1
|
Karunaraj P, Cao E, Singh H, Luf M, Baker SJ, Reddy EP, Pfleger CM. A Drosophila model for Costello Syndrome caused by Ras mutation K117R. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.635762. [PMID: 39974993 PMCID: PMC11838535 DOI: 10.1101/2025.02.03.635762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Germline mutations that increase signaling through the Ras pathway can cause developmental disorders called RASopathies. The RASopathy Costello syndrome has been described to present with hallmarks that include short stature, intellectual disability, cardiac issues, and characteristic facial abnormalities and has been associated with gain-of-function mutations in HRas. The most common HRas mutations in Costello Syndrome occur at G12 and G13, but there are also other rare mutation sites such as K117 including HRas K117R . Ras K117R mutations are also found in colorectal cancer. Drosophila studies modeling gain-of-function in Ras primarily utilize the common cancer-associated mutation G12V, and previous Drosophila RASopathy models assessing Ras gain-of-function mutations have used human sequences for KRas G12D and HRas G12S. To augment these studies, we characterized the phenotype of engineering the rare gain-of-function mutation K117R in the Drosophila Ras sequence. We report here that constitutive low-level expression of Ras K117R increased lethality and reduced body size while also causing rough eye and ectopic wing vein phenotypes in those flies that survived to adulthood. Ras pathway inhibitors Trametinib and Rigosertib suppressed the lethality but not the reduced size phenotypes. Trametinib strongly suppressed the K117R wing vein phenotype whereas Rigosertib had only subtle effects. Trametinib is a direct MEK inhibitor. Rigosertib has been reported to have strong effects on PI3K signaling and to indirectly inhibit the Raf-ERK branch. Therefore, this data is consistent with an interpretation that some lethality in the fly Ras K117R model depends on elevated signaling through the Raf-ERK branch and potentially some lethality depends on the PI3K branch. In contrast, the lack of effects on the reduced size phenotypes would be consistent with small stature resulting from Raf- and PI3K-independent processes. We propose that this model can be useful for future mechanistic analysis and pharmacological screening and evaluation.
Collapse
|
2
|
Xiao YC, Chen FE. The vinyl sulfone motif as a structural unit for novel drug design and discovery. Expert Opin Drug Discov 2024; 19:239-251. [PMID: 37978948 DOI: 10.1080/17460441.2023.2284201] [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: 09/01/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Vinyl sulfones are a special sulfur-containing structural unit that have attracted considerable attention, owing to their important role in serving as key structural motifs of various biologically active compounds as well as serving as versatile building blocks for organic transformations. The synthetic strategy of vinyl sulfone derivatives has been substantially upgraded over the past 30 years, and the wide application of this functional group in drug design and discovery has been promoted. AREA COVERED In this review, the authors review the application of vinyl sulfones in drug discovery and select optimized compounds which might have significant impact or potential inspiration for drug design. EXPERT OPINION Vinyl sulfones have been reported to target various macromolecular targets via non-covalent or covalent interactions, including multiple kinases, tubulin, cysteine protease, transcription factor, and so on. Thus, it has been significantly applied as a privileged scaffold in the design of anticancer, anti-infective, anti-inflammatory, and neuroprotective agents. However, much work remains to be done to improve the drug-like properties, such as chemical and metabolic stability, ADME, and toxicity. Besides, the chemical space of vinyl sulfones needs to be expanded, including but not limited to the design of constrained endocyclic and exocyclic vinyl sulfones.
Collapse
Affiliation(s)
- You-Cai Xiao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Ministry of Education and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Fen-Er Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Ministry of Education and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Ghorbanzadeh Neghab M, Jalili-Nik M, Soltani A, Afshari AR, Hassanian SM, Rafatpanah H, Rezaee SA, Sadeghnia HR, Ataei Azimi S, Mashkani B. Rigosertib is more potent than wortmannin and rapamycin against adult T-cell leukemia-lymphoma. Biofactors 2023; 49:1174-1188. [PMID: 37345860 DOI: 10.1002/biof.1985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/09/2023] [Indexed: 06/23/2023]
Abstract
Human T lymphotropic virus type 1 (HTLV-1) infection can cause adult T-cell lymphoblastic leukemia (ATLL), an incurable, chemotherapy-resistant malignancy. In a quest for new therapeutic targets, our study sought to determine the levels of AKT, mTOR, and PI3K in ATLL MT-2 cells, HTLV-1 infected NIH/3T3 cells (Inf-3T3), and HTLV-1 infected patients (Carrier, HAM/TSP, and ATLL). Furthermore, the effects of rigosertib, wortmannin, and rapamycin on the PI3K/Akt/mTOR pathway to inhibit the proliferation of ATLL cells were examined. The results showed that mRNA expression of Akt/PI3K/mTOR was down-regulated in carrier, HAM/TSP, and ATLL patients, as well as MT-2, and Inf-3T3 cells, compared to the healthy individuals and untreated MT-2 and Inf-3T3 as controls. However, western blotting revealed an increase in the phosphorylated and activated forms of AKT and mTOR. Treating the cells with rapamycin, wortmannin, and rigosertib decreased the phosphorylated forms of Akt and mTOR and restored their mRNA expression levels. Using these inhibitors also significantly boosted the expression of the pro-apoptotic genes, Bax/Bcl-2 ratio as well as the expression of the tumor suppressor gene p53 in the MT-2 and Inf-3T3cells. Rigosertib was more potent than wortmannin and rapamycin in inducing sub-G1 and G2-M cell cycle arrest, as well as late apoptosis in the Inf-3T3 and MT-2 cells. It also synergized the cytotoxic effects of vincristine. These findings demonstrate that HTLV-1 downregulation of the mRNA level may occur as a negative feedback response to increased PI3K-Akt-mTOR phosphorylation by HTLV-1. Therefore, using rigosertib alone or in combination with common chemotherapy drugs may be beneficial in ATLL patients.
Collapse
Affiliation(s)
| | - Mohammad Jalili-Nik
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Soltani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Department of Medical Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Abdolrahim Rezaee
- Department of Medical Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid R Sadeghnia
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajad Ataei Azimi
- Department of Hematology Oncology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Baratali Mashkani
- Bioinformatics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Aguayo F, Perez-Dominguez F, Osorio JC, Oliva C, Calaf GM. PI3K/AKT/mTOR Signaling Pathway in HPV-Driven Head and Neck Carcinogenesis: Therapeutic Implications. BIOLOGY 2023; 12:biology12050672. [PMID: 37237486 DOI: 10.3390/biology12050672] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023]
Abstract
High-risk human papillomaviruses (HR-HPVs) are the causal agents of cervical, anogenital and a subset of head and neck carcinomas (HNCs). Indeed, oropharyngeal cancers are a type of HNC highly associated with HR-HPV infections and constitute a specific clinical entity. The oncogenic mechanism of HR-HPV involves E6/E7 oncoprotein overexpression for promoting cell immortalization and transformation, through the downregulation of p53 and pRB tumor suppressor proteins, among other cellular targets. Additionally, E6/E7 proteins are involved in promoting PI3K/AKT/mTOR signaling pathway alterations. In this review, we address the relationship between HR-HPV and PI3K/AKT/mTOR signaling pathway activation in HNC with an emphasis on its therapeutic importance.
Collapse
Affiliation(s)
- Francisco Aguayo
- Departamento de Biomedicina, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile
| | - Francisco Perez-Dominguez
- Laboratorio de Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| | - Julio C Osorio
- Laboratorio de Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| | - Carolina Oliva
- Laboratorio de Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| | - Gloria M Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile
| |
Collapse
|
5
|
Monfort-Vengut A, de Cárcer G. Lights and Shadows on the Cancer Multi-Target Inhibitor Rigosertib (ON-01910.Na). Pharmaceutics 2023; 15:pharmaceutics15041232. [PMID: 37111716 PMCID: PMC10145883 DOI: 10.3390/pharmaceutics15041232] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Rigosertib (ON-01910.Na) is a small-molecule member of the novel synthetic benzyl-styryl-sulfonate family. It is currently in phase III clinical trials for several myelodysplastic syndromes and leukemias and is therefore close to clinical translation. The clinical progress of rigosertib has been hampered by a lack of understanding of its mechanism of action, as it is currently considered a multi-target inhibitor. Rigosertib was first described as an inhibitor of the mitotic master regulator Polo-like kinase 1 (Plk1). However, in recent years, some studies have shown that rigosertib may also interact with the PI3K/Akt pathway, act as a Ras-Raf binding mimetic (altering the Ras signaling pathway), as a microtubule destabilizing agent, or as an activator of a stress-induced phospho-regulatory circuit that ultimately hyperphosphorylates and inactivates Ras signaling effectors. Understanding the mechanism of action of rigosertib has potential clinical implications worth exploring, as it may help to tailor cancer therapies and improve patient outcomes.
Collapse
Affiliation(s)
- Ana Monfort-Vengut
- Cell Cycle and Cancer Biomarkers Group, Instituto de Investigaciones Biomédicas Alberto Sols (IIBM) CSIC-UAM, 28029 Madrid, Spain
| | - Guillermo de Cárcer
- Cell Cycle and Cancer Biomarkers Group, Instituto de Investigaciones Biomédicas Alberto Sols (IIBM) CSIC-UAM, 28029 Madrid, Spain
| |
Collapse
|
6
|
Subversion of a protein-microRNA signaling pathway by hepatitis C virus. Proc Natl Acad Sci U S A 2023; 120:e2220406120. [PMID: 36649406 PMCID: PMC9942813 DOI: 10.1073/pnas.2220406120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
7
|
Malacrida A, Deschamps-Wright M, Rigolio R, Cavaletti G, Miloso M. Another Brick to Confirm the Efficacy of Rigosertib as Anticancer Agent. Int J Mol Sci 2023; 24:1721. [PMID: 36675237 PMCID: PMC9866497 DOI: 10.3390/ijms24021721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Rigosertib is a small molecule in preclinical development that, due to its characteristics as a dual PLK1 and PI3K inhibitor, is particularly effective in counteracting the advance of different types of tumors. In this work, we evaluated the efficacy of Rigosertib and the expression of p53 in five different human tumor cell lines in vitro, A549 (lung adenocarcinoma), MCF-7 and MDA-MB231 (breast cancer cells), RPMI 8226 (multiple myeloma), and U87-MG (glioblastoma). We demonstrated that in all cell lines, the effect was dose- and time-dependent, but A549 cells were the most sensible to the treatment while higher concentrations were required for the most resistant cell line U87-MG. Moreover, the highest and lowest p53 levels have been observed, respectively, in A459 and U87-MG cells. The alterations in the cell cycle and in cell-cycle-related proteins were observed in A549 at lower concentrations than U87-MG. In conclusion, with this article we have demonstrated that Rigosertib has different efficacy depending on the cell line considered and that it could be a potential antineoplastic agent against lung cancer in humans.
Collapse
Affiliation(s)
- Alessio Malacrida
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, MB, Italy
| | | | | | | | | |
Collapse
|
8
|
Hsueh WT, Chen SH, Chien CH, Chou SW, Chi PI, Chu JM, Chang KY. SOD2 Enhancement by Long-Term Inhibition of the PI3K Pathway Confers Multi-Drug Resistance and Enhanced Tumor-Initiating Features in Head and Neck Cancer. Int J Mol Sci 2021; 22:ijms222011260. [PMID: 34681918 PMCID: PMC8537886 DOI: 10.3390/ijms222011260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 11/16/2022] Open
Abstract
The phosphoinositide-3-kinase (PI3K) pathway has widely been considered as a potential therapeutic target for head and neck cancer (HNC); however, the application of PI3K inhibitors is often overshadowed by the induction of drug resistance with unknown mechanisms. In this study, PII3K inhibitor resistant cancer cells were developed by prolonged culturing of cell lines with BEZ235, a dual PI3K and mammalian target of rapamycin (mTOR) inhibitor. The drug resistant HNC cells showed higher IC50 of the proliferation to inhibitors specifically targeting PI3K and/or mTOR, as compared to their parental cells. These cells also showed profound resistance to drugs of other classes. Molecular analysis revealed persistent activation of phosphorylated AKT at threonine 308 in the drug resistant cells and increased expression of markers for tumor-initiating cells. Interestingly, increased intra-cellular ROS levels were observed in the drug resistant cells. Among anti-oxidant molecules, the expression of SOD2 was increased and was associated with the ALDH-positive tumor-initiating cell features. Co-incubation of SOD inhibitors and BEZ235 decreased the stemness feature of the cells in vitro, as shown by results of the spheroid formation assay. In conclusion, dysregulation of SOD2 might contribute to the profound resistance to PI3K inhibitors and the other drugs in HNC cells.
Collapse
Affiliation(s)
- Wei-Ting Hsueh
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70456, Taiwan; (W.-T.H.); (S.-H.C.)
| | - Shang-Hung Chen
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70456, Taiwan; (W.-T.H.); (S.-H.C.)
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
| | - Chia-Hung Chien
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
- School of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Shao-Wen Chou
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
| | - Pei-I Chi
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
| | - Jui-Mei Chu
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
| | - Kwang-Yu Chang
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70456, Taiwan; (W.-T.H.); (S.-H.C.)
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
- Correspondence: ; Tel.: +886-6-208-3422
| |
Collapse
|
9
|
Rahmani F, Hashemzehi M, Avan A, Barneh F, Asgharzadeh F, Moradi Marjaneh R, Soleimani A, Parizadeh M, Ferns GA, Ghayour Mobarhan M, Ryzhikov M, Afshari AR, Ahmadian MR, Giovannetti E, Jafari M, Khazaei M, Hassanian SM. Rigosertib elicits potent anti-tumor responses in colorectal cancer by inhibiting Ras signaling pathway. Cell Signal 2021; 85:110069. [PMID: 34214591 DOI: 10.1016/j.cellsig.2021.110069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 06/02/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND The therapeutic potency of Rigosertib (RGS) in the treatment of the myelodysplastic syndrome has been investigated previously, but little is known about its mechanisms of action. METHODS The present study integrates systems and molecular biology approaches to investigate the mechanisms of the anti-tumor effects of RGS, either alone or in combination with 5-FU in cellular and animal models of colorectal cancer (CRC). RESULTS The effects of RGS were more pronounced in dedifferentiated CRC cell types, compared to cell types that were epithelial-like. RGS inhibited cell proliferation and cell cycle progression in a cell-type specific manner, and that was dependent on the presence of mutations in KRAS, or its down-stream effectors. RGS increased both early and late apoptosis, by regulating the expression of p53, BAX and MDM2 in tumor model. We also found that RGS induced cell senescence in tumor tissues by increasing ROS generation, and impairing oxidant/anti-oxidant balance. RGS also inhibited angiogenesis and metastatic behavior of CRC cells, by regulating the expression of CD31, E-cadherin, and matrix metalloproteinases-2 and 9. CONCLUSION Our findings support the therapeutic potential of this potent RAS signaling inhibitor either alone or in combination with standard regimens for the management of patients with CRC.
Collapse
Affiliation(s)
- Farzad Rahmani
- Iranshahr University of Medical Sciences, Iranshahr, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Milad Hashemzehi
- Tropical and Communicable Diseases Research Centre, Iranshahr University of Medical Sciences, Iranshahr, Iran; Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Science, Mashhad, Iran
| | - Farnaz Barneh
- Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Asgharzadeh
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhaneh Moradi Marjaneh
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atena Soleimani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Parizadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Science, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Majid Ghayour Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Science, Mashhad, Iran
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, Washington University, School of Medicine, Saint Louis, MO, USA
| | - Amir Reza Afshari
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine University, Düsseldorf, Germany
| | - Elisa Giovannetti
- Cancer Pharmacology Lab, AIRC Start-up, University Hospital of Pisa, Pisa, Italy; Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Mohieddin Jafari
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Finland
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Science, Mashhad, Iran.
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Science, Mashhad, Iran.
| |
Collapse
|
10
|
Malacrida A, Rigolio R, Celio L, Damian S, Cavaletti G, Mazzaferro V, Miloso M. In Vitro Evaluation of Rigosertib Antitumoral and Radiosensitizing Effects against Human Cholangiocarcinoma Cells. Int J Mol Sci 2021; 22:8230. [PMID: 34360994 PMCID: PMC8348961 DOI: 10.3390/ijms22158230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022] Open
Abstract
Cholangiocarcinoma is the first most common cancer of the biliary tract. To date, surgical resection is the only potentially curative option, but it is possible only for a limited percentage of patients, and in any case survival rate is quite low. Moreover, cholangiocarcinoma is often chemotherapy-resistant, and the only drug with a significant benefit for patient's survival is Gemcitabine. It is necessary to find new drugs or combination therapies to treat nonresectable cholangiocarcinoma and improve the overall survival rate of patients. In this work, we evaluate in vitro the antitumoral effects of Rigosertib, a multi-kinase inhibitor in clinical development, against cholangiocarcinoma EGI-1 cell lines. Rigosertib impairs EGI-1 cell viability in a dose- and time-dependent manner, reversibility is dose-dependent, and significant morphological and nuclear alterations occur. Moreover, Rigosertib induces the arrest of the cell cycle in the G2/M phase, increases autophagy, and inhibits proteasome, cell migration, and invasion. Lastly, Rigosertib shows to be a stronger radiosensitizer than Gemcitabine and 5-Fluorouracil. In conclusion, Rigosertib could be a potential therapeutic option, alone or in combination with radiations, for nonresectable patients with cholangiocarcinoma.
Collapse
Affiliation(s)
- Alessio Malacrida
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano—Bicocca, Via Cadore 48, 20900 Monza, Italy; (R.R.); (G.C.); (M.M.)
| | - Roberta Rigolio
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano—Bicocca, Via Cadore 48, 20900 Monza, Italy; (R.R.); (G.C.); (M.M.)
| | - Luigi Celio
- Medical Oncology Unit, ASST del Garda, 25015 Desenzano del Garda, Italy;
| | - Silvia Damian
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori—Via Giacomo Venezian 1, 20133 Milano, Italy;
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano—Bicocca, Via Cadore 48, 20900 Monza, Italy; (R.R.); (G.C.); (M.M.)
| | - Vincenzo Mazzaferro
- Department of Hepatology, Hepato Pancreatic Biliary Surgery and Liver Transplantation, Istituto Nazionale Tumori, Foundation IRCCS, 20133 Milan, Italy;
- Department of Oncology, University of Milan, 20122 Milan, Italy
| | - Mariarosaria Miloso
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano—Bicocca, Via Cadore 48, 20900 Monza, Italy; (R.R.); (G.C.); (M.M.)
| |
Collapse
|
11
|
Yan C, Saleh N, Yang J, Nebhan CA, Vilgelm AE, Reddy EP, Roland JT, Johnson DB, Chen SC, Shattuck-Brandt RL, Ayers GD, Richmond A. Novel induction of CD40 expression by tumor cells with RAS/RAF/PI3K pathway inhibition augments response to checkpoint blockade. Mol Cancer 2021; 20:85. [PMID: 34092233 PMCID: PMC8182921 DOI: 10.1186/s12943-021-01366-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND While immune checkpoint blockade (ICB) is the current first-line treatment for metastatic melanoma, it is effective for ~ 52% of patients and has dangerous side effects. The objective here was to identify the feasibility and mechanism of RAS/RAF/PI3K pathway inhibition in melanoma to sensitize tumors to ICB therapy. METHODS Rigosertib (RGS) is a non-ATP-competitive small molecule RAS mimetic. RGS monotherapy or in combination therapy with ICB were investigated using immunocompetent mouse models of BRAFwt and BRAFmut melanoma and analyzed in reference to patient data. RESULTS RGS treatment (300 mg/kg) was well tolerated in mice and resulted in ~ 50% inhibition of tumor growth as monotherapy and ~ 70% inhibition in combination with αPD1 + αCTLA4. RGS-induced tumor growth inhibition depends on CD40 upregulation in melanoma cells followed by immunogenic cell death, leading to enriched dendritic cells and activated T cells in the tumor microenvironment. The RGS-initiated tumor suppression was partially reversed by either knockdown of CD40 expression in melanoma cells or depletion of CD8+ cytotoxic T cells. Treatment with either dabrafenib and trametinib or with RGS, increased CD40+SOX10+ melanoma cells in the tumors of melanoma patients and patient-derived xenografts. High CD40 expression level correlates with beneficial T-cell responses and better survival in a TCGA dataset from melanoma patients. Expression of CD40 by melanoma cells is associated with therapeutic response to RAF/MEK inhibition and ICB. CONCLUSIONS Our data support the therapeutic use of RGS + αPD1 + αCTLA4 in RAS/RAF/PI3K pathway-activated melanomas and point to the need for clinical trials of RGS + ICB for melanoma patients who do not respond to ICB alone. TRIAL REGISTRATION NCT01205815 (Sept 17, 2010).
Collapse
Affiliation(s)
- Chi Yan
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Nabil Saleh
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jinming Yang
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Caroline A Nebhan
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anna E Vilgelm
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - E Premkumar Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph T Roland
- Departments of Surgery and Pediatrics and the Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sheau-Chiann Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rebecca L Shattuck-Brandt
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Gregory D Ayers
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ann Richmond
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA. .,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
12
|
Škubník J, Jurášek M, Ruml T, Rimpelová S. Mitotic Poisons in Research and Medicine. Molecules 2020; 25:E4632. [PMID: 33053667 PMCID: PMC7587177 DOI: 10.3390/molecules25204632] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the greatest challenges of the modern medicine. Although much effort has been made in the development of novel cancer therapeutics, it still remains one of the most common causes of human death in the world, mainly in low and middle-income countries. According to the World Health Organization (WHO), cancer treatment services are not available in more then 70% of low-income countries (90% of high-income countries have them available), and also approximately 70% of cancer deaths are reported in low-income countries. Various approaches on how to combat cancer diseases have since been described, targeting cell division being among them. The so-called mitotic poisons are one of the cornerstones in cancer therapies. The idea that cancer cells usually divide almost uncontrolled and far more rapidly than normal cells have led us to think about such compounds that would take advantage of this difference and target the division of such cells. Many groups of such compounds with different modes of action have been reported so far. In this review article, the main approaches on how to target cancer cell mitosis are described, involving microtubule inhibition, targeting aurora and polo-like kinases and kinesins inhibition. The main representatives of all groups of compounds are discussed and attention has also been paid to the presence and future of the clinical use of these compounds as well as their novel derivatives, reviewing the finished and ongoing clinical trials.
Collapse
Affiliation(s)
- Jan Škubník
- Department of Biochemistry and Microbiology, University of Chemistry and Technology in Prague, Technická 3, 166 28, Prague 6, Czech Republic; (J.Š.); (T.R.)
| | - Michal Jurášek
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology in Prague, Technická 3, 166 28, Prague 6, Czech Republic;
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology in Prague, Technická 3, 166 28, Prague 6, Czech Republic; (J.Š.); (T.R.)
| | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology in Prague, Technická 3, 166 28, Prague 6, Czech Republic; (J.Š.); (T.R.)
| |
Collapse
|
13
|
Mechanism analysis of toxicity of sodium sulfite to human hepatocytes L02. Mol Cell Biochem 2020; 473:25-37. [PMID: 32632612 DOI: 10.1007/s11010-020-03805-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/18/2020] [Indexed: 10/23/2022]
Abstract
Food additives are widely used in various food products to preserve the taste, color, and other qualities. However, if they are used improperly or exceed the standard, they will cause damage to the human body. Sulfite is a commonly used food additive to prevent oxidation from deteriorating the nutrients in foods, it has been widely used as a bleaching agent in the food industry for a long time. In this study, human hepatocytes L02 cells were used as a model cell line to evaluate the toxicity of sodium sulfite. The cell morphology and cell proliferation were affected by sodium sulfite treatment, and apoptosis was detected. Transcriptome sequencing showed 97 differentially expressed genes (DEGs) between the experimental group (IC50) and the control group (MOCK), and 27 differentially expressed genes related to cell apoptosis, metabolism and inflammation were selected for validation by qPCR. Among them, 13 significantly upregulated genes and 14 significantly downregulated genes were identified by qPCR. The results showed that with increase of sodium sulfite concentration, the morphology of L02 changed, cell proliferation and activity were inhibited, and sodium sulfite caused apoptosis in a concentration- and time-dependent manner. The resulting toxic mechanism inhibits proliferation, damages the mitochondrial integrity, and promotes apoptosis.
Collapse
|
14
|
Rahmani F, Asgharzadeh F, Avan A, Barneh F, Parizadeh MR, Ferns GA, Ryzhikov M, Ahmadian MR, Giovannetti E, Jafari M, Khazaei M, Hassanian SM. Rigosertib potently protects against colitis-associated intestinal fibrosis and inflammation by regulating PI3K/AKT and NF-κB signaling pathways. Life Sci 2020; 249:117470. [PMID: 32135184 DOI: 10.1016/j.lfs.2020.117470] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 12/26/2019] [Accepted: 02/24/2020] [Indexed: 01/02/2023]
Abstract
AIMS Rigosertib (RGS) is a PI3K inhibitor that exerts protective effects against tumor progression and cancer-related inflammation. This study was aimed to explore the regulatory effects of RGS on proliferative, pro-fibrotic and inflammatory factors in DSS- induced colitis mice model. MATERIALS AND METHODS The present study integrates systems and molecular biology approaches to investigate the therapeutic potency of RGS in an experimental model of colitis specifically examining its effects on the PI3K/AKT and NF-κB signaling pathways. KEY FINDINGS Analysis of time-resolved proteome profiling showed that PI3K-AKT inhibitors regulate expression of many proteins in all stages of inflammation, fibrogenesis and extracellular matrix remodeling. Consistent with our in-silico findings, RGS improved colitis disease activity as assessed by changes in body weight, degree of stool consistency, rectal bleeding and prolapse. RGS also reduced oxidative stress markers and colon histopathological score by decreasing inflammatory responses in colon tissues. Moreover, expression of pro-fibrotic and pro-inflammatory factors including Acta 2, Col 1a1, Col 1a2, IL-1β, TNF-α, INF-γ, and MCP-1 were suppressed in the mice treated with RGS compared to the control group. The protective effects of RGS were mediated by inactivation of PI3K/AKT and NF-kB signaling pathways. SIGNIFICANCE This study clearly demonstrates the anti-proliferative, anti-inflammatory and anti-fibrotic effects of RGS in colitis that may have implications for the treatment of colitis and colitis-associated cancer.
Collapse
Affiliation(s)
- Farzad Rahmani
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.; Iranshahr University of Medical Siences, Iranshahr, Iran
| | - Fereshteh Asgharzadeh
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farnaz Barneh
- Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Parizadeh
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.; Metabolic syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, Washington University, School of Medicine, Saint Louis, MO, USA
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, 40223, Germany
| | - Elisa Giovannetti
- Cancer Pharmacology Lab, AIRC Start-up, University Hospital of Pisa, Pisa, Italy; Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Mohieddin Jafari
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Finland
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.; Metabolic syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Design, Synthesis and Preliminary Biological Evaluation of Benzylsulfone Coumarin Derivatives as Anti-Cancer Agents. Molecules 2019; 24:molecules24224034. [PMID: 31703373 PMCID: PMC6891324 DOI: 10.3390/molecules24224034] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/02/2019] [Accepted: 11/04/2019] [Indexed: 12/19/2022] Open
Abstract
In this work, a series of benzylsulfone coumarin derivatives 5a–5o were synthesized and characterized. Kinase inhibitory activity assay indicated that most of the compounds showed considerable activity against PI3K. Anti-tumor activity studies of the active compounds were also carried out in vitro on the Hela, HepG2, H1299, HCT-116, and MCF-7 tumor cell lines by MTS assay. The structure–activity relationships (SARs) of these compounds were analyzed in detail. Compound 5h exhibited the most potent activities against the mentioned cell lines with IC50 values ranging from 18.12 to 32.60 μM, followed by 5m with IC50 values of 29.30–42.14 μM. Furthermore, 5h and 5m clearly retarded the migration of Hela cells in vitro. Next, an in silico molecular docking study was conducted to evaluate the binding models of 5h and 5m towards PI3Kα and PI3Kβ. Collectively, the above findings suggested that compounds 5h and 5m might be promising PI3K inhibitors deserving further investigation for cancer treatment.
Collapse
|
16
|
Lebelo MT, Joubert AM, Visagie MH. Warburg effect and its role in tumourigenesis. Arch Pharm Res 2019; 42:833-847. [PMID: 31473944 DOI: 10.1007/s12272-019-01185-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/27/2019] [Indexed: 12/17/2022]
Abstract
Glucose is a crucial molecule in energy production and produces different end products in non-tumourigenic- and tumourigenic tissue metabolism. Tumourigenic cells oxidise glucose by fermentation and generate lactate and adenosine triphosphate even in the presence of oxygen (Warburg effect). The Na+/H+-antiporter is upregulated in tumourigenic cells resulting in release of lactate- and H+ ions into the extracellular space. Accumulation of lactate- and proton ions in the extracellular space results in an acidic environment that promotes invasion and metastasis. Otto Warburg reported that tumourigenic cells have defective mitochondria that produce less energy. However, decades later it became evident that these mitochondria have adapted with alterations in mitochondrial content, structure, function and activity. Mitochondrial biogenesis and mitophagy regulate the formation of new mitochondria and degradation of defective mitochondria in order to combat accumulation of mutagenic mitochondrial deoxyribonucleic acid. Tumourigenic cells also produce increase reactive oxygen species (ROS) resulting from upregulated glycolysis leading to pathogenesis including cancer. Moderate ROS levels exert proliferative- and prosurvival signaling, while high ROS quantities induce cell death. Understanding the crosstalk between aberrant metabolism, redox regulation, mitochondrial adaptions and pH regulation provides scientific- and medical communities with new opportunities to explore cancer therapies.
Collapse
Affiliation(s)
- Maphuti T Lebelo
- Department of Physiology, University of Pretoria, Private Bag X323, Arcadia, Pretoria, 0007, South Africa
| | - Anna M Joubert
- Department of Physiology, University of Pretoria, Private Bag X323, Arcadia, Pretoria, 0007, South Africa
| | - Michelle H Visagie
- Department of Physiology, University of Pretoria, Private Bag X323, Arcadia, Pretoria, 0007, South Africa.
| |
Collapse
|
17
|
Balaian E, Weidner H, Wobus M, Baschant U, Jacobi A, Mies A, Bornhäuser M, Guck J, Hofbauer LC, Rauner M, Platzbecker U. Effects of rigosertib on the osteo-hematopoietic niche in myelodysplastic syndromes. Ann Hematol 2019; 98:2063-2072. [PMID: 31312928 DOI: 10.1007/s00277-019-03756-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022]
Abstract
Rigosertib is a novel multi-kinase inhibitor, which has clinical activity towards leukemic progenitor cells of patients with high-risk myelodysplastic syndromes (MDS) after failure or progression on hypomethylating agents. Since the bone marrow microenvironment plays an important role in MDS pathogenesis, we investigated the impact of rigosertib on cellular compartments within the osteo-hematopoietic niche. Healthy C57BL/6J mice treated with rigosertib for 3 weeks showed a mild suppression of hematopoiesis (hemoglobin and red blood cells, both - 16%, p < 0.01; white blood cells, - 34%, p < 0.05; platelets, - 38%, p < 0.05), whereas there was no difference in the number of hematopoietic stem cells in the bone marrow. Trabecular bone mass of the spine was reduced by rigosertib (- 16%, p = 0.05). This was accompanied by a lower trabecular number and thickness (- 6% and - 10%, respectively, p < 0.05), partly explained by the increase in osteoclast number and surface (p < 0.01). Milder effects of rigosertib on bone mass were detected in an MDS mouse model system (NHD13). However, rigosertib did not further aggravate MDS-associated cytopenia in NHD13 mice. Finally, we tested the effects of rigosertib on human mesenchymal stromal cells (MSC) in vitro and demonstrated reduced cell viability at nanomolar concentrations. Deterioration of the hematopoietic supportive capacity of MDS-MSC after rigosertib pretreatment demonstrated by decreased number of colony-forming units, especially in the monocytic lineage, further supports the idea of disturbed crosstalk within the osteo-hematopoietic niche mediated by rigosertib. Thus, rigosertib exerts inhibitory effects on the stromal components of the osteo-hematopoietic niche which may explain the dissociation between anti-leukemic activity and the absence of hematological improvement.
Collapse
Affiliation(s)
- Ekaterina Balaian
- Medical Clinic I, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Heike Weidner
- Medical Clinic III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany.,Center for Healthy Aging, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Manja Wobus
- Medical Clinic I, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Ulrike Baschant
- Medical Clinic III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany.,Center for Healthy Aging, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Angela Jacobi
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, TU Dresden, Dresden, Germany
| | - Anna Mies
- Medical Clinic I, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Martin Bornhäuser
- Medical Clinic I, University Hospital Carl Gustav Carus Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT Partner Site Dresden), DKFZ, Heidelberg, Germany
| | - Jochen Guck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, TU Dresden, Dresden, Germany
| | - Lorenz C Hofbauer
- Medical Clinic III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany.,Center for Healthy Aging, University Hospital Carl Gustav Carus Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martina Rauner
- Medical Clinic III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany.,Center for Healthy Aging, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Uwe Platzbecker
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, Leipzig University Hospital, Liebigstraße 22, 04103, Leipzig, Germany. .,German MDS Study Group (G-MDS), Leipzig, Germany. .,European Myelodysplastic Syndromes Cooperative Group (EMSCO group), .
| |
Collapse
|
18
|
Stratmann JA, Sebastian M. Polo-like kinase 1 inhibition in NSCLC: mechanism of action and emerging predictive biomarkers. LUNG CANCER-TARGETS AND THERAPY 2019; 10:67-80. [PMID: 31308774 PMCID: PMC6612950 DOI: 10.2147/lctt.s177618] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 05/24/2019] [Indexed: 12/16/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer death worldwide. Due to often unspecific disease symptoms, locally advanced or metastatic disease is diagnosed in the majority of all cases. Palliative treatment options comprise of conventional cytotoxic agents, immunotherapy with checkpoint inhibitors and the use of specific small-molecule tyrosine kinase inhibitors (TKI). However, these TKIs are mainly restricted to a small proportion of patients with lung cancer that harbor activating driver mutations. Still, the effectiveness and favorable safety profile of these compounds have prompted a systematic search for specific driver mechanisms of tumorigenesis and moreover the development of corresponding kinase inhibitors. In recent years, the Polo-like kinase (PLK) family has emerged as a key regulator in mitotic regulation. Its role in cell proliferation and the frequently observed overexpression in various tumor entities have raised much interest in basic and clinical oncology aiming to attenuate tumor growth by targeting the PLK. In this review, we give a comprehensive summary on the (pre-) clinical development of the different types of PLK inhibitors in lung cancer and summarize their mechanisms of action, safety and efficacy data and give an overview on translational research aiming to identify predictive biomarkers for a rational use of PLK inhibitors.
Collapse
Affiliation(s)
- Jan A Stratmann
- Department of Internal Medicine II, University Clinic of Frankfurt, 60596 Frankfurt, Germany
| | - Martin Sebastian
- Department of Internal Medicine II, University Clinic of Frankfurt, 60596 Frankfurt, Germany
| |
Collapse
|
19
|
Atanasova VS, Pourreyron C, Farshchian M, Lawler M, Brown CA, Watt SA, Wright S, Warkala M, Guttmann-Gruber C, Hofbauer JP, Fuentes I, Prisco M, Rashidghamat E, Has C, Salas-Alanis JC, Palisson F, Hovnanian A, McGrath JA, Mellerio JE, Bauer JW, South AP. Identification of Rigosertib for the Treatment of Recessive Dystrophic Epidermolysis Bullosa-Associated Squamous Cell Carcinoma. Clin Cancer Res 2019; 25:3384-3391. [PMID: 30846478 PMCID: PMC8185613 DOI: 10.1158/1078-0432.ccr-18-2661] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/11/2019] [Accepted: 02/21/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Squamous cell carcinoma (SCC) of the skin is the leading cause of death in patients with the severe generalized form of the genetic disease recessive dystrophic epidermolysis bullosa (RDEB). Although emerging data are identifying why patients suffer this fatal complication, therapies for treatment of RDEB SCC are in urgent need.Experimental Design: We previously identified polo-like kinase 1 (PLK1) as a therapeutic target in skin SCC, including RDEB SCC. Here, we undertake a screen of 6 compounds originally designated as PLK1 inhibitors, and detail the efficacy of the lead compound, the multipathway allosteric inhibitor ON-01910, for targeting RDEB SCC in vitro and in vivo. RESULTS ON-01910 (or rigosertib) exhibited significant specificity for RDEB SCC: in culture rigosertib induced apoptosis in 10 of 10 RDEB SCC keratinocyte populations while only slowing the growth of normal primary skin cells at doses 2 orders of magnitude higher. Furthermore, rigosertib significantly inhibited the growth of two RDEB SCC in murine xenograft studies with no apparent toxicity. Mechanistically, rigosertib has been shown to inhibit multiple signaling pathways. Comparison of PLK1 siRNA with MEK inhibition, AKT inhibition, and the microtubule-disrupting agent vinblastine in RDEB SCC shows that only PLK1 reduction exhibits a similar sensitivity profile to rigosertib. CONCLUSIONS These data support a "first in RDEB" phase II clinical trial of rigosertib to assess tumor targeting in patients with late stage, metastatic, and/or unresectable SCC.
Collapse
Affiliation(s)
- Velina S Atanasova
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Celine Pourreyron
- Division of Cellular Medicine, University of Dundee, Dundee, United Kingdom
| | - Mehdi Farshchian
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Michael Lawler
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Christian A Brown
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Stephen A Watt
- Division of Cellular Medicine, University of Dundee, Dundee, United Kingdom
| | - Sheila Wright
- Division of Cellular Medicine, University of Dundee, Dundee, United Kingdom
| | - Michael Warkala
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Christina Guttmann-Gruber
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus Medical University Salzburg, Austria
| | - Josefina Piñón Hofbauer
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus Medical University Salzburg, Austria
| | - Ignacia Fuentes
- Fundación DEBRA Chile, Santiago, Chile
- Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Marco Prisco
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Elham Rashidghamat
- St. John's Institute of Dermatology, King's College London (Guy's Campus), London, United Kingdom
| | - Cristina Has
- Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | | | - Francis Palisson
- Fundación DEBRA Chile, Santiago, Chile
- Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Alain Hovnanian
- INSERM UMR 1163, Paris, France
- Imagine Institute, Paris, France
| | - John A McGrath
- St. John's Institute of Dermatology, King's College London (Guy's Campus), London, United Kingdom
| | - Jemima E Mellerio
- St. John's Institute of Dermatology, King's College London (Guy's Campus), London, United Kingdom
| | - Johann W Bauer
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus Medical University Salzburg, Austria
| | - Andrew P South
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
20
|
Son YS, Choi K, Lee H, Kwon O, Jung KB, Cho S, Baek J, Son B, Kang SM, Kang M, Yoon J, Shen H, Lee S, Oh JH, Lee HA, Lee MO, Cho HS, Jung CR, Kim J, Cho S, Son MY. A SMN2 Splicing Modifier Rescues the Disease Phenotypes in an In Vitro Human Spinal Muscular Atrophy Model. Stem Cells Dev 2019; 28:438-453. [PMID: 30667343 DOI: 10.1089/scd.2018.0181] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Spinal muscular atrophy (SMA) is caused by the mutation or deletion of the survival motor neuron 1 (SMN1) gene. Only ∼10% of the products of SMN2, a paralogue of SMN1, are functional full-length SMN (SMN-FL) proteins, whereas SMN2 primarily produces alternatively spliced transcripts lacking exon 7. Reduced SMN protein levels in SMA patients lead to progressive degeneration of spinal motor neurons (MNs). In this study, we report an advanced platform based on an SMN2 splicing-targeting approach for SMA drug screening and validation using an SMN2 splicing reporter cell line and an in vitro human SMA model through induced pluripotent stem cell (iPSC) technology. Through drug screening using a robust cell-based luciferase assay to quantitatively measure SMN2 splicing, the small-molecule candidate compound rigosertib was identified as an SMN2 splicing modulator that led to enhanced SMN protein expression. The therapeutic potential of the candidate compound was validated in MN progenitors differentiated from SMA patient-derived iPSCs (SMA iPSC-pMNs) as an in vitro human SMA model, which recapitulated the biochemical and molecular phenotypes of SMA, including lower levels of SMN-FL transcripts and protein, enhanced cell death, and reduced neurite length. The candidate compound exerted strong splicing correction activity for SMN2 and potently alleviated the disease-related phenotypes of SMA iPSC-pMNs by modulating various cellular and molecular abnormalities. Our combined screening platform representing a pMN model of human SMA provides an efficient and reliable drug screening system and is a promising resource for drug evaluation and the exploration of drug modes of action.
Collapse
Affiliation(s)
- Ye Seul Son
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,2 Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kwangman Choi
- 3 Natural Medicine Research Center, KRIBB, Cheongju, Chungbuk, Republic of Korea
| | - Hana Lee
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,2 Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Ohman Kwon
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Kwang Bo Jung
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,2 Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Sunwha Cho
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Jiyeon Baek
- 3 Natural Medicine Research Center, KRIBB, Cheongju, Chungbuk, Republic of Korea
| | - Bora Son
- 3 Natural Medicine Research Center, KRIBB, Cheongju, Chungbuk, Republic of Korea
| | - Sung-Min Kang
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Mingu Kang
- 3 Natural Medicine Research Center, KRIBB, Cheongju, Chungbuk, Republic of Korea.,4 Department of Biomolecular Science, KRIBB School of Bioscience, UST, Daejeon, Republic of Korea
| | - Jihee Yoon
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,3 Natural Medicine Research Center, KRIBB, Cheongju, Chungbuk, Republic of Korea
| | - Haihong Shen
- 5 School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Sangku Lee
- 3 Natural Medicine Research Center, KRIBB, Cheongju, Chungbuk, Republic of Korea
| | - Jung-Hwa Oh
- 6 Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Hyang-Ae Lee
- 6 Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Mi-Ok Lee
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Hyun-Soo Cho
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,2 Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Cho-Rok Jung
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,2 Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Janghwan Kim
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,2 Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Sungchan Cho
- 3 Natural Medicine Research Center, KRIBB, Cheongju, Chungbuk, Republic of Korea.,4 Department of Biomolecular Science, KRIBB School of Bioscience, UST, Daejeon, Republic of Korea
| | - Mi-Young Son
- 1 Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,2 Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
21
|
Saei A, Palafox M, Benoukraf T, Kumari N, Jaynes PW, Iyengar PV, Muñoz-Couselo E, Nuciforo P, Cortés J, Nötzel C, Kumarakulasinghe NB, Richard JLC, Bin Adam Isa ZF, Pang B, Guzman M, Siqin Z, Yang H, Tam WL, Serra V, Eichhorn PJA. Loss of USP28-mediated BRAF degradation drives resistance to RAF cancer therapies. J Exp Med 2018; 215:1913-1928. [PMID: 29880484 PMCID: PMC6028519 DOI: 10.1084/jem.20171960] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 03/09/2018] [Accepted: 05/09/2018] [Indexed: 11/04/2022] Open
Abstract
RAF kinase inhibitors are clinically active in patients with BRAF (V600E) mutant melanoma. However, rarely do tumors regress completely, with the majority of responses being short-lived. This is partially mediated through the loss of negative feedback loops after MAPK inhibition and reactivation of upstream signaling. Here, we demonstrate that the deubiquitinating enzyme USP28 functions through a feedback loop to destabilize RAF family members. Loss of USP28 stabilizes BRAF enhancing downstream MAPK activation and promotes resistance to RAF inhibitor therapy in culture and in vivo models. Importantly, we demonstrate that USP28 is deleted in a proportion of melanoma patients and may act as a biomarker for response to BRAF inhibitor therapy in patients. Furthermore, we identify Rigosertib as a possible therapeutic strategy for USP28-depleted tumors. Our results show that loss of USP28 enhances MAPK activity through the stabilization of RAF family members and is a key factor in BRAF inhibitor resistance.
Collapse
Affiliation(s)
- Azad Saei
- Genome Institute of Singapore, A*STAR, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Marta Palafox
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Touati Benoukraf
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Nishi Kumari
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | | | | | - Paolo Nuciforo
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Javier Cortés
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Cancer Ramon y Cajal University Hospital, Madrid, Spain
- IOB Institute of Oncology, Quironsalud group, Madrid & Barcelona, Spain
| | - Christopher Nötzel
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Nesaretnam Barr Kumarakulasinghe
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore, Singapore
| | | | | | - Brendan Pang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Marta Guzman
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Zhou Siqin
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Wai Leong Tam
- Genome Institute of Singapore, A*STAR, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Violeta Serra
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Pieter Johan Adam Eichhorn
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Australia
| |
Collapse
|
22
|
Qian X, Nie X, Yao W, Klinghammer K, Sudhoff H, Kaufmann AM, Albers AE. Reactive oxygen species in cancer stem cells of head and neck squamous cancer. Semin Cancer Biol 2018; 53:248-257. [PMID: 29935313 DOI: 10.1016/j.semcancer.2018.06.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/08/2018] [Accepted: 06/17/2018] [Indexed: 12/12/2022]
Abstract
One of the greatest challenges in systemic treatment of head and neck squamous cell carcinoma (HNSCC) is a small tumor cell population, namely, cancer stem-like cells (CSC). CSC can regenerate and maintain a heterogenic tumor by their self-renewal capacity. Their potential ability to be more resistant to and survival after chemo- and radiation therapy was also identified. Further studies have shown that reactive oxygen species (ROS) contribute to this CSC-associated resistance. In this review, we focus on the current knowledge of HNSCC-CSC, with regard to ROS as a possible and novel therapeutic approach in targeting CSC.
Collapse
Affiliation(s)
- Xu Qian
- Department of Otorhinolaryngology, Head and Neck Surgery, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Berlin, Germany; Division of Molecular Diagnostics, Department of Laboratory Medicine, Zhejiang Cancer Hospital, Hangzhou, PR China; Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou Medical University, Wenzhou, PR China
| | - Xiaobo Nie
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, PR China
| | - Wenhao Yao
- Department of Otorhinolaryngology, Head and Neck Surgery, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Konrad Klinghammer
- Department of Hematology and Oncology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Holger Sudhoff
- Department of Otorhinolaryngology, Head and Neck Surgery, Klinikum Bielefeld, Bielefeld, Germany
| | - Andreas M Kaufmann
- Clinic for Gynecology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Andreas E Albers
- Department of Otorhinolaryngology, Head and Neck Surgery, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Berlin, Germany.
| |
Collapse
|
23
|
McCubrey JA, Abrams SL, Lertpiriyapong K, Cocco L, Ratti S, Martelli AM, Candido S, Libra M, Murata RM, Rosalen PL, Lombardi P, Montalto G, Cervello M, Gizak A, Rakus D, Steelman LS. Effects of berberine, curcumin, resveratrol alone and in combination with chemotherapeutic drugs and signal transduction inhibitors on cancer cells-Power of nutraceuticals. Adv Biol Regul 2018; 67:190-211. [PMID: 28988970 DOI: 10.1016/j.jbior.2017.09.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 09/29/2017] [Indexed: 06/07/2023]
Abstract
Over the past fifty years, society has become aware of the importance of a healthy diet in terms of human fitness and longevity. More recently, the concept of the beneficial effects of certain components of our diet and other compounds, that are consumed often by different cultures in various parts of the world, has become apparent. These "healthy" components of our diet are often referred to as nutraceuticals and they can prevent/suppress: aging, bacterial, fungal and viral infections, diabetes, inflammation, metabolic disorders and cardiovascular diseases and have other health-enhancing effects. Moreover, they are now often being investigated because of their anti-cancer properties/potentials. Understanding the effects of various natural products on cancer cells may enhance their usage as anti-proliferative agents which may be beneficial for many health problems. In this manuscript, we discuss and demonstrate how certain nutraceuticals may enhance other anti-cancer drugs to suppress proliferation of cancer cells.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA.
| | - Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA; Center of Comparative Medicine and Pathology, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medicine and the Hospital for Special Surgery, New York City, New York, USA
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences - Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences - Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy
| | - Ramiro M Murata
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA; Department of Foundational Sciences, School of Dental Medicine, East Carolina University, USA
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Paolo Lombardi
- Naxospharma, Via Giuseppe Di Vittorio 70, Novate Milanese 20026, Italy
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale Delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale Delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| |
Collapse
|