1
|
Huang QJ, Liao GC, Zhuang XR, Yang ML, Yao JJ, Deng JH, Zhang YM, Wang Y, Qi XX, Pan DF, Guan Y, Huang ZY, Zhang FX, Liu ZQ, Lu LL. Ras inhibitor farnesylthiosalicylic acid conjugated with IR783 dye exhibits improved tumor-targeting and altered anti-breast cancer mechanisms in mice. Acta Pharmacol Sin 2022; 43:1843-1856. [PMID: 34845369 PMCID: PMC9253319 DOI: 10.1038/s41401-021-00775-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/05/2021] [Indexed: 11/09/2022]
Abstract
Ras has long been viewed as a promising target for cancer therapy. Farnesylthiosalicylic acid (FTS), as the only Ras inhibitor has ever entered phase II clinical trials, has yielded disappointing results due to its strong hydrophobicity, poor tumor-targeting capacity, and low therapeutic efficiency. Thus, enhancing hydrophilicity and tumor-targeting capacity of FTS for improving its therapeutic efficacy is of great significance. In this study we conjugated FTS with a cancer-targeting small molecule dye IR783 and characterized the anticancer properties of the conjugate FTS-IR783. We showed that IR783 conjugation greatly improved the hydrophilicity, tumor-targeting and therapeutic potential of FTS. After a single oral administration in Balb/c mice, the relative bioavailability of FTS-IR783 was increased by 90.7% compared with FTS. We demonstrated that organic anion transporting polypeptide (OATP) and endocytosis synergistically drove the uptake of the FTS-IR783 conjugate in breast cancer MDA-MB-231 cells, resulting in superior tumor-targeting ability of the conjugate both in vitro and in vivo. We further revealed that FTS-IR783 conjugate could bind with and directly activate AMPK rather than affecting Ras, and subsequently regulate the TSC2/mTOR signaling pathway, thus achieving 2-10-fold increased anti-cancer therapeutic efficacy against 6 human breast cancer cell lines compared to FTS both in vivo and in vitro. Overall, our data highlights a promising approach for the modification of the anti-tumor drug FTS using IR783 and makes it possible to return FTS back to the clinic with a better efficacy.
Collapse
Affiliation(s)
- Qiu-ju Huang
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China ,grid.411866.c0000 0000 8848 7685School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Guo-chao Liao
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Xue-rong Zhuang
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Meng-lan Yang
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Jing-jing Yao
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Jian-hua Deng
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Yan-min Zhang
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Ying Wang
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Xiao-xiao Qi
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Dong-feng Pan
- grid.27755.320000 0000 9136 933XDepartment of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903 USA
| | - Yang Guan
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Zhi-ying Huang
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Feng-xue Zhang
- grid.411866.c0000 0000 8848 7685School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Zhong-qiu Liu
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine/ Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, SAR China
| | - Lin-lin Lu
- grid.411866.c0000 0000 8848 7685Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine/ Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, SAR China
| |
Collapse
|
2
|
Ramazani A, Karimi M, Hosseinzadeh Z, Rezayati S, Hanifehpour Y, Joo SW. Syntheses and Antitumor Properties of Furoxan Derivatives. CURR ORG CHEM 2021. [DOI: 10.2174/1385272825666210208183751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cancer is the second leading cause of death in Iran, next to heart disease. Current
therapy suffers from the major limitations of side effects and drug resistance, so the characterization
of new structures that can be power-selective and less-toxic anticancer agents is the
main challenge to medicinal chemistry research. Furoxan (1,2,5-oxadiazole-2-oxide) is a crucial
compound with many medicinal and pharmaceutical properties. The most important aspect
of furoxan is the nitric oxide (NO) molecule. One of the most essential furoxan derivatives,
which could be utilized in medicinal goals and pharmaceutical affairs, is benzofuroxan.
Furoxan could be described as a NO-donating compound in a variety of reactions, which
could also appear as hybridised with different medicinal compounds. This review article presents
a summary of syntheses and antitumor properties of furoxan derivatives as possible
chemotherapy agents for cancer. Furoxan can inhibit tumor growth in vivo without any side
effects in normal cells. Furthermore, due to NO-releasing in high levels in vivo and a wide
range of anticancer compounds, furoxan derivatives and especially its hybridised compounds could be considered as
antitumor, cytotoxic and apoptosis compounds to be applied in the human body.
Collapse
Affiliation(s)
- Ali Ramazani
- Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan 45371-38791, Iran
| | - Masoud Karimi
- Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan 45371-38791, Iran
| | - Zahra Hosseinzadeh
- Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan 45371-38791, Iran
| | - Sobhan Rezayati
- Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan 45371-38791, Iran
| | - Younes Hanifehpour
- School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Korea
| | - Sang Woo Joo
- School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Korea
| |
Collapse
|
3
|
Targeting KRAS mutant cancers by preventing signaling transduction in the MAPK pathway. Eur J Med Chem 2020; 211:113006. [PMID: 33228976 DOI: 10.1016/j.ejmech.2020.113006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 01/06/2023]
Abstract
KRAS genes are the most commonly mutated oncogenes in cancer. Unfortunately, effective therapeutic strategies for targeting KRAS mutant cancers have proven to be difficult to obtain. A key reason for this setback is due to the lack of success direct KRAS mutant inhibitors have received. Researchers have turned their efforts away from targeting the KRAS nucleotide-binding site directly and towards targeting other areas of the MAPK signaling pathway to block KRAS function. Researchers found that inhibiting enzymes and protein-protein interactions involved in the MAPK signaling pathway inhibit the activation of KRAS mutant therefore can lead to a potential therapeutic for KRAS mutated cancers. Throughout the past two decades, various indirect inhibitors have been designed and tested. EGFR and MEK inhibitors have presented with less success; however, significant advances have been made when targeting the plasma membrane localization process and the allosteric site of KRAS mutant. Farnesyltransferase and allosteric inhibitors have both advanced to human clinical trials. This comprehensive review presents the most recent developments of direct and indirect KRAS mutant inhibitors. This review summarizes published data on the inhibitory and anti-cancer activity of compounds that target KRAS activation as well as highlights the most promising strategies for targeting KRAS mutant cancers.
Collapse
|
4
|
Ye N, Xu Q, Li W, Wang P, Zhou J. Recent Advances in Developing K-Ras Plasma Membrane Localization Inhibitors. Curr Top Med Chem 2019; 19:2114-2127. [PMID: 31475899 DOI: 10.2174/1568026619666190902145116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/02/2019] [Accepted: 07/02/2019] [Indexed: 12/22/2022]
Abstract
The Ras proteins play an important role in cell growth, differentiation, proliferation and survival by regulating diverse signaling pathways. Oncogenic mutant K-Ras is the most frequently mutated class of Ras superfamily that is highly prevalent in many human cancers. Despite intensive efforts to combat various K-Ras-mutant-driven cancers, no effective K-Ras-specific inhibitors have yet been approved for clinical use to date. Since K-Ras proteins must be associated to the plasma membrane for their function, targeting K-Ras plasma membrane localization represents a logical and potentially tractable therapeutic approach. Here, we summarize the recent advances in the development of K-Ras plasma membrane localization inhibitors including natural product-based inhibitors achieved from high throughput screening, fragment-based drug design, virtual screening, and drug repurposing as well as hit-to-lead optimizations.
Collapse
Affiliation(s)
- Na Ye
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.,Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.,Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Qingfeng Xu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wanwan Li
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| |
Collapse
|
5
|
Porta EOJ, Bofill Verdaguer I, Perez C, Banchio C, Ferreira de Azevedo M, Katzin AM, Labadie GR. Repositioning Salirasib as a new antimalarial agent. MEDCHEMCOMM 2019; 10:1599-1605. [PMID: 31803400 DOI: 10.1039/c9md00298g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 06/18/2019] [Indexed: 12/16/2022]
Abstract
Malaria is a serious tropical disease that kills thousands of people every year, mainly in Africa, due to Plasmodium falciparum infections. Salirasib is a promising cancer drug candidate that interferes with the post-translational modification of Ras. This S-farnesyl thiosalicylate inhibits isoprenylcysteine carboxyl methyltransferase (ICMT), a validated target for cancer drug development. There is a high homology between the human and the parasite enzyme isoforms, in addition to being a druggable target. Looking to repurpose its structure as an antimalarial drug, a collection of S-substituted derivatives of thiosalicylic acid were prepared by introducing 1,2,3-triazole as a diversity entry point or by direct alkylation of the thiol. We further investigated the in vitro toxicity of FTS analogues to Plasmodium falciparum in the asexual stages and in Vero cells. An antiplasmodial activity assay was performed using a simple, high-sensitivity methodology based on nanoluciferase (NLuc)-transfected P. falciparum parasites. The results showed that some of the analogs were active at low micromolar concentration, including Salirasib. The most potent member of the series has S-farnesyl and the 1,2,3-triazole moiety substituted with phytyl. However, the compound substituted with methyl-naphthyl shows promising physicochemical and activity values. The low cytotoxicity in eukaryotic cells of the most active analogs provided good therapeutic indices, being starting-point candidates for future antimalarial drug development.
Collapse
Affiliation(s)
- Exequiel O J Porta
- Instituto de Química Rosario , UNR , CONICET , Suipacha 531 , S2002LRK , Rosario , Argentina . ; ; Tel: +54 341 4370477
| | - Ignasi Bofill Verdaguer
- Departamento de Parasitología , Instituto de Ciências Biomédicas , Universidade de São Paulo , São Paulo , Brazil .
| | - Consuelo Perez
- Instituto de Biología Molecular y Celular (IBR-CONICET-UNR) , Facultad de Ciencias Bioquímicas y Farmacéuticas , Universidad Nacional de Rosario , Suipacha 531 , S2002LRK , Rosario , Argentina
| | - Claudia Banchio
- Instituto de Biología Molecular y Celular (IBR-CONICET-UNR) , Facultad de Ciencias Bioquímicas y Farmacéuticas , Universidad Nacional de Rosario , Suipacha 531 , S2002LRK , Rosario , Argentina
| | | | - Alejandro M Katzin
- Departamento de Parasitología , Instituto de Ciências Biomédicas , Universidade de São Paulo , São Paulo , Brazil .
| | - Guillermo R Labadie
- Instituto de Química Rosario , UNR , CONICET , Suipacha 531 , S2002LRK , Rosario , Argentina . ; ; Tel: +54 341 4370477.,Departamento de Química Orgánica , Facultad de Ciencias Bioquímicas y Farmacéuticas , Universidad Nacional de Rosario , Suipacha 531 , S2002LRK , Rosario , Argentina
| |
Collapse
|
6
|
Misra P, Singh S. Role of cytokines in combinatorial immunotherapeutics of non-small cell lung cancer through systems perspective. Cancer Med 2019; 8:1976-1995. [PMID: 30997737 PMCID: PMC6536974 DOI: 10.1002/cam4.2112] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 02/22/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022] Open
Abstract
Lung cancer is the leading cause of deaths related to cancer and accounts for more than a million deaths per year. Various new strategies have been developed and adapted for treatment; still the survival for 5 years is just 16% in patients with non‐small cell lung cancer (NSCLC). Most of these strategies to combat NSCLC whether it is a drug molecule or immunotherapy/vaccine candidate require a big cost and time. Integration of computational modeling with systems biology has opened new avenues for understanding complex cancer biology. Resolving the complex interactions of various pathways and their crosstalk leading to oncogenic changes could identify new therapeutic targets with lesser cost and time. Herein, this review provides an overview of various aspects of NSCLC along with available strategies for its cure concluding with our insight into how systems approach could serve as a therapeutic intervention dissecting the immunologic parameters and cross talk between various pathways involved.
Collapse
Affiliation(s)
- Pragya Misra
- National Centre for Cell ScienceSP Pune University CampusPuneIndia
| | - Shailza Singh
- National Centre for Cell ScienceSP Pune University CampusPuneIndia
| |
Collapse
|
7
|
Furuse J, Kurata T, Okano N, Fujisaka Y, Naruge D, Shimizu T, Kitamura H, Iwasa T, Nagashima F, Nakagawa K. An early clinical trial of Salirasib, an oral RAS inhibitor, in Japanese patients with relapsed/refractory solid tumors. Cancer Chemother Pharmacol 2018; 82:511-519. [PMID: 29992354 PMCID: PMC6105164 DOI: 10.1007/s00280-018-3618-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/04/2018] [Indexed: 12/23/2022]
Abstract
Purpose Patients with RAS-positive tumors respond poorly to chemotherapies and have a few treatment options. Salirasib is an oral RAS inhibitor that competitively blocks the membrane association of RAS proteins. The aim of this phase I multiple-ascending-dose clinical trial was to investigate the safety and pharmacokinetics of Salirasib in Japanese patients with relapsed/refractory solid tumors and to explore its efficacy. Methods Salirasib was started at a dose of 100-mg twice-daily and escalated to a maximum of 1000-mg twice-daily from days 1 to 21 of a 28-day regimen. The pharmacokinetics was evaluated on days 1 and 21. Dose-limiting toxicity (DLT) and adverse events (AEs) were monitored throughout the trial. Patients with stable disease or better repeated the dosing regimen. Results A total of 21 patients received Salirasib. Among 14 patients tested, 4 had KRAS mutations. Cmax and AUCinf were maximal at 800 mg. No maximum tolerable dose was discerned, as no DLT was observed in any dosing group. The most frequently observed AEs were gastrointestinal disturbances, including diarrhea, abdominal pain, and nausea. No AEs led to discontinuation. All patients completed the first regimen and 11 patients repeated the regimen (median: 2 cycles; range: 1–13). Patients with KRAS mutations showed median progression-free survival of 227 days (range: 79–373). Conclusion Salirasib was safe and well tolerated in Japanese patients, and 800-mg twice-daily is recommended for phase II trials. Although the number of participants with KRAS mutations was limited, the remarkably long progression-free period warrants further investigation. Clinical trial registration JAPIC Clinical Trials Information; JapicCTI-121751. Electronic supplementary material The online version of this article (10.1007/s00280-018-3618-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Junji Furuse
- Department of Medical Oncology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan.
| | - Takayasu Kurata
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Yasuhito Fujisaka
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Daisuke Naruge
- Department of Medical Oncology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Toshio Shimizu
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Hiroshi Kitamura
- Department of Medical Oncology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Tsutomu Iwasa
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Fumio Nagashima
- Department of Medical Oncology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| |
Collapse
|
8
|
Sekerdag E, Lüle S, Bozdağ Pehlivan S, Öztürk N, Kara A, Kaffashi A, Vural I, Işıkay I, Yavuz B, Oguz KK, Söylemezoğlu F, Gürsoy-Özdemir Y, Mut M. A potential non-invasive glioblastoma treatment: Nose-to-brain delivery of farnesylthiosalicylic acid incorporated hybrid nanoparticles. J Control Release 2017; 261:187-198. [PMID: 28684169 DOI: 10.1016/j.jconrel.2017.06.032] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/29/2017] [Accepted: 06/28/2017] [Indexed: 11/26/2022]
Abstract
New drug delivery systems are highly needed in research and clinical area to effectively treat gliomas by reaching a high antineoplastic drug concentration at the target site without damaging healthy tissues. Intranasal (IN) administration, an alternative route for non-invasive drug delivery to the brain, bypasses the blood-brain-barrier (BBB) and eliminates systemic side effects. This study evaluated the antitumor efficacy of farnesylthiosalicylic acid (FTA) loaded (lipid-cationic) lipid-PEG-PLGA hybrid nanoparticles (HNPs) after IN application in rats. FTA loaded HNPs were prepared, characterized and evaluated for cytotoxicity. Rat glioma 2 (RG2) cells were implanted unilaterally into the right striatum of female Wistar rats. 10days later, glioma bearing rats received either no treatment, or 5 repeated doses of 500μM freshly prepared FTA loaded HNPs via IN or intravenous (IV) application. Pre-treatment and post-treatment tumor sizes were determined with MRI. After a treatment period of 5days, IN applied FTA loaded HNPs achieved a significant decrease of 55.7% in tumor area, equal to IV applied FTA loaded HNPs. Herewith, we showed the potential utility of IN application of FTA loaded HNPs as a non-invasive approach in glioblastoma treatment.
Collapse
Affiliation(s)
- Emine Sekerdag
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey; Neuroscience Research Lab, Research Center for Translational Medicine, Koҫ University, Istanbul, Turkey.
| | - Sevda Lüle
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey; Neuroscience Center and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Sibel Bozdağ Pehlivan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Naile Öztürk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Aslı Kara
- Department of Nanotechnology and Nanomedicine, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey; Department of Biology, Faculty of Art and Science, Hitit University, Çorum, Turkey
| | - Abbas Kaffashi
- Department of Nanotechnology and Nanomedicine, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Imran Vural
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Ilkay Işıkay
- Department of Neurosurgery, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Burҫin Yavuz
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Kader Karlı Oguz
- Department of Radiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Figen Söylemezoğlu
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Yasemin Gürsoy-Özdemir
- Neuroscience Research Lab, Research Center for Translational Medicine, Koҫ University, Istanbul, Turkey; Department of Neurology, School of Medicine, Koҫ University, Istanbul, Turkey
| | - Melike Mut
- Department of Neurosurgery, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
9
|
Targeting KRAS mutated non-small cell lung cancer: A history of failures and a future of hope for a diverse entity. Crit Rev Oncol Hematol 2017; 110:1-12. [DOI: 10.1016/j.critrevonc.2016.12.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/10/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023] Open
|
10
|
Yang S, Liu G. Targeting the Ras/Raf/MEK/ERK pathway in hepatocellular carcinoma. Oncol Lett 2017; 13:1041-1047. [PMID: 28454211 DOI: 10.3892/ol.2017.5557] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 09/28/2016] [Indexed: 12/12/2022] Open
Abstract
Although the biological basis of hepatocellular carcinoma (HCC) remains unclear, effective treatments and improvement of the survival rate remain worthwhile research goals. Abnormal protein signaling pathways contributing to uncontrolled cell proliferation, differentiation, survival and apoptosis are biomarkers of the carcinogenic process. Certain mutated components or overexpression of the rat sarcoma virus (Ras)/rapidly accelerated fibrosarcoma (Raf)/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling pathway are increasingly being studied in HCC carcinogenesis. The present review addresses the effect of the Ras/Raf/MEK/ERK signaling pathway on the pathogenesis of HCC, and provides an update on the preclinical and clinical development of various inhibitors targeting this core signaling pathway, which include various Ras inhibitors, Raf inhibitors and MEK inhibitors for HCC.
Collapse
Affiliation(s)
- Sufang Yang
- Department of Pharmacy, The First Affiliated Hospital of Shantou University Medical Collage, Shantou, Guangdong 515041, P.R. China
| | - Guohua Liu
- Department of Pharmacy, The First Affiliated Hospital of Shantou University Medical Collage, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
11
|
Gupta A, Misra A, Deretic V. Targeted pulmonary delivery of inducers of host macrophage autophagy as a potential host-directed chemotherapy of tuberculosis. Adv Drug Deliv Rev 2016; 102:10-20. [PMID: 26829287 DOI: 10.1016/j.addr.2016.01.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/26/2015] [Accepted: 01/21/2016] [Indexed: 12/19/2022]
Abstract
One of the promising host-directed chemotherapeutic interventions in tuberculosis (TB) is based on inducing autophagy as an immune effector. Here we consider the strengths and weaknesses of potential autophagy-based pharmacological intervention. Using the existing drugs that induce autophagy is an option, but it has limitations given the broad role of autophagy in most cells, tissues, and organs. Thus, it may be desirable that the agent being used to modulate autophagy is applied in a targeted manner, e.g. delivered to affected tissues, with infected macrophages being an obvious choice. This review addresses the advantages and disadvantages of delivering drugs to induce autophagy in M. tuberculosis-infected macrophages. One option, already being tested in models, is to design particles for inhalation delivery to lung macrophages. The choice of drugs, drug release kinetics and intracellular residence times, non-target cell exposure and feasibility of use by patients is discussed. We term here this (still experimental) approach, of compartment-targeting, autophagy-based, host-directed therapy as "Track-II antituberculosis chemotherapy."
Collapse
|
12
|
Bhattacharya S, Socinski MA, Burns TF. KRAS mutant lung cancer: progress thus far on an elusive therapeutic target. Clin Transl Med 2015; 4:35. [PMID: 26668062 PMCID: PMC4678136 DOI: 10.1186/s40169-015-0075-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/25/2015] [Indexed: 01/22/2023] Open
Abstract
The KRAS mutation remains the most common driver mutation in patients with non-small cell lung cancer (NSCLC) and confers a poor prognosis. Thus far, efforts to target this mutation over the last two decades have been unsuccessful. Over the past 5 years, many efforts to develop drugs that target the RAS-RAF-MEK-ERK (MAPK) pathway have resulted in enhanced understanding of the KRAS mutant NSCLC and have provided optimism that this disease can be targeted.
Collapse
Affiliation(s)
- Saveri Bhattacharya
- University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Room 461, Pittsburgh, PA, 15232, USA.
| | - Mark A Socinski
- Medicine and Cardiothoracic Surgery, University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Room 556, Pittsburgh, PA, 15232, USA.
| | - Timothy F Burns
- University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Office: Suite 2.18e, Pittsburgh, PA, 15232, USA.
| |
Collapse
|
13
|
Ling Y, Wang X, Wang C, Xu C, Zhang W, Zhang Y, Zhang Y. Hybrids from Farnesylthiosalicylic Acid and Hydroxamic Acid as Dual Ras-Related Signaling and Histone Deacetylase (HDAC) Inhibitors: Design, Synthesis and Biological Evaluation. ChemMedChem 2015; 10:971-6. [PMID: 25882299 DOI: 10.1002/cmdc.201500019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/18/2015] [Indexed: 01/05/2023]
Abstract
A novel series of hybrids was designed and synthesized by combining key elements from farnesylthiosalicylic acid (FTS) and hydroxamic acid. Several 3,7,11-trimethyldodeca-2,6,10-trien-1-yl) thio)benzamide derivatives, particularly those with branched and linear aliphatic linkers between the hydroxamic zinc binding group (ZBG) and the benzamide core, not only displayed significant antitumor activities against six human cancer cells but also exhibited histone deacetylase (HDAC) inhibitory effects in vitro. Among them, N-(4-(hydroxyamino)-4-oxobutyl)-2-(((2E,6E)-3,7,11-trimethyldodeca-2,6, 10-trien-1-yl)thio)benzamide (8 d) was the most potent, with IC50 values of 4.9-7.6 μM; these activities are eight- to sixteen-fold more potent than FTS and comparable to that of suberoylanilide hydroxamic acid (SAHA). Derivative 8 d induced cell cycle arrest in the G0/G1 phase, inhibited the acetylation of histone H3 and α-tubulin, and blocked Ras-related signaling pathways in a dose-dependent manner. The improved tumor growth inhibition and cell-cycle arrest in vitro might result from the dual inhibition. These findings suggest dual inhibitors of Ras-related signaling pathway and HDAC hold promise as therapeutic agents for the treatment of cancer.
Collapse
Affiliation(s)
- Yong Ling
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China). .,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009 (P. R. China).
| | - Xuemin Wang
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China)
| | - Chenniu Wang
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China)
| | - Chenjun Xu
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China)
| | - Wei Zhang
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China)
| | - Yihua Zhang
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China).,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009 (P. R. China)
| | - Yanan Zhang
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China).
| |
Collapse
|
14
|
Badar T, Cortes JE, Ravandi F, O'Brien S, Verstovsek S, Garcia-Manero G, Kantarjian H, Borthakur G. Phase I study of S-trans, trans-farnesylthiosalicylic acid (salirasib), a novel oral RAS inhibitor in patients with refractory hematologic malignancies. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2015; 15:433-438.e2. [PMID: 25795639 DOI: 10.1016/j.clml.2015.02.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/09/2015] [Accepted: 02/12/2015] [Indexed: 01/13/2023]
Abstract
BACKGROUND Rat sarcoma (RAS)/rapidly accelerated fibrosarcoma (RAF)/mitogen-activated protein kinase activation (mutational or nonmutational) is a key pathway for survival and proliferative advantage of leukemic cells. Salirasib (Concordia Pharmaceuticals) is an oral RAS inhibitor that causes dislocation of RAS by competing directly with farnesylated RAS in binding to its putative membrane-binding proteins. Salirasib does not inhibit farnesyl transferase enzyme. PATIENTS AND METHODS We report on a phase I study of Salirasib in patients with relapsed/refractory hematologic malignancies. Salirasib was administered orally twice daily on days 1 to 21 of a 28-day cycle in a "3+3" dose escalation design. RESULTS Seventeen patients with relapsed/refractory leukemia were treated for a median of 4 cycles (range, 1-29). Three patients each were enrolled at a dose level of 100, 200, 400, 600, and 800 mg twice daily and 2 patients at a dose level of 900 mg twice daily. No dose-limiting toxicities were encountered. Grade 1/2 diarrhea was the only frequent nonhematologic toxicity observed in 14 of 17 (82%) patients and was resolved with oral antidiarrheal agents. Eight (47%) patients (4 with myelodysplastic syndrome, 2 with acute myeloid leukemia, 1 with chronic myelomonocytic leukemia, and 1 with chronic myeloid leukemia) had hematological improvement; 1 in 3 lineages, 1 in 2 lineages, and 6 in 1 lineage. None of the patients achieved complete remission. The responses lasted for a median of 10 weeks (range, 5-115). The study was discontinued because of financial constraints. CONCLUSION Salirasib was well tolerated and showed modest activity in relapsed/refractory hematological malignancies. The safety profile of Salirasib and its hematological malignancy relevant target makes it a potential drug to be used in combination therapy.
Collapse
Affiliation(s)
- Talha Badar
- Department of Leukemia, M.D. Anderson Cancer Center, Houston, TX
| | - Jorge E Cortes
- Department of Leukemia, M.D. Anderson Cancer Center, Houston, TX
| | - Farhad Ravandi
- Department of Leukemia, M.D. Anderson Cancer Center, Houston, TX
| | - Susan O'Brien
- Department of Leukemia, M.D. Anderson Cancer Center, Houston, TX
| | - Srdan Verstovsek
- Department of Leukemia, M.D. Anderson Cancer Center, Houston, TX
| | | | - Hagop Kantarjian
- Department of Leukemia, M.D. Anderson Cancer Center, Houston, TX
| | - Gautam Borthakur
- Department of Leukemia, M.D. Anderson Cancer Center, Houston, TX.
| |
Collapse
|
15
|
Wang ZQ, Chang RA, Huang HY, Wang XM, Wang XY, Chen L, Ling Y. Synthesis and biological evaluation of novel farnesylthiosalicylic acid/salicylic acid hybrids as potential anti-tumor agents. CHINESE CHEM LETT 2014. [DOI: 10.1016/j.cclet.2014.06.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
16
|
The dual RAF/MEK inhibitor CH5126766/RO5126766 may be a potential therapy for RAS-mutated tumor cells. PLoS One 2014; 9:e113217. [PMID: 25422890 PMCID: PMC4244135 DOI: 10.1371/journal.pone.0113217] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 10/20/2014] [Indexed: 01/07/2023] Open
Abstract
Although melanoma is the most aggressive skin cancer, recent advances in BRAF and/or MEK inhibitors against BRAF-mutated melanoma have improved survival rates. Despite these advances, a treatment strategy targeting NRAS-mutated melanoma has not yet been elucidated. We discovered CH5126766/RO5126766 as a potent and selective dual RAF/MEK inhibitor currently under early clinical trials. We examined the activity of CH5126766/RO5126766 in a panel of malignant tumor cell lines including melanoma with a BRAF or NRAS mutation. Eight cell lines including melanoma were assessed for their sensitivity to the BRAF, MEK, or RAF/MEK inhibitor using in vitro growth assays. CH5126766/RO5126766 induced G1 cell cycle arrest in two melanoma cell lines with the BRAF V600E or NRAS mutation. In these cells, the G1 cell cycle arrest was accompanied by up-regulation of the cyclin-dependent kinase inhibitor p27 and down-regulation of cyclinD1. CH5126766/RO5126766 was more effective at reducing colony formation than a MEK inhibitor in NRAS- or KRAS-mutated cells. In the RAS-mutated cells, CH5126766/RO5126766 suppressed the MEK reactivation caused by a MEK inhibitor. In addition, CH5126766/RO5126766 suppressed the tumor growth in SK-MEL-2 xenograft model. The present study indicates that CH5126766/RO5126766 is an attractive RAF/MEK inhibitor in RAS-mutated malignant tumor cells including melanoma.
Collapse
|
17
|
Ling Y, Wang Z, Wang X, Li X, Wang X, Zhang W, Dai H, Chen L, Zhang Y. Hybrid molecule from Farnesylthiosalicylic acid-diamine and phenylpropenoic acid as Ras-related signaling inhibitor with potent antitumor activities. Chem Biol Drug Des 2014; 85:145-52. [PMID: 25043275 DOI: 10.1111/cbdd.12393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/13/2014] [Accepted: 06/25/2014] [Indexed: 12/30/2022]
Abstract
Novel series of Farnesylthiosalicylic acid-diamine/phenylpropenoic acid hybrids were designed and synthesized. Their in vitro growth inhibitory assays showed that most compounds displayed strong antiproliferation activity against seven cancer cells. Especially, the new hybrid 12 f, by the conjugation of 10a with ferulic acid, could selectively suppress the proliferation of tumor cells and display significantly lower toxicities to normal cells than its intermediate 10a. Furthermore, 12 f dose-dependently induced SMMC-7721 cell apoptosis. Additionally, our observations demonstrated that 12 f inhibited both Ras-related signaling and phosphorylated NF-κB synergistically, which may be advantageous to the strong antitumor activities of 12 f. Our findings suggest that these novel hybrids may hold a great promise as therapeutic agents for the intervention of human cancers.
Collapse
Affiliation(s)
- Yong Ling
- School of Pharmacy, Nantong University, Nantong, 226001, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Ling Y, Wang X, Zhu H, Wang Z, Xu C, Wang X, Chen L, Zhang W. Synthesis and biological evaluation of novel farnesylthiosalicylic acid derivatives for cancer treatment. Arch Pharm (Weinheim) 2014; 347:327-33. [PMID: 24435839 DOI: 10.1002/ardp.201300325] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 11/22/2013] [Accepted: 11/22/2013] [Indexed: 12/21/2022]
Abstract
Novel farnesylthiosalicylic acid (FTS) derivatives were synthesized by coupling with different substituted diamines. Their in vitro growth inhibitory activities against seven human cancer cell lines were evaluated. The results revealed that the synthetic farnesylthiosalicylamides displayed significant antitumor activities compared to the positive control FTS. Especially, compound 8f exhibited the strongest antitumor activities with IC50 values of 6.20-7.83 µM, which were one- to threefold less than those of sorafenib and six- to tenfold less than that of FTS against each cell line in vitro. Furthermore, 8f could inhibit the Ras-related signaling pathway and induce SMMC-7721 cell apoptosis superior to FTS in a dose-dependent manner. These data indicate that 8f may hold greater promise as therapeutic agent for the intervention of human cancers.
Collapse
Affiliation(s)
- Yong Ling
- School of Pharmacy, Nantong University, Nantong, P. R. China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Ling Y, Wang Z, Zhu H, Wang X, Zhang W, Wang X, Chen L, Huang Z, Zhang Y. Synthesis and biological evaluation of farnesylthiosalicylamides as potential anti-tumor agents. Bioorg Med Chem 2014; 22:374-80. [DOI: 10.1016/j.bmc.2013.11.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 10/17/2013] [Accepted: 11/08/2013] [Indexed: 10/26/2022]
|
20
|
Wang D, Zhu Q, Liu B, Chen G, Ling Y. Identification of Gut Microbial Metabolites of a Synthetic Nitric Oxide-Releasing Farnesylthiosalicylic Acid Derivative, an Antitumor Agent. ANAL LETT 2013. [DOI: 10.1080/00032719.2013.834443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
21
|
Abstract
INTRODUCTION The Ras proteins (K-Ras, N-Ras, and H-Ras) are GTPases that function as molecular switches for a variety of critical cellular activities and their function is tightly and temporally regulated in normal cells. Oncogenic mutations in the RAS genes, which create constitutively-active Ras proteins, can result in uncontrolled proliferation or survival in tumor cells. AREAS COVERED The paper discusses three therapeutic approaches targeting the Ras pathway in cancer: i) Ras itself, ii) Ras downstream pathways, and iii) synthetic lethality. The most adopted approach is targeting Ras downstream signaling, and specifically the PI3K-AKT-mTOR and Raf-MEK pathways, as they are frequently major oncogenic drivers in cancers with high Ras signaling. Although direct targeting of Ras has not been successful clinically, newer approaches being investigated in preclinical studies, such as RNA interference-based and synthetic lethal approaches, promise great potential for clinical application. EXPERT OPINION The challenges of current and emerging therapeutics include the lack of "tumor specificity" and their limitation to those cancers which are "dependent" on aberrant Ras signaling for survival. While the newer approaches have the potential to overcome these limitations, they also highlight the importance of robust preclinical studies and bidirectional translational research for successful clinical development of Ras-related targeted therapies.
Collapse
Affiliation(s)
- Asami Takashima
- Boston University School of Medicine, Cancer Research Center , 72 E. Concord St. Boston MA, 02118 , USA
| | | |
Collapse
|
22
|
Salirasib sensitizes hepatocarcinoma cells to TRAIL-induced apoptosis through DR5 and survivin-dependent mechanisms. Cell Death Dis 2013; 4:e471. [PMID: 23348585 PMCID: PMC3563988 DOI: 10.1038/cddis.2012.200] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Ras activation is a frequent event in human hepatocarcinoma that may contribute to resistance towards apoptosis. Salirasib is a ras and mTOR inhibitor that induces a pro-apoptotic phenotype in human hepatocarcinoma cell lines. In this work, we evaluate whether salirasib sensitizes those cells to TRAIL-induced apoptosis. Cell viability, cell death and apoptosis were evaluated in vitro in HepG2, Hep3B and Huh7 cells treated with DMSO, salirasib and YM155 (a survivin inhibitor), alone or in combination with recombinant TRAIL. Our results show that pretreatment with salirasib sensitized human hepatocarcinoma cell lines, but not normal human hepatocytes, to TRAIL-induced apoptosis. Indeed, FACS analysis showed that 25 (Huh7) to 50 (HepG2 and Hep3B) percent of the cells treated with both drugs were apoptotic. This occurred through activation of the extrinsic and the intrinsic pathways, as evidenced by a marked increase in caspase 3/7 (five to ninefold), caspase 8 (four to sevenfold) and caspase 9 (eight to 12-fold) activities in cells treated with salirasib and TRAIL compared with control. Survivin inhibition had an important role in this process and was sufficient to sensitize hepatocarcinoma cells to apoptosis. Furthermore, TRAIL-induced apoptosis in HCC cells pretreated with salirasib was dependent on activation of death receptor (DR) 5. In conclusion, salirasib sensitizes hepatocarcinoma cells to TRAIL-induced apoptosis by a mechanism involving the DR5 receptor and survivin inhibition. These results in human hepatocarcinoma cell lines and primary hepatocytes provide a rationale for testing the combination of salirasib and TRAIL agonists in human hepatocarcinoma.
Collapse
|
23
|
Sacco E, Spinelli M, Vanoni M. Approaches to Ras signaling modulation and treatment of Ras-dependent disorders: a patent review (2007--present). Expert Opin Ther Pat 2012; 22:1263-87. [PMID: 23009088 DOI: 10.1517/13543776.2012.728586] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Ras proteins are small GTPases molecular switches that cycle through two alternative conformational states, a GDP-bound inactive state and a GTP-bound active state. In the active state, Ras proteins interact with and modulate the activity of several downstream effectors regulating key cellular processes including proliferation, differentiation, survival, senescence, migration and metabolism. Activating mutations of RAS genes and of genes encoding Ras signaling members have a great incidence in proliferative disorders, such as cancer, immune and inflammatory diseases and developmental syndromes. Therefore, Ras and Ras signaling represent important clinical targets for the design and development of pharmaceutically active agents, including anticancer agents. AREAS COVERED The authors summarize methods available to down-regulate the Ras pathway and review recent patents covering Ras signaling modulators, as well as methods designed to kill specifically cancer cells bearing activated RAS oncogene. EXPERT OPINION Targeted therapy approach based on direct targeting of molecules specifically altered in Ras-dependent diseases is pursued with molecules that down-regulate expression or inhibit the biological function of mutant Ras or Ras signaling members. The low success rate in a clinical setting of molecules targeting activated members of the Ras pathway may require development of novel approaches, including combined and synthetic lethal therapies.
Collapse
Affiliation(s)
- Elena Sacco
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Milano, Italy
| | | | | |
Collapse
|
24
|
Laheru D, Shah P, Rajeshkumar NV, McAllister F, Taylor G, Goldsweig H, Le DT, Donehower R, Jimeno A, Linden S, Zhao M, Song D, Rudek MA, Hidalgo M. Integrated preclinical and clinical development of S-trans, trans-Farnesylthiosalicylic Acid (FTS, Salirasib) in pancreatic cancer. Invest New Drugs 2012; 30:2391-9. [PMID: 22547163 DOI: 10.1007/s10637-012-9818-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 04/03/2012] [Indexed: 12/14/2022]
Abstract
PURPOSE S-trans,trans-Farnesylthiosalicylic Acid (FTS, salirasib) inhibits Ras-dependent cell growth by dislodging all isoforms of Ras, including mutant Ras, from the plasma membrane. This study evaluated the activity, safety, and toxicity of salirasib in preclinical models and patients with metastatic pancreatic adenocarcinoma (PDA). PATIENTS AND METHODS In the preclinical study, salirasib was tested, alone and in combination with gemcitabine, in patient derived xenografts (PDX) of PDA. In the clinical study, treatment-naïve patients with advanced, metastatic PDA were treated with a standard dose schedule of gemcitabine and salirasib 200-800 mg orally (PO) twice daily (bid) for 21 days every 28 days. Tissue from preclinical models and patients' biopsies were collected pre-treatment and on Cycle (C) 1, Day (D) 9 to characterize the effect of gemcitabine and salirasib on activated Ras protein levels. Plasma samples for pharmacokinetics were collected for salirasib administered alone and in combination. RESULTS Salirasib inhibited the growth of 2/14 PDX models of PDA and modulated Ras signaling in these tumors. Nineteen patients were enrolled. No DLTs occurred. Common adverse events included hematologic and gastrointestinal toxicities and fatigue. The median overall survival was 6.2 months and the 1 year survival 37 %. In 2 patients in whom paired tissue biopsies were available, Ras and KRas protein levels were decreased on C1D9. Salirasib exposure was not altered by gemcitabine and did not correlate with PD outcomes. CONCLUSION The combination of gemcitabine and salirasib appears well-tolerated, with no alteration of salirasib exposure, and exerted clinical and PD activity in PDA.
Collapse
Affiliation(s)
- Daniel Laheru
- Department of Medical Oncology, Skip Viragh Center for Pancreatic Cancer Research and Patient Care, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Bunting- Blaustein Cancer Research Building, Room 4M09, Baltimore, MD 21231, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bergman JA, Hahne K, Song J, Hrycyna CA, Gibbs RA. S-Farnesyl-Thiopropionic Acid (FTPA) Triazoles as Potent Inhibitors of Isoprenylcysteine Carboxyl Methyltransferase. ACS Med Chem Lett 2012; 3:15-19. [PMID: 22754607 DOI: 10.1021/ml200106d] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
We report the design and synthesis of novel FTPA-triazole compounds as potent inhibitors of isoprenylcysteine carboxyl methyltransferase (Icmt), through a focus on thioether and isoprenoid mimetics. These mimetics were coupled utilizing a copper-assisted cycloaddition to assemble the potential inhibitors. Using the resulting triazole from the coupling as an isoprenyl mimetic resulted in the biphenyl substituted FTPA triazole 10n. This lipid-modified analog is a potent inhibitor of Icmt (IC(50) = 0.8 ± 0.1 μM; calculated K(i) = 0.4 μM).
Collapse
Affiliation(s)
- Joel A. Bergman
- Department
of Medicinal Chemistry and Molecular Pharmacology and the Center for
Cancer Research, and ‡Department of Chemistry and the Center for Cancer Research, Purdue University, West Lafayette, Indiana
47907, United States
| | - Kalub Hahne
- Department
of Medicinal Chemistry and Molecular Pharmacology and the Center for
Cancer Research, and ‡Department of Chemistry and the Center for Cancer Research, Purdue University, West Lafayette, Indiana
47907, United States
| | - Jiao Song
- Department
of Medicinal Chemistry and Molecular Pharmacology and the Center for
Cancer Research, and ‡Department of Chemistry and the Center for Cancer Research, Purdue University, West Lafayette, Indiana
47907, United States
| | - Christine A. Hrycyna
- Department
of Medicinal Chemistry and Molecular Pharmacology and the Center for
Cancer Research, and ‡Department of Chemistry and the Center for Cancer Research, Purdue University, West Lafayette, Indiana
47907, United States
| | - Richard A. Gibbs
- Department
of Medicinal Chemistry and Molecular Pharmacology and the Center for
Cancer Research, and ‡Department of Chemistry and the Center for Cancer Research, Purdue University, West Lafayette, Indiana
47907, United States
| |
Collapse
|
26
|
Peptide Scaffolds: Flexible Molecular Structures With Diverse Therapeutic Potentials. Int J Pept Res Ther 2012. [DOI: 10.1007/s10989-011-9286-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
27
|
Subbiah V, Slopis J, Hong DS, Ketonen LM, Hamilton J, McCutcheon IE, Kurzrock R. Treatment of patients with advanced neurofibromatosis type 2 with novel molecularly targeted therapies: from bench to bedside. J Clin Oncol 2011; 30:e64-8. [PMID: 22203760 DOI: 10.1200/jco.2011.38.2614] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
INTRODUCTION KRAS mutations are present in 30% of lung adenocarcinomas. Salirasib prevents Ras membrane binding thereby blocking the function of all Ras isoforms. This phase II study determined the activity of salirasib in patients with advanced lung adenocarcinomas with KRAS mutations. METHODS Two cohorts of patients with stage IIIB/IV lung adenocarcinoma were eligible: patients with tumors with KRAS mutations who were previously treated with chemotherapy and patients receiving initial therapy who had ≥15 pack-year smoking history. Salirasib was given orally from days 1 to 28 of a 35-day cycle. The primary end point was the rate of nonprogression at 10 weeks. RESULTS Thirty-three patients were enrolled. Thirty patients had KRAS mutations (23 patients who were previously treated and 7/10 patients who had no prior therapy). Of the previously treated patients, 7 of 23 (30%) had stable disease at 10 weeks, and 4 of 10 (40%) previously untreated patients had stable disease at 10 weeks. No patient had a radiographic partial response (0% observed rate, 95% confidence interval 0-12%). The median overall survival was not reached (>9 months) for previously untreated patients and it was 15 months for patients who received prior chemotherapy. Diarrhea, nausea, and fatigue were the most common toxicities. CONCLUSIONS Salirasib at the current dose and schedule has insufficient activity in the treatment of KRAS mutant lung adenocarcinoma to warrant further evaluation. The successful enrollment of 30 patients with tumors with KRAS mutant lung adenocarcinoma over 15 months at a single site demonstrates that drug trials directed at a KRAS-specific genotype in lung cancer are feasible.
Collapse
|
29
|
Amide-modified prenylcysteine based Icmt inhibitors: Structure-activity relationships, kinetic analysis and cellular characterization. Bioorg Med Chem 2011; 20:283-95. [PMID: 22142613 DOI: 10.1016/j.bmc.2011.10.087] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 10/27/2011] [Accepted: 10/30/2011] [Indexed: 11/23/2022]
Abstract
Human protein isoprenylcysteine carboxyl methyltransferase (hIcmt) is the enzyme responsible for the α-carboxyl methylation of the C-terminal isoprenylated cysteine of CaaX proteins, including Ras proteins. This specific posttranslational methylation event has been shown to be important for cellular transformation by oncogenic Ras isoforms. This finding led to interest in hIcmt inhibitors as potential anti-cancer agents. Previous analog studies based on N-acetyl-S-farnesylcysteine identified two prenylcysteine-based low micromolar inhibitors (1a and 1b) of hIcmt, each bearing a phenoxyphenyl amide modification. In this study, a focused library of analogs of 1a and 1b was synthesized and screened versus hIcmt, delineating structural features important for inhibition. Kinetic characterization of the most potent analogs 1a and 1b established that both inhibitors exhibited mixed-mode inhibition and that the competitive component predominated. Using the Cheng-Prusoff method, the K(i) values were determined from the IC(50) values. Analog 1a has a K(IC) of 1.4±0.2μM and a K(IU) of 4.8±0.5μM while 1b has a K(IC) of 0.5±0.07μM and a K(IU) of 1.9±0.2μM. Cellular evaluation of 1b revealed that it alters the subcellular localization of GFP-KRas, and also inhibits both Ras activation and Erk phosphorylation in Jurkat cells.
Collapse
|
30
|
Bustinza-Linares E, Kurzrock R, Tsimberidou AM. Salirasib in the treatment of pancreatic cancer. Future Oncol 2010; 6:885-91. [PMID: 20528225 DOI: 10.2217/fon.10.71] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The Ras family of genes is involved in the cellular regulation of proliferation, differentiation, cell adhesion and apoptosis. The K-ras gene is mutated in over 90% of pancreatic cancer cases. Salirasib (S-trans,trans-farnesylthiosalycilic acid [FTS]) is a synthetic small molecule that acts as a potent Ras inhibitor. It is a farnesylcysteine mimetic that selectively disrupts the association of active RAS proteins with the plasma membrane. Animal studies demonstrated that salirasib inhibited tumor growth, downregulated gene expression in the cell cycle and Ras signaling pathways. In a clinical study of salirasib combined with standard doses of gemcitabine, it was demonstrated that the two drugs have no overlapping pharmacokinetics. The salirasib recommended dose was 600 mg twice daily and the progression-free survival was 4.7 months. Future studies will determine whether salirasib adds to the anti-tumor activity of drugs approved by the US FDA for pancreatic cancer.
Collapse
Affiliation(s)
- Ernesto Bustinza-Linares
- Department of Investigational Cancer Therapeutics, The Phase I Clinical Trials Program, Unit 455, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | |
Collapse
|
31
|
Charette N, De Saeger C, Lannoy V, Horsmans Y, Leclercq I, Stärkel P. Salirasib inhibits the growth of hepatocarcinoma cell lines in vitro and tumor growth in vivo through ras and mTOR inhibition. Mol Cancer 2010; 9:256. [PMID: 20860815 PMCID: PMC2955616 DOI: 10.1186/1476-4598-9-256] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 09/22/2010] [Indexed: 03/01/2023] Open
Abstract
Background Dysregulation of epidermal growth factor and insulin-like growth factor signaling play important roles in human hepatocellular carcinoma (HCC), leading to frequent activation of their downstream targets, the ras/raf/extracellular signal-regulated kinase (ERK) and the phosphoinositide 3-kinase (PI3K)/Akt/mammalian Target of Rapamycin (mTOR) pathways. Salirasib is an S-prenyl-cysteine analog that has been shown to block ras and/or mTOR activation in several non hepatic tumor cell lines. We investigated in vitro the effect of salirasib on cell growth as well as its mechanism of action in human hepatoma cell lines (HepG2, Huh7, and Hep3B) and its in vivo effect in a subcutaneous xenograft model with HepG2 cells. Results Salirasib induced a time and dose dependent growth inhibition in hepatocarcinoma cells through inhibition of proliferation and partially through induction of apoptosis. A 50 percent reduction in cell growth was obtained in all three cell lines at a dose of 150 μM when they were cultured with serum. By contrast, salirasib was more potent at reducing cell growth after stimulation with EGF or IGF2 under serum-free conditions, with an IC50 ranging from 60 μM to 85 μM. The drug-induced anti-proliferative effect was associated with downregulation of cyclin A and to a lesser extent of cyclin D1, and upregulation of p21 and p27. Apoptosis induction was related to a global pro-apoptotic balance with caspase 3 activation, cytochrome c release, death receptor upregulation, and a reduced mRNA expression of the apoptosis inhibitors cFLIP and survivin. These effects were associated with ras downregulation and mTOR inhibition, without reduction of ERK and Akt activation. In vivo, salirasib reduced tumour growth from day 5 onwards. After 12 days of treatment, mean tumor weight was diminished by 56 percent in the treated animals. Conclusions Our results show for the first time that salirasib inhibits the growth of human hepatoma cell lines through inhibition of proliferation and induction of apoptosis, which is associated with ras and mTOR inhibition. The therapeutic potential of salirasib in human HCC was further confirmed in a subcutaneous xenograft model.
Collapse
Affiliation(s)
- Nicolas Charette
- Laboratory of Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
32
|
|