1
|
Beygi F, Mostoufi A, Mojaddami A. Novel Hydrazone Derivatives of 3-Bromopyruvate: Synthesis, Evaluation of the Cytotoxic Effects, Molecular Docking and ADME Studies. Chem Biodivers 2022; 19:e202100754. [PMID: 35427437 DOI: 10.1002/cbdv.202100754] [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: 09/16/2021] [Accepted: 04/14/2022] [Indexed: 11/09/2022]
Abstract
A series of 3-bromopyruvate (3-BP) derivatives were synthesized to develop new potent anticancer agents. The chemical structures of the compounds were characterized using FT-IR, 1 H-, 13 C-NMR spectroscopy, and elemental analysis (CHN). Their cytotoxic activities were investigated against four cancer cell lines, including colon (SW1116), breast (MDA-MB-231), lung (A549), and liver (HepG2) cancer cell lines. Among the synthesized compounds, 3b showed promising cytotoxic activity compared to 3-BP, with IC50 values of 16.3 μM, 19.1 μM, 27.8 μM, and 14.5 μM against A549, MDA-MB-231, SW1116 and, HepG2 cell lines, respectively. Furthermore, the effect of these compounds on MCF-10A (a normal breast cell lines) was investigated to determine their selectivity between tumorigenic and non-tumorigenic cells. Since the 3-BP inhibits hexokinase II (HK II), molecular docking of 3-BP derivatives was carried out using AutoDock 4.2. The binding energies of these derivatives were greater than 3-BP, indicating that they had a higher affinity for HK II. For validation of docking, a 40 ns MD simulation was performed. SwissADME was used to predict pharmacokinetics, drug-likeness, and ADME parameters of the screened compounds. The results demonstrated that these derivatives are suitable candidates for developing orally potent HK II inhibitors.
Collapse
Affiliation(s)
- Farzaneh Beygi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Azar Mostoufi
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ayyub Mojaddami
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
2
|
Huang R, Zhou PK. DNA damage repair: historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduct Target Ther 2021; 6:254. [PMID: 34238917 PMCID: PMC8266832 DOI: 10.1038/s41392-021-00648-7] [Citation(s) in RCA: 390] [Impact Index Per Article: 97.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 04/28/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Genomic instability is the hallmark of various cancers with the increasing accumulation of DNA damage. The application of radiotherapy and chemotherapy in cancer treatment is typically based on this property of cancers. However, the adverse effects including normal tissues injury are also accompanied by the radiotherapy and chemotherapy. Targeted cancer therapy has the potential to suppress cancer cells' DNA damage response through tailoring therapy to cancer patients lacking specific DNA damage response functions. Obviously, understanding the broader role of DNA damage repair in cancers has became a basic and attractive strategy for targeted cancer therapy, in particular, raising novel hypothesis or theory in this field on the basis of previous scientists' findings would be important for future promising druggable emerging targets. In this review, we first illustrate the timeline steps for the understanding the roles of DNA damage repair in the promotion of cancer and cancer therapy developed, then we summarize the mechanisms regarding DNA damage repair associated with targeted cancer therapy, highlighting the specific proteins behind targeting DNA damage repair that initiate functioning abnormally duo to extrinsic harm by environmental DNA damage factors, also, the DNA damage baseline drift leads to the harmful intrinsic targeted cancer therapy. In addition, clinical therapeutic drugs for DNA damage and repair including therapeutic effects, as well as the strategy and scheme of relative clinical trials were intensive discussed. Based on this background, we suggest two hypotheses, namely "environmental gear selection" to describe DNA damage repair pathway evolution, and "DNA damage baseline drift", which may play a magnified role in mediating repair during cancer treatment. This two new hypothesis would shed new light on targeted cancer therapy, provide a much better or more comprehensive holistic view and also promote the development of new research direction and new overcoming strategies for patients.
Collapse
Affiliation(s)
- Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Ping-Kun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, China.
| |
Collapse
|
3
|
Personalized Medicine: The Path to New Medicine. Per Med 2016. [DOI: 10.1007/978-3-319-39349-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
4
|
Morgan RJ, Synold TW, Longmate JA, Quinn DI, Gandara D, Lenz HJ, Ruel C, Xi B, Lewis MD, Colevas AD, Doroshow J, Newman EM. Pharmacodynamics (PD) and pharmacokinetics (PK) of E7389 (eribulin, halichondrin B analog) during a phase I trial in patients with advanced solid tumors: a California Cancer Consortium trial. Cancer Chemother Pharmacol 2015; 76:897-907. [PMID: 26362045 DOI: 10.1007/s00280-015-2868-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 09/02/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND The California Cancer Consortium completed a phase I trial of E7389 (eribulin mesylate), an analog of the marine natural product halichondrin B. This trial was to determine the pharmacodynamics, pharmacokinetics, and MTD of E7389 administered by bolus injection weekly for 3 weeks out of four. METHODS This trial included a rapid titration design. Real-time pharmacokinetics were utilized to guide dose escalation. Initially, single-patient cohorts were enrolled with intra- and inter-patient dose doubling. The second phase was a standard 3 + 3 dose escalation schedule. At the MTD, a cohort of patients was enrolled for target validation studies (separate manuscript). The starting dose was 0.125 mg/m(2), and doses were doubled within and between patients in the first phase. Blood and urine sampling for E7389 pharmacokinetics was performed on doses 1 and 3 of cycle 1. Levels were determined using a LC/MS/MS assay. RESULTS Forty patients were entered. Thirty-eight were evaluable for toxicity and 35 for response. The rapid escalation ended with a grade 3 elevation of alkaline phosphatase at 0.5 mg/m(2)/week. The second phase ended at 2.0 mg/m(2)/week with dose-limiting toxicities of grades 3 and 4 febrile neutropenia. Other toxicities included hypoglycemia, hypophosphatemia, and fatigue. The MTD was 1.4 mg/m(2)/week. Responses included four partial responses (lung cancer [2], urothelial [1], and melanoma [1]). CONCLUSIONS E7389 was well tolerated in this trial with the major toxicity being myelosuppression. PD shows that E7389 induces significant morphologic changes (bundle formation) in the microtubules of peripheral blood mononuclear cells and tumor cells in vivo. The data suggest that lower intra-tumoral levels of β-tubulin III or higher intra-tumoral levels of MAP4 may correlate with response to E7389, while lower intra-tumoral levels of stathmin may be associated with progression. PK data reveal that E7389 exhibits a tri-exponential elimination from the plasma of patients receiving a rapid i.v. infusion. At sub-toxic doses, plasma concentrations of E7389 are maintained well above the levels required for activity in vitro for >72 h.
Collapse
Affiliation(s)
- Robert J Morgan
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd., Duarte, CA, 91010, USA.
| | - Timothy W Synold
- Department of Cancer Biology, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Jeffrey A Longmate
- Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - David I Quinn
- Division of Medical Oncology, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA, 90033, USA
| | - David Gandara
- Division of Medical Oncology, University of California, Davis Cancer Center, Sacramento, CA, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA, 90033, USA
| | - Christopher Ruel
- Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Bixin Xi
- Department of Cancer Biology, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Michael D Lewis
- Eisai Research Institute, Andover, MA, 01810, USA.,Edward P. Evans Foundation, Casanova, VA, 20139, USA
| | - A Dimitrios Colevas
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, 20892, USA.,Stanford Cancer Center, Stanford, CA, 94305, USA
| | - James Doroshow
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd., Duarte, CA, 91010, USA.,Division of Cancer Treatment and Diagnosis6, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Edward M Newman
- Department of Cancer Biology, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| |
Collapse
|
5
|
Liew K, Yong PVC, Navaratnam V, Lim YM, Ho ASH. Differential proteomic analysis on the effects of 2-methoxy-1,4-naphthoquinone towards MDA-MB-231 cell line. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:517-527. [PMID: 25981917 DOI: 10.1016/j.phymed.2015.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/31/2015] [Accepted: 03/05/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND We have previously reported the anti-metastatic effects of 2-methoxy-1,4-naphthoquinone (MNQ) against MDA-MB-231 cell line. PURPOSE To investigate the molecular mechanism underlying the anti-metastatic effects of MNQ towards MDA-MB-231 cell line via the comparative proteomic approach. STUDY DESIGN/METHODS Differentially expressed proteins in MNQ-treated MDA-MB-231 cells were identified by using two-dimensional gel electrophoresis coupled with tandem mass spectrometry. Proteins and signalling pathways associated with the identified MNQ-altered proteins were studied by using Western blotting. RESULTS Significant modulation of MDA-MB-231 cell proteome was observed upon treatment with MNQ in which the expressions of 19 proteins were found to be downregulated whereas another eight were upregulated (>1.5 fold, p < 0.05). The altered proteins were mainly related to cytoskeletal functions and regulations, mRNA processing, protein modifications and oxidative stress response. Notably, two of the downregulated proteins, protein S100-A4 (S100A4) and laminin-binding protein (RPSA) are known to play key roles in driving metastasis and were verified using Western blotting. Further investigation using Western blotting also revealed that MNQ decreased the activations of pro-metastatic ERK1/2 and NF-κB signalling pathways. Moreover, MNQ was shown to stimulate the expression of the metastatic suppressor, E-cadherin. CONCLUSION This study reports a proposed mechanism by which MNQ exerts its anti-metastatic effects against MDA-MB-231 cell line. The findings from this study offer new insights on the potential of MNQ to be developed as a novel anti-metastatic agent.
Collapse
Affiliation(s)
- Kitson Liew
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Phelim Voon Chen Yong
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Visweswaran Navaratnam
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Yang Mooi Lim
- Department of Pre-Clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Lot PT21144, Jalan Sungai Long, Bandar Sungai Long, 43000 Kajang, Selangor Darul Ehsan, Malaysia.
| | - Anthony Siong Hock Ho
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
6
|
Markova-Car EP, Jurišić D, Ilić N, Kraljević Pavelić S. Running for time: circadian rhythms and melanoma. Tumour Biol 2014; 35:8359-68. [PMID: 24729125 DOI: 10.1007/s13277-014-1904-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/27/2014] [Indexed: 12/22/2022] Open
Abstract
Circadian timing system includes an input pathway transmitting environmental signals to a core oscillator that generates circadian signals responsible for the peripheral physiological or behavioural events. Circadian 24-h rhythms regulate diverse physiologic processes. Deregulation of these rhythms is associated with a number of pathogenic conditions including depression, diabetes, metabolic syndrome and cancer. Melanoma is a less common type of skin cancer yet more aggressive often with a lethal ending. However, little is known about circadian control in melanoma and exact functional associations between core clock genes and development of melanoma skin cancer. This paper, therefore, comprehensively analyses current literature data on the involvement of circadian clock components in melanoma development. In particular, the role of circadian rhythm deregulation is discussed in the context of DNA repair mechanisms and influence of UV radiation and artificial light exposure on cancer development. The role of arylalkylamine N-acetyltransferase (AANAT) enzyme and impact of melatonin, as a major output factor of circadian rhythm, and its protective role in melanoma are discussed in details. We hypothesise that further understanding of clock genes' involvement and circadian regulation might foster discoveries in the field of melanoma diagnostics and treatment.
Collapse
Affiliation(s)
- Elitza P Markova-Car
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000, Rijeka, Croatia,
| | | | | | | |
Collapse
|
7
|
Nagarapu L, Paparaju V, Satyender A, Bantu R. Total synthesis of (+)-varitriol via a symmetrical furanose diol as the key intermediate. Tetrahedron Lett 2011. [DOI: 10.1016/j.tetlet.2011.10.076] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
8
|
Patel GS, Kiuchi T, Lawler K, Ofo E, Fruhwirth GO, Kelleher M, Shamil E, Zhang R, Selvin PR, Santis G, Spicer J, Woodman N, Gillett CE, Barber PR, Vojnovic B, Kéri G, Schaeffter T, Goh V, O'Doherty MJ, Ellis PA, Ng T. The challenges of integrating molecular imaging into the optimization of cancer therapy. Integr Biol (Camb) 2011; 3:603-31. [PMID: 21541433 DOI: 10.1039/c0ib00131g] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We review novel, in vivo and tissue-based imaging technologies that monitor and optimize cancer therapeutics. Recent advances in cancer treatment centre around the development of targeted therapies and personalisation of treatment regimes to individual tumour characteristics. However, clinical outcomes have not improved as expected. Further development of the use of molecular imaging to predict or assess treatment response must address spatial heterogeneity of cancer within the body. A combination of different imaging modalities should be used to relate the effect of the drug to dosing regimen or effective drug concentration at the local site of action. Molecular imaging provides a functional and dynamic read-out of cancer therapeutics, from nanometre to whole body scale. At the whole body scale, an increase in the sensitivity and specificity of the imaging probe is required to localise (micro)metastatic foci and/or residual disease that are currently below the limit of detection. The use of image-guided endoscopic biopsy can produce tumour cells or tissues for nanoscopic analysis in a relatively patient-compliant manner, thereby linking clinical imaging to a more precise assessment of molecular mechanisms. This multimodality imaging approach (in combination with genetics/genomic information) could be used to bridge the gap between our knowledge of mechanisms underlying the processes of metastasis, tumour dormancy and routine clinical practice. Treatment regimes could therefore be individually tailored both at diagnosis and throughout treatment, through monitoring of drug pharmacodynamics providing an early read-out of response or resistance.
Collapse
Affiliation(s)
- G S Patel
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, King's College London, Guy's Medical School Campus, London, SE1 1UL, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Milli A, Perego P, Beretta GL, Corvo A, Righetti PG, Carenini N, Corna E, Zuco V, Zunino F, Cecconi D. Proteomic Analysis of Cellular Response to Novel Proapoptotic Agents Related to Atypical Retinoids in Human IGROV-1 Ovarian Carcinoma Cells. J Proteome Res 2010; 10:1191-207. [DOI: 10.1021/pr100963n] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Alberto Milli
- Dipartimento di Biotecnologie, Laboratorio di Proteomica e Spettrometria di Massa, University of Verona, Strada le Grazie 15, 37134, Verona, Italy
| | - Paola Perego
- Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, 20133, Milano, Italy
| | - Giovanni L. Beretta
- Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, 20133, Milano, Italy
| | - Alice Corvo
- Dipartimento di Biotecnologie, Laboratorio di Proteomica e Spettrometria di Massa, University of Verona, Strada le Grazie 15, 37134, Verona, Italy
| | - Pier Giorgio Righetti
- Dipartimento di Chimica, Materiali e Ingegneria Chimica “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131, Milano, Italy
| | - Nives Carenini
- Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, 20133, Milano, Italy
| | - Elisabetta Corna
- Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, 20133, Milano, Italy
| | - Valentina Zuco
- Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, 20133, Milano, Italy
| | - Franco Zunino
- Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, 20133, Milano, Italy
| | - Daniela Cecconi
- Dipartimento di Biotecnologie, Laboratorio di Proteomica e Spettrometria di Massa, University of Verona, Strada le Grazie 15, 37134, Verona, Italy
| |
Collapse
|
10
|
Cheng KW, Wong CC, Wang M, He QY, Chen F. Identification and characterization of molecular targets of natural products by mass spectrometry. MASS SPECTROMETRY REVIEWS 2010; 29:126-155. [PMID: 19319922 DOI: 10.1002/mas.20235] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Natural products, and their derivatives and mimics, have contributed to the development of important therapeutics to combat diseases such as infections and cancers over the past decades. The value of natural products to modern drug discovery is still considerable. However, its development is hampered by a lack of a mechanistic understanding of their molecular action, as opposed to the emerging molecule-targeted therapeutics that are tailored to a specific protein target(s). Recent advances in the mass spectrometry-based proteomic approaches have the potential to offer unprecedented insights into the molecular action of natural products. Chemical proteomics is established as an invaluable tool for the identification of protein targets of natural products. Small-molecule affinity selection combined with mass spectrometry is a successful strategy to "fish" cellular targets from the entire proteome. Mass spectrometry-based profiling of protein expression is also routinely employed to elucidate molecular pathways involved in the therapeutic and possible toxicological responses upon treatment with natural products. In addition, mass spectrometry is increasingly utilized to probe structural aspects of natural products-protein interactions. Limited proteolysis, photoaffinity labeling, and hydrogen/deuterium exchange in conjunction with mass spectrometry are sensitive and high-throughput strategies that provide low-resolution structural information of non-covalent natural product-protein complexes. In this review, we provide an overview on the applications of mass spectrometry-based techniques in the identification and characterization of natural product-protein interactions, and we describe how these applications might revolutionize natural product-based drug discovery.
Collapse
Affiliation(s)
- Ka-Wing Cheng
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | | | | | | | | |
Collapse
|
11
|
Li RX, Zeng R. Dynamic proteomics for investigating the response of individual cancer cells under drug action. Expert Rev Proteomics 2009; 6:19-21. [PMID: 19210123 DOI: 10.1586/14789450.6.1.19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Evaluation of: Cohen AA, Geva-Zatorsky N, Eden E et al. Dynamic proteomics of individual cancer cells in response to a drug. Science 322(5907), 1511-1516 (2008). One of the greatest challenges in cancer chemotherapy is that seemingly identical cancer cells can respond differently to drug treatment. The pioneering work reported by Cohen and colleagues moves one step closer to solving this challenge. They develop a dynamic proteomics approach that utilizes fluorescent markers and a time-lapse microscope to detect the fluctuating locations and levels of approximately 1000 proteins in individual cancer cells at high temporal resolution. After adminstration of the cancer drug camptothecin, certain proteins display similar spatiotemporal distribution patterns in individual cells; for example, the drug target topoisomerase-1 shows a rapid decrease in protein level and in nuclear location. However, two particular proteins demonstrate cell-cell variability in their behavior corresponding to cell fate, which may help to explain drug resistance. This method offers an effective way to investigate drug mechanisms in individual cells.
Collapse
Affiliation(s)
- Rong-Xia Li
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | | |
Collapse
|
12
|
Rajic Z, Butula I, Zorc B, Kraljevic Pavelic S, Hock K, Pavelic K, Naesens L, De Clercq E, Balzarini J, Przyborowska M, Ossowski T, Mintas M. Cytostatic and Antiviral Activity Evaluations of Hydroxamic Derivatives of Some Non-steroidal Anti-inflammatory Drugs. Chem Biol Drug Des 2009; 73:328-38. [DOI: 10.1111/j.1747-0285.2009.00774.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
13
|
Kraljevic Pavelic S, Sedic M, Hock K, Vucinic S, Jurisic D, Gehrig P, Scott M, Schlapbach R, Cacev T, Kapitanovic S, Pavelic K. An integrated proteomics approach for studying the molecular pathogenesis of Dupuytren's disease. J Pathol 2009; 217:524-33. [DOI: 10.1002/path.2483] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
14
|
Bosnjak H, Pavelic K, Kraljevic Pavelic S. Towards preventive medicine. High-throughput methods from molecular biology are about to change daily clinical practice. EMBO Rep 2008; 9:1056-60. [PMID: 18846103 DOI: 10.1038/embor.2008.198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Hrvojka Bosnjak
- Rudjer Boskovic Institute, Division of Molecular Medicine, in Zagreb, Croatia.
| | | | | |
Collapse
|
15
|
Sedic M, Poznic M, Gehrig P, Scott M, Schlapbach R, Hranjec M, Karminski-Zamola G, Pavelic K, Kraljevic Pavelic S. Differential antiproliferative mechanisms of novel derivative of benzimidazo[1,2-alpha]quinoline in colon cancer cells depending on their p53 status. Mol Cancer Ther 2008; 7:2121-32. [PMID: 18645022 DOI: 10.1158/1535-7163.mct-07-2261] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the present article, we describe a mechanistic study of a novel derivative of N-amidino-substituted benzimidazo[1,2-alpha]quinoline in two human colorectal cancer cell lines differing in p53 gene status. We used a proteomic approach based on two-dimensional gel electrophoresis coupled with mass spectrometry to complement the results obtained by common molecular biology methods for analyzing cell proliferation, cell cycle, and apoptosis. Tested quinoline derivative inhibited colon cancer cell growth, whereby p53 gene status seemed to be critical for its differential response patterns. DNA damage and oxidative stress are likely to be the common triggers of molecular events underlying its antiproliferative effects. In HCT 116 (wild-type p53), this compound induced a p53-dependent response resulting in accumulation of the G(1)- and S-phase cells and induction of apoptosis via both caspase-3-dependent and caspase-independent pathways. Quinoline derivative triggered transient, p53-independent G(2)-M arrest in mutant p53 cells (SW620) and succeeding mitotic transition, whereby these cells underwent cell death probably due to aberrant mitosis (mitotic catastrophe). Proteomic approach used in this study proved to be a valuable tool for investigating cancer cell response to newly synthesized compound, as it specifically unraveled some molecular changes that would not have been otherwise detected (e.g., up-regulation of the p53-dependent chemotherapeutic response marker maspin in HCT 116 and impairment in ribosome biogenesis in SW620). Finally, antiproliferative effects of tested quinoline derivative on SW620 cells strongly support its possible role as an antimetastatic agent and encourage further in vivo studies on the chemotherapeutic potential of this compound against colorectal carcinoma.
Collapse
Affiliation(s)
- Mirela Sedic
- Division of Molecular Medicine, Ruder Boskovic Institute, Bijenicka cesta 54, 10000 Zagreb, Croatia.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Gazivoda T, Raić-Malić S, Krištafor V, Makuc D, Plavec J, Bratulić S, Kraljević-Pavelić S, Pavelić K, Naesens L, Andrei G, Snoeck R, Balzarini J, Mintas M. Synthesis, cytostatic and anti-HIV evaluations of the new unsaturated acyclic C-5 pyrimidine nucleoside analogues. Bioorg Med Chem 2008; 16:5624-34. [PMID: 18424155 PMCID: PMC7127491 DOI: 10.1016/j.bmc.2008.03.074] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2008] [Revised: 03/20/2008] [Accepted: 03/28/2008] [Indexed: 11/26/2022]
Abstract
A series of the novel C-5 alkynyl pyrimidine nucleoside analogues (1-14) in which the sugar moiety was replaced by the conformationally restricted Z- and E-2-butenyl spacer between the phthalimido and pyrimidine ring were synthesized by using Sonogashira cross-coupling reaction. Cytostatic activity evaluation of the novel compounds showed that E-isomers exhibited, in general, better cytostatic activities than the corresponding Z-isomers. E-isomer 14 exhibited the best cytostatic effect against all evaluated malignant cell lines, particularly against hepatocellular carcinoma (Hep G2, IC(50)=4.3microM). However, this compound was also cytotoxic to human normal fibroblasts (WI 38). Its Z-isomer 7 showed highly specific antiproliferative activity against Hep G2 (IC(50)=18microM) and no cytotoxicity to WI 38. Moreover, compounds 3, 4 and 14 expressed some marginal inhibitory activity against HIV-1 and HIV-2.
Collapse
Affiliation(s)
- Tatjana Gazivoda
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, HR-10000 Zagreb, Croatia
| | - Silvana Raić-Malić
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, HR-10000 Zagreb, Croatia
| | - Vedran Krištafor
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, HR-10000 Zagreb, Croatia
| | - Damjan Makuc
- Slovenian NMR Centre, National Institute of Chemistry, Hajdrihova 19, POB 660, SL-1001 Ljubljana, Slovenia
| | - Janez Plavec
- Slovenian NMR Centre, National Institute of Chemistry, Hajdrihova 19, POB 660, SL-1001 Ljubljana, Slovenia
| | - Siniša Bratulić
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička 54, POB 1016, HR-10001 Zagreb, Croatia
| | - Sandra Kraljević-Pavelić
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička 54, POB 1016, HR-10001 Zagreb, Croatia
| | - Krešimir Pavelić
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička 54, POB 1016, HR-10001 Zagreb, Croatia
| | - Lieve Naesens
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | - Graciela Andrei
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | - Robert Snoeck
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | - Jan Balzarini
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | - Mladen Mintas
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, HR-10000 Zagreb, Croatia
| |
Collapse
|
17
|
Azevedo LG, Muccillo-Baisch AL, Filgueira DDMVB, Boyle RT, Ramos DF, Soares AD, Lerner C, Silva PA, Trindade GS. Comparative cytotoxic and anti-tuberculosis activity of Aplysina caissara marine sponge crude extracts. Comp Biochem Physiol C Toxicol Pharmacol 2008; 147:36-42. [PMID: 17826358 DOI: 10.1016/j.cbpc.2007.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Revised: 07/23/2007] [Accepted: 07/24/2007] [Indexed: 11/16/2022]
Abstract
Three crude extracts of Aplysina caissara, a marine sponge endemic to Brazil, were tested against a hepatoma cell line and Mycobacterium tuberculosis. The results demonstrate that all extracts are toxic and capable of inhibiting cellular growth. Additionally, the extracts produced morphological aberrations and inhibited cell attachment to culture substrates. These effects were dose/time dependent. Our results also suggest that reactive oxygen species (ROS) production is not involved in the cytotoxic processes levied by the extracts employed in this study and that active metabolites are likely to be present in the polar fractions of the crude extracts. Finally, our results indicate that all three extracts exhibit a moderate anti-tuberculosis capacity, and that the removal of an extract's lipid fraction appears to diminish this activity.
Collapse
Affiliation(s)
- Luciana G Azevedo
- Departamento de Ciências Fisiológicas, Fundação Universidade Federal do Rio Grande (FURG), Rio Grande, (96201-900), Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Oncoproteomics is the application of proteomics technologies in oncology. Functional proteomics is a promising technique for the rational identification of biomarkers and novel therapeutic targets for cancers. Recent progress in proteomics has opened new avenues for tumor-associated biomarker discovery. With the advent of new and improved proteomics technologies, such as the development of quantitative proteomic methods, high-resolution, -speed and -sensitivity mass spectrometry and protein arrays, as well as advanced bioinformatics for data handling and interpretation, it is now possible to discover biomarkers that can reliably and accurately predict outcomes during cancer management and treatment. However, there are several difficulties in the study of proteins/peptides that are not inherent in the study of nucleic acids. New challenges arise in large-scale proteomic profiling when dealing with complex biological mixtures. Nevertheless, oncoproteomics offers great promise for unveiling the complex molecular events of tumorigenesis, as well as those that control clinically important tumor behaviors, such as metastasis, invasion and resistance to therapy. In this review, the development and advancement of oncoproteomics technologies for cancer research in recent years are expounded.
Collapse
Affiliation(s)
- William C S Cho
- Queen Elizabeth Hospital, Department of Clinical Oncology, Kowloon, Hong Kong SAR, PR China.
| | | |
Collapse
|
19
|
Kraljevic Pavelic S, Saban N. Evolving ‘-omics’ technologies in the drug development process. Expert Opin Drug Discov 2007; 2:431-6. [DOI: 10.1517/17460441.2.4.431] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|