LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 35

Search options

  1. Article ; Online: High FOXA1 levels induce ER transcriptional reprogramming, a pro-metastatic secretome, and metastasis in endocrine-resistant breast cancer.

    Fu, Xiaoyong / Pereira, Resel / Liu, Chia-Chia / De Angelis, Carmine / Shea, Martin J / Nanda, Sarmistha / Qin, Lanfang / Mitchell, Tamika / Cataldo, Maria L / Veeraraghavan, Jamunarani / Sethunath, Vidyalakshmi / Giuliano, Mario / Gutierrez, Carolina / Győrffy, Balázs / Trivedi, Meghana V / Cohen, Ofir / Wagle, Nikhil / Nardone, Agostina / Jeselsohn, Rinath /
    Rimawi, Mothaffar F / Osborne, C Kent / Schiff, Rachel

    Cell reports

    2023  Volume 42, Issue 8, Page(s) 112821

    Abstract: Aberrant activation of the forkhead protein FOXA1 is observed in advanced hormone-related cancers. However, the key mediators of high FOXA1 signaling remain elusive. We demonstrate that ectopic high FOXA1 (H-FOXA1) expression promotes estrogen receptor- ... ...

    Abstract Aberrant activation of the forkhead protein FOXA1 is observed in advanced hormone-related cancers. However, the key mediators of high FOXA1 signaling remain elusive. We demonstrate that ectopic high FOXA1 (H-FOXA1) expression promotes estrogen receptor-positive (ER+) breast cancer (BC) metastasis in a xenograft mouse model. Mechanistically, H-FOXA1 reprograms ER-chromatin binding to elicit a core gene signature (CGS) enriched in ER+ endocrine-resistant (EndoR) cells. We identify Secretome14, a CGS subset encoding ER-dependent cancer secretory proteins, as a strong predictor for poor outcomes of ER+ BC. It is elevated in ER+ metastases vs. primary tumors, irrespective of ESR1 mutations. Genomic ER binding near Secretome14 genes is also increased in mutant ER-expressing or mitogen-treated ER+ BC cells and in ER+ metastatic vs. primary tumors, suggesting a convergent pathway including high growth factor receptor signaling in activating pro-metastatic secretome genes. Our findings uncover H-FOXA1-induced ER reprogramming that drives EndoR and metastasis partly via an H-FOXA1/ER-dependent secretome.
    Language English
    Publishing date 2023-07-18
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2649101-1
    ISSN 2211-1247 ; 2211-1247
    ISSN (online) 2211-1247
    ISSN 2211-1247
    DOI 10.1016/j.celrep.2023.112821
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: A Multiparameter Molecular Classifier to Predict Response to Neoadjuvant Lapatinib plus Trastuzumab without Chemotherapy in HER2+ Breast Cancer.

    Veeraraghavan, Jamunarani / Gutierrez, Carolina / De Angelis, Carmine / Davis, Robert / Wang, Tao / Pascual, Tomas / Selenica, Pier / Sanchez, Katherine / Nitta, Hiroaki / Kapadia, Monesh / Pavlick, Anne C / Galvan, Patricia / Rexer, Brent / Forero-Torres, Andres / Nanda, Rita / Storniolo, Anna M / Krop, Ian E / Goetz, Matthew P / Nangia, Julie R /
    Wolff, Antonio C / Weigelt, Britta / Reis-Filho, Jorge S / Hilsenbeck, Susan G / Prat, Aleix / Osborne, C Kent / Schiff, Rachel / Rimawi, Mothaffar F

    Clinical cancer research : an official journal of the American Association for Cancer Research

    2024  Volume 29, Issue 16, Page(s) 3101–3109

    Abstract: Purpose: Clinical trials reported 25% to 30% pathologic complete response (pCR) rates in HER2+ patients with breast cancer treated with anti-HER2 therapies without chemotherapy. We hypothesize that a multiparameter classifier can identify patients with ... ...

    Abstract Purpose: Clinical trials reported 25% to 30% pathologic complete response (pCR) rates in HER2+ patients with breast cancer treated with anti-HER2 therapies without chemotherapy. We hypothesize that a multiparameter classifier can identify patients with HER2-"addicted" tumors who may benefit from a chemotherapy-sparing strategy.
    Experimental design: Baseline HER2+ breast cancer specimens from the TBCRC023 and PAMELA trials, which included neoadjuvant treatment with lapatinib and trastuzumab, were used. In the case of estrogen receptor-positive (ER+) tumors, endocrine therapy was also administered. HER2 protein and gene amplification (ratio), HER2-enriched (HER2-E), and PIK3CA mutation status were assessed by dual gene protein assay (GPA), research-based PAM50, and targeted DNA-sequencing. GPA cutoffs and classifier of response were constructed in TBCRC023 using a decision tree algorithm, then validated in PAMELA.
    Results: In TBCRC023, 72 breast cancer specimens had GPA, PAM50, and sequencing data, of which 15 had pCR. Recursive partitioning identified cutoffs of HER2 ratio ≥ 4.6 and %3+ IHC staining ≥ 97.5%. With PAM50 and sequencing data, the model added HER2-E and PIK3CA wild-type (WT). For clinical implementation, the classifier was locked as HER2 ratio ≥ 4.5, %3+ IHC staining ≥ 90%, and PIK3CA-WT and HER2-E, yielding 55% and 94% positive (PPV) and negative (NPV) predictive values, respectively. Independent validation using 44 PAMELA cases with all three biomarkers yielded 47% PPV and 82% NPV. Importantly, our classifier's high NPV signifies its strength in accurately identifying patients who may not be good candidates for treatment deescalation.
    Conclusions: Our multiparameter classifier differentially identifies patients who may benefit from HER2-targeted therapy alone from those who need chemotherapy and predicts pCR to anti-HER2 therapy alone comparable with chemotherapy plus dual anti-HER2 therapy in unselected patients.
    MeSH term(s) Female ; Humans ; Antineoplastic Combined Chemotherapy Protocols ; Biomarkers, Tumor/metabolism ; Breast Neoplasms/diagnosis ; Breast Neoplasms/drug therapy ; Breast Neoplasms/genetics ; Class I Phosphatidylinositol 3-Kinases/genetics ; Lapatinib ; Neoadjuvant Therapy ; Quinazolines ; Receptor, ErbB-2/metabolism ; Trastuzumab ; Treatment Outcome
    Chemical Substances Biomarkers, Tumor ; Class I Phosphatidylinositol 3-Kinases (EC 2.7.1.137) ; Lapatinib (0VUA21238F) ; Quinazolines ; Receptor, ErbB-2 (EC 2.7.10.1) ; Trastuzumab (P188ANX8CK)
    Language English
    Publishing date 2024-03-14
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 1225457-5
    ISSN 1557-3265 ; 1078-0432
    ISSN (online) 1557-3265
    ISSN 1078-0432
    DOI 10.1158/1078-0432.CCR-22-3753
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: A Novel Neoplastic Fusion Transcript,

    Liu, Chia-Chia / Veeraraghavan, Jamunarani / Tan, Ying / Kim, Jin-Ah / Wang, Xian / Loo, Suet Kee / Lee, Sanghoon / Hu, Yiheng / Wang, Xiao-Song

    Clinical cancer research : an official journal of the American Association for Cancer Research

    2020  Volume 27, Issue 3, Page(s) 785–798

    Abstract: Purpose: Luminal B breast tumors are more aggressive estrogen receptor-positive (ER: Experimental design: We performed large-scale RNA sequencing analysis to identify chimerical transcripts preferentially expressed in luminal B breast cancer. The ... ...

    Abstract Purpose: Luminal B breast tumors are more aggressive estrogen receptor-positive (ER
    Experimental design: We performed large-scale RNA sequencing analysis to identify chimerical transcripts preferentially expressed in luminal B breast cancer. The lead candidate was validated by reverse transcription PCR in breast cancer tissues. The effects of inducible ectopic expression or genetic silencing were assessed by phenotypic assays such as MTS, transwell, and transendothelial migration assays, and by clonogenic assays to assess MEK inhibitor sensitivity. Subcellular fractionation, Western blots, and immunoprecipitation were performed to characterize the protein products and elucidate the engaged mechanisms.
    Results: Here we report a novel tumor-specific chimeric transcript
    Conclusions: This discovery sheds light on a new area of molecular pathobiology of luminal B tumors and implies potential new therapeutic opportunities for more aggressive breast tumors overexpressing this fusion.
    MeSH term(s) Breast/pathology ; Breast Neoplasms/drug therapy ; Breast Neoplasms/genetics ; Breast Neoplasms/pathology ; Cell Line, Tumor ; DNA-Binding Proteins/genetics ; Datasets as Topic ; Drug Resistance, Neoplasm/genetics ; Female ; Gene Expression Regulation, Neoplastic ; Gene Knockdown Techniques ; Humans ; MAP Kinase Signaling System/drug effects ; MAP Kinase Signaling System/genetics ; Mitogen-Activated Protein Kinase Kinases/metabolism ; Oncogene Proteins, Fusion/genetics ; Protein Serine-Threonine Kinases/genetics ; Protein-Tyrosine Kinases/genetics ; Pyridones/pharmacology ; Pyridones/therapeutic use ; Pyrimidinones/pharmacology ; Pyrimidinones/therapeutic use ; RNA-Binding Proteins/genetics ; RNA-Seq ; Dyrk Kinases
    Chemical Substances DNA-Binding Proteins ; Oncogene Proteins, Fusion ; Pyridones ; Pyrimidinones ; RAD51AP1 protein, human ; RNA-Binding Proteins ; trametinib (33E86K87QN) ; Protein-Tyrosine Kinases (EC 2.7.10.1) ; Protein Serine-Threonine Kinases (EC 2.7.11.1) ; Mitogen-Activated Protein Kinase Kinases (EC 2.7.12.2)
    Language English
    Publishing date 2020-11-10
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, Non-P.H.S.
    ZDB-ID 1225457-5
    ISSN 1557-3265 ; 1078-0432
    ISSN (online) 1557-3265
    ISSN 1078-0432
    DOI 10.1158/1078-0432.CCR-20-2769
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: High FOXA1 levels induce ER transcriptional reprogramming, a pro-metastatic secretome, and metastasis in endocrine-resistant breast cancer

    Xiaoyong Fu / Resel Pereira / Chia-Chia Liu / Carmine De Angelis / Martin J. Shea / Sarmistha Nanda / Lanfang Qin / Tamika Mitchell / Maria L. Cataldo / Jamunarani Veeraraghavan / Vidyalakshmi Sethunath / Mario Giuliano / Carolina Gutierrez / Balázs Győrffy / Meghana V. Trivedi / Ofir Cohen / Nikhil Wagle / Agostina Nardone / Rinath Jeselsohn /
    Mothaffar F. Rimawi / C. Kent Osborne / Rachel Schiff

    Cell Reports, Vol 42, Iss 8, Pp 112821- (2023)

    2023  

    Abstract: Summary: Aberrant activation of the forkhead protein FOXA1 is observed in advanced hormone-related cancers. However, the key mediators of high FOXA1 signaling remain elusive. We demonstrate that ectopic high FOXA1 (H-FOXA1) expression promotes estrogen ... ...

    Abstract Summary: Aberrant activation of the forkhead protein FOXA1 is observed in advanced hormone-related cancers. However, the key mediators of high FOXA1 signaling remain elusive. We demonstrate that ectopic high FOXA1 (H-FOXA1) expression promotes estrogen receptor-positive (ER+) breast cancer (BC) metastasis in a xenograft mouse model. Mechanistically, H-FOXA1 reprograms ER-chromatin binding to elicit a core gene signature (CGS) enriched in ER+ endocrine-resistant (EndoR) cells. We identify Secretome14, a CGS subset encoding ER-dependent cancer secretory proteins, as a strong predictor for poor outcomes of ER+ BC. It is elevated in ER+ metastases vs. primary tumors, irrespective of ESR1 mutations. Genomic ER binding near Secretome14 genes is also increased in mutant ER-expressing or mitogen-treated ER+ BC cells and in ER+ metastatic vs. primary tumors, suggesting a convergent pathway including high growth factor receptor signaling in activating pro-metastatic secretome genes. Our findings uncover H-FOXA1-induced ER reprogramming that drives EndoR and metastasis partly via an H-FOXA1/ER-dependent secretome.
    Keywords CP: Cancer ; Biology (General) ; QH301-705.5
    Language English
    Publishing date 2023-08-01T00:00:00Z
    Publisher Elsevier
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

  5. Article ; Online: Therapeutic role of recurrent ESR1-CCDC170 gene fusions in breast cancer endocrine resistance.

    Li, Li / Lin, Ling / Veeraraghavan, Jamunarani / Hu, Yiheng / Wang, Xian / Lee, Sanghoon / Tan, Ying / Schiff, Rachel / Wang, Xiao-Song

    Breast cancer research : BCR

    2020  Volume 22, Issue 1, Page(s) 84

    Abstract: Background: Endocrine therapy is the most common treatment for estrogen receptor (ER)-positive breast cancer, but its effectiveness is limited by high rates of primary and acquired resistance. There are likely many genetic causes, and recent studies ... ...

    Abstract Background: Endocrine therapy is the most common treatment for estrogen receptor (ER)-positive breast cancer, but its effectiveness is limited by high rates of primary and acquired resistance. There are likely many genetic causes, and recent studies suggest the important role of ESR1 mutations and fusions in endocrine resistance. Previously, we reported a recurrent ESR1 fusion called ESR1-CCDC170 in 6-8% of the luminal B breast cancers that has a worse clinical outcome after endocrine therapy. Despite being the most frequent ESR1 fusion, its functional role in endocrine resistance has not been studied in vivo, and the engaged mechanism and therapeutic relevance remain uncharacterized.
    Methods: The endocrine sensitivities of HCC1428 or T47D breast cancer cells following genetic perturbations of ESR1-CCDC170 were assessed using clonogenic assays and/or xenograft mouse models. The underlying mechanisms were investigated by reverse phase protein array, western blotting, immunoprecipitation, and bimolecular fluorescence complementation assays. The sensitivity of ESR1-CCDC170 expressing breast cancer cells to concomitant treatments of tamoxifen and HER/SRC inhibitors was assessed by clonogenic assays.
    Results: Our results suggested that different ESR1-CCDC170 fusions endow different levels of reduced endocrine sensitivity in vivo, resulting in significant survival disadvantages. Further investigation revealed a novel mechanism that ESR1-CCDC170 binds to HER2/HER3/SRC and activates SRC/PI3K/AKT signaling. Silencing of ESR1-CCDC170 in the fusion-positive cell line, HCC1428, downregulates HER2/HER3, represses pSRC/pAKT, and improves endocrine sensitivity. More important, breast cancer cells expressing ectopic or endogenous ESR1-CCDC170 are highly sensitive to treatment regimens combining endocrine agents with the HER2 inhibitor lapatinib and/or the SRC inhibitor dasatinib.
    Conclusion: ESR1-CCDC170 may endow breast cancer cell survival under endocrine therapy via maintaining/activating HER2/HER3/SRC/AKT signaling which implies a potential therapeutic strategy for managing these fusion positive tumors.
    MeSH term(s) Animals ; Antineoplastic Combined Chemotherapy Protocols/pharmacology ; Breast Neoplasms/drug therapy ; Breast Neoplasms/genetics ; Breast Neoplasms/pathology ; Carrier Proteins/genetics ; Cell Line, Tumor ; Dasatinib/administration & dosage ; Drug Resistance, Neoplasm ; Estrogen Receptor alpha/genetics ; Female ; Fulvestrant/administration & dosage ; Humans ; Lapatinib/administration & dosage ; Mice ; Mice, Nude ; Oncogene Proteins, Fusion ; Receptor, ErbB-2/antagonists & inhibitors ; Receptor, ErbB-2/metabolism ; Signal Transduction ; Tamoxifen/administration & dosage ; Xenograft Model Antitumor Assays ; src-Family Kinases/antagonists & inhibitors ; src-Family Kinases/metabolism
    Chemical Substances CCDC170 protein, human ; Carrier Proteins ; ESR1 protein, human ; Estrogen Receptor alpha ; Oncogene Proteins, Fusion ; Tamoxifen (094ZI81Y45) ; Lapatinib (0VUA21238F) ; Fulvestrant (22X328QOC4) ; ERBB2 protein, human (EC 2.7.10.1) ; Receptor, ErbB-2 (EC 2.7.10.1) ; src-Family Kinases (EC 2.7.10.2) ; Dasatinib (RBZ1571X5H)
    Language English
    Publishing date 2020-08-08
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2015059-3
    ISSN 1465-542X ; 1465-5411
    ISSN (online) 1465-542X
    ISSN 1465-5411
    DOI 10.1186/s13058-020-01325-3
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Recurrent and pathological gene fusions in breast cancer: current advances in genomic discovery and clinical implications.

    Veeraraghavan, Jamunarani / Ma, Jiacheng / Hu, Yiheng / Wang, Xiao-Song

    Breast cancer research and treatment

    2016  Volume 158, Issue 2, Page(s) 219–232

    Abstract: Gene fusions have long been considered principally as the oncogenic events of hematologic malignancies, but have recently gained wide attention in solid tumors due to several milestone discoveries and the advancement of deep sequencing technologies. With ...

    Abstract Gene fusions have long been considered principally as the oncogenic events of hematologic malignancies, but have recently gained wide attention in solid tumors due to several milestone discoveries and the advancement of deep sequencing technologies. With the progress in deep sequencing studies of breast cancer transcriptomes and genomes, the discovery of recurrent and pathological gene fusions in breast cancer is on the focus. Recently, driven by new deep sequencing studies, several recurrent or pathological gene fusions have been identified in breast cancer, including ESR1-CCDC170, SEC16A-NOTCH1, SEC22B-NOTCH2, and ESR1-YAP1 etc. More important, most of these gene fusions are preferentially identified in the more aggressive breast cancers, such as luminal B, basal-like, or endocrine-resistant breast cancer, suggesting recurrent gene fusions as additional key driver events in these tumors other than the known drivers such as the estrogen receptor. In this paper, we have comprehensively summarized the newly identified recurrent or pathological gene fusion events in breast cancer, reviewed the contributions of new genomic and deep sequencing technologies to new fusion discovery and the integrative bioinformatics tools to analyze these data, highlighted the biological relevance and clinical implications of these fusion discoveries, and discussed future directions of gene fusion research in breast cancer.
    Language English
    Publishing date 2016-07
    Publishing country Netherlands
    Document type Journal Article ; Review
    ZDB-ID 604563-7
    ISSN 1573-7217 ; 0167-6806
    ISSN (online) 1573-7217
    ISSN 0167-6806
    DOI 10.1007/s10549-016-3876-y
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Towards personalized treatment for early stage HER2-positive breast cancer.

    Goutsouliak, Kristina / Veeraraghavan, Jamunarani / Sethunath, Vidyalakshmi / De Angelis, Carmine / Osborne, C Kent / Rimawi, Mothaffar F / Schiff, Rachel

    Nature reviews. Clinical oncology

    2019  Volume 17, Issue 4, Page(s) 233–250

    Abstract: Advances in HER2-targeted therapies have improved the survival of patients with HER2-positive breast cancer. The standard-of-care treatment for localized disease has been chemotherapy and 1 year of adjuvant HER2-targeted therapy, typically with the anti- ... ...

    Abstract Advances in HER2-targeted therapies have improved the survival of patients with HER2-positive breast cancer. The standard-of-care treatment for localized disease has been chemotherapy and 1 year of adjuvant HER2-targeted therapy, typically with the anti-HER2 antibody trastuzumab. Despite the effectiveness of this treatment, disease relapse occurs in a subset of patients; thus, focus has been placed on escalating treatment by either combining different HER2-targeted agents or extending the duration of HER2-targeted therapy. Indeed, dual HER2-targeted therapies and extended-duration anti-HER2 therapy, as well as adjuvant therapy with the anti-HER2 antibody-drug conjugate T-DM1, have all been approved for clinical use. Emerging evidence suggests, however, that some patients do not derive sufficient benefit from these additional therapies to offset the associated toxicities and/or costs. Similarly, the universal use of chemotherapy might not benefit all patients, and treatment de-escalation through omission of chemotherapy has shown promise in clinical trials and is currently being explored further. The future of precision medicine should therefore involve tailoring of therapy based on the genetics and biology of each tumour and the clinical characteristics of each patient. Predictive biomarkers that enable the identification of patients who will benefit from either escalated or de-escalated treatment will be crucial to this approach. In this Review, we summarize the available HER2-targeted agents and associated mechanisms of resistance, and describe the current therapeutic landscape of early stage HER2-positive breast cancer, focusing on strategies for treatment escalation or de-escalation.
    MeSH term(s) Antineoplastic Combined Chemotherapy Protocols/therapeutic use ; Breast Neoplasms/drug therapy ; Breast Neoplasms/metabolism ; Breast Neoplasms/pathology ; Chemotherapy, Adjuvant ; Female ; Humans ; Molecular Targeted Therapy/methods ; Neoplasm Staging ; Precision Medicine/methods ; Receptor, ErbB-2/antagonists & inhibitors ; Receptor, ErbB-2/metabolism ; Trastuzumab/administration & dosage ; Trastuzumab/therapeutic use
    Chemical Substances Receptor, ErbB-2 (EC 2.7.10.1) ; Trastuzumab (P188ANX8CK)
    Language English
    Publishing date 2019-12-13
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, Non-P.H.S. ; Review
    ZDB-ID 2491410-1
    ISSN 1759-4782 ; 1759-4774
    ISSN (online) 1759-4782
    ISSN 1759-4774
    DOI 10.1038/s41571-019-0299-9
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Amplification of TLK2 Induces Genomic Instability via Impairing the G2-M Checkpoint.

    Kim, Jin-Ah / Anurag, Meenakshi / Veeraraghavan, Jamunarani / Schiff, Rachel / Li, Kaiyi / Wang, Xiao-Song

    Molecular cancer research : MCR

    2016  Volume 14, Issue 10, Page(s) 920–927

    Abstract: Managing aggressive breast cancers with enhanced chromosomal instability (CIN) is a significant challenge in clinics. Previously, we described that a cell cycle-associated kinase called Tousled-like kinase 2 (TLK2) is frequently deregulated by genomic ... ...

    Abstract Managing aggressive breast cancers with enhanced chromosomal instability (CIN) is a significant challenge in clinics. Previously, we described that a cell cycle-associated kinase called Tousled-like kinase 2 (TLK2) is frequently deregulated by genomic amplifications in aggressive estrogen receptor-positive (ER
    Implications: Targeting TLK2 presents an attractive therapeutic strategy for the TLK2-amplified breast cancers that possess enhanced genomic instability and aggressiveness. Mol Cancer Res; 14(10); 920-7. ©2016 AACR.
    MeSH term(s) Breast Neoplasms/genetics ; Cell Line, Tumor ; DNA Damage ; Female ; G2 Phase Cell Cycle Checkpoints ; Gene Amplification ; Gene Expression Regulation, Neoplastic ; Genomic Instability ; Humans ; Protein Kinases/genetics ; Up-Regulation
    Chemical Substances Protein Kinases (EC 2.7.-) ; protein kinase U (EC 2.7.1.-)
    Language English
    Publishing date 2016-10
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2098788-2
    ISSN 1557-3125 ; 1541-7786
    ISSN (online) 1557-3125
    ISSN 1541-7786
    DOI 10.1158/1541-7786.MCR-16-0161
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Therapeutic Targeting of Nemo-like Kinase in Primary and Acquired Endocrine-resistant Breast Cancer.

    Wang, Xian / Veeraraghavan, Jamunarani / Liu, Chia-Chia / Cao, Xixi / Qin, Lanfang / Kim, Jin-Ah / Tan, Ying / Loo, Suet Kee / Hu, Yiheng / Lin, Ling / Lee, Sanghoon / Shea, Martin J / Mitchell, Tamika / Li, Shunqiang / Ellis, Matthew J / Hilsenbeck, Susan G / Schiff, Rachel / Wang, Xiao-Song

    Clinical cancer research : an official journal of the American Association for Cancer Research

    2021  Volume 27, Issue 9, Page(s) 2648–2662

    Abstract: Purpose: Endocrine resistance remains a major clinical challenge in estrogen receptor (ER)-positive breast cancer. Despite the encouraging results from clinical trials for the drugs targeting known survival signaling, relapse is still inevitable. There ... ...

    Abstract Purpose: Endocrine resistance remains a major clinical challenge in estrogen receptor (ER)-positive breast cancer. Despite the encouraging results from clinical trials for the drugs targeting known survival signaling, relapse is still inevitable. There is an unmet need to discover new drug targets in the unknown escape pathways. Here, we report Nemo-like kinase (NLK) as a new actionable kinase target that endows previously uncharacterized survival signaling in endocrine-resistant breast cancer.
    Experimental design: The effects of NLK inhibition on the viability of endocrine-resistant breast cancer cell lines were examined by MTS assay. The effect of VX-702 on NLK activity was verified by kinase assay. The modulation of ER and its coactivator, SRC-3, by NLK was examined by immunoprecipitation, kinase assay, luciferase assay, and RNA sequencing. The therapeutic effects of VX-702 and everolimus were tested on cell line- and patient-derived xenograft (PDX) tumor models.
    Results: NLK overexpression endows reduced endocrine responsiveness and is associated with worse outcome of patients treated with tamoxifen. Mechanistically, NLK may function, at least in part, via enhancing the phosphorylation of ERα and its key coactivator, SRC-3, to modulate ERα transcriptional activity. Through interrogation of a kinase profiling database, we uncovered and verified a highly selective dual p38/NLK inhibitor, VX-702. Coadministration of VX-702 with the mTOR inhibitor, everolimus, demonstrated a significant therapeutic effect in cell line-derived xenograft and PDX tumor models of acquired or
    Conclusions: Together, this study reveals the potential of therapeutic modulation of NLK for the management of the endocrine-resistant breast cancers with active NLK signaling.
    MeSH term(s) Animals ; Antineoplastic Agents, Hormonal/pharmacology ; Antineoplastic Agents, Hormonal/therapeutic use ; Biomarkers, Tumor ; Breast Neoplasms/diagnosis ; Breast Neoplasms/drug therapy ; Breast Neoplasms/etiology ; Breast Neoplasms/metabolism ; Cell Line, Tumor ; Cell Survival/drug effects ; Disease Models, Animal ; Dose-Response Relationship, Drug ; Drug Resistance, Neoplasm/drug effects ; Estrogen Receptor alpha/metabolism ; Female ; Gene Expression Profiling ; Gene Expression Regulation, Neoplastic ; Humans ; Mice ; Phosphorylation ; Prognosis ; Protein Kinase Inhibitors/pharmacology ; Protein Kinase Inhibitors/therapeutic use ; Protein Serine-Threonine Kinases/antagonists & inhibitors ; Protein Serine-Threonine Kinases/metabolism ; Signal Transduction/drug effects ; Xenograft Model Antitumor Assays
    Chemical Substances Antineoplastic Agents, Hormonal ; Biomarkers, Tumor ; Estrogen Receptor alpha ; Protein Kinase Inhibitors ; NLK protein, human (EC 2.7.1.-) ; Protein Serine-Threonine Kinases (EC 2.7.11.1)
    Language English
    Publishing date 2021-02-04
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, Non-P.H.S.
    ZDB-ID 1225457-5
    ISSN 1557-3265 ; 1078-0432
    ISSN (online) 1557-3265
    ISSN 1078-0432
    DOI 10.1158/1078-0432.CCR-20-2961
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Correction: Activation of the IFN Signaling Pathway is Associated with Resistance to CDK4/6 Inhibitors and Immune Checkpoint Activation in ER-Positive Breast Cancer.

    De Angelis, Carmine / Fu, Xiaoyong / Cataldo, Maria Letizia / Nardone, Agostina / Pereira, Resel / Veeraraghavan, Jamunarani / Nanda, Sarmistha / Qin, Lanfang / Sethunath, Vidyalakshmi / Wang, Tao / Hilsenbeck, Susan G / Benelli, Matteo / Migliaccio, Ilenia / Guarducci, Cristina / Malorni, Luca / Litchfield, Lacey M / Liu, Jiangang / Donaldson, Joshua / Selenica, Pier /
    Brown, David N / Weigelt, Britta / Reis-Filho, Jorge S / Park, Ben H / Hurvitz, Sara A / Slamon, Dennis J / Rimawi, Mothaffar F / Jansen, Valerie M / Jeselsohn, Rinath / Osborne, C Kent / Schiff, Rachel

    Clinical cancer research : an official journal of the American Association for Cancer Research

    2021  Volume 27, Issue 17, Page(s) 4939

    Language English
    Publishing date 2021-09-01
    Publishing country United States
    Document type Journal Article ; Published Erratum
    ZDB-ID 1225457-5
    ISSN 1557-3265 ; 1078-0432
    ISSN (online) 1557-3265
    ISSN 1078-0432
    DOI 10.1158/1078-0432.CCR-21-2431
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top