1
|
Almutairi S, Kalloush HM, Manoon NA, Bardaweel SK. Matrix Metalloproteinases Inhibitors in Cancer Treatment: An Updated Review (2013-2023). Molecules 2023; 28:5567. [PMID: 37513440 PMCID: PMC10384300 DOI: 10.3390/molecules28145567] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/09/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are identifiable members of proteolytic enzymes that can degrade a wide range of proteins in the extracellular matrix (ECM). MMPs can be categorized into six groups based on their substrate specificity and structural differences: collagenases, gelatinases, stromelysins, matrilysins, metalloelastase, and membrane-type MMPs. MMPs have been linked to a wide variety of biological processes, such as cell transformation and carcinogenesis. Over time, MMPs have been evaluated for their role in cancer progression, migration, and metastasis. Accordingly, various MMPs have become attractive therapeutic targets for anticancer drug development. The first generations of broad-spectrum MMP inhibitors displayed effective inhibitory activities but failed in clinical trials due to poor selectivity. Thanks to the evolution of X-ray crystallography, NMR analysis, and homology modeling studies, it has been possible to characterize the active sites of various MMPs and, consequently, to develop more selective, second-generation MMP inhibitors. In this review, we summarize the computational and synthesis approaches used in the development of MMP inhibitors and their evaluation as potential anticancer agents.
Collapse
Affiliation(s)
- Shriefa Almutairi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
| | - Hanin Moh'd Kalloush
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Nour A Manoon
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
| | - Sanaa K Bardaweel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
| |
Collapse
|
2
|
Rahman F, Wushur I, Malla N, Åstrand OAH, Rongved P, Winberg JO, Sylte I. Zinc-Chelating Compounds as Inhibitors of Human and Bacterial Zinc Metalloproteases. Molecules 2021; 27:molecules27010056. [PMID: 35011288 PMCID: PMC8746695 DOI: 10.3390/molecules27010056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
Inhibition of bacterial virulence is believed to be a new treatment option for bacterial infections. In the present study, we tested dipicolylamine (DPA), tripicolylamine (TPA), tris pyridine ethylene diamine (TPED), pyridine and thiophene derivatives as putative inhibitors of the bacterial virulence factors thermolysin (TLN), pseudolysin (PLN) and aureolysin (ALN) and the human zinc metalloproteases, matrix metalloprotease-9 (MMP-9) and matrix metalloprotease-14 (MMP-14). These compounds have nitrogen or sulfur as putative donor atoms for zinc chelation. In general, the compounds showed stronger inhibition of MMP-14 and PLN than of the other enzymes, with Ki values in the lower μM range. Except for DPA, none of the compounds showed significantly stronger inhibition of the virulence factors than of the human zinc metalloproteases. TPA and Zn230 were the only compounds that inhibited all five zinc metalloproteinases with a Ki value in the lower μM range. The thiophene compounds gave weak or no inhibition. Docking indicated that some of the compounds coordinated zinc by one oxygen atom from a hydroxyl or carbonyl group, or by oxygen atoms both from a hydroxyl group and a carbonyl group, and not by pyridine nitrogen as in DPA and TPA.
Collapse
Affiliation(s)
- Fatema Rahman
- Molecular Pharmacology and Toxicology, Department of Medical Biology, Faculty of Health Sciences, UiT—The Arctic University of Norway, NO-9037 Tromsø, Norway; (F.R.); (I.W.); (N.M.); (J.-O.W.)
| | - Imin Wushur
- Molecular Pharmacology and Toxicology, Department of Medical Biology, Faculty of Health Sciences, UiT—The Arctic University of Norway, NO-9037 Tromsø, Norway; (F.R.); (I.W.); (N.M.); (J.-O.W.)
| | - Nabin Malla
- Molecular Pharmacology and Toxicology, Department of Medical Biology, Faculty of Health Sciences, UiT—The Arctic University of Norway, NO-9037 Tromsø, Norway; (F.R.); (I.W.); (N.M.); (J.-O.W.)
| | - Ove Alexander Høgmoen Åstrand
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, NO-0316 Oslo, Norway; (O.A.H.Å.); (P.R.)
| | - Pål Rongved
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, NO-0316 Oslo, Norway; (O.A.H.Å.); (P.R.)
| | - Jan-Olof Winberg
- Molecular Pharmacology and Toxicology, Department of Medical Biology, Faculty of Health Sciences, UiT—The Arctic University of Norway, NO-9037 Tromsø, Norway; (F.R.); (I.W.); (N.M.); (J.-O.W.)
| | - Ingebrigt Sylte
- Molecular Pharmacology and Toxicology, Department of Medical Biology, Faculty of Health Sciences, UiT—The Arctic University of Norway, NO-9037 Tromsø, Norway; (F.R.); (I.W.); (N.M.); (J.-O.W.)
- Correspondence: ; Tel.: +47-7764-4705
| |
Collapse
|
3
|
Ayoup MS, Abu-Serie MM, Awad LF, Teleb M, Ragab HM, Amer A. Halting colorectal cancer metastasis via novel dual nanomolar MMP-9/MAO-A quinoxaline-based inhibitors; design, synthesis, and evaluation. Eur J Med Chem 2021; 222:113558. [PMID: 34116327 DOI: 10.1016/j.ejmech.2021.113558] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 12/25/2022]
Abstract
Matrix metalloproteinase-9 (MMP-9) and monoamine oxidase-A (MAO-A) are central signaling nodes in CRC and promotors of distant metastasis associated with high mortality rates. Novel series of quinoxaline-based dual MMP-9/MAO-A inhibitors were synthesized to suppress CRC progression. The design rationale combines the thematic pharmacophoric features of MMP-9 and MAO-A inhibitors in hybrid scaffolds. All derivatives were initially screened via MTT assay for cytotoxic effects on normal colonocytes to assess their safety profiles, then evaluated for their anticancer potential on HCT116 cells overexpressing MMP-9 and MAO-A. The most promising derivatives 8, 16, 17, 19, and 28 exhibited single digit nanomolar IC50 against HCT116 cells within their safe doses (EC100) on normal colonocytes. They suppressed HCT116 cell migration by 73.32, 61.29, 21.27, 28.82, and 27.48%, respectively as detected by wound healing assay. Enzymatic assays revealed that the selected derivatives were superior to the reference MMP-9 and MAO-A inhibitors (quercetin and clorgyline, respectively). The nanomolar dual MMP-9/MAO-A inhibitor 19 was identified as the most potent and balanced dual inhibitor among the evaluated series with considerable selectivity against MAO-A over MAO-B. Besides, qRT-PCR analysis was conducted to explore the hit compounds' potential to downregulate hypoxia-inducing factor (HIF-1α) in HCT116 cells being correlated with MAO-A mediated CRC migration and invasion. The five above-mentioned compounds significantly downregulated HIF-1α by more than 5 folds. Docking simulations predicted their possible binding modes with MMP-9 and MAO-A and highlighted their essential structural features. Finally, they recorded drug-like in silico physicochemical parameters and ADMET profiles.
Collapse
Affiliation(s)
- Mohammed Salah Ayoup
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria, 21321, Egypt.
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Egypt
| | - Laila F Awad
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria, 21321, Egypt
| | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Hanan M Ragab
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Adel Amer
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria, 21321, Egypt; Department of Chemistry, College of Science, Taibah University, Al-Madinah Al-Munawarah, Saudi Arabia.
| |
Collapse
|
4
|
Molecular Interactions Stabilizing the Promatrix Metalloprotease-9·Serglycin Heteromer. Int J Mol Sci 2020; 21:ijms21124205. [PMID: 32545641 PMCID: PMC7352350 DOI: 10.3390/ijms21124205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/03/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
Previous studies have shown that THP-1 cells produced an SDS-stable and reduction-sensitive complex between proMMP-9 and a chondroitin sulfate proteoglycan (CSPG) core protein. The complex could be reconstituted in vitro using purified serglycin (SG) and proMMP-9 and contained no inter-disulfide bridges. It was suggested that the complex involved both the FnII module and HPX domain of proMMP-9. The aims of the present study were to resolve the interacting regions of the molecules that form the complex and the types of interactions involved. In order to study this, we expressed and purified full-length and deletion variants of proMMP-9, purified CSPG and SG, and performed in vitro reconstitution assays, peptide arrays, protein modelling, docking, and molecular dynamics (MD) simulations. ProMMP-9 variants lacking both the FnII module and the HPX domain did not form the proMMP-9∙CSPG/SG complex. Deletion variants containing at least the FnII module or the HPX domain formed the proMMP-9∙CSPG/SG complex, as did the SG core protein without CS chains. The interacting parts covered large surface areas of both molecules and implicated dynamic and complementary ionic, hydrophobic, and hydrogen bond interactions. Hence, no short single interacting linear motifs in the two macromolecules could explain the strong SDS-stable and reduction-sensitive binding.
Collapse
|
5
|
Boudreau MW, Peh J, Hergenrother PJ. Procaspase-3 Overexpression in Cancer: A Paradoxical Observation with Therapeutic Potential. ACS Chem Biol 2019; 14:2335-2348. [PMID: 31260254 PMCID: PMC6858495 DOI: 10.1021/acschembio.9b00338] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many anticancer strategies rely on the promotion of apoptosis in cancer cells as a means to shrink tumors. Crucial for apoptotic function are executioner caspases, most notably caspase-3, that proteolyze a variety of proteins, inducing cell death. Paradoxically, overexpression of procaspase-3 (PC-3), the low-activity zymogen precursor to caspase-3, has been reported in a variety of cancer types. Until recently, this counterintuitive overexpression of a pro-apoptotic protein in cancer has been puzzling. Recent studies suggest subapoptotic caspase-3 activity may promote oncogenic transformation, a possible explanation for the enigmatic overexpression of PC-3. Herein, the overexpression of PC-3 in cancer and its mechanistic basis is reviewed; collectively, the data suggest the potential for exploitation of PC-3 overexpression with PC-3 activators as a targeted anticancer strategy.
Collapse
Affiliation(s)
- Matthew W. Boudreau
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Jessie Peh
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Paul J. Hergenrother
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| |
Collapse
|
6
|
Yurttaş L, Öztürk Ö, Cantürk Z. New Procaspase Activating Compound (PAC-1) Like Molecules as Potent Antitumoral Agents Against Lung Cancer. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180815666180926113040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
In this study, novel ortho-hydroxy N-acyl hydrazone moiety including
compounds (3a-l) were designed, based on procaspase activating compound (PAC-1) which is a
small molecule known with antitumor activity. The antitumor activity was evaluated on A549 (human
lung cancer cell line) and CCD 19Lu (human lung normal cell line).
Methods:
Twelve N'-arylidene-2-[4-(methylsulfonyl)piperazin-1-yl]acetohydrazide derivatives
(3a-l) were synthesized starting from ethyl 1-piperazinylacetate. All compounds were tested using
MTT method and Xcelligence-Real time cell analysis system (RTCA DP) to determine their antitumor
activity.
Results:
Some physicochemical properties of four active compounds were also predicted using
MolSoft, PreADMET and PROTOX software. Four of them, 3h, 3j, 3k and 3l bearing 3-hydroxy,
4-dimethylamino, 2,6-dichloro and 3,4-dichloro substituents in order exhibited selective cytotoxicity.
Conclusion:
Eligible values were obtained in the specified ranges as to be an oral/intravenous drug
considering the physicochemical calculations.
Collapse
Affiliation(s)
- Leyla Yurttaş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey
| | - Ömer Öztürk
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey
| | - Zerrin Cantürk
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey
| |
Collapse
|
7
|
Schlein LJ, Fadl-Alla B, Pondenis HC, Lezmi S, Eberhart CG, LeBlanc AK, Dickinson PJ, Hergenrother PJ, Fan TM. Immunohistochemical Characterization of Procaspase-3 Overexpression as a Druggable Target With PAC-1, a Procaspase-3 Activator, in Canine and Human Brain Cancers. Front Oncol 2019; 9:96. [PMID: 30859090 PMCID: PMC6397847 DOI: 10.3389/fonc.2019.00096] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 02/04/2019] [Indexed: 11/24/2022] Open
Abstract
Gliomas and meningiomas are the most common brain neoplasms affecting both humans and canines, and identifying druggable targets conserved across multiple brain cancer histologies and comparative species could broadly improve treatment outcomes. While satisfactory cure rates for low grade, non-invasive brain cancers are achievable with conventional therapies including surgery and radiation, the management of non-resectable or recurrent brain tumors remains problematic and necessitates the discovery of novel therapies that could be accelerated through a comparative approach, such as the inclusion of pet dogs with naturally-occurring brain cancers. Evidence supports procaspase-3 as a druggable brain cancer target with PAC-1, a pro-apoptotic, small molecule activator of procaspase-3 that crosses the blood-brain barrier. Procaspase-3 is frequently overexpressed in malignantly transformed tissues and provides a preferential target for inducing cancer cell apoptosis. While preliminary evidence supports procaspase-3 as a viable target in preclinical models, with PAC-1 demonstrating activity in rodent models and dogs with spontaneous brain tumors, the broader applicability of procaspase-3 as a target in human brain cancers, as well as the comparability of procaspase-3 expressions between differing species, requires further investigation. As such, a large-scale validation of procaspase-3 as a druggable target was undertaken across 651 human and canine brain tumors. Relative to normal brain tissues, procaspase-3 was overexpressed in histologically diverse cancerous brain tissues, supporting procaspase-3 as a broad and conserved therapeutic target. Additionally, procaspase-3 expressing glioma and meningioma cell lines were sensitive to the apoptotic effects of PAC-1 at biologically relevant exposures achievable in cancer patients. Importantly, the clinical relevance of procaspase-3 as a potential prognostic variable was demonstrated in human astrocytomas of variable histologic grades and associated clinical outcomes, whereby tumoral procaspase-3 expression was negatively correlated with survival; findings which suggest that PAC-1 might provide the greatest benefit for patients with the most guarded prognoses.
Collapse
Affiliation(s)
- Lisa J. Schlein
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Bahaa Fadl-Alla
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Holly C. Pondenis
- Department of Veterinary Clinical Medicine and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Stéphane Lezmi
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Charles G. Eberhart
- Department of Neuropathology and Ophthalmic Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Amy K. LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Peter J. Dickinson
- Department of Surgical and Radiological Sciences, University of California, Davis, Davis, CA, United States
| | - Paul J. Hergenrother
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Timothy M. Fan
- Department of Veterinary Clinical Medicine and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
8
|
Sylte I, Dawadi R, Malla N, von Hofsten S, Nguyen TM, Solli AI, Berg E, Adekoya OA, Svineng G, Winberg JO. The selectivity of galardin and an azasugar-based hydroxamate compound for human matrix metalloproteases and bacterial metalloproteases. PLoS One 2018; 13:e0200237. [PMID: 30075004 PMCID: PMC6075749 DOI: 10.1371/journal.pone.0200237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 06/22/2018] [Indexed: 02/07/2023] Open
Abstract
Inhibitors targeting bacterial enzymes should not interfere with enzymes of the host, and knowledge about structural determinants for selectivity is important for designing inhibitors with a therapeutic potential. We have determined the binding strengths of two hydroxamate compounds, galardin and compound 1b for the bacterial zinc metalloproteases, thermolysin, pseudolysin and auerolysin, known to be bacterial virulence factors, and the two human zinc metalloproteases MMP-9 and MMP-14. The active sites of the bacterial and human enzymes have huge similarities. In addition, we also studied the enzyme-inhibitor interactions by molecular modelling. The obtained Ki values of galardin for MMP-9 and MMP-14 and compound 1b for MMP-9 are approximately ten times lower than previously reported. Compound 1b binds stronger than galardin to both MMP-9 and MMP-14, and docking studies indicated that the diphenyl ether moiety of compound 1b obtains more favourable interactions within the S´1-subpocket than the 4-methylpentanoyl moiety of galardin. Both compounds bind stronger to MMP-9 than to MMP-14, which appears to be due to a larger S´1-subpocket in the former enzyme. Galardin, but not 1b, inhibits the bacterial enzymes, but the galardin Ki values were much larger than for the MMPs. The docking indicates that the S´1-subpockets of the bacterial proteases are too small to accommodate the diphenyl ether moiety of 1b, while the 4-methylpentanoyl moiety of galardin enters the pocket. The present study indicates that the size and shape of the ligand structural moiety entering the S´1-subpocket is an important determinant for selectivity between the studied MMPs and bacterial MPs.
Collapse
Affiliation(s)
- Ingebrigt Sylte
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Rangita Dawadi
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Nabin Malla
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Susannah von Hofsten
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Tra-Mi Nguyen
- Department of Pharmacy, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Ann Iren Solli
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Eli Berg
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Olayiwola A. Adekoya
- Department of Pharmacy, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Gunbjørg Svineng
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Jan-Olof Winberg
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
- * E-mail:
| |
Collapse
|
9
|
Wang F, Wang L, Li Y, Wang N, Wang Y, Cao Q, Gong P, Yang J, Wu C. PAC-1 and its derivative WF-210 Inhibit Angiogenesis by inhibiting VEGF/VEGFR pathway. Eur J Pharmacol 2017; 821:29-38. [PMID: 29269017 DOI: 10.1016/j.ejphar.2017.12.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 11/29/2022]
Abstract
Procaspase Activating Compound-1 (PAC-1) and its derivative WF-210 induce apoptosis in cancer cells by activating procaspase-3 to caspase-3. The aim of this study was to extend current knowledge about the mechanisms of PAC-1 and WF-210, particularly about their effects on tumor angiogenesis. PAC-1 and WF-210 restrained VEGF-induced human umbilical vascular endothelial cells (HUVECs) proliferation, invasion, and tube formation. PAC-1 and WF-210 abrogated VEGF-induced vessel sprouting from rat aortic rings and inhibited vascular formation in the Matrigel plug assay. PAC-1 and WF-210 suppressed phosphorylation of VEGFR2 and its downstream protein kinases c-Src, FAK, and AKT in both HUVECs and U-87 cells. When given to mice bearing subcutaneous or orthotopic xenograft, PAC-1 and WF-210 inhibited the tumor growth and tumor angiogenesis. Further tests showed that PAC-1 and WF-210 inhibited stemness and induced autophagy flux of U-87 cells. This study revealed mechanisms of PAC-1 and WF-210 other than inducing apoptosis, which provides additional support for their using in the clinic.
Collapse
Affiliation(s)
- Fangyang Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Yi Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Nannan Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Yating Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Qi Cao
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Ping Gong
- Department of Pharmaceutical Chemistry, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China.
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China.
| |
Collapse
|
10
|
Smirnov AS, Nikolaev DN, Gurzhiy VV, Smirnov SN, Suslonov VS, Garabadzhiu AV, Davidovich PB. Conformational stabilization of isatin Schiff bases – biologically active chemical probes. RSC Adv 2017. [DOI: 10.1039/c6ra26779c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Two ways of conformation stabilization for isatin Schiff bases (group of biologically active compounds) are reported here: complexation with metals that stabilize E-conformer and substitution in the 4th position of isatin core stabilizing Z-form.
Collapse
Affiliation(s)
- Andrey S. Smirnov
- Saint-Petersburg Technological Institute
- St. Petersburg
- Russia
- Saint-Petersburg State University
- St. Petersburg
| | | | | | | | | | | | | |
Collapse
|
11
|
Roth HS, Hergenrother PJ. Derivatives of Procaspase-Activating Compound 1 (PAC-1) and their Anticancer Activities. Curr Med Chem 2016; 23:201-41. [PMID: 26630918 PMCID: PMC4968085 DOI: 10.2174/0929867323666151127201829] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/04/2015] [Accepted: 11/27/2015] [Indexed: 01/26/2023]
Abstract
PAC-1 induces the activation of procaspase-3 in vitro and in cell culture by chelation of inhibitory labile zinc ions via its ortho-hydroxy-N-acylhydrazone moiety. First reported in 2006, PAC-1 has shown promise in cell culture and animal models of cancer, and a Phase I clinical trial in cancer patients began in March 2015 (NCT02355535). Because of the considerable interest in this compound and a well-defined structure-activity relationship, over 1000 PAC-1 derivatives have been synthesized in an effort to vary pharmacological properties such as potency and pharmacokinetics. This article provides a comprehensive examination of all PAC-1 derivatives reported to date. A survey of PAC-1 derivative libraries is provided, with an indepth discussion of four derivatives on which extensive studies have been performed.
Collapse
Affiliation(s)
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois, 261 Roger Adams Laboratory, Box 36-5, 600 S. Mathews Ave., Urbana, IL, 61801, USA.
| |
Collapse
|
12
|
Lobo G, Monasterios M, Rodrigues J, Gamboa N, Capparelli MV, Martínez-Cuevas J, Lein M, Jung K, Abramjuk C, Charris J. Synthesis, crystal structure and effect of indeno[1,2-b]indole derivatives on prostate cancer in vitro. Potential effect against MMP-9. Eur J Med Chem 2015; 96:281-95. [DOI: 10.1016/j.ejmech.2015.04.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 01/03/2023]
|
13
|
Roth HS, Botham RC, Schmid SC, Fan TM, Dirikolu L, Hergenrother PJ. Removal of Metabolic Liabilities Enables Development of Derivatives of Procaspase-Activating Compound 1 (PAC-1) with Improved Pharmacokinetics. J Med Chem 2015; 58:4046-65. [PMID: 25856364 DOI: 10.1021/acs.jmedchem.5b00413] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Procaspase-activating compound 1 (PAC-1) is an o-hydroxy-N-acylhydrazone that induces apoptosis in cancer cells by chelation of labile inhibitory zinc from procaspase-3. PAC-1 has been assessed in a wide variety of cell culture experiments and in vivo models of cancer, with promising results, and a phase 1 clinical trial in cancer patients has been initiated (NCT02355535). For certain applications, however, the in vivo half-life of PAC-1 could be limiting. Thus, with the goal of developing a compound with enhanced metabolic stability, a series of PAC-1 analogues were designed containing modifications that systematically block sites of metabolic vulnerability. Evaluation of the library of compounds identified four potentially superior candidates with comparable anticancer activity in cell culture, enhanced metabolic stability in liver microsomes, and improved tolerability in mice. In head-to-head experiments with PAC-1, pharmacokinetic evaluation in mice demonstrated extended elimination half-lives and greater area under the curve values for each of the four compounds, suggesting them as promising candidates for further development.
Collapse
Affiliation(s)
- Howard S Roth
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Rachel C Botham
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Steven C Schmid
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Timothy M Fan
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Levent Dirikolu
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Paul J Hergenrother
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| |
Collapse
|