Multi-Targeted Kinase Inhibition Alleviates Mtor Inhibitor

Total Page:16

File Type:pdf, Size:1020Kb

Multi-Targeted Kinase Inhibition Alleviates Mtor Inhibitor Breast Cancer Research and Treatment https://doi.org/10.1007/s10549-019-05380-z PRECLINICAL STUDY Multi‑targeted kinase inhibition alleviates mTOR inhibitor resistance in triple‑negative breast cancer Jichao He1 · Ronan P. McLaughlin1 · Vera van der Noord1 · John A. Foekens2 · John W. M. Martens2 · Gerard van Westen1 · Yinghui Zhang1 · Bob van de Water1 Received: 21 February 2019 / Accepted: 27 July 2019 © The Author(s) 2019 Abstract Purpose Owing to its genetic heterogeneity and acquired resistance, triple-negative breast cancer (TNBC) is not responsive to single-targeted therapy, causing disproportional cancer-related death worldwide. Combined targeted therapy strategies to block interactive oncogenic signaling networks are being explored for efective treatment of the refractory TNBC subtype. Methods A broad kinase inhibitor screen was applied to profle the proliferative responses of TNBC cells, revealing resist- ance of TNBC cells to inhibition of the mammalian target of rapamycin (mTOR). A systematic drug combination screen was subsequently performed to identify that AEE788, an inhibitor targeting multiple receptor tyrosine kinases (RTKs) EGFR/ HER2 and VEGFR, synergizes with selective mTOR inhibitor rapamycin as well as its analogs (rapalogs) temsirolimus and everolimus to inhibit TNBC cell proliferation. Results The combination treatment with AEE788 and rapalog efectively inhibits phosphorylation of mTOR and 4EBP1, relieves mTOR inhibition-mediated upregulation of cyclin D1, and maintains suppression of AKT and ERK signaling, thereby sensitizing TNBC cells to the rapalogs. siRNA validation of cheminformatics-based predicted AEE788 targets has further revealed the mTOR interactive RPS6K members (RPS6KA3, RPS6KA6, RPS6KB1, and RPS6KL1) as synthetic lethal targets for rapalog combination treatment. Conclusions mTOR signaling is highly activated in TNBC tumors. As single rapalog treatment is insufcient to block mTOR signaling in rapalog-resistant TNBC cells, our results thus provide a potential multi-kinase inhibitor combinatorial strategy to overcome mTOR-targeted therapy resistance in TNBC cells. Keywords Multi-kinase inhibitor · mTOR-targeted therapy · Drug resistance · Triple-negative breast cancer (TNBC) · Polypharmacology Abbreviations FC Fold change BL Basal-like IC50 Half-maximal inhibitory concentration CDK4 Cyclin-dependent kinase 4 IM Immunomodulatory CI Combination index KI Kinase inhibitor EGF Human epidermal growth factor LAR Luminal androgen receptor-like MSL Mesenchymal stem-like mTOR The mammalian target of rapamycin Electronic supplementary material The online version of this article (https ://doi.org/10.1007/s1054 9-019-05380 -z) contains RTK Receptor tyrosine kinase supplementary material, which is available to authorized users. SRB Sulforhodamine B TNBC Triple-negative breast cancer * Bob van de Water [email protected] 1 Division of Drug Discovery and Safety, Leiden Background Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, The Netherlands Triple-negative breast cancer (TNBC) constitutes a small 2 Department of Medical Oncology and Cancer Genomic subtype (10–20%) of breast cancer, but causes the major- Netherlands, Erasmus MC Cancer Institute, Erasmus Medical ity of breast cancer-related deaths [1, 2]. As defned by the Centre, 3000 CA Rotterdam, The Netherlands Vol.:(0123456789)1 3 Breast Cancer Research and Treatment absence of ER and PR expression and HER2 overexpression, with mTOR signaling. Most importantly, our study provided TNBC is not curable by hormone receptor or HER2-targeted an efcacious approach for exploring cancer combination therapies [3]. Furthermore, TNBC is highly heterogeneous. treatment. Moreover, the combinatorial therapy is more Gene expression profling has further classifed TNBC into efective than single drug application and thus demonstrates six unique molecular subtypes, namely basal-like (BL1 and a therapeutic advantage over either agents as a monotherapy BL2), mesenchymal (M), mesenchymal stem-like (MSL), in TNBC treatment. immunomodulatory (IM), and luminal androgen receptor- like (LAR) subtype [4]. The TNBC molecular signatures have been explored for targeted therapies in clinical trials, Methods including those targeting receptor tyrosine kinases (RTKs, e.g., EGFR, VEGFR, c-Met), PI3K/AKT, Ras/MAPK, JAK/ Kinase inhibitor library combination screen STAT, cell cycle regulators [5, 6]. Yet, TNBC has not ben- efted from above mono-targeted therapies so far, due to One day post seeding into 96-well plates, cells were treated intrinsic or acquired resistance [6]. with individual kinase inhibitors alone or combined with The mammalian target of rapamycin (mTOR), a con- rapamycin at 1 µM. After 4-day treatment, proliferation was served serine/threonine protein kinase, is a central regulator evaluated by sulforhodamine B (SRB) colorimetric assay of cell growth and proliferation, by sensing and integrating [23]. Detailed information on materials and methods can be multiple signals from growth factors and nutrient signals found in Supplementary fle ESM_3. [7, 8]. mTOR hyperactivity is frequently observed in TNBC compared to other breast cancer subtypes and is often cor- related with poor prognosis, underpinning the potential Results of mTOR-targeted therapy for TNBC treatment [9–11]. Although mTOR-targeted interventions, such as rapamy- TNBC cell lines are diferentially responsive to mTOR cin and its analogs (rapalogs) temsirolimus and everoli- inhibitor rapalogs mus, delay progression and extend survival, patients with TNBC eventually develop resistance to mTOR inhibitors To gain insights into TNBC dependency on mTOR signaling with undesired outcome [9, 12]. Evidence has shown that integration for proliferation and cell survival, a KI library rapalog treatment could release mTOR negative feedback on (Selleckchem®) containing 378 small molecular inhibitors upstream kinases and activate compensatory pathways, for targeting various kinase signaling pathways was screened instance, PI3K/AKT and MAPK/ERK signaling pathways, across 19 TNBC cell lines (Suppl. Table S1), which are thereby bypassing mTOR inhibition [13–15]. This obser- representative for the six transcriptome-based subtypes of vation underscores the need for alternative combinatorial TNBC [4]. All TNBC cell lines were exposed to individual therapeutic approaches for TNBC treatment. inhibitors at 1 µM for 4 days, followed by measurement of Since oncogenic pathways incorporate multiple signaling cell proliferation. The efect of each inhibitor on prolifera- components and axes to promote tumor malignancy, mono- tion was assessed by Z scores normalized to overall pro- therapy may not be sufcient for long-term control of TNBC liferative response. TNBC cell lines were largely resistant [9, 13, 16]. Hence, simultaneously targeting diferent sign- to the majority of the kinase inhibitors, without any clear aling molecules represents a promising strategy to impede correlation to the TNBC molecular subtypes (Fig. 1a). The tumor growth and progression [8, 17]. Several reports have proliferative response towards mTOR inhibitors was vari- documented that co-targeting growth factor receptors and able among TNBC cell lines. We distinguished 11 TNBC mTOR exerts cooperative anti-cancer efects in various can- cell lines insensitive to diferent mTOR inhibitors (Fig. 1b), cer types, including TNBC [18–22]. However, these studies including rapamycin (Rap) and its analogs (i.e., rapalogs), focus on a particular combination in the questioned cancer zotarolimus, everolimus, ridaforolimus, and temsirolimus. type. Little is known about the interactive kinases involved HCC1806 and SUM149PT were most resistant to rapologs, in rapalog resistance and the mechanisms of the combinato- while Hs578T was most sensitive. rial efect remain unclear. Here, we systematically screened Rapalogs are highly selective allosteric inhibitors of a broad collection of kinase inhibitors across a large panel of mTOR, by binding to FKBP12/rapamycin-binding domain TNBC lines treated with rapamycin. Our data demonstrated to block mTOR Ser2448 phosphorylation and function [24, that multiple targeted kinase inhibition, for instance, by 25]. mTOR Ser2448 is a predominant phosphorylation inhibitor AEE788, sensitizes TNBC cells to various mTOR residue for mTOR kinase activity in response to mitogen- inhibitors, rapamycin, temsirolimus, and everolimus. Inte- derived stimuli [25]. Therefore, we examined the inhibi- grated cheminformatics study and siRNA validation revealed tory efect of rapamycin (Rap), temsirolimus (Tem), and additional putative targets of AEE788, which interact closely everolimus (Eve), on Ser2448-mTOR phosphorylation with 1 3 Breast Cancer Research and Treatment A TNBC Subtype LAR BL1 BL2 C Rap Tem Eve D Rap Tem Eve MSL M unclassified M M M M M M M M M M M M 1µ 1µ 1µ 1µ 1µ 1µ 0.1µ 0.1µ 0.1µ 0.1µ 0.1µ 0.1µ Pathway DMSO DMSO Angiogenesis 0.01µM 0.01µM 0.01µM 0.01µM 0.01µM 0.01µM Autophagy 1.5 Cell Cycle p-mTOR Cytoskeletal Signaling DNA Damage 1.0 Epigenetics mTOR GPCR & G Protein JAK/STAT 0.5 HCC1806 MAPK Tubulin NF-κB p-mTOR / mTOR 0.0 Others HCC1806 PI3K/AKT/mTOR 1.5 Protein Tyrosine Kinase p-mTOR TGF-β/Smad T 7 y 1.0 T T E E E Z score 49P mTOR 4 BT20 M1 thwa BT549 2 0.5 HCC38 SKBR HCC70 Hs578T SU Pa 0 Tubulin HCC1806 HCC1937 HCC1143 A-MB-468 A-MB-453 A-MB-231 A-MB-436 SUM52P −2 p-mTOR / mTOR SUM149P SUM159P SUM229P SUM185P −4 0.0 MD MD MD MD SUM149PT SUM1315MO2 1.5 p-mTOR B mTORi T 1.0 Zotarolimus mTOR Rapamycin Everolimus Hs578
Recommended publications
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Expressed Gene Fusions As Frequent Drivers of Poor Outcomes in Hormone Receptor–Positive Breast Cancer
    Published OnlineFirst December 14, 2017; DOI: 10.1158/2159-8290.CD-17-0535 RESEARCH ARTICLE Expressed Gene Fusions as Frequent Drivers of Poor Outcomes in Hormone Receptor–Positive Breast Cancer Karina J. Matissek1,2, Maristela L. Onozato3, Sheng Sun1,2, Zongli Zheng2,3,4, Andrew Schultz1, Jesse Lee3, Kristofer Patel1, Piiha-Lotta Jerevall2,3, Srinivas Vinod Saladi1,2, Allison Macleay3, Mehrad Tavallai1,2, Tanja Badovinac-Crnjevic5, Carlos Barrios6, Nuran Beşe7, Arlene Chan8, Yanin Chavarri-Guerra9, Marcio Debiasi6, Elif Demirdögen10, Ünal Egeli10, Sahsuvar Gökgöz10, Henry Gomez11, Pedro Liedke6, Ismet Tasdelen10, Sahsine Tolunay10, Gustavo Werutsky6, Jessica St. Louis1, Nora Horick12, Dianne M. Finkelstein2,12, Long Phi Le2,3, Aditya Bardia1,2, Paul E. Goss1,2, Dennis C. Sgroi2,3, A. John Iafrate2,3, and Leif W. Ellisen1,2 ABSTRACT We sought to uncover genetic drivers of hormone receptor–positive (HR+) breast cancer, using a targeted next-generation sequencing approach for detecting expressed gene rearrangements without prior knowledge of the fusion partners. We identified inter- genic fusions involving driver genes, including PIK3CA, AKT3, RAF1, and ESR1, in 14% (24/173) of unselected patients with advanced HR+ breast cancer. FISH confirmed the corresponding chromo- somal rearrangements in both primary and metastatic tumors. Expression of novel kinase fusions in nontransformed cells deregulates phosphoprotein signaling, cell proliferation, and survival in three- dimensional culture, whereas expression in HR+ breast cancer models modulates estrogen-dependent growth and confers hormonal therapy resistance in vitro and in vivo. Strikingly, shorter overall survival was observed in patients with rearrangement-positive versus rearrangement-negative tumors. Cor- respondingly, fusions were uncommon (<5%) among 300 patients presenting with primary HR+ breast cancer.
    [Show full text]
  • Expressed Gene Fusions As Frequent Drivers of Poor Outcomes in Hormone Receptor–Positive Breast Cancer
    Published OnlineFirst December 14, 2017; DOI: 10.1158/2159-8290.CD-17-0535 RESEARCH ARTICLE Expressed Gene Fusions as Frequent Drivers of Poor Outcomes in Hormone Receptor–Positive Breast Cancer Karina J. Matissek1,2, Maristela L. Onozato3, Sheng Sun1,2, Zongli Zheng2,3,4, Andrew Schultz1, Jesse Lee3, Kristofer Patel1, Piiha-Lotta Jerevall2,3, Srinivas Vinod Saladi1,2, Allison Macleay3, Mehrad Tavallai1,2, Tanja Badovinac-Crnjevic5, Carlos Barrios6, Nuran Beşe7, Arlene Chan8, Yanin Chavarri-Guerra9, Marcio Debiasi6, Elif Demirdögen10, Ünal Egeli10, Sahsuvar Gökgöz10, Henry Gomez11, Pedro Liedke6, Ismet Tasdelen10, Sahsine Tolunay10, Gustavo Werutsky6, Jessica St. Louis1, Nora Horick12, Dianne M. Finkelstein2,12, Long Phi Le2,3, Aditya Bardia1,2, Paul E. Goss1,2, Dennis C. Sgroi2,3, A. John Iafrate2,3, and Leif W. Ellisen1,2 ABSTRACT We sought to uncover genetic drivers of hormone receptor–positive (HR+) breast cancer, using a targeted next-generation sequencing approach for detecting expressed gene rearrangements without prior knowledge of the fusion partners. We identified inter- genic fusions involving driver genes, including PIK3CA, AKT3, RAF1, and ESR1, in 14% (24/173) of unselected patients with advanced HR+ breast cancer. FISH confirmed the corresponding chromo- somal rearrangements in both primary and metastatic tumors. Expression of novel kinase fusions in nontransformed cells deregulates phosphoprotein signaling, cell proliferation, and survival in three- dimensional culture, whereas expression in HR+ breast cancer models modulates estrogen-dependent growth and confers hormonal therapy resistance in vitro and in vivo. Strikingly, shorter overall survival was observed in patients with rearrangement-positive versus rearrangement-negative tumors. Cor- respondingly, fusions were uncommon (<5%) among 300 patients presenting with primary HR+ breast cancer.
    [Show full text]
  • LIM and Cysteine-Rich Domains 1 (LMCD1) Regulates Skeletal Muscle Hypertrophy, Calcium Handling, and Force Duarte M
    Ferreira et al. Skeletal Muscle (2019) 9:26 https://doi.org/10.1186/s13395-019-0214-1 RESEARCH Open Access LIM and cysteine-rich domains 1 (LMCD1) regulates skeletal muscle hypertrophy, calcium handling, and force Duarte M. S. Ferreira1, Arthur J. Cheng2,3, Leandro Z. Agudelo1,4, Igor Cervenka1, Thomas Chaillou2,5, Jorge C. Correia1, Margareta Porsmyr-Palmertz1, Manizheh Izadi1,6, Alicia Hansson1, Vicente Martínez-Redondo1, Paula Valente-Silva1, Amanda T. Pettersson-Klein1, Jennifer L. Estall7, Matthew M. Robinson8, K. Sreekumaran Nair8, Johanna T. Lanner2 and Jorge L. Ruas1* Abstract Background: Skeletal muscle mass and strength are crucial determinants of health. Muscle mass loss is associated with weakness, fatigue, and insulin resistance. In fact, it is predicted that controlling muscle atrophy can reduce morbidity and mortality associated with diseases such as cancer cachexia and sarcopenia. Methods: We analyzed gene expression data from muscle of mice or human patients with diverse muscle pathologies and identified LMCD1 as a gene strongly associated with skeletal muscle function. We transiently expressed or silenced LMCD1 in mouse gastrocnemius muscle or in mouse primary muscle cells and determined muscle/cell size, targeted gene expression, kinase activity with kinase arrays, protein immunoblotting, and protein synthesis levels. To evaluate force, calcium handling, and fatigue, we transduced the flexor digitorum brevis muscle with a LMCD1-expressing adenovirus and measured specific force and sarcoplasmic reticulum Ca2+ release in individual fibers. Finally, to explore the relationship between LMCD1 and calcineurin, we ectopically expressed Lmcd1 in the gastrocnemius muscle and treated those mice with cyclosporine A (calcineurin inhibitor). In addition, we used a luciferase reporter construct containing the myoregulin gene promoter to confirm the role of a LMCD1- calcineurin-myoregulin axis in skeletal muscle mass control and calcium handling.
    [Show full text]
  • Human Kinases Info Page
    Human Kinase Open Reading Frame Collecon Description: The Center for Cancer Systems Biology (Dana Farber Cancer Institute)- Broad Institute of Harvard and MIT Human Kinase ORF collection from Addgene consists of 559 distinct human kinases and kinase-related protein ORFs in pDONR-223 Gateway® Entry vectors. All clones are clonal isolates and have been end-read sequenced to confirm identity. Kinase ORFs were assembled from a number of sources; 56% were isolated as single cloned isolates from the ORFeome 5.1 collection (horfdb.dfci.harvard.edu); 31% were cloned from normal human tissue RNA (Ambion) by reverse transcription and subsequent PCR amplification adding Gateway® sequences; 11% were cloned into Entry vectors from templates provided by the Harvard Institute of Proteomics (HIP); 2% additional kinases were cloned into Entry vectors from templates obtained from collaborating laboratories. All ORFs are open (stop codons removed) except for 5 (MST1R, PTK7, JAK3, AXL, TIE1) which are closed (have stop codons). Detailed information can be found at: www.addgene.org/human_kinases Handling and Storage: Store glycerol stocks at -80oC and minimize freeze-thaw cycles. To access a plasmid, keep the plate on dry ice to prevent thawing. Using a sterile pipette tip, puncture the seal above an individual well and spread a portion of the glycerol stock onto an agar plate. To patch the hole, use sterile tape or a portion of a fresh aluminum seal. Note: These plasmid constructs are being distributed to non-profit institutions for the purpose of basic
    [Show full text]
  • Gene Symbol Accession Alias/Prev Symbol Official Full Name AAK1 NM 014911.2 KIAA1048, Dkfzp686k16132 AP2 Associated Kinase 1
    Gene Symbol Accession Alias/Prev Symbol Official Full Name AAK1 NM_014911.2 KIAA1048, DKFZp686K16132 AP2 associated kinase 1 (AAK1) AATK NM_001080395.2 AATYK, AATYK1, KIAA0641, LMR1, LMTK1, p35BP apoptosis-associated tyrosine kinase (AATK) ABL1 NM_007313.2 ABL, JTK7, c-ABL, p150 v-abl Abelson murine leukemia viral oncogene homolog 1 (ABL1) ABL2 NM_007314.3 ABLL, ARG v-abl Abelson murine leukemia viral oncogene homolog 2 (arg, Abelson-related gene) (ABL2) ACVR1 NM_001105.2 ACVRLK2, SKR1, ALK2, ACVR1A activin A receptor ACVR1B NM_004302.3 ACVRLK4, ALK4, SKR2, ActRIB activin A receptor, type IB (ACVR1B) ACVR1C NM_145259.2 ACVRLK7, ALK7 activin A receptor, type IC (ACVR1C) ACVR2A NM_001616.3 ACVR2, ACTRII activin A receptor ACVR2B NM_001106.2 ActR-IIB activin A receptor ACVRL1 NM_000020.1 ACVRLK1, ORW2, HHT2, ALK1, HHT activin A receptor type II-like 1 (ACVRL1) ADCK1 NM_020421.2 FLJ39600 aarF domain containing kinase 1 (ADCK1) ADCK2 NM_052853.3 MGC20727 aarF domain containing kinase 2 (ADCK2) ADCK3 NM_020247.3 CABC1, COQ8, SCAR9 chaperone, ABC1 activity of bc1 complex like (S. pombe) (CABC1) ADCK4 NM_024876.3 aarF domain containing kinase 4 (ADCK4) ADCK5 NM_174922.3 FLJ35454 aarF domain containing kinase 5 (ADCK5) ADRBK1 NM_001619.2 GRK2, BARK1 adrenergic, beta, receptor kinase 1 (ADRBK1) ADRBK2 NM_005160.2 GRK3, BARK2 adrenergic, beta, receptor kinase 2 (ADRBK2) AKT1 NM_001014431.1 RAC, PKB, PRKBA, AKT v-akt murine thymoma viral oncogene homolog 1 (AKT1) AKT2 NM_001626.2 v-akt murine thymoma viral oncogene homolog 2 (AKT2) AKT3 NM_181690.1
    [Show full text]
  • Koch Shrna Gene Webpage
    Symbol SEPT9 ADAM30 AEN AMBP ARHGEF12 ATG16L2 BCAS3 A1CF ADAM32 AFF3 AMBRA1 ARHGEF17 ATG2A BCKDK AAK1 ADAM33 AGAP2 AMHR2 ARHGEF2 ATG3 BCL10 AATK ADAM7 AGER AMPH ARHGEF4 ATG4B BCL11A ABCA1 ADAM8 AGK ANAPC2 ARHGEF6 ATG4C BCL11B ABCA3 ADAM9 AGL ANG ARHGEF7 ATG4D BCL2 ABCB1 ADAMDEC1 AGPAT9 ANGPT2 ARID1A ATG5 BCL2L1 ABCB4 ADAMTS1 AGR3 ANGPTL4 ARID1B ATG7 BCL2L11 ABCC1 ADAMTS10 AHR ANKK1 ARID2 ATM BCL2L2 ABCC10 ADAMTS12 AIMP2 ANKRD30A ARID3A ATMIN BCL3 ABCC2 ADAMTS13 AIP ANO1 ARID3B ATP1B3 BCL6 ABCG2 ADAMTS14 AJAP1 ANXA1 ARID4B ATP2B4 BCL7A ABI1 ADAMTS15 AK1 ANXA2 ARID5A ATP7A BCL9 ABL1 ADAMTS16 AK2 ANXA6 ARID5B ATP7B BCR ABL2 ADAMTS17 AK3 ANXA7 ARL11 ATR BECN1 ACIN1 ADAMTS18 AK4 APAF1 ARNT ATRX BFAR ACP1 ADAMTS19 AK5 APC ARSB ATXN1 BIK ACPP ADAMTS2 AK7 APCDD1 ARSG ATXN2 BIN1 ACSL4 ADAMTS20 AK8 APEX1 ASAP1 AURKA BIN2 ACTN1 ADAMTS3 AKAP1 APOBEC1 ASAP3 AURKB BIRC2 ACVR1 ADAMTS4 AKAP13 APOBEC2 ASB15 AURKC BIRC3 ACVR1B ADAMTS5 AKAP3 APOBEC3G ASCC1 AXIN1 BIRC5 ACVR1C ADAMTS7 AKAP8L AQP1 ASCC3 AXIN2 BIRC7 ACVR2A ADAMTS8 AKR1B10 AQP5 ASCL1 AXL BLCAP ACVR2B ADAMTS9 AKR1C1 AQP7 ASCL2 AZGP1 BLK ACVRL1 ADAR AKR1C3 AR ASF1A BACE1 BLM AD026 ADARB1 AKT1 ARAF ASH1L BAD BMI1 ADAM10 ADARB2 AKT2 AREG ASH2L BAG1 BMP2 ADAM11 ADAT2 AKT3 ARF1 ASNS BAG4 BMP2K ADAM12 ADCK1 ALCAM ARF4 ASPH BANF1 BMP2KL ADAM15 ADCK2 ALDH18A1 ARF5 ASPSCR1 BAP1 BMPR1A ADAM17 ADCK3 ALK ARF6 ASS1 BARD1 BMPR1B ADAM18 ADCK4 ALKBH2 ARHGAP12 ASTE1 BAX BMPR2 ADAM19 ADCK5 ALKBH3 ARHGAP22 ASXL1 BAZ1A BMX ADAM2 ADCY6 ALKBH8 ARHGAP25 ATF1 BAZ1B BNIP3 ADAM20 ADK ALOX15 ARHGAP26 ATF2 BAZ2A BPTF ADAM21
    [Show full text]
  • Multi-Targeted Kinase Inhibition Alleviates Mtor Inhibitor
    Breast Cancer Research and Treatment (2019) 178:263–274 https://doi.org/10.1007/s10549-019-05380-z PRECLINICAL STUDY Multi‑targeted kinase inhibition alleviates mTOR inhibitor resistance in triple‑negative breast cancer Jichao He1 · Ronan P. McLaughlin1 · Vera van der Noord1 · John A. Foekens2 · John W. M. Martens2 · Gerard van Westen1 · Yinghui Zhang1 · Bob van de Water1 Received: 21 February 2019 / Accepted: 27 July 2019 / Published online: 6 August 2019 © The Author(s) 2019 Abstract Purpose Owing to its genetic heterogeneity and acquired resistance, triple-negative breast cancer (TNBC) is not responsive to single-targeted therapy, causing disproportional cancer-related death worldwide. Combined targeted therapy strategies to block interactive oncogenic signaling networks are being explored for efective treatment of the refractory TNBC subtype. Methods A broad kinase inhibitor screen was applied to profle the proliferative responses of TNBC cells, revealing resist- ance of TNBC cells to inhibition of the mammalian target of rapamycin (mTOR). A systematic drug combination screen was subsequently performed to identify that AEE788, an inhibitor targeting multiple receptor tyrosine kinases (RTKs) EGFR/ HER2 and VEGFR, synergizes with selective mTOR inhibitor rapamycin as well as its analogs (rapalogs) temsirolimus and everolimus to inhibit TNBC cell proliferation. Results The combination treatment with AEE788 and rapalog efectively inhibits phosphorylation of mTOR and 4EBP1, relieves mTOR inhibition-mediated upregulation of cyclin D1, and maintains suppression of AKT and ERK signaling, thereby sensitizing TNBC cells to the rapalogs. siRNA validation of cheminformatics-based predicted AEE788 targets has further revealed the mTOR interactive RPS6K members (RPS6KA3, RPS6KA6, RPS6KB1, and RPS6KL1) as synthetic lethal targets for rapalog combination treatment.
    [Show full text]
  • Targeting CDK4 Overcomes EMT-Mediated Tumor Heterogeneity and Therapeutic Resistance in KRAS Mutant Lung Cancer
    Targeting CDK4 overcomes EMT-mediated tumor heterogeneity and therapeutic resistance in KRAS mutant lung cancer Aparna Padhye1,2, Jessica M. Konen1, B. Leticia Rodriguez1, Jared J. Fradette1, Joshua K. Ochieng1, Lixia Diao3, Jing Wang3, Wei Lu4, Luisa S. Solis4, Harsh Batra4, Maria G. Raso4, Michael D. Peoples5, Rosalba Minelli5, Alessandro Carugo5, Christopher A. Bristow5, Don L. Gibbons1,6* 1. Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. 2. University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA. 3. Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. 4. Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA 5. TRACTION Platform, Division of Therapeutics Development, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. 6. Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. *Corresponding author. Email: [email protected] Supplemental Methods Plasmids, Transfections, and Lentiviral Generation and Transduction Transfections of si-RNAs werr performed using the Lipofectamine 2000 Transfection Reagent (Thermo Fisher Scientific). Constitutive Cdkn1a overexpression cell lines were generated by using Cdkn1a mouse Tagged ORF Clone (Origene (NM_007669)). Cdkn1a ORF was also subcloned into dox-inducible pTRIPZ-GFP vector to generate doxycycline inducible cell lines using EcoRI and AgeI restriction cut sites. Constitutive Cdkn1a shRNAs were purchased from Milipore sigma. The sequences used in the experiments are listed in table S11. Dox- inducible shRNAs were expressed in Tet-pLKO-puro vector with a scramble sequence as the non-targeting control.
    [Show full text]
  • Autocrine IFN Signaling Inducing Profibrotic Fibroblast Responses By
    Downloaded from http://www.jimmunol.org/ by guest on September 23, 2021 Inducing is online at: average * The Journal of Immunology , 11 of which you can access for free at: 2013; 191:2956-2966; Prepublished online 16 from submission to initial decision 4 weeks from acceptance to publication August 2013; doi: 10.4049/jimmunol.1300376 http://www.jimmunol.org/content/191/6/2956 A Synthetic TLR3 Ligand Mitigates Profibrotic Fibroblast Responses by Autocrine IFN Signaling Feng Fang, Kohtaro Ooka, Xiaoyong Sun, Ruchi Shah, Swati Bhattacharyya, Jun Wei and John Varga J Immunol cites 49 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2013/08/20/jimmunol.130037 6.DC1 This article http://www.jimmunol.org/content/191/6/2956.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2013 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 23, 2021. The Journal of Immunology A Synthetic TLR3 Ligand Mitigates Profibrotic Fibroblast Responses by Inducing Autocrine IFN Signaling Feng Fang,* Kohtaro Ooka,* Xiaoyong Sun,† Ruchi Shah,* Swati Bhattacharyya,* Jun Wei,* and John Varga* Activation of TLR3 by exogenous microbial ligands or endogenous injury-associated ligands leads to production of type I IFN.
    [Show full text]
  • Mouse Rps6kc1 Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Rps6kc1 Knockout Project (CRISPR/Cas9) Objective: To create a Rps6kc1 knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Rps6kc1 gene (NCBI Reference Sequence: NM_178775 ; Ensembl: ENSMUSG00000089872 ) is located on Mouse chromosome 1. 15 exons are identified, with the ATG start codon in exon 1 and the TGA stop codon in exon 15 (Transcript: ENSMUST00000061611). Exon 2~3 will be selected as target site. Cas9 and gRNA will be co-injected into fertilized eggs for KO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Exon 2 starts from about 3.35% of the coding region. Exon 2~3 covers 4.96% of the coding region. The size of effective KO region: ~5668 bp. The KO region does not have any other known gene. Page 1 of 9 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele 5' gRNA region gRNA region 3' 1 2 3 15 Legends Exon of mouse Rps6kc1 Knockout region Page 2 of 9 https://www.alphaknockout.com Overview of the Dot Plot (up) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 2000 bp section upstream of Exon 2 is aligned with itself to determine if there are tandem repeats. No significant tandem repeat is found in the dot plot matrix. So this region is suitable for PCR screening or sequencing analysis. Overview of the Dot Plot (down) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 2000 bp section downstream of Exon 3 is aligned with itself to determine if there are tandem repeats.
    [Show full text]