1
|
Pu X, Qi L, Yan JW, Ai Z, Wu P, Yang F, Fu Y, Li X, Zhang M, Sun B, Yue S, Chen J. Oncogenic activation revealed by FGFR2 genetic alterations in intrahepatic cholangiocarcinomas. Cell Biosci 2023; 13:208. [PMID: 37964396 PMCID: PMC10644541 DOI: 10.1186/s13578-023-01156-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Except for gene fusions, FGFR2 genetic alterations in intrahepatic cholangiocarcinomas (ICCs) have received limited attention, leaving patients harboring activating FGFR2 gene mutations with inadequate access to targeted therapies. EXPERIMENTAL DESIGN We sought to survey FGFR2 genetic alterations in ICC and pan-cancers using fluorescence in situ hybridization and next-generation sequencing. We conducted an analysis of the clinical and pathological features of ICCs with different FGFR2 alterations, compared FGFR2 lesion spectrum through public databases and multicenter data, and performed cellular experiments to investigate the oncogenic potential of different FGFR2 mutants. RESULTS FGFR2 gene fusions were identified in 30 out of 474 ICC samples, while five FGFR2 genetic alterations aside from fusion were present in 290 ICCs. The tumors containing FGFR2 translocations exhibited unique features, which we designated as the "FGFR2 fusion subtypes of ICC". Molecular analysis revealed that FGFR2 fusions were not mutually exclusive with other oncogenic driver genes/mutations, whereas FGFR2 in-frame deletions and site mutations often co-occurred with TP53 mutations. Multicenter and pan-cancer studies demonstrated that FGFR2 in-frame deletions were more prevalent in ICCs (0.62%) than in other cancers, and were not limited to the extracellular domain. We selected representative FGFR2 genetic alterations, including in-frame deletions, point mutations, and frameshift mutations, to analyze their oncogenic activity and responsiveness to targeted drugs. Cellular experiments revealed that different FGFR2 genetic alterations promoted ICC tumor growth, invasion, and metastasis but responded differently to FGFR-selective small molecule kinase inhibitors (SMKIs). CONCLUSIONS FGFR2 oncogenic alterations have different clinicopathological features and respond differently to SMKIs.
Collapse
Affiliation(s)
- Xiaohong Pu
- Department of Pathology, Drum Tower Hospital, Affiliated Hospital of Medical School,Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Liang Qi
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
| | - Jia Wu Yan
- Department of Hepatobiliary Surgery, Drum Tower Hospital, Affiliated Hospital of Medical School,Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Zihe Ai
- Department of Medical Genetics, Nanjing Medical University, Nanjing, 210008, Jiangsu, China
| | - Ping Wu
- Department of Medical Genetics, Nanjing Medical University, Nanjing, 210008, Jiangsu, China
| | - Fei Yang
- Department of Hepatobiliary Surgery, Drum Tower Hospital, Affiliated Hospital of Medical School,Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Yao Fu
- Department of Pathology, Drum Tower Hospital, Affiliated Hospital of Medical School,Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Xing Li
- Shanghai Origimed Limited Company, Shanghai, 20000, China
| | - Min Zhang
- Beijing Gene Plus Limited Company, Beijing, 10000, China
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, Drum Tower Hospital, Affiliated Hospital of Medical School,Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Shen Yue
- Department of Medical Genetics, Nanjing Medical University, Nanjing, 210008, Jiangsu, China.
| | - Jun Chen
- Department of Pathology, Drum Tower Hospital, Affiliated Hospital of Medical School,Nanjing University, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
2
|
Attalla SS, Boucher J, Proud H, Taifour T, Zuo D, Sanguin-Gendreau V, Ling C, Johnson G, Li V, Luo RB, Kuasne H, Papavasiliou V, Walsh LA, Barok M, Joensuu H, Park M, Roux PP, Muller WJ. HER2Δ16 Engages ENPP1 to Promote an Immune-Cold Microenvironment in Breast Cancer. Cancer Immunol Res 2023; 11:1184-1202. [PMID: 37311021 DOI: 10.1158/2326-6066.cir-22-0140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/07/2023] [Accepted: 06/09/2023] [Indexed: 06/15/2023]
Abstract
The tumor-immune microenvironment (TIME) is a critical determinant of therapeutic response. However, the mechanisms regulating its modulation are not fully understood. HER2Δ16, an oncogenic splice variant of the HER2, has been implicated in breast cancer and other tumor types as a driver of tumorigenesis and metastasis. Nevertheless, the underlying mechanisms of HER2Δ16-mediated oncogenicity remain poorly understood. Here, we show that HER2∆16 expression is not exclusive to the clinically HER2+ subtype and associates with a poor clinical outcome in breast cancer. To understand how HER2 variants modulated the tumor microenvironment, we generated transgenic mouse models expressing either proto-oncogenic HER2 or HER2Δ16 in the mammary epithelium. We found that HER2∆16 tumors were immune cold, characterized by low immune infiltrate and an altered cytokine profile. Using an epithelial cell surface proteomic approach, we identified ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) as a functional regulator of the immune cold microenvironment. We generated a knock-in model of HER2Δ16 under the endogenous promoter to understand the role of Enpp1 in aggressive HER2+ breast cancer. Knockdown of Enpp1 in HER2Δ16-derived tumor cells resulted in decreased tumor growth, which correlated with increased T-cell infiltration. These findings suggest that HER2Δ16-dependent Enpp1 activation associates with aggressive HER2+ breast cancer through its immune modulatory function. Our study provides a better understanding of the mechanisms underlying HER2Δ16-mediated oncogenicity and highlights ENPP1 as a potential therapeutic target in aggressive HER2+ breast cancer.
Collapse
Affiliation(s)
- Sherif Samer Attalla
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Jonathan Boucher
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
| | - Hailey Proud
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Tarek Taifour
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Department of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Dongmei Zuo
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Virginie Sanguin-Gendreau
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Chen Ling
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Gabriella Johnson
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Vincent Li
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Robin B Luo
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Hellen Kuasne
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Vasilios Papavasiliou
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Logan A Walsh
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Mark Barok
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Heikki Joensuu
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Morag Park
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Department of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Philippe P Roux
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
- Department of Pathology and Cell Biology, Université de Montréal, Montreal, Canada
| | - William J Muller
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Department of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| |
Collapse
|
3
|
Kamashev D, Shaban N, Lebedev T, Prassolov V, Suntsova M, Raevskiy M, Gaifullin N, Sekacheva M, Garazha A, Poddubskaya E, Sorokin M, Buzdin A. Human Blood Serum Can Diminish EGFR-Targeted Inhibition of Squamous Carcinoma Cell Growth through Reactivation of MAPK and EGFR Pathways. Cells 2023; 12:2022. [PMID: 37626832 PMCID: PMC10453612 DOI: 10.3390/cells12162022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Regardless of the presence or absence of specific diagnostic mutations, many cancer patients fail to respond to EGFR-targeted therapeutics, and a personalized approach is needed to identify putative (non)responders. We found previously that human peripheral blood and EGF can modulate the activities of EGFR-specific drugs on inhibiting clonogenity in model EGFR-positive A431 squamous carcinoma cells. Here, we report that human serum can dramatically abolish the cell growth rate inhibition by EGFR-specific drugs cetuximab and erlotinib. We show that this phenomenon is linked with derepression of drug-induced G1S cell cycle transition arrest. Furthermore, A431 cell growth inhibition by cetuximab, erlotinib, and EGF correlates with a decreased activity of ERK1/2 proteins. In turn, the EGF- and human serum-mediated rescue of drug-treated A431 cells restores ERK1/2 activity in functional tests. RNA sequencing revealed 1271 and 1566 differentially expressed genes (DEGs) in the presence of cetuximab and erlotinib, respectively. Erlotinib- and cetuximab-specific DEGs significantly overlapped. Interestingly, the expression of 100% and 75% of these DEGs restores to the no-drug level when EGF or a mixed human serum sample, respectively, is added along with cetuximab. In the case of erlotinib, EGF and human serum restore the expression of 39% and 83% of DEGs, respectively. We further assessed differential molecular pathway activation levels and propose that EGF/human serum-mediated A431 resistance to EGFR drugs can be largely explained by reactivation of the MAPK signaling cascade.
Collapse
Affiliation(s)
- Dmitri Kamashev
- I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia;
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (N.S.); (A.B.)
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia;
| | - Nina Shaban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (N.S.); (A.B.)
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia;
| | - Timofey Lebedev
- Engelhardt Institute of Molecular Biology, Moscow 119991, Russia; (T.L.); (V.P.)
| | - Vladimir Prassolov
- Engelhardt Institute of Molecular Biology, Moscow 119991, Russia; (T.L.); (V.P.)
| | - Maria Suntsova
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia;
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (M.R.); (E.P.)
| | - Mikhail Raevskiy
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (M.R.); (E.P.)
| | - Nurshat Gaifullin
- Department of Pathology, Faculty of Medicine, Lomonosov Moscow State University, Moscow 119992, Russia;
| | - Marina Sekacheva
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (M.R.); (E.P.)
| | - Andrew Garazha
- Oncobox Ltd., Moscow 121205, Russia;
- Omicsway Corp., Walnut, CA 91789, USA
| | - Elena Poddubskaya
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (M.R.); (E.P.)
| | - Maksim Sorokin
- I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia;
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia;
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (N.S.); (A.B.)
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia;
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (M.R.); (E.P.)
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| |
Collapse
|
4
|
Asif M, Alvi SS, Azaz T, Khan AR, Tiwari B, Hafeez BB, Nasibullah M. Novel Functionalized Spiro [Indoline-3,5'-pyrroline]-2,2'dione Derivatives: Synthesis, Characterization, Drug-Likeness, ADME, and Anticancer Potential. Int J Mol Sci 2023; 24:ijms24087336. [PMID: 37108498 PMCID: PMC10139052 DOI: 10.3390/ijms24087336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
A highly stereo-selective, one-pot, multicomponent method was chosen to synthesize the novel functionalized 1, 3-cycloaddition spirooxindoles (SOXs) (4a-4h). Synthesized SOXs were analyzed for their drug-likeness and ADME parameters and screened for their anticancer activity. Our molecular docking analysis revealed that among all derivatives of SOXs (4a-4h), 4a has a substantial binding affinity (∆G) -6.65, -6.55, -8.73, and -7.27 Kcal/mol with CD-44, EGFR, AKR1D1, and HER-2, respectively. A functional study demonstrated that SOX 4a has a substantial impact on human cancer cell phenotypes exhibiting abnormality in cytoplasmic and nuclear architecture as well as granule formation leading to cell death. SOX 4a treatment robustly induced reactive oxygen species (ROS) generation in cancer cells as observed by enhanced DCFH-DA signals. Overall, our results suggest that SOX (4a) targets CD-44, EGFR, AKR1D1, and HER-2 and induces ROS generation in cancer cells. We conclude that SOX (4a) could be explored as a potential chemotherapeutic molecule against various cancers in appropriate pre-clinical in vitro and in vivo model systems.
Collapse
Affiliation(s)
- Mohd Asif
- Department of Chemistry, Integral University, Lucknow 226026, Uttar Pradesh, India
| | - Sahir Sultan Alvi
- Department of Immunology and Microbiology, South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Tazeen Azaz
- Department of Biological and Synthetic Chemistry, Centre of Biomedical Research, SGPGIMS-Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| | - Abdul Rahman Khan
- Department of Chemistry, Integral University, Lucknow 226026, Uttar Pradesh, India
| | - Bhoopendra Tiwari
- Department of Biological and Synthetic Chemistry, Centre of Biomedical Research, SGPGIMS-Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| | - Bilal Bin Hafeez
- Department of Immunology and Microbiology, South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Malik Nasibullah
- Department of Chemistry, Integral University, Lucknow 226026, Uttar Pradesh, India
| |
Collapse
|
5
|
Porth I, Hirsch D, Ceribas Y, Weidner P, Weichert W, Götze TO, Perner S, Luley K, Heyer CM, de la Torre C, Hofheinz RD, Lorenzen S, Gaiser T. Comprehensive biomarker analysis of long-term response to trastuzumab in patients with HER2-positive advanced gastric or gastroesophageal adenocarcinoma. Eur J Cancer 2023; 183:119-130. [PMID: 36848831 DOI: 10.1016/j.ejca.2023.01.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND A subgroup of patients with HER2-positive metastatic gastric and gastroesophageal junction cancers shows long-term response under trastuzumab maintenance monotherapy. Obviously, HER2 status alone is not able to identify these patients. We performed this study to identify potential new prognostic biomarkers for this long-term responding patient group. PATIENTS AND METHODS Tumour samples of 19 patients with HER2-positive metastatic gastric and gastroesophageal junction cancer who underwent trastuzumab treatment were retrospectively collected from multiple centres. Patients were divided into long-term responding (n = 7) or short-term responding group (n = 12) according to progression-free survival (PFS≥12 months vs. PFS < 12 months). Next-generation sequencing and microarray-based gene expression analysis were performed along with HER2 and PD-L1 immunohistochemistry. RESULTS Long-term responding patients had significantly higher PD-L1 combined positive scores (CPS) and CPS correlated with longer progression-free survival. PD-L1 positivity (CPS ≥ 1) was further associated with an increased CD4+ memory T-cell score. The ERBB2 copy number as well as the tumour mutational burden could not discriminate between short-term and long-term responding patients. Genetic alterations and coamplifications in HER2 pathway associated genes such as EGFR, which were connected to trastuzumab resistance, were present in 10% of the patients and equally distributed between the groups. CONCLUSION The study highlights the clinical relevance of PD-L1 testing also in the context of trastuzumab treatment and offers a biological rational by demonstrating elevated CD4+ memory T-cells scores in the PD-L1-positive group.
Collapse
Affiliation(s)
- Isabel Porth
- Institute of Pathology, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany.
| | - Daniela Hirsch
- Institute of Pathology, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Yonca Ceribas
- Institute of Pathology, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Wilko Weichert
- Institute of Pathology, School of Medicine, Technical University of Munich, Klinikum rechts der Isar, 81675 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich; Bavarian Cancer Center (BZKF), Germany
| | - Thorsten Oliver Götze
- Institute of Clinical Cancer Research, Krankenhaus Nordwest, UCT-University Cancer Center, Frankfurt, Germany; IKF Klinische Krebsforschung GmbH am Krankenhaus Nordwest, 60488 Frankfurt, Germany
| | - Sven Perner
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein Campus Luebeck, 23538 Luebeck, Germany; Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Kim Luley
- Clinic for Hematology and Oncology, University Hospital Schleswig-Holstein-Campus Luebeck, 23538 Luebeck, Germany
| | - Christian Moritz Heyer
- Biomedical Informatics, Data Mining and Data Analytics, Augsburg University, 86159 Augsburg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Carolina de la Torre
- NGS Core Facility, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Ralf-Dieter Hofheinz
- Interdisciplinary Tumor Center Mannheim, University Hospital Mannheim, University Heidelberg, 68167 Mannheim, Germany
| | - Sylvie Lorenzen
- Medical Clinic III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Timo Gaiser
- Institute of Pathology, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany; Institute of Applied Pathology, 67346, Speyer, Germany
| |
Collapse
|
6
|
Whole-Exome Sequencing Revealed New Candidate Genes for Human Dilated Cardiomyopathy. Diagnostics (Basel) 2022; 12:diagnostics12102411. [PMID: 36292100 PMCID: PMC9600457 DOI: 10.3390/diagnostics12102411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/29/2022] [Accepted: 10/02/2022] [Indexed: 11/17/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a complex disease affecting young adults. It is a pathological condition impairing myocardium activity that leads to heart failure and, in the most severe cases, transplantation, which is currently the only possible therapy for the disease. DCM can be attributed to many genetic determinants interacting with environmental factors, resulting in a highly variable phenotype. Due to this complexity, the early identification of causative gene mutations is an important goal to provide a genetic diagnosis, implement pre-symptomatic interventions, and predict prognosis. The advent of next-generation sequencing (NGS) has opened a new path for mutation screening, and exome sequencing provides a promising approach for identifying causal variants in known genes and novel disease-associated candidates. We analyzed the whole-exome sequencing (WES) of 15 patients affected by DCM without overloading (hypertension, valvular, or congenital heart disease) or chronic ischemic conditions. We identified 70 pathogenic or likely pathogenic variants and 1240 variants of uncertain clinical significance. Gene ontology enrichment analysis was performed to assess the potential connections between affected genes and biological or molecular function, identifying genes directly related to extracellular matrix organization, transcellular movement through the solute carrier and ATP-binding cassette transporter, and vitamin B12 metabolism. We found variants in genes implicated to a different extent in cardiac function that may represent new players in the complex genetic scenario of DCM.
Collapse
|
7
|
Human Blood Serum Inhibits Ductal Carcinoma Cells BT474 Growth and Modulates Effect of HER2 Inhibition. Biomedicines 2022; 10:biomedicines10081914. [PMID: 36009461 PMCID: PMC9405390 DOI: 10.3390/biomedicines10081914] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022] Open
Abstract
Trastuzumab, a HER2-targeted antibody, is widely used for targeted therapy of HER2-positive breast cancer (BC) patients; yet, not all of them respond to this treatment. We investigated here whether trastuzumab activity on the growth of HER2-overexpressing BT474 cells may interfere with human peripheral blood endogenous factors. Among 33 individual BC patient blood samples supplemented to the media, BT474 sensitivity to trastuzumab varied up to 14 times. In the absence of trastuzumab, human peripheral blood serum samples could inhibit growth of BT474, and this effect varied ~10 times for 50 individual samples. In turn, the epidermal growth factor (EGF) suppressed the trastuzumab effect on BT474 cell growth. Trastuzumab treatment increased the proportion of BT474 cells in the G0/G1 phases of cell cycle, while simultaneous addition of EGF decreased it, yet not to the control level. We used RNA sequencing profiling of gene expression to elucidate the molecular mechanisms involved in EGF- and human-sera-mediated attenuation of the trastuzumab effect on BT474 cell growth. Bioinformatic analysis of the molecular profiles suggested that trastuzumab acts similarly to the inhibition of PI3K/Akt/mTOR signaling axis, and the mechanism of EGF suppression of trastuzumab activity may be associated with parallel activation of PKC and transcriptional factors ETV1-ETV5.
Collapse
|
8
|
Out-of-distribution generalization from labelled and unlabelled gene expression data for drug response prediction. NAT MACH INTELL 2021. [DOI: 10.1038/s42256-021-00408-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
9
|
Braunstein EM, Chen H, Juarez F, Yang F, Tao L, Makhlin I, Williams DM, Chaturvedi S, Pallavajjala A, Karantanos T, Martin R, Wohler E, Sobreira N, Gocke CD, Moliterno AR. Germline ERBB2/ HER2 Coding Variants Are Associated with Increased Risk of Myeloproliferative Neoplasms. Cancers (Basel) 2021; 13:cancers13133246. [PMID: 34209587 PMCID: PMC8268839 DOI: 10.3390/cancers13133246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 10/29/2022] Open
Abstract
Familial cases of myeloproliferative neoplasms (MPN) are relatively common, yet few inherited risk factors have been identified. Exome sequencing of a kindred with a familial cancer syndrome characterized by both MPN and melanoma produced a germline variant in the ERBB2/HER2 gene that co-segregates with disease. To further investigate whether germline ERBB2 variants contribute to MPN predisposition, the frequency of ERBB2 variants was analyzed in 1604 cases that underwent evaluation for hematologic malignancy, including 236 cases of MPN. MPN cases had a higher frequency of rare germline ERBB2 coding variants compared to non-MPN hematologic malignancies (8.9% vs. 4.1%, OR 2.4, 95% CI: 1.4 to 4.0, p = 0.0028) as well as cases without a blood cancer diagnosis that served as an internal control (8.9% vs. 2.7%, OR 3.5, 95% CI: 1.4 to 8.3, p = 0.0053). This finding was validated via comparison to an independent control cohort of 1587 cases without selection for hematologic malignancy (8.9% in MPN cases vs. 5.2% in controls, p = 0.040). The most frequent variant identified, ERBB2 c.1960A > G; p.I654V, was present in MPN cases at more than twice its expected frequency. These data indicate that rare germline coding variants in ERBB2 are associated with an increased risk for development of MPN. The ERBB2 gene is a novel susceptibility locus which likely contributes to cancer risk in combination with additional risk alleles.
Collapse
Affiliation(s)
- Evan M. Braunstein
- Department of Medicine, Division of Haematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (H.C.); (F.J.); (F.Y.); (L.T.); (D.M.W.); (S.C.); (A.R.M.)
- Correspondence:
| | - Hang Chen
- Department of Medicine, Division of Haematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (H.C.); (F.J.); (F.Y.); (L.T.); (D.M.W.); (S.C.); (A.R.M.)
| | - Felicia Juarez
- Department of Medicine, Division of Haematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (H.C.); (F.J.); (F.Y.); (L.T.); (D.M.W.); (S.C.); (A.R.M.)
| | - Fanghan Yang
- Department of Medicine, Division of Haematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (H.C.); (F.J.); (F.Y.); (L.T.); (D.M.W.); (S.C.); (A.R.M.)
| | - Lindsay Tao
- Department of Medicine, Division of Haematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (H.C.); (F.J.); (F.Y.); (L.T.); (D.M.W.); (S.C.); (A.R.M.)
| | - Igor Makhlin
- Department of Medicine, Division of Hematology & Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Donna M. Williams
- Department of Medicine, Division of Haematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (H.C.); (F.J.); (F.Y.); (L.T.); (D.M.W.); (S.C.); (A.R.M.)
| | - Shruti Chaturvedi
- Department of Medicine, Division of Haematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (H.C.); (F.J.); (F.Y.); (L.T.); (D.M.W.); (S.C.); (A.R.M.)
| | - Aparna Pallavajjala
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (A.P.); (C.D.G.)
| | - Theodoros Karantanos
- Department of Medical Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Renan Martin
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.M.); (E.W.); (N.S.)
| | - Elizabeth Wohler
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.M.); (E.W.); (N.S.)
| | - Nara Sobreira
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.M.); (E.W.); (N.S.)
| | - Christopher D. Gocke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (A.P.); (C.D.G.)
- Department of Medical Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Alison R. Moliterno
- Department of Medicine, Division of Haematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (H.C.); (F.J.); (F.Y.); (L.T.); (D.M.W.); (S.C.); (A.R.M.)
| |
Collapse
|
10
|
Cleary JM, Raghavan S, Wu Q, Li YY, Spurr LF, Gupta HV, Rubinson DA, Fetter IJ, Hornick JL, Nowak JA, Siravegna G, Goyal L, Shi L, Brais LK, Loftus M, Shinagare AB, Abrams TA, Clancy TE, Wang J, Patel AK, Brichory F, Vaslin Chessex A, Sullivan RJ, Keller RB, Denning S, Hill ER, Shapiro GI, Pokorska-Bocci A, Zanna C, Ng K, Schrag D, Janne PA, Hahn WC, Cherniack AD, Corcoran RB, Meyerson M, Daina A, Zoete V, Bardeesy N, Wolpin BM. FGFR2 Extracellular Domain In-Frame Deletions are Therapeutically Targetable Genomic Alterations that Function as Oncogenic Drivers in Cholangiocarcinoma. Cancer Discov 2021; 11:2488-2505. [PMID: 33926920 DOI: 10.1158/2159-8290.cd-20-1669] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/10/2021] [Accepted: 04/26/2021] [Indexed: 11/16/2022]
Abstract
We conducted next generation DNA sequencing on 335 biliary tract cancers and characterized the genomic landscape by anatomic site within the biliary tree. In addition to frequent FGFR2 fusions among patients with intrahepatic cholangiocarcinoma (IHCC), we identified FGFR2 extracellular domain in-frame deletions (EIDs) in 5 of 178 (2.8%) patients with IHCC, including two patients with FGFR2 p.H167_N173del. Expression of this FGFR2 EID in NIH3T3 cells resulted in constitutive FGFR2 activation, oncogenic transformation, and sensitivity to FGFR inhibitors. Three patients with FGFR2 EIDs were treated with Debio 1347, an oral FGFR-1/2/3 inhibitor, and all showed partial responses. One patient developed an acquired L618F FGFR2 kinase domain mutation at disease progression and experienced a further partial response for 17 months to an irreversible FGFR2 inhibitor, futibatinib. Together, these findings reveal FGFR2 EIDs as an alternative mechanism of FGFR2 activation in IHCC that predict sensitivity to FGFR inhibitors in the clinic.
Collapse
Affiliation(s)
- James M Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | | | | | - Yvonne Y Li
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | - Liam F Spurr
- Dana-Farber Cancer Institute, Harvard Medical School
| | - Hersh V Gupta
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | | | | | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School
| | | | | | - Lipika Goyal
- Internal Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School
| | - Lei Shi
- Center for Cancer Research, Massachusetts General Hospital Cancer Center, Harvard Medical School
| | - Lauren K Brais
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | | | - Atul B Shinagare
- Department of Radiology, Brigham and Women's Hospital/ Dana-Farber Cancer Institute
| | | | | | - Jiping Wang
- Department of Surgery, Brigham and Women's Hospital
| | - Anuj K Patel
- Department of Gastrointestinal Oncology, Dana-Farber Cancer Institute
| | | | | | - Ryan J Sullivan
- Center for Melanoma, Massachusetts General Hospital Cancer Center
| | | | | | - Emma R Hill
- Dana-Farber/Brigham and Women's Cancer Center
| | | | | | | | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | | | - Pasi A Janne
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute
| | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | - Andrew D Cherniack
- Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School
| | | | | | | | | | | | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber/Harvard Cancer Center
| |
Collapse
|
11
|
Kam AYF, Piryani SO, Lee CL, Rizzieri DA, Spector NL, Sarantopoulos S, Doan PL. Selective ERBB2 and BCL2 Inhibition Is Synergistic for Mitochondrial-Mediated Apoptosis in MDS and AML Cells. Mol Cancer Res 2021; 19:886-899. [PMID: 33514658 DOI: 10.1158/1541-7786.mcr-20-0973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/23/2020] [Accepted: 01/21/2021] [Indexed: 11/16/2022]
Abstract
The ERBB2 proto-oncogene is associated with an aggressive phenotype in breast cancer. Its role in hematologic malignancies is incompletely defined, in part because ERBB2 is not readily detected on the surface of cancer cells. We demonstrate that truncated ERBB2, which lacks the extracellular domain, is overexpressed on primary CD34+ myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) cells compared with healthy hematopoietic cells. This overexpression of ERBB2 is associated with aberrant, oncogenic signaling with autophosphorylation of multiple tyrosine sites. Like in breast cancers, ERBB2 can exist as truncated isoforms p95ERBB2 and p110ERBB2 in MDS and AML. Neutralization of ERBB2 signaling with ERBB2 tyrosine kinase inhibitors (i.e., lapatinib, afatinib, and neratinib) increases apoptotic cell death and reduces human engraftment of MDS cells in mice at 21 weeks posttransplantation. Inhibition of ERBB2 modulates the expression of multiple pro- and anti-apoptotic mitochondrial proteins, including B-cell lymphoma 2 (BCL2). Dual blockade with ERBB2 and BCL2 inhibitors triggers additional reductions of BCL2 phosphorylation and myeloid cell leukemia-1 (MCL1) expression compared with single drug treatment. Dual therapy was synergistic at all tested doses, with a dose reduction index of up to 29 for lapatinib + venetoclax compared with venetoclax alone. Notably, these agents operated together and shifted cancer cells to a pro-apoptotic phenotype, resulting in increased mitochondrial cytochrome c release and activated caspase-3-mediated cell death. IMPLICATIONS: These findings warrant study of ERBB2 and BCL2 combination therapy in patients with MDS and AML. VISUAL OVERVIEW: http://mcr.aacrjournals.org/content/molcanres/19/5/886/F1.large.jpg.
Collapse
Affiliation(s)
- Angel Y F Kam
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University, Durham, North Carolina
| | - Sadhna O Piryani
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University, Durham, North Carolina
| | - Chang-Lung Lee
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina.,Duke Cancer Institute, Duke University, Durham, North Carolina
| | - David A Rizzieri
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University, Durham, North Carolina.,Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Neil L Spector
- Duke Cancer Institute, Duke University, Durham, North Carolina.,Division of Medical Oncology, Department of Medicine, Duke University, Durham, North Carolina
| | - Stefanie Sarantopoulos
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University, Durham, North Carolina.,Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Phuong L Doan
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University, Durham, North Carolina. .,Duke Cancer Institute, Duke University, Durham, North Carolina
| |
Collapse
|
12
|
Sun ZG, Zhao LH, Li ZN, Zhu HL. Development and Challenges of the Discovery of HER2 Inhibitors. Mini Rev Med Chem 2020; 20:2123-2134. [PMID: 32727326 DOI: 10.2174/1389557520666200729162118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/01/2020] [Accepted: 06/23/2020] [Indexed: 11/22/2022]
Abstract
The treatment of cancer has always been a major problem in the world. Some cancers cannot be treated with surgery, but only with cancer drugs. Among many cancer drugs, small molecule inhibitors play an irreplaceable role. HER2 is one of the HER families, and the development of HER2 inhibitors has made a huge contribution to the treatment of cancer. Some HER2 inhibitors are already on the market, and some HER2 inhibitors are undergoing clinical research. The design, synthesis and development of new HER2 inhibitors targeting different targets are also ongoing, and some are even under clinical research. The HER2 inhibitors that are on the market have developed resistance, which brings great challenges to the HER2 inhibitor development in the future. This article reviews the development and challenges of the discovery of HER2 inhibitors.
Collapse
Affiliation(s)
- Zhi-Gang Sun
- Central Laboratory, Linyi Central Hospital, No.17 Jiankang Road, Linyi 276400, China
| | - Liang-Hui Zhao
- Weifang Medical University, No. 7166 Baotong West Street, Weifang 261000, China
| | - Zhi-Na Li
- Central Laboratory, Linyi Central Hospital, No.17 Jiankang Road, Linyi 276400, China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing 210023, China
| |
Collapse
|