1
|
Hossain MA. Targeting the RAS upstream and downstream signaling pathway for cancer treatment. Eur J Pharmacol 2024; 979:176727. [PMID: 38866361 DOI: 10.1016/j.ejphar.2024.176727] [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: 03/08/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Cancer often involves the overactivation of RAS/RAF/MEK/ERK (MAPK) and PI3K-Akt-mTOR pathways due to mutations in genes like RAS, RAF, PTEN, and PIK3CA. Various strategies are employed to address the overactivation of these pathways, among which targeted therapy emerges as a promising approach. Directly targeting specific proteins, leads to encouraging results in cancer treatment. For instance, RTK inhibitors such as imatinib and afatinib selectively target these receptors, hindering ligand binding and reducing signaling initiation. These inhibitors have shown potent efficacy against Non-Small Cell Lung Cancer. Other inhibitors, like lonafarnib targeting Farnesyltransferase and GGTI 2418 targeting geranylgeranyl Transferase, disrupt post-translational modifications of proteins. Additionally, inhibition of proteins like SOS, SH2 domain, and Ras demonstrate promising anti-tumor activity both in vivo and in vitro. Targeting downstream components with RAF inhibitors such as vemurafenib, dabrafenib, and sorafenib, along with MEK inhibitors like trametinib and binimetinib, has shown promising outcomes in treating cancers with BRAF-V600E mutations, including myeloma, colorectal, and thyroid cancers. Furthermore, inhibitors of PI3K (e.g., apitolisib, copanlisib), AKT (e.g., ipatasertib, perifosine), and mTOR (e.g., sirolimus, temsirolimus) exhibit promising efficacy against various cancers such as Invasive Breast Cancer, Lymphoma, Neoplasms, and Hematological malignancies. This review offers an overview of small molecule inhibitors targeting specific proteins within the RAS upstream and downstream signaling pathways in cancer.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
2
|
Zubaș A, Ghinet A, Farce A, Dubois J, Bîcu E. Phenothiazine- and Carbazole-Cyanochalcones as Dual Inhibitors of Tubulin Polymerization and Human Farnesyltransferase. Pharmaceuticals (Basel) 2023; 16:888. [PMID: 37375835 DOI: 10.3390/ph16060888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
In the search for innovative approaches to cancer chemotherapy, a chemical library of 49 cyanochalcones, 1a-r, 2a-o, and 3a-p, was designed as dual inhibitors of human farnesyltransferase (FTIs) and tubulin polymerization (MTIs) (FTIs/MTIs), two important biological targets in oncology. This approach is innovative since the same molecule would be able to interfere with two different mitotic events of the cancer cells and prevent these cells from developing an emergency route and becoming resistant to anticancer agents. Compounds were synthesized by the Claisen-Schmidt condensation of aldehydes with N-3-oxo-propanenitriles under classical magnetic stirring and under sonication. Newly synthesized compounds were screened for their potential to inhibit human farnesyltransferase, tubulin polymerization, and cancer cell growth in vitro. This study allowed for the identification of 22 FTIs and 8 dual FTIs/MTIs inhibitors. The most effective molecule was carbazole-cyanochalcone 3a, bearing a 4-dimethylaminophenyl group (IC50 (h-FTase) = 0.12 µM; IC50 (tubulin) = 0.24 µM) with better antitubulin activity than the known inhibitors that were previously reported, phenstatin and (-)-desoxypodophyllotoxin. The docking of the dual inhibitors was realized in both the active site of FTase and in the colchicine binding site of tubulin. Such compounds with a dual inhibitory profile are excellent clinical candidates for the treatment of human cancers and offer new research perspectives in the search for new anti-cancer drugs.
Collapse
Affiliation(s)
- Andreea Zubaș
- Faculty of Chemistry, 'Alexandru Ioan Cuza' University of Iasi, Bulevardul Carol I, nr. 11, 700506 Iasi, Romania
| | - Alina Ghinet
- Faculty of Chemistry, 'Alexandru Ioan Cuza' University of Iasi, Bulevardul Carol I, nr. 11, 700506 Iasi, Romania
- Junia, Health and Environment, Laboratory of Sustainable Chemistry and Health, 59000 Lille, France
- Institut National de la Santé et de la Recherche Médicale, CHU Lille, Institut Pasteur Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, University of Lille, 59000 Lille, France
| | - Amaury Farce
- Institut National de la Santé et de la Recherche Médicale, CHU Lille, U1286-Infinite-Institute for Translational Research in Inflammation, University of Lille, 59000 Lille, France
| | - Joëlle Dubois
- Institut de Chimie des Substances Naturelles, UPR2301, CNRS, Centre de Recherche de Gif, 91190 Gif-sur-Yvette, France
| | - Elena Bîcu
- Faculty of Chemistry, 'Alexandru Ioan Cuza' University of Iasi, Bulevardul Carol I, nr. 11, 700506 Iasi, Romania
| |
Collapse
|
3
|
Kim JH, Lee CH, Baek MC. Dissecting exosome inhibitors: therapeutic insights into small-molecule chemicals against cancer. Exp Mol Med 2022; 54:1833-1843. [PMID: 36446847 PMCID: PMC9707221 DOI: 10.1038/s12276-022-00898-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022] Open
Abstract
Intensive research in the field of cancer biology has revealed unique methods of communication between cells through extracellular vesicles called exosomes. Exosomes are released from a broad spectrum of cell types and serve as functional mediators under physiological or pathological conditions. Hence, blocking the release of exosome bio carriers may prove useful for slowing the progression of certain types of cancers. Therefore, efforts are being made to develop exosome inhibitors to be used both as research tools and as therapies in clinical trials. Thus, studies on exosomes may lead to a breakthrough in cancer research, for which new clinical targets for different types of cancers are urgently needed. In this review, we briefly outline exosome inhibitors and discuss their modes of action and potential for use as therapeutic tools for cancer.
Collapse
Affiliation(s)
- Jong Hyun Kim
- grid.412072.20000 0004 0621 4958Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu, 42472 South Korea
| | - Chan-Hyeong Lee
- grid.258803.40000 0001 0661 1556Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu, 41944 South Korea
| | - Moon-Chang Baek
- grid.258803.40000 0001 0661 1556Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu, 41944 South Korea
| |
Collapse
|
4
|
Exosomes and cancer - Diagnostic and prognostic biomarkers and therapeutic vehicle. Oncogenesis 2022; 11:54. [PMID: 36109501 PMCID: PMC9477829 DOI: 10.1038/s41389-022-00431-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/08/2022] Open
Abstract
AbstractExosomes belong to a subpopulation of extracellular vesicles secreted by the dynamic multistep endocytosis process and carry diverse functional molecular cargoes, including proteins, lipids, nucleic acids (DNA, messenger and noncoding RNA), and metabolites to promote intercellular communication. Proteins and noncoding RNA are among the most abundant contents in exosomes; they have biological functions and are selectively packaged into exosomes. Exosomes derived from tumor, stromal and immune cells contribute to the multiple stages of cancer progression as well as resistance to therapy. In this review, we will discuss the biogenesis of exosomes and their roles in cancer development. Since specific contents within exosomes originate from their cells of origin, this property allows exosomes to function as valuable biomarkers. We will also discuss the potential use of exosomes as diagnostic and prognostic biomarkers or predictors for different therapeutic strategies for multiple cancers. Furthermore, the applications of exosomes as direct therapeutic targets or engineered vehicles for drugs are an important field of exosome study. Better understanding of exosome biology may pave the way to promising exosome-based clinical applications.
Collapse
|
5
|
Targeting Cancer by Using Nanoparticles to Modulate RHO GTPase Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1357:115-127. [DOI: 10.1007/978-3-030-88071-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
6
|
Rajpurohit T, Bhattacharya S. Moving Towards Dawn: KRas Signaling and Treatment in Pancreatic Ductal Adenocarcinoma. Curr Mol Pharmacol 2022; 15:904-928. [PMID: 35088684 DOI: 10.2174/1874467215666220128161647] [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: 07/26/2021] [Revised: 10/20/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022]
Abstract
"Pancreatic ductal adenocarcinoma (PDAC)" is robust, nearly clueless, and all-around deadly among all tumors. Below 10 %, the general 5-year endurance period has remained adamantly unaltered in the last 30 years, regardless of enormous clinical and therapeutic endeavors. The yearly number of deaths is more than the number of recently analyzed cases. Not a classic one, but "Carbohydrate Antigen CA19- 9" remains the prevailing tool for diagnosis. MicroRNAs and non-invasive techniques are now incorporated for the effective prognosis of PDAC than just CA19-9. Mutated "Rat sarcoma virus Ras" conformation "V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog KRas" is 95 % accountable for PDAC, and its active (GTP-bound) formation activates signaling cascade comprising "Rapidly accelerated fibrosarcoma Raf"/"Mitogen-activated protein kinase MEK"/ "Extracellular signal-regulated kinase ERK" with "Phosphoinositide 3-kinase PI3K"/ "protein kinase B Akt"/ "mammalian target of rapamycin mTOR" pathways. KRas has acquired the label of 'undruggable' since the crosstalk in the nexus of pathways compensates for Raf and PI3K signaling cascade blocking. It is arduous to totally regulate KRascoordinated PDAC with traditional medicaments like "gemcitabine GEM" plus nabpaclitaxel/ FOLFIRINOX. For long-haul accomplishments aiming at KRas, future endeavors should be directed to combinatorial methodologies to adequately block KRas pathways at different standpoints. Currently they are contributing to healing PDAC. In this review article, we outline the function of KRas in carcinogenesis in PDAC, its signaling cascade, former techniques utilized in hindering Kras, current and future possibilities for targeting Kras.
Collapse
Affiliation(s)
- Tarun Rajpurohit
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| |
Collapse
|
7
|
Mourot L, Schmitt M, Mouray E, Spichty M, Florent I, Albrecht S. Structure-activity relationship and molecular modelling studies of quinazolinedione derivatives MMV665916 as potential antimalarial agent. Bioorg Med Chem 2021; 51:116513. [PMID: 34798379 DOI: 10.1016/j.bmc.2021.116513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/11/2021] [Accepted: 10/17/2021] [Indexed: 10/19/2022]
Abstract
A series of new quinazolinedione derivatives have been readily synthesized and evaluated for their in vitro antiplasmodial growth inhibition activity. Most of the compounds inhibited P. falciparum FcB1 strain in the low to medium micromolar concentration. The 2-ethoxy 8ag', 2-trifluoromethoxy 8ai' and 4-fluoro-2-methoxy 8ak' showed the best inhibitory activity with EC50 values around 5 µM and were non-toxic to the primary human fibroblast cell line AB943. However, these compounds were less potent than the original hit MMV665916, which showed remarkable growth inhibition with EC50 value of 0.4 µM and presented the highest selectivity index (SI > 250). In addition, a novel approach for determining the docking poses of these quinazolinedione derivatives with their potential protein target, the P. falciparum farnesyltransferase PfFT, was investigated.
Collapse
Affiliation(s)
- Laura Mourot
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA UMR 7042, F-68000 Mulhouse, France
| | - Marjorie Schmitt
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA UMR 7042, F-68000 Mulhouse, France
| | - Elisabeth Mouray
- Unité Molécules de Communication et Adaptation des Micro-organismes, UMR7245, Muséum National d'Histoire Naturelle, CNRS, Sorbonne Universités, Paris, France
| | - Martin Spichty
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA UMR 7042, F-68000 Mulhouse, France
| | - Isabelle Florent
- Unité Molécules de Communication et Adaptation des Micro-organismes, UMR7245, Muséum National d'Histoire Naturelle, CNRS, Sorbonne Universités, Paris, France
| | - Sébastien Albrecht
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA UMR 7042, F-68000 Mulhouse, France.
| |
Collapse
|
8
|
Pegylation of phenothiazine – A synthetic route towards potent anticancer drugs. J Adv Res 2021; 37:279-290. [PMID: 35499049 PMCID: PMC9040145 DOI: 10.1016/j.jare.2021.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 11/22/2022] Open
Abstract
Antitumor activity of two PEGylated phenotiazines was investigated The compounds showed cytotoxic activity against six tumor lines They inhibited the tumor growth in experimental mice The PEGylation improved the phenothiazine biocompatibility A synergistic effect of PEG and phenothiazine toward properties improvement was proved
Introduction Cancer is a big challenge of the 21 century, whose defeat requires efficient antitumor drugs. Objectives The paper aims to investigate the synergistic effect of two structural building blocks, phenothiazine and poly(ethylene glycol), towards efficient antitumor drugs. Methods Two PEGylated phenothiazine derivatives were synthetized by attaching poly(ethylene glycol) of 550 Da to the nitrogen atom of phenothiazine by ether or ester linkage. Their antitumor activity has been investigated on five human tumour lines and a mouse tumor line as well, by determination of IC50. The in vivo toxicity was determined by measuring the LD50 in BALB/c mice by the sequential method and the in vivo antitumor potential was measured by the tumours growth test. The antitumor mechanism was investigated by complexation studies of zinc and magnesium ions characteristic to the farnesyltransferase enzyme, by studies of self-aggregation in the cells proximity and by investigation of the antitumor properties of the acid species resulted by enzymatic cleavage of the PEGylated derivatives. Results The two compounds showed antitumor activity, with IC50 against mouse colon carcinoma cell line comparable with that of the traditional antitumor drugs 5-Fluorouracil and doxorubicin. The phenothiazine PEGylation resulted in a significant toxicity diminishing, the LD50 in BALB/c mice increasing from 952.38 up to 1450 mg/kg, in phenothiazine equivalents. Both compounds inflicted a 92% inhibition of the tumour growth for doses much smaller than LD50. The investigation of the possible tumour inhibition mechanism suggested the nanoaggregate formation and the cleavage of ester bonds as key factors for the inhibition of cancer cell proliferation and biocompatibility improvement. Conclusion Phenothiazine and PEG building blocks have a synergetic effect working for both tumour growth inhibition and biocompatibility improvement. All these findings recommend the PEGylated phenothiazine derivatives as a valuable workbench for a next generation of antitumor drugs.
Collapse
|
9
|
Zhang H, Lu J, Liu J, Zhang G, Lu A. Advances in the discovery of exosome inhibitors in cancer. J Enzyme Inhib Med Chem 2020; 35:1322-1330. [PMID: 32543905 PMCID: PMC7717571 DOI: 10.1080/14756366.2020.1754814] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/07/2020] [Indexed: 12/18/2022] Open
Abstract
Exosomes are small membrane vesicles released by most eukaryotic cells. They are considered to play an essential role in cell-to-cell communication, and It is also found that they serve as functional mediators in many severe diseases, including progression of various types of cancers. Inhibition of exosome release may slow the progression of some cancers; thus, exosome has been an attractive target for cancer treatment. Over the years, considerable efforts have been made to discover novel, highly potent and excellently selective exosome inhibitors. Most of these inhibitors are derived from synthetic compounds, some of which are currently existed drugs and found to have the potential to inhibit exosome release. In this review, we briefly discussed the development of exosome inhibitors that are currently discovered and provided guidance for the future development of inhibitors.
Collapse
Affiliation(s)
- Huarui Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute of Integrated Bioinfomedicine and Translational Science, Hong Kong Baptist University Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Jun Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Integrated Bioinfomedicine and Translational Science, Hong Kong Baptist University Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute of Integrated Bioinfomedicine and Translational Science, Hong Kong Baptist University Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute of Integrated Bioinfomedicine and Translational Science, Hong Kong Baptist University Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute of Integrated Bioinfomedicine and Translational Science, Hong Kong Baptist University Shenzhen Research Institute and Continuing Education, Shenzhen, China
| |
Collapse
|
10
|
Uprety D, Adjei AA. KRAS: From undruggable to a druggable Cancer Target. Cancer Treat Rev 2020; 89:102070. [DOI: 10.1016/j.ctrv.2020.102070] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023]
|
11
|
Dysregulation of Rho GTPases in Human Cancers. Cancers (Basel) 2020; 12:cancers12051179. [PMID: 32392742 PMCID: PMC7281333 DOI: 10.3390/cancers12051179] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/24/2020] [Accepted: 04/30/2020] [Indexed: 01/28/2023] Open
Abstract
Rho GTPases play central roles in numerous cellular processes, including cell motility, cell polarity, and cell cycle progression, by regulating actin cytoskeletal dynamics and cell adhesion. Dysregulation of Rho GTPase signaling is observed in a broad range of human cancers, and is associated with cancer development and malignant phenotypes, including metastasis and chemoresistance. Rho GTPase activity is precisely controlled by guanine nucleotide exchange factors, GTPase-activating proteins, and guanine nucleotide dissociation inhibitors. Recent evidence demonstrates that it is also regulated by post-translational modifications, such as phosphorylation, ubiquitination, and sumoylation. Here, we review the current knowledge on the role of Rho GTPases, and the precise mechanisms controlling their activity in the regulation of cancer progression. In addition, we discuss targeting strategies for the development of new drugs to improve cancer therapy.
Collapse
|
12
|
Pesquet A, Marzag H, Knorr M, Strohmann C, Lawson AM, Ghinet A, Dubois J, Amaury F, Daïch A, Othman M. Access to 3-spiroindolizines containing an isoindole ring through intra-molecular arylation of spiro-N-acyliminium species: a new family of potent farnesyltransferase inhibitors. Org Biomol Chem 2019; 17:2798-2808. [PMID: 30793727 DOI: 10.1039/c8ob02612b] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Based on N-acyliminium species, two efficient and rapid approaches to diversify spirocyclic systems connected by two different carbon centers to the isoindole ring have been developed. The imide reduction and the tandem oxidative cleavage of olefin/formyl-amide equilibration were at first selected as the key steps for these strategies. Ultimately the intramolecular α-amidoalkylation reaction was achieved through the arylation of α-acetoxy lactams or α-hydroxy lactams using, respectively, a Lewis acid or a Brønsted acid depending on the nature of N-acyliminium precursors. The latter led, in addition to the spiro-6-membered aza-heterocycles, to the formation of scarce spiro-5-membered analogues which show promising inhibitory activities on human farnesyltransferase in the nanomolar range demonstrating improved IC50 values of up to 1.5 nM.
Collapse
Affiliation(s)
- Anthony Pesquet
- Normandie Univ, UNILEHAVRE, CNRS, URCOM, 76600 Le Havre, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
High-throughput screening identified selective inhibitors of exosome biogenesis and secretion: A drug repurposing strategy for advanced cancer. Sci Rep 2018; 8:8161. [PMID: 29802284 PMCID: PMC5970137 DOI: 10.1038/s41598-018-26411-7] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 03/22/2018] [Indexed: 12/12/2022] Open
Abstract
Targeting exosome biogenesis and release may have potential clinical implications for cancer therapy. Herein, we have optimized a quantitative high throughput screen (qHTS) assay to identify compounds that modulate exosome biogenesis and/or release by aggressive prostate cancer (PCa) CD63-GFP-expressing C4-2B cells. A total of 4,580 compounds were screened from the LOPAC library (a collection of 1,280 pharmacologically active compounds) and the NPC library (NCGC collection of 3,300 compounds approved for clinical use). Twenty-two compounds were found to be either potent activators or inhibitors of intracellular GFP signal in the CD63-GFP-expressing C4-2B cells. The activity of lead compounds in modulating the secretion of exosomes was validated by a tunable resistive pulse sensing (TRPS) system (qNano-IZON) and flow cytometry. The mechanism of action of the lead compounds in modulating exosome biogenesis and/or secretion were delineated by immunoblot analysis of protein markers of the endosomal sorting complex required for transport (ESCRT)-dependent and ESCRT-independent pathways. The lead compounds tipifarnib, neticonazole, climbazole, ketoconazole, and triademenol were validated as potent inhibitors and sitafloxacin, forskolin, SB218795, fenoterol, nitrefazole and pentetrazol as activators of exosome biogenesis and/or secretion in PC cells. Our findings implicate the potential utility of drug-repurposing as novel adjunct therapeutic strategies in advanced cancer.
Collapse
|
14
|
Kaminskyy D, Kryshchyshyn A, Lesyk R. 5-Ene-4-thiazolidinones - An efficient tool in medicinal chemistry. Eur J Med Chem 2017; 140:542-594. [PMID: 28987611 PMCID: PMC7111298 DOI: 10.1016/j.ejmech.2017.09.031] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 07/14/2017] [Accepted: 09/17/2017] [Indexed: 02/02/2023]
Abstract
The presented review is an attempt to summarize a huge volume of data on 5-ene-4-thiazolidinones being a widely studied class of small molecules used in modern organic and medicinal chemistry. The manuscript covers approaches to the synthesis of 5-ene-4-thiazolidinone derivatives: modification of the C5 position of the basic core; synthesis of the target compounds in the one-pot or multistage reactions or transformation of other related heterocycles. The most prominent pharmacological profiles of 5-ene derivatives of different 4-thiazolidinone subtypes belonging to hit-, lead-compounds, drug-candidates and drugs as well as the most studied targets have been discussed. Currently target compounds (especially 5-en-rhodanines) are assigned as frequent hitters or pan-assay interference compounds (PAINS) within high-throughput screening campaigns. Nevertheless, the crucial impact of the presence/nature of C5 substituent (namely 5-ene) on the pharmacological effects of 5-ene-4-thiazolidinones was confirmed by the numerous listed findings from the original articles. The main directions for active 5-ene-4-thiazolidinones optimization have been shown: i) complication of the fragment in the C5 position; ii) introduction of the substituents in the N3 position (especially fragments with carboxylic group or its derivatives); iii) annealing in complex heterocyclic systems; iv) combination with other pharmacologically attractive fragments within hybrid pharmacophore approach. Moreover, the utilization of 5-ene-4-thiazolidinones in the synthesis of complex compounds with potent pharmacological application is described. The chemical transformations cover mainly the reactions which involve the exocyclic double bond in C5 position of the main core and correspond to the abovementioned direction of the 5-ene-4-thiazolidinone modification.
Collapse
Affiliation(s)
- Danylo Kaminskyy
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, Lviv-10, 79010, Ukraine
| | - Anna Kryshchyshyn
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, Lviv-10, 79010, Ukraine
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, Lviv-10, 79010, Ukraine.
| |
Collapse
|
15
|
Shrestha G, MacNeil SM, McQuerry JA, Jenkins DF, Sharma S, Bild AH. The value of genomics in dissecting the RAS-network and in guiding therapeutics for RAS-driven cancers. Semin Cell Dev Biol 2016; 58:108-17. [PMID: 27338857 PMCID: PMC5951171 DOI: 10.1016/j.semcdb.2016.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 06/18/2016] [Indexed: 12/11/2022]
Abstract
The rise in genomic knowledge over the past decade has revealed the molecular etiology of many diseases, and has identified intricate signaling network activity in human cancers. Genomics provides the opportunity to determine genome structure and capture the activity of thousands of molecular events concurrently, which is important for deciphering highly complex genetic diseases such as cancer. In this review, we focus on genomic efforts directed towards one of cancer's most frequently mutated networks, the RAS pathway. Genomic tools such as gene expression signatures and assessment of mutations across the RAS network enable the capture of RAS signaling complexity. Due to this high level of interaction and cross-talk within the network, efforts to target RAS signaling in the clinic have generally failed, and we currently lack the ability to directly inhibit the RAS protein with high efficacy. We propose that the use of gene expression data can identify effective treatments that broadly inhibit the RAS network as this approach measures pathway activity independent of mutation status or any single mechanism of activation. Here, we review the genomic studies that map the complexity of the RAS network in cancer, and that show how genomic measurements of RAS pathway activation can identify effective RAS inhibition strategies. We also address the challenges and future directions for treating RAS-driven tumors. In summary, genomic assessment of RAS signaling provides a level of complexity necessary to accurately map the network that matches the intricacy of RAS pathway interactions in cancer.
Collapse
Affiliation(s)
- Gajendra Shrestha
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Shelley M MacNeil
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Jasmine A McQuerry
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - David F Jenkins
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Sunil Sharma
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA; Center for Investigational Therapeutics, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Andrea H Bild
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
16
|
Basha NM, Reddy PR, Padmaja A, Padmavathi V. Synthesis and Antioxidant Activity of Bis-oxazolyl/thiazolyl/imidazolyl 1,3,4-Oxadiazoles and 1,3,4-Thiadiazoles. J Heterocycl Chem 2016. [DOI: 10.1002/jhet.2436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | | | - Adivireddy Padmaja
- Department of Chemistry; Sri Venkateswara University; Tirupati 517 502 Andhra Pradesh India
| | | |
Collapse
|
17
|
Abstract
Mutations in the KRAS oncogene represent one of the most prevalent genetic alterations in colorectal cancer (CRC), the third leading cause of cancer-related death in the US. In addition to their well-characterized function in driving tumor progression, KRAS mutations have been recognized as a critical determinant of the therapeutic response of CRC. Recent studies demonstrate that KRAS-mutant tumors are intrinsically insensitive to clinically-used epidermal growth factor receptor (EGFR) targeting antibodies, including cetuximab and panitumumab. Acquired resistance to the anti-EGFR therapy was found to be associated with enrichment of KRAS-mutant tumor cells. However, the underlying molecular mechanism of mutant-KRAS-mediated therapeutic resistance has remained unclear. Despite intensive efforts, directly targeting mutant KRAS has been largely unsuccessful. This review summarizes the recent advances in understanding the biological function of KRAS mutations in determining the therapeutic response of CRC, highlighting several recently developed agents and strategies for targeting mutant KRAS, such as synthetic lethal interactions.
Collapse
|
18
|
Novel indolizine derivatives with unprecedented inhibitory activity on human farnesyltransferase. Bioorg Med Chem Lett 2014; 24:5777-5781. [DOI: 10.1016/j.bmcl.2014.10.044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/13/2014] [Accepted: 10/14/2014] [Indexed: 11/18/2022]
|
19
|
Premakumari C, Seenaiah D, Srilakshmi A, Sai Gopal DVR, Padmavathi V. Synthesis and Antimicrobial Activity of Amido Sulfonamido Methane Linked Bisoxazoles, Bisthiazoles, and Bisimidazoles. J Heterocycl Chem 2014. [DOI: 10.1002/jhet.1884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Dandu Seenaiah
- Department of Chemistry; Sri Venkateswara University; Tirupati 517 502 Andhra Pradesh India
| | - Akula Srilakshmi
- Department of Microbiology; Sri Venkateswara University; Tirupati 517 502 Andhra Pradesh India
| | | | | |
Collapse
|
20
|
Ma Y, Gong Y, Cheng Z, Loganathan S, Kao C, Sarkaria JN, Abel TW, Wang J. Critical functions of RhoB in support of glioblastoma tumorigenesis. Neuro Oncol 2014; 17:516-25. [PMID: 25216671 DOI: 10.1093/neuonc/nou228] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND RhoB is a member of the Rho small GTPase family that regulates cytoskeletal dynamics and vesicle trafficking. The RhoB homologs, RhoA and RhoC, have been shown to promote cancer progression and metastasis. In contrast, the functions of RhoB in human cancers are context dependent. Although expression of RhoB inversely correlates with disease progression in several epithelial cancers, recent data suggest that RhoB may support malignant phenotypes in certain cancer types. METHODS We assessed RhoB protein levels in glioma surgical specimens and patient-derived xenografts. The roles of RhoB in glioblastoma were determined by loss-of-function and gain-of-function assays in vitro and in vivo. The impact on p53 and STAT3 signaling was investigated. RESULTS RhoB expression was similar in tumor specimens compared with normal neural tissues obtained from epilepsy surgery. RhoB was expressed in the vast majority of xenograft tumors and spheroid cultures. Knockdown of RhoB induced cell-cycle arrest and apoptosis and compromised in vivo tumorigenic potential. However, overexpression of wild-type RhoB or a constitutively active mutant (RhoB-V14) did not significantly affect cell growth, which suggests that RhoB is not a rate-limiting oncogenic factor and is consistent with the scarcity of RhoB mutations in human cancer. Knockdown of RhoB reduced basal STAT3 activity and impaired cytokine-induced STAT3 activation. In glioblastoma tumors retaining wild-type p53, depletion of RhoB also activated p53 and induced expression of p21(CIP1) (/WAF1). CONCLUSIONS Our data suggest that RhoB belongs to an emerging class of "nononcogene addiction" factors that are essential for maintenance of malignant phenotypes in human cancers.
Collapse
Affiliation(s)
- Yufang Ma
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (Y.M., Y.G., Z.C., C.K., J.W.); Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (T.W.A.); Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee (J.W.); Department of Pain Management and Oncology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China (Z.C.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Yuanying Gong
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (Y.M., Y.G., Z.C., C.K., J.W.); Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (T.W.A.); Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee (J.W.); Department of Pain Management and Oncology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China (Z.C.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Zhixiang Cheng
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (Y.M., Y.G., Z.C., C.K., J.W.); Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (T.W.A.); Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee (J.W.); Department of Pain Management and Oncology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China (Z.C.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Sudan Loganathan
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (Y.M., Y.G., Z.C., C.K., J.W.); Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (T.W.A.); Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee (J.W.); Department of Pain Management and Oncology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China (Z.C.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Crystal Kao
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (Y.M., Y.G., Z.C., C.K., J.W.); Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (T.W.A.); Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee (J.W.); Department of Pain Management and Oncology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China (Z.C.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Jann N Sarkaria
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (Y.M., Y.G., Z.C., C.K., J.W.); Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (T.W.A.); Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee (J.W.); Department of Pain Management and Oncology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China (Z.C.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Ty W Abel
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (Y.M., Y.G., Z.C., C.K., J.W.); Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (T.W.A.); Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee (J.W.); Department of Pain Management and Oncology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China (Z.C.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Jialiang Wang
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (Y.M., Y.G., Z.C., C.K., J.W.); Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (T.W.A.); Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee (J.W.); Department of Pain Management and Oncology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China (Z.C.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| |
Collapse
|
21
|
Sharma GVM, Ramesh A, Singh A, Srikanth G, Jayaram V, Duscharla D, Jun JH, Ummanni R, Malhotra SV. Imidazole derivatives show anticancer potential by inducing apoptosis and cellular senescence. MEDCHEMCOMM 2014. [DOI: 10.1039/c4md00277f] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
22
|
Synthesis, antioxidant, and cytotoxic activities of bis(oxazolyl/thiazolyl/imidazolyl)amidomethanesulfonyl Acetamides. Med Chem Res 2014. [DOI: 10.1007/s00044-014-1109-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
23
|
Peptide chemistry applied to a new family of phenothiazine-containing inhibitors of human farnesyltransferase. Bioorg Med Chem Lett 2014; 24:3180-5. [DOI: 10.1016/j.bmcl.2014.04.102] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/23/2014] [Accepted: 04/25/2014] [Indexed: 02/02/2023]
|
24
|
Divya K, Sravya G, Padmaja A, Padmavathi V. Synthesis and antimicrobial activity of bis-heterocyclic sulfamoyl acetamides. RESEARCH ON CHEMICAL INTERMEDIATES 2014. [DOI: 10.1007/s11164-014-1540-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
25
|
Abuhaie CM, Ghinet A, Farce A, Dubois J, Rigo B, Bîcu E. Synthesis and biological evaluation of a new series of N-ylides as protein farnesyltransferase inhibitors. Bioorg Med Chem Lett 2013; 23:5887-92. [DOI: 10.1016/j.bmcl.2013.08.088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/20/2013] [Accepted: 08/23/2013] [Indexed: 11/24/2022]
|
26
|
Wang Y, Kaiser CE, Frett B, Li HY. Targeting mutant KRAS for anticancer therapeutics: a review of novel small molecule modulators. J Med Chem 2013; 56:5219-30. [PMID: 23566315 PMCID: PMC4666308 DOI: 10.1021/jm3017706] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The RAS proteins play a role in cell differentiation, proliferation, and survival. Aberrant RAS signaling has been found to play a role in 30% of all cancers. KRAS, a key member of the RAS protein family, is an attractive cancer target, as frequent point mutations in the KRAS gene render the protein constitutively active. A number of attempts have been made to target aberrant KRAS signaling by identifying small molecule compounds that (1) are synthetic lethal to mutant KRAS, (2) block KRAS/GEF interactions, (3) inhibit downstream KRAS effectors, or (4) inhibit the post-translational processing of RAS proteins. In addition, inhibition of novel targets outside the main KRAS signaling pathway, specifically the cell cycle related kinase PLK1, has been shown have an effect in cells that harbor mutant KRAS. Herein we review the use of various high-throughput screening assays utilized to identify new small-molecule compounds capable of targeting mutant KRAS-driven cancers.
Collapse
Affiliation(s)
- Yuanxiang Wang
- Department of Pharmacoloy and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
- BIO5 Oro Valley, The University of Arizona, 1580 Hanley Boulevard, Oro Valley, Arizona 85737, United States
| | - Christine E. Kaiser
- Department of Pharmacoloy and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Brendan Frett
- Department of Pharmacoloy and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
- BIO5 Oro Valley, The University of Arizona, 1580 Hanley Boulevard, Oro Valley, Arizona 85737, United States
| | - Hong-yu Li
- Department of Pharmacoloy and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
- BIO5 Oro Valley, The University of Arizona, 1580 Hanley Boulevard, Oro Valley, Arizona 85737, United States
| |
Collapse
|
27
|
Mahaboob Basha N, Lavanya G, Padmaja A, Padmavathi V. Synthesis and Antioxidant Activities of Acetamidomethylsulfonyl Bis Heterocycles-Oxazolyl/Thiazolyl/Imidazolyl-1,3,4-Oxadiazoles. Arch Pharm (Weinheim) 2013; 346:511-20. [DOI: 10.1002/ardp.201300115] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/01/2013] [Accepted: 05/06/2013] [Indexed: 11/10/2022]
Affiliation(s)
| | - Gopala Lavanya
- Department of Chemistry; Sri Venkateswara University; Tirupati; Andhra Pradesh; India
| | - Adivireddy Padmaja
- Department of Chemistry; Sri Venkateswara University; Tirupati; Andhra Pradesh; India
| | | |
Collapse
|
28
|
Synthesis, crystal structure and antibacterial activity of new highly functionalized ionic compounds based on the imidazole nucleus. Bioorg Med Chem Lett 2013; 23:1274-8. [DOI: 10.1016/j.bmcl.2013.01.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 12/21/2012] [Accepted: 01/02/2013] [Indexed: 11/20/2022]
|
29
|
Abuhaie CM, Ghinet A, Farce A, Dubois J, Gautret P, Rigo B, Belei D, Bîcu E. Synthesis and biological evaluation of a new series of phenothiazine-containing protein farnesyltransferase inhibitors. Eur J Med Chem 2013. [DOI: 10.1016/j.ejmech.2012.11.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
30
|
Synthesis and biological evaluation of new phenothiazine derivatives bearing a pyrazole unit as protein farnesyltransferase inhibitors. Bioorg Med Chem Lett 2012; 22:6896-902. [DOI: 10.1016/j.bmcl.2012.09.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 09/07/2012] [Accepted: 09/10/2012] [Indexed: 01/23/2023]
|
31
|
Belei D, Dumea C, Samson A, Farce A, Dubois J, Bîcu E, Ghinet A. New farnesyltransferase inhibitors in the phenothiazine series. Bioorg Med Chem Lett 2012; 22:4517-22. [DOI: 10.1016/j.bmcl.2012.06.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 06/01/2012] [Accepted: 06/03/2012] [Indexed: 02/07/2023]
|
32
|
Marra M, Salzano G, Leonetti C, Tassone P, Scarsella M, Zappavigna S, Calimeri T, Franco R, Liguori G, Cigliana G, Ascani R, La Rotonda MI, Abbruzzese A, Tagliaferri P, Caraglia M, De Rosa G. Nanotechnologies to use bisphosphonates as potent anticancer agents: the effects of zoledronic acid encapsulated into liposomes. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 7:955-64. [PMID: 21453789 DOI: 10.1016/j.nano.2011.03.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 02/20/2011] [Accepted: 03/11/2011] [Indexed: 01/09/2023]
Abstract
UNLABELLED Zoledronic acid (ZOL) is a potent amino-bisphosphonate used for the treatment of bone metastases with recently reported antitumor activity. However, the short plasma half-life and rapid accumulation in bone limits the use of ZOL as an antitumor agent in extraskeletal tissues. Therefore, we developed stealth liposomes encapsulating ZOL (LipoZOL) to increase extraskeletal drug availability. Compared to free ZOL, LipoZOL induced a stronger inhibition of growth of a range of different cancer cell lines in vitro. LipoZOL also caused significantly larger inhibition of tumor growth and increased the overall survival in murine models of human prostate cancer and multiple myeloma, in comparison with ZOL. Moreover, a strong inhibition of vasculogenetic events without evidence of necrosis in the tumor xenografts from prostate cancer was recorded after treatment with LipoZOL. We demonstrated both antitumor activity and tolerability of LipoZOL in preclinical animal models of both solid and hematopoietic malignancies, providing a rationale for early exploration of use of LipoZOL as a potential anticancer agent in cancer patients. FROM THE CLINICAL EDITOR The short plasma half-life and rapid accumulation in bone limits the use of zoledronic acid as an antitumor agent in extraskeletal tissues. Therefore, stealth liposomes encapsulating ZOL (LipoZOL) have been developed to increase extraskeletal drug availability.
Collapse
Affiliation(s)
- Monica Marra
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kauh J, Chanel-Vos C, Escuin D, Fanucchi MP, Harvey RD, Saba N, Shin DM, Gal A, Pan L, Kutner M, Ramalingam SS, Bender L, Marcus A, Giannakakou P, Khuri FR. Farnesyl transferase expression determines clinical response to the docetaxel-lonafarnib combination in patients with advanced malignancies. Cancer 2011; 117:4049-59. [PMID: 21365629 DOI: 10.1002/cncr.26004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 12/10/2010] [Accepted: 01/10/2011] [Indexed: 11/11/2022]
Abstract
BACKGROUND Lonafarnib (LNF) is a protein farnesyl transferase (FTase) inhibitor that has shown synergistic activity with taxanes in preclinical models and early stage clinical trials. Preclinical findings suggested tubulin acetylation and FTase expression levels may be important determinants of drug sensitivity that would help identify patient populations more likely to benefit from this regimen. This pilot study evaluated the biological effects of LNF and docetaxel (DTX) combination therapy in refractory solid tumors by comparing pretreatment and post-treatment tumor biopsies. METHODS Patients with histologically confirmed locally advanced or metastatic solid malignancies refractory to standard therapies or with no effective therapies available were eligible. Patients were randomized to 1 of 4 dosing cohorts: 1) 30 mg/m², 100 mg; 2) 36 mg/m², 100 mg; 3) 30 mg/m², 150 mg; or 4) 36 mg/m², 150 mg of DTX intravenously weekly, LNF orally twice daily, respectively. RESULTS Of the 38 patients enrolled, 36 were treated, and 29 were evaluable for toxicity and response assessment. The combination of LNF and DTX was tolerated in all cohorts with the exception of a 28% incidence of grade 3/4 diarrhea, which was manageable with aggressive antidiarrheal regimens. Seven patients derived clinically meaningful benefit from this combination treatment; these patients had significantly lower basal FTase-beta mRNA expression levels than the mean study population level (P < .05). Correlation of clinical benefit with tubulin acetylation content as well as basal acetyl-tubulin content were evaluated. However, no significant correlation was found. CONCLUSIONS Despite the small number of patients, these findings support our preclinical mechanistic studies and warrant further clinical investigations using FTase-beta mRNA expression as a potential predictive biomarker to select for an enriched patient population to study the effects of taxane and FTase inhibitor combination therapies.
Collapse
Affiliation(s)
- John Kauh
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Duverna R, Ablordeppey SY, Lamango NS. Biochemical and docking analysis of substrate interactions with polyisoprenylated methylated protein methyl esterase. Curr Cancer Drug Targets 2010; 10:634-48. [PMID: 20491620 PMCID: PMC2947619 DOI: 10.2174/156800910791859443] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2009] [Accepted: 05/04/2010] [Indexed: 11/22/2022]
Abstract
Polyisoprenylated proteins (PPs) methylation by polyisoprenylated protein methyl transferase (PPMTase) is counteracted by polyisoprenylated methylated protein methyl esterase (PMPMEase). This is the only reversible step of the polyisoprenylation pathway as the relative amounts of the acid and ester forms are determined by the two competing reactions. Since PMPMEase and PPMTase may influence both the structural/functional conformations of PPs, a thorough study of these enzymes is essential to our understanding of the structural/functional features of PPs. PMPMEase has been reported under such pseudonyms as human carboxylesterase 1 (hCE1) because of its apparent broad substrate spectrum. The current study aimed to show the complementarity between its active site and the polyisoprenylated substrates that it metabolizes. Kinetics analysis was conducted with N-, S- and O-substituted substrates using porcine liver PMPMEase and docking analysis using Arguslab. Consistent with the biochemical analysis, the S-ethyl analog yielded an AScore binding energy of -11.32 compared to -13.48, -14.88, -16.15, and -16.81 kcal/mol for S-prenyl (C-5), S-trans-geranyl (C-10), S-trans,trans-farnesyl (C-15) and S-all trans-geranylgeranyl (C-20), respectively. The all trans-geranylgeranyl moiety provides the optimal size for active site interactions. The data reveal that the trans,trans-farnesyl and all trans-geranylgeranyl groups, which are reminiscent of endogenous PPs modifications, have the highest affinity for PMPMEase. Since PPs such as monomeric G-proteins are oncogenic, PMPMEase may be viewed as a viable target for anticancer drug development. The analyses reveal the important structural elements for the design of specific PMPMEase inhibitors to serve in the modulation of oncogenic PPs activities. The results also show that hCE1's repertoire of substrates extends beyond xenobiotics to include PPs as its endogenous substrates.
Collapse
Affiliation(s)
- R Duverna
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | | | | |
Collapse
|
35
|
Kaminskyy DV, Lesyk RB. Structure–anticancer activity relationships among 4-azolidinone-3-carboxylic acids derivatives. ACTA ACUST UNITED AC 2010. [DOI: 10.7124/bc.000150] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
| | - R. B. Lesyk
- Danylo Halytsky Lviv National Medical University
| |
Collapse
|
36
|
Hersey P, Zhang XD. Treatment combinations targeting apoptosis to improve immunotherapy of melanoma. Cancer Immunol Immunother 2009; 58:1749-59. [PMID: 19551381 PMCID: PMC11030855 DOI: 10.1007/s00262-009-0732-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Accepted: 06/11/2009] [Indexed: 10/20/2022]
Abstract
Immunotherapy based on T cell responses to the tumor is believed to involve killing of cancer cells by induction of apoptosis. The predominant mechanisms are death ligand-induced signaling mainly by TNF-related apoptosis-inducing ligand (TRAIL) mediated by CD4 T cells, monocytes and dendritic cells, and perforin/granzyme mediated apoptosis mediated by CD8 T cells and NK cells. Resistance against TRAIL involves loss of TRAIL death receptors and/or activation of the MEK and/or Akt signal pathways. Resistance to CD8 CTL responses also involves activation of the MEK and/or Akt pathways. Apoptosis induced by immune responses is regulated by the Bcl-2 family of proteins. Many reagents have been developed against the Bcl-2 antiapoptotic proteins and clinical trials combining them with immunotherapy are awaited. The second group of agents that regulate the Bcl-2 family of proteins are the signal pathway inhibitors. Clinical trials with inhibitors of RAS, RAF or MEK are in progress and would appear an exciting combination with immunotherapy. One of the main drivers of resistance to apoptosis are adaptive mechanisms that allow cancer cells to overcome endoplasmic reticulum (ER) stress. These adaptive mechanisms inhibit practically all known apoptotic pathways and create an acidic environment that may reduce infiltration of lymphocytes against the tumor. The signal pathway inhibitors may be effective against these adaptive processes but additional agents that target ER stress pathways are in development. In conclusion, combination of immunotherapy with agents that target antiapoptotic mechanisms in cancer cells offers a new approach that requires evaluation in clinical trials.
Collapse
Affiliation(s)
- Peter Hersey
- Oncology and Immunology Unit, Calvary Mater Newcastle Hospital, Room 443, David Maddison Clinical Sciences Building, Cnr. King and Watt Streets, Newcastle, NSW 2300, Australia.
| | | |
Collapse
|
37
|
Hersey P, Bastholt L, Chiarion-Sileni V, Cinat G, Dummer R, Eggermont AMM, Espinosa E, Hauschild A, Quirt I, Robert C, Schadendorf D. Small molecules and targeted therapies in distant metastatic disease. Ann Oncol 2009; 20 Suppl 6:vi35-40. [PMID: 19617296 PMCID: PMC2712592 DOI: 10.1093/annonc/mdp254] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy, biological agents or combinations of both have had little impact on survival of patients with metastatic melanoma. Advances in understanding the genetic changes associated with the development of melanoma resulted in availability of promising new agents that inhibit specific proteins up-regulated in signal cell pathways or inhibit anti-apoptotic proteins. Sorafenib, a multikinase inhibitor of the RAF/RAS/MEK pathway, elesclomol (STA-4783) and oblimersen (G3139), an antisense oligonucleotide targeting anti-apoptotic BCl-2, are in phase III clinical studies in combination with chemotherapy. Agents targeting mutant B-Raf (RAF265 and PLX4032), MEK (PD0325901, AZD6244), heat-shock protein 90 (tanespimycin), mTOR (everolimus, deforolimus, temsirolimus) and VEGFR (axitinib) showed some promise in earlier stages of clinical development. Receptor tyrosine-kinase inhibitors (imatinib, dasatinib, sunitinib) may have a role in treatment of patients with melanoma harbouring c-Kit mutations. Although often studied as single agents with disappointing results, new targeted drugs should be more thoroughly evaluated in combination therapies. The future of rational use of new targeted agents also depends on successful application of analytical techniques enabling molecular profiling of patients and leading to selection of likely therapy responders.
Collapse
Affiliation(s)
- P Hersey
- Immunology and Oncology Unit, Calvary Mater Newcastle Hospital, New South Wales, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Molecular biology of mantle cell lymphoma: From profiling studies to new therapeutic strategies. Blood Rev 2009; 23:205-16. [DOI: 10.1016/j.blre.2009.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
39
|
Chow LQM, Eckhardt SG, O’Bryant CL, Schultz MK, Morrow M, Grolnic S, Basche M, Gore L. A phase I safety, pharmacological, and biological study of the farnesyl protein transferase inhibitor, lonafarnib (SCH 663366), in combination with cisplatin and gemcitabine in patients with advanced solid tumors. Cancer Chemother Pharmacol 2008; 62:631-46. [PMID: 18058098 PMCID: PMC2813768 DOI: 10.1007/s00280-007-0646-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Accepted: 11/12/2007] [Indexed: 10/22/2022]
Abstract
PURPOSE This phase I study was conducted to evaluate the safety, tolerability, pharmacological properties and biological activity of the combination of the lonafarnib, a farnesylproteintransferase (FTPase) inhibitor, with gemcitabine and cisplatin in patients with advanced solid malignancies. EXPERIMENTAL DESIGN This was a single institution study to determine the maximal tolerated dose (MTD) of escalating lonafarnib (75-125 mg po BID) with gemcitabine (750-1,000 mg/m(2) on days 1, 8, 15) and fixed cisplatin (75 mg/m(2) day 1) every 28 days. Due to dose-limiting toxicities (DLTs) of neutropenia and thrombocytopenia in initial patients, these patients were considered "heavily pre-treated" and the protocol was amended to limit prior therapy and re-escalate lonafarnib in "less heavily pre-treated patients" on 28-day and 21-day schedules. Cycle 1 and 2 pharmacokinetics (PK), and farnesylation of the HDJ2 chaperone protein and FPTase activity were analyzed. RESULTS Twenty-two patients received 53 courses of therapy. Nausea, vomiting, and fatigue were frequent in all patients. Severe toxicities were observed in 91% of patients: neutropenia (41%), nausea (36%), thrombocytopenia (32%), anemia (23%) and vomiting (23%). Nine patients withdrew from the study due to toxicity. DLTs of neutropenia, febrile neutropenia, thrombocytopenia, and fatigue limited dose-escalation on the 28-day schedule. The MTD was established as lonafarnib 75 mg BID, gemcitabine 750 mg/m(2) days 1, 8, 15, and cisplatin 75 mg/m(2) in heavily pre-treated patients. The MTD in the less heavily pre-treated patients could not be established on the 28-day schedule as DLTs were observed at the lowest dose level, and dose escalation was not completed on the 21-day schedule due to early study termination by the Sponsor. No PK interactions were observed. FTPase inhibition was not observed at the MTD, however HDJ-2 gel shift was observed in one patient at the 100 mg BID lonafarnib dose. Anti-cancer activity was observed: four patients had stable disease lasting >2 cycles, one subject had a complete response, and another had a partial response, both with metastatic breast cancer. CONCLUSION Lonafarnib 75 mg BID, gemcitabine 750 mg/m(2) days 1, 8, 15, and cisplatin 75 mg/m(2) day 1 on a 28-day schedule was established as the MTD. Lonafarnib did not demonstrate FTPase inhibition at these doses. Despite the observed efficacy, substantial toxicity and questionable contribution of anti-tumor activity of lonafarnib to gemcitabine and cisplatin limits further exploration of this combination.
Collapse
Affiliation(s)
| | | | | | | | - Mark Morrow
- University of Colorado Cancer Center, Aurora, CO, USA
| | - Stacy Grolnic
- University of Colorado Cancer Center, Aurora, CO, USA
| | | | - Lia Gore
- University of Colorado Cancer Center, Aurora, CO, USA
- The Children’s Hospital, Denver, CO, USA
- University of Colorado Health Sciences Center at Fitzsimons, Mail Stop 8302, PO Box 6511, Aurora, CO 80045, USA
| |
Collapse
|
40
|
Field KA, Charoenthongtrakul S, Bishop JM, Refaeli Y. Farnesyl transferase inhibitors induce extended remissions in transgenic mice with mature B cell lymphomas. Mol Cancer 2008; 7:39. [PMID: 18489761 PMCID: PMC2409375 DOI: 10.1186/1476-4598-7-39] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Accepted: 05/19/2008] [Indexed: 12/16/2022] Open
Abstract
Background We have used a mouse model based on overexpression of c-Myc in B cells genetically engineered to be self-reactive to test the hypothesis that farnesyl transferase inhibitors (FTIs) can effectively treat mature B cell lymphomas. FTIs are undergoing clinical trials to treat both lymphoid and non-lymphoid malignancies and we wished to obtain evidence to support the inclusion of B cell lymphomas in future trials. Results We report that two FTIs, L-744,832 and SCH66336, blocked the growth of mature B cell lymphoma cells in vitro and in vivo. The FTI treatment affected the proliferation and survival of the transformed B cells to a greater extent than naïve B cells stimulated with antigen. In syngeneic mice transplanted with the transgenic lymphoma cells, L-744,832 treatment prevented the growth of the tumor cells and the morbidity associated with the resulting lymphoma progression. Tumors that arose from transplantation of the lymphoma cells regressed with as little as three days of treatment with L-744,832 or SCH66336. Treatment of these established lymphomas with L-744,832 for seven days led to long-term remission of the disease in approximately 25% of animals. Conclusion FTI treatment can block the proliferation and survival of self-reactive transformed B cells that overexpress Myc. In mice transplanted with mature B cell lymphomas, we found that FTI treatment led to regression of disease. FTIs warrant further consideration as therapeutic agents for mature B cell lymphomas and other lymphoid tumors.
Collapse
Affiliation(s)
- Kenneth A Field
- Cell Biology and Biochemistry Program, Biology Department, Bucknell University, Lewisburg, PA, USA.
| | | | | | | |
Collapse
|
41
|
|
42
|
Gilleron P, Wlodarczyk N, Houssin R, Farce A, Laconde G, Goossens JF, Lemoine A, Pommery N, Hénichart JP, Millet R. Design, synthesis and biological evaluation of substituted dioxodibenzothiazepines and dibenzocycloheptanes as farnesyltransferase inhibitors. Bioorg Med Chem Lett 2007; 17:5465-71. [PMID: 17827015 DOI: 10.1016/j.bmcl.2007.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Revised: 06/29/2007] [Accepted: 07/01/2007] [Indexed: 12/24/2022]
Abstract
A new series of FTase inhibitors containing a tricyclic moiety--dioxodibenzothiazepine or dibenzocycloheptane--has been designed and synthesized. Among them, dioxodibenzothiazepine 18d displayed significant inhibitory FTase activity (IC(50)=17.3 nM) and antiproliferative properties.
Collapse
Affiliation(s)
- Pauline Gilleron
- Institut de Chimie Pharmaceutique Albert Lespagnol, EA 2692, IFR 114, Université de Lille 2, 3 rue du Professeur Laguesse, BP 83, 59006 Lille, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Johnson MD, Sade B, Milano MT, Lee JH, Toms SA. New prospects for management and treatment of inoperable and recurrent skull base meningiomas. J Neurooncol 2007; 86:109-22. [PMID: 17624496 DOI: 10.1007/s11060-007-9434-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Accepted: 06/07/2007] [Indexed: 12/25/2022]
Abstract
Skull base, including optic nerve, cavernous sinus, clival and foramen magnum tumors represent a major challenge for neurosurgeons and neuro-oncologists. Growth regulatory signaling pathways for these tumors are of increasing interest as potential targets for new chemotherapy. Those differentially activated in various grades of meningiomas are currently being identified as well. This article reviews some recent findings pathways that appear to regulate meningioma growth. Potential targets for novel therapies are also discussed.
Collapse
Affiliation(s)
- Mahlon D Johnson
- Department of Pathology and Laboratory Medicine, Division of Neuropathology, University of Rochester Medical Center, 601 Elmwood Ave. Box 626, Rochester, NY 14623, USA.
| | | | | | | | | |
Collapse
|
44
|
Puntambekar DS, Giridhar R, Yadav MR. Inhibition of farnesyltransferase: a rational approach to treat cancer? J Enzyme Inhib Med Chem 2007; 22:127-40. [PMID: 17518338 DOI: 10.1080/14756360601072841] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
This article presents in brief the development of farnesyltransferase inhibitors (FTIs) and their preclinical and clinical status. In this review the mechanism of action of FTIs is discussed and their selectivity issue towards tumor cells is also addressed. The significant efficacy of FTIs as single or combined agents in preclinical studies stands in contrast with only moderate effects in Clinical Phase II-III studies. This suggests that there is a need to further explore and understand the complex mechanism of action of FTIs and their interaction with cytotoxic agents.
Collapse
Affiliation(s)
- Devendra S Puntambekar
- Pharmacy Department, Faculty of Technology and Engineering, The Maharaja Sayajirao University of Baroda, Vadodara 390 001, Gujarat, India
| | | | | |
Collapse
|
45
|
Balducci L. Molecular insight in cancer treatment and prevention. Int J Biochem Cell Biol 2007; 39:1329-36. [PMID: 17531523 DOI: 10.1016/j.biocel.2007.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2006] [Revised: 03/08/2007] [Accepted: 03/09/2007] [Indexed: 12/17/2022]
Abstract
This article explores the impact of new insights in the biology of cancer on the treatment and the prevention of this disease. There are two types of targeted cancer treatment, afforded by the molecular profile of cancer. One concerns the use of agents targeted on a specific component of the cancer cells (e.g., CD20 in lymphoma) or on a specific survival function of the cancer cell (growth-factor-receptor interaction; transduction cascade). The other concerns the recognition of tumors that are more or less likely to benefit from cytotoxic chemotherapy according to their genomic or proteomic profile. Cancer prevention may benefit from new molecular insight in cancer biology as these processes allow early diagnosis of cancer, identification of patients at risk for cancer, and may provide intermediate markers for chemoprevention studies.
Collapse
Affiliation(s)
- Lodovico Balducci
- Interdisciplinary Oncology, University of South Florida College of Medicine, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| |
Collapse
|
46
|
Schafer-Hales K, Iaconelli J, Snyder JP, Prussia A, Nettles JH, El-Naggar A, Khuri FR, Giannakakou P, Marcus AI. Farnesyl transferase inhibitors impair chromosomal maintenance in cell lines and human tumors by compromising CENP-E and CENP-F function. Mol Cancer Ther 2007; 6:1317-28. [PMID: 17431110 DOI: 10.1158/1535-7163.mct-06-0703] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Farnesyl transferase inhibitors (FTI) exhibit anticancer activity as a single agent in preclinical studies and show promise in combination with other therapeutics in clinical trials. Previous studies show that FTIs arrest cancer cells in mitosis; however, the mechanism by which this occurs is unclear. Here, we observed that treatment of various cancer cell lines with the FTI lonafarnib caused mitotic chromosomal alignment defects, leaving cells in a pseudometaphase state, whereby both aligned chromosomes and chromosomes juxtaposed to the spindle poles (termed "lagging chromosomes") were observed in the same cell. To determine how this occurs, we investigated the functionality of two farnesylated mitotic proteins, CENP-E and CENP-F, which mediate chromosomal capture and alignment. The data show that lonafarnib in proliferating cancer cells depletes CENP-E and CENP-F from metaphase but not prometaphase kinetochores. Loss of CENP-E and CENP-F metaphase localization triggered aberrant chromosomal maintenance, causing aligned chromosomes to be prematurely released from the spindle equator and become lagging chromosomes, resulting in a mitotic delay. Furthermore, lonafarnib treatment reduces sister kinetochore tension and activates the BubR1 spindle checkpoint, suggesting that farnesylation of CENP-E and CENP-F is critical for their functionality in maintaining kinetochore-microtubule interactions. Importantly, apparently similar chromosomal alignment defects were observed in head and neck tumors samples from a phase I trial with lonafarnib, providing support that lonafarnib disrupts chromosomal maintenance in human cancers. Lastly, to examine how farnesylation could regulate CENP-E in mediating kinetochore-microtubule attachments, we examined possible docking motifs of a farnesyl group on the outer surface of the microtubule. This analysis revealed three hydrophobic patches on the tubulin dimer for insertion of a farnesyl group, alluding to the possibility of an association between a farnesyl group and the microtubule.
Collapse
|
47
|
Caraglia M, Marra M, Leonetti C, Meo G, D'Alessandro AM, Baldi A, Santini D, Tonini G, Bertieri R, Zupi G, Budillon A, Abbruzzese A. R115777 (Zarnestra)/Zoledronic acid (Zometa) cooperation on inhibition of prostate cancer proliferation is paralleled by Erk/Akt inactivation and reduced Bcl-2 and bad phosphorylation. J Cell Physiol 2007; 211:533-43. [PMID: 17192846 DOI: 10.1002/jcp.20960] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Zoledronic acid (ZOL) has proved activity in bone metastases from prostate cancer through inhibition of mevalonate pathway and of prenylation of intracellular proteins. We have reported that ZOL synergizes with R115777 farnesyltransferase inhibitor (FTI, Zarnestra) in inducing apoptosis and growth inhibition on epidermoid cancer cells. Here, we have studied the effects of the combination of these agents in prostate adenocarcinoma models and, specifically, on androgen-independent (PC3 and DU145) and -dependent (LNCaP) prostate cancer cell lines. We have found that ZOL and R115777 were synergistic in inducing both growth inhibition and apoptosis in prostate adenocarcinoma cells. These effects were paralleled by disruption of Ras-->Erk and Akt survival pathways, consequent decreased phosphorylation of both mitochondrial bcl-2 and bad proteins, and caspase activation. Finally, ZOL/R115777 combination induced cooperative effects also in vivo on tumor growth inhibition of prostate cancer xenografts in nude mice with a significant survival increase. These effects were paralleled by enhanced apoptosis and inactivation of both Erk and Akt. In conclusions, the combination between ZOL and FTI leads to enhanced anti-tumor activity in human prostate adenocarcinoma cells likely through a more efficacious inhibition of ras-dependent survival pathways and consequent bcl-related proteins-dependent apoptosis.
Collapse
Affiliation(s)
- Michele Caraglia
- Experimental Pharmacology Unit, National Cancer Institute of Naples "Fondazione G. Pascale," Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Equbal T, Silakari O, Ravikumar M. Exploring three-dimensional quantitative structural activity relationship (3D-QSAR) analysis of SCH 66336 (Sarasar) analogues of farnesyltransferase inhibitors. Eur J Med Chem 2007; 43:204-9. [PMID: 17442459 DOI: 10.1016/j.ejmech.2007.02.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Revised: 02/03/2007] [Accepted: 02/13/2007] [Indexed: 11/18/2022]
Abstract
3D-QSAR analysis of a set of 37 analogues of SCH 66336 (Sarasar) was performed by most widely used computational tool, molecular field analysis (MFA) to investigate the substitutional requirements for the favorable receptor-drug interaction and to derive a predictive model that may be used for the designing of a novel farnesyltransferase inhibitors (FTIs). Regression analysis was carried out using genetic partial least squares (G/PLS) method. A highly predictive and statistically significant model was generated. The predictive ability of the model developed was assessed using a test set of six compounds (r(2)(pred) as high as 0.791). The analyzed MFA model has demonstrated a good fit, having r(2) value of 0.967 and cross-validated coefficient r(2)(cv) value as 0.921.
Collapse
Affiliation(s)
- Tabish Equbal
- Department of Pharmaceutical Science and Drug Research, Punjabi University, Patiala, Punjab 147-002, India
| | | | | |
Collapse
|
49
|
Iwasaki S, Omura S. Search for Protein Farnesyltransferase Inhibitors of Microbial Origin: Our Strategy and Results as well as the Results Obtained by Other Groups. J Antibiot (Tokyo) 2007; 60:1-12. [PMID: 17390583 DOI: 10.1038/ja.2007.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mutant ras oncogenes are associated with various human tumors, being found in approximately 25% of all human cancers. Since its identification, the enzyme Ras protein farnesyltransferase (PFTase), which catalyzes the initial step of Ras-processing, has been viewed as a most promising target for cancer therapy. Consequently, a number of synthetic and natural small molecules have been searched and developed according to this concept during the 1990s. Among these, microbial metabolites have provided diverse structural classes of compounds which exhibit PFTase inhibitory activity. This article reviews our work on PFTase inhibitors originating from microbial metabolites, and the results of similar works carried out by several other research groups.
Collapse
Affiliation(s)
- Shigeo Iwasaki
- The Kitasato Institute, 5-9-1, Shirokane, Minatoku, Tokyo 108-8641, Japan
| | | |
Collapse
|
50
|
Eastman RT, White J, Hucke O, Yokoyama K, Verlinde CLMJ, Hast MA, Beese LS, Gelb MH, Rathod PK, Van Voorhis WC. Resistance mutations at the lipid substrate binding site of Plasmodium falciparum protein farnesyltransferase. Mol Biochem Parasitol 2006; 152:66-71. [PMID: 17208314 PMCID: PMC2875941 DOI: 10.1016/j.molbiopara.2006.11.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Revised: 11/30/2006] [Accepted: 11/30/2006] [Indexed: 11/23/2022]
Abstract
The post-translational farnesylation of proteins serves to anchor a subset of intracellular proteins to membranes in eukaryotic organisms and also promotes protein-protein interactions. This enzymatic reaction is carried out by protein farnesyltransferase (PFT), which catalyzes the transfer of a 15-carbon isoprenoid lipid unit, a farnesyl group, from farnesyl pyrophosphate to the C-termini of proteins containing a CaaX motif. Inhibition of PFT is lethal to the pathogenic protozoa Plasmodium falciparum. Previously, we have shown that parasites resistant to a tetrahydroquinoline (THQ)-based PFT inhibitor BMS-388891 have mutations leading to amino acid substitutions in PFT that map to the peptide substrate binding domain. We now report the selection of parasites resistant to another THQ PFT inhibitor BMS-339941. In whole cell assays sensitivity to BMS-339941 was reduced by 33-fold in a resistant clone, and biochemical analysis demonstrated a corresponding 33-fold increase in the BMS-339941 K(i) for the mutant PFT enzyme. More detailed kinetic analysis revealed that the mutant enzyme required higher concentration of peptide and farnesyl pyrophosphate substrates for optimum catalysis. Unlike previously characterized parasites resistant to BMS-388891, the resistant parasites have a mutation which is predicted to be in a distinct location of the enzymatic pocket, near the farnesyl pyrophosphate binding pocket. This is the first description of a mutation from any species affecting the farnesyl pyrophosphate binding pocket with reduced efficacy of PFT inhibitors. These data provide further support that PFT is the target of THQ inhibitors in P. falciparum and suggest that PFT inhibitors should be combined with other antimalarial agents to minimize the development of resistant parasites.
Collapse
Affiliation(s)
| | - John White
- Department of Pathobiology, University of Washington, Seattle, WA, USA
| | - Oliver Hucke
- Biochemistry, University of Washington, Seattle, WA, USA
| | | | | | - Michael A. Hast
- Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Lorena S. Beese
- Biochemistry, Duke University Medical Center, Durham, NC, USA
| | | | - Pradipsinh K. Rathod
- Department of Pathobiology, University of Washington, Seattle, WA, USA
- Chemistry, University of Washington, Seattle, WA, USA
| | - Wesley C. Van Voorhis
- Department of Pathobiology, University of Washington, Seattle, WA, USA
- Medicine, University of Washington, Seattle, WA, USA
- Corresponding author: Wesley C. Van Voorhis, Dept. of Medicine, University of Washington, Box 357185, 1959 N.E. Pacific, Seattle, WA 98195-7185, Tel.: + 1-206-543-2447; fax: + 1-206-685-8681, E. mail addresses:
| |
Collapse
|