Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Neoplasia ; 22(10): 511-521, 2020 10.
Article in English | MEDLINE | ID: mdl-32896761

ABSTRACT

Bone metastatic prostate cancer significantly impacts patient quality of life and overall survival, and despite available therapies, it is presently incurable with an unmet need for improved treatment options. As mediators of tumor progression, matrix metalloproteinases (MMPs) can degrade extracellular matrix components and regulate growth factor and cytokine bioactivity. Depending on tissue context, MMPs can either promote or inhibit tumorigenesis. Therefore, it is essential to study individual MMPs in specific cancer contexts and microenvironments to support the design and application of selective MMP inhibitors. Here we report that tumor-derived MMP-3 contributes to bone metastatic prostate cancer progression via intrinsic and extrinsic routes. MMP-3 ablation in prostate cancer cell lines significantly reduced in vitro growth combined with lowered AKT and ERK phosphorylation and total VEGFR1 and FGFR3 protein levels. In vivo, MMP-3 ablated tumors grew at a slower rate and were significantly less vascularized. Quantitative PCR analyses of wild type and MMP-3 silenced prostate cancer cells also demonstrate downregulation of a wide array of angiogenic factors. The extrinsic role for MMP-3 in angiogenesis was supported by in vitro endothelial tube formation assays where the lack of MMP-3 in prostate cancer conditioned media resulted in slower rates of tube formation. Taken together, our results suggest that tumor-derived MMP-3 contributes to prostate cancer growth in bone. These data indicate that selective inhibition of MMP-3 and/or targeting MMP generated products could be efficacious for the treatment of prostate to bone metastases.


Subject(s)
Bone Neoplasms/blood supply , Bone Neoplasms/secondary , Matrix Metalloproteinase 3/metabolism , Neovascularization, Pathologic/pathology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Animals , Apoptosis , Bone Neoplasms/enzymology , Cell Proliferation , Disease Models, Animal , Humans , Male , Matrix Metalloproteinase 3/genetics , Mice , Neovascularization, Pathologic/enzymology , Prostatic Neoplasms/enzymology , Tumor Cells, Cultured
2.
Pharmaceuticals (Basel) ; 13(6)2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32492898

ABSTRACT

Matrix metalloproteinases (MMPs) are a family of enzymes involved at different stages of cancer progression and metastasis. We previously identified a novel class of bisphosphonic inhibitors, selective for MMPs crucial for bone remodeling, such as MMP-2. Due to the increasing relevance of specific MMPs at various stages of tumor malignancy, we focused on improving potency towards certain isoforms. Here, we tackled MMP-9 because of its confirmed role in tumor invasion, metastasis, angiogenesis, and immuno-response, making it an ideal target for cancer therapy. Using a computational analysis, we designed and characterized potent MMP-2/MMP-9 inhibitors. This is a promising approach to develop and clinically translate inhibitors that could be used in combination with standard care therapy for the treatment of skeletal malignancies.

3.
Int J Cancer ; 147(10): 2811-2823, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32599665

ABSTRACT

Overall survival rates for patients with advanced osteosarcoma have remained static for over three decades. An in vitro analysis of osteosarcoma cell lines for sensitivity to an array of approved cancer therapies revealed that panobinostat, a broad spectrum histone deacetalyase (HDAC) inhibitor, is highly effective at triggering osteosarcoma cell death. Using in vivo models of orthotopic and metastatic osteosarcoma, here we report that panobinostat impairs the growth of primary osteosarcoma in bone and spontaneous metastasis to the lung, the most common site of metastasis for this disease. Further, pretreatment of mice with panobinostat prior to tail vein inoculation of osteosarcoma prevents the seeding and growth of lung metastases. Additionally, panobinostat impaired the growth of established lung metastases and improved overall survival, and these effects were also manifest in the lung metastatic SAOS2-LM7 model. Mechanistically, the efficacy of panobinostat was linked to high expression of HDAC1 and HDAC2 in osteosarcoma, and silencing of HDAC1 and 2 greatly reduced osteosarcoma growth in vitro. In accordance with these findings, treatment with the HDAC1/2 selective inhibitor romidepsin compromised the growth of osteosarcoma in vitro and in vivo. Analysis of patient-derived xenograft osteosarcoma cell lines further demonstrated the sensitivity of the disease to panobinostat or romidepsin. Collectively, these studies provide rationale for clinical trials in osteosarcoma patients using the approved therapies panobinostat or romidepsin.


Subject(s)
Bone Neoplasms/drug therapy , Histone Deacetylase Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Osteosarcoma/drug therapy , Animals , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Depsipeptides/administration & dosage , Depsipeptides/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Lung Neoplasms/metabolism , Mice , Osteosarcoma/metabolism , Panobinostat/administration & dosage , Panobinostat/pharmacology , Survival Analysis , Xenograft Model Antitumor Assays
4.
Cancers (Basel) ; 10(6)2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29874869

ABSTRACT

Bone metastatic breast cancer is currently incurable and will be evident in more than 70% of patients that succumb to the disease. Understanding the factors that contribute to the progression and metastasis of breast cancer can reveal therapeutic opportunities. Matrix metalloproteinases (MMPs) are a family of proteolytic enzymes whose role in cancer has been widely documented. They are capable of contributing to every step of the metastatic cascade, but enthusiasm for the use of MMP inhibition as a therapeutic approach has been dampened by the disappointing results of clinical trials conducted more than 20 years ago. Since the trials, our knowledge of MMP biology has expanded greatly. Combined with advances in the selective targeting of individual MMPs and the specific delivery of therapeutics to the tumor microenvironment, we may be on the verge of finally realizing the promise of MMP inhibition as a treatment strategy. Here, as a case in point, we focus specifically on MMP-2 as an example to show how it can contribute to each stage of breast-cancer-to-bone metastasis and also discuss novel approaches for the selective targeting of MMP-2 in the setting of the bone-cancer microenvironment.

5.
Oncotarget ; 8(26): 41827-41840, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28611279

ABSTRACT

Multiple myeloma is a plasma cell malignancy that homes aberrantly to bone causing extensive skeletal destruction. Despite the development of novel therapeutic agents that have significantly improved overall survival, multiple myeloma remains an incurable disease. Matrix metalloproteinase-2 (MMP-2) is associated with cancer and is significantly overexpressed in the bone marrow of myeloma patients. These data provide rationale for selectively inhibiting MMP-2 activity as a multiple myeloma treatment strategy. Given that MMP-2 is systemically expressed, we used novel "bone-seeking" bisphosphonate based MMP-2 specific inhibitors (BMMPIs) to target the skeletal tissue thereby circumventing potential off-target effects of MMP-2 inhibition outside the bone marrow-tumor microenvironment. Using in vivo models of multiple myeloma (5TGM1, U266), we examined the impact of MMP-2 inhibition on disease progression using BMMPIs. Our data demonstrate that BMMPIs can decrease multiple myeloma burden and protect against cancer-induced osteolysis. Additionally, we have shown that MMP-2 can be specifically inhibited in the multiple myeloma-bone microenvironment, underscoring the feasibility of developing targeted and tissue selective MMP inhibitors. Given the well-tolerated nature of bisphosphonates in humans, we anticipate that BMMPIs could be rapidly translated to the clinical setting for the treatment of multiple myeloma.


Subject(s)
Bone and Bones/pathology , Cellular Microenvironment/drug effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Animals , Biopsy , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cellular Microenvironment/genetics , Disease Models, Animal , Enzyme Activation/drug effects , Female , Gene Expression , Humans , Male , Matrix Metalloproteinase 2/genetics , Mice , Mice, Knockout , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteolysis/drug therapy , Osteolysis/metabolism , Osteolysis/pathology , Protein Transport
6.
Mol Cancer Ther ; 16(6): 1054-1067, 2017 06.
Article in English | MEDLINE | ID: mdl-28336808

ABSTRACT

Synergistic action of kinase and BET bromodomain inhibitors in cell killing has been reported for a variety of cancers. Using the chemical scaffold of the JAK2 inhibitor TG101348, we developed and characterized single agents which potently and simultaneously inhibit BRD4 and a specific set of oncogenic tyrosine kinases including JAK2, FLT3, RET, and ROS1. Lead compounds showed on-target inhibition in several blood cancer cell lines and were highly efficacious at inhibiting the growth of hematopoietic progenitor cells from patients with myeloproliferative neoplasm. Screening across 931 cancer cell lines revealed differential growth inhibitory potential with highest activity against bone and blood cancers and greatly enhanced activity over the single BET inhibitor JQ1. Gene drug sensitivity analyses and drug combination studies indicate synergism of BRD4 and kinase inhibition as a plausible reason for the superior potency in cell killing. Combined, our findings indicate promising potential of these agents as novel chemical probes and cancer therapeutics. Mol Cancer Ther; 16(6); 1054-67. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Design , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor/methods , Drug Synergism , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Mice , Models, Molecular , Molecular Conformation , Protein Kinase Inhibitors/chemistry , Proteins/chemistry , Structure-Activity Relationship , Xenograft Model Antitumor Assays
7.
Mol Cancer Ther ; 16(3): 494-505, 2017 03.
Article in English | MEDLINE | ID: mdl-28069877

ABSTRACT

Bone metastasis is common during breast cancer progression. Matrix metalloproteinase-2 (MMP-2) is significantly associated with aggressive breast cancer and poorer overall survival. In bone, tumor- or host-derived MMP-2 contributes to breast cancer growth and does so by processing substrates, including type I collagen and TGFß latency proteins. These data provide strong rationale for the application of MMP-2 inhibitors to treat the disease. However, in vivo, MMP-2 is systemically expressed. Therefore, to overcome potential toxicities noted with previous broad-spectrum MMP inhibitors (MMPIs), we used highly selective bisphosphonic-based MMP-2 inhibitors (BMMPIs) that allowed for specific bone targeting. In vitro, BMMPIs affected the viability of breast cancer cell lines and osteoclast precursors, but not osteoblasts. In vivo, we demonstrated using two bone metastatic models (PyMT-R221A and 4T1) that BMMPI treatment significantly reduced tumor growth and tumor-associated bone destruction. In addition, BMMPIs are superior in promoting tumor apoptosis compared with the standard-of-care bisphosphonate, zoledronate. We demonstrated MMP-2-selective inhibition in the bone microenvironment using specific and broad-spectrum MMP probes. Furthermore, compared with zoledronate, BMMPI-treated mice had significantly lower levels of TGFß signaling and MMP-generated type I collagen carboxy-terminal fragments. Taken together, our data show the feasibility of selective inhibition of MMPs in the bone metastatic breast cancer microenvironment. We posit that BMMPIs could be easily translated to the clinical setting for the treatment of bone metastases given the well-tolerated nature of bisphosphonates. Mol Cancer Ther; 16(3); 494-505. ©2017 AACR.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Animals , Bone Density Conservation Agents/pharmacology , Bone Neoplasms/diagnosis , Bone Neoplasms/mortality , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Survival/drug effects , Diphosphonates/pharmacology , Disease Progression , Enzyme Activation/drug effects , Female , Humans , Imidazoles/pharmacology , Kaplan-Meier Estimate , Matrix Metalloproteinase 2/genetics , Mice , Models, Biological , Multimodal Imaging , Osteoclasts/drug effects , Osteoclasts/metabolism , Tumor Burden/drug effects , Tumor Microenvironment , Zoledronic Acid
8.
J Enzyme Inhib Med Chem ; 31(sup4): 25-37, 2016.
Article in English | MEDLINE | ID: mdl-27556138

ABSTRACT

New catechol-containing chemical entities have been investigated as matrix metalloproteinase inhibitors as well as antioxidant molecules. The combination of the two properties could represent a useful feature due to the potential application in all the pathological processes characterized by increased proteolytic activity and radical oxygen species (ROS) production, such as inflammation and photoaging. A series of catechol-based molecules were synthesized and tested for both proteolytic and oxidative inhibitory activity, and the detailed binding mode was assessed by crystal structure determination of the complex between a catechol derivative and the matrix metalloproteinase-8. Surprisingly, X-ray structure reveals that the catechol oxygens do not coordinates the zinc atom.


Subject(s)
Antioxidants/pharmacology , Catechols/pharmacology , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Antioxidants/chemical synthesis , Antioxidants/chemistry , Catechols/chemical synthesis , Catechols/chemistry , Dose-Response Relationship, Drug , Humans , Matrix Metalloproteinase 8/isolation & purification , Matrix Metalloproteinase Inhibitors/chemical synthesis , Matrix Metalloproteinase Inhibitors/chemistry , Models, Molecular , Molecular Structure , Quantitative Structure-Activity Relationship , Reactive Oxygen Species/metabolism
9.
ChemMedChem ; 11(6): 612-9, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26915684

ABSTRACT

A small library of 2,3-dihydroxybenzamide- and N-(2,3-dihydroxyphenyl)-4-sulfonamide-based microsomal prostaglandin E2 synthase-1 (mPGES-1) inhibitors was identified following a step-by-step optimization of small aromatic fragments selected to interact in focused regions in the active site of mPGES-1. During the virtual optimization process, the 2,3-dihydroxybenzamide moiety was first selected as a backbone of the proposed new chemical entities; the identified compounds were then synthesized and biologically evaluated, identifying derivatives with very promising inhibitory activities in the micromolar range. Subsequent structure-guided replacement of the 2,3-dihydroxybenzamide by the N-(2,3-dihydroxyphenyl)sulfonamide moiety led to the identification of N-(2,3-dihydroxyphenyl)-4-biphenylsulfonamide (6), the most potent small molecule of the series (IC50 =0.53 ± 0.04 µM). The simple synthetic procedure and the possibility of enhancing the potency of this class of inhibitors through additional structural modifications pave the way for further development of new molecules with mPGES-1-inhibitory activity, with potential application as anti-inflammatory and anticancer agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzamides/chemical synthesis , Cyclooxygenase 1/metabolism , Cyclooxygenase Inhibitors/chemical synthesis , Sulfonamides/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Cyclooxygenase Inhibitors/pharmacology , Humans , Molecular Docking Simulation , Quantitative Structure-Activity Relationship , Sulfonamides/pharmacology
10.
Clin Oral Investig ; 20(8): 2013-2021, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26689567

ABSTRACT

OBJECTIVES: Bisphosphonates (BPs) are drugs clinically used in resorptive diseases. It was already proved that some clinically relevant BPs can inhibit a class of enzymes called matrix metalloproteinases (MMPs), required during tissue remodelling. Combining the arylsulfonamide function with the bisphosphonic group, several compounds were synthesized to obtain selective inhibitors of MMPs. The aim of the present study was to compare the effect of zoledronic acid (ZA), the most potent bisphosphonate available as therapy, with new sulfonamide containing BPs in an in vitro model of human gingival fibroblasts (HGFs). MATERIALS AND METHODS: Western blot was used to measure procollagen I, ß1 integrin MMP-8 and MMP-9, phase contrast and MTT for cell viability; L-lactate-dehydrogenase (LDH) measurement was performed for toxicity evaluation and ELISA for prostaglandin E2 (PGE2) secretion assessment. RESULTS: When compared with ZA, the treatment with the newly synthesized compounds shows increasing viability, procollagen I expression and decreased expression of ß1 integrin in HGFs. Higher levels of released LDH, PGE2 and MMP-9 expression are recorded in ZA-treated HGFs. Increased levels of MMP-8 are recorded in newly synthesized compounds-treated samples. CONCLUSIONS: These findings allowed to conclude that new tested BPs did not affect HGFs viability and adhesion, did not induce cellular toxicity, were not responsible for inflammatory event induction and could preserve the physiological matrix turnover. CLINICAL RELEVANCE: It could be hypothesized that the new molecules were better tolerated by soft tissues, resulting in lesser side effects.


Subject(s)
Bone Density Conservation Agents/pharmacology , Diphosphonates/pharmacology , Fibroblasts/drug effects , Gingiva/cytology , Imidazoles/pharmacology , Adult , Biomarkers/analysis , Blotting, Western , Bone Density Conservation Agents/chemical synthesis , Cell Adhesion/drug effects , Cell Survival/drug effects , Diphosphonates/chemical synthesis , Enzyme-Linked Immunosorbent Assay , Humans , Imidazoles/chemical synthesis , In Vitro Techniques , Inflammation , Molar, Third , Zoledronic Acid
11.
Curr Drug Targets ; 16(14): 1634-44, 2015.
Article in English | MEDLINE | ID: mdl-25585125

ABSTRACT

Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases, capable to degrade the extracellular matrix (ECM) in physiologic conditions. Because of their overexpression and pivotal role in many pathological events, they have been proposed as a therapeutic and prognostic target for a number of diseases. Selectivity among MMPs is essential for realizing the clinical potential of inhibitors. The design of MMP inhibitors (MMPIs) has largely focused on development of various compounds containing a zinc binding group (ZBG) in their structure, with hydroxamate being the most potent one. Due to the high degree of homology in the catalytic domain of all the MMPs, the specificity and selectivity of first generation hydroxamate MMPIs were minimal, with several off-target effects and binding to other metzincins. This review highlights the role of phosphonate as ZBG in the design and development of new MMPIs.


Subject(s)
Matrix Metalloproteinase Inhibitors/chemical synthesis , Organophosphonates/chemistry , Binding Sites , Clinical Trials as Topic , Humans , Matrix Metalloproteinase Inhibitors/chemistry , Models, Molecular
12.
Eur J Med Chem ; 90: 583-94, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25497132

ABSTRACT

The effects resulting from the introduction of an oxime group in place of the distal aromatic ring of the diphenyl moiety of LT175, previously reported as a PPARα/γ dual agonist, have been investigated. This modification allowed the identification of new bioisosteric ligands with fairly good activity on PPARα and fine-tuned moderate activity on PPARγ. For the most interesting compound (S)-3, docking studies in PPARα and PPARγ provided a molecular explanation for its different behavior as full and partial agonist of the two receptor isotypes, respectively. A further investigation of this compound was carried out performing gene expression studies on HepaRG cells. The results obtained allowed to hypothesize a possible mechanism through which this ligand could be useful in the treatment of metabolic disorders. The higher induction of the expression of some genes, compared to selective agonists, seems to confirm the importance of a dual PPARα/γ activity which probably involves a synergistic effect on both receptor subtypes.


Subject(s)
Drug Design , Oximes/pharmacology , PPAR alpha/agonists , PPAR alpha/metabolism , PPAR gamma/agonists , PPAR gamma/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Ligands , Molecular Docking Simulation , Molecular Structure , Oximes/chemical synthesis , Oximes/chemistry , PPAR alpha/genetics , PPAR gamma/genetics , Structure-Activity Relationship
13.
Cancer Metastasis Rev ; 33(4): 1043-57, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25325988

ABSTRACT

Heightened matrix metalloproteinase (MMP) activity has been noted in the context of the tumor microenvironment for many years, and causal roles for MMPs have been defined across the spectrum of cancer progression. This is primarily due to the ability of the MMPs to process extracellular matrix (ECM) components and to regulate the bioavailability/activity of a large repertoire of cytokines and growth factors. These characteristics made MMPs an attractive target for therapeutic intervention but notably clinical trials performed in the 1990s did not fulfill the promise of preclinical studies. The reason for the failure of early MMP inhibitor (MMPI) clinical trials that are multifold but arguably principal among them was the inability of early MMP-based inhibitors to selectively target individual MMPs and to distinguish between MMPs and other members of the metzincin family. In the decades that have followed the MMP inhibitor trials, innovations in chemical design, antibody-based strategies, and nanotechnologies have greatly enhanced our ability to specifically target and measure the activity of MMPs. These advances provide us with the opportunity to generate new lines of highly selective MMPIs that will not only extend the overall survival of cancer patients, but will also afford us the ability to utilize heightened MMP activity in the tumor microenvironment as a means by which to deliver MMPIs or MMP activatable prodrugs.


Subject(s)
Carcinogenesis/drug effects , Matrix Metalloproteinase Inhibitors/therapeutic use , Matrix Metalloproteinases/genetics , Neoplasms/genetics , Extracellular Matrix/drug effects , Humans , Matrix Metalloproteinases/biosynthesis , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic , Tumor Microenvironment/drug effects
14.
Bioorg Med Chem Lett ; 24(12): 2617-20, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24813742

ABSTRACT

A set of bisphosphonate matrix metalloproteinase (MMP) inhibitors was investigated for inhibitory activity against several carbonic anhydrase (CA, EC 4.2.1.1) isozymes, some of which are overexpressed in hypoxic tumors. Some of the bisphosphonate revealed to be very potent inhibitors (in the low nanomolar range) of the cytosolic isoform CA II and the membrane-bound CA IX, XII and XIV isozymes, a feature useful for considering them as interesting compounds for bone resorption inhibition applications. We suggest here that it is possible to develop dual enzyme inhibitors bearing bisphosphonate moieties that may target both MMPs and CAs, two families of enzymes involved in tumor formation, growth, and metastasis.


Subject(s)
Bone Neoplasms/enzymology , Carbonic Anhydrase Inhibitors/pharmacology , Diphosphonates/pharmacology , Imidazoles/pharmacology , Matrix Metalloproteinase Inhibitors/pharmacology , Bone Neoplasms/drug therapy , Carbonic Anhydrase Inhibitors/therapeutic use , Cell Line , Diphosphonates/therapeutic use , Enzyme Activation/drug effects , Humans , Imidazoles/therapeutic use , Inhibitory Concentration 50 , Matrix Metalloproteinase Inhibitors/therapeutic use , Molecular Structure , Zoledronic Acid
15.
Bioorg Med Chem Lett ; 24(8): 1941-3, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24650641

ABSTRACT

A set of matrix metalloproteinases (MMPs) inhibitors, containing a bisphosphonate moiety (BP), has been evaluated for the inhibitory activity of carbonic anhydrases (CAs, EC 4.2.1.1). Human (h) isoforms hCA I, II, IX, XII and XIV were included in the study due to their involvement in crucial physiologic and pathologic processes. Some of these molecules selectively inhibited CA XII in the nanomolar range, showing an attractive dual mechanism (anti-MMP and anti-CA) of action as potential antitumor agents. The BP inhibitors investigated in this study are also excellent leads for obtaining even more effective compounds able to selectively target membrane-bound CA XII and having the potential to be used as tools for understanding physiologic processes regulated by this isoform.


Subject(s)
Antineoplastic Agents/chemical synthesis , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/chemistry , Diphosphonates/chemical synthesis , Diphosphonates/pharmacology , Acetazolamide/chemistry , Acetazolamide/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carbonic Anhydrase Inhibitors/chemistry , Diphosphonates/chemistry , Drug Delivery Systems , Enzyme Activation/drug effects , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Inhibitory Concentration 50 , Molecular Structure , Protein Isoforms/chemistry , Structure-Activity Relationship , Zoledronic Acid
16.
Bioorg Med Chem ; 21(21): 6456-65, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24071448

ABSTRACT

The complexity of matrix metalloproteinase inhibitors (MMPIs) design derives from the difficulty in carefully addressing their inhibitory activity towards the MMP isoforms involved in many pathological conditions. In particular, specific metalloproteinases, such as MMP-2 and MMP-9, are key regulators of the 'vicious cycle' occurring between tumor metastases growth and bone remodeling. In an attempt to devise new approaches to selective inhibitor derivatives, we describe novel bisphosphonate bone seeking MMP inhibitors (BP-MMPIs), capable to be selectively targeted and to overcome undesired side effects of broad spectrum MMPIs. In vitro activity (IC50 values) for each inhibitor was determined against MMP-2, -8, -9 and -14, because of their relevant role in skeletal development and renewal. The results show that BP-MMPIs reached IC50 values of enzymatic inhibition in the low micromolar range. Computational studies, used to rationalize some trends in the observed inhibitory profiles, suggest a possible differential binding mode in MMP-2 that explains the selective inhibition of this isoform. In addition, survival assay was conducted on J774 cell line, a well known model system used to evaluate the structure-activity relationship of BPs for inhibiting bone resorption. The resulting data, confirming the specific activity of BP-MMPIs, and their additional proved propensity to bind hydroxyapatite powder in vitro, suggest a potential use of BP-MMPIs in skeletal malignancies.


Subject(s)
Diphosphonates/chemistry , Matrix Metalloproteinase Inhibitors/chemistry , Matrix Metalloproteinases/chemistry , Animals , Binding Sites , Cell Line , Cell Survival/drug effects , Diphosphonates/metabolism , Diphosphonates/toxicity , Durapatite/chemistry , Durapatite/metabolism , Hep G2 Cells , Humans , Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 8/chemistry , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/metabolism , Matrix Metalloproteinase Inhibitors/toxicity , Matrix Metalloproteinases/metabolism , Mice , Molecular Docking Simulation , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...