1
|
Emelyanova AG, Zolotovskaia MA, Poddubskaya EV, Modestov AA, Prassolov VS, Kuzmin DV, Buzdin AA. Activation of the ERK1/2 Molecular Pathways and Its Relation to the Pathogenicity of Human Malignant Tumors. Acta Naturae 2025; 17:36-51. [PMID: 40264578 PMCID: PMC12011182 DOI: 10.32607/actanaturae.27497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/05/2024] [Indexed: 04/24/2025] Open
Abstract
Mitogen-activated protein kinases, ERK1/2 (MAPK3/1), play a key role in the regulation of cell growth, differentiation, and apoptosis. We have previously presented evidence proving that activation of the ERK1/2 axis in cancer cells following the administration of therapeutics leads to the overexpression of growth factor receptors and drug resistance. Recently, we have proposed a new bioinformatic technique that enables direct construction of interactome network-based molecular pathways for gene products of interest, as well as quantitation of their activation levels using high-throughput gene expression data. In this study, we, for the first time, algorithmically constructed ERK1/2 molecular pathways and investigated how their activation levels (PALs) affect survival and responsiveness to targeted drugs at the pan-cancer level based on transcriptomic data. We examined a total of 11 287 human tumor profiles from 31 types of cancer, drawn from 53 of our previously published and other literature datasets, looking at patient survival and clinical response to 29 chemo- and targeted therapy regimens. We found that activation of the ERK1/2 pathways has different prognostic significance depending on cancer type. In glioblastoma, sarcoma, lung, kidney, bladder, gastric, colon, and several other cancer types, ERK pathway activation was associated with worse survival. In contrast, the same phenomenon was associated with a better chance of survival in HER2+, luminal A and luminal B breast cancer, and uterine corpus cancer. These trends were consistent with treatment response analysis. At the same time, we found significantly worse associations with the expression levels of individual MAPK1 and MAPK3 genes: hence, ERK1/2 pathway activation levels can be considered putative biomarkers for predicting clinical outcomes and selecting new personalized treatment strategies, such as the use of MAPK inhibitors.
Collapse
Affiliation(s)
- A. G. Emelyanova
- Moscow Institute of Physics and Technology, Dolgoprudny, 141701 Russian Federation
- I.M. Sechenov First Moscow State Medical University, Moscow, 119991 Russian Federation
| | - M. A. Zolotovskaia
- Moscow Institute of Physics and Technology, Dolgoprudny, 141701 Russian Federation
- I.M. Sechenov First Moscow State Medical University, Moscow, 119991 Russian Federation
| | - E. V. Poddubskaya
- I.M. Sechenov First Moscow State Medical University, Moscow, 119991 Russian Federation
| | - A. A. Modestov
- Moscow Institute of Physics and Technology, Dolgoprudny, 141701 Russian Federation
- I.M. Sechenov First Moscow State Medical University, Moscow, 119991 Russian Federation
| | - V. S. Prassolov
- Engelhardt Institute of Molecular Biology, Moscow, 119991 Russian Federation
| | - D. V. Kuzmin
- Moscow Institute of Physics and Technology, Dolgoprudny, 141701 Russian Federation
| | - A. A. Buzdin
- Moscow Institute of Physics and Technology, Dolgoprudny, 141701 Russian Federation
- I.M. Sechenov First Moscow State Medical University, Moscow, 119991 Russian Federation
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| |
Collapse
|
2
|
Poddubskaya E, Suntsova M, Lyadova M, Luppov D, Guryanova A, Lyadov V, Garazha A, Sorokin M, Semenova A, Shatalov V, Biakhova M, Simonov A, Moisseev A, Buzdin A. Biomarkers of success of anti-PD-(L)1 immunotherapy for non-small cell lung cancer derived from RNA- and whole-exome sequencing: results of a prospective observational study on a cohort of 85 patients. Front Immunol 2024; 15:1493877. [PMID: 39723204 PMCID: PMC11669362 DOI: 10.3389/fimmu.2024.1493877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) treatment have shown high efficacy for about 15 cancer types. However, this therapy is only effective in 20-30% of cancer patients. Thus, the precise biomarkers of ICI response are an urgent need. Methods We conducted a prospective observational study of the prognostic potential ofseveral existing and putative biomarkers of response to immunotherapy in acohort of 85 patients with lung cancer (LC) receiving PD-1 or PD-L1 targeted ICIs. Tumor biosamples were obtained prior to ICI treatment and profiled by whole exome and RNA sequencing. The entire 403 putative biomarkers were screened, including tumor mutation burden (TMB) and number of cancer neoantigens, 131 specific HLA alleles, homozygous state of 11 HLA alleles and their superfamilies; four gene mutation biomarkers, expression of 45 immune checkpoint genes and closely related genes, and three previously published diagnostic gene signatures; for the first time, activation levels of 188 molecular pathways containing immune checkpoint genes and activation levels of 19 pathways algorithmically generated using a human interactome model centered around immune checkpoint genes. Treatment outcomes and/or progression-free survival (PFS) times were available for 61 of 85 patients with LC, including 24 patients with adenocarcinoma and 27 patients with squamous cell LC, whose samples were further analyzed. For the rest 24 patients, both treatment outcomes and PFS data could not be collected. Of these, 54 patients were treated with PD1-specific and 7 patients with PD-L1-specific ICIs. We evaluated the potential of biomarkers based on PFS and RECIST treatment response data. Results In our sample, 45 biomarkers were statistically significantly associated with PFS and 44 with response to treatment, of which eight were shared. Five of these (CD3G and NCAM1 gene expression levels, and levels of activation of Adrenergic signaling in cardiomyocytes, Growth hormone signaling, and Endothelin molecular pathways) were used in our signature that showed an AUC of 0.73 and HR of 0.27 (p=0.00034) on the experimental dataset. This signature was also reliable (AUC 0.76, 0.87) for the independent publicly available LC datasets GSE207422, GSE126044 annotated with ICI response data and demonstrated same survival trends on independent dataset GSE135222 annotated with PFS data. In both experimental and one independent datasets annotated with samples' histotypes, the signature worked better for lung adenocarcinoma than for squamous cell LC. Conclusion The high reliability of our signature to predict response and PFS after ICI treatment was demonstrated using experimental and 3 independent datasets. Additionally, annotated molecular profiles obtained in this study were made publicly accessible.
Collapse
Affiliation(s)
- Elena Poddubskaya
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Vitamed Clinic, Moscow, Russia
| | - Maria Suntsova
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Translational Genomic Bioinformatic, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Marina Lyadova
- Oncology Center No. 1, Moscow City Hospital Named after S. S. Yudin, Moscow Healthcare Department, Moscow, Russia
| | - Daniil Luppov
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Translational Genomic Bioinformatic, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Department of Molecular Genetic Research, Endocrinology Research Center, Moscow, Russia
| | - Anastasia Guryanova
- Laboratory of Translational Genomic Bioinformatic, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Vladimir Lyadov
- Oncology Center No. 1, Moscow City Hospital Named after S. S. Yudin, Moscow Healthcare Department, Moscow, Russia
- Branch Campus of the Federal State Budgetary Educational Institution of Further Professional Education «Russian Medical Academy of Continuous Professional Education» of the Ministry of Healthcare of the Russian Federation, Novokuznetsk, Russia, Novokuznetsk, Russia
| | | | - Maksim Sorokin
- Department of Molecular Genetic Research, Endocrinology Research Center, Moscow, Russia
- Department of Research, Oncobox Ltd., Moscow, Russia
- Laboratory for Genomic Analysis of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Anna Semenova
- Oncology Center No. 1, Moscow City Hospital Named after S. S. Yudin, Moscow Healthcare Department, Moscow, Russia
| | - Vitaly Shatalov
- Oncology Center No. 1, Moscow City Hospital Named after S. S. Yudin, Moscow Healthcare Department, Moscow, Russia
| | - Maria Biakhova
- Oncology Center No. 1, Moscow City Hospital Named after S. S. Yudin, Moscow Healthcare Department, Moscow, Russia
| | - Alexander Simonov
- Laboratory of Translational Genomic Bioinformatic, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Aleksey Moisseev
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Molecular Genetic Research, Endocrinology Research Center, Moscow, Russia
| | - Anton Buzdin
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Translational Genomic Bioinformatic, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Laboratory for Genomic Analysis of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| |
Collapse
|
3
|
Shaban N, Kamashev D, Emelianova A, Buzdin A. Targeted Inhibitors of EGFR: Structure, Biology, Biomarkers, and Clinical Applications. Cells 2023; 13:47. [PMID: 38201251 PMCID: PMC10778338 DOI: 10.3390/cells13010047] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Members of the EGFR family of tyrosine kinase receptors are major regulators of cellular proliferation, differentiation, and survival. In humans, abnormal activation of EGFR is associated with the development and progression of many cancer types, which makes it an attractive target for molecular-guided therapy. Two classes of EGFR-targeted cancer therapeutics include monoclonal antibodies (mAbs), which bind to the extracellular domain of EGFR, and tyrosine kinase inhibitors (TKIs), which mostly target the intracellular part of EGFR and inhibit its activity in molecular signaling. While EGFR-specific mAbs and three generations of TKIs have demonstrated clinical efficacy in various settings, molecular evolution of tumors leads to apparent and sometimes inevitable resistance to current therapeutics, which highlights the need for deeper research in this field. Here, we tried to provide a comprehensive and systematic overview of the rationale, molecular mechanisms, and clinical significance of the current EGFR-targeting drugs, highlighting potential candidate molecules in development. We summarized the underlying mechanisms of resistance and available personalized predictive approaches that may lead to improved efficacy of EGFR-targeted therapies. We also discuss recent developments and the use of specific therapeutic strategies, such as multi-targeting agents and combination therapies, for overcoming cancer resistance to EGFR-specific drugs.
Collapse
Affiliation(s)
- Nina Shaban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (D.K.); (A.B.)
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Dmitri Kamashev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (D.K.); (A.B.)
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Aleksandra Emelianova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia;
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (D.K.); (A.B.)
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| |
Collapse
|
4
|
Zolotovskaia M, Kovalenko M, Pugacheva P, Tkachev V, Simonov A, Sorokin M, Seryakov A, Garazha A, Gaifullin N, Sekacheva M, Zakharova G, Buzdin AA. Algorithmically Reconstructed Molecular Pathways as the New Generation of Prognostic Molecular Biomarkers in Human Solid Cancers. Proteomes 2023; 11:26. [PMID: 37755705 PMCID: PMC10535530 DOI: 10.3390/proteomes11030026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Individual gene expression and molecular pathway activation profiles were shown to be effective biomarkers in many cancers. Here, we used the human interactome model to algorithmically build 7470 molecular pathways centered around individual gene products. We assessed their associations with tumor type and survival in comparison with the previous generation of molecular pathway biomarkers (3022 "classical" pathways) and with the RNA transcripts or proteomic profiles of individual genes, for 8141 and 1117 samples, respectively. For all analytes in RNA and proteomic data, respectively, we found a total of 7441 and 7343 potential biomarker associations for gene-centric pathways, 3020 and 2950 for classical pathways, and 24,349 and 6742 for individual genes. Overall, the percentage of RNA biomarkers was statistically significantly higher for both types of pathways than for individual genes (p < 0.05). In turn, both types of pathways showed comparable performance. The percentage of cancer-type-specific biomarkers was comparable between proteomic and transcriptomic levels, but the proportion of survival biomarkers was dramatically lower for proteomic data. Thus, we conclude that pathway activation level is the advanced type of biomarker for RNA and proteomic data, and momentary algorithmic computer building of pathways is a new credible alternative to time-consuming hypothesis-driven manual pathway curation and reconstruction.
Collapse
Affiliation(s)
- Marianna Zolotovskaia
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology (State University), 141701 Dolgoprudny, Russia
- Omicsway Corp., Walnut, CA 91789, USA
- Laboratory of Clinical and Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, 119048 Moscow, Russia
| | - Maks Kovalenko
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology (State University), 141701 Dolgoprudny, Russia
| | - Polina Pugacheva
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology (State University), 141701 Dolgoprudny, Russia
| | | | - Alexander Simonov
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology (State University), 141701 Dolgoprudny, Russia
- Omicsway Corp., Walnut, CA 91789, USA
| | - Maxim Sorokin
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology (State University), 141701 Dolgoprudny, Russia
- Laboratory of Clinical and Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, 119048 Moscow, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| | | | | | - Nurshat Gaifullin
- Department of Pathology, Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Marina Sekacheva
- Laboratory of Clinical and Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, 119048 Moscow, Russia
| | - Galina Zakharova
- Laboratory of Clinical and Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, 119048 Moscow, Russia
| | - Anton A. Buzdin
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology (State University), 141701 Dolgoprudny, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119048 Moscow, Russia
- Laboratory of Systems Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| |
Collapse
|
5
|
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
|
6
|
Pun FW, Liu BHM, Long X, Leung HW, Leung GHD, Mewborne QT, Gao J, Shneyderman A, Ozerov IV, Wang J, Ren F, Aliper A, Bischof E, Izumchenko E, Guan X, Zhang K, Lu B, Rothstein JD, Cudkowicz ME, Zhavoronkov A. Identification of Therapeutic Targets for Amyotrophic Lateral Sclerosis Using PandaOmics – An AI-Enabled Biological Target Discovery Platform. Front Aging Neurosci 2022; 14:914017. [PMID: 35837482 PMCID: PMC9273868 DOI: 10.3389/fnagi.2022.914017] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disease with ill-defined pathogenesis, calling for urgent developments of new therapeutic regimens. Herein, we applied PandaOmics, an AI-driven target discovery platform, to analyze the expression profiles of central nervous system (CNS) samples (237 cases; 91 controls) from public datasets, and direct iPSC-derived motor neurons (diMNs) (135 cases; 31 controls) from Answer ALS. Seventeen high-confidence and eleven novel therapeutic targets were identified and will be released onto ALS.AI (http://als.ai/). Among the proposed targets screened in the c9ALS Drosophila model, we verified 8 unreported genes (KCNB2, KCNS3, ADRA2B, NR3C1, P2RY14, PPP3CB, PTPRC, and RARA) whose suppression strongly rescues eye neurodegeneration. Dysregulated pathways identified from CNS and diMN data characterize different stages of disease development. Altogether, our study provides new insights into ALS pathophysiology and demonstrates how AI speeds up the target discovery process, and opens up new opportunities for therapeutic interventions.
Collapse
Affiliation(s)
- Frank W. Pun
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Bonnie Hei Man Liu
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Xi Long
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Hoi Wing Leung
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Geoffrey Ho Duen Leung
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Quinlan T. Mewborne
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, United States
| | - Junli Gao
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, United States
| | - Anastasia Shneyderman
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Ivan V. Ozerov
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Ju Wang
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Feng Ren
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Alexander Aliper
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Evelyne Bischof
- College of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- International Center for Multimorbidity and Complexity in Medicine (ICMC), Universität Zürich, Zurich, Switzerland
| | - Evgeny Izumchenko
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, United States
| | - Xiaoming Guan
- 4B Technologies Limited, Suzhou BioBay, Suzhou, China
| | - Ke Zhang
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, United States
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, United States
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Jeffrey D. Rothstein
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Merit E. Cudkowicz
- Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- *Correspondence: Merit E. Cudkowicz,
| | - Alex Zhavoronkov
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
- Buck Institute for Research on Aging, Novato, CA, United States
- Alex Zhavoronkov,
| |
Collapse
|
7
|
Konovalov N, Timonin S, Asyutin D, Raevskiy M, Sorokin M, Buzdin A, Kaprovoy S. Transcriptomic Portraits and Molecular Pathway Activation Features of Adult Spinal Intramedullary Astrocytomas. Front Oncol 2022; 12:837570. [PMID: 35387112 PMCID: PMC8978956 DOI: 10.3389/fonc.2022.837570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 11/30/2022] Open
Abstract
In this study, we report 31 spinal intramedullary astrocytoma (SIA) RNA sequencing (RNA-seq) profiles for 25 adult patients with documented clinical annotations. To our knowledge, this is the first clinically annotated RNA-seq dataset of spinal astrocytomas derived from the intradural intramedullary compartment. We compared these tumor profiles with the previous healthy central nervous system (CNS) RNA-seq data for spinal cord and brain and identified SIA-specific gene sets and molecular pathways. Our findings suggest a trend for SIA-upregulated pathways governing interactions with the immune cells and downregulated pathways for the neuronal functioning in the context of normal CNS activity. In two patient tumor biosamples, we identified diagnostic KIAA1549-BRAF fusion oncogenes, and we also found 16 new SIA-associated fusion transcripts. In addition, we bioinformatically simulated activities of targeted cancer drugs in SIA samples and predicted that several tyrosine kinase inhibitory drugs and thalidomide analogs could be potentially effective as second-line treatment agents to aid in the prevention of SIA recurrence and progression.
Collapse
Affiliation(s)
| | | | | | - Mikhail Raevskiy
- Omicsway Corp., Walnut, CA, United States
- Moscow Institute of Physics and Technology, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maxim Sorokin
- Moscow Institute of Physics and Technology, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anton Buzdin
- Omicsway Corp., Walnut, CA, United States
- Moscow Institute of Physics and Technology, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Oncobox Ltd., Moscow, Russia
| | | |
Collapse
|
8
|
Sorokin M, Gorelyshev A, Efimov V, Zotova E, Zolotovskaia M, Rabushko E, Kuzmin D, Seryakov A, Kamashev D, Li X, Poddubskaya E, Suntsova M, Buzdin A. RNA Sequencing Data for FFPE Tumor Blocks Can Be Used for Robust Estimation of Tumor Mutation Burden in Individual Biosamples. Front Oncol 2021; 11:732644. [PMID: 34650919 PMCID: PMC8506044 DOI: 10.3389/fonc.2021.732644] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/06/2021] [Indexed: 01/16/2023] Open
Abstract
Tumor mutation burden (TMB) is a well-known efficacy predictor for checkpoint inhibitor immunotherapies. Currently, TMB assessment relies on DNA sequencing data. Gene expression profiling by RNA sequencing (RNAseq) is another type of analysis that can inform clinical decision-making and including TMB estimation may strongly benefit this approach, especially for the formalin-fixed, paraffin-embedded (FFPE) tissue samples. Here, we for the first time compared TMB levels deduced from whole exome sequencing (WES) and RNAseq profiles of the same FFPE biosamples in single-sample mode. We took TCGA project data with mean sequencing depth 23 million gene-mapped reads (MGMRs) and found 0.46 (Pearson)–0.59 (Spearman) correlation with standard mutation calling pipelines. This was converted into low (<10) and high (>10) TMB per megabase classifier with area under the curve (AUC) 0.757, and application of machine learning increased AUC till 0.854. We then compared 73 experimental pairs of WES and RNAseq profiles with lower (mean 11 MGMRs) and higher (mean 68 MGMRs) RNA sequencing depths. For higher depth, we observed ~1 AUC for the high/low TMB classifier and 0.85 (Pearson)–0.95 (Spearman) correlation with standard mutation calling pipelines. For the lower depth, the AUC was below the high-quality threshold of 0.7. Thus, we conclude that using RNA sequencing of tumor materials from FFPE blocks with enough coverage can afford for high-quality discrimination of tumors with high and low TMB levels in a single-sample mode.
Collapse
Affiliation(s)
- Maxim Sorokin
- Biostatistics and Bioinformatics Subgroup, European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium.,The Laboratory of Clinical and Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,OmicsWay Corp., Walnut, CA, United States
| | - Alexander Gorelyshev
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,OmicsWay Corp., Walnut, CA, United States
| | - Victor Efimov
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Evgenia Zotova
- The Laboratory of Clinical and Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Marianna Zolotovskaia
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Elizaveta Rabushko
- The Laboratory of Clinical and Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Denis Kuzmin
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | | | - Dmitry Kamashev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Xinmin Li
- Department of Pathology & Laboratory Medicine, University of California Los Angeles (UCLA) Technology Center for Genomics & Bioinformatics, Los Angeles, CA, United States
| | - Elena Poddubskaya
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maria Suntsova
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anton Buzdin
- Biostatistics and Bioinformatics Subgroup, European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium.,Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,OmicsWay Corp., Walnut, CA, United States.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
9
|
Using proteomic and transcriptomic data to assess activation of intracellular molecular pathways. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 127:1-53. [PMID: 34340765 DOI: 10.1016/bs.apcsb.2021.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Analysis of molecular pathway activation is the recent instrument that helps to quantize activities of various intracellular signaling, structural, DNA synthesis and repair, and biochemical processes. This may have a deep impact in fundamental research, bioindustry, and medicine. Unlike gene ontology analyses and numerous qualitative methods that can establish whether a pathway is affected in principle, the quantitative approach has the advantage of exactly measuring the extent of a pathway up/downregulation. This results in emergence of a new generation of molecular biomarkers-pathway activation levels, which reflect concentration changes of all measurable pathway components. The input data can be the high-throughput proteomic or transcriptomic profiles, and the output numbers take both positive and negative values and positively reflect overall pathway activation. Due to their nature, the pathway activation levels are more robust biomarkers compared to the individual gene products/protein levels. Here, we review the current knowledge of the quantitative gene expression interrogation methods and their applications for the molecular pathway quantization. We consider enclosed bioinformatic algorithms and their applications for solving real-world problems. Besides a plethora of applications in basic life sciences, the quantitative pathway analysis can improve molecular design and clinical investigations in pharmaceutical industry, can help finding new active biotechnological components and can significantly contribute to the progressive evolution of personalized medicine. In addition to the theoretical principles and concepts, we also propose publicly available software for the use of large-scale protein/RNA expression data to assess the human pathway activation levels.
Collapse
|
10
|
Kamashev D, Sorokin M, Kochergina I, Drobyshev A, Vladimirova U, Zolotovskaia M, Vorotnikov I, Shaban N, Raevskiy M, Kuzmin D, Buzdin A. Human blood serum can donor-specifically antagonize effects of EGFR-targeted drugs on squamous carcinoma cell growth. Heliyon 2021; 7:e06394. [PMID: 33748471 PMCID: PMC7966997 DOI: 10.1016/j.heliyon.2021.e06394] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/29/2020] [Accepted: 02/25/2021] [Indexed: 02/09/2023] Open
Abstract
Many patients fail to respond to EGFR-targeted therapeutics, and personalized diagnostics is needed to identify putative responders. We investigated 1630 colorectal and lung squamous carcinomas and 1357 normal lung and colon samples and observed huge variation in EGFR pathway activation in both cancerous and healthy tissues, irrespectively on EGFR gene mutation status. We investigated whether human blood serum can affect squamous carcinoma cell growth and EGFR drug response. We demonstrate that human serum antagonizes the effects of EGFR-targeted drugs erlotinib and cetuximab on A431 squamous carcinoma cells by increasing IC50 by about 2- and 20-fold, respectively. The effects on clonogenicity varied significantly across the individual serum samples in every experiment, with up to 100% differences. EGF concentration could explain many effects of blood serum samples, and EGFR ligands-depleted serum showed lesser effect on drug sensitivity.
Collapse
Affiliation(s)
- Dmitry Kamashev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
| | - Maksim Sorokin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Irina Kochergina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
| | - Aleksey Drobyshev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Uliana Vladimirova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
| | - Marianna Zolotovskaia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Igor Vorotnikov
- Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russia
| | - Nina Shaban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Mikhail Raevskiy
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
- OmicsWay Corp., Walnut, CA, USA
| | - Denis Kuzmin
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| |
Collapse
|
11
|
Sorokin M, Borisov N, Kuzmin D, Gudkov A, Zolotovskaia M, Garazha A, Buzdin A. Algorithmic Annotation of Functional Roles for Components of 3,044 Human Molecular Pathways. Front Genet 2021; 12:617059. [PMID: 33633781 PMCID: PMC7900570 DOI: 10.3389/fgene.2021.617059] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/20/2021] [Indexed: 12/16/2022] Open
Abstract
Current methods of high-throughput molecular and genomic analyses enabled to reconstruct thousands of human molecular pathways. Knowledge of molecular pathways structure and architecture taken along with the gene expression data can help interrogating the pathway activation levels (PALs) using different bioinformatic algorithms. In turn, the pathway activation profiles can characterize molecular processes, which are differentially regulated and give numeric characteristics of the extent of their activation or inhibition. However, different pathway nodes may have different functions toward overall pathway regulation, and calculation of PAL requires knowledge of molecular function of every node in the pathway in terms of its activator or inhibitory role. Thus, high-throughput annotation of functional roles of pathway nodes is required for the comprehensive analysis of the pathway activation profiles. We proposed an algorithm that identifies functional roles of the pathway components and applied it to annotate 3,044 human molecular pathways extracted from the Biocarta, Reactome, KEGG, Qiagen Pathway Central, NCI, and HumanCYC databases and including 9,022 gene products. The resulting knowledgebase can be applied for the direct calculation of the PALs and establishing large scale profiles of the signaling, metabolic, and DNA repair pathway regulation using high throughput gene expression data. We also provide a bioinformatic tool for PAL data calculations using the current pathway knowledgebase.
Collapse
Affiliation(s)
- Maxim Sorokin
- Omicsway Corp., Walnut, CA, United States.,Laboratory of Clinical Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Laboratory for Translational Bioinformatics, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Nicolas Borisov
- Omicsway Corp., Walnut, CA, United States.,Laboratory for Translational Bioinformatics, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Denis Kuzmin
- Laboratory for Translational Bioinformatics, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Alexander Gudkov
- Laboratory of Clinical Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Marianna Zolotovskaia
- Laboratory for Translational Bioinformatics, Moscow Institute of Physics and Technology, Moscow, Russia
| | | | - Anton Buzdin
- Omicsway Corp., Walnut, CA, United States.,Laboratory for Translational Bioinformatics, Moscow Institute of Physics and Technology, Moscow, Russia.,Laboratory of Systems Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
12
|
Qi J, Hu Z, Liu S, Li F, Wang S, Wang W, Sheng X, Feng L. Comprehensively Analyzed Macrophage-Regulated Genes Indicate That PSMA2 Promotes Colorectal Cancer Progression. Front Oncol 2021; 10:618902. [PMID: 33537240 PMCID: PMC7849682 DOI: 10.3389/fonc.2020.618902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/30/2020] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. Here, we identified tumor-associated macrophages (TAMs) as regulators of genes in CRC. In total, the expressions of 457 genes were dysregulated after TAM coculture; specifically, 344 genes were up-regulated, and 113 genes were down-regulated. Bioinformatic analysis implied that these TAM-related genes were associated with regulation of the processes of macromolecule metabolism, apoptosis, cell death, programmed cell death, and the response to stress. To further uncover the interplay among these proteins, we constructed a PPI network; 15 key regulators were identified in CRC, including VEGFA, FN1, JUN, CDH1, MAPK8, and FOS. Among the identified genes, we focused on PSMA2 and conducted loss-of-function experiments to validate the functions of PSMA2 in CRC. To further determine the mechanism by which PSMA2 affected CRC, we conducted multiple assays in CRC cell lines and tissues. PSMA2 enhanced the proliferation, migration and invasion of CRC cells. Moreover, our data indicated that PSMA2 expression was dramatically increased in stage 1, stage 2, stage 3, and stage 4 CRC samples. Our data indicated that PSMA2 was one target of miR-132. A miR-132 mimic greatly hindered CRC cell proliferation. In addition, the luciferase assay results revealed that miR-132 directly regulated PSMA2. Moreover, our data indicated that miR-132 expression was greatly decreased in CRC samples, which was associated with longer survival times of CRC patients, implying that miR-132 was a probable biomarker for CRC. Collectively, these data indicate that PSMA2 is a promising target for the therapy of CRC.
Collapse
Affiliation(s)
- Jingbo Qi
- Endoscopy Center, Minhang Hospital, Fudan University and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhiqiu Hu
- Department of Surgical, Minhang Hospital, Fudan University, Shanghai, China
| | - Shaoqun Liu
- Department of Surgical, Minhang Hospital, Fudan University, Shanghai, China
| | - Fan Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Sheng Wang
- Endoscopy Center, Minhang Hospital, Fudan University and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wuqing Wang
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Xia Sheng
- Department of Pathology, Minhang Hospital, Fudan University, Shanghai, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Sorokin M, Ignatev K, Barbara V, Vladimirova U, Muraveva A, Suntsova M, Gaifullin N, Vorotnikov I, Kamashev D, Bondarenko A, Baranova M, Poddubskaya E, Buzdin A. Molecular Pathway Activation Markers Are Associated with Efficacy of Trastuzumab Therapy in Metastatic HER2-Positive Breast Cancer Better than Individual Gene Expression Levels. BIOCHEMISTRY (MOSCOW) 2020; 85:758-772. [DOI: 10.1134/s0006297920070044] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
14
|
Sorokin M, Kholodenko I, Kalinovsky D, Shamanskaya T, Doronin I, Konovalov D, Mironov A, Kuzmin D, Nikitin D, Deyev S, Buzdin A, Kholodenko R. RNA Sequencing-Based Identification of Ganglioside GD2-Positive Cancer Phenotype. Biomedicines 2020; 8:E142. [PMID: 32486168 PMCID: PMC7344710 DOI: 10.3390/biomedicines8060142] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/20/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
The tumor-associated ganglioside GD2 represents an attractive target for cancer immunotherapy. GD2-positive tumors are more responsive to such targeted therapy, and new methods are needed for the screening of GD2 molecular tumor phenotypes. In this work, we built a gene expression-based binary classifier predicting the GD2-positive tumor phenotypes. To this end, we compared RNA sequencing data from human tumor biopsy material from experimental samples and public databases as well as from GD2-positive and GD2-negative cancer cell lines, for expression levels of genes encoding enzymes involved in ganglioside biosynthesis. We identified a 2-gene expression signature combining ganglioside synthase genes ST8SIA1 and B4GALNT1 that serves as a more efficient predictor of GD2-positive phenotype (Matthews Correlation Coefficient (MCC) 0.32, 0.88, and 0.98 in three independent comparisons) compared to the individual ganglioside biosynthesis genes (MCC 0.02-0.32, 0.1-0.75, and 0.04-1 for the same independent comparisons). No individual gene showed a higher MCC score than the expression signature MCC score in two or more comparisons. Our diagnostic approach can hopefully be applied for pan-cancer prediction of GD2 phenotypes using gene expression data.
Collapse
Affiliation(s)
- Maxim Sorokin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
- Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., 119992 Moscow, Russia
- Omicsway Corp., 340 S Lemon Ave, 6040, Walnut, CA 91789, USA
| | - Irina Kholodenko
- Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., 119121 Moscow, Russia;
| | - Daniel Kalinovsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
| | - Tatyana Shamanskaya
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., 117997 Moscow, Russia; (T.S.); (D.K.)
| | - Igor Doronin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
- Real Target LLC, 108841 Moscow, Russia
| | - Dmitry Konovalov
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., 117997 Moscow, Russia; (T.S.); (D.K.)
| | - Aleksei Mironov
- Skolkovo Institute of Science and Technology, 3, Nobelya St., 121205 Moscow, Russia;
| | - Denis Kuzmin
- Moscow Institute of Physics and Technology (National Research University), 141700 Moscow, Russia;
| | - Daniil Nikitin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
| | - Sergey Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
- Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., 119992 Moscow, Russia
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
- Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., 119992 Moscow, Russia
- Moscow Institute of Physics and Technology (National Research University), 141700 Moscow, Russia;
- Oncobox ltd., 121205 Moscow, Russia
| | - Roman Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
- Real Target LLC, 108841 Moscow, Russia
| |
Collapse
|
15
|
Intratumoral Heterogeneity and Longitudinal Changes in Gene Expression Predict Differential Drug Sensitivity in Newly Diagnosed and Recurrent Glioblastoma. Cancers (Basel) 2020; 12:cancers12020520. [PMID: 32102350 PMCID: PMC7072286 DOI: 10.3390/cancers12020520] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Inevitable recurrence after radiochemotherapy is the major problem in the treatment of glioblastoma, the most prevalent type of adult brain malignancy. Glioblastomas are notorious for a high degree of intratumor heterogeneity manifest through a diversity of cell types and molecular patterns. The current paradigm of understanding glioblastoma recurrence is that cytotoxic therapy fails to target effectively glioma stem cells. Recent advances indicate that therapy-driven molecular evolution is a fundamental trait associated with glioblastoma recurrence. There is a growing body of evidence indicating that intratumor heterogeneity, longitudinal changes in molecular biomarkers and specific impacts of glioma stem cells need to be taken into consideration in order to increase the accuracy of molecular diagnostics still relying on readouts obtained from a single tumor specimen. Methods: This study integrates a multisampling strategy, longitudinal approach and complementary transcriptomic investigations in order to identify transcriptomic traits of recurrent glioblastoma in whole-tissue specimens of glioblastoma or glioblastoma stem cells. In this study, 128 tissue samples of 44 tumors including 23 first diagnosed, 19 recurrent and 2 secondary recurrent glioblastomas were analyzed along with 27 primary cultures of glioblastoma stem cells by RNA sequencing. A novel algorithm was used to quantify longitudinal changes in pathway activities and model efficacy of anti-cancer drugs based on gene expression data. Results: Our study reveals that intratumor heterogeneity of gene expression patterns is a fundamental characteristic of not only newly diagnosed but also recurrent glioblastomas. Evidence is provided that glioblastoma stem cells recapitulate intratumor heterogeneity, longitudinal transcriptomic changes and drug sensitivity patterns associated with the state of recurrence. Conclusions: Our results provide a transcriptional rationale for the lack of significant therapeutic benefit from temozolomide in patients with recurrent glioblastoma. Our findings imply that the spectrum of potentially effective drugs is likely to differ between newly diagnosed and recurrent glioblastomas and underscore the merits of glioblastoma stem cells as prognostic models for identifying alternative drugs and predicting drug response in recurrent glioblastoma. With the majority of recurrent glioblastomas being inoperable, glioblastoma stem cell models provide the means of compensating for the limited availability of recurrent glioblastoma specimens.
Collapse
|
16
|
Mutation Enrichment and Transcriptomic Activation Signatures of 419 Molecular Pathways in Cancer. Cancers (Basel) 2020; 12:cancers12020271. [PMID: 31979117 PMCID: PMC7073226 DOI: 10.3390/cancers12020271] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/13/2022] Open
Abstract
Carcinogenesis is linked with massive changes in regulation of gene networks. We used high throughput mutation and gene expression data to interrogate involvement of 278 signaling, 72 metabolic, 48 DNA repair and 47 cytoskeleton molecular pathways in cancer. Totally, we analyzed 4910 primary tumor samples with individual cancer RNA sequencing and whole exome sequencing profiles including ~1.3 million DNA mutations and representing thirteen cancer types. Gene expression in cancers was compared with the corresponding 655 normal tissue profiles. For the first time, we calculated mutation enrichment values and activation levels for these pathways. We found that pathway activation profiles were largely congruent among the different cancer types. However, we observed no correlation between mutation enrichment and expression changes both at the gene and at the pathway levels. Overall, positive median cancer-specific activation levels were seen in the DNA repair, versus similar slightly negative values in the other types of pathways. The DNA repair pathways also demonstrated the highest values of mutation enrichment. However, the signaling and cytoskeleton pathways had the biggest proportions of representatives among the outstandingly frequently mutated genes thus suggesting their initiator roles in carcinogenesis and the auxiliary/supporting roles for the other groups of molecular pathways.
Collapse
|
17
|
Prediction of Drug Efficacy in Colon Cancer Preclinical Models Using a Novel Ranking Method of Gene Expression. Cancers (Basel) 2020; 12:cancers12010149. [PMID: 31936310 PMCID: PMC7016638 DOI: 10.3390/cancers12010149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/31/2019] [Accepted: 01/06/2020] [Indexed: 01/03/2023] Open
Abstract
The presence of stromal cells in tumors is altering the significance of molecular profiling when using standard methods of gene expression quantification. We developed a novel normalization method to rank target gene expression in tumor samples by comparisons with reference samples representing the different cell types found in a tumor. The score for each target gene obtained after normalization, is aimed to be predictive of targeted therapies efficiency. We performed this qPCR analysis on human colorectal cancers to demonstrate the importance of reference samples to obtain accurate data and on a collection of patient-derived xenografted (PDX) colon tumors treated with Cetuximab (anti-EGFR) to demonstrate that the calculated EGFR score is predictive of Cetuximab efficacy. Interestingly, the score allowed to select an efficient treatment in a PDX model refractory to standard of care. This method is opening a novel way to predict targeted therapy efficiency which could be extended to several tumor types, and to unlimited target genes.
Collapse
|
18
|
Tkachev V, Sorokin M, Garazha A, Borisov N, Buzdin A. Oncobox Method for Scoring Efficiencies of Anticancer Drugs Based on Gene Expression Data. Methods Mol Biol 2020; 2063:235-255. [PMID: 31667774 DOI: 10.1007/978-1-0716-0138-9_17] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We describe here the Oncobox method for scoring efficiencies of anticancer target drugs (ATDs) using high throughput gene expression data. The method rationale, design, and validation are given along with the examples of its practical applications in biomedicine. The method is based on the analysis of intracellular molecular pathways activation and measuring expressions of molecular target genes for every ATD under consideration. Using Oncobox method requires collection of normal (control) expression profiles and annotated databases of molecular pathways and drug target genes. Both microarray and RNA sequencing profiles are acceptable, although the latter type of data prevails in the most recent applications of this technique.
Collapse
Affiliation(s)
| | - Maxim Sorokin
- Omicsway Corp., Walnut, CA, USA
- Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Nicolas Borisov
- Omicsway Corp., Walnut, CA, USA
- Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anton Buzdin
- Omicsway Corp., Walnut, CA, USA.
- Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.
| |
Collapse
|
19
|
Zolotovskaia M, Sorokin M, Garazha A, Borisov N, Buzdin A. Molecular Pathway Analysis of Mutation Data for Biomarkers Discovery and Scoring of Target Cancer Drugs. Methods Mol Biol 2020; 2063:207-234. [PMID: 31667773 DOI: 10.1007/978-1-0716-0138-9_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
DNA mutations govern cancer development. Cancer mutation profiles vary dramatically among the individuals. In some cases, they may serve as the predictors of disease progression and response to therapies. However, the biomarker potential of cancer mutations can be dramatically (several orders of magnitude) enhanced by applying molecular pathway-based approach. We developed Oncobox system for calculation of pathway instability (PI) values for the molecular pathways that are aggregated mutation frequencies of the pathway members normalized on gene lengths and on number of genes in the pathway. PI scores can be effective biomarkers in different types of comparisons, for example, as the cancer type biomarkers and as the predictors of tumor response to target therapies. The latter option is implemented using mutation drug score (MDS) values, which algorithmically rank the drugs capacity of interfering with the mutated molecular pathways. Here, describe the mathematical basis and algorithms for PI and MDS values calculation, validation and implementation. The example analysis is provided encompassing 5956 human tumor mutation profiles of 15 cancer types from The Cancer Genome Atlas (TCGA) project, that totally make 2,316,670 mutations in 19,872 genes and 1748 molecular pathways, thus enabling ranking of 128 clinically approved target drugs. Our results evidence that the Oncobox PI and MDS approaches are highly useful for basic and applied aspects of molecular oncology and pharmacology research.
Collapse
Affiliation(s)
- Marianna Zolotovskaia
- Omicsway Corp., Walnut, CA, USA
- Department of Oncology, Hematology and Radiotherapy of Pediatric Faculty, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Maxim Sorokin
- Omicsway Corp., Walnut, CA, USA
- Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | | | - Nikolay Borisov
- Omicsway Corp., Walnut, CA, USA
- Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anton Buzdin
- Omicsway Corp., Walnut, CA, USA.
- Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.
| |
Collapse
|
20
|
Koga Y, Ochiai A. Systematic Review of Patient-Derived Xenograft Models for Preclinical Studies of Anti-Cancer Drugs in Solid Tumors. Cells 2019; 8:cells8050418. [PMID: 31064068 PMCID: PMC6562882 DOI: 10.3390/cells8050418] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 04/26/2019] [Accepted: 05/04/2019] [Indexed: 01/06/2023] Open
Abstract
Patient-derived xenograft (PDX) models are used as powerful tools for understanding cancer biology in PDX clinical trials and co-clinical trials. In this systematic review, we focus on PDX clinical trials or co-clinical trials for drug development in solid tumors and summarize the utility of PDX models in the development of anti-cancer drugs, as well as the challenges involved in this approach, following the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. Recently, the assessment of drug efficacy by PDX clinical and co-clinical trials has become an important method. PDX clinical trials can be used for the development of anti-cancer drugs before clinical trials, with their efficacy assessed by the modified response evaluation criteria in solid tumors (mRECIST). A few dozen cases of PDX models have completed enrollment, and the efficacy of the drugs is assessed by 1 × 1 × 1 or 3 × 1 × 1 approaches in the PDX clinical trials. Furthermore, co-clinical trials can be used for personalized care or precision medicine with the evaluation of a new drug or a novel combination. Several PDX models from patients in clinical trials have been used to assess the efficacy of individual drugs or drug combinations in co-clinical trials.
Collapse
Affiliation(s)
- Yoshikatsu Koga
- Department of Strategic Programs, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan.
| | - Atsushi Ochiai
- Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan.
| |
Collapse
|
21
|
Zolotovskaia MA, Sorokin MI, Roumiantsev SA, Borisov NM, Buzdin AA. Pathway Instability Is an Effective New Mutation-Based Type of Cancer Biomarkers. Front Oncol 2019; 8:658. [PMID: 30662873 PMCID: PMC6328788 DOI: 10.3389/fonc.2018.00658] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/12/2018] [Indexed: 01/20/2023] Open
Abstract
DNA mutations play a crucial role in cancer development and progression. Mutation profiles vary dramatically in different cancer types and between individual tumors. Mutations of several individual genes are known as reliable cancer biomarkers, although the number of such genes is tiny and does not enable differential diagnostics for most of the cancers. We report here a technique enabling dramatically increased efficiency of cancer biomarkers development using DNA mutations data. It includes a quantitative metric termed Pathway instability (PI) based on mutations enrichment of intracellular molecular pathways. This method was tested on 5,956 tumor mutation profiles of 15 cancer types from The Cancer Genome Atlas (TCGA) project. Totally, we screened 2,316,670 mutations in 19,872 genes and 1,748 molecular pathways. Our results demonstrated considerable advantage of pathway-based mutation biomarkers over individual gene mutation profiles, as reflected by more than two orders of magnitude greater numbers by high-quality [ROC area-under-curve (AUC)>0.75] biomarkers. For example, the number of such high-quality mutational biomarkers distinguishing between different cancer types was only six for the individual gene mutations, and already 660 for the pathway-based biomarkers. These results evidence that PI value can be used as a new generation of complex cancer biomarkers significantly outperforming the existing gene mutation biomarkers.
Collapse
Affiliation(s)
- Marianna A Zolotovskaia
- Department of Oncology, Hematology and Radiotherapy of Pediatric Faculty, Pirogov Russian National Research Medical University, Moscow, Russia.,Oncobox Ltd., Moscow, Russia
| | - Maxim I Sorokin
- The Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Omicsway Corp., Walnut, CA, United States
| | - Sergey A Roumiantsev
- Department of Oncology, Hematology and Radiotherapy of Pediatric Faculty, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Nikolay M Borisov
- Oncobox Ltd., Moscow, Russia.,The Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anton A Buzdin
- The Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Omicsway Corp., Walnut, CA, United States.,The Laboratory of Systems Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
22
|
Molecular pathway activation – New type of biomarkers for tumor morphology and personalized selection of target drugs. Semin Cancer Biol 2018; 53:110-124. [DOI: 10.1016/j.semcancer.2018.06.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 02/06/2023]
|
23
|
Kovalchuk A, Ilnytskyy Y, Rodriguez-Juarez R, Shpyleva S, Melnyk S, Pogribny I, Katz A, Sidransky D, Kovalchuk O, Kolb B. Chemo brain or tumor brain - that is the question: the presence of extracranial tumors profoundly affects molecular processes in the prefrontal cortex of TumorGraft mice. Aging (Albany NY) 2018; 9:1660-1676. [PMID: 28758896 PMCID: PMC5559168 DOI: 10.18632/aging.101243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 05/22/2017] [Indexed: 01/17/2023]
Abstract
Cancer chemotherapy causes numerous persistent central nervous system complications. This condition is known as chemo brain. Cognitive impairments occur even before treatment, and hence are referred to as cancer associated cognitive changes, or tumor brain. There is much yet to be learned about the mechanisms of both chemo brain and tumor brain. The frequency and timing of chemo brain and tumor brain occurrence and persistence strongly suggest they may be epigenetic in nature and associated with altered gene expression. Here we used TumorGraftTM models wherein part of a patient's tumor is removed and grafted into immune-deficient mice and conducted global gene expression and DNA methylation analysis. We show that malignant non-central nervous system tumor growth causes profound molecular alterations in the brain. Mice harbouring triple negative or progesterone positive breast cancer TumorGrafts exhibited altered gene expression, decreased levels of DNA methylation, increased levels of DNA hydroxymethylation, and oxidative stress in the prefrontal cortex. Interestingly, chemotherapy did not have any additional synergistic effects on the analyzed processes. The molecular changes observed in this study are known signs of neurodegeneration and brain aging. This study provides an important roadmap for future large-scale analysis of the molecular and cellular mechanisms of tumor brain.
Collapse
Affiliation(s)
- Anna Kovalchuk
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 6T5, Canada.,Leaders in Medicine Program, Cumming School of Medicine, University of Calgary, Calgary, T2N 1N4, Canada
| | - Yaroslav Ilnytskyy
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 6T5, Canada
| | - Rocio Rodriguez-Juarez
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 6T5, Canada
| | - Svitlana Shpyleva
- Division of Biochemical Toxicology, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stepan Melnyk
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Igor Pogribny
- Division of Biochemical Toxicology, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA
| | - Amanda Katz
- Department of Oncology, Champions Oncology, Baltimore, MD 21205, USA
| | - David Sidransky
- Department of Oncology, Champions Oncology, Baltimore, MD 21205, USA
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 6T5, Canada
| | - Bryan Kolb
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 6T5, Canada
| |
Collapse
|
24
|
Aliper A, Jellen L, Cortese F, Artemov A, Karpinsky-Semper D, Moskalev A, Swick AG, Zhavoronkov A. Towards natural mimetics of metformin and rapamycin. Aging (Albany NY) 2017; 9:2245-2268. [PMID: 29165314 PMCID: PMC5723685 DOI: 10.18632/aging.101319] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/02/2017] [Indexed: 12/14/2022]
Abstract
Aging is now at the forefront of major challenges faced globally, creating an immediate need for safe, widescale interventions to reduce the burden of chronic disease and extend human healthspan. Metformin and rapamycin are two FDA-approved mTOR inhibitors proposed for this purpose, exhibiting significant anti-cancer and anti-aging properties beyond their current clinical applications. However, each faces issues with approval for off-label, prophylactic use due to adverse effects. Here, we initiate an effort to identify nutraceuticals-safer, naturally-occurring compounds-that mimic the anti-aging effects of metformin and rapamycin without adverse effects. We applied several bioinformatic approaches and deep learning methods to the Library of Integrated Network-based Cellular Signatures (LINCS) dataset to map the gene- and pathway-level signatures of metformin and rapamycin and screen for matches among over 800 natural compounds. We then predicted the safety of each compound with an ensemble of deep neural network classifiers. The analysis revealed many novel candidate metformin and rapamycin mimetics, including allantoin and ginsenoside (metformin), epigallocatechin gallate and isoliquiritigenin (rapamycin), and withaferin A (both). Four relatively unexplored compounds also scored well with rapamycin. This work revealed promising candidates for future experimental validation while demonstrating the applications of powerful screening methods for this and similar endeavors.
Collapse
Affiliation(s)
- Alexander Aliper
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | - Leslie Jellen
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | - Franco Cortese
- Biogerontology Research Foundation, Research Department, Oxford, United Kingdom
- Department of Biomedical and Molecular Science, Queen's University School of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Artem Artemov
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | | | - Alexey Moskalev
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, 167982, Russia
| | | | - Alex Zhavoronkov
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
- Biogerontology Research Foundation, Research Department, Oxford, United Kingdom
| |
Collapse
|
25
|
Mamoshina P, Ojomoko L, Yanovich Y, Ostrovski A, Botezatu A, Prikhodko P, Izumchenko E, Aliper A, Romantsov K, Zhebrak A, Ogu IO, Zhavoronkov A. Converging blockchain and next-generation artificial intelligence technologies to decentralize and accelerate biomedical research and healthcare. Oncotarget 2017; 9:5665-5690. [PMID: 29464026 PMCID: PMC5814166 DOI: 10.18632/oncotarget.22345] [Citation(s) in RCA: 238] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/02/2017] [Indexed: 12/19/2022] Open
Abstract
The increased availability of data and recent advancements in artificial intelligence present the unprecedented opportunities in healthcare and major challenges for the patients, developers, providers and regulators. The novel deep learning and transfer learning techniques are turning any data about the person into medical data transforming simple facial pictures and videos into powerful sources of data for predictive analytics. Presently, the patients do not have control over the access privileges to their medical records and remain unaware of the true value of the data they have. In this paper, we provide an overview of the next-generation artificial intelligence and blockchain technologies and present innovative solutions that may be used to accelerate the biomedical research and enable patients with new tools to control and profit from their personal data as well with the incentives to undergo constant health monitoring. We introduce new concepts to appraise and evaluate personal records, including the combination-, time- and relationship-value of the data. We also present a roadmap for a blockchain-enabled decentralized personal health data ecosystem to enable novel approaches for drug discovery, biomarker development, and preventative healthcare. A secure and transparent distributed personal data marketplace utilizing blockchain and deep learning technologies may be able to resolve the challenges faced by the regulators and return the control over personal data including medical records back to the individuals.
Collapse
Affiliation(s)
- Polina Mamoshina
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA.,Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Lucy Ojomoko
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA
| | | | | | | | | | - Eugene Izumchenko
- Department of Otolaryngology-Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander Aliper
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA
| | - Konstantin Romantsov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA
| | - Alexander Zhebrak
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA
| | - Iraneus Obioma Ogu
- Africa Blockchain Artificial Intelligence for Healthcare Initiative, Insilico Medicine, Inc, Abuja, Nigeria
| | - Alex Zhavoronkov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA.,The Biogerontology Research Foundation, London, United Kingdom
| |
Collapse
|
26
|
Pan Y, Zhang H, Zhang M, Zhu J, Yu J, Wang B, Qiu J, Zhang J. A five-gene based risk score with high prognostic value in colorectal cancer. Oncol Lett 2017; 14:6724-6734. [PMID: 29344121 PMCID: PMC5754913 DOI: 10.3892/ol.2017.7097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 08/31/2017] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently occurring malignancies worldwide. The outcomes of patients with similar clinical symptoms or at similar pathological stages remain unpredictable. This inherent clinical diversity is most likely due to the genetic heterogeneity. The present study aimed to create a predicting tool to evaluate patient survival based on genetic profile. Firstly, three Gene Expression Omnibus (GEO) datasets (GSE9348, GSE44076 and GSE44861) were utilized to identify and validate differentially expressed genes (DEGs) in CRC. The GSE14333 dataset containing survival information was then introduced in order to screen and verify prognosis-associated genes. Of the 66 DEGs, the present study screened out 46 biomarkers closely associated to patient overall survival. By Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis, it was demonstrated that these genes participated in multiple biological processes which were highly associated with cancer proliferation, drug-resistance and metastasis, thus further affecting patient survival. The five most important genes, MET proto-oncogene, receptor tyrosine kinase, carboxypeptidase M, serine hydroxymethyltransferase 2, guanylate cyclase activator 2B and sodium voltage-gated channel a subunit 9 were selected by a random survival forests algorithm, and were further made up to a linear risk score formula by multivariable cox regression. Finally, the present study tested and verified this risk score within three independent GEO datasets (GSE14333, GSE17536 and GSE29621), and observed that patients with a high risk score had a lower overall survival (P<0.05). Furthermore, this risk score was the most significant compared with other predicting factors including age and American Joint Committee on Cancer stage, in the model, and was able to predict patient survival independently and directly. The findings suggest that this survival associated DEGs-based risk score is a powerful and accurate prognostic tool and is promisingly implemented in a clinical setting.
Collapse
Affiliation(s)
- Yida Pan
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Hongyang Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Mingming Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing 210008, P.R. China
| | - Jie Zhu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jianghong Yu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Bangting Wang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jigang Qiu
- Department of General Surgery, Huadong Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jun Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
27
|
Borisov N, Suntsova M, Sorokin M, Garazha A, Kovalchuk O, Aliper A, Ilnitskaya E, Lezhnina K, Korzinkin M, Tkachev V, Saenko V, Saenko Y, Sokov DG, Gaifullin NM, Kashintsev K, Shirokorad V, Shabalina I, Zhavoronkov A, Mishra B, Cantor CR, Buzdin A. Data aggregation at the level of molecular pathways improves stability of experimental transcriptomic and proteomic data. Cell Cycle 2017; 16:1810-1823. [PMID: 28825872 DOI: 10.1080/15384101.2017.1361068] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
High throughput technologies opened a new era in biomedicine by enabling massive analysis of gene expression at both RNA and protein levels. Unfortunately, expression data obtained in different experiments are often poorly compatible, even for the same biologic samples. Here, using experimental and bioinformatic investigation of major experimental platforms, we show that aggregation of gene expression data at the level of molecular pathways helps to diminish cross- and intra-platform bias otherwise clearly seen at the level of individual genes. We created a mathematical model of cumulative suppression of data variation that predicts the ideal parameters and the optimal size of a molecular pathway. We compared the abilities to aggregate experimental molecular data for the 5 alternative methods, also evaluated by their capacity to retain meaningful features of biologic samples. The bioinformatic method OncoFinder showed optimal performance in both tests and should be very useful for future cross-platform data analyses.
Collapse
Affiliation(s)
- Nicolas Borisov
- a Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, National Research Centre "Kurchatov Institute" , Moscow , Russia.,b Department of R&D, First Oncology Research and Advisory Center , Moscow , Russia
| | - Maria Suntsova
- c Department of R&D, Center for Biogerontology and Regenerative Medicine , Moscow , Russia.,d Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology , Moscow , Russia
| | - Maxim Sorokin
- a Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, National Research Centre "Kurchatov Institute" , Moscow , Russia.,e Group for Genomic Regulation of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry , Moscow , Russia
| | - Andrew Garazha
- c Department of R&D, Center for Biogerontology and Regenerative Medicine , Moscow , Russia.,f Department of R&D, OmicsWay Corporation , Walnut , CA , USA
| | - Olga Kovalchuk
- g Department of Biological Sciences , University of Lethbridge , Lethbridge , AB , Canada
| | - Alexander Aliper
- d Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology , Moscow , Russia
| | - Elena Ilnitskaya
- c Department of R&D, Center for Biogerontology and Regenerative Medicine , Moscow , Russia
| | - Ksenia Lezhnina
- b Department of R&D, First Oncology Research and Advisory Center , Moscow , Russia
| | - Mikhail Korzinkin
- c Department of R&D, Center for Biogerontology and Regenerative Medicine , Moscow , Russia
| | - Victor Tkachev
- f Department of R&D, OmicsWay Corporation , Walnut , CA , USA
| | - Vyacheslav Saenko
- h Technological Research Institute S.P. Kapitsa , Ulyanovsk State University , Ulyanovsk , Russia
| | - Yury Saenko
- h Technological Research Institute S.P. Kapitsa , Ulyanovsk State University , Ulyanovsk , Russia
| | - Dmitry G Sokov
- i Chemotherapy Department, Moscow 1st Oncological Hospital , Moscow , Russia
| | - Nurshat M Gaifullin
- j Faculty of Fundamental Medicine , Lomonosov Moscow State University , Moscow , Russia.,k Department of Oncology, Russian Medical Postgraduate Academy , Moscow , Russia
| | - Kirill Kashintsev
- l Chemotherapy Department, Moscow Oncological Hospital 62 , Stepanovskoye , Russia
| | - Valery Shirokorad
- l Chemotherapy Department, Moscow Oncological Hospital 62 , Stepanovskoye , Russia
| | - Irina Shabalina
- m Faculty of Mathematics and Information Technologies , Petrozavodsk State University , Petrozavodsk , Russia
| | - Alex Zhavoronkov
- d Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology , Moscow , Russia
| | | | - Charles R Cantor
- o Department of Biomedical Engineering , Boston University , Boston , MA , USA
| | - Anton Buzdin
- a Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, National Research Centre "Kurchatov Institute" , Moscow , Russia.,b Department of R&D, First Oncology Research and Advisory Center , Moscow , Russia.,e Group for Genomic Regulation of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry , Moscow , Russia.,f Department of R&D, OmicsWay Corporation , Walnut , CA , USA
| |
Collapse
|
28
|
Kadurin A, Aliper A, Kazennov A, Mamoshina P, Vanhaelen Q, Khrabrov K, Zhavoronkov A. The cornucopia of meaningful leads: Applying deep adversarial autoencoders for new molecule development in oncology. Oncotarget 2017; 8:10883-10890. [PMID: 28029644 PMCID: PMC5355231 DOI: 10.18632/oncotarget.14073] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 11/24/2016] [Indexed: 12/19/2022] Open
Abstract
Recent advances in deep learning and specifically in generative adversarial networks have demonstrated surprising results in generating new images and videos upon request even using natural language as input. In this paper we present the first application of generative adversarial autoencoders (AAE) for generating novel molecular fingerprints with a defined set of parameters. We developed a 7-layer AAE architecture with the latent middle layer serving as a discriminator. As an input and output the AAE uses a vector of binary fingerprints and concentration of the molecule. In the latent layer we also introduced a neuron responsible for growth inhibition percentage, which when negative indicates the reduction in the number of tumor cells after the treatment. To train the AAE we used the NCI-60 cell line assay data for 6252 compounds profiled on MCF-7 cell line. The output of the AAE was used to screen 72 million compounds in PubChem and select candidate molecules with potential anti-cancer properties. This approach is a proof of concept of an artificially-intelligent drug discovery engine, where AAEs are used to generate new molecular fingerprints with the desired molecular properties.
Collapse
Affiliation(s)
- Artur Kadurin
- Search Department, Mail.Ru Group Ltd., Moscow, Russia.,Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA.,Big Data and Text Analysis Laboratory, Kazan Federal University, Kazan, Republic of Tatarstan, Russia.,St. Petersburg Department of V.A. Steklov Institute of Mathematics of the Russian Academy of Sciences, Petersburg, Russia
| | - Alexander Aliper
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA
| | - Andrey Kazennov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Polina Mamoshina
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA.,Department of Computer Science, University of Oxford, Oxford, UK
| | - Quentin Vanhaelen
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA
| | | | - Alex Zhavoronkov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA.,The Biogerontology Research Foundation, Trevissome Park, Truro TR4 8UN, UK.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| |
Collapse
|
29
|
Stamatas GN, Wu J, Pappas A, Mirmirani P, McCormick TS, Cooper KD, Consolo M, Schastnaya J, Ozerov IV, Aliper A, Zhavoronkov A. An analysis of gene expression data involving examination of signaling pathways activation reveals new insights into the mechanism of action of minoxidil topical foam in men with androgenetic alopecia. Cell Cycle 2017; 16:1578-1584. [PMID: 28594262 DOI: 10.1080/15384101.2017.1327492] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Androgenetic alopecia is the most common form of hair loss. Minoxidil has been approved for the treatment of hair loss, however its mechanism of action is still not fully clarified. In this study, we aimed to elucidate the effects of 5% minoxidil topical foam on gene expression and activation of signaling pathways in vertex and frontal scalp of men with androgenetic alopecia. We identified regional variations in gene expression and perturbed signaling pathways using in silico Pathway Activation Network Decomposition Analysis (iPANDA) before and after treatment with minoxidil. Vertex and frontal scalp of patients showed a generally similar response to minoxidil. Both scalp regions showed upregulation of genes that encode keratin associated proteins and downregulation of ILK, Akt, and MAPK signaling pathways after minoxidil treatment. Our results provide new insights into the mechanism of action of minoxidil topical foam in men with androgenetic alopecia.
Collapse
Affiliation(s)
- Georgios N Stamatas
- a Emerging Science & Innovation, Johnson & Johnson Santé Beauté France , Johnson & Johnson Group of Consumer Companies , Issy-les-Moulineaux , France
| | - Jeff Wu
- b Hair Care R&D, Johnson & Johnson Consumer Worldwide , Johnson & Johnson Family of Consumer Companies, Inc. , Skillman , NJ , USA
| | - Apostolos Pappas
- c Emerging Science & Innovation, Johnson & Johnson Consumer Worldwide , Johnson & Johnson Family of Consumer Companies, Inc. , Skillman , NJ , USA
| | - Paradi Mirmirani
- d Department of Dermatology , The Permanente Medical Group , Vallejo , CA , USA.,e Department of Dermatology , University of California , San Francisco , CA , USA.,f Department of Dermatology , Case Western Reserve University, University Hospitals Cleveland Medical Center , Cleveland , OH , USA
| | - Thomas S McCormick
- f Department of Dermatology , Case Western Reserve University, University Hospitals Cleveland Medical Center , Cleveland , OH , USA
| | - Kevin D Cooper
- f Department of Dermatology , Case Western Reserve University, University Hospitals Cleveland Medical Center , Cleveland , OH , USA
| | - Mary Consolo
- f Department of Dermatology , Case Western Reserve University, University Hospitals Cleveland Medical Center , Cleveland , OH , USA
| | - Jane Schastnaya
- g Insilico Medicine, Inc., Emerging Technology Centers , Johns Hopkins University at Eastern , Baltimore , MD , USA
| | - Ivan V Ozerov
- g Insilico Medicine, Inc., Emerging Technology Centers , Johns Hopkins University at Eastern , Baltimore , MD , USA
| | - Alexander Aliper
- g Insilico Medicine, Inc., Emerging Technology Centers , Johns Hopkins University at Eastern , Baltimore , MD , USA
| | - Alex Zhavoronkov
- g Insilico Medicine, Inc., Emerging Technology Centers , Johns Hopkins University at Eastern , Baltimore , MD , USA
| |
Collapse
|
30
|
Ozawa H, Ranaweera RS, Izumchenko E, Makarev E, Zhavoronkov A, Fertig EJ, Howard JD, Markovic A, Bedi A, Ravi R, Perez J, Le QT, Kong CS, Jordan RC, Wang H, Kang H, Quon H, Sidransky D, Chung CH. SMAD4 Loss Is Associated with Cetuximab Resistance and Induction of MAPK/JNK Activation in Head and Neck Cancer Cells. Clin Cancer Res 2017; 23:5162-5175. [PMID: 28522603 DOI: 10.1158/1078-0432.ccr-16-1686] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 12/01/2016] [Accepted: 05/15/2017] [Indexed: 01/01/2023]
Abstract
Purpose: We previously demonstrated an association between decreased SMAD4 expression and cetuximab resistance in head and neck squamous cell carcinoma (HNSCC). The purpose of this study was to further elucidate the clinical relevance of SMAD4 loss in HNSCC.Experimental Design: SMAD4 expression was assessed by IHC in 130 newly diagnosed and 43 patients with recurrent HNSCC. Correlative statistical analysis with clinicopathologic data was also performed. OncoFinder, a bioinformatics tool, was used to analyze molecular signaling in TCGA tumors with low or high SMAD4 mRNA levels. The role of SMAD4 was investigated by shRNA knockdown and gene reconstitution of HPV-negative HNSCC cell lines in vitro and in vivoResults: Our analysis revealed that SMAD4 loss was associated with an aggressive, HPV-negative, cetuximab-resistant phenotype. We found a signature of prosurvival and antiapoptotic pathways that were commonly dysregulated in SMAD4-low cases derived from TCGA-HNSCC dataset and an independent oral cavity squamous cell carcinoma (OSCC) cohort obtained from GEO. We show that SMAD4 depletion in an HNSCC cell line induces cetuximab resistance and results in worse survival in an orthotopic mouse model in vivo We implicate JNK and MAPK activation as mediators of cetuximab resistance and provide the foundation for the concomitant EGFR and JNK/MAPK inhibition as a potential strategy for overcoming cetuximab resistance in HNSCCs with SMAD4 loss.Conclusions: Our study demonstrates that loss of SMAD4 expression is a signature characterizing the cetuximab-resistant phenotype and suggests that SMAD4 expression may be a determinant of sensitivity/resistance to EGFR/MAPK or EGFR/JNK inhibition in HPV-negative HNSCC tumors. Clin Cancer Res; 23(17); 5162-75. ©2017 AACR.
Collapse
Affiliation(s)
- Hiroyuki Ozawa
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Ruchira S Ranaweera
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Evgeny Izumchenko
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eugene Makarev
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, Maryland
| | - Alex Zhavoronkov
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, Maryland
| | - Elana J Fertig
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
- Department of Health Science Informatics, Johns Hopkins University, Baltimore, Maryland
| | - Jason D Howard
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Ana Markovic
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Atul Bedi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rajani Ravi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jimena Perez
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Quynh-Thu Le
- Department of Pathology, Stanford University School of Medicine Stanford, California
| | - Christina S Kong
- Department of Pathology, Stanford University School of Medicine Stanford, California
| | - Richard C Jordan
- Departments of Orofacial Sciences and Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Hao Wang
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Hyunseok Kang
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Harry Quon
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine H Chung
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
31
|
Zhavoronkov A, Izumchenko E, Kanherkar RR, Teka M, Cantor C, Manaye K, Sidransky D, West MD, Makarev E, Csoka AB. Pro-fibrotic pathway activation in trabecular meshwork and lamina cribrosa is the main driving force of glaucoma. Cell Cycle 2017; 15:1643-52. [PMID: 27229292 PMCID: PMC4934076 DOI: 10.1080/15384101.2016.1170261] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
While primary open-angle glaucoma (POAG) is a leading cause of blindness worldwide, it still does not have a clear mechanism that can explain all clinical cases of the disease. Elevated IOP is associated with increased accumulation of extracellular matrix (ECM) proteins in the trabecular meshwork (TM) that prevents normal outflow of aqueous humor (AH) and has damaging effects on the fine mesh-like lamina cribrosa (LC) through which the optic nerve fibers pass. Applying a pathway analysis algorithm, we discovered that an elevated level of TGFβ observed in glaucoma-affected tissues could lead to pro-fibrotic pathway activation in TM and in LC. In turn, activated pro-fibrotic pathways lead to ECM remodeling in TM and LC, making TM less efficient in AH drainage and making LC more susceptible to damage from elevated IOP via ECM transformation in LC. We propose pathway targets for potential therapeutic interventions to delay or avoid fibrosis initiation in TM and LC tissues.
Collapse
Affiliation(s)
- Alex Zhavoronkov
- a Insilico Medicine, Inc., ETC, Johns Hopkins University , Baltimore , MD , USA.,b The Biogerontology Research Foundation , London , UK
| | - Evgeny Izumchenko
- e Johns Hopkins University , Department of Otolaryngology-Head and Neck Surgery
| | - Riya R Kanherkar
- c Vision Genomics, LLC , Washington, DC , USA.,d Epigenetics Laboratory, Howard University , Washington, DC , USA
| | - Mahder Teka
- c Vision Genomics, LLC , Washington, DC , USA
| | - Charles Cantor
- f Boston University , Boston , MA , USA.,g Retrotope, Inc ; Los Altos Hills , CA , USA
| | - Kebreten Manaye
- d Epigenetics Laboratory, Howard University , Washington, DC , USA
| | | | | | - Eugene Makarev
- a Insilico Medicine, Inc., ETC, Johns Hopkins University , Baltimore , MD , USA
| | - Antonei Benjamin Csoka
- c Vision Genomics, LLC , Washington, DC , USA.,d Epigenetics Laboratory, Howard University , Washington, DC , USA
| |
Collapse
|
32
|
Brijwani N, Jain M, Dhandapani M, Zahed F, Mukhopadhyay P, Biswas M, Khatri D, Radhakrishna VD, Majumder B, Radhakrishnan P, Thiyagarajan S. Rationally co-targeting divergent pathways in KRAS wild-type colorectal cancers by CANscript technology reveals tumor dependence on Notch and Erbb2. Sci Rep 2017; 7:1502. [PMID: 28473715 PMCID: PMC5431418 DOI: 10.1038/s41598-017-01566-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 03/29/2017] [Indexed: 02/07/2023] Open
Abstract
KRAS mutation status can distinguish between metastatic colorectal carcinoma (mCRC) patients who may benefit from therapies that target the epidermal growth factor receptor (EGFR), such as cetuximab. However, patients whose tumors harbor mutant KRAS (codons 12/13, 61 and 146) are often excluded from EGFR-targeted regimens, while other patients with wild type KRAS will sometimes respond favorably to these same drugs. These conflicting observations suggest that a more robust approach to individualize therapy may enable greater frequency of positive clinical outcome for mCRC patients. Here, we utilized alive tumor tissues in ex-vivo platform termed CANscript, which preserves the native tumor heterogeneity, in order to interrogate the antitumor effects of EGFR-targeted drugs in mCRC (n = 40). We demonstrated that, irrespective of KRAS status, cetuximab did not induce an antitumor response in a majority of patient tumors. In the subset of non-responsive tumors, data showed that expression levels of EGFR ligands contributed to a mechanism of resistance. Transcriptomic and phosphoproteomic profiling revealed deregulation of multiple pathways, significantly the Notch and Erbb2. Targeting these nodes concurrently resulted in antitumor efficacy in a majority of cetuximab-resistant tumors. These findings highlight the importance of integrating molecular profile and functional testing tools for optimization of alternate strategies in resistant population.
Collapse
Affiliation(s)
- Nilesh Brijwani
- Division of Molecular Profiling, Mitra Biotech, Bangalore, Karnataka, 560099, India.,Symbiosis International University (SIU), Lavale, Mulshi Taluka, Pune, Maharashtra, 412115, India
| | - Misti Jain
- Division of Molecular Profiling, Mitra Biotech, Bangalore, Karnataka, 560099, India
| | - Muthu Dhandapani
- Division of Cancer Biology, Mitra Biotech, Bangalore, Karnataka, 560099, India
| | - Farrah Zahed
- Division of Molecular Profiling, Mitra Biotech, Bangalore, Karnataka, 560099, India
| | | | - Manjusha Biswas
- Division of Molecular Pathology, Mitra Biotech, Bangalore, Karnataka, 560099, India
| | - Deepak Khatri
- Division of Cancer Biology, Mitra Biotech, Bangalore, Karnataka, 560099, India
| | - Vinod D Radhakrishna
- Division of Oncology Pharmacology, Mitra Biotech, Bangalore, Karnataka, 560099, India
| | - Biswanath Majumder
- Division of Molecular Pathology, Mitra Biotech, Bangalore, Karnataka, 560099, India
| | | | - Saravanan Thiyagarajan
- Division of Cancer Biology, Mitra Biotech, Bangalore, Karnataka, 560099, India. .,Division of Oncology Pharmacology, Mitra Biotech, Bangalore, Karnataka, 560099, India.
| |
Collapse
|
33
|
Buzdin AA, Prassolov V, Zhavoronkov AA, Borisov NM. Bioinformatics Meets Biomedicine: OncoFinder, a Quantitative Approach for Interrogating Molecular Pathways Using Gene Expression Data. Methods Mol Biol 2017; 1613:53-83. [PMID: 28849558 DOI: 10.1007/978-1-4939-7027-8_4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We propose a biomathematical approach termed OncoFinder (OF) that enables performing both quantitative and qualitative analyses of the intracellular molecular pathway activation. OF utilizes an algorithm that distinguishes the activator/repressor role of every gene product in a pathway. This method is applicable for the analysis of any physiological, stress, malignancy, and other conditions at the molecular level. OF showed a strong potential to neutralize background-caused differences between experimental gene expression data obtained using NGS, microarray and modern proteomics techniques. Importantly, in most cases, pathway activation signatures were better markers of cancer progression compared to the individual gene products. OF also enables correlating pathway activation with the success of anticancer therapy for individual patients. We further expanded this approach to analyze impact of micro RNAs (miRs) on the regulation of cellular interactome. Many alternative sources provide information about miRs and their targets. However, instruments elucidating higher level impact of the established total miR profiles are still largely missing. A variant of OncoFinder termed MiRImpact enables linking miR expression data with its estimated outcome on the regulation of molecular processes, such as signaling, metabolic, cytoskeleton, and DNA repair pathways. MiRImpact was used to establish cancer-specific and cytomegaloviral infection-linked interactomic signatures for hundreds of molecular pathways. Interestingly, the impact of miRs appeared orthogonal to pathway regulation at the mRNA level, which stresses the importance of combining all available levels of gene regulation to build a more objective molecular model of cell.
Collapse
Affiliation(s)
- Anton A Buzdin
- Pathway Pharmaceuticals, Wan Chai, Hong Kong SAR.
- Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, National Research Centre "Kurchatov Institute", Bldg 140, Suite 415, 1, Akademika Kurchatova sq., Moscow, 123182, Russia.
- Group for Genomic Regulation of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia.
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.
| | - Vladimir Prassolov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova street 32, Mosow, 119991, Russia
| | - Alex A Zhavoronkov
- Pathway Pharmaceuticals, Wan Chai, Hong Kong SAR
- Group for Genomic Regulation of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Nikolay M Borisov
- Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, National Research Centre "Kurchatov Institute", Bldg 140, Suite 415, 1, Akademika Kurchatova sq., Moscow, 123182, Russia
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia
| |
Collapse
|
34
|
Aliper AM, Korzinkin MB, Kuzmina NB, Zenin AA, Venkova LS, Smirnov PY, Zhavoronkov AA, Buzdin AA, Borisov NM. Mathematical Justification of Expression-Based Pathway Activation Scoring (PAS). Methods Mol Biol 2017; 1613:31-51. [PMID: 28849557 DOI: 10.1007/978-1-4939-7027-8_3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Although modeling of activation kinetics for various cell signaling pathways has reached a high grade of sophistication and thoroughness, most such kinetic models still remain of rather limited practical value for biomedicine. Nevertheless, recent advancements have been made in application of signaling pathway science for real needs of prescription of the most effective drugs for individual patients. The methods for such prescription evaluate the degree of pathological changes in the signaling machinery based on two types of data: first, on the results of high-throughput gene expression profiling, and second, on the molecular pathway graphs that reflect interactions between the pathway members. For example, our algorithm OncoFinder evaluates the activation of molecular pathways on the basis of gene/protein expression data in the objects of the interest.Yet, the question of assessment of the relative importance for each gene product in a molecular pathway remains unclear unless one call for the methods of parameter sensitivity /stiffness analysis in the interactomic kinetic models of signaling pathway activation in terms of total concentrations of each gene product.Here we show two principal points: 1. First, the importance coefficients for each gene in pathways that were obtained using the extremely time- and labor-consuming stiffness analysis of full-scaled kinetic models generally differ from much easier-to-calculate expression-based pathway activation score (PAS) not more than by 30%, so the concept of PAS is kinetically justified. 2. Second, the use of pathway-based approach instead of distinct gene analysis, due to the law of large numbers, allows restoring the correlation between the similar samples that were examined using different transcriptome investigation techniques.
Collapse
Affiliation(s)
- Alexander M Aliper
- Drug Research and Design Department, Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Michael B Korzinkin
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Natalia B Kuzmina
- Laboratory of Systems Biology, A.I. Burnazyan Federal Medical Biophysical Center, Moscow, 123182, Russia
| | - Alexander A Zenin
- Laboratory of Systems Biology, A.I. Burnazyan Federal Medical Biophysical Center, Moscow, 123182, Russia
| | - Larisa S Venkova
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Philip Yu Smirnov
- Laboratory of Systems Biology, A.I. Burnazyan Federal Medical Biophysical Center, Moscow, 123182, Russia
| | - Alex A Zhavoronkov
- Drug Research and Design Department, Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Anton A Buzdin
- Drug Research and Design Department, Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Group for Genomic Regulation of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- National Research Centre "Kurchatov Institute", Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, Moscow, Russia
| | - Nikolay M Borisov
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia.
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.
- National Research Centre "Kurchatov Institute", Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, Moscow, Russia.
| |
Collapse
|
35
|
Artemov A, Aliper A, Korzinkin M, Lezhnina K, Jellen L, Zhukov N, Roumiantsev S, Gaifullin N, Zhavoronkov A, Borisov N, Buzdin A. A method for predicting target drug efficiency in cancer based on the analysis of signaling pathway activation. Oncotarget 2016; 6:29347-56. [PMID: 26320181 PMCID: PMC4745731 DOI: 10.18632/oncotarget.5119] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 07/24/2015] [Indexed: 02/07/2023] Open
Abstract
A new generation of anticancer therapeutics called target drugs has quickly developed in the 21st century. These drugs are tailored to inhibit cancer cell growth, proliferation, and viability by specific interactions with one or a few target proteins. However, despite formally known molecular targets for every "target" drug, patient response to treatment remains largely individual and unpredictable. Choosing the most effective personalized treatment remains a major challenge in oncology and is still largely trial and error. Here we present a novel approach for predicting target drug efficacy based on the gene expression signature of the individual tumor sample(s). The enclosed bioinformatic algorithm detects activation of intracellular regulatory pathways in the tumor in comparison to the corresponding normal tissues. According to the nature of the molecular targets of a drug, it predicts whether the drug can prevent cancer growth and survival in each individual case by blocking the abnormally activated tumor-promoting pathways or by reinforcing internal tumor suppressor cascades. To validate the method, we compared the distribution of predicted drug efficacy scores for five drugs (Sorafenib, Bevacizumab, Cetuximab, Sorafenib, Imatinib, Sunitinib) and seven cancer types (Clear Cell Renal Cell Carcinoma, Colon cancer, Lung adenocarcinoma, non-Hodgkin Lymphoma, Thyroid cancer and Sarcoma) with the available clinical trials data for the respective cancer types and drugs. The percent of responders to a drug treatment correlated significantly (Pearson's correlation 0.77 p = 0.023) with the percent of tumors showing high drug scores calculated with the current algorithm.
Collapse
Affiliation(s)
- Artem Artemov
- Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR.,D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexander Aliper
- D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.,First Oncology Research and Advisory Center, Moscow, Russia
| | | | | | - Leslie Jellen
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nikolay Zhukov
- D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.,First Oncology Research and Advisory Center, Moscow, Russia.,Pirogov Russian National Research Medical University, Department of Oncology, Hematology and Radiotherapy, Moscow, Russia
| | - Sergey Roumiantsev
- D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.,Pirogov Russian National Research Medical University, Department of Oncology, Hematology and Radiotherapy, Moscow, Russia
| | - Nurshat Gaifullin
- Moscow State University, Faculty of Fundamental Medicine, Moscow, Russia
| | - Alex Zhavoronkov
- Insilico Medicine, Inc., ETC, Johns Hopkins University, Baltimore, MD, USA
| | | | - Anton Buzdin
- Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR.,D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.,Group for Genomic Regulation of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
36
|
Makarev E, Fortney K, Litovchenko M, Braunewell KH, Zhavoronkov A, Atala A. Quantifying signaling pathway activation to monitor the quality of induced pluripotent stem cells. Oncotarget 2016; 6:23204-12. [PMID: 26327604 PMCID: PMC4695112 DOI: 10.18632/oncotarget.4673] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/10/2015] [Indexed: 11/25/2022] Open
Abstract
Many attempts have been made to evaluate the safety and potency of human induced pluripotent stem cells (iPSCs) for clinical applications using transcriptome data, but results so far have been ambiguous or even contradictory. Here, we characterized stem cells at the pathway level, rather than at the gene level as has been the focus of previous work. We meta-analyzed publically-available gene expression data sets and evaluated signaling and metabolic pathway activation profiles for 20 human embryonic stem cell (ESC) lines, 12 human iPSC lines, five embryonic body lines, and six fibroblast cell lines. We demonstrated the close resemblance of iPSCs with ESCs at the pathway level, and provided examples of how pathway activity can be applied to identify iPSC line abnormalities or to predict in vitro differentiation potential. Our results indicate that pathway activation profiling is a promising strategy for evaluating the safety and potency of iPSC lines in translational medicine applications.
Collapse
Affiliation(s)
- Eugene Makarev
- Atlas Regeneration, Inc, Winston-Salem, NC, USA.,Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD, USA
| | - Kristen Fortney
- Atlas Regeneration, Inc, Winston-Salem, NC, USA.,Department of Developmental Biology, Stanford University Medical Center, Stanford, CA, USA
| | - Maria Litovchenko
- Department of Computational Genomics, Ludwig Maximilian University of Munich, Germany
| | - Karl H Braunewell
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Germany
| | - Alex Zhavoronkov
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD, USA.,The Biogerontology Research Foundation, London, UK
| | - Anthony Atala
- Atlas Regeneration, Inc, Winston-Salem, NC, USA.,Department of Urology, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
37
|
Aliper A, Plis S, Artemov A, Ulloa A, Mamoshina P, Zhavoronkov A. Deep Learning Applications for Predicting Pharmacological Properties of Drugs and Drug Repurposing Using Transcriptomic Data. Mol Pharm 2016; 13:2524-30. [PMID: 27200455 DOI: 10.1021/acs.molpharmaceut.6b00248] [Citation(s) in RCA: 282] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Deep learning is rapidly advancing many areas of science and technology with multiple success stories in image, text, voice and video recognition, robotics, and autonomous driving. In this paper we demonstrate how deep neural networks (DNN) trained on large transcriptional response data sets can classify various drugs to therapeutic categories solely based on their transcriptional profiles. We used the perturbation samples of 678 drugs across A549, MCF-7, and PC-3 cell lines from the LINCS Project and linked those to 12 therapeutic use categories derived from MeSH. To train the DNN, we utilized both gene level transcriptomic data and transcriptomic data processed using a pathway activation scoring algorithm, for a pooled data set of samples perturbed with different concentrations of the drug for 6 and 24 hours. In both pathway and gene level classification, DNN achieved high classification accuracy and convincingly outperformed the support vector machine (SVM) model on every multiclass classification problem, however, models based on pathway level data performed significantly better. For the first time we demonstrate a deep learning neural net trained on transcriptomic data to recognize pharmacological properties of multiple drugs across different biological systems and conditions. We also propose using deep neural net confusion matrices for drug repositioning. This work is a proof of principle for applying deep learning to drug discovery and development.
Collapse
Affiliation(s)
- Alexander Aliper
- Insilico Medicine, ETC, B301, Johns Hopkins University , Baltimore, Maryland 21218, United States
| | - Sergey Plis
- Datalytic Solutions , 1101 Yale Boulevard NE, Albuquerque, New Mexico 87106, United States.,The Mind Research Network , Albuquerque, New Mexico 87106, United States
| | - Artem Artemov
- Insilico Medicine, ETC, B301, Johns Hopkins University , Baltimore, Maryland 21218, United States
| | - Alvaro Ulloa
- The Mind Research Network , Albuquerque, New Mexico 87106, United States
| | - Polina Mamoshina
- Insilico Medicine, ETC, B301, Johns Hopkins University , Baltimore, Maryland 21218, United States
| | - Alex Zhavoronkov
- Insilico Medicine, ETC, B301, Johns Hopkins University , Baltimore, Maryland 21218, United States.,The Biogerontology Research Foundation , Oxford, U.K
| |
Collapse
|
38
|
Makarev E, Izumchenko E, Aihara F, Wysocki PT, Zhu Q, Buzdin A, Sidransky D, Zhavoronkov A, Atala A. Common pathway signature in lung and liver fibrosis. Cell Cycle 2016; 15:1667-73. [PMID: 27267766 PMCID: PMC4957589 DOI: 10.1080/15384101.2016.1152435] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Fibrosis, a progressive accumulation of extracellular matrix components, encompasses a wide spectrum of distinct organs, and accounts for an increasing burden of morbidity and mortality worldwide. Despite the tremendous clinical impact, the mechanisms governing the fibrotic process are not yet understood, and to date, no clinically reliable therapies for fibrosis have been discovered. Here we applied Regeneration Intelligence, a new bioinformatics software suite for qualitative analysis of intracellular signaling pathway activation using transcriptomic data, to assess a network of molecular signaling in lung and liver fibrosis. In both tissues, our analysis detected major conserved signaling pathways strongly associated with fibrosis, suggesting that some of the pathways identified by our algorithm but not yet wet-lab validated as fibrogenesis related, may be attractive targets for future research. While the majority of significantly disrupted pathways were specific to histologically distinct organs, several pathways have been concurrently activated or downregulated among the hepatic and pulmonary fibrosis samples, providing new evidence of evolutionary conserved pathways that may be relevant as possible therapeutic targets. While future confirmatory studies are warranted to validate these observations, our platform proposes a promising new approach for detecting fibrosis-promoting pathways and tailoring the right therapy to prevent fibrogenesis.
Collapse
Affiliation(s)
- Eugene Makarev
- a Atlas Regeneration, Inc. , Winston-Salem , NC , USA.,b Insilico Medicine, Inc., ETC, Johns Hopkins University , Baltimore , MD , USA
| | - Evgeny Izumchenko
- c Department of Otolaryngology-Head & Neck Surgery , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Fumiaki Aihara
- d Advanced Academic Programs, Johns Hopkins University , Baltimore , MD , USA
| | - Piotr T Wysocki
- c Department of Otolaryngology-Head & Neck Surgery , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Qingsong Zhu
- b Insilico Medicine, Inc., ETC, Johns Hopkins University , Baltimore , MD , USA
| | - Anton Buzdin
- e The Biogerontology Research Foundation , London , UK
| | - David Sidransky
- c Department of Otolaryngology-Head & Neck Surgery , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Alex Zhavoronkov
- b Insilico Medicine, Inc., ETC, Johns Hopkins University , Baltimore , MD , USA.,f Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine , Winston-Salem , NC , USA
| | - Anthony Atala
- a Atlas Regeneration, Inc. , Winston-Salem , NC , USA.,g Pathway Pharmaceuticals, Ltd , Hong Kong , Hong Kong
| |
Collapse
|
39
|
Izumchenko E, Meir J, Bedi A, Wysocki PT, Hoque MO, Sidransky D. Patient-derived xenografts as tools in pharmaceutical development. Clin Pharmacol Ther 2016; 99:612-21. [PMID: 26874468 DOI: 10.1002/cpt.354] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/20/2016] [Accepted: 02/11/2016] [Indexed: 12/16/2022]
Abstract
Successful drug development in oncology is grossly suboptimal, manifested by the very low percentage of new agents being developed that ultimately succeed in clinical approval. This poor success is in part due to the inability of standard cell-line xenograft models to accurately predict clinical success and to tailor chemotherapy specifically to a group of patients more likely to benefit from the therapy. Patient-derived xenografts (PDXs) maintain the histopathological architecture and molecular features of human tumors, and offer a potential solution to maximize drug development success and ultimately generate better outcomes for patients. Although imperfect in mimicking all aspects of human cancer, PDXs are a more predictable platform for preclinical evaluation of treatment effect and in selected cases can guide therapeutic decision making in the clinic. This article summarizes the current status of PDX models, challenges associated with modeling human cancer, and various approaches that have been applied to overcome these challenges and improve the clinical relevance of PDX cancer models.
Collapse
Affiliation(s)
- E Izumchenko
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - J Meir
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - A Bedi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - P T Wysocki
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - M O Hoque
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - D Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
40
|
Amadoz A, Sebastian-Leon P, Vidal E, Salavert F, Dopazo J. Using activation status of signaling pathways as mechanism-based biomarkers to predict drug sensitivity. Sci Rep 2015; 5:18494. [PMID: 26678097 PMCID: PMC4683444 DOI: 10.1038/srep18494] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/19/2015] [Indexed: 12/22/2022] Open
Abstract
Many complex traits, as drug response, are associated with changes in biological pathways rather than being caused by single gene alterations. Here, a predictive framework is presented in which gene expression data are recoded into activity statuses of signal transduction circuits (sub-pathways within signaling pathways that connect receptor proteins to final effector proteins that trigger cell actions). Such activity values are used as features by a prediction algorithm which can efficiently predict a continuous variable such as the IC50 value. The main advantage of this prediction method is that the features selected by the predictor, the signaling circuits, are themselves rich-informative, mechanism-based biomarkers which provide insight into or drug molecular mechanisms of action (MoA).
Collapse
Affiliation(s)
- Alicia Amadoz
- Computational Genomics Department, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Patricia Sebastian-Leon
- Computational Genomics Department, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Enrique Vidal
- Computational Genomics Department, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
- Bioinformatics of Rare Diseases (BIER), CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Francisco Salavert
- Computational Genomics Department, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
- Bioinformatics of Rare Diseases (BIER), CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Joaquin Dopazo
- Computational Genomics Department, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
- Bioinformatics of Rare Diseases (BIER), CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
- Functional Genomics Node, (INB) at CIPF, Valencia, Spain
| |
Collapse
|