AACR 2020 HMGA2 Poster V5.1

Total Page:16

File Type:pdf, Size:1020Kb

AACR 2020 HMGA2 Poster V5.1 Association between TGF-β signaling and HMGA2, a potential biomarker for Poster No. 1573 bintrafusp alfa in triple-negative breast cancer T.-L. Yeung, G. Locke, A. Lazorchak, G. Qin, H. Yu, J. Qi, B. Marelli, M. H. Jenkins, A. Rolfe, L. S. Ojalvo, I. Dussault, Y. Lan EMD Serono Research & Development Institute, Inc., Billerica, MA, USA; a business of Merck KGaA, Darmstadt, Germany • HMGA2 expression signicantly correlated with the INTRODUCTION Figure 3. Best change in target lesions from RESULTS CONCLUSIONS baseline assessed by investigator expression of TGF-β receptors, ligands, collagen, • Bintrafusp alfa is a rst-in-class bifunctional fusion 150 and EMT-related genes in bintrafusp alfa–treated BOR • HMGA2 expression signicantly correlated with TGF-β signaling–related genes⁴ • Patients with TNBC who experienced disease 130 PR SD mice (Figure 7) protein composed of the extracellular domain of the 110 PD in tumors treated with isotype control (29%) or bintrafusp alfa (55%) (Figure 5), control with bintrafusp alfa had high expression of TGF- RII receptor, to function as a TGF- “trap”, fused to 90 * Figure 7. Correlation between HMGA2 and TGF-β genes β β 70 indicating a strong association between HMGA2 and TGF-β signaling HMGA2 PD-L1 50 Tgfbr1 Core genes Tgfbr2 a human IgG1 antibody blocking PD-L1 (Figure 1) expression 6 5 30 0 Figure 5. Association between HMGA2, TGF-β signaling, and bintrafusp alfa • In a murine model, the correlation of HMGA2 and * ≥1 10 Figure 1. Proposed mechanism of action of bintrafusp alfa * * * * * ≥5 pharmacodynamic effects in a TNBC model 4 -10 ≥10 TGF-β signaling was stronger in bintrafusp alfa–treated 4 Tumor cells 20 -30 Isotype control Bintrafusp alfa 3 NK cell Mesenchymal-like tumors compared with isotype control–treated tumors tumor cell Change in sum of diameters, % -50 expression expression 2 TAM -70 29% correlated 55% correlated 2 Immune Hmga2 1.0 Hmga2 1.0 HMGA2 Smad3 HMGA2 Smad3 Tgfb1 Tgfb1 N=15 N=15 Dendritic Tgfb2 Tgfb2 • These data demonstrate a link between HMGA2 phenotype Tgfb3 Tgfb3 1 cell EMT (leading to metastasis and resistance Tgfbr1 Tgfbr1 Tgfbr1 r=0.846 Tgfbr2 r=0.911 TC PD-L1 expression Tgfbr2 Tgfbr2 Tumor angiogenesis Tgfbr3 Tgfbr3 to therapy [including checkpoint inhibition]) Fibroblast I Bmp2 Bmp2 Bmp3 Bmp3 p<0.001 p<0.001 Bmp4 Bmp4 TME PD-L1 expression E Excluded Bmp5 Bmp5 Bmp6 Bmp6 0 0 expression and TGF- ; bintrafusp alfa and Trap control CAF Bmp7 Bmp7 β D Desert Bmp10 Bmp10 Bmp15 Bmp15 Cytotoxic Immune phenotype E E E E E E D E E E ‡ D D E E E E D E E E D Bmpr1a Bmpr1a 2.53.0 3.54.0 4.55.0 5.5 2.53.0 3.54.0 4.55.0 5.5 † Bmpr1b Bmpr1b T cell Bmpr2 Bmpr2 Acvr1 Acvr1 Hmga2 expression Hmga2 expression Acvr1b Acvr1b Acvr1c Acvr1c reduced expression of HMGA2 and individual TGF- Acvr2a Acvr2a β Patients with a BOR of NE (n=5) are not included in this gure. Five additional patients were not Acvr2b 0.5 Acvr2b 0.5 Acvrl1 Acvrl1 Suppression of immune response TGF-β trap moiety sequesters Fibrosis and impaired Nodal Nodal Gdf1 Gdf1 Tgfb1 Tgfb2 TGF- to inhibit downstream signaling drug access Gdf11 Gdf11 β included because they lacked a valid postbaseline target lesion measurement. Inha Inha Inhba Inhba TGF- * Inhbb Inhbb signaling–related genes relative to anti–PD-L1 β Inhbc Inhbc 6 0.0 Inhbe Inhbe *Patients with HMGA2-high tumors who had an evaluable response. Smad2 Smad2 Smad1 Smad1 Smad5 Smad5 † Smad4 Smad4 T cell Smad6 Smad6 Sample had an indeterminate immune phenotype. Smad7 Smad7 -0.2 Bintrafusp Tumor cells Smad9 Smad9 Sptbn1 Sptbn1 ‡ Tgfbrap1 Tgfbrap1 • Collectively, these observations warrant further alfa Zfyve9 Zfyve9 PD-L1 expression and immune phenotype were not determinable. Aldh1a1 Aldh1a1 PD-1 PD-L1 Bcl2l11 Bcl2l11 4 Cdh1 Cdh1 Cdh2 0 Cdh2 0 -0.4 Cdkn1a Cdkn1a Cdkn2b Cdkn2b Fos Fos Jun Jun analysis of the potential link between bintrafusp alfa Myc Myc Anti–PD-L1 mAb moiety Col1a1 Col1a1 expression expression HMGA2 expression was 32-fold higher in patients who Col1a2 • Col1a2 blocks PD-L1 interactions with PD-1 Col3a1 Col3a1 -0.6 Dab2 Dab2 Dapk1 Dapk1 2 Fn1 Fn1 Foxk1 Foxk1 Foxk2 Foxk2 antitumor activity and HMGA2 Foxo1 N=15 Foxo3 Foxo1 N=15 experienced disease control with bintrafusp alfa Foxo4 Foxo3 Tgfb1 Tgfb2 -0.8 Gadd45b Foxo4 Id1 Gadd45b r= 0.590 r=0.696 Igf2 Id1 Il6 Igf2 Itgb6 Il6 Itgb8 Itgb6 p=0.023 p=0.005 Mapk14 -0.5 Itgb8 Mmp2 Mapk14 -0.5 0 -1.0 Mmp2 • Based on phase 1 trial results and biomarker compared with those who had PD (Figure 4) Mmp9 Pard6a Mmp9 Pja1 Pard6a Pja2 Pja1 2.53.0 3.54.0 4.55.0 5.5 2.53.0 3.54.0 4.55.0 5.5 CAF, cancer-associated broblast; EMT, epithelial-mesenchymal transition; NK, natural killer; Pja2 Runx1 Runx2 Runx1 Runx3 Runx2 Hmga2 expression Hmga2 expression Serpine1 Runx3 Smad7 Serpine1 TAM, tumor-associated macrophage. Smad7 analysis, a phase 2 study is planned to evaluate the Snai1 Figure 4. HMGA2 expression by investigator-assessed Snai2 Snai1 Stat3 Snai2 Tert Stat3 Twist1 Tert *Tumor cells are also a major source of TGF- in the tumor microenvironment (TME). Twist2 Twist1 β Twist2 Vegfa Vim Vegfa Tgfb3 Smad3 10 Zeb1 Vim BOR Zeb2 Zeb1 clinical activity of bintrafusp alfa in patients with † Zeb2 l 4.5 5 3 5 2 1 1 0 1 2 1 5 4 6 7 9 c 1 2 3 7 1 2 a 2 3 1 1 1 1 2 3 n f2 m Il6 a1 a2 e1 l b2 p2 p9 p2 p3 p4 p5 p6 p7 ai ai 3 l1 1 7 1 2 a 1 2 3 c 2 1 0 5 1 2 1 5 4 6 7 9 2 3 at 1 1 1 1 2 3 Id n gf • In a global, phase 1, open-label trial of bintrafusp alfa pr p1 pk p1 ad ad ad ad ad ad ad ad ga ad f2 m Fos Ju Fn1 Ig gb6 gb8 Vi Tert My l1a1 l1a2 l3a1 Il6 a1 a2 e1 in p2 p9 b2 p2 p3 p4 p5 p6 p7 Pj Pj Inha pr1a rd6a tbn1 9 pr1b l2 ai ai l1 kn1a Gdf1 at Id Zeb1 Zeb2 kn2b gf It It pr Da St Cdh1 Cdh2 pk ad p1 p1 ad ad ad ad ad ad ad ga ad Fn1 Ig Fos Ju gb6 gb8 Inhba Inhbc Inhbe Sn Sn Vi Inhbb Tgfb1 Tgfb2 Tgfb3 Ve Tert Acvr Noda Bm Bm Bm Bm Bm Bm My l1a1 l1a2 l3a1 Gdf1 in Foxk1 Foxk2 Mm Mm dh1a1 Pj Pj Inha Foxo1 Foxo3 Foxo4 rd6a pr1a tbn1 Da rp pr1b Acvrl1 Runx Runx Runx l2 Tgfbr Tgfbr Tgfbr Twist1 Twist2 kn1a Gdf1 Bm Zfyve9 Zeb1 Zeb2 kn2b It It Sm Sm Sm Sm Sm Sm Sm Co Co Co Sm Sm St Da Acvr1c Acvr2a Pa Bm Bm Hm Cdh1 Cdh2 Acvr1b Acvr2b Sp Bc Sn Sn Inhba Inhbc Inhbe Inhbb Tgfb1 Tgfb2 Tgfb3 Ve Cd Acvr Bm Bm Bm Bm Bm Bm Noda Cd Mapk14 Gdf1 Bm Foxk1 Foxk2 Mm Mm dh1a1 Bm Foxo1 Foxo3 Foxo4 Da rp Al Runx Runx Runx Acvrl1 Twist1 Twist2 Tgfbr Tgfbr Tgfbr Bm Zfyve9 Sm Co Co Co Sm Sm Sm Sm Sm Sm Sm Sm Pa Acvr1c Acvr2a Bm Bm Hm Se Acvr1b Acvr2b Sp Tgfbrap Gadd45b Bc Cd Mapk14 Cd Bm Bm Al Se Gadd45b Tgfbrap 4.0 4 HMGA2-high TNBC (NCT02517398), 33 patients with triple-negative breast 8 TGF-β core genes TGF-β target genes TGF-β core genes TGF-β target genes 7 3.5 3 cancer (TNBC), who received bintrafusp alfa 1200 mg 6 expression • Bintrafusp alfa or Trap control (TGF-β trap linked to an inactive anti–PD-L1) reduced expression (TPM+0.5) 2 3.0 2 5 every 2 weeks until conrmed progression, were log REFERENCES 4 expression of these HMGA2-correlated TGF-β signaling–related genes relative to Tgfb3 2.5 N=15 Smad3 1 N=15 r=0.850 r=0.943 analyzed for best overall response (BOR) 3 p<0.001 p<0.001 anti–PD-L1 or isotype control treatment (Figure 6), suggesting that HMGA2 is correlated 2.0 0 1. Lan Y, et al. Sci Transl Med. 2018;10:eaan5488. 2 2.53.0 3.54.0 4.55.0 5.5 2.53.0 3.54.0 4.55.0 5.5 NE PD* SD PR Hmga2 expression Hmga2 expression Response with TGF- –related signaling more strongly in the presence of TGF- sequestration – The investigator-assessed median duration of TPM, transcripts per million. β β 2. Spira A, et al. Cancer Res. 2020;80(Suppl 4):Abstract P3-09-06. response was 5.4 months (range, 4.7-5.5) in patients *Three samples from patients with a BOR of PD are not included in this gure due to lack of Figure 6. Effects of bintrafusp alfa on HMGA2 expression and TGF- signaling activity Target genes 3. Locke G, et al. Cancer Res. 2020;80(Suppl 4):Abstract P3-09-13. sample or sequencing failure. β Col1a2 Col1a1 4. Korkut A, et al. Cell Syst. 2018;7:422-37. with conrmed responses (Figure 2) †One sample from a patient with a BOR of PR was excluded from the formal biomarker Core genes Target genes 8 7.0 assessment due to failing quality control. Tgfbr1 Tgfbr2 Col1a1 Col1a2 6.5 5 p=0.434 1 8 8 p=0.002 6 – 4 patients had a BOR of partial response (PR) per p<0.001 6.0 • HMGA2 expression is upregulated by TGF-β signaling p=0.001 7 investigator assessment (3 conrmed, 1 unconrmed; 7 4 5.5 ACKNOWLEDGMENTS and is an important factor in mediating TGF-β–induced 4 6 expression expression 5.0 objective response rate,12.1%) (Figures 2 and 3) 0 5 6 2 N=15 N=15 EMT Col1a1 r=0.714 Col1a2 4.5 r=0.746 The authors thank the patients and their families, investigators, 3 4 p=0.004 p=0.002 – An additional patient had stable disease (SD), giving a 5 0 4.0 co-investigators, and the study teams at each of the participating • Using the same cutoff for HMGA2-high expression, 3 2.53.0 3.54.0 4.55.0 5.5 2.53.0 3.54.0 4.55.0 5.5 disease control rate of 15.2% Gene expression Gene expression Gene expression Gene expression Hmga2 expression Hmga2 expression centers and at Merck KGaA, Darmstadt, Germany, and EMD approximately 13.0% of tumor samples annotated as 2 -1 2 4 Vim Vegfa Serono Research & Development Institute, Inc., Billerica, MA, USA; • Tumor cell (TC) PD-L1 expression was not associated Hmga2 Tgfb1 Tgfb2 Vim Vegfa 10.5 6 TNBC in the Breast Invasive Carcinoma TCGA (The 6 6 p<0.001 1 11 6 p=0.475 p=0.004 p<0.001 p=0.036 a business of Merck KGaA with response to treatment (Figure 3) 10.0 Cancer Genome Atlas) data set had high expression of 5 4 4 9.5 This study was funded by Merck KGaA and is part of an alliance Figure 2.
Recommended publications
  • ACVR1 Antibody Cat
    ACVR1 Antibody Cat. No.: 4791 Western blot analysis of ACVR1 in A549 cell lysate with ACVR1 antibody at 1 μg/mL in (A) the absence and (B) the presence of blocking peptide. Specifications HOST SPECIES: Rabbit SPECIES REACTIVITY: Human, Mouse HOMOLOGY: Predicted species reactivity based on immunogen sequence: Bovine: (100%), Rat: (93%) ACVR1 antibody was raised against a 14 amino acid synthetic peptide near the amino terminus of the human ACVR1. IMMUNOGEN: The immunogen is located within the first 50 amino acids of ACVR1. TESTED APPLICATIONS: ELISA, WB ACVR1 antibody can be used for detection of ACVR1 by Western blot at 1 μg/mL. APPLICATIONS: Antibody validated: Western Blot in human samples. All other applications and species not yet tested. At least four isoforms of ACVR1 are known to exist. This antibody is predicted to have no SPECIFICITY: cross-reactivity to ACVR1B or ACVR1C. POSITIVE CONTROL: 1) Cat. No. 1203 - A549 Cell Lysate Properties October 1, 2021 1 https://www.prosci-inc.com/acvr1-antibody-4791.html PURIFICATION: ACVR1 Antibody is affinity chromatography purified via peptide column. CLONALITY: Polyclonal ISOTYPE: IgG CONJUGATE: Unconjugated PHYSICAL STATE: Liquid BUFFER: ACVR1 Antibody is supplied in PBS containing 0.02% sodium azide. CONCENTRATION: 1 mg/mL ACVR1 antibody can be stored at 4˚C for three months and -20˚C, stable for up to one STORAGE CONDITIONS: year. As with all antibodies care should be taken to avoid repeated freeze thaw cycles. Antibodies should not be exposed to prolonged high temperatures. Additional Info OFFICIAL SYMBOL: ACVR1 ACVR1 Antibody: FOP, ALK2, SKR1, TSRI, ACTRI, ACVR1A, ACVRLK2, Activin receptor type-1, ALTERNATE NAMES: Activin receptor type I, ACTR-I ACCESSION NO.: NP_001096 PROTEIN GI NO.: 4501895 GENE ID: 90 USER NOTE: Optimal dilutions for each application to be determined by the researcher.
    [Show full text]
  • Characterization of Pulmonary Arteriovenous Malformations in ACVRL1 Versus ENG Mutation Carriers in Hereditary Hemorrhagic Telangiectasia
    © American College of Medical Genetics and Genomics ORIGINAL RESEARCH ARTICLE Characterization of pulmonary arteriovenous malformations in ACVRL1 versus ENG mutation carriers in hereditary hemorrhagic telangiectasia Weiyi Mu, ScM1, Zachary A. Cordner, MD, PhD2, Kevin Yuqi Wang, MD3, Kate Reed, MPH, ScM4, Gina Robinson, RN5, Sally Mitchell, MD5 and Doris Lin, MD, PhD5 Purpose: Pulmonary arteriovenous malformations (pAVMs) are mutation carriers to have pAVMs (P o 0.001) or multiple lesions major contributors to morbidity and mortality in hereditary (P = 0.03), and to undergo procedural intervention (P = 0.02). hemorrhagic telangiectasia (HHT). Mutations in ENG and ACVRL1 Additionally, pAVMs in ENG carriers were more likely to exhibit underlie the vast majority of clinically diagnosed cases. The aims of bilateral lung involvement and growth over time, although this did this study were to characterize and compare the clinical and not reach statistical significance. The HHT severity score was morphologic features of pAVMs between these two genotype significantly higher in ENG than in ACVRL1 (P = 0.02). groups. Conclusion: The propensity and multiplicity of ENG-associated Methods: Sixty-six patients with HHT and affected family pAVMs may contribute to the higher disease severity in this members were included. Genotype, phenotypic data, and imaging genotype, as reflected by the HHT severity score and the frequency were obtained from medical records. Morphologic features of of interventional procedures. pAVMs were analyzed using computed tomography angiography. Genet Med HHT symptoms, pAVM imaging characteristics, frequency of advance online publication 19 October 2017 procedural intervention, and HHT severity scores were compared Key Words: ACVRL1; ENG; genotype-phenotype correlation; between ENG and ACVRL1 genotype groups.
    [Show full text]
  • Tumor Promoting Effect of BMP Signaling in Endometrial Cancer
    International Journal of Molecular Sciences Article Tumor Promoting Effect of BMP Signaling in Endometrial Cancer Tomohiko Fukuda 1,* , Risa Fukuda 1, Kohei Miyazono 1,2,† and Carl-Henrik Heldin 1,*,† 1 Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Box 582, Uppsala University, SE-751 23 Uppsala, Sweden; [email protected] (R.F.); [email protected] (K.M.) 2 Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan * Correspondence: [email protected] (T.F.); [email protected] (C.-H.H.); Tel.: +46-18-4714738 (T.F.); +46-18-4714738 (C.-H.H.) † These authors contributed equally to this work. Abstract: The effects of bone morphogenetic proteins (BMPs), members of the transforming growth factor-β (TGF-β) family, in endometrial cancer (EC) have yet to be determined. In this study, we analyzed the TCGA and MSK-IMPACT datasets and investigated the effects of BMP2 and of TWSG1, a BMP antagonist, on Ishikawa EC cells. Frequent ACVR1 mutations and high mRNA expressions of BMP ligands and receptors were observed in EC patients of the TCGA and MSK-IMPACT datasets. Ishikawa cells secreted higher amounts of BMP2 compared with ovarian cancer cell lines. Exogenous BMP2 stimulation enhanced EC cell sphere formation via c-KIT induction. BMP2 also induced EMT of EC cells, and promoted migration by induction of SLUG. The BMP receptor kinase inhibitor LDN193189 augmented the growth inhibitory effects of carboplatin. Analyses of mRNAs of several BMP antagonists revealed that TWSG1 mRNA was abundantly expressed in Ishikawa cells.
    [Show full text]
  • Saracatinib Is an Efficacious Clinical Candidate for Fibrodysplasia Ossificans Progressiva
    RESEARCH ARTICLE Saracatinib is an efficacious clinical candidate for fibrodysplasia ossificans progressiva Eleanor Williams,1 Jana Bagarova,2 Georgina Kerr,1 Dong-Dong Xia,2 Elsie S. Place,3 Devaveena Dey,2 Yue Shen,2 Geoffrey A. Bocobo,2 Agustin H. Mohedas,2 Xiuli Huang,4 Philip E. Sanderson,4 Arthur Lee,4 Wei Zheng,4 Aris N. Economides,5 James C. Smith,3 Paul B. Yu,2 and Alex N. Bullock1 1Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom. 2Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. 3Developmental Biology Laboratory, Francis Crick Institute, London, United Kingdom. 4National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA. 5Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA. Currently, no effective therapies exist for fibrodysplasia ossificans progressiva (FOP), a rare congenital syndrome in which heterotopic bone is formed in soft tissues owing to dysregulated activity of the bone morphogenetic protein (BMP) receptor kinase ALK2 (also known as ACVR1). From a screen of known biologically active compounds, we identified saracatinib as a potent ALK2 kinase inhibitor. In enzymatic and cell-based assays, saracatinib preferentially inhibited ALK2, compared with other receptors of the BMP/TGF-β signaling pathway, and induced dorsalization in zebrafish embryos consistent with BMP antagonism. We further tested the efficacy of saracatinib using an inducible ACVR1Q207D-transgenic mouse line, which provides a model of heterotopic ossification (HO), as well as an inducible ACVR1R206H-knockin mouse, which serves as a genetically and physiologically faithful FOP model. In both models, saracatinib was well tolerated and potently inhibited the development of HO, even when administered transiently following soft tissue injury.
    [Show full text]
  • Supplementary Table 1: Adhesion Genes Data Set
    Supplementary Table 1: Adhesion genes data set PROBE Entrez Gene ID Celera Gene ID Gene_Symbol Gene_Name 160832 1 hCG201364.3 A1BG alpha-1-B glycoprotein 223658 1 hCG201364.3 A1BG alpha-1-B glycoprotein 212988 102 hCG40040.3 ADAM10 ADAM metallopeptidase domain 10 133411 4185 hCG28232.2 ADAM11 ADAM metallopeptidase domain 11 110695 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 195222 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 165344 8751 hCG20021.3 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 189065 6868 null ADAM17 ADAM metallopeptidase domain 17 (tumor necrosis factor, alpha, converting enzyme) 108119 8728 hCG15398.4 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 117763 8748 hCG20675.3 ADAM20 ADAM metallopeptidase domain 20 126448 8747 hCG1785634.2 ADAM21 ADAM metallopeptidase domain 21 208981 8747 hCG1785634.2|hCG2042897 ADAM21 ADAM metallopeptidase domain 21 180903 53616 hCG17212.4 ADAM22 ADAM metallopeptidase domain 22 177272 8745 hCG1811623.1 ADAM23 ADAM metallopeptidase domain 23 102384 10863 hCG1818505.1 ADAM28 ADAM metallopeptidase domain 28 119968 11086 hCG1786734.2 ADAM29 ADAM metallopeptidase domain 29 205542 11085 hCG1997196.1 ADAM30 ADAM metallopeptidase domain 30 148417 80332 hCG39255.4 ADAM33 ADAM metallopeptidase domain 33 140492 8756 hCG1789002.2 ADAM7 ADAM metallopeptidase domain 7 122603 101 hCG1816947.1 ADAM8 ADAM metallopeptidase domain 8 183965 8754 hCG1996391 ADAM9 ADAM metallopeptidase domain 9 (meltrin gamma) 129974 27299 hCG15447.3 ADAMDEC1 ADAM-like,
    [Show full text]
  • ACVR1C Antibody Cat
    ACVR1C Antibody Cat. No.: 4795 ACVR1C Antibody Specifications HOST SPECIES: Rabbit SPECIES REACTIVITY: Human, Mouse, Rat ACVR1C antibody was raised against a 15 amino acid synthetic peptide near the amino terminus of the human ACVR1C. IMMUNOGEN: The immunogen is located within amino acids 130 - 180 of ACVR1C. TESTED APPLICATIONS: ELISA, WB ACVR1C antibody can be used for detection of ACVR1C by Western blot at 1 and 2 μg/mL. APPLICATIONS: Antibody validated: Western Blot in human samples. All other applications and species not yet tested. SPECIFICITY: This antibody is predicted to have no cross-reactivity to ACVR1 or ACVR1B. POSITIVE CONTROL: 1) Cat. No. 1309 - Human Placenta Tissue Lysate Properties PURIFICATION: ACVR1C Antibody is affinity chromatography purified via peptide column. CLONALITY: Polyclonal September 25, 2021 1 https://www.prosci-inc.com/acvr1c-antibody-4795.html ISOTYPE: IgG CONJUGATE: Unconjugated PHYSICAL STATE: Liquid BUFFER: ACVR1C Antibody is supplied in PBS containing 0.02% sodium azide. CONCENTRATION: 1 mg/mL ACVR1C antibody can be stored at 4˚C for three months and -20˚C, stable for up to one STORAGE CONDITIONS: year. As with all antibodies care should be taken to avoid repeated freeze thaw cycles. Antibodies should not be exposed to prolonged high temperatures. Additional Info OFFICIAL SYMBOL: ACVR1 ACVR1C Antibody: FOP, ALK2, SKR1, TSRI, ACTRI, ACVR1A, ACVRLK2, Activin receptor ALTERNATE NAMES: type-1, Activin receptor type I, ACTR-I ACCESSION NO.: Q8NER5 PROTEIN GI NO.: 4501895 GENE ID: 90 USER NOTE: Optimal dilutions for each application to be determined by the researcher. Background and References ACVR1C Antibody: Activins are dimeric growth and differentiation factors which belong to the transforming growth factor-beta (TGF-beta) superfamily of structurally related signaling proteins.
    [Show full text]
  • Human ALK-7 / ACVR1C Protein (ECD, Fc Tag)
    Human ALK-7 / ACVR1C Protein (ECD, Fc Tag) Catalog Number: 10869-H02H General Information SDS-PAGE: Gene Name Synonym: ACVRLK7; ALK7 Protein Construction: A DNA sequence encoding the human ACVR1C (NP_660302.2) (Met1- Glu113) was expressed with the Fc region of human IgG1 at the C- terminus. Source: Human Expression Host: HEK293 Cells QC Testing Purity: > 95 % as determined by SDS-PAGE. Endotoxin: Protein Description < 1.0 EU per μg protein as determined by the LAL method. ALK-7, also known as ALK7 and ACVR1C, belongs to the ALK family. It is a type I receptor for the TGFB family of signaling molecules. TGF-β is the Stability: prototype of a protein superfamily which, in humans, contains at least 35 members, including activins, inhibins, bone morphogenetic proteins, Samples are stable for up to twelve months from date of receipt at -70 ℃ growth/differentiation factors, and Müllerian inhibiting substance. ALK-7 is a serine-threonine kinase that can cause the activation of one of the SMAD Predicted N terminal: Leu 22 signal transducers, SMAD2. ALK-7 has a ligand known as Nodal. Nodal Molecular Mass: stimulates the secretion of TIMP-1 and inhibits matrix metalloproteinases MMP-2 and MMP-9 activity. The overexpression of Nodal or constitutively The recombinant human ACVR1C consists 330 amino acids and predicts active ALK-7 decreases cell migration and invasion, whereas knock-down a molecular mass of 36.6 kDa. of Nodal and ALK-7 has the opposite effects. Formulation: References Lyophilized from sterile PBS, pH 7.4. 1.Lin YY, et al. (2012) Functional dissection of lysine deacetylases reveals that HDAC1 and p300 regulate AMPK.
    [Show full text]
  • Molecular Classification of Patients with Unexplained Hamartomatous and Hyperplastic Polyposis
    ORIGINAL CONTRIBUTION Molecular Classification of Patients With Unexplained Hamartomatous and Hyperplastic Polyposis Kevin Sweet, MS, CGC Context Significant proportions of patients with hamartomatous polyposis or with Joseph Willis, MD hyperplastic/mixed polyposis remain without specific clinical and molecular diagnosis Xiao-Ping Zhou, MD, PhD or present atypically. Assigning a syndromic diagnosis is important because it guides management, especially surveillance and prophylactic surgery. Carol Gallione, PhD Objective To systematically classify patients with unexplained hamartomatous or hy- Takeshi Sawada, MD, PhD perplastic/mixed polyposis by extensive molecular analysis in the context of central Pia Alhopuro, MD rereview of histopathology results. Sok Kean Khoo, PhD Design, Setting, and Patients Prospective, referral-based study of 49 unrelated patients from outside institutions (n=28) and at a comprehensive cancer center (n=21), Attila Patocs, MD, PhD conducted from May 2, 2002, until December 15, 2004. Germline analysis of PTEN, Cossette Martin, PhD BMPR1A, STK11 (sequence, deletion), SMAD4, and ENG (sequence), specific exon screen- Scott Bridgeman, BSc ing of BRAF, MYH, and BHD, and rereview of polyp histology results were performed. John Heinz, PhD Main Outcome Measures Molecular, clinical, and histopathological findings in pa- tients with unexplained polyposis. Robert Pilarski, MS, CGC Results Of the 49 patients, 11 (22%) had germline mutations. Of 14 patients with Rainer Lehtonen, BSc juvenile polyposis, 2 with early-onset disease had mutations in ENG, encoding endo- Thomas W. Prior, PhD glin, previously only associated with hereditary hemorrhagic telangiectasia; 1 had hemi- zygous deletion encompassing PTEN and BMPR1A; and 1 had an SMAD4 mutation. Thierry Frebourg, MD, PhD One individual previously classified with Peutz-Jeghers syndrome had a PTEN dele- Bin Tean Teh, MD, PhD tion.
    [Show full text]
  • ACVR2B Recombinant Protein Cat
    ACVR2B Recombinant Protein Cat. No.: 96-009 ACVR2B Recombinant Protein Specifications SPECIES: Human SOURCE SPECIES: HEK293 cells SEQUENCE: Ser 19 - Thr 137 FUSION TAG: C-His Tag TESTED APPLICATIONS: WB APPLICATIONS: This recombinant protein can be used for WB. For research use only. PREDICTED MOLECULAR 14.5 kDa WEIGHT: Properties PURITY: >97% as determined by SDS-PAGE. PHYSICAL STATE: Lyophilized BUFFER: PBS, pH7.4 Lyophilized Protein should be stored at -20˚C or lower for long term storage. Upon STORAGE CONDITIONS: reconstitution, working aliquots should be stored at -20˚C or -70˚C. Avoid repeated freeze-thaw cycles. September 27, 2021 1 https://www.prosci-inc.com/acvr2b-recombinant-protein-96-009.html Additional Info OFFICIAL SYMBOL: ACVR2B ALTERNATE NAMES: ACVR2B, ACTRIIB, MGC116908 ACCESSION NO.: NP_001097 GENE ID: 93 Background and References Activin receptor type-2B (ACVR2B) is also known as ActR-IIB and MGC116908, ACVR2B is an activin type 2 receptor. Activins are dimeric growth and differentiation factors which belong to the transforming growth factor-beta (TGF-beta) superfamily of structurally related signaling proteins. Activins signal through a heteromeric complex of receptor serine kinases which include at least two type I (I and IB) and two type II (II and IIB) receptors. These receptors are all transmembrane proteins, composed of a ligand-binding extracellular domain with cysteine-rich region, a transmembrane domain, and a BACKGROUND: cytoplasmic domain with predicted serine/threonine specificity. Type I receptors are essential for signaling; and type II receptors are required for binding ligands and for expression of type I receptors. Type I and II receptors form a stable complex after ligand binding, resulting in phosphorylation of type I receptors by type II receptors.
    [Show full text]
  • A Novel Recombinant Anti-CD22 Immunokinase Delivers
    Published OnlineFirst January 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0685 Large Molecule Therapeutics Molecular Cancer Therapeutics A Novel Recombinant Anti-CD22 Immunokinase Delivers Proapoptotic Activity of Death- Associated Protein Kinase (DAPK) and Mediates Cytotoxicity in Neoplastic B Cells Nils Lilienthal1,2, Gregor Lohmann1, Giuliano Crispatzu1, Elena Vasyutina1, Stefan Zittrich3, Petra Mayer1, Carmen Diana Herling4, Mehmet Kemal Tur5, Michael Hallek4, Gabriele Pfitzer3, Stefan Barth6,7, and Marco Herling1,4 Abstract The serine/threonine death-associated protein kinases (DAPK) SGIII against the B-cell–exclusive endocytic glyco-receptor CD22 provide pro-death signals in response to (oncogenic) cellular stres- was created. Its high purity and large-scale recombinant production ses. Lost DAPK expression due to (epi)genetic silencing is found in a provided a stable, selectively binding, and efficiently internalizing broad spectrum of cancers. Within B-cell lymphomas, deficiency of construct with preserved robust catalytic activity. DK1KD-SGIII the prototypic family member DAPK1 represents a predisposing or specifically and efficiently killed CD22-positive cells of lymphoma early tumorigenic lesion and high-frequency promoter methylation lines and primary CLL samples, sparing healthy donor– or CLL marks more aggressive diseases. On the basis of protein studies and patient–derived non-B cells. The mode of cell death was predom- meta-analyzed gene expression profiling data, we show here that inantly PARP-mediated and caspase-dependent conventional apo- within the low-level context of B-lymphocytic DAPK, particularly ptosis as well as triggering of an autophagic program. The notori- CLL cells have lost DAPK1 expression. To target this potential ously high apoptotic threshold of CLL could be overcome by vulnerability, we conceptualized B-cell–specific cytotoxic reconsti- DK1KD-SGIII in vitro also in cases with poor prognostic features, tution of the DAPK1 tumor suppressor in the format of an immu- such as therapy resistance.
    [Show full text]
  • Bidirectional Crosstalk Between PD-L1 Expression and Epithelial To
    Alsuliman et al. Molecular Cancer (2015) 14:149 DOI 10.1186/s12943-015-0421-2 RESEARCH Open Access Bidirectional crosstalk between PD-L1 expression and epithelial to mesenchymal transition: Significance in claudin-low breast cancer cells Abdullah Alsuliman1, Dilek Colak2, Olfat Al-Harazi2, Hanaa Fitwi1, Asma Tulbah3, Taher Al-Tweigeri4, Monther Al-Alwan1,5 and Hazem Ghebeh1,5* Abstract Background: The T-cell inhibitory molecule PD-L1 (B7-H1, CD274) is expressed on tumor cells of a subset of breast cancer patients. However, the mechanism that regulates PD-L1 expression in this group of patients is still not well-identified. Methods: We have used loss and gain of function gene manipulation approach, multi-parametric flow cytometry, large scale gene expression dataset analysis and immunohistochemistry of breast cancer tissue sections. Results: Induction of epithelial to mesenchymal transition (EMT) in human mammary epithelial cells upregulated PD-L1 expression, which was dependent mainly on the activation of the PI3K/AKT pathway. Interestingly, gene expression signatures available from large cohort of breast tumors showed a significant correlation between EMT score and the PD-L1 mRNA level (p < 0.001). Strikingly, very strong association (p < 0.0001) was found between PD-L1 expression and claudin-low subset of breast cancer, which is known to have high EMT score. On the protein level, significant correlation was found between PD-L1 expression and standard markers of EMT (p =0.005)in67breast cancer patients. Importantly, specific downregulation of PD-L1 in claudin-low breast cancer cells showed signs of EMT reversal as manifested by CD44 and Vimentin downregulation and CD24 upregulation.
    [Show full text]
  • FOP) Can Be Rescued by the Drug Candidate Saracatinib
    Stem Cell Reviews and Reports https://doi.org/10.1007/s12015-020-10103-9 ActivinA Induced SMAD1/5 Signaling in an iPSC Derived EC Model of Fibrodysplasia Ossificans Progressiva (FOP) Can Be Rescued by the Drug Candidate Saracatinib Susanne Hildebrandt1,2,3 & Branka Kampfrath1 & Kristin Fischer3,4 & Laura Hildebrand2,3 & Julia Haupt1 & Harald Stachelscheid3,4 & Petra Knaus 1,2 Accepted: 1 December 2020 # The Author(s) 2021 Abstract Balanced signal transduction is crucial in tissue patterning, particularly in the vasculature. Heterotopic ossification (HO) is tightly linked to vascularization with increased vessel number in hereditary forms of HO, such as Fibrodysplasia ossificans progressiva (FOP). FOP is caused by mutations in the BMP type I receptor ACVR1 leading to aberrant SMAD1/5 signaling in response to ActivinA. Whether observed vascular phenotype in human FOP lesions is connected to aberrant ActivinA signaling is unknown. Blocking of ActivinA prevents HO in FOP mice indicating a central role of the ligand in FOP. Here, we established a new FOP endothelial cell model generated from induced pluripotent stem cells (iECs) to study ActivinA signaling. FOP iECs recapitulate pathogenic ActivinA/SMAD1/5 signaling. Whole transcriptome analysis identified ActivinA mediated activation of the BMP/ NOTCH pathway exclusively in FOP iECs, which was rescued to WT transcriptional levels by the drug candidate Saracatinib. We propose that ActivinA causes transcriptional pre-patterning of the FOP endothelium, which might contribute to differential vascularity in FOP lesions compared to non-hereditary HO. Keywords FOP . BMP-receptor . Activin . iPSCs . Human endothelial cells . HO . Saracatinib Introduction pathological conditions such as cancer and chronic inflamma- tion [2].
    [Show full text]