvant chemotherapy, recent studies have shown that patients with androgen receptor positive (AR+) TNBC are far less likely to benefit from the current standard of care chemotherapy regimens and novel targeted approaches need to be investigated. In this study, we show that activating PIK3CA mutations are enriched in AR+ TNBC; and, the growth and viability of AR+ TNBC cell line models is significantly reduced after treatment with PI3K inhibitors used in combination with an AR antagonist. These results provide rationale for pre-selection of TNBC patients with a biomarker (AR expression) to investigate the use of AR antagonists in combination with PI3K/mTOR inhibitors. Citation Format: Brian D Lehmann, Joshua A Bauer, Johanna M Schafer, Christopher S Pendleton, Luojia Tang, Kimberly C Johnson, Xi Chen, Justin M Balko, Henry Gomez, Carlos L Arteaga, Gordon B Mills, Melinda E Sanders, Jennifer A Pietenpol. PIK3CA mutations in androgen receptor-positive triple negative breast cancer confer sensitivity to the combination of PI3K and androgen receptor inhibitors [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr P3-04-03.
- Published
- 2015
18. Abstract P6-05-03: Targeted inhibition of recurrent PIK3CA mutations synergizes with bicalutamide in AR-expressing triple negative breast cancer
- Author
-
Christopher S Pendleton, Brian D. Lehmann, JA Bauer, Luojia Tang, Johanna M. Schafer, Jennifer A. Pietenpol, and Melinda E. Sanders
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Gene knockdown ,Bicalutamide ,business.industry ,Cancer ,medicine.disease ,Breast cancer ,Internal medicine ,Medicine ,business ,Receptor ,Protein kinase B ,Triple-negative breast cancer ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
Triple negative breast cancers (TNBC) are generally more aggressive than other types of breast cancer, with higher rates of relapse in the early stage and decreased overall survival in the metastatic setting. TNBC has been particularly difficult to treat given that the biology of the disease is not well understood, and until recently, molecular targets for therapeutic intervention have remained elusive. Lacking HER2 amplification as well as estrogen and progesterone receptors, TNBC appears to be a heterogeneous collection of biologically diverse cancers, which likely contributes to variable clinical outcomes for patients suffering from this disease. Using gene expression analyses, we recently identified six distinct molecular TNBC subtypes with unique biologies that include two basal-like (BL1 and BL2), an immunomodulatory (IM), a mesenchymal (M), a mesenchymal stem-like (MSL), and a luminal androgen receptor (LAR) subtype. We demonstrated that the growth of LAR cell line models was dependent on AR signaling, as siRNA-mediated AR knockdown or pharmacological inhibition by bicalutamide greatly diminished viability. In addition we previously demonstrated that LAR cell lines are highly sensitive to the PI3K/mTOR inhibitor NVP-BEZ235. Interestingly, we observed that all AR-positive TNBC cell lines examined contain PIK3CA mutations (H1047R) suggesting selection for deregulation of the PI3K pathways during evolution of AR expressing breast cancers. Further investigation of a panel of AR-positive TNBC tumors showed that PIK3CA kinase mutations are a frequent event in this TNBC subtype (69.2% vs. 26.9%) and not just an artifact of the LAR cell line models. We show that PI3K pathway activation plays a role in resistance to AR antagonists, as there are elevated levels of activated AKT in residual tumors after bicalutamide treatment of xenograft tumors. Furthermore, we demonstrate that genetic/pharmacological targeting of AR synergizes with PI3K/mTOR inhibition in both two- and three-dimensional cell culture models. Moreover, the combination of bicalutamide with PI3K/mTOR inhibitors GDC0980 or NVP-BEZ235 decreased tumor volume in mouse xenograft studies. Our preclinical data provide the rationale for pre-selecting AR-positive TNBC patients for future clinical trials evaluating the combination of AR antagonists with PI3K/mTOR inhibitors. Citation Information: Cancer Res 2012;72(24 Suppl):Abstract nr P6-05-03.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.