Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers After Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets

Total Page:16

File Type:pdf, Size:1020Kb

Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers After Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets Published OnlineFirst December 19, 2013; DOI: 10.1158/2159-8290.CD-13-0286 RESEARCH ARTICLE Molecular Profi ling of the Residual Disease of Triple-Negative Breast Cancers after Neoadjuvant Chemotherapy Identifi es Actionable Therapeutic Targets Justin M. Balko1 , 5 , Jennifer M. Giltnane2 , 5 , Kai Wang 8 , Luis J. Schwarz1 , 9 , 10 , Christian D. Young1 , Rebecca S. Cook3 , 5 , Phillip Owens 3 , Melinda E. Sanders 2 , 5 , Maria G. Kuba 2 , Violeta Sánchez 1 , Richard Kurupi 1 , Preston D. Moore 1 , Joseph A. Pinto 9 , Franco D. Doimi 9 , Henry Gómez 10 , Dai Horiuchi 6 , 7 , Andrei Goga 6 , 7 , Brian D. Lehmann 4 , Joshua A. Bauer 4 , Jennifer A. Pietenpol 4 , 5 , Jeffrey S. Ross 8 , Gary A. Palmer 8 , Roman Yelensky 8 , Maureen Cronin8 , Vincent A. Miller 8 , Phillip J. Stephens 8 , and Carlos L. Arteaga 1 , 3 , 5 Downloaded from cancerdiscovery.aacrjournals.org on September 29, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst December 19, 2013; DOI: 10.1158/2159-8290.CD-13-0286 ABSTRACT Neoadjuvant chemotherapy (NAC) induces a pathologic complete response (pCR) in approximately 30% of patients with triple-negative breast cancers (TNBC). In patients lacking a pCR, NAC selects a subpopulation of chemotherapy-resistant tumor cells. To under- stand the molecular underpinnings driving treatment-resistant TNBCs, we performed comprehensive molecular analyses on the residual disease of 74 clinically defi ned TNBCs after NAC, including next- generation sequencing (NGS) on 20 matched pretreatment biopsies. Combined NGS and digital RNA expression analysis identifi ed diverse molecular lesions and pathway activation in drug-resistant tumor cells. Ninety percent of the tumors contained a genetic alteration potentially treatable with a currently available targeted therapy. Thus, profi ling residual TNBCs after NAC identifi es targetable molecular lesions in the chemotherapy-resistant component of the tumor, which may mirror micro- metastases destined to recur clinically. These data can guide biomarker-driven adjuvant studies targeting these micrometastases to improve the outcome of patients with TNBC who do not respond completely to NAC. SIGNIFICANCE: This study demonstrates the spectrum of genomic alterations present in residual TNBC after NAC. Because TNBCs that do not achieve a CR after NAC are likely to recur as metastatic disease at variable times after surgery, these alterations may guide the selection of targeted thera- pies immediately after mastectomy before these metastases become evident. Cancer Discov; 4(2); 232–45. ©2013 AACR. INTRODUCTION have recently shown that these subtypes differ vastly in their post-NAC Ki67 scores, confounding the prognostic utility Neoadjuvant chemotherapy (NAC) is used increasingly in of Ki67 in this setting ( 10 ), and this has been confi rmed by patients with triple-negative breast cancer (TNBC), a subtype other investigators ( 9 ). Furthermore, Ki67 scoring is diffi cult lacking expression of estrogen receptor (ER), progesterone to standardize among clinical laboratories and many stud- receptor (PR), or HER2 amplifi cation. The goals of NAC are ies have defi ned different “cutoffs” for patient stratifi cation, to increase the likelihood of breast-conserving surgery and ranging from 14% to 50% ( 4–6 ). Finally, the Ki67 scoring of to eliminate clinically silent micrometastases. Approximately the post-NAC residual tumor is not actionable as it does not 30% of TNBC patients who receive NAC achieve a pathologic identify a pathogenic driver of the tumor and, as such, a drug complete response (pCR). These patients have a favorable target and rational treatment decision. recurrence-free survival (RFS) and overall survival (OS; refs. Intuition suggests that tumor cells remaining after NAC 1–3 ). The remaining patients with residual viable cancer in contain the cancer cell population intrinsically resistant to the breast or lymph nodes exhibit high rates of metastatic chemotherapy. These tumor cells likely mirror the micrometa- recurrence and an overall poor long-term outcome ( 1–3 ). static component of the disease that is ultimately responsible Immunohistochemistry (IHC) of the proliferation marker for distant metastases, and is unlikely to be highly sensitive Ki67 in post-NAC residual disease has been shown to corre- to further chemotherapy once clinical metastases become evi- late with patient outcome ( 4–6 ). Previous studies showing the dent. The standard of care for patients with TNBC who have prognostic ability of Ki67 after NAC included all subtypes of residual disease after NAC is observation, as therapies that breast cancer (i.e., HER2-enriched, luminal A, luminal B, and would be effective in reducing recurrences are unknown. Thus, basal-like), which also offer prognostic information ( 7–9 ). We we molecularly profi led the residual disease remaining after NAC in a cohort of 111 TNBCs [including gene expression analysis of 89 tumors and next-generation sequencing (NGS) Authors’ Affi liations: Departments of 1 Medicine, 2 Pathology, Microbi- ology & Immunology, 3 Cancer Biology, and 4 Biochemistry; 5 Breast Can- of 80 tumors, 74 of which were TNBC] to identify lesions that cer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt could be therapeutically targeted in adjuvant trials. University, Nashville, Tennessee; Departments of 6 Cell & Tissue Biology and 7 Medicine, University of California, San Francisco, San Francisco, California; 8 Foundation Medicine, Cambridge, Massachusetts ; 9 Oncosalud; RESULTS and 10 Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Perú Ki67 Does Not Predict Clinical Outcome in TNBCs Note: Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/). Because TNBC is a heterogeneous subtype of breast can- Corresponding Author: Carlos L. Arteaga, Vanderbilt University Medical cer ( 11 ), we determined whether Ki67 could predict patient Center, 2200 Pierce Ave, 777 PRB, Nashville, TN 37232-6307. Phone: 615- outcome within this clinical subtype by scoring Ki67 in the 936-3524; Fax: 615-936-1790; E-mail: [email protected] residual disease of a cohort of 111 TNBCs after NAC. Patient doi: 10.1158/2159-8290.CD-13-0286 demographics are listed in Supplementary Table S1. Molecu- © 2013 American Association for Cancer Research. lar subtyping based on gene expression using the PAM50 FEBRUARY 2014CANCER DISCOVERY | 233 Downloaded from cancerdiscovery.aacrjournals.org on September 29, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst December 19, 2013; DOI: 10.1158/2159-8290.CD-13-0286 RESEARCH ARTICLE Balko et al. centroids ( 7 ) revealed a predominance of tumors with basal- (trend, P = 0.0697), and JAK2 amplifi cations (trend, P = 0.08) like gene expression (Supplementary Fig. S1A). After adjust- in the residual disease. Amplifi cations in CDK6 and CCND1, ing for seven tumors that exhibited HER2 amplifi cation (see CCND2 , and CCND3 were collectively enriched as well (24% below), 70% of TNBCs were basal-like, which is similar to in post-NAC TNBC vs. 10% in TCGA basal-like tumors). This previously published rates in larger datasets ( 12 ). Basal-like difference suggests that these alterations are present at higher status was associated with a trend toward worse RFS and frequency in chemotherapy-treated TNBCs, and may play a OS (log-rank test, P = 0.12 and 0.058, respectively; Supple- role in de novo or acquired therapeutic resistance. However, it mentary Fig. S1B). As we have previously demonstrated ( 10 ), is important to note that these comparisons of CNAs with the the Ki67 score in the residual disease varied signifi cantly TCGA data are made between platforms [NGS vs. Affymetrix among molecular subtypes within this TNBC cohort, but single-nucleotide polymorphism (SNP) arrays], and thus some was not prognostic (Supplementary Fig. S1C and S1D). Ki67 variation in calling rates and detection of alterations may be staining decreased signifi cantly in response to chemotherapy platform-specifi c. ( P < 0.0001, paired t test; Supplementary Fig. S1E), but this Identifi ed alterations were categorized into several key path- change was not different among the molecular subtypes way or functional groups: cell-cycle alterations (amplifi cations (Supplementary Fig. S1F). Tumor cellularity was signifi cantly in CDK4 , CDK6 , CCND1, CCND2, CCND3 , CCNE1 , or AURKA decreased between the pre- and post-NAC samples (paired and loss of CDKN2A CDKN2B , , or RB1 ); phosphoinositide t test, P < 0.0001; Supplementary Fig. S1G). Node status at 3-kinase (PI3K)/mTOR alterations (amplifi cations of AKT1, surgery (an established prognostic marker), but not a change AKT2, AKT3 , PIK3CA , RAPTOR , or RICTOR ; loss or mutation in Ki67, was predictive of both RFS and OS, although this of PTEN ; truncations or nonsense mutations in TSC1 ; ampli- effect seemed to be confi ned only to postmenopausal women fi cations or mutations in PIK3CA or PIK3R1 ); growth factor (Supplementary Fig. S2). These data suggest that the underly- receptor (GFR) amplifi cations ( EGFR , MET , KIT , FGFR1, 2 , and ing molecular subtype may confound the prognostic ability 4 , or IGF1R); RAS/mitogen-activated protein kinase (MAPK) of Ki67 score in the residual disease after NAC. alterations (amplifi cations/gains of KRAS , BRAF , or RAF1 , or truncations of NF1 ); or DNA repair alterations (truncations, Genomic Alterations in Drug-Resistant loss or mutations of BRCA1 or BRCA2 , or mutations in ATM ; Residual
Recommended publications
  • Profiling Data
    Compound Name DiscoveRx Gene Symbol Entrez Gene Percent Compound Symbol Control Concentration (nM) JNK-IN-8 AAK1 AAK1 69 1000 JNK-IN-8 ABL1(E255K)-phosphorylated ABL1 100 1000 JNK-IN-8 ABL1(F317I)-nonphosphorylated ABL1 87 1000 JNK-IN-8 ABL1(F317I)-phosphorylated ABL1 100 1000 JNK-IN-8 ABL1(F317L)-nonphosphorylated ABL1 65 1000 JNK-IN-8 ABL1(F317L)-phosphorylated ABL1 61 1000 JNK-IN-8 ABL1(H396P)-nonphosphorylated ABL1 42 1000 JNK-IN-8 ABL1(H396P)-phosphorylated ABL1 60 1000 JNK-IN-8 ABL1(M351T)-phosphorylated ABL1 81 1000 JNK-IN-8 ABL1(Q252H)-nonphosphorylated ABL1 100 1000 JNK-IN-8 ABL1(Q252H)-phosphorylated ABL1 56 1000 JNK-IN-8 ABL1(T315I)-nonphosphorylated ABL1 100 1000 JNK-IN-8 ABL1(T315I)-phosphorylated ABL1 92 1000 JNK-IN-8 ABL1(Y253F)-phosphorylated ABL1 71 1000 JNK-IN-8 ABL1-nonphosphorylated ABL1 97 1000 JNK-IN-8 ABL1-phosphorylated ABL1 100 1000 JNK-IN-8 ABL2 ABL2 97 1000 JNK-IN-8 ACVR1 ACVR1 100 1000 JNK-IN-8 ACVR1B ACVR1B 88 1000 JNK-IN-8 ACVR2A ACVR2A 100 1000 JNK-IN-8 ACVR2B ACVR2B 100 1000 JNK-IN-8 ACVRL1 ACVRL1 96 1000 JNK-IN-8 ADCK3 CABC1 100 1000 JNK-IN-8 ADCK4 ADCK4 93 1000 JNK-IN-8 AKT1 AKT1 100 1000 JNK-IN-8 AKT2 AKT2 100 1000 JNK-IN-8 AKT3 AKT3 100 1000 JNK-IN-8 ALK ALK 85 1000 JNK-IN-8 AMPK-alpha1 PRKAA1 100 1000 JNK-IN-8 AMPK-alpha2 PRKAA2 84 1000 JNK-IN-8 ANKK1 ANKK1 75 1000 JNK-IN-8 ARK5 NUAK1 100 1000 JNK-IN-8 ASK1 MAP3K5 100 1000 JNK-IN-8 ASK2 MAP3K6 93 1000 JNK-IN-8 AURKA AURKA 100 1000 JNK-IN-8 AURKA AURKA 84 1000 JNK-IN-8 AURKB AURKB 83 1000 JNK-IN-8 AURKB AURKB 96 1000 JNK-IN-8 AURKC AURKC 95 1000 JNK-IN-8
    [Show full text]
  • Functional Analysis of Somatic Mutations Affecting Receptor Tyrosine Kinase Family in Metastatic Colorectal Cancer
    Author Manuscript Published OnlineFirst on March 29, 2019; DOI: 10.1158/1535-7163.MCT-18-0582 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Functional analysis of somatic mutations affecting receptor tyrosine kinase family in metastatic colorectal cancer Leslie Duplaquet1, Martin Figeac2, Frédéric Leprêtre2, Charline Frandemiche3,4, Céline Villenet2, Shéhérazade Sebda2, Nasrin Sarafan-Vasseur5, Mélanie Bénozène1, Audrey Vinchent1, Gautier Goormachtigh1, Laurence Wicquart6, Nathalie Rousseau3, Ludivine Beaussire5, Stéphanie Truant7, Pierre Michel8, Jean-Christophe Sabourin9, Françoise Galateau-Sallé10, Marie-Christine Copin1,6, Gérard Zalcman11, Yvan De Launoit1, Véronique Fafeur1 and David Tulasne1 1 Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T – Mechanisms of Tumorigenesis and Target Therapies, F-59000 Lille, France. 2 Univ. Lille, Plateau de génomique fonctionnelle et structurale, CHU Lille, F-59000 Lille, France 3 TCBN - Tumorothèque Caen Basse-Normandie, F-14000 Caen, France. 4 Réseau Régional de Cancérologie – OncoBasseNormandie – F14000 Caen – France. 5 Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, F-76000 Rouen, France. 6 Tumorothèque du C2RC de Lille, F-59037 Lille, France. 7 Department of Digestive Surgery and Transplantation, CHU Lille, Univ Lille, 2 Avenue Oscar Lambret, 59037, Lille Cedex, France. 8 Department of hepato-gastroenterology, Rouen University Hospital, Normandie Univ, UNIROUEN, Inserm U1245, IRON group, F-76000 Rouen, France. 9 Department of Pathology, Normandy University, INSERM 1245, Rouen University Hospital, F 76 000 Rouen, France. 10 Department of Pathology, MESOPATH-MESOBANK, Centre León Bérard, Lyon, France. 11 Thoracic Oncology Department, CIC1425/CLIP2 Paris-Nord, Hôpital Bichat-Claude Bernard, Paris, France.
    [Show full text]
  • Rabbit Anti-Ephrin-A1 Rabbit Anti-Ephrin-A1
    Qty: 100 µg/400 µl Rabbit anti-ephrin-A1 Catalog No. 34-3300 Lot No. See product label Rabbit anti-ephrin-A1 FORM This polyclonal antibody is supplied as a 400 µl aliquot at a concentration of 0.25 mg/ml in phosphate buffered saline (pH 7.4) containing 0.1% sodium azide. The antibody is epitope-affinity-purified from rabbit antiserum. PAD: ZMD.39 IMMUNOGEN Synthetic peptide derived from the C-terminal end of mouse ephrin-A1 protein. SPECIFICITY This antibody is specific for the ephrin-A1 protein. REACTIVITY Reactivity is confirmed with a chimeric protein consisting of the extracellular domain of mouse ephrin-A1 and the Fc region of human IgG1. Cross-reactivity with human ephrin-A1 is confirmed with IHC experiments on human tissue sections and this reactivity was expected because of the 85% shared amino acid identity in the extracellular domain. Sample Western Immunoprecipitation Immunohistochemistry Blotting (FFPE) Mouse +++ +++ NT Human NT NT ++ (Excellent +++, Good++, Poor +, No reactivity 0, Not tested NT) USAGE Working concentrations for specific applications should be determined by the investigator. Appropriate concentrations will be affected by several factors, including secondary antibody affinity, antigen concentration, sensitivity of detection method, temperature and length of incubations, etc. The suitability of this antibody for applications other than those listed below has not been determined. The following concentration ranges are recommended starting points for this product. Western Blotting: 1-5 µg/mL Immunoprecipitation: 5-10 µg/ IP reaction Immunohistochemistry: 4-10 µg/mL Please note that immunohistochemical assays were optimized on formalin-fixed, paraffin-embedded tissue sections and Heat Induced Epitope Retrieval (HIER) with EDTA, pH 8.0 was required for optimal staining.
    [Show full text]
  • EPH/Ephrin Profile and EPHB2 Expression Predicts Patient Survival in Breast Cancer
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 16 EPH/ephrin profile and EPHB2 expression predicts patient survival in breast cancer Anna-Maria Husa1,2, Željana Magić1, Malin Larsson3, Tommy Fornander4, Gizeh Pérez-Tenorio1 1Department of Clinical and Experimental Medicine, Division of Oncology, Linköping University, Linköping, Sweden 2Current address: CCRI, Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung e.V., Vienna, Austria 3Bioinformatics Infrastructure for Life Sciences (BILS) and Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden 4Department of Oncology, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden Correspondence to: Gizeh Pérez-Tenorio, e-mail: [email protected] Keywords: EPHB2, EPH family, TaqMan array, gene expression, protein expression Received: July 03, 2015 Accepted: January 23, 2016 Published: February 8, 2016 ABSTRACT The EPH and ephrins function as both receptor and ligands and the output on their complex signaling is currently investigated in cancer. Previous work shows that some EPH family members have clinical value in breast cancer, suggesting that this family could be a source of novel clinical targets. Here we quantified the mRNA expression levels of EPH receptors and their ligands, ephrins, in 65 node positive breast cancer samples by RT-PCR with TaqMan® Micro Fluidics Cards Microarray. Upon hierarchical clustering of the mRNA expression levels, we identified a subgroup of patients with high expression, and poor clinical outcome. EPHA2, EPHA4, EFNB1, EFNB2, EPHB2 and EPHB6 were significantly correlated with the cluster groups and particularly EPHB2 was an independent prognostic factor in multivariate analysis and in four public databases. The EPHB2 protein expression was also analyzed by immunohistochemistry in paraffin embedded material (cohort 2).
    [Show full text]
  • Eph Receptor Signalling: from Catalytic to Non-Catalytic Functions
    Oncogene (2019) 38:6567–6584 https://doi.org/10.1038/s41388-019-0931-2 REVIEW ARTICLE Eph receptor signalling: from catalytic to non-catalytic functions 1,2 1,2 3 1,2 1,2 Lung-Yu Liang ● Onisha Patel ● Peter W. Janes ● James M. Murphy ● Isabelle S. Lucet Received: 20 March 2019 / Revised: 23 July 2019 / Accepted: 24 July 2019 / Published online: 12 August 2019 © The Author(s) 2019. This article is published with open access Abstract Eph receptors, the largest subfamily of receptor tyrosine kinases, are linked with proliferative disease, such as cancer, as a result of their deregulated expression or mutation. Unlike other tyrosine kinases that have been clinically targeted, the development of therapeutics against Eph receptors remains at a relatively early stage. The major reason is the limited understanding on the Eph receptor regulatory mechanisms at a molecular level. The complexity in understanding Eph signalling in cells arises due to following reasons: (1) Eph receptors comprise 14 members, two of which are pseudokinases, EphA10 and EphB6, with relatively uncharacterised function; (2) activation of Eph receptors results in dimerisation, oligomerisation and formation of clustered signalling centres at the plasma membrane, which can comprise different combinations of Eph receptors, leading to diverse downstream signalling outputs; (3) the non-catalytic functions of Eph receptors have been overlooked. This review provides a structural perspective of the intricate molecular mechanisms that 1234567890();,: 1234567890();,: drive Eph receptor signalling, and investigates the contribution of intra- and inter-molecular interactions between Eph receptors intracellular domains and their major binding partners. We focus on the non-catalytic functions of Eph receptors with relevance to cancer, which are further substantiated by exploring the role of the two pseudokinase Eph receptors, EphA10 and EphB6.
    [Show full text]
  • Original Article a RTK-Based Functional Rnai Screen Reveals Determinants of PTX-3 Expression
    Int J Clin Exp Pathol 2013;6(4):660-668 www.ijcep.com /ISSN:1936-2625/IJCEP1301062 Original Article A RTK-based functional RNAi screen reveals determinants of PTX-3 expression Hua Liu*, Xin-Kai Qu*, Fang Yuan, Min Zhang, Wei-Yi Fang Department of Cardiology, Shanghai Chest Hospital affiliated to Shanghai JiaoTong University, Shanghai, China. *These authors contributed equally to this work. Received January 30, 2013; Accepted February 15, 2013; Epub March 15, 2013; Published April 1, 2013 Abstract: Aim: The aim of the present study was to explore the role of receptor tyrosine kinases (RTKs) in the regu- lation of expression of PTX-3, a protector in atherosclerosis. Methods: Human monocytic U937 cells were infected with a shRNA lentiviral vector library targeting human RTKs upon LPS stimuli and PTX-3 expression was determined by ELISA analysis. The involvement of downstream signaling in the regulation of PTX-3 expression was analyzed by both Western blotting and ELISA assay. Results: We found that knocking down of ERBB2/3, EPHA7, FGFR3 and RET impaired PTX-3 expression without effects on cell growth or viability. Moreover, inhibition of AKT, the downstream effector of ERBB2/3, also reduced PTX-3 expression. Furthermore, we showed that FGFR3 inhibition by anti-cancer drugs attenuated p38 activity, in turn induced a reduction of PTX-3 expression. Conclusion: Altogether, our study demonstrates the role of RTKs in the regulation of PTX-3 expression and uncovers a potential cardiotoxicity effect of RTK inhibitor treatments in cancer patients who have symptoms of atherosclerosis or are at the risk of athero- sclerosis.
    [Show full text]
  • Tyrosine Kinase Panel
    KinaseSeeker™ TK Panel (126 Wild-Type and Full-Length Kinases) Kinase Group Kinase Group ABL1 TK EPHA3 intracellular module TK ABL1 full-length TK EPHA4 TK ABL2 TK EPHA4 intracellular module TK ALK TK EPHA5 TK AXL TK EPHA5 intracellular module TK BLK TK EPHA6 TK BLK full-length TK EPHA6 intracellular module TK BMX TK EPHA7 TK BTK TK EPHA7 intracellular module TK BTK full-length TK EPHA8 TK CSF1R TK EPHA8 intracellular module TK CSF1R intracellular module TK EPHB2 TK CSK TK EPHB3 TK CSK full-length TK EPHB3 intracellular module TK DDR1 TK EPHB4 TK DDR1 intracellular module TK EPHB4 intracellular module TK DDR2 TK EPHB6 TK DDR2 intracellular module TK ERBB2 TK EGFR TK ERBB2 intracellular module TK EGFR intracellular module TK ERBB3 TK EPHA1 TK ERBB4 intracellular module TK EPHA1 intracellular module TK FAK TK EPHA2 TK FAK full-length TK EPHA2 intracellular module TK FER TK EPHA3 TK FES TK Legend: Full-Length: Construct contains Full-length kinase Intracellular Module: Construct contains Cytoplasmic Region in Receptor Tyrosine Kinases Page 1 of 3 KinaseSeeker™ TK Panel (126 Wild-Type and Full-Length Kinases) Kinase Group Kinase Group FGFR2 TK INSRR TK FGFR2 intracellular module TK ITK TK FGFR3 TK JAK2 TK FGFR3 intracellular module TK JAK3 TK FGFR4 TK KIT TK FGR TK KIT intracellular module TK FGR full-length TK LCK TK FLT1 TK LCK full-length TK FLT1 intracellular module TK LYN TK FLT2 TK LYN full-length TK FLT2 intracellular module TK MET TK FLT3 TK MET intracellular module TK FLT4 TK MST1R/RON TK FLT4 intracellular module TK MUSK TK FRK
    [Show full text]
  • Phospho-Ephb3 Ptyr608 Antibody
    Lot Number: RG2231302 Phospho-EphB3 pTyr608 Antibody Product Data Sheet Tested Species Reactivity Details Human (Hu) Catalog Number: PA5-37348 Mouse (Ms) Size: 100 µl Rat (Rt) Class: Polyclonal Type: Antibody Tested Applications Dilution * Clone: Western Blot (WB) 1:500-1:1000 Host / Isotype: Rabbit / IgG * Suggested working dilutions are given as a guide only. It is recommended that the user titrates the product for use in their own Synthetic phosphopeptide derived experiment using appropriate negative and positive controls. from human EphB3 around the Immunogen: phosphorylation site of Tyrosine 608 Form Information Form: Liquid Concentration: 1mg/ml Purification: Antigen affinity chromatography Storage Buffer: PBS with 50% glycerol Preservative: 0.1% sodium azide Store at 4°C short term. For long Storage Conditions: term storage, store at -20°C, avoiding freeze/thaw cycles. Product Specific Information General Information This antibody detects endogenous protein at a molecular weight of 110 kDa. The Eph subfamily represents the largest group of receptor protein tyrosine kinases identified to date. While the biological activities of these receptors Purity is >95% by SDS-PAGE. have yet to be determined, there is increasing evidence that they are involved For Research Use Only. Not for use in diagnostic procedures. Not for in central nervous system function and in development. The Eph subfamily resale without express authorization. receptors of human origin (and their murine/avian homologs) include EphA1 (Eph), EphA2 (Eck), EphA3 (Hek4), EphA4 (Hek8), EphA5 (Hek7), EphA6 (Hek12), EphA7 (Hek11/MDK1), EphA8 (Hek3), EphB1 (Hek6), EphB2 (Hek5), EphB3 (Cek10, Hek2), EphB4 (Htk), EphB5 (Hek9) and EphB6 (Mep). Ligands for Eph receptors include ephrin-A4 (LERK-4) which binds EphA3 and EphB1.
    [Show full text]
  • The Role of the Eph Receptor Family in Tumorigenesis. Cancers 2021
    cancers Review The Role of the Eph Receptor Family in Tumorigenesis Meg Anderton 1,2 , Emma van der Meulen 1,2 , Melissa J. Blumenthal 1,2,3,* and Georgia Schäfer 1,2,3,* 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) Cape Town, Observatory, Cape Town 7925, South Africa; [email protected] (M.A.); [email protected] (E.v.d.M.) 2 Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa 3 Division of Medical Biochemistry and Structural Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa * Correspondence: [email protected] (M.J.B.); [email protected] (G.S.); Tel.: +27-21-4047630 (M.J.B.) Simple Summary: The Eph receptor family is implicated in both tumour promotion and suppression, depending on the tissue-specific context of available receptor interactions with ligands, adaptor proteins and triggered downstream signalling pathways. This complex interplay has not only consequences for tumorigenesis but also offers a basis from which new cancer-targeting strategies can be developed. This review comprehensively summarises the current knowledge of Eph receptor implications in oncogenesis in a tissue- and receptor-specific manner, with the aim to develop a better understanding of Eph signalling pathways for potential targeting in novel cancer therapies. Abstract: The Eph receptor tyrosine kinase family, activated by binding to their cognate ephrin ligands, are important components of signalling pathways involved in animal development. More recently, they have received significant interest due to their involvement in oncogenesis.
    [Show full text]
  • Evolution of Mutational Landscape and Tumor Immune-Microenvironment in Liver Oligo-Metastatic Colorectal Cancer
    Article Evolution of Mutational Landscape and Tumor Immune-Microenvironment in Liver Oligo-Metastatic Colorectal Cancer Alessandro Ottaiano, Michele Caraglia, Annabella Di Mauro, Gerardo Botti, Angela Lombardi, Jerome Galon, Amalia Luce, Luigi D’Amore, Francesco Perri, Mariachiara Santorsola, Fabienne Hermitte, Giovanni Savarese, Fabiana Tatangelo, Vincenza Granata, Francesco Izzo, Andrea Belli, Stefania Scala, Paolo Delrio, Luisa Circelli and Guglielmo Nasti Supplementary Materials: Figure S1. Inclusion and exclusion criteria flow-chart for patients’ selection. Figure S2. Clinical courses of group A and group B patients. Figure S3. Representative model showing divergent mutational and immunologic dynamics between oligo- and poly-metastatic colorectal cancer. Table S1. List of genes mutations gained by liver metastases (all coding variants). AMP/ACMG Patient Mutated genes Role ClinVar ID prioritization Group A PAT1 PRKDC p.Gln281His8:g.48855892C>A DNA repair and recombination Tier 4 NR ACVR1B 12:g.52374752G>T (SAV) Proliferation, differentiation Tier 4 NR ALK p.Ala96Thr2:g.30143240C>T Proliferation, differentiation Tier 3 451139 CREBBP p.Pro1983Thr16:g.3779101G>T Transcriptional coactivation Tier 3 NR DNMT3B p.Gly76Arg20:g.31372585G>A Epigenetic modifications Tier 3 NR FAS p.Cys135ValfsTer5210:g.90768707CT>C Programmed cell death Tier 3 NR FGF2 p.Asp179ArgfsTer54:g.123748460A>AC Proliferation, angiogenesis Tier 4 NR PAT2 IFNGR1 p.Pro431Ser6:g.137519347G>A Immune response Tier 4 NR IRF2 p.Gly270Cys4:g.185310154C>A Transcription factor
    [Show full text]
  • AXL-Driven EMT State As a Targetable Conduit in Cancer Jane Antony1,2,3 and Ruby Yun-Ju Huang1,4,5
    Published OnlineFirst June 30, 2017; DOI: 10.1158/0008-5472.CAN-17-0392 Cancer Review Research AXL-Driven EMT State as a Targetable Conduit in Cancer Jane Antony1,2,3 and Ruby Yun-Ju Huang1,4,5 Abstract The receptor tyrosine kinase (RTK) AXL has been intrinsically further elucidation to define targetable conduits. Therapeuti- linked to epithelial–mesenchymal transition (EMT) and pro- cally, as AXL inhibition has shown EMT reversal and resensi- moting cell survival, anoikis resistance, invasion, and metas- tization to other tyrosine kinase inhibitors, mitotic inhibitors, tasis in several cancers. AXL signaling has been shown to and platinum-based therapy, there is a need to stratify patients directly affect the mesenchymal state and confer it with aggres- based on AXL dependence. This review elucidates the role of sive phenotype and drug resistance. Recently, the EMT gradient AXL in EMT-mediated oncogenesis and highlights the recipro- has also been shown to rewire the kinase signaling nodes that cal control between AXL signaling and the EMT state. In facilitate AXL–RTK cross-talk, protracted signaling, converging addition, we review the potential in inhibiting AXL for the on ERK, and PI3K axes. The molecular mechanisms under- development of different therapeutic strategies and inhibitors. playing the regulation between the kinome and EMT require Cancer Res; 77(14); 1–8. Ó2017 AACR. Introduction was originally identified in 1991 as a transforming gene in chronic myeloid leukemia (CML; ref. 3). Under normal phys- AXL, which stems from the Greek word for uncontrolled, iologic conditions, it is ubiquitously expressed in several tissues "anexelekto," is a receptor tyrosine kinase (RTK) belonging to and organs, including but not limited to the hippocampus, the tumor-associated macrophage (TAM) family, comprising of cerebellum, macrophages, platelets, endothelial cells, heart, TYRO-3, AXL, and MER (Fig.
    [Show full text]
  • Sequenced Genes
    UvA-DARE (Digital Academic Repository) Mutational profiling of glioblastoma Bleeker, F.E. Publication date 2009 Document Version Final published version Link to publication Citation for published version (APA): Bleeker, F. E. (2009). Mutational profiling of glioblastoma. General rights It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons). Disclaimer/Complaints regulations If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible. UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl) Download date:28 Sep 2021 Sequenced genes Gene Gene Name Cytoband Ch ABCA1 ATP-binding cassette, sub-family A (ABC1), member 1 9q31.1 4 ADAM12 A disintegrin and metalloproteinase domain 12 10q26.3 5 ADAMTS18 A disintegrin and metalloproteinase with thrombospondin 16q23 5 motifs 18 ADAMTSL3 ADAMTS-like 3 15q25.2 4 AIM1 Absent in melanoma 1 protein 5p13.3 5 AKT1 V-akt murine
    [Show full text]