1
|
Dechow A, Timonen S, Ianevski A, Jiang Q, Wahnschaffe L, Peng Y, Jungherz D, Becker K, Neubauer HA, Schönefeldt S, de Araujo E, Gunning P, Fleck R, Schrader A, Hallek M, Pflug N, Moriggl R, Aittokallio T, Mustjoki S, Braun T, Herling M. Dual STAT3/STAT5 inhibition as a novel treatment strategy in T-prolymphocytic leukemia. Leukemia 2025:10.1038/s41375-025-02577-8. [PMID: 40234614 DOI: 10.1038/s41375-025-02577-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/24/2025] [Accepted: 03/19/2025] [Indexed: 04/17/2025]
Abstract
T-prolymphocytic leukemia (T-PLL) is a rare, aggressive T-cell malignancy with poor outcomes and an urgent need for new therapeutic approaches. Integrating genomic data and new transcriptomic profiling, we identified recurrent JAK/STAT mutations (predominantly in JAK3 and STAT5B) as hallmarks in a cohort of 335 T-PLL cases. In line, transcriptomic and protein analyses revealed constitutive JAK/STAT activation in virtually all samples. Consequently, we explored the anti-leukemic potential of dual STAT3/STAT5 non-PROTAC degraders in T-PLL, with JPX-1244 as our lead substance. JPX-1244 efficiently and selectively induced cell death in primary T-PLL samples, including those resistant to conventional therapies, by blocking STAT3 and STAT5 phosphorylation and by inducing their degradation. The extent of STAT3/STAT5 degradation directly correlated with cytotoxicity. RNA-sequencing confirmed the treatment-related downregulation of STAT5 target genes. While JAK/STAT mutations did not predict responses to pharmacologic STAT3/STAT5 degradation, elevated expression of TOX, PAK6, and SPINT1 were associated with drug sensitivity. In subsequent combination screenings, cladribine, venetoclax, and azacytidine emerged as most effective combination partners of STAT3/STAT5 degraders, even in low-responding T-PLL samples, all synergistically reducing STAT5 phosphorylation. These findings highlight dual STAT3/STAT5 inhibition, particularly in combination with hypomethylating and BCL2-targeting agents, as a promising interventional approach in T-PLL, warranting further investigation.
Collapse
Affiliation(s)
- Annika Dechow
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf, University Hospital Cologne, Cologne, Germany
| | - Sanna Timonen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Qu Jiang
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf, University Hospital Cologne, Cologne, Germany
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Hospital Leipzig, University of Leipzig Medical Center and Comprehensive Cancer Center Central Germany (CCCG), Leipzig-, Jena, Germany
| | - Linus Wahnschaffe
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf, University Hospital Cologne, Cologne, Germany
| | - Yayi Peng
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf, University Hospital Cologne, Cologne, Germany
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Hospital Leipzig, University of Leipzig Medical Center and Comprehensive Cancer Center Central Germany (CCCG), Leipzig-, Jena, Germany
| | - Dennis Jungherz
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf, University Hospital Cologne, Cologne, Germany
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Hospital Leipzig, University of Leipzig Medical Center and Comprehensive Cancer Center Central Germany (CCCG), Leipzig-, Jena, Germany
| | - Kerstin Becker
- Cologne Center for Genomics (CCG), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Heidi A Neubauer
- Department for Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Susann Schönefeldt
- Department for Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Elvin de Araujo
- Centre for Medicinal Chemistry, University of Toronto at Mississauga, Mississauga, ON, L5L 1C6, Canada
| | - Patrick Gunning
- Centre for Medicinal Chemistry, University of Toronto at Mississauga, Mississauga, ON, L5L 1C6, Canada
| | | | - Alexandra Schrader
- Lymphoma Immuno Biology Team, Equipe Labellisée LIGUE 2023, Centre International de Recherche en Infectiologie, INSERM U1111-CNRS UMR5308, Faculté de Médecine Lyon-Sud, Hospices Civils de Lyon, Université Claude Bernard Lyon I-ENS de Lyon, Lyon, France
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf, University Hospital Cologne, Cologne, Germany
| | - Natali Pflug
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf, University Hospital Cologne, Cologne, Germany
| | - Richard Moriggl
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Till Braun
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf, University Hospital Cologne, Cologne, Germany
- Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf (MSSO ABCD), Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Marco Herling
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf, University Hospital Cologne, Cologne, Germany.
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Hospital Leipzig, University of Leipzig Medical Center and Comprehensive Cancer Center Central Germany (CCCG), Leipzig-, Jena, Germany.
| |
Collapse
|
2
|
Chidambaram K, Rekha A, Goyal A, Rana M. Targeting KRAS-G12C in lung cancer: The emerging role of PROTACs in overcoming resistance. Pathol Res Pract 2025; 270:155954. [PMID: 40233529 DOI: 10.1016/j.prp.2025.155954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/30/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
In lung cancer, KRAS mutations, especially the G12C, favor aggressive tumor growth and resistance to standard therapies. Although first-generation inhibitors of KRAS G12C, such as sotorasib and adagrasib, are highly effective in early-phase studies, resistance invariably develops under selective inhibition pressure and rarely leads to sustained long-term treatment benefits. As a novel approach to targeting KRAS mutations in lung cancer, PROTAC (Proteolysis Targeting Chimera) technology is explored in this review. The PROTACs take advantage of the cell's ubiquitin-proteasome system to selectively degrade KRAS proteins, overcoming the dilemma of a lack of traditional binding sites and the means of resistance. We review recent progress with KRAS-specific PROTACs and their mechanisms, clinical application, and effectiveness at targeting primary KRAS oncogenes and secondary drivers and signaling pathways contributing to therapeutic resistance. Also, the synergies between PROTACs and immunotherapies or chemotherapies are further amplified. This review also underscores PROTAC technology's promise to advance precision medicine by providing durable treatment options for KRAS-driven lung cancers. It addresses future directions for optimizing PROTAC efficacy, bioavailability, and patient-specific applications.
Collapse
Affiliation(s)
- Kumarappan Chidambaram
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia.
| | - A Rekha
- Dr DY Patil Medical college , Hospital and Research Centre, Pimpri , Pune, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, UP 281406, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
3
|
Paul JK, Azmal M, Haque Shohan MN, Mrinmoy M, Newaz Been Haque ANMS, Talukder OF, Ghosh A. Identification of natural phytochemicals as AKT2 inhibitors using molecular docking and dynamics simulations as potential cancer therapeutics. Heliyon 2025; 11:e41897. [PMID: 39897896 PMCID: PMC11783009 DOI: 10.1016/j.heliyon.2025.e41897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
The PI3K/AKT/mTOR pathway is central in regulating key cellular processes such as proliferation, survival, metabolism, and angiogenesis. Dysregulation of this pathway, particularly in the AKT2 isoform, is commonly observed in cancers such as breast, ovarian, and pancreatic cancers, leading to enhanced tumor progression, metastasis, and therapeutic resistance. Therefore, targeting AKT2 for inhibition is a promising strategy for cancer therapy. This study utilized molecular docking and dynamics simulations to identify natural phytochemicals that inhibit AKT2. Molecular docking results revealed that millettone (CID 442810) exhibited the highest binding affinity to AKT2, with a docking score of -9.5 kcal/mol, followed by uzarigenin (CID 92760), dihydrobiochanin A (CID 439784), and abyssinone I (CID 442152) with docking scores of -9.0 kcal/mol, -8.9 kcal/mol, and -8.7 kcal/mol respectively, outperforming the control inhibitor, ipatasertib (CID 24788740) docking score of -7.56 kcal/mol. Molecular dynamics simulations indicated that millettone, uzarigenin, and dihydrobiochanin A demonstrated strong binding affinities and stable interactions with AKT2, suggesting their potential as therapeutic agents for cancers that involve AKT2 hyperactivation. Notably, uzarigenin's superior stability, as evidenced by its lower root mean square deviation (RMSD), which measures structural stability, and solvent-accessible surface area (SASA), which indicates molecular compactness, highlights its promise as a potent inhibitor of AKT2. Future in vitro and in vivo studies will be crucial to confirm the efficacy of these inhibitors in reducing tumor progression and their potential applications. Given that AKT2 also plays a role in neuronal survival and plasticity, these compounds may have potential applications in neurodegenerative diseases such as Alzheimer's, warranting further investigation into their dual therapeutic relevance.
Collapse
Affiliation(s)
- Jibon Kumar Paul
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Md Naimul Haque Shohan
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Mohua Mrinmoy
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - ANM Shah Newaz Been Haque
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Omar Faruk Talukder
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| |
Collapse
|
4
|
Ganguly S, Burikhanov R, Sviripa VM, Ellingson S, Jiang J, Gosser CM, Orren D, Goellner EM, Shenoy GG, Rao M, D'Orazio J, Brainson CF, Zhan CG, Spielmann PH, Watt DS, Rangnekar VM. S6K1 is a Targetable Vulnerability in Tumors Exhibiting Plasticity and Therapy Resistance. Int J Biol Sci 2025; 21:454-472. [PMID: 39781466 PMCID: PMC11705648 DOI: 10.7150/ijbs.96672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025] Open
Abstract
Background: Most tumors initially respond to treatment, yet refractory clones subsequently develop owing to resistance mechanisms associated with cancer cell plasticity and heterogeneity. Methods: We used a chemical biology approach to identify protein targets in cancer cells exhibiting diverse driver mutations and representing models of tumor lineage plasticity and therapy resistance. An unbiased screen of a drug library was performed against cancer cells followed by synthesis of chemical analogs of the most effective drug. The cancer subtype target range of the leading drug was determined by PRISM analysis of over 900 cancer cell lines at the Broad Institute, MA. RNA-sequencing and enrichment analysis of differentially expressed genes, as well as computational molecular modeling and pull-down with biotinylated small molecules were used to identify and validate RPS6KB1 (p70S6K or S6K1) as an essential target. Genetic restoration was used to test the functional role of S6K1 in cell culture and xenograft models. Results: We identified a novel derivative of the antihistamine drug ebastine, designated Super-ebastine (Super-EBS), that inhibited the viability of cancer cells representing diverse KRAS and EGFR driver mutations and models of plasticity and treatment resistance. Interestingly, PRISM analysis indicated that over 95% of the diverse cancer cell lines tested were sensitive to Super-EBS and the predicted target was the serine/threonine kinase S6K1. S6K1 is upregulated in various cancers relative to counterpart normal/benign tissues and phosphorylated-S6K1 predicts poor prognosis for cancer patients. We noted that inhibition of S6K1 phosphorylation was necessary for tumor cell growth inhibition, and restoration of phospho-S6K1 rendered tumor cells resistant to Super-EBS. Inhibition of S6K1 phosphorylation by Super-EBS induced caspase-2 dependent apoptosis via inhibition of the Cdc42/Rac-1/p-PAK1 pathway that led to actin depolymerization and caspase-2 activation. The essential role of S6K1 in the action of Super-EBS was recapitulated in xenografts, and knockout of S6K1 abrogated tumor growth in mice. Conclusion: S6K1 is a therapeutic vulnerability in tumors exhibiting intrinsic and/or acquired resistance to treatment.
Collapse
Affiliation(s)
- Saptadwipa Ganguly
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Ravshan Burikhanov
- Department of Radiation Medicine, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Vitaliy M. Sviripa
- Department of Molecular and Cellular Biochemistry and Molecular Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Sally Ellingson
- Division of Internal Medicine, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Jieyun Jiang
- Department of Radiation Medicine, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Christian M. Gosser
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - David Orren
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Eva M. Goellner
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Gautham G. Shenoy
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Center for Translational Research, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - John D'Orazio
- Department of Pediatrics, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Christine F. Brainson
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky, USA
- Molecular Modeling and Pharmaceutical Center, College of Pharmacy, University of Kentucky, Lexington, Kentucky, USA
| | - Peter H. Spielmann
- Department of Molecular and Cellular Biochemistry and Molecular Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - David S. Watt
- Department of Molecular and Cellular Biochemistry and Molecular Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Vivek M. Rangnekar
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Department of Radiation Medicine, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
5
|
Geijerman E, Terrana F, Peters GJ, Deng D, Diana P, Giovannetti E, Xu G. Targeting a key FAK-tor: the therapeutic potential of combining focal adhesion kinase (FAK) inhibitors and chemotherapy for chemoresistant non-small cell lung cancer. Expert Opin Investig Drugs 2024; 33:1103-1118. [PMID: 39435477 DOI: 10.1080/13543784.2024.2417762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024]
Abstract
INTRODUCTION NSCLC is the leading cause of cancer-related deaths globally, with a low survival rate primarily due to NSCLC frequently becoming chemoresistant. Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase involved in pathways regulating multiple processes in the cell, including survival, migration, and the TME, that contribute to both tumor progression and drug resistance. Recently, FAK inhibitors (FAKi) have shown promising potential for the treatment of NSCLC. AREAS COVERED This narrative review aims to summarize key signaling pathways involving FAK that contribute to tumor progression and drug resistance. It will further provide an overview of FAKi currently in pre- and early-phase clinical trials for solid tumors, as well as the therapeutic potential of combining FAKi with chemotherapy, as this has emerged as a promising strategy to overcome chemoresistance in NSCLC. EXPERT OPINION It is becoming increasingly clear that FAK is not an oncogenic driver but rather contributes to tumor progression and drug resistance. Hence, while FAKi have only demonstrated modest results in clinical trials when given by themselves, treatment regimens combining other therapies with FAKi have shown promising potential to overcome drug resistance. Lastly, of particular novelty are FAK-PROTACs (proteolysis-targeting chimaeras), which uniquely target both cytosolic and nuclear FAK.
Collapse
Affiliation(s)
- Emma Geijerman
- Amsterdam University College, Amsterdam, The Netherlands
| | - Francesca Terrana
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Fondazione Pisana per la Scienza, Pisa, Italy
| | - Geng Xu
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Saadh MJ, Rasulova I, Almoyad MAA, Kiasari BA, Ali RT, Rasheed T, Faisal A, Hussain F, Jawad MJ, Hani T, Sârbu I, Lakshmaiya N, Ciongradi CI. Recent progress and the emerging role of lncRNAs in cancer drug resistance; focusing on signaling pathways. Pathol Res Pract 2024; 253:154999. [PMID: 38118218 DOI: 10.1016/j.prp.2023.154999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/22/2023]
Abstract
It is becoming more and more apparent that many of the genetic alterations associated with cancer are located in areas that do not encode proteins. lncRNAs are a class of RNAs that do not code for proteins but play a crucial role in maintaining cell function and regulating various cellular processes. By doing this, they have recently introduced what may be a brand-new and essential layer of biological control. These have more than 200 nucleotides and are linked to several diseases; as a result, they have become potential tools for therapeutic intervention. Emerging technologies suggest the presence of mutations on genomic loci that give rise to lncRNAs rather than proteins in a disease as complex as cancer. These lncRNAs play essential parts in gene regulation, which impacts several cellular homeostasis processes, including proliferation, survival, migration, and genomic stability. The leading cause of death in the world today is cancer. Delays in diagnosis and a lack of standard and efficient treatments are the leading causes of the high death rate. Clinically, surgery is frequently used successfully to remove cancers that have not spread, but it is less successful in treating metastatic cancer, which has a drastically lower chance of survival. Chemotherapeutic drugs are a typical therapy to treat the cancer that has spread to other organs. Drug resistance to chemotherapy, however, presents a significant challenge to achieving positive outcomes and is frequently the cause of treatment failure. A substantial barrier to progress in medical oncology is cancer drug resistance. Resistance can develop clinically either before or after cancer treatment. According to this study, lncRNAs influence drug resistance through several different methods. LncRNAs often impact drug resistance by controlling the expression of a few intermediary regulatory variables rather than by directly affecting drug resistance. Additionally, lncRNAs have a variety of roles in cancer medication resistance. Most lncRNAs induce drug resistance when overexpressed; however, other lncRNAs have inhibitory effects. This study provides an overview of the current understanding of lncRNAs, relevance to cancer, and potential therapeutic applications.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | - Irodakhon Rasulova
- School of Humanities, Natural & Social Sciences, New Uzbekistan University, 54 Mustaqillik Ave., Tashkent 100007, Uzbekistan; Department of Public Health, Samarkand State Medical University, Amir Temur Street 18, Samarkand, Uzbekistan
| | - Muhammad Ali Abdullah Almoyad
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University, P.O. Box 4536, 47 Abha Mushait, 61412, Saudi Arabia
| | - Bahman Abedi Kiasari
- Microbiology & Immunology Group, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ronak Taher Ali
- College of Medical Technology, Al-Kitab University, Kirkuk, Iraq
| | - Tariq Rasheed
- College of Science and Humanities, Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Ahmed Faisal
- Department of Pharmacy, Al-Noor University College, Nineveh, Iraq
| | - Farah Hussain
- Medical Technical College, Al-Farahidi University, Iraq
| | | | - Thamer Hani
- Dentistry Department, Al-Turath University College, Baghdad, Iraq
| | - Ioan Sârbu
- 2nd Department of Surgery-Pediatric Surgery and Orthopedics, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania.
| | - Natrayan Lakshmaiya
- Department of Mechanical Engineering, Saveetha School of Engineering, SIMATS, Chennai, Tamil Nadu, India
| | - Carmen Iulia Ciongradi
- 2nd Department of Surgery-Pediatric Surgery and Orthopedics, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania.
| |
Collapse
|
7
|
Zhang X, Chen J, Xi B, Liu Y, Wang S, Gu L, Zhao H, Tao L, Hua Y, Wang Y, Chen M. Agrimoniin is a dual inhibitor of AKT and ERK pathways that inhibit pancreatic cancer cell proliferation. Phytother Res 2023; 37:4076-4091. [PMID: 37156642 DOI: 10.1002/ptr.7867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 04/08/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
Molecular-targeted therapy has shown its effectiveness in pancreatic cancer, while single-targeted drug often cannot provide long-term benefit because of drug resistance. Fortunately, multitarget combination therapy can reverse drug resistance and achieve better efficacy. The typical treatment characteristics of traditional Chinese medicine monomer on tumor are multiple targets, with small side effects, low toxicity, and so forth. Agrimoniin has been reported to be effective on some cancers, while the mechanism still needs to be clarified. In this study, we used 5-ethynyl-2'-deoxyuridine, cell counting kit-8, flow cytometry, and western blot experiments to confirm that agrimoniin can significantly inhibit the proliferation of pancreatic cancer cell PANC-1 by inducing apoptosis and cell cycle arrest. In addition, by using SC79, LY294002 (the agonist or inhibitor of AKT pathway), and U0126 (the inhibitor of ERK pathway), we found that agrimoniin inhibited cell proliferation by simultaneously inhibiting AKT and ERK pathways. Moreover, agrimoniin could significantly increase the inhibitory effect of LY294002 and U0126 on pancreatic cancer cells. Meanwhile, in vivo experiments also supported the above results. In general, agrimoniin is a double-target inhibitor of AKT and ERK pathways in pancreatic cancer cells; it is expected to be used as a resistance reversal agent of targeted drugs or a synergistic drug of the inhibitor of AKT pathway or ERK pathway.
Collapse
Affiliation(s)
- Xiongfei Zhang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianping Chen
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Beili Xi
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yutong Liu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shaojun Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ling Gu
- College of Traditional Chinese Medicine & Integrated Chinese and Western Medicine College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huanhuan Zhao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Tao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Hua
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Wang
- Endoscopy Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meijuan Chen
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
8
|
Ávila-Mendoza J, Delgado-Rueda K, Urban-Sosa VA, Carranza M, Luna M, Martínez-Moreno CG, Arámburo C. KLF13 Regulates the Activity of the GH-Induced JAK/STAT Signaling by Targeting Genes Involved in the Pathway. Int J Mol Sci 2023; 24:11187. [PMID: 37446365 DOI: 10.3390/ijms241311187] [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: 06/15/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
The Krüppel-like factor 13 (KLF13) has emerged as an important transcription factor involved in essential processes of the central nervous system (CNS). It predominantly functions as a transcriptional repressor, impacting the activity of several signaling pathways with essential roles in the CNS, including the JAK/STAT pathway, which is the canonical mediator of growth hormone (GH) signaling. It is now recognized that GH has important actions as a neurotrophic factor. Therefore, we analyzed the effects of KLF13 on the activity of the JAK/STAT signaling pathway in the hippocampus-derived cell line HT22. Results showed that KLF13 directly regulates the expression of several genes involved in the JAK-STAT pathway, including Jak1, Jak2, Jak3, and Socs1, by associating with their proximal gene promoters. In addition, it was found that in KLF13-deficient HT22 neurons, the expression of Jak1, Stat3, Socs1, Socs3, and Igf1 was dysregulated, exhibiting mRNA levels that went up to 7-fold higher than the control cell line. KLF13 displayed a differential effect on the GH-induced JAK/STAT pathway activity, decreasing the STAT3 branch while enhancing the STAT5 branch. In KLF13-deficient HT22 cells, the activity of the STAT3 branch was enhanced, mediating the GH-dependent augmented expression of the JAK/STAT output genes Socs1, Socs3, Igf1, and Bdnf. Furthermore, GH treatment increased both the nuclear content of KLF13 and Klf13 mRNA levels, suggesting that KLF13 could be part of the mechanisms that maintain the homeostatic state of this pathway. These findings support the notion that KLF13 is a regulator of JAK/STAT activity.
Collapse
Affiliation(s)
- José Ávila-Mendoza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Karen Delgado-Rueda
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Valeria A Urban-Sosa
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Martha Carranza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Maricela Luna
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Carlos G Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Carlos Arámburo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| |
Collapse
|
9
|
Stulpinas A, Sereika M, Vitkeviciene A, Imbrasaite A, Krestnikova N, Kalvelyte AV. Crosstalk between protein kinases AKT and ERK1/2 in human lung tumor-derived cell models. Front Oncol 2023; 12:1045521. [PMID: 36686779 PMCID: PMC9848735 DOI: 10.3389/fonc.2022.1045521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/28/2022] [Indexed: 01/06/2023] Open
Abstract
There is no doubt that cell signaling manipulation is a key strategy for anticancer therapy. Furthermore, cell state determines drug response. Thus, establishing the relationship between cell state and therapeutic sensitivity is essential for the development of cancer therapies. In the era of personalized medicine, the use of patient-derived ex vivo cell models is a promising approach in the translation of key research findings into clinics. Here, we were focused on the non-oncogene dependencies of cell resistance to anticancer treatments. Signaling-related mechanisms of response to inhibitors of MEK/ERK and PI3K/AKT pathways (regulators of key cellular functions) were investigated using a panel of patients' lung tumor-derived cell lines with various stemness- and EMT-related markers, varying degrees of ERK1/2 and AKT phosphorylation, and response to anticancer treatment. The study of interactions between kinases was the goal of our research. Although MEK/ERK and PI3K/AKT interactions are thought to be cell line-specific, where oncogenic mutations have a decisive role, we demonstrated negative feedback loops between MEK/ERK and PI3K/AKT signaling pathways in all cell lines studied, regardless of genotype and phenotype differences. Our work showed that various and distinct inhibitors of ERK signaling - selumetinib, trametinib, and SCH772984 - increased AKT phosphorylation, and conversely, inhibitors of AKT - capivasertib, idelalisib, and AKT inhibitor VIII - increased ERK phosphorylation in both control and cisplatin-treated cells. Interaction between kinases, however, was dependent on cellular state. The feedback between ERK and AKT was attenuated by the focal adhesion kinase inhibitor PF573228, and in cells grown in suspension, showing the possible role of extracellular contacts in the regulation of crosstalk between kinases. Moreover, studies have shown that the interplay between MEK/ERK and PI3K/AKT signaling pathways may be dependent on the strength of the chemotherapeutic stimulus. The study highlights the importance of spatial location of the cells and the strength of the treatment during anticancer therapy.
Collapse
|
10
|
Kim Y, Suh BC. Editorial: Brain cells' compensatory mechanisms in response to disease risk factors. Front Mol Neurosci 2022; 15:1096287. [PMID: 36606142 PMCID: PMC9808396 DOI: 10.3389/fnmol.2022.1096287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Yong Kim
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States,Brain Health Institute, Rutgers University, Piscataway, NJ, United States,*Correspondence: Yong Kim ✉
| | - Byung-Chang Suh
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea,Byung-Chang Suh ✉
| |
Collapse
|
11
|
Kareva I. Different costs of therapeutic resistance in cancer: Short- and long-term impact of population heterogeneity. Math Biosci 2022; 352:108891. [PMID: 35998834 DOI: 10.1016/j.mbs.2022.108891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/28/2022] [Accepted: 08/13/2022] [Indexed: 11/29/2022]
Abstract
Therapeutic resistance continues to undercut long-term success of many promising cancer treatments. At times, development of therapeutic resistance can come at a fitness cost for the cancer cell population, which could potentially be leveraged to the patient's advantage. A mathematical formulation of such a situation was proposed by Pressley et al. (2020), who discussed two scenarios, namely, when developing therapeutic resistance can come at a cost to proliferative capacity (such as when a drug targets a growth receptor), or to the total tumor carrying capacity (such as when a drug targets neovascularization). Here we expand the analysis of the two models and evaluate both short- and long-term dynamics of a population heterogeneous with respect to resistance. We analyze the four initial distributions with respect to resistance at the time of treatment initiation: uniform, bell-shaped, exponential, and U-shaped. We show that final population composition is invariant to the initial distribution, with a single clone eventually dominating within the population; the value of the resistance parameter of the final clone depends on other system parameters but not on the initial distribution. Transitional behaviors, however, which may have more significant implications for immediate treatment decisions, depend critically on the initial distribution. Furthermore, we show that depending on the mechanism for the cost of resistance (i.e., proliferation vs carrying capacity), increase in natural cell death rate has opposite effects, with higher natural death rate selecting for less resistant cell clones in the long term for proliferation-dependent model, and selecting for more resistant cell clones for carrying capacity-dependent model, a prediction that may have implications for combination therapy with cytotoxic agents. We conclude with a discussion of strengths and limitations of using modeling for understanding treatment trajectory, as well as the promise of model-informed evolutionary steering for improved long-term therapeutic outcomes.
Collapse
Affiliation(s)
- Irina Kareva
- Department of Biomedical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
12
|
Ju M, Fan J, Zou Y, Yu M, Jiang L, Wei Q, Bi J, Hu B, Guan Q, Song X, Dong M, Wang L, Yu L, Wang Y, Kang H, Xin W, Zhao L. Computational Recognition of a Regulatory T-cell-specific Signature With Potential Implications in Prognosis, Immunotherapy, and Therapeutic Resistance of Prostate Cancer. Front Immunol 2022; 13:807840. [PMID: 35812443 PMCID: PMC9259848 DOI: 10.3389/fimmu.2022.807840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
Prostate cancer, recognized as a "cold" tumor, has an immunosuppressive microenvironment in which regulatory T cells (Tregs) usually play a major role. Therefore, identifying a prognostic signature of Tregs has promising benefits of improving survival of prostate cancer patients. However, the traditional methods of Treg quantification usually suffer from bias and variability. Transcriptional characteristics have recently been found to have a predictive power for the infiltration of Tregs. Thus, a novel machine learning-based computational framework has been presented using Tregs and 19 other immune cell types using 42 purified immune cell datasets from GEO to identify Treg-specific mRNAs, and a prognostic signature of Tregs (named "TILTregSig") consisting of five mRNAs (SOCS2, EGR1, RRM2, TPP1, and C11orf54) was developed and validated to monitor the prognosis of prostate cancer using the TCGA and ICGC datasets. The TILTregSig showed a stronger predictive power for tumor immunity compared with tumor mutation burden and glycolytic activity, which have been reported as immune predictors. Further analyses indicate that the TILTregSig might influence tumor immunity mainly by mediating tumor-infiltrating Tregs and could be a powerful predictor for Tregs in prostate cancer. Moreover, the TILTregSig showed a promising potential for predicting cancer immunotherapy (CIT) response in five CIT response datasets and therapeutic resistance in the GSCALite dataset in multiple cancers. Our TILTregSig derived from PBMCs makes it possible to achieve a straightforward, noninvasive, and inexpensive detection assay for prostate cancer compared with the current histopathological examination that requires invasive tissue puncture, which lays the foundation for the future development of a panel of different molecules in peripheral blood comprising a biomarker of prostate cancer.
Collapse
Affiliation(s)
- Mingyi Ju
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Jingyi Fan
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Yuanjiang Zou
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Mingjie Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Longyang Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Jia Bi
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Baohui Hu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Qiutong Guan
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Xinyue Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Mingyan Dong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Lin Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Yan Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Hui Kang
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Xin
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|
13
|
Sharma M, Bakshi AK, Mittapelly N, Gautam S, Marwaha D, Rai N, Singh N, Tiwari P, Aggarwal N, Kumar A, Mishra PR. Recent updates on innovative approaches to overcome drug resistance for better outcomes in cancer. J Control Release 2022; 346:43-70. [PMID: 35405165 DOI: 10.1016/j.jconrel.2022.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 02/07/2023]
|
14
|
Mengie Ayele T, Tilahun Muche Z, Behaile Teklemariam A, Bogale Kassie A, Chekol Abebe E. Role of JAK2/STAT3 Signaling Pathway in the Tumorigenesis, Chemotherapy Resistance, and Treatment of Solid Tumors: A Systemic Review. J Inflamm Res 2022; 15:1349-1364. [PMID: 35241923 PMCID: PMC8887966 DOI: 10.2147/jir.s353489] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/11/2022] [Indexed: 12/12/2022] Open
Abstract
Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway is a common signaling pathway used to transduce signals from the extracellular to the intracellular (nucleus) upon the binding of cytokines and growth factors to the extracellular domain of specific cell surface receptors. This signaling pathway is tightly regulated and has a multitude of biological functions such as cell proliferation, differentiation, and apoptosis. Besides, the regulated JAK2/STAT3 signaling plays a crucial role in embryonic development, hemopoiesis, and controlling the immune system. Conversely, aberrantly activated JAK2/STAT3 is frequently detected in varieties of tumors and involved in oncogenesis, angiogenesis, and metastasis of many cancer diseases that are usually refractory to the standard chemotherapy. However, the JAK3/STAT3 pathway recently emerged interestingly as a new site for the development of novel anti-tumor agents and becomes a promising therapeutic target in the treatment of many solid malignancies. Herein, this review aimed to provide insight into the JAK2/STAT3 pathway, in the hope to gain an understanding of its potential role in the pathogenesis, progression, chemotherapy resistance, and cancer therapy of solid tumors.
Collapse
Affiliation(s)
- Teklie Mengie Ayele
- Department of Pharmacy, Debre Tabor University, Debre Tabor, Amhara, Ethiopia
| | | | | | | | - Endeshaw Chekol Abebe
- Department of Medical Biochemistry, Debre Tabor University, Debre Tabor, Amhara, Ethiopia
- Correspondence: Endeshaw Chekol Abebe, Tel +251928428133, Email
| |
Collapse
|
15
|
Mason LD, Chava S, Reddi KK, Gupta R. The BRD9/7 Inhibitor TP-472 Blocks Melanoma Tumor Growth by Suppressing ECM-Mediated Oncogenic Signaling and Inducing Apoptosis. Cancers (Basel) 2021; 13:cancers13215516. [PMID: 34771678 PMCID: PMC8582741 DOI: 10.3390/cancers13215516] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Melanoma accounts for the majority of all skin cancer-related deaths and only 1/3rd of melanoma patients with distal metastasis survive beyond five years. However, current therapies including BRAF/MEK targeted therapies or immunotherapies only benefit a subset of melanoma patients due to the emergence of intrinsic or extrinsic resistance mechanisms. Effective treatment of melanoma will thus require new and more effective therapeutic agents. Towards the goal of identifying new therapeutic agents, we conducted an unbiased, druggable epigenetic drug screen using a library of 32 epigenetic inhibitors obtained from the Structural Genome Consortium that targets proteins encoding for epigenetic regulators. This chemical genetic screening identified TP-472, which targets bromodomain-7/9, as the strongest inhibitor of melanoma growth in both short- and long-term survival assays and in mouse models of melanoma tumor growth. Mechanistically, using a transcriptome-wide mRNA sequencing profile we identified TP-472 treatment downregulates genes encoding various extracellular matrix (ECM) proteins, including integrins, collagens, and fibronectins. Reactome-based functional pathway analyses revealed that many of the ECM proteins are involved in extracellular matrix interactions required for cancer cell growth and proliferation. TP-472 treatment also upregulated several pro-apoptotic genes that can inhibit melanoma growth. Collectively, our results identify BRD7/9 inhibitor TP-472 as a potentially useful therapeutic agent for melanoma therapy.
Collapse
Affiliation(s)
- Lawrence David Mason
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
| | - Suresh Chava
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
| | - Kiran Kumar Reddi
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35322, USA
- Correspondence: ; Tel.: +1-205-934-6207
| |
Collapse
|
16
|
AboulWafa OM, Daabees HMG, Hammad A, Badawi WA. New functionalized 6-thienylpyrimidine-5-carbonitriles as antiproliferative agents against human breast cancer cells. Arch Pharm (Weinheim) 2021; 354:e2100177. [PMID: 34347303 DOI: 10.1002/ardp.202100177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 01/02/2023]
Abstract
6-Thienylpyrimidine-5-carbonitrile derivatives were synthesized and screened for their in vitro antiproliferative activities against two human breast cancer cell lines in comparison to 5-fluorouracil as a reference. Compounds 2, 3a-c, and 6b evolved as the most active congeners against both cell lines, while others showed selectivity for only one cell line. Compound 2 exerted its effect through inhibition of the epidermal growth factor receptor (EGFR), while 6b showed less aromatase inhibitory activity than letrozole. The rest of the tested compounds did not show significant inhibition, and it can be assumed that they exert their antiproliferative activity through different target mechanisms. In addition, caspase-9 protein activation assays, cell cycle analysis using flow cytometry, and annexin V-fluorescein isothiocyanate-propidium iodide (FITC/PI) dual staining assays were performed for the most active compounds. All the tested compounds were found to be potent pyrimidine derivatives able to initiate apoptosis in MCF-7 and MDA-MB-231 cells.
Collapse
Affiliation(s)
- Omaima M AboulWafa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Hoda M G Daabees
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Ali Hammad
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Waleed A Badawi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| |
Collapse
|
17
|
Cha JE, Bae WY, Choi JS, Lee SH, Jeong JW. Angiogenic activities are increased via upregulation of HIF-1α expression in gefitinib-resistant non-small cell lung carcinoma cells. Oncol Lett 2021; 22:671. [PMID: 34345296 PMCID: PMC8323004 DOI: 10.3892/ol.2021.12932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) have been used to treat patients with non-small cell lung cancer (NSCLC) and activating EGFR mutations; however, the emergence of secondary mutations in EGFR or the acquisition of resistance to EGFR-TKIs can develop and is involved in clinical failure. Since angiogenesis is associated with tumor progression and the blockade of antitumor drugs, inhibition of angiogenesis could be a rational strategy for developing anticancer drugs combined with EGFR-TKIs to treat patients with NSCLC. The signaling pathway mediated by hypoxia-inducible factor-1 (HIF-1) is essential for tumor angiogenesis. The present study aimed to identify the dependence of gefitinib resistance on HIF-1α activity using angiogenesis assays, western blot analysis, colony formation assay, xenograft tumor mouse model and immunohistochemical analysis of tumor tissues. In the NSCLC cell lines, HIF-1α protein expression levels and hypoxia-induced angiogenic activities were found to be increased. In a xenograft mouse tumor model, tumor tissues derived from gefitinib-resistant PC9 cells showed increased protein expression of HIF-1α and angiogenesis within the tumors. Furthermore, inhibition of HIF-1α suppressed resistance to gefitinib, whereas overexpression of HIF-1α increased resistance to gefitinib. The results from the present study provides evidence that HIF-1α was associated with the acquisition of resistance to gefitinib and suggested that inhibiting HIF-1α alleviated gefitinib resistance in NSCLC cell lines.
Collapse
Affiliation(s)
- Jeong Eun Cha
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woom-Yee Bae
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.,Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae-Sun Choi
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.,Medical Science Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Hyeun Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joo-Won Jeong
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.,Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
18
|
Chen K, Gorgen A, Ding A, Du L, Jiang K, Ding Y, Sapisochin G, Ghanekar A. Dual-Specificity Phosphatase 9 Regulates Cellular Proliferation and Predicts Recurrence After Surgery in Hepatocellular Carcinoma. Hepatol Commun 2021; 5:1310-1328. [PMID: 34278178 PMCID: PMC8279460 DOI: 10.1002/hep4.1701] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/19/2021] [Accepted: 02/07/2021] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (CC) is a common and deadly cancer with complex molecular pathogenesis. Little is known about dual-specificity phosphatases (DUSPs) in HCC. We investigated DUSP9 expression in human HCC, associations between DUSP9 and patient outcomes, and effects of altered DUSP9 expression on HCC biology. We studied public data sets as well as 196 patients at our institution who had HCC resections. Quantitative real-time reverse transcription polymerase chain reaction and western blot demonstrated that DUSP9 expression was increased >10-fold in HCC compared to adjacent liver and healthy controls (P = 0.005). Kaplan-Meier and multivariable regression analyses revealed that higher DUSP9 expression was associated with shorter disease-free survival (high DUSP9, 1.6; 95% confidence interval, 0.9-2.3 vs. low DUSP9, 3.4; 95% confidence interval, 1.8-5.0 years; P = 0.04) and increased risk of recurrence (hazard ratio 1.55; 95% confidence interval, 1.01-2.67; P = 0.05) after resection. DUSP9 complementary DNA (cDNA) was cloned using rapid amplification of cDNA ends, revealing two DUSP9 isoforms in human HCC cells. Studies of transcriptional regulation using promoter-luciferase reporter constructs suggested that DUSP9 transcription is regulated by E26 transformation-specific transcription factors. Proliferation of hepatic cells in vitro was enhanced by lentiviral-mediated overexpression of DUSP9. In contrast, DUSP9 knockout HCC cells generated using clustered regularly interspaced short palindromic repeats (CRISPR) demonstrated decreased HCC proliferation and doxorubicin resistance in vitro and impaired xenograft growth in vivo. RNA sequencing, gene set enrichment, and network/pathway analysis revealed that DUSP9 knockout is associated with activation of protein kinase activity and apoptosis. Conclusion: DUSP9 regulates cell proliferation and predicts recurrence after surgery in HCC. DUSP9 may represent a novel prognostic candidate and therapeutic target. Additional studies are warranted to further explore the role and regulation of DUSP9 in HCC.
Collapse
Affiliation(s)
- Kui Chen
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
| | - Andre Gorgen
- Division of General SurgeryUniversity Health NetworkTorontoONCanada
- Department of SurgeryUniversity of TorontoTorontoONCanada
| | - Avrilynn Ding
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
| | - Lulu Du
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
| | - Keruo Jiang
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
| | - Yu Ding
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
| | - Gonzalo Sapisochin
- Division of General SurgeryUniversity Health NetworkTorontoONCanada
- Department of SurgeryUniversity of TorontoTorontoONCanada
| | - Anand Ghanekar
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
- Division of General SurgeryUniversity Health NetworkTorontoONCanada
- Department of SurgeryUniversity of TorontoTorontoONCanada
| |
Collapse
|
19
|
Walters ZS, Aladowicz E, Villarejo-Balcells B, Nugent G, Selfe JL, Eve P, Blagg J, Rossanese O, Shipley J. Role for the Histone Demethylase KDM4B in Rhabdomyosarcoma via CDK6 and CCNA2: Compensation by KDM4A and Apoptotic Response of Targeting Both KDM4B and KDM4A. Cancers (Basel) 2021; 13:1734. [PMID: 33917420 PMCID: PMC8038694 DOI: 10.3390/cancers13071734] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/02/2021] [Accepted: 04/04/2021] [Indexed: 01/10/2023] Open
Abstract
Histone demethylases are epigenetic modulators that play key roles in regulating gene expression related to many critical cellular functions and are emerging as promising therapeutic targets in a number of tumor types. We previously identified histone demethylase family members as overexpressed in the pediatric sarcoma, rhabdomyosarcoma. Here we show high sensitivity of rhabdomyosarcoma cells to a pan-histone demethylase inhibitor, JIB-04 and identify a key role for the histone demethylase KDM4B in rhabdomyosarcoma cell growth through an RNAi-screening approach. Decreasing KDM4B levels affected cell cycle progression and transcription of G1/S and G2/M checkpoint genes including CDK6 and CCNA2, which are bound by KDM4B in their promoter regions. However, after sustained knockdown of KDM4B, rhabdomyosarcoma cell growth recovered. We show that this can be attributed to acquired molecular compensation via recruitment of KDM4A to the promoter regions of CDK6 and CCNA2 that are otherwise bound by KDM4B. Furthermore, upfront silencing of both KDM4B and KDM4A led to RMS cell apoptosis, not seen by reducing either alone. To circumvent compensation and elicit stronger therapeutic responses, our study supports targeting histone demethylase sub-family proteins through selective poly-pharmacology as a therapeutic approach.
Collapse
Affiliation(s)
- Zoë S. Walters
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Sutton, London SM2 5NG, UK; (Z.S.W.); (E.A.); (B.V.-B.); (J.L.S.)
- Cancer Sciences, Faculty of Medicine, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Ewa Aladowicz
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Sutton, London SM2 5NG, UK; (Z.S.W.); (E.A.); (B.V.-B.); (J.L.S.)
| | - Barbara Villarejo-Balcells
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Sutton, London SM2 5NG, UK; (Z.S.W.); (E.A.); (B.V.-B.); (J.L.S.)
| | - Gary Nugent
- Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, London SM2 5NG, UK; (G.N.); (P.E.); (J.B.); (O.R.)
| | - Joanna L. Selfe
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Sutton, London SM2 5NG, UK; (Z.S.W.); (E.A.); (B.V.-B.); (J.L.S.)
| | - Paul Eve
- Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, London SM2 5NG, UK; (G.N.); (P.E.); (J.B.); (O.R.)
| | - Julian Blagg
- Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, London SM2 5NG, UK; (G.N.); (P.E.); (J.B.); (O.R.)
| | - Olivia Rossanese
- Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, London SM2 5NG, UK; (G.N.); (P.E.); (J.B.); (O.R.)
| | - Janet Shipley
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, Sutton, London SM2 5NG, UK; (Z.S.W.); (E.A.); (B.V.-B.); (J.L.S.)
| |
Collapse
|
20
|
Chong ZX, Yeap SK, Ho WY. Role of miRNAs in regulating responses to radiotherapy in human breast cancer. Int J Radiat Biol 2021; 97:289-301. [PMID: 33356761 DOI: 10.1080/09553002.2021.1864048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Breast cancer is the most common type of cancer that affects females globally. Radiotherapy is a standard treatment option for breast cancer, where one of its most significant limitations is radioresistance development. MicroRNAs (miRNAs) are small, non-protein-coding RNAs that have been widely studied for their roles as disease biomarkers. To date, several in vitro, in vivo, and clinical studies have reported the roles of miRNAs in regulating radiosensitivity and radioresistance in breast cancer cells. This article reviews the roles of miRNAs in regulating treatment response toward radiotherapy and the associating cellular pathways. We identified 36 miRNAs that play a role in mediating radio-responses; 22 were radiosensitizing, 12 were radioresistance-promoting, and two miRNAs were reported to promote both effects. A brief overview of breast cancer therapy options, mechanism of action of radiation, and molecular mechanism of radioresistance was provided in this article. A summary of the latest clinical researches involving miRNAs in breast cancer radiotherapy was also included.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, Selangor, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Selangor, Malaysia
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, Selangor, Malaysia
| |
Collapse
|
21
|
Ciocan-Cȃrtiţă CA, Jurj A, Raduly L, Cojocneanu R, Moldovan A, Pileczki V, Pop LA, Budişan L, Braicu C, Korban SS, Berindan-Neagoe I. New perspectives in triple-negative breast cancer therapy based on treatments with TGFβ1 siRNA and doxorubicin. Mol Cell Biochem 2020; 475:285-299. [PMID: 32888160 DOI: 10.1007/s11010-020-03881-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/07/2020] [Indexed: 12/22/2022]
Abstract
Triple-negative breast cancer (TNBC), which accounts for 10-20% of all breast cancers, has the worst prognosis. Although chemotherapy treatment is a standard for TNBC, it lacks a specific target. Therefore, new therapeutic strategies are required to be investigated. In this study, a combined doxorubicin (DOX) and small interfering RNA (siRNA) therapy is proposed as therapeutic strategy for targeting TGFβ1 gene. Hs578T cell line is used as in vitro model for TNBC, wherein TGFβ1siRNA therapy is employed to enhance therapeutic effects. Cell proliferation rate is measured using an MTT test, and morphological alterations are assed using microscopically approached, while gene expression is determined by qRT-PCR analysis. The combined treatment of TGFβ1siRNA and DOX reduced levels of cell proliferation and mitochondrial activity and promoted the alteration of cell morphology (dark-field microscopy). DOX treatment caused downregulation of six genes and upregulation of another six genes. The combined effects of DOX and TGFβ1siRNA resulted in upregulation of 13 genes and downregulation of four genes. Silencing of TGFβ1 resulted in activation of cell death mechanisms in Hs578T cells, to potentiate the effects of DOX, but not in an additive manner, due to the activation of genes involved in resistance to therapy (ABCB1 and IL-6).
Collapse
Affiliation(s)
- Cristina Alexandra Ciocan-Cȃrtiţă
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Ancuţa Jurj
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Lajos Raduly
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Roxana Cojocneanu
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Alin Moldovan
- MedFuture Research Center for Advanced Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400349, Cluj-Napoca, Romania
| | - Valentina Pileczki
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Laura-Ancuta Pop
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Liviuţa Budişan
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Cornelia Braicu
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania.
| | - Schuyler S Korban
- Department of Natural and Environmental Sciences, University of Illinois At Urbana-Champaign, Urbana, IL, 61801, USA
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania. .,Department of Functional Genomics and Experimental Pathology, "Prof. Dr. Ion Chiricuţă" Oncology Institute, 34-36 Republicii Street, 400015, Cluj-Napoca, Romania.
| |
Collapse
|
22
|
A scalable insect cell-based production process of the human recombinant BMX for in-vitro covalent ligand high-throughput screening. Bioprocess Biosyst Eng 2020; 44:209-215. [PMID: 32816073 DOI: 10.1007/s00449-020-02421-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/31/2020] [Indexed: 10/23/2022]
Abstract
Bone Marrow Tyrosine kinase in the chromosome X (BMX) is a TEC family kinase associated with numerous pathological pathways in cancer cells. Covalent inhibition of BMX activity holds promise as a therapeutic approach against cancer. To screen for potent and selective covalent BMX inhibitors, large quantities of highly pure BMX are normally required which is challenging with the currently available production and purification processes. Here, we developed a scalable production process for the human recombinant BMX (hrBMX) using the insect cell-baculovirus expression vector system. Comparable expression levels were obtained in small-scale shake flasks (13 mL) and in stirred-tank bioreactors (STB, 5 L). A two-step chromatographic-based process was implemented, reducing purification times by 75% when compared to traditional processes, while maintaining hrBMX stability. The final production yield was 24 mg of purified hrBMX per litter of cell culture, with a purity of > 99%. Product quality was assessed and confirmed through a series of biochemical and biophysical assays, including circular dichroism and dynamic light scattering. Overall, the platform herein developed was capable of generating 100 mg purified hrBMX from 5 L STB in just 34 days, thus having the potential to assist in-vitro covalent ligand high-throughput screening for BMX activity inhibition.
Collapse
|
23
|
Zhou Y, Mowlazadeh Haghighi S, Liu Z, Wang L, Hruby VJ, Cai M. Development of Ligand-Drug Conjugates Targeting Melanoma through the Overexpressed Melanocortin 1 Receptor. ACS Pharmacol Transl Sci 2020; 3:921-930. [PMID: 33073191 DOI: 10.1021/acsptsci.0c00072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Indexed: 12/31/2022]
Abstract
Melanoma is a lethal form of skin cancer. Despite recent breakthroughs of BRAF-V600E and PD-1 inhibitors showing remarkable clinical responses, melanoma can eventually survive these targeted therapies and become resistant. To solve the drug resistance issue, we designed and synthesized ligand-drug conjugates that couple cytotoxic drugs, which have a low cancer resistance issue, with the melanocortin 1 receptor (MC1R) agonist melanotan-II (MT-II), which provides specificity to MC1R-overexpressing melanoma. The drug-MT-II conjugates maintain strong binding interactions to MC1R and induce selective drug delivery to A375 melanoma cells through its MT-II moiety in vitro. Furthermore, using camptothecin as the cytotoxic drug, camptothecin-MT-II (compound 1) can effectively inhibit A375 melanoma cell growth with an IC50 of 16 nM. By providing selectivity to melanoma cells through its MT-II moiety, this approach of drug-MT-II conjugates enables us to have many more options for cytotoxic drug selection, which can be the key to solving the cancer resistant problem for melanoma.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Saghar Mowlazadeh Haghighi
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Zekun Liu
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Lingzhi Wang
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Victor J Hruby
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Minying Cai
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
24
|
Samaržija I, Dekanić A, Humphries JD, Paradžik M, Stojanović N, Humphries MJ, Ambriović-Ristov A. Integrin Crosstalk Contributes to the Complexity of Signalling and Unpredictable Cancer Cell Fates. Cancers (Basel) 2020; 12:E1910. [PMID: 32679769 PMCID: PMC7409212 DOI: 10.3390/cancers12071910] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/10/2020] [Accepted: 07/12/2020] [Indexed: 12/12/2022] Open
Abstract
Integrins are heterodimeric cell surface receptors composed of α and β subunits that control adhesion, proliferation and gene expression. The integrin heterodimer binding to ligand reorganises the cytoskeletal networks and triggers multiple signalling pathways that can cause changes in cell cycle, proliferation, differentiation, survival and motility. In addition, integrins have been identified as targets for many different diseases, including cancer. Integrin crosstalk is a mechanism by which a change in the expression of a certain integrin subunit or the activation of an integrin heterodimer may interfere with the expression and/or activation of other integrin subunit(s) in the very same cell. Here, we review the evidence for integrin crosstalk in a range of cellular systems, with a particular emphasis on cancer. We describe the molecular mechanisms of integrin crosstalk, the effects of cell fate determination, and the contribution of crosstalk to therapeutic outcomes. Our intention is to raise awareness of integrin crosstalk events such that the contribution of the phenomenon can be taken into account when researching the biological or pathophysiological roles of integrins.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (I.S.); (M.P.); (N.S.)
| | - Ana Dekanić
- Laboratory for Protein Dynamics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | - Jonathan D. Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, UK; (J.D.H.); (M.J.H.)
| | - Mladen Paradžik
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (I.S.); (M.P.); (N.S.)
| | - Nikolina Stojanović
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (I.S.); (M.P.); (N.S.)
| | - Martin J. Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, UK; (J.D.H.); (M.J.H.)
| | - Andreja Ambriović-Ristov
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (I.S.); (M.P.); (N.S.)
| |
Collapse
|
25
|
Willoughby JLS, George K, Roberto MP, Chin HG, Stoiber P, Shin H, Pedamallu CS, Schaus SE, Fitzgerald K, Shah J, Hansen U. Targeting the oncogene LSF with either the small molecule inhibitor FQI1 or siRNA causes mitotic delays with unaligned chromosomes, resulting in cell death or senescence. BMC Cancer 2020; 20:552. [PMID: 32539694 PMCID: PMC7296649 DOI: 10.1186/s12885-020-07039-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/04/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The oncogene LSF (encoded by TFCP2) has been proposed as a novel therapeutic target for multiple cancers. LSF overexpression in patient tumors correlates with poor prognosis in particular for both hepatocellular carcinoma and colorectal cancer. The limited treatment outcomes for these diseases and disappointing clinical results, in particular, for hepatocellular carcinoma in molecularly targeted therapies targeting cellular receptors and kinases, underscore the need for molecularly targeting novel mechanisms. LSF small molecule inhibitors, Factor Quinolinone Inhibitors (FQIs), have exhibited robust anti-tumor activity in multiple pre-clinical models, with no observable toxicity. METHODS To understand how the LSF inhibitors impact cancer cell proliferation, we characterized the cellular phenotypes that result from loss of LSF activity. Cell proliferation and cell cycle progression were analyzed, using HeLa cells as a model cancer cell line responsive to FQI1. Cell cycle progression was studied either by time lapse microscopy or by bulk synchronization of cell populations to ensure accuracy in interpretation of the outcomes. In order to test for biological specificity of targeting LSF by FQI1, results were compared after treatment with either FQI1 or siRNA targeting LSF. RESULTS Highly similar cellular phenotypes are observed upon treatments with FQI1 and siRNA targeting LSF. Along with similar effects on two cellular biomarkers, inhibition of LSF activity by either mechanism induced a strong delay or arrest prior to metaphase as cells progressed through mitosis, with condensed, but unaligned, chromosomes. This mitotic disruption in both cases resulted in improper cellular division leading to multiple outcomes: multi-nucleation, apoptosis, and cellular senescence. CONCLUSIONS These data strongly support that cellular phenotypes observed upon FQI1 treatment are due specifically to the loss of LSF activity. Specific inhibition of LSF by either small molecules or siRNA results in severe mitotic defects, leading to cell death or senescence - consequences that are desirable in combating cancer. Taken together, these findings confirm that LSF is a promising target for cancer treatment. Furthermore, this study provides further support for developing FQIs or other LSF inhibitory strategies as treatment for LSF-related cancers with high unmet medical needs.
Collapse
Affiliation(s)
- Jennifer L S Willoughby
- Alnylam Pharmaceuticals, Inc., Cambridge, MA, 02142, USA.,Department of Biology, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA
| | - Kelly George
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Mark P Roberto
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA
| | - Hang Gyeong Chin
- MCBB Graduate Program, Boston University, Boston, MA, 02215, USA.,New England BioLabs, Ipswich, MA, 01938, USA
| | - Patrick Stoiber
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA.,MCBB Graduate Program, Boston University, Boston, MA, 02215, USA
| | - Hyunjin Shin
- Data Science Institute, Takeda Pharmaceuticals International, Inc., Cambridge, MA, 02139, USA
| | - Chandra Sekhar Pedamallu
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Scott E Schaus
- Center for Molecular Discovery, Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | | | - Jagesh Shah
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Ulla Hansen
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA. .,MCBB Graduate Program, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
26
|
Resistance to MET/VEGFR2 Inhibition by Cabozantinib Is Mediated by YAP/TBX5-Dependent Induction of FGFR1 in Castration-Resistant Prostate Cancer. Cancers (Basel) 2020; 12:cancers12010244. [PMID: 31963871 PMCID: PMC7016532 DOI: 10.3390/cancers12010244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/31/2019] [Accepted: 01/06/2020] [Indexed: 12/20/2022] Open
Abstract
The overall goal of this study was to elucidate the role of FGFR1 induction in acquired resistance to MET and VEGFR2 inhibition by cabozantinib in prostate cancer (PCa) and leverage this understanding to improve therapy outcomes. The response to cabozantinib was examined in mice bearing patient-derived xenografts in which FGFR1 was overexpressed. Using a variety of cell models that reflect different PCa disease states, the mechanism underpinning FGFR1 signaling activation by cabozantinib was investigated. We performed parallel investigations in specimens from cabozantinib-treated patients to confirm our in vitro and in vivo data. FGFR1 overexpression was sufficient to confer resistance to cabozantinib. Our results demonstrate transcriptional activation of FGF/FGFR1 expression in cabozantinib-resistant models. Further analysis of molecular pathways identified a YAP/TBX5-driven mechanism of FGFR1 and FGF overexpression induced by MET inhibition. Importantly, knockdown of YAP and TBX5 led to decreased FGFR1 protein expression and decreased mRNA levels of FGFR1, FGF1, and FGF2. This association was confirmed in a cohort of hormone-naïve patients with PCa receiving androgen deprivation therapy and cabozantinib, further validating our findings. These findings reveal that the molecular basis of resistance to MET inhibition in PCa is FGFR1 activation through a YAP/TBX5-dependent mechanism. YAP and its downstream target TBX5 represent a crucial mediator in acquired resistance to MET inhibitors. Thus, our studies provide insight into the mechanism of acquired resistance and will guide future development of clinical trials with MET inhibitors.
Collapse
|
27
|
Brachet-Botineau M, Polomski M, Neubauer HA, Juen L, Hédou D, Viaud-Massuard MC, Prié G, Gouilleux F. Pharmacological Inhibition of Oncogenic STAT3 and STAT5 Signaling in Hematopoietic Cancers. Cancers (Basel) 2020; 12:E240. [PMID: 31963765 PMCID: PMC7016966 DOI: 10.3390/cancers12010240] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022] Open
Abstract
Signal Transducer and Activator of Transcription (STAT) 3 and 5 are important effectors of cellular transformation, and aberrant STAT3 and STAT5 signaling have been demonstrated in hematopoietic cancers. STAT3 and STAT5 are common targets for different tyrosine kinase oncogenes (TKOs). In addition, STAT3 and STAT5 proteins were shown to contain activating mutations in some rare but aggressive leukemias/lymphomas. Both proteins also contribute to drug resistance in hematopoietic malignancies and are now well recognized as major targets in cancer treatment. The development of inhibitors targeting STAT3 and STAT5 has been the subject of intense investigations during the last decade. This review summarizes the current knowledge of oncogenic STAT3 and STAT5 functions in hematopoietic cancers as well as advances in preclinical and clinical development of pharmacological inhibitors.
Collapse
Affiliation(s)
- Marie Brachet-Botineau
- Leukemic Niche and Oxidative metabolism (LNOx), CNRS ERL 7001, University of Tours, 37000 Tours, France;
| | - Marion Polomski
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, A-1210 Vienna, Austria;
| | - Ludovic Juen
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Damien Hédou
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Marie-Claude Viaud-Massuard
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Gildas Prié
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Fabrice Gouilleux
- Leukemic Niche and Oxidative metabolism (LNOx), CNRS ERL 7001, University of Tours, 37000 Tours, France;
| |
Collapse
|
28
|
van der Kooij MK, Speetjens FM, van der Burg SH, Kapiteijn E. Uveal Versus Cutaneous Melanoma; Same Origin, Very Distinct Tumor Types. Cancers (Basel) 2019; 11:E845. [PMID: 31248118 PMCID: PMC6627906 DOI: 10.3390/cancers11060845] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/16/2019] [Accepted: 06/17/2019] [Indexed: 02/06/2023] Open
Abstract
Here, we critically evaluated the knowledge on cutaneous melanoma (CM) and uveal melanoma (UM). Both cancer types derive from melanocytes that share the same embryonic origin and display the same cellular function. Despite their common origin, both CM and UM display extreme differences in their genetic alterations and biological behavior. We discuss the differences in genetic alterations, metastatic routes, tumor biology, and tumor-host interactions in the context of their clinical responses to targeted- and immunotherapy.
Collapse
Affiliation(s)
- Monique K van der Kooij
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300 RC Leiden, The Netherlands.
| | - Frank M Speetjens
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300 RC Leiden, The Netherlands.
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300 RC Leiden, The Netherlands.
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300 RC Leiden, The Netherlands.
| |
Collapse
|
29
|
Maher M, Kassab AE, Zaher AF, Mahmoud Z. Novel pyrazolo[3,4-d]pyrimidines: design, synthesis, anticancer activity, dual EGFR/ErbB2 receptor tyrosine kinases inhibitory activity, effects on cell cycle profile and caspase-3-mediated apoptosis. J Enzyme Inhib Med Chem 2019; 34:532-546. [PMID: 30688116 PMCID: PMC6352943 DOI: 10.1080/14756366.2018.1564046] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A series of novel pyrazolo[3,4-d]pyrimidines was synthesised. Twelve synthesised compounds were evaluated for their anticancer activity against 60 human tumour cell lines by NCI (USA). Compound 7d proved prominent anticancer activity. It showed 1.6-fold more potent anti-proliferative activity against OVCAR-4 cell line with IC50 = 1.74 μM. It also exhibited promising potent anticancer activity against ACHN cell line with IC50 value 5.53 μM, representing 2.2-fold more potency than Erlotinib. Regarding NCI-H460 cell line, compound 7d (IC50 = 4.44 μM) was 1.9-fold more potent than Erlotinib. It inhibited EGFR and ErbB2 kinases at sub-micromolar level (IC50 = 0.18 and 0.25 µM, respectively). Dual inhibition of EGFR and ErbB2 caused induction of apoptosis which was confirmed by a significant increase in the level of active caspase-3 (11-fold). It showed accumulation of cells in pre-G1 phase and cell cycle arrest at G2/M phase.
Collapse
Affiliation(s)
- Mai Maher
- a Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy , Cairo University , Cairo , Egypt
| | - Asmaa E Kassab
- a Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy , Cairo University , Cairo , Egypt
| | - Ashraf F Zaher
- a Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy , Cairo University , Cairo , Egypt
| | - Zeinab Mahmoud
- a Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy , Cairo University , Cairo , Egypt
| |
Collapse
|
30
|
Giovannini C, Salzano AM, Baglioni M, Vitale M, Scaloni A, Zambrano N, Giannone FA, Vasuri F, D'Errico A, Svegliati Baroni G, Bolondi L, Gramantieri L. Brivanib in combination with Notch3 silencing shows potent activity in tumour models. Br J Cancer 2019; 120:601-611. [PMID: 30765875 PMCID: PMC6461893 DOI: 10.1038/s41416-018-0375-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 12/03/2018] [Accepted: 12/14/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Sorafenib is the first targeted agent proven to improve survival of patients with advanced hepatocellular carcinoma (HCC) and it has been used in first line treatments with heterogeneous response across patients. Most of the promising agents evaluated in first-line or second-line phase III trials for HCC failed to improve patient survival. The absence of molecular characterisation, including the identification of pathways driving resistance might be responsible for these disappointing results. METHODS 2D DIGE and MS analyses were used to reveal proteomic signatures resulting from Notch3 inhibition in HepG2 cells, combined with brivanib treatment. The therapeutic potential of Notch3 inhibition combined with brivanib treatment was also demonstrated in a rat model of HCC and in cell lines derived from different human cancers. RESULTS Using a proteomic approach, we have shown that Notch3 is strongly involved in brivanib resistance through a p53-dependent regulation of enzymes of the tricarboxylic acid (TCA), both in vitro and in vivo. CONCLUSION We have demonstrated that regulation of the TCA cycle is a common mechanism in different human cancers, suggesting that Notch3 inhibitors combined with brivanib treatment may represent a strong formulation for the treatment of HCC as well as Notch3-driven cancers.
Collapse
Affiliation(s)
- Catia Giovannini
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy. .,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.
| | - Anna Maria Salzano
- Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147, Napoli, Italy
| | - Michele Baglioni
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy
| | - Monica Vitale
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Napoli, Italy.,CEINGE Biotecnologie Avanzate S.C.aR.L, Napoli, Italy
| | - Andrea Scaloni
- Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147, Napoli, Italy
| | - Nicola Zambrano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Napoli, Italy.,CEINGE Biotecnologie Avanzate S.C.aR.L, Napoli, Italy
| | | | - Francesco Vasuri
- Pathology Unit, St. Orsola-Malpighi University Hospital, 40138, Bologna, Italy
| | - Antonia D'Errico
- Pathology Unit, St. Orsola-Malpighi University Hospital, 40138, Bologna, Italy
| | | | - Luigi Bolondi
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Laura Gramantieri
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
31
|
Hurwitz H, Van Cutsem E, Bendell J, Hidalgo M, Li CP, Salvo MG, Macarulla T, Sahai V, Sama A, Greeno E, Yu KH, Verslype C, Dawkins F, Walker C, Clark J, O'Reilly EM. Ruxolitinib + capecitabine in advanced/metastatic pancreatic cancer after disease progression/intolerance to first-line therapy: JANUS 1 and 2 randomized phase III studies. Invest New Drugs 2018; 36:683-695. [PMID: 29508247 PMCID: PMC6752723 DOI: 10.1007/s10637-018-0580-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/20/2018] [Indexed: 12/18/2022]
Abstract
Background Ruxolitinib, a Janus kinase 1 (JAK1)/JAK2 inhibitor, plus capecitabine improved overall survival (OS) vs capecitabine in a subgroup analysis of patients with metastatic pancreatic cancer and systemic inflammation (C-reactive protein [CRP] >13 mg/dL) in the randomized phase II RECAP study. We report results from two randomized phase III studies, JANUS 1 (NCT02117479) and JANUS 2 (NCT02119663). Patients and Methods Adults with advanced/metastatic pancreatic cancer, one prior chemotherapy regimen and CRP >10 mg/L were randomized 1:1 (stratified by modified Glasgow Prognostic Score [1 vs 2] and Eastern Cooperative Oncology Group performance status [0/1 vs 2]) to 21-day cycles of ruxolitinib 15 mg twice daily plus capecitabine 2000 mg/m2/day (Days 1-14) or placebo plus capecitabine. The primary endpoint was OS. Results Both studies were terminated following a planned interim futility/efficacy analysis of JANUS 1. Overall, 321 and 86 patients were randomized in JANUS 1 (ruxolitinib: n = 161; placebo: n = 160) and JANUS 2 (ruxolitinib: n = 43; placebo: n = 43). There was no significant difference in OS or progression-free survival (PFS) between treatments in JANUS 1 (OS: hazard ratio [HR], 0.969, 95% confidence interval [CI], 0.747-1.256; PFS: HR, 1.056; 95% CI, 0.827-1.348) or JANUS 2 (OS: HR, 1.584; 95% CI, 0.886-2.830; PFS: HR, 1.166; 95% CI, 0.687-1.978). The most common hematologic adverse event was anemia. No new safety signals with ruxolitinib or capecitabine were identified. Conclusions Ruxolitinib plus capecitabine was well tolerated in refractory pancreatic cancer patients; this combination did not improve survival.
Collapse
Affiliation(s)
- Herbert Hurwitz
- Duke University Medical Center, Campus mail 439 Seeley-mudd Bldg, 10 Bryan Searle Drive, Duke University M, Durham, NC, 27710, USA
| | - Eric Van Cutsem
- Clinical Digestive Oncology, University Hospitals Leuven and KU Leuven, UZ Herestraat 49, 3000, Leuven, Belgium
| | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, 250 25th Ave N, Nashville, TN, 37203, USA
| | - Manuel Hidalgo
- Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Chung-Pin Li
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Road, Beitou District, Taipei, 11217, Taiwan
- School of Medicine, National Yang-Ming University, No. 155, Sec. 2, Li-Nong Street, Beitou District, Taipei, 112, Taiwan
| | - Marcelo Garrido Salvo
- Pontificia Universidad Católica de Chile, Av Libertador Bernardo O'Higgins, 340, Santiago, Región Metropolitana, Chile
| | - Teresa Macarulla
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital (HUVH), Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Vaibhav Sahai
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Cancer Center Floor B1 Reception E, 1500 E Medical Center Dr SPC 5912, Ann Arbor, MI, 48109-5912, USA
| | - Ashwin Sama
- Thomas Jefferson University Hospital, 925 Chestnut Street, Suite 320A, Philadelphia, PA, 19107, USA
| | - Edward Greeno
- Division of Hematology, Oncology and Transplantation, University of Minnesota, 420 Delaware Street SE, MMC 480, Minneapolis, MN, 55455, USA
| | - Kenneth H Yu
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY, 10065, USA
| | - Chris Verslype
- Clinical Digestive Oncology, University Hospitals Leuven and KU Leuven, UZ Herestraat 49, 3000, Leuven, Belgium
| | - Fitzroy Dawkins
- Incyte Corporation, 1801 Augustine Cut-off, Wilmington, DE, 19803, USA
| | - Chris Walker
- Incyte Corporation, 1801 Augustine Cut-off, Wilmington, DE, 19803, USA
| | - Jason Clark
- Incyte Corporation, 1801 Augustine Cut-off, Wilmington, DE, 19803, USA
| | - Eileen M O'Reilly
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY, 10065, USA.
- Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
32
|
Bhayadia R, Krowiorz K, Haetscher N, Jammal R, Emmrich S, Obulkasim A, Fiedler J, Schwarzer A, Rouhi A, Heuser M, Wingert S, Bothur S, Döhner K, Mätzig T, Ng M, Reinhardt D, Döhner H, Zwaan CM, van den Heuvel Eibrink M, Heckl D, Fornerod M, Thum T, Humphries RK, Rieger MA, Kuchenbauer F, Klusmann JH. Endogenous Tumor Suppressor microRNA-193b: Therapeutic and Prognostic Value in Acute Myeloid Leukemia. J Clin Oncol 2018; 36:1007-1016. [DOI: 10.1200/jco.2017.75.2204] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose Dysregulated microRNAs are implicated in the pathogenesis and aggressiveness of acute myeloid leukemia (AML). We describe the effect of the hematopoietic stem-cell self-renewal regulating miR-193b on progression and prognosis of AML. Methods We profiled miR-193b-5p/3p expression in cytogenetically and clinically characterized de novo pediatric AML (n = 161) via quantitative real-time polymerase chain reaction and validated our findings in an independent cohort of 187 adult patients. We investigated the tumor suppressive function of miR-193b in human AML blasts, patient-derived xenografts, and miR-193b knockout mice in vitro and in vivo. Results miR-193b exerted important, endogenous, tumor-suppressive functions on the hematopoietic system. miR-193b-3p was downregulated in several cytogenetically defined subgroups of pediatric and adult AML, and low expression served as an independent indicator for poor prognosis in pediatric AML (risk ratio ± standard error, −0.56 ± 0.23; P = .016). miR-193b-3p expression improved the prognostic value of the European LeukemiaNet risk-group stratification or a 17-gene leukemic stemness score. In knockout mice, loss of miR-193b cooperated with Hoxa9/Meis1 during leukemogenesis, whereas restoring miR-193b expression impaired leukemic engraftment. Similarly, expression of miR-193b in AML blasts from patients diminished leukemic growth in vitro and in mouse xenografts. Mechanistically, miR-193b induced apoptosis and a G1/S-phase block in various human AML subgroups by targeting multiple factors of the KIT-RAS-RAF-MEK-ERK (MAPK) signaling cascade and the downstream cell cycle regulator CCND1. Conclusion The tumor-suppressive function is independent of patient age or genetics; therefore, restoring miR-193b would assure high antileukemic efficacy by blocking the entire MAPK signaling cascade while preventing the emergence of resistance mechanisms.
Collapse
Affiliation(s)
- Raj Bhayadia
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Kathrin Krowiorz
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Nadine Haetscher
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Razan Jammal
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Stephan Emmrich
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Askar Obulkasim
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Jan Fiedler
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Adrian Schwarzer
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Arefeh Rouhi
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Michael Heuser
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Susanne Wingert
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Sabrina Bothur
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Konstanze Döhner
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Tobias Mätzig
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Michelle Ng
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Dirk Reinhardt
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Hartmut Döhner
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - C. Michel Zwaan
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Marry van den Heuvel Eibrink
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Dirk Heckl
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Maarten Fornerod
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Thomas Thum
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - R. Keith Humphries
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Michael A. Rieger
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Florian Kuchenbauer
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| | - Jan-Henning Klusmann
- Raj Bhayadia, Razan Jammal, Stephan Emmrich, Jan Fiedler, Adrian Schwarzer, Michael Heuser, Michelle Ng, Dirk Heckl, and Thomas Thum, Hannover Medical School, Hannover; Raj Bhayadia, Michelle Ng, and Jan-Henning Klusmann, University of Halle, Halle; Kathrin Krowiorz, Arefeh Rouhi, Konstanze Döhner, Hartmut Döhner, and Florian Kuchenbauer, University Hospital of Ulm, Ulm; Nadine Haetscher, Susanne Wingert, Sabrina Bothur, and Michael A. Rieger, Goethe University Frankfurt, Frankfurt; Sabrina Bothur and
| |
Collapse
|
33
|
Houshmand M, Yazdi N, Kazemi A, Atashi A, Hamidieh AA, Anjam Najemdini A, Mohammadi Pour M, Nikougoftar Zarif M. Long non-coding RNA PVT1 as a novel candidate for targeted therapy in hematologic malignancies. Int J Biochem Cell Biol 2018; 98:54-64. [PMID: 29510227 DOI: 10.1016/j.biocel.2018.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 02/22/2018] [Accepted: 03/02/2018] [Indexed: 01/10/2023]
Abstract
Cancerous cells show resistance to various forms of therapy, so applying up to the minute targeted therapy is crucial. For this purpose, long non-coding RNA PVT1 as shown by recent studies is an important oncogene that interacts with vital cellular signaling pathways and different proteins such as c-Myc, NOP2 and LATS2. Due to the enormous role of long non-coding RNAs in development of leukemias, we aimed to show the role of PVT1 knock-down on fate of different hematologic cell lines. owing to this matter, various experiments such as Real-time PCR, cell cycle analysis and apoptosis assay were performed. Meanwhile, proliferation rate by CFSE, protein expression of c-Myc and hTERT by western blot and flow cytometry analysis were investigated. Our results demonstrated that PVT1 knock-down results in c-Myc degradation, proliferation down-regulation, induction of apoptosis and G0/G1 arrest. Simultaneously, for the first time, we posited the relation between this oncogene with hTERT that reduced after PVT1 knock-down. Considering these results, long non-coding RNA PVT1 may be a potential option for targeted therapy in hematologic malignancies.
Collapse
Affiliation(s)
- Mohammad Houshmand
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran; Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Narjes Yazdi
- Department of Molecular Genetics, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Alireza Kazemi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Atashi
- Stem Cell and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Amir Ali Hamidieh
- Hematology, Oncology and Stem Cell Transplantation Research Centre, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Anjam Najemdini
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahshid Mohammadi Pour
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mahin Nikougoftar Zarif
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.
| |
Collapse
|
34
|
Chen D, Song X, Zhang Y, Kong L, Wang H, Yu J. Optimizing intrapleural bevacizumab dosing in non-small-cell lung cancer-mediated malignant pleural effusion: less is more. Future Oncol 2018; 14:2131-2138. [PMID: 29546764 DOI: 10.2217/fon-2018-0089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
AIM Intrapleural infusion of bevacizumab (BEV) is an emerging clinical treatment for malignant pleural effusion, but many details of usage need to be determined, especially for the effective dose. PATIENTS & METHODS We performed a retrospective study of the records of malignant pleural effusion patients from non-small-cell lung cancer who underwent intrapleural infusion of BEV. According to the BEV dose commonly used in clinical, patients were allocated into either low-dose group or high-dose group. RESULTS A total of 71 patients were enrolled in this study. Administration with intrapleural BEV in low dose has less toxicity. For survival data, low- and high-dose groups have no difference. CONCLUSION Lower rates of serious BEV-related toxicities and similar survival date are noted when lower dosages are used without diminishing positive clinical impact.
Collapse
Affiliation(s)
- Dawei Chen
- Department of Radiotherapy, Shandong Cancer Hospital Affiliated to Shandong University, Jinan 250117, Shandong, PR China
| | - Xinyu Song
- School of Medicine & Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan 250000, Shandong, PR China.,Department of Internal Medicine-Oncology, Shandong Cancer Hospital & Institute, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan 250117, Shandong, PR China
| | - Yan Zhang
- Department of Radiotherapy, Shandong Cancer Hospital Affiliated to Shandong University, Jinan 250117, Shandong, PR China
| | - Li Kong
- Department of Radiotherapy, Shandong Cancer Hospital Affiliated to Shandong University, Jinan 250117, Shandong, PR China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital & Institute, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan 250117, Shandong, PR China
| | - Jinming Yu
- Department of Radiotherapy, Shandong Cancer Hospital Affiliated to Shandong University, Jinan 250117, Shandong, PR China.,School of Medicine & Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan 250000, Shandong, PR China
| |
Collapse
|
35
|
Perez-Castillo Y, Sánchez-Rodríguez A, Tejera E, Cruz-Monteagudo M, Borges F, Cordeiro MNDS, Le-Thi-Thu H, Pham-The H. A desirability-based multi objective approach for the virtual screening discovery of broad-spectrum anti-gastric cancer agents. PLoS One 2018; 13:e0192176. [PMID: 29420638 PMCID: PMC5805264 DOI: 10.1371/journal.pone.0192176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/17/2018] [Indexed: 01/09/2023] Open
Abstract
Gastric cancer is the third leading cause of cancer-related mortality worldwide and despite advances in prevention, diagnosis and therapy, it is still regarded as a global health concern. The efficacy of the therapies for gastric cancer is limited by a poor response to currently available therapeutic regimens. One of the reasons that may explain these poor clinical outcomes is the highly heterogeneous nature of this disease. In this sense, it is essential to discover new molecular agents capable of targeting various gastric cancer subtypes simultaneously. Here, we present a multi-objective approach for the ligand-based virtual screening discovery of chemical compounds simultaneously active against the gastric cancer cell lines AGS, NCI-N87 and SNU-1. The proposed approach relays in a novel methodology based on the development of ensemble models for the bioactivity prediction against each individual gastric cancer cell line. The methodology includes the aggregation of one ensemble per cell line using a desirability-based algorithm into virtual screening protocols. Our research leads to the proposal of a multi-targeted virtual screening protocol able to achieve high enrichment of known chemicals with anti-gastric cancer activity. Specifically, our results indicate that, using the proposed protocol, it is possible to retrieve almost 20 more times multi-targeted compounds in the first 1% of the ranked list than what is expected from a uniform distribution of the active ones in the virtual screening database. More importantly, the proposed protocol attains an outstanding initial enrichment of known multi-targeted anti-gastric cancer agents.
Collapse
Affiliation(s)
- Yunierkis Perez-Castillo
- Escuela de Ciencias Físicas y Matemáticas, Universidad de Las Américas, Quito, Ecuador
- * E-mail: (YPC); (HPT)
| | | | - Eduardo Tejera
- Facultad de Ingenieria y Ciencias Agropecuarias, Universidad de Las Américas, Quito, Ecuador
| | - Maykel Cruz-Monteagudo
- CIQUP/Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- REQUIMTE/Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- Department of General Education, West Coast University—Miami Campus, Doral, Florida, United States of America
| | - Fernanda Borges
- CIQUP/Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - M. Natália D. S. Cordeiro
- REQUIMTE/Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Huong Le-Thi-Thu
- VNU School of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
| | - Hai Pham-The
- Hanoi University of Pharmacy, Hanoi, Vietnam
- * E-mail: (YPC); (HPT)
| |
Collapse
|
36
|
Wu J, Chen Y, Liu X, Gao Y, Hu J, Chen H. Discovery of novel negletein derivatives as potent anticancer agents for acute myeloid leukemia. Chem Biol Drug Des 2018; 91:924-932. [PMID: 29240303 DOI: 10.1111/cbdd.13159] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/03/2017] [Accepted: 12/01/2017] [Indexed: 12/26/2022]
Abstract
Baicalin and its aglycone baicalein derived from Scutellaria baicalensis exhibited potent anticancer effects in various types of cancer cell lines. However, the unfavorable pharmaceutical properties became the main obstacle for their potential clinical development. With the aim of development of novel anticancer agents based on the skeleton of baicalin, a series of novel negletein derivatives were designed and synthesized. Among them, compound 8 (FZU-02,006) with an N,N-dimethylamino ethoxyl moiety at the C-6 position exhibited significant enhanced antiproliferative effect against HL-60 cells in vitro through regulating multisignaling pathways. These results revealed that compound 8 with the improved aqueous solubility (as HCl salt, >1 mg/ml) and enhanced antileukemia potency might serve as a promising lead for further development.
Collapse
Affiliation(s)
- Jianlei Wu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Yingyu Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xuanping Liu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Yu Gao
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Jianda Hu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Haijun Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| |
Collapse
|
37
|
Simpson A, Petnga W, Macaulay VM, Weyer-Czernilofsky U, Bogenrieder T. Insulin-Like Growth Factor (IGF) Pathway Targeting in Cancer: Role of the IGF Axis and Opportunities for Future Combination Studies. Target Oncol 2017; 12:571-597. [PMID: 28815409 PMCID: PMC5610669 DOI: 10.1007/s11523-017-0514-5] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite a strong preclinical rationale for targeting the insulin-like growth factor (IGF) axis in cancer, clinical studies of IGF-1 receptor (IGF-1R)-targeted monotherapies have been largely disappointing, and any potential success has been limited by the lack of validated predictive biomarkers for patient enrichment. A large body of preclinical evidence suggests that the key role of the IGF axis in cancer is in driving treatment resistance, via general proliferative/survival mechanisms, interactions with other mitogenic signaling networks, and class-specific mechanisms such as DNA damage repair. Consequently, combining IGF-targeted agents with standard cytotoxic agents, other targeted agents, endocrine therapies, or immunotherapies represents an attractive therapeutic approach. Anti-IGF-1R monoclonal antibodies (mAbs) do not inhibit IGF ligand 2 (IGF-2) activation of the insulin receptor isoform-A (INSR-A), which may limit their anti-proliferative activity. In addition, due to their lack of specificity, IGF-1R tyrosine kinase inhibitors are associated with hyperglycemia as a result of interference with signaling through the classical metabolic INSR-B isoform; this may preclude their use at clinically effective doses. Conversely, IGF-1/IGF-2 ligand-neutralizing mAbs inhibit proliferative/anti-apoptotic signaling via IGF-1R and INSR-A, without compromising the metabolic function of INSR-B. Therefore, combination regimens that include these agents may be more efficacious and tolerable versus IGF-1R-targeted combinations. Herein, we review the preclinical and clinical experience with IGF-targeted therapies to-date, and discuss the rationale for future combination approaches as a means to overcome treatment resistance.
Collapse
Affiliation(s)
- Aaron Simpson
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | | | - Thomas Bogenrieder
- Boehringer Ingelheim RCV, Dr. Boehringer Gasse 5-11, 1121, Vienna, Austria.
- Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Marchioninistrasse 15, 81377, Munich, Germany.
| |
Collapse
|
38
|
Ricciardi MR, Mirabilii S, Licchetta R, Piedimonte M, Tafuri A. Targeting the Akt, GSK-3, Bcl-2 axis in acute myeloid leukemia. Adv Biol Regul 2017; 65:36-58. [PMID: 28549531 DOI: 10.1016/j.jbior.2017.05.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 06/07/2023]
Abstract
Over the last few decades, there has been significant progress in the understanding of the pathogenetic mechanisms of the Acute Myeloid Leukemia (AML). However, despite important advances in elucidating molecular mechanisms, the treatment of AML has not improved significantly, remaining anchored at the standard chemotherapy regimen "3 + 7", with the prognosis of patients remaining severe, especially for the elderly and for those not eligible for transplant procedures. The biological and clinical heterogeneity of AML represents the major obstacle that hinders the improvement of prognosis and the identification of new effective therapeutic approaches. To date, abundant information has been collected on the genetic and molecular alterations of AML carrying prognostic significance. However, not enough is known on how AML progenitors regulate proliferation and survival by redundant and cross-talking signal transduction pathways (STP). Furthermore, it remains unclear how such complicated network affects prognosis and therapeutic treatment options, although many of these molecular determinants are potentially attractive for their druggable characteristics. In this review, some of the key STP frequently deregulated in AML, such as PI3k/Akt/mTOR pathway, GSK3 and components of Bcl-2 family of proteins, are summarized, highlighting in addition their interplay. Based on this information, we reviewed new targeted therapeutic approaches, focusing on the aberrant networks that sustain the AML blast proliferation, survival and drug resistance, aiming to improve disease treatment. Finally, we reported the approaches aimed at disrupting key signaling cross-talk overcoming resistances based on the combination of different targeting therapeutic strategies.
Collapse
Affiliation(s)
- Maria Rosaria Ricciardi
- Hematology, "Sant'Andrea" Hospital-Sapienza, University of Rome, Department of Clinical and Molecular Medicine, Rome, Italy
| | - Simone Mirabilii
- Hematology, "Sant'Andrea" Hospital-Sapienza, University of Rome, Department of Clinical and Molecular Medicine, Rome, Italy.
| | - Roberto Licchetta
- Hematology, "Sant'Andrea" Hospital-Sapienza, University of Rome, Department of Clinical and Molecular Medicine, Rome, Italy
| | - Monica Piedimonte
- Hematology, "Sant'Andrea" Hospital-Sapienza, University of Rome, Department of Clinical and Molecular Medicine, Rome, Italy
| | - Agostino Tafuri
- Hematology, "Sant'Andrea" Hospital-Sapienza, University of Rome, Department of Clinical and Molecular Medicine, Rome, Italy
| |
Collapse
|
39
|
Cruz-Monteagudo M, Schürer S, Tejera E, Pérez-Castillo Y, Medina-Franco JL, Sánchez-Rodríguez A, Borges F. Systemic QSAR and phenotypic virtual screening: chasing butterflies in drug discovery. Drug Discov Today 2017; 22:994-1007. [PMID: 28274840 PMCID: PMC5487293 DOI: 10.1016/j.drudis.2017.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/02/2017] [Accepted: 02/27/2017] [Indexed: 12/20/2022]
Abstract
Current advances in systems biology suggest a new change of paradigm reinforcing the holistic nature of the drug discovery process. According to the principles of systems biology, a simple drug perturbing a network of targets can trigger complex reactions. Therefore, it is possible to connect initial events with final outcomes and consequently prioritize those events, leading to a desired effect. Here, we introduce a new concept, 'Systemic Chemogenomics/Quantitative Structure-Activity Relationship (QSAR)'. To elaborate on the concept, relevant information surrounding it is addressed. The concept is challenged by implementing a systemic QSAR approach for phenotypic virtual screening (VS) of candidate ligands acting as neuroprotective agents in Parkinson's disease (PD). The results support the suitability of the approach for the phenotypic prioritization of drug candidates.
Collapse
Affiliation(s)
- Maykel Cruz-Monteagudo
- CIQUP/Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto 4169-007, Portugal.
| | - Stephan Schürer
- Department of Pharmacology, Miller School of Medicine and Center for Computational Science, University of Miami, Miami, FL 33136, USA
| | - Eduardo Tejera
- Instituto de Investigaciones Biomédicas (IIB), Universidad de Las Américas, 170513 Quito, Ecuador
| | - Yunierkis Pérez-Castillo
- Sección Físico Química y Matemáticas, Departamento de Química, Universidad Técnica Particular de Loja, San Cayetano Alto S/N, EC1101608 Loja, Ecuador
| | - José L Medina-Franco
- Universidad Nacional Autónoma de México, Departamento de Farmacia, Facultad de Química, Avenida Universidad 3000, Mexico City, 04510, Mexico
| | - Aminael Sánchez-Rodríguez
- Departamento de Ciencias Naturales, Universidad Técnica Particular de Loja, Calle París S/N, EC1101608 Loja, Ecuador
| | - Fernanda Borges
- CIQUP/Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto 4169-007, Portugal.
| |
Collapse
|
40
|
Ajlan A, Thomas P, Albakr A, Nagpal S, Recht L. Optimizing bevacizumab dosing in glioblastoma: less is more. J Neurooncol 2017; 135:99-105. [PMID: 28667595 DOI: 10.1007/s11060-017-2553-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 06/27/2017] [Indexed: 12/20/2022]
Abstract
Compared to traditional chemotherapies, where dose limiting toxicities represent the maximum possible dose, monoclonal antibody therapies are used at doses well below maximum tolerated dose. However, there has been little effort to ascertain whether there is a submaximal dose at which the efficacy/complication ratio is maximized. Thus, despite the general practice of using Bevacizumab (BEV) at dosages of 10 mg/kg every other week for glioma patients, there has not been much prior work examining whether the relatively high complication rates reported with this agent can be decreased by lowering the dose without impairing efficacy. We assessed charts from 80 patients who received BEV for glioblastoma to survey the incidence of complications relative to BEV dose. All patients were treated with standard upfront chemoradiation. The toxicity was graded based on the NCI CTCAE, version 4.03. The rate of BEV serious related adverse events was 12.5% (n = 10/80). There were no serious adverse events (≥grade 3) when the administered dose was (<3 mg/kg/week), compared to a 21% incidence in those who received higher doses (≥3 mg/kg/week) (P < 0.01). Importantly, the three patient deaths attributable to BEV administration occurred in patients receiving higher doses. Patients who received lower doses also had a better survival rate, although this did not reach statistical significance [median OS 39 for low dose group vs. 17.3 for high dose group (P = 0.07)]. Lower rates of serious BEV related toxicities are noted when lower dosages are used without diminishing positive clinical impact. Further work aimed at optimizing BEV dosage is justified.
Collapse
Affiliation(s)
- Abdulrazag Ajlan
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Neurosurgery, King Saud University, Riyadh, Saudi Arabia.
| | - Piia Thomas
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Seema Nagpal
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lawrence Recht
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
41
|
Thibault B, Jean-Claude B. Dasatinib + Gefitinib, a non platinum-based combination with enhanced growth inhibitory, anti-migratory and anti-invasive potency against human ovarian cancer cells. J Ovarian Res 2017; 10:31. [PMID: 28446239 PMCID: PMC5405511 DOI: 10.1186/s13048-017-0319-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/21/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovarian cancer is the leading cause of death for gynecological cancers and the 6th cause of women cancer death in developed countries. The late stage detection, the peritoneal dissemination and the acquisition of resistance against carboplatin are the main reasons to explain this poor prognosis and strengthen the need of alternative treatments to improve the management of ovarian cancer and/or to sensitize tumors to platinum salts. Epidermal growth factor receptor (EGFR), hepatocyte growth factor receptor (Met) and cellular Src kinase (c-Src) are crucial kinases implied in ovarian tumor growth, survival, invasion and resistance to carboplatin. Their expression is increased in advanced ovarian cancers and is correlated with poor prognosis. Despite a clear potential in inhibiting these proteins in ovarian cancer, as a single agent or in combination with a carboplatin treatment, we need to target kinases in tandem because of their capacity to trigger compensatory pathways that synergize to promote drug resistance. METHODS Here we target EGFR, c-Src and Met individually or in combination with carboplatin, using Gefitinib, Dasatinib and Crizotinib respectively, in a panel of carboplatin-sensitive (OVCAR-3, IGROV-1 and A2780) and carboplatin-resistant cells (SKOV-3 and EFO-21). We studied the ability of the most potent combination to induce apoptosis, regulate migration, invasion and to modulate the activation of proliferation and survival proteins. RESULTS Crizotinib, Dasatinib and Gefitinib, alone or in combination with carboplatin, showed a cell-specific cytotoxic synergy in ovarian cancer cells. The Dasatinib plus Gefitinib combination was synergistic in OVCAR-3, SKOV-3 and, in IGROV-1 cells (high concentrations). This combination was unable to induce apoptosis but suppressed cell migration, invasion and the activation of EGFR, Erk, c-Src and Akt compared to single treatments. CONCLUSIONS Combining carboplatin with kinase inhibitors lead to synergistic interactions in a cell-specific manner. Unlike platinum-based combinations, mixing Dasatinib with Gefitinib led to cytotoxic activity, inhibition of cell migration and invasion. Thus, the Dasatinib + Gefitinib combination presents anti-tumour properties that are superior to those of platinum-based combinations, indicating that it may well represent a promising new treatment modality to be tested in the clinic.
Collapse
Affiliation(s)
- Benoît Thibault
- Research Institute - McGill University Health Center (MUHC), 1001 Décarie Blvd, Block E, Montreal, QC, H4A 3J1, Canada.,Present Address: INSERM - Cancer Research Center of Toulouse (CRCT), 2 avenue Hubert Curien, Toulouse, France
| | - Bertrand Jean-Claude
- Research Institute - McGill University Health Center (MUHC), 1001 Décarie Blvd, Block E, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
42
|
Cho SH, Park SM, Lee HS, Lee HY, Cho KH. Attractor landscape analysis of colorectal tumorigenesis and its reversion. BMC SYSTEMS BIOLOGY 2016; 10:96. [PMID: 27765040 PMCID: PMC5072344 DOI: 10.1186/s12918-016-0341-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 10/10/2016] [Indexed: 02/08/2023]
Abstract
Background Colorectal cancer arises from the accumulation of genetic mutations that induce dysfunction of intracellular signaling. However, the underlying mechanism of colorectal tumorigenesis driven by genetic mutations remains yet to be elucidated. Results To investigate colorectal tumorigenesis at a system-level, we have reconstructed a large-scale Boolean network model of the human signaling network by integrating previous experimental results on canonical signaling pathways related to proliferation, metastasis, and apoptosis. Throughout an extensive simulation analysis of the attractor landscape of the signaling network model, we found that the attractor landscape changes its shape by expanding the basin of attractors for abnormal proliferation and metastasis along with the accumulation of driver mutations. A further hypothetical study shows that restoration of a normal phenotype might be possible by reversely controlling the attractor landscape. Interestingly, the targets of approved anti-cancer drugs were highly enriched in the identified molecular targets for the reverse control. Conclusions Our results show that the dynamical analysis of a signaling network based on attractor landscape is useful in acquiring a system-level understanding of tumorigenesis and developing a new therapeutic strategy. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0341-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sung-Hwan Cho
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sang-Min Park
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Ho-Sung Lee
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hwang-Yeol Lee
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Kwang-Hyun Cho
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea. .,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
43
|
Ma YC, Wang ZX, Jin SJ, Zhang YX, Hu GQ, Cui DT, Wang JS, Wang M, Wang FQ, Zhao ZJ. Dual Inhibition of Topoisomerase II and Tyrosine Kinases by the Novel Bis-Fluoroquinolone Chalcone-Like Derivative HMNE3 in Human Pancreatic Cancer Cells. PLoS One 2016; 11:e0162821. [PMID: 27760157 PMCID: PMC5070812 DOI: 10.1371/journal.pone.0162821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 08/29/2016] [Indexed: 01/05/2023] Open
Abstract
Both tyrosine kinase and topoisomerase II (TopII) are important anticancer targets, and their respective inhibitors are widely used in cancer therapy. However, some combinations of anticancer drugs could exhibit mutually antagonistic actions and drug resistance, which further limit their therapeutic efficacy. Here, we report that HMNE3, a novel bis-fluoroquinolone chalcone-like derivative that targets both tyrosine kinase and TopII, induces tumor cell proliferation and growth inhibition. The viabilities of 6 different cancer cell lines treated with a range of HMNE3 doses were detected using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cellular apoptosis was determined using Hoechst 33258 fluorescence staining and the terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling (TUNEL) assay. The expression of activated Caspase-3 was examined by immunocytochemistry. The tyrosine kinase activity was measured with a human receptor tyrosine kinase (RTK) detection kit using a horseradish peroxidase (HRP)-conjugated phosphotyrosine (pY20) antibody as the substrate. The topoisomerase II activity was measured using agarose gel electrophoresis with the DNA plasmid pBR322 as the substrate. The expression levels of the P53, Bax, Bcl-2, Caspase-3, -8, -9, p-cSrc, c-Src and topoisomerase II proteins were detected by western blot analysis. The proliferation of five of the six cancer cell lines was significantly inhibited by HMNE3 at 0.312 to 10 μmol/L in a time- and dose-dependent manner. Treatment of the Capan-1 and Panc-1 cells with 1.6 to 3.2 μM HMNE3 for 48 h significantly increased the percentage of apoptotic cells (P<0.05), and this effect was accompanied by a decrease in tyrosine kinase activity. HMNE3 potentially inhibited tyrosine kinase activity in vitro with an IC50 value of 0.64±0.34 μmol/L in Capan-1 cells and 3.1±0.86 μmol/L in Panc-1 cells. The activity of c-Src was significantly inhibited by HMNE3 in a dose- and time-dependent manner in different cellular contexts. Compared with the control group, HMNE3 induced increased expression of cellular apoptosis-related proteins. Consistent with cellular apoptosis data, a significant decrease in topoisomerase IIβ activity was noted following treatment with HMNE3 for 24 h. Our data suggest that HMNE3 induced apoptosis in Capan-1 and Panc-1 cells by inhibiting the activity of both tyrosine kinases and topoisomerase II.
Collapse
Affiliation(s)
- Yong-Chao Ma
- Clinical Medical Institute of Luohe Medical College, Luohe, Henan, P.R. China
- The First Affiliated Hospital of Luohe Medical College, Luohe, Henan, P.R. China
- Tumor Occurrence and Prevention Research Innovation team of Luohe, Luohe, Henan, P.R. China
| | - Zhi-Xin Wang
- Basic Medical Institute of ZhengZhou University, ZhengZhou, Henan, P.R. China
| | - Shao-Ju Jin
- Institute of pharmacology of Luohe Medical College, Luohe, Henan, P.R. China
| | - Yan-Xin Zhang
- Basic Medical Institute of Luohe Medical College, Luohe, Henan, P.R. China
| | - Guo-Qiang Hu
- Institute of Chemistry and Biology of Henan University, Kaifeng, Henan, P.R. China
| | - Dong-Tao Cui
- Clinical Medical Institute of Luohe Medical College, Luohe, Henan, P.R. China
| | - Jiang-Shuan Wang
- Basic Medical Institute of Luohe Medical College, Luohe, Henan, P.R. China
| | - Min Wang
- Clinical Medical Institute of Luohe Medical College, Luohe, Henan, P.R. China
| | - Fu-Qing Wang
- Basic Medical Institute of Luohe Medical College, Luohe, Henan, P.R. China
| | - Zhi-Jun Zhao
- Clinical Medical Institute of Luohe Medical College, Luohe, Henan, P.R. China
| |
Collapse
|
44
|
von Manstein V, Groner B. Tumor cell resistance against targeted therapeutics: the density of cultured glioma tumor cells enhances Stat3 activity and offers protection against the tyrosine kinase inhibitor canertinib. MEDCHEMCOMM 2016; 8:96-102. [PMID: 30108694 PMCID: PMC6072326 DOI: 10.1039/c6md00463f] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 10/04/2016] [Indexed: 01/03/2023]
Abstract
Tumor cell resistance to drug treatment severely limits the therapeutic success of treatment.
Tumor cell resistance to drug treatment severely limits the therapeutic success of treatment. Tumor cells, exposed to chemotherapeutic drugs, have developed intricate strategies to escape the cytotoxic effects and adapt to adverse conditions. The molecular mechanisms causing drug resistance can be based upon modifications of drug transport or metabolism, structural alterations of drug targets or adaptation of cellular signaling. An important component in the transformation of cells and the emergence of drug resistance is the activation of the transcription factor Stat3. The persistent, inappropriate activation of Stat3 causes the expression of target genes which promote tumor cell proliferation, survival, invasion and immune suppression, and it is instrumental in the process of the emergence of resistance to both conventional chemotherapeutic agents and novel targeted compounds. For these reasons, Stat3 inhibition is being pursued as a promising therapeutic strategy. We have investigated the effects of the tyrosine kinase inhibitor canertinib on the glioma cell line Tu-2449. In these cells Stat3 is persistently phosphorylated and activated downstream of the oncogenic driver v-Src and its effector, the cytoplasmic tyrosine kinase Bmx. Canertinib exposure of Tu-2449 cells rapidly caused the inhibition of the Bmx kinase and the deactivation of Stat3. Prolonged exposure of the cells to canertinib caused the death of the large majority of the cells. Only a few cells became resistant to canertinib and survived in tight clusters. These cells have become drug resistant. When the canertinib resistant cells were expanded and cultured at lower cell densities, they regained their sensitivity towards canertinib. We measured the extent of Stat3 activation as a function of cell density and found that higher cell densities are accompanied by increased Stat3 activation and a higher expression of Stat3 target genes. We suggest that Stat3 induction through tight cell–cell interactions, most likely through the engagement of cadherins, can counteract the inhibitory effects exerted by canertinib on Bmx. Cell–cell interactions induced Stat3 and compensated for the suppression of Stat3 by canertinib, thus transiently protecting the cells from the cytotoxic effects of the inhibitor.
Collapse
Affiliation(s)
- V von Manstein
- Georg Speyer Haus , Institute for Tumor Biology and Experimental Therapy , Paul Ehrlich Str. 42 , 60596 Frankfurt am Main , Germany . ; Tel: +49 6963395180
| | - B Groner
- Georg Speyer Haus , Institute for Tumor Biology and Experimental Therapy , Paul Ehrlich Str. 42 , 60596 Frankfurt am Main , Germany . ; Tel: +49 6963395180
| |
Collapse
|
45
|
Kathiriya JJ, Pathak RR, Bezginov A, Xue B, Uversky VN, Tillier ERM, Davé V. Structural pliability adjacent to the kinase domain highlights contribution of FAK1 IDRs to cytoskeletal remodeling. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1865:43-54. [PMID: 27718363 DOI: 10.1016/j.bbapap.2016.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/27/2016] [Accepted: 10/03/2016] [Indexed: 12/24/2022]
Abstract
Therapeutic protein kinase inhibitors are designed on the basis of kinase structures. Here, we define intrinsically disordered regions (IDRs) in structurally hybrid kinases. We reveal that 65% of kinases have an IDR adjacent to their kinase domain (KD). These IDRs are evolutionarily more conserved than IDRs distant to KDs. Strikingly, 36 kinases have adjacent IDRs extending into their KDs, defining a unique structural and functional subset of the kinome. Functional network analysis of this subset of the kinome uncovered FAK1 as topologically the most connected hub kinase. We identify that KD-flanking IDR of FAK1 is more conserved and undergoes more post-translational modifications than other IDRs. It preferentially interacts with proteins regulating scaffolding and kinase activity, which contribute to cytoskeletal remodeling. In summary, spatially and evolutionarily conserved IDRs in kinases may influence their functions, which can be exploited for targeted therapies in diseases including those that involve aberrant cytoskeletal remodeling.
Collapse
Affiliation(s)
- Jaymin J Kathiriya
- Morsani College of Medicine, Department of Pathology and Cell Biology, University of South Florida, Tampa, FL 33612, United States
| | - Ravi Ramesh Pathak
- Morsani College of Medicine, Department of Pathology and Cell Biology, University of South Florida, Tampa, FL 33612, United States
| | - Alexandr Bezginov
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Bin Xue
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; USF Health Byrd Alzheimer's Research Institute, University of South Florida, Tampa, FL 33612, United States; Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russian Federation
| | | | - Vrushank Davé
- Morsani College of Medicine, Department of Pathology and Cell Biology, University of South Florida, Tampa, FL 33612, United States; Department of Cancer Biology and Evolution, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States.
| |
Collapse
|
46
|
Jimbo M, Blanco FF, Huang YH, Telonis AG, Screnci BA, Cosma GL, Alexeev V, Gonye GE, Yeo CJ, Sawicki JA, Winter JM, Brody JR. Targeting the mRNA-binding protein HuR impairs malignant characteristics of pancreatic ductal adenocarcinoma cells. Oncotarget 2016; 6:27312-31. [PMID: 26314962 PMCID: PMC4694992 DOI: 10.18632/oncotarget.4743] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/13/2015] [Indexed: 12/21/2022] Open
Abstract
Post-transcriptional regulation is a powerful mediator of gene expression, and can rapidly alter the expression of numerous transcripts involved in tumorigenesis. We have previously shown that the mRNA-binding protein HuR (ELAVL1) is elevated in human pancreatic ductal adenocarcinoma (PDA) specimens compared to normal pancreatic tissues, and its cytoplasmic localization is associated with increased tumor stage. To gain a better insight into HuR’s role in PDA biology and to assess it as a candidate therapeutic target, we altered HuR expression in PDA cell lines and characterized the resulting phenotype in preclinical models. HuR silencing by short hairpin and small interfering RNAs significantly decreased cell proliferation and anchorage-independent growth, as well as impaired migration and invasion. In comparison, HuR overexpression increased migration and invasion, but had no significant effects on cell proliferation and anchorage-independent growth. Importantly, two distinct targeted approaches to HuR silencing showed marked impairment in tumor growth in mouse xenografts. NanoString nCounter® analyses demonstrated that HuR regulates core biological processes, highlighting that HuR inhibition likely thwarts PDA viability through post-transcriptional regulation of diverse signaling pathways (e.g. cell cycle, apoptosis, DNA repair). Taken together, our study suggests that targeted inhibition of HuR may be a novel, promising approach to the treatment of PDA.
Collapse
Affiliation(s)
- Masaya Jimbo
- Department of Surgery and The Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Fernando F Blanco
- Department of Surgery and The Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Pharmacology & Experimental Therapeutics, Division of Clinical Pharmacology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Yu-Hung Huang
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Aristeidis G Telonis
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Brad A Screnci
- Department of Surgery and The Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gabriela L Cosma
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Vitali Alexeev
- Department of Dermatology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Charles J Yeo
- Department of Surgery and The Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Jordan M Winter
- Department of Surgery and The Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jonathan R Brody
- Department of Surgery and The Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
47
|
Bon G, Loria R, Amoreo CA, Verdina A, Sperduti I, Mastrofrancesco A, Soddu S, Diodoro MG, Mottolese M, Todaro M, Stassi G, Milella M, De Maria R, Falcioni R. Dual targeting of HER3 and MEK may overcome HER3-dependent drug-resistance of colon cancers. Oncotarget 2016; 8:108463-108479. [PMID: 29312543 PMCID: PMC5752456 DOI: 10.18632/oncotarget.11400] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/10/2016] [Indexed: 01/06/2023] Open
Abstract
Although the medical treatment of colorectal cancer has evolved greatly in the last years, a significant portion of early-stage patients develops recurrence after therapies. The current clinical trials are directed to evaluate new drug combinations and treatment schedules. By the use of patient-derived or established colon cancer cell lines, we found that the tyrosine kinase receptor HER3 is involved in the mechanisms of resistance to therapies. In agreement, the immunohistochemical analysis of total and phospho-HER3 expression in 185 colorectal cancer specimens revealed a significant correlation with lower disease-free survival. Targeting HER3 by the use of the monoclonal antibody patritumab we found induction of growth arrest in all cell lines. Despite the high efficiency of patritumab in abrogating the HER3-dependent activation of PI3K pathway, the HER2 and EGFR-dependent MAPK pathway is activated as a compensatory mechanism. Interestingly, we found that the MEK-inhibitor trametinib inhibits, as expected, the MAPK pathway but induces the HER3-dependent activation of PI3K pathway. The combined treatment results in the abrogation of both PI3K and MAPK pathways and in a significant reduction of cell proliferation and survival. These data suggest a new strategy of therapy for HER3-overexpressing colon cancers.
Collapse
Affiliation(s)
- Giulia Bon
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Rossella Loria
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Carla Azzurra Amoreo
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alessandra Verdina
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Isabella Sperduti
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Arianna Mastrofrancesco
- Physiopathology Laboratory of Skin, IRCCS San Gallicano Dermatological Institute, Rome, Italy
| | - Silvia Soddu
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Grazia Diodoro
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marcella Mottolese
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Matilde Todaro
- Surgical and Oncological Sciences, University of Palermo, Palermo, Italy
| | - Giorgio Stassi
- Surgical and Oncological Sciences, University of Palermo, Palermo, Italy
| | - Michele Milella
- Department of Experimental Clinical Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Rita Falcioni
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
48
|
El-Damasy AK, Cho NC, Nam G, Pae AN, Keum G. Discovery of a Nanomolar Multikinase Inhibitor (KST016366): A New Benzothiazole Derivative with Remarkable Broad-Spectrum Antiproliferative Activity. ChemMedChem 2016; 11:1587-95. [PMID: 27405013 DOI: 10.1002/cmdc.201600224] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/25/2016] [Indexed: 01/07/2023]
Abstract
Herein we report the discovery of compound 6 [KST016366; 4-((2-(3-(4-((4-ethylpiperazin-1-yl)methyl)-3-(trifluoromethyl)phenyl)ureido)benzo[d]thiazol-6-yl)oxy)picolinamide] as a new potent multikinase inhibitor through minor structural modification of our previously reported RAF kinase inhibitor A. In vitro anticancer evaluation of 6 showed substantial broad-spectrum antiproliferative activity against 60 human cancer cell lines. In particular, it showed GI50 values of 51.4 and 19 nm against leukemia K-562 and colon carcinoma KM12 cell lines, respectively. Kinase screening of compound 6 revealed its nanomolar-level inhibitory activity of certain oncogenic kinases implicated in both tumorigenesis and angiogenesis. Interestingly, 6 displays IC50 values of 0.82, 3.81, and 53 nm toward Tie2, TrkA, and ABL-1 (wild-type and T315I mutant) kinases, respectively. Moreover, 6 is orally bioavailable with a favorable in vivo pharmacokinetic profile. Compound 6 may serve as a promising candidate for further development of potent anticancer chemotherapeutics.
Collapse
Affiliation(s)
- Ashraf Kareem El-Damasy
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarangro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Republic of Korea.,Department of Biological Chemistry, Korea University of Science and Technology (UST), Gajungro 217, Youseong-gu, Daejeon, 305-350, Republic of Korea.,Department of Medicinal Chemistry, Faculty of Pharmacy, University of Mansoura, Mansoura, 35516, Egypt
| | - Nam-Chul Cho
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarangro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Republic of Korea
| | - Ghilsoo Nam
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarangro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Republic of Korea.,Department of Biological Chemistry, Korea University of Science and Technology (UST), Gajungro 217, Youseong-gu, Daejeon, 305-350, Republic of Korea
| | - Ae Nim Pae
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarangro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Republic of Korea.,Department of Biological Chemistry, Korea University of Science and Technology (UST), Gajungro 217, Youseong-gu, Daejeon, 305-350, Republic of Korea
| | - Gyochang Keum
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarangro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Republic of Korea. .,Department of Biological Chemistry, Korea University of Science and Technology (UST), Gajungro 217, Youseong-gu, Daejeon, 305-350, Republic of Korea.
| |
Collapse
|
49
|
Pantziarka P. Emergent properties of a computational model of tumour growth. PeerJ 2016; 4:e2176. [PMID: 27413638 PMCID: PMC4933089 DOI: 10.7717/peerj.2176] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 06/05/2016] [Indexed: 01/06/2023] Open
Abstract
While there have been enormous advances in our understanding of the genetic drivers and molecular pathways involved in cancer in recent decades, there also remain key areas of dispute with respect to fundamental theories of cancer. The accumulation of vast new datasets from genomics and other fields, in addition to detailed descriptions of molecular pathways, cloud the issues and lead to ever greater complexity. One strategy in dealing with such complexity is to develop models to replicate salient features of the system and therefore to generate hypotheses which reflect on the real system. A simple tumour growth model is outlined which displays emergent behaviours that correspond to a number of clinically relevant phenomena including tumour growth, intra-tumour heterogeneity, growth arrest and accelerated repopulation following cytotoxic insult. Analysis of model data suggests that the processes of cell competition and apoptosis are key drivers of these emergent behaviours. Questions are raised as to the role of cell competition and cell death in physical cancer growth and the relevance that these have to cancer research in general is discussed.
Collapse
Affiliation(s)
- Pan Pantziarka
- The George Pantziarka TP53 Trust , London , United Kingdom
| |
Collapse
|
50
|
McIntosh K, Balch C, Tiwari AK. Tackling multidrug resistance mediated by efflux transporters in tumor-initiating cells. Expert Opin Drug Metab Toxicol 2016; 12:633-44. [DOI: 10.1080/17425255.2016.1179280] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Kyle McIntosh
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Curt Balch
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Amit K. Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| |
Collapse
|