Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Antiviral Res ; 226: 105888, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641024

ABSTRACT

296 million people worldwide are predisposed to developing severe end-stage liver diseases due to chronic hepatitis B virus (HBV) infection. HBV forms covalently closed circular DNA (cccDNA) molecules that persist as episomal DNA in the nucleus of infected hepatocytes and drive viral replication. Occasionally, the HBV genome becomes integrated into host chromosomal DNA, a process that is believed to significantly contribute to circulating HBsAg levels and HCC development. Neither cccDNA accumulation nor expression from integrated HBV DNA are directly targeted by current antiviral treatments. In this study, we investigated the antiviral properties of a newly described allosteric modulator, FLS-359, that targets sirtuin 2 (SIRT2), an NAD+-dependent deacylase. Our results demonstrate that SIRT2 modulation by FLS-359 and by other tool compounds inhibits cccDNA synthesis following de novo infection of primary human hepatocytes and HepG2 (C3A)-NTCP cells, and FLS-359 substantially reduces cccDNA recycling in HepAD38 cells. While pre-existing cccDNA is not eradicated by short-term treatment with FLS-359, its transcriptional activity is substantially impaired, likely through inhibition of viral promoter activities. Consistent with the inhibition of viral transcription, HBsAg production by HepG2.2.15 cells, which contain integrated HBV genomes, is also suppressed by FLS-359. Our study provides further insights on SIRT2 regulation of HBV infection and supports the development of potent SIRT2 inhibitors as HBV antivirals.


Subject(s)
Antiviral Agents , DNA, Circular , DNA, Viral , Hepatitis B virus , Hepatocytes , Sirtuin 2 , Virus Replication , Humans , DNA, Circular/metabolism , Sirtuin 2/antagonists & inhibitors , Sirtuin 2/metabolism , Hepatitis B virus/drug effects , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Hepatocytes/virology , Hepatocytes/drug effects , Antiviral Agents/pharmacology , Virus Replication/drug effects , Hep G2 Cells , Allosteric Regulation/drug effects , Transcription, Genetic/drug effects
2.
Antiviral Res ; 217: 105698, 2023 09.
Article in English | MEDLINE | ID: mdl-37562606

ABSTRACT

Peripheral blood monocytes are the cells predominantly responsible for systemic dissemination of human cytomegalovirus (HCMV) and a significant cause of morbidity and mortality in immunocompromised patients. HCMV establishes a silent/quiescent infection in monocytes, which is defined by the lack of viral replication and lytic gene expression. The absence of replication shields the virus within infected monocytes from the current available antiviral drugs that are designed to suppress active replication. Our previous work has shown that HCMV stimulates a noncanonical phosphorylation of Akt and the subsequent upregulation of a distinct subset of prosurvival proteins in normally short-lived monocytes. In this study, we found that SIRT2 activity is required for the unique activation profile of Akt induced within HCMV-infected monocytes. Importantly, both therapeutic and prophylactic treatment with a novel SIRT2 inhibitor, FLS-379, promoted death of infected monocytes via both the apoptotic and necroptotic cell death pathways. Mechanistically, SIRT2 inhibition reduced expression of Mcl-1, an Akt-dependent antiapoptotic Bcl-2 family member, and enhanced activation of MLKL, the executioner kinase of necroptosis. We have previously reported HCMV to block necroptosis by stimulating cellular autophagy. Here, we additionally demonstrate that inhibition of SIRT2 suppressed Akt-dependent HCMV-induced autophagy leading to necroptosis of infected monocytes. Overall, our data show that SIRT2 inhibition can simultaneously promote death of quiescently infected monocytes by two distinct death pathways, apoptosis and necroptosis, which may be vital for limiting viral dissemination to peripheral organs in immunosuppressed patients.


Subject(s)
Cytomegalovirus , Monocytes , Humans , Monocytes/metabolism , Cytomegalovirus/physiology , Proto-Oncogene Proteins c-akt/metabolism , Necroptosis , Sirtuin 2/metabolism , Apoptosis , Cells, Cultured
3.
J Clin Invest ; 133(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37317966

ABSTRACT

Most drugs used to treat viral disease target a virus-coded product. They inhibit a single virus or virus family, and the pathogen can readily evolve resistance. Host-targeted antivirals can overcome these limitations. The broad-spectrum activity achieved by host targeting can be especially useful in combating emerging viruses and for treatment of diseases caused by multiple viral pathogens, such as opportunistic agents in immunosuppressed patients. We have developed a family of compounds that modulate sirtuin 2, an NAD+-dependent deacylase, and now report the properties of a member of that family, FLS-359. Biochemical and x-ray structural studies show that the drug binds to sirtuin 2 and allosterically inhibits its deacetylase activity. FLS-359 inhibits the growth of RNA and DNA viruses, including members of the coronavirus, orthomyxovirus, flavivirus, hepadnavirus, and herpesvirus families. FLS-359 acts at multiple levels to antagonize cytomegalovirus replication in fibroblasts, causing modest reductions in viral RNAs and DNA, together with a much greater reduction in infectious progeny, and it exhibits antiviral activity in humanized mouse models of infection. Our results highlight the potential of sirtuin 2 inhibitors as broad-spectrum antivirals and set the stage for further understanding of how host epigenetic mechanisms impact the growth and spread of viral pathogens.


Subject(s)
Coronavirus Infections , Coronavirus , Animals , Mice , Antiviral Agents/pharmacology , Sirtuin 2/genetics , RNA, Viral
4.
Nat Cancer ; 3(1): 43-59, 2022 01.
Article in English | MEDLINE | ID: mdl-35121987

ABSTRACT

Metastatic breast cancer is a leading health burden worldwide. Previous studies have shown that metadherin (MTDH) promotes breast cancer initiation, metastasis and therapy resistance; however, the therapeutic potential of targeting MTDH remains largely unexplored. Here, we used genetically modified mice and demonstrate that genetic ablation of Mtdh inhibits breast cancer development through disrupting the interaction with staphylococcal nuclease domain-containing 1 (SND1), which is required to sustain breast cancer progression in established tumors. We performed a small-molecule compound screening to identify a class of specific inhibitors that disrupts the protein-protein interaction (PPI) between MTDH and SND1 and show that our lead candidate compounds C26-A2 and C26-A6 suppressed tumor growth and metastasis and enhanced chemotherapy sensitivity in preclinical models of triple-negative breast cancer (TNBC). Our results demonstrate a significant therapeutic potential in targeting the MTDH-SND1 complex and identify a new class of therapeutic agents for metastatic breast cancer.


Subject(s)
Endonucleases/metabolism , Membrane Proteins/metabolism , Micrococcal Nuclease , RNA-Binding Proteins/metabolism , Triple Negative Breast Neoplasms , Animals , Cell Adhesion Molecules/genetics , Humans , Membrane Proteins/genetics , Mice , RNA-Binding Proteins/genetics , Transcription Factors
5.
J Med Chem ; 56(20): 7788-803, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24093940

ABSTRACT

The IAPs are key regulators of the apoptotic pathways and are commonly overexpressed in many cancer cells. IAPs contain one to three BIR domains that are crucial for their inhibitory function. The pro-survival properties of XIAP come from binding of the BIR domains to the pro-apoptotic caspases. The BIR3 domain of XIAP binds and inhibits caspase 9, while the BIR2 domain binds and inhibits the terminal caspases 3 and 7. While XIAP BIR3 inhibitors have previously been reported, they also inhibit cIAP1/2 and promote the release of TNFα, potentially limiting their therapeutic utility. This paper will focus on the optimization of selective XIAP BIR2 inhibitors leading to the discovery of highly potent benzodiazepinone 36 (IC50 = 45 nM), which has high levels of selectivity over XIAP BIR3 and cIAP1 BIR2/3 and shows efficacy in a xenograft pharmacodynamic model monitoring caspase activity while not promoting the release of TNFα in vitro.


Subject(s)
Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Alanine/analogs & derivatives , Alanine/chemical synthesis , Alanine/pharmacokinetics , Alanine/pharmacology , Animals , Apoptosis/drug effects , Benzodiazepinones/chemical synthesis , Benzodiazepinones/pharmacokinetics , Benzodiazepinones/pharmacology , Blotting, Western , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Female , Heterocyclic Compounds/pharmacokinetics , Humans , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, Nude , Models, Chemical , Models, Molecular , Molecular Structure , Protein Structure, Tertiary , Ubiquitin-Protein Ligases , X-Linked Inhibitor of Apoptosis Protein/chemistry , X-Linked Inhibitor of Apoptosis Protein/metabolism , Xenograft Model Antitumor Assays
6.
J Med Chem ; 56(20): 7772-87, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24083782

ABSTRACT

XIAP is a key regulator of apoptosis, and its overexpression in cancer cells may contribute to their survival. The antiapoptotic function of XIAP derives from its BIR domains, which bind to and inhibit pro-apoptotic caspases. Most known IAP inhibitors are selective for the BIR3 domain and bind to cIAP1 and cIAP2 as well as XIAP. Pathways activated upon cIAP binding contribute to the function of these compounds. Inhibitors selective for XIAP should exert pro-apoptotic effects through competition with the terminal caspases. This paper details our synthetic explorations of a novel XIAP BIR2-selective benzazepinone screening hit with a focus on increasing BIR2 potency and overcoming high in vivo clearance. These efforts led to the discovery of benzoxazepinone 40, a potent BIR2-selective inhibitor with good in vivo pharmacokinetic properties which potentiates apoptotic signaling in a manner mechanistically distinct from that of known pan-IAP inhibitors.


Subject(s)
Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Alanine/analogs & derivatives , Alanine/chemical synthesis , Alanine/pharmacokinetics , Alanine/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Female , Heterocyclic Compounds/pharmacokinetics , Humans , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, Nude , Models, Chemical , Models, Molecular , Molecular Structure , Oxazepines/chemical synthesis , Oxazepines/pharmacokinetics , Oxazepines/pharmacology , Protein Structure, Tertiary , Rats , Ubiquitin-Protein Ligases , X-Linked Inhibitor of Apoptosis Protein/chemistry , X-Linked Inhibitor of Apoptosis Protein/metabolism , Xenograft Model Antitumor Assays
7.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 9): 1717-25, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23999295

ABSTRACT

XIAP, a member of the inhibitor of apoptosis family of proteins, is a critical regulator of apoptosis. Inhibition of the BIR domain-caspase interaction is a promising approach towards treating cancer. Previous work has been directed towards inhibiting the BIR3-caspase-9 interaction, which blocks the intrinsic apoptotic pathway; selectively inhibiting the BIR2-caspase-3 interaction would also block the extrinsic pathway. The BIR2 domain of XIAP has successfully been crystallized; peptides and small-molecule inhibitors can be soaked into these crystals, which diffract to high resolution. Here, the BIR2 apo crystal structure and the structures of five BIR2-tetrapeptide complexes are described. The structural flexibility observed on comparing these structures, along with a comparison with XIAP BIR3, affords an understanding of the structural elements that drive selectivity between BIR2 and BIR3 and which can be used to design BIR2-selective inhibitors.


Subject(s)
Caspase 3/chemistry , Caspase 3/metabolism , Caspase Inhibitors/chemistry , Inhibitor of Apoptosis Proteins/chemistry , Nucleopolyhedroviruses/chemistry , Viral Proteins/chemistry , X-Linked Inhibitor of Apoptosis Protein/chemistry , Amino Acid Sequence , Apoproteins/chemistry , Apoproteins/genetics , Apoptosis/genetics , Crystallography, X-Ray , Humans , Inhibitor of Apoptosis Proteins/genetics , Molecular Sequence Data , Multigene Family/genetics , Nucleopolyhedroviruses/genetics , Oligopeptides/chemistry , Oligopeptides/genetics , Protein Interaction Mapping , Protein Structure, Tertiary/genetics , Viral Proteins/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics
8.
J Med Chem ; 54(13): 4752-72, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21650221

ABSTRACT

Histone deacetylase (HDAC) inhibitors have shown promise in treating various forms of cancer. However, many HDAC inhibitors from diverse structural classes have been associated with QT prolongation in humans. Inhibition of the human ether a-go-go related gene (hERG) channel has been associated with QT prolongation and fatal arrhythmias. To determine if the observed cardiac effects of HDAC inhibitors in humans is due to hERG blockade, a highly potent HDAC inhibitor devoid of hERG activity was required. Starting with dacinostat (LAQ824), a highly potent HDAC inhibitor, we explored the SAR to determine the pharmacophores required for HDAC and hERG inhibition. We disclose here the results of these efforts where a high degree of pharmacophore homology between these two targets was discovered. This similarity prevented traditional strategies for mitigating hERG binding/modulation from being successful and novel approaches for reducing hERG inhibition were required. Using a hERG homology model, two compounds, 11r and 25i, were discovered to be highly efficacious with weak affinity for the hERG and other ion channels.


Subject(s)
Acrylamides/toxicity , Antineoplastic Agents/toxicity , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Histone Deacetylase Inhibitors/toxicity , Hydroxamic Acids/toxicity , Acrylamides/chemical synthesis , Acrylamides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , ERG1 Potassium Channel , HCT116 Cells , Half-Life , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacology , In Vitro Techniques , Mice , Mice, Nude , Microsomes, Liver/metabolism , Models, Molecular , Neoplasm Transplantation , Patch-Clamp Techniques , Radioligand Assay , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Tissue Distribution , Transplantation, Heterologous
9.
Novartis Found Symp ; 259: 249-66; discussion 266-8, 285-8, 2004.
Article in English | MEDLINE | ID: mdl-15171259

ABSTRACT

We have developed a cinnamic hydroxamic class of histone deacetylase inhibitors of which a prototype was designated as NVP-LAQ824. NVP-LAQ824, inhibits histone deacetylase enzymatic activities in vitro and transcriptionally activated the p21 promoter in reporter gene assays. When tested on a variety of solid tumour cell lines, NVP-LAQ824 exhibited selective anti-proliferative effects, inducing cell growth inhibition in some, while inducing cell death in others. To induce cell death, a minimum of 16 h exposure to NVP-LAQ824 is required. Flow cytometry studies revealed that both tumour cell lines and normal diploid fibroblasts arrested in the G2/M phase of the cell cycle after compound treatment. However, an increased sub-G1 population at 48 h (reminiscent of apoptotic cells) was only observed in the cancer cell lines. Annexin V staining data confirmed that NVP-LAQ824 induced apoptosis in tumour cells, but not in normal cells. To relate HDAC inhibition to the anti-proliferative effects of NVP-LAQ824, expression of HDAC 1 was inhibited using antisense and this was sufficient to activate p21 expression, hypophosphorylate Rb and inhibit cell growth. Furthermore, tumour cells treated with NVP-LAQ824 caused acetylation of HSP90 and degradation of its cargo oncoproteins. Finally, NVP-LAQ824 exhibited antitumour effects in a xenograft animal model. To determine if NVP-LAQ824 inhibited histone deacetylases in vivo, tumours treated with the drug were immunoblotted with an antibody specific for acetylated histones H3 and H4 and the results indicated increased histone H3 and 114 acetylation levels in NVP-LAQ824 treated cancer cells. Together, our data indicated that the activity of NVP-LAQ824 was consistent with its intended mechanism of action. This novel HDAC inhibitor is currently in clinical trials as an anticancer agent.


Subject(s)
Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Humans , Hydroxamic Acids/chemistry , Mice , Mice, Nude , Promoter Regions, Genetic , Tumor Cells, Cultured
10.
Haematologica ; 89(4): 419-26, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15075075

ABSTRACT

BACKGROUND AND OBJECTIVES: Chromatin structure and thereby transcription is controlled by the level of acetylation of histones, which is determined by the balance between histone acetyl transferase (HAT) activity and histone deacetylase (HDAC) activity. HDAC inhibitors are a class of compounds able to regulate gene expression by modulating chromatin structure. There are two major classes of HDAC inhibitors: the hydroxamic acid derivatives such as trichostatin A (TSA) or SAHA, and the butyrates such as phenyl-butyrate. HDAC inhibitors interfere with differentiation, proliferation and apoptosis in tumor cells. Here, we investigated the activity of a new hydroxamic acid derivative, LAQ824, on lymphoblastic cells. DESIGN AND METHODS: Four different pre-B lymphoblastic cell lines: Sup-B15 and TMD-5, both t(9;22) positive, SEM, t(4;11) positive, and NALM-6 cells were exposed to the hydroxamic acid derivatives, LAQ824 and TSA. Histone hyperacetylation, apoptosis, cell cycle and related pathways were assessed by flow cytometry and Western blotting. RESULTS: LAQ824 significantly inhibited the proliferation of leukemic lymphoblastic cell lines. The effect of LAQ824 was due to increased apoptosis accompanied by activation of caspase-3 and caspase-9, cleavage of poly(ADP-ribose)-polymerase (PARP) as well as by down-regulation of Bcl-2 and disruption of the mitochondrial membrane potential. Surprisingly, LAQ824-induced apoptosis was at least partially independent of caspase activation as indicated by the fact that LAQ824-induced apoptosis was inhibited only partially in both t(9;22) positive Sup-B15 and TMD-5 cells, whereas no inhibition was observed in t(4;11) positive SEM cells upon exposure to the polycaspase inhibitor zVAD-fmk. INTERPRETATION AND CONCLUSIONS: Our study establishes that LAQ824 is a promising agent for the therapy of acute lymphoblastic leukemia.


Subject(s)
Apoptosis/drug effects , Burkitt Lymphoma/pathology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Cell Line, Tumor , Humans , Hydroxamic Acids/pharmacology , Translocation, Genetic
11.
Cancer Res ; 64(2): 689-95, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14744786

ABSTRACT

We have synthesized a histone deacetylase inhibitor, NVP-LAQ824, a cinnamic hydroxamic acid, that inhibited in vitro enzymatic activities and transcriptionally activated the p21 promoter in reporter gene assays. NVP-LAQ824 selectively inhibited growth of cancer cell lines at submicromolar levels after 48-72 h of exposure, whereas higher concentrations and longer exposure times were required to retard the growth of normal dermal human fibroblasts. Flow cytometry studies revealed that both tumor and normal cells arrested in the G(2)-M phase of the cell cycle after compound treatment. However, an increased sub-G(1) population at 48 h (reminiscent of apoptotic cells) was observed only in the cancer cell line. Annexin V staining data supported our hypothesis that NVP-LAQ824 induced apoptosis in tumor and transformed cells but not in normal cells. Western blotting experiments showed an increased histone H3 and H4 acetylation level in NVP-LAQ824-treated cancer cells, suggesting that the likely in vivo target of NVP-LAQ824 was histone deacetylase(s). Finally, NVP-LAQ824 exhibited antitumor effects in a xenograft animal model. Together, our data indicated that the activity of NVP-LAQ824 was consistent with its intended mechanism of action. This novel histone deacetylase inhibitor is currently in clinical trials as an anticancer agent.


Subject(s)
Antineoplastic Agents/toxicity , Colonic Neoplasms/drug therapy , Histone Deacetylase Inhibitors , Hydroxamic Acids/toxicity , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Cell Division/genetics , Cell Line, Tumor , Colonic Neoplasms/pathology , Enzyme Inhibitors/toxicity , Fluorouracil/therapeutic use , Histone Deacetylases/isolation & purification , Humans , Kinetics , Male , Mice , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Transplantation, Heterologous
12.
J Med Chem ; 46(21): 4609-24, 2003 Oct 09.
Article in English | MEDLINE | ID: mdl-14521422

ABSTRACT

A series of N-hydroxy-3-phenyl-2-propenamides were prepared as novel inhibitors of human histone deacetylase (HDAC). These compounds were potent enzyme inhibitors, having IC(50)s < 400 nM in a partially purified enzyme assay. However, potency in cell growth inhibition assays ranged over 2 orders of magnitude in two human carcinoma cell lines. Selected compounds having cellular IC(50) < 750 nM were tested for maximum tolerated dose (MTD) and for efficacy in the HCT116 human colon tumor xenograft assay. Four compounds having an MTD > or = 100 mg/kg were selected for dose-response studies in the HCT116 xenograft model. One compound, 9 (NVP-LAQ824), had significant dose-related activity in the HCT116 colon and A549 lung tumor models, high MTD, and low gross toxicity. On the basis, in part, of these properties, 9 has entered human clinical trials in 2002.


Subject(s)
Acetyltransferases/antagonists & inhibitors , Acrylamides/chemical synthesis , Acrylamides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Animals , Body Weight/drug effects , Cell Division/drug effects , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Female , Histone Acetyltransferases , Humans , Indicators and Reagents , Mice , Mice, Nude , Molecular Conformation , Neoplasm Transplantation
13.
Curr Med Chem ; 10(22): 2393-402, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14529481

ABSTRACT

The natural products trapoxin B and trichostatin A, as well as the novel marine natural product psammaplin A (PSMA) were found in a cell-based screen for compounds that induced the expression of the cyclin dependent kinase inhibitor p21(waf1). The mechanism of p21(waf1) induction for these compounds was via histone deacetylase (HDAC) inhibition. Of these compounds, PSMA was of interest because of its novel structure, but the physiological stability of it, and its analogs was poor. Thus, a directed medicinal chemistry effort was undertaken to prepare analogs of the simple HDAC inhibitor dimethylaminobenzamidylcaprylic hydroxamate (DBCH), which led to chromenone amide 9. This compound was efficacious in the HCT116 colon xenograft assay, but was difficult to formulate in pharmaceutically acceptable vehicles. In parallel with these efforts, a screen of the Novartis compound archive for novel HDAC inhibitors uncovered the cinnamyl hydroxamic acid NVP-LAK974. This compound had good enzyme and cellular potency, but poor efficacy in vivo. A systematic structural exploration of cinnamyl hydroxamates based on NVP-LAK974 was undertaken with the goal of finding a novel, well-tolerated and efficacious HDAC inhibitor. Several derivatives were found to be efficacious in the xenograft assay. Of those compounds, NVP-LAQ824 distinguished itself due to its tolerability, efficacy and potency. Based, in part, on these properties, NVP-LAQ824 is currently undergoing human clinical trials as a novel anti-cancer agent.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/chemistry , Structure-Activity Relationship
14.
Blood ; 102(7): 2615-22, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-12816865

ABSTRACT

Histone deacetylase (HDAC) inhibitors are emerging as a promising new treatment strategy in hematologic malignancies. Here we show that NVP-LAQ824, a novel hydroxamic acid derivative, induces apoptosis at physiologically achievable concentrations (median inhibitory concentration [IC50] of 100 nM at 24 hours) in multiple myeloma (MM) cell lines resistant to conventional therapies. MM.1S myeloma cell proliferation was also inhibited when cocultured with bone marrow stromal cells, demonstrating ability to overcome the stimulatory effects of the bone marrow microenvironment. Importantly, NVP-LAQ824 also inhibited patient MM cell growth in a dose- and time-dependent manner. NVP-LAQ824-induced apoptotic signaling includes up-regulation of p21, caspase cascade activation, and poly (adenosine diphosphate [ADP]) ribose (PARP) cleavage. Apoptosis was confirmed with cell cycle analysis and annexin-propidium iodide staining. Interestingly, treatment of MM cells with NVPLAQ824 also led to proteasome inhibition, as determined by reduced proteasome chymotrypsin-like activity and increased levels of cellular polyubiquitin conjugates. Finally, a study using NVP-LAQ824 in a preclinical murine myeloma model provides in vivo relevance to our in vitro studies. Taken together, these findings provide the framework for NVP-LAQ824 as a novel therapeutic in MM.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Multiple Myeloma/drug therapy , Acetylation/drug effects , Animals , Cell Cycle/drug effects , Cysteine Endopeptidases/drug effects , Cysteine Endopeptidases/metabolism , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Humans , Immunocompromised Host , Interleukin-6/pharmacology , Mice , Mice, Nude , Multienzyme Complexes/drug effects , Multienzyme Complexes/metabolism , NF-kappa B/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
15.
J Org Chem ; 68(10): 3866-73, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12737565

ABSTRACT

Four novel bisulfide bromotyrosine derivatives, psammaplins E (9), F (10), G (11), and H (12), and two new bromotyrosine derivatives, psammaplins I (13) and J (14), were isolated from the sponge Pseudoceratina purpurea, along with known psammaplins A (4), B (6), C (7), and D (8) and bisaprasin (5). The structures of psammaplins E (9) and F (10), which each contain an oxalyl group rarely found in marine organisms, were determined by spectroscopic analysis. Compounds 4, 5, and 10 are potent histone deacetylase inhibitors and also show mild cytotoxicity. Furthermore, compounds 4, 5, and 11 are potent DNA methyltransferase inhibitors. The biogenetic pathway previously proposed for the psammaplins class is also revisited.


Subject(s)
DNA Modification Methylases/antagonists & inhibitors , Disulfides/isolation & purification , Enzyme Inhibitors/isolation & purification , Histone Deacetylase Inhibitors , Porifera/chemistry , Sulfuric Acid Esters/isolation & purification , Tyrosine/analogs & derivatives , Tyrosine/isolation & purification , Animals , Disulfides/chemistry , Disulfides/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Papua New Guinea , Sulfuric Acid Esters/chemistry , Sulfuric Acid Esters/pharmacology , Tyrosine/chemistry , Tyrosine/pharmacology
16.
Curr Opin Drug Discov Devel ; 5(4): 487-99, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12197307

ABSTRACT

The reversible acetylation of lysine amino groups in nuclear histones is an important mechanism by which gene expression is regulated, and may act by disrupting higher order chromatin structure. In normal cells, the competing activities of two enzyme classes, the histone acetyl transferases (HATs) and histone deacetylases (HDACs), result in cell-specific patterns of gene expression. Aberrant patterns of histone acetylation have been linked to cancer, and natural and synthetic HDAC inhibitors (HDAIs) have shown antiproliferative effects on tumor cells in culture. Thus, there has been an increasing interest in histone deacetylase inhibitors as novel anticancer agents, with clinical investigation underway for several molecules. This review summarizes recent progress in the development of new HDAIs and their reported activities.


Subject(s)
Enzyme Inhibitors , Histone Deacetylase Inhibitors , Technology, Pharmaceutical/trends , Animals , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Humans , Technology, Pharmaceutical/methods
17.
J Med Chem ; 45(4): 753-7, 2002 Feb 14.
Article in English | MEDLINE | ID: mdl-11831887

ABSTRACT

Inhibitors of histone deacetylase (HDAC) have been shown to induce terminal differentiation of human tumor cell lines and to have antitumor effects in vivo. We have prepared analogues of suberoylanilide hydroxamic acid (SAHA) and trichostatin A and have evaluated them in a human HDAC enzyme inhibition assay, a p21(waf1) (p21) promoter assay, and in monolayer growth inhibition assays. One compound, 4-(dimethylamino)-N-[7-(hydroxyamino)-7-oxoheptyl]-benzamide, was found to affect the growth of a panel of eight human tumor cell lines differentially.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzamides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors , Hydroxamic Acids/chemical synthesis , Hydroxylamines/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzamides/chemistry , Benzamides/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Hydroxylamines/chemistry , Hydroxylamines/pharmacology , Models, Molecular , Promoter Regions, Genetic , Structure-Activity Relationship , Tumor Cells, Cultured
18.
J Org Chem ; 63(3): 445-451, 1998 Feb 06.
Article in English | MEDLINE | ID: mdl-11672031

ABSTRACT

A comprehensive study of nitration reaction of azatricyclic systems has been carried out. Whereas classical nitrations using KNO(3)-H(2)SO(4) at low temperatures gave nitrated products mainly at the 9-position, use of tetrabutylammonium nitrate-trifluoroacetic anhydride (TBAN-TFAA) resulted in exclusive nitration of the 3-position in the case carbamates 1, and 4-6 and the tricyclic ketone 7. These 3-nitro tricyclic derivatives have been valuable intermediates for the preparation of the very potent farnesyl protein transferase inhibitors such as the tricyclic pyridyl acetamide 32 and other new analogues.

SELECTION OF CITATIONS
SEARCH DETAIL
...