Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 295(44): 15013-15028, 2020 10 30.
Article in English | MEDLINE | ID: mdl-32855235

ABSTRACT

The antimalarial agents artemisinins inhibit cytomegalovirus (CMV) in vitro and in vivo, but their target(s) has been elusive. Using a biotin-labeled artemisinin, we identified the intermediate filament protein vimentin as an artemisinin target, validated by detailed biochemical and biological assays. We provide insights into the dynamic and unique modulation of vimentin, depending on the stage of human CMV (HCMV) replication. In vitro, HCMV entry and viral progeny are reduced in vimentin-deficient fibroblasts, compared with control cells. Similarly, mouse CMV (MCMV) replication in vimentin knockout mice is significantly reduced compared with controls in vivo, confirming the requirement of vimentin for establishment of infection. Early after HCMV infection of human foreskin fibroblasts vimentin level is stable, but as infection proceeds, vimentin is destabilized, concurrent with its phosphorylation and virus-induced calpain activity. Intriguingly, in vimentin-overexpressing cells, HCMV infection is reduced compared with control cells. Binding of artesunate, an artemisinin monomer, to vimentin prevents virus-induced vimentin degradation, decreasing vimentin phosphorylation at Ser-55 and Ser-83 and resisting calpain digestion. In vimentin-deficient fibroblasts, the anti-HCMV activity of artesunate is reduced compared with controls. In summary, an intact and stable vimentin network is important for the initiation of HCMV replication but hinders its completion. Artesunate binding to vimentin early during infection stabilizes it and antagonizes subsequent HCMV-mediated vimentin destabilization, thus suppressing HCMV replication. Our target discovery should enable the identification of vimentin-binding sites and compound moieties for binding.


Subject(s)
Antiviral Agents/pharmacology , Artemisinins/pharmacology , Cytomegalovirus/drug effects , Vimentin/antagonists & inhibitors , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Artemisinins/chemistry , Artemisinins/metabolism , Artesunate/pharmacology , Binding Sites , Calpain/metabolism , Cell Cycle/drug effects , Cells, Cultured , Cytomegalovirus/physiology , Cytomegalovirus Infections/metabolism , Drug Repositioning , Humans , Mass Spectrometry , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Vimentin/genetics , Vimentin/metabolism , Virus Replication/drug effects , Withanolides/pharmacology
2.
Oncotarget ; 7(6): 7268-79, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26771236

ABSTRACT

Artemisinins, endoperoxide-containing molecules, best known as antimalarials, have potent antineoplastic activity. The established antimalarial, artesunate (AS), and the novel artemisinin-derived trioxane diphenylphosphate dimer 838 (ART-838) inhibited growth of all 23 tested acute leukemia cell lines, reduced cell proliferation and clonogenicity, induced apoptosis, and increased intracellular levels of reactive oxygen species (ROS). ART-838 was 88-fold more potent that AS in vitro, inhibiting all leukemia cell lines at submicromolar concentrations. Both ART-838 and AS cooperated with several established antileukemic drugs and newer kinase inhibitors to inhibit leukemia cell growth. ART-838 had a longer plasma half-life than AS in immunodeficient NOD-SCID-IL2Rgnull (NSG) mice, remaining at effective antileukemic concentrations for >8h. Intermittent cycles of ART-838 inhibited growth of acute leukemia xenografts and primagrafts in NSG mice, at higher potency than AS. Based on these preclinical data, we propose that AS, with its established low toxicity and low cost, and ART-838, with its higher potency and longer persistence in vivo, should be further developed toward integration into antileukemic regimens.


Subject(s)
Antineoplastic Agents/pharmacology , Artemisinins/pharmacology , Leukemia, Myeloid, Acute/prevention & control , Precursor Cell Lymphoblastic Leukemia-Lymphoma/prevention & control , Animals , Antimalarials/chemistry , Antimalarials/pharmacology , Apoptosis/drug effects , Artemisinins/chemistry , Blotting, Western , Cell Proliferation/drug effects , Drug Synergism , Drug Therapy, Combination , Humans , Immunoenzyme Techniques , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Antimicrob Agents Chemother ; 59(7): 3870-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25870074

ABSTRACT

Artemisinin-derived monomers and dimers inhibit human cytomegalovirus (CMV) replication in human foreskin fibroblasts (HFFs). The monomer artesunate (AS) inhibits CMV at micromolar concentrations, while dimers inhibit CMV replication at nanomolar concentrations, without increased toxicity in HFFs. We report on the variable anti-CMV activity of AS compared to the consistent and reproducible CMV inhibition by dimer 606 and ganciclovir (GCV). Investigation of this phenomenon revealed that the anti-CMV activity of AS correlated with HFFs synchronized to the G0/G1 stage of the cell cycle. In contact-inhibited serum-starved HFFs or cells arrested at early/late G1 with specific checkpoint regulators, AS and dimer 606 efficiently inhibited CMV replication. However, in cycling HFFs, in which CMV replication was productive, virus inhibition by AS was significantly reduced, but inhibition by dimer 606 and GCV was maintained. Cell cycle analysis in noninfected HFFs revealed that AS induced early G1 arrest, while dimer 606 partially blocked cell cycle progression. In infected HFFs, AS and dimer 606 prevented the progression of cell cycle toward the G1/S checkpoint. AS reduced the expression of cyclin-dependent kinases (CDK) 2, 4, and 6 in noninfected cycling HFFs, while the effect of dimer 606 on these CDKs was moderate. Neither compound affected CDK expression in noninfected contact-inhibited HFFs. In CMV-infected cells, AS activity correlated with reduced CDK2 levels. CMV inhibition by AS and dimer 606 also correlated with hypophosphorylation (activity) of the retinoblastoma protein (pRb). AS activity was strongly associated with pRb hypophosphorylation, while its reduced anti-CMV activity was marked by pRb phosphorylation. Roscovitine, a CDK2 inhibitor, antagonized the anti-CMV activities of AS and dimer 606. These data suggest that cell cycle modulation through CDKs and pRb might play a role in the anti-CMV activities of artemisinins. Proteins involved in this modulation may be identified and targeted for CMV inhibition.


Subject(s)
Antimalarials/pharmacology , Antiviral Agents/pharmacology , Artemisinins/pharmacology , Cell Cycle/drug effects , Cytomegalovirus/drug effects , Cells, Cultured , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/biosynthesis , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/biosynthesis , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/biosynthesis , Drug Resistance, Viral , Enzyme Inhibitors/pharmacology , Fibroblasts/virology , G1 Phase/drug effects , Ganciclovir/pharmacology , Humans , Phosphorylation , Purines/pharmacology , Resting Phase, Cell Cycle/drug effects , Retinoblastoma Protein/metabolism , Roscovitine , Virus Replication/drug effects
4.
Bioorg Med Chem Lett ; 25(2): 245-8, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25481079

ABSTRACT

Eight new artemisinin-derived trioxane dimer esters 5 have been prepared and tested for antimalarial efficacy in malaria-infected mice. At a single oral dose of only 6mg/kg combined with 18mg/kg of mefloquine, each of the dimer esters 5 outperformed the antimalarial drug artemether (2). The most efficacious dimer, dichlorobenzoate ester 5h, prolonged mouse survival past day 30 of infection with three of the four mice in this group having no detectable parasitemia and appearing and acting healthy on day 30.


Subject(s)
Antimalarials/administration & dosage , Antimalarials/chemistry , Artemisinins/administration & dosage , Artemisinins/chemistry , Malaria/drug therapy , Plasmodium berghei/drug effects , Administration, Oral , Animals , Dimerization , Mice
5.
Bioorg Med Chem Lett ; 24(11): 2440-3, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24775306

ABSTRACT

Several 2-carbon-linked trioxane dimer secondary alcohol carbonates 14 and thiocarbonates 15, combined with mefloquine and administered in a low single oral dose, prolonged the survival times of malaria-infected mice much more effectively than the popular monomeric antimalarial drug artemether plus mefloquine. Three dimer carbonates 14 and one dimer thiocarbonate 15 partially cured malaria-infected mice.


Subject(s)
Antimalarials/therapeutic use , Artemisinins/chemistry , Carbonates/therapeutic use , Malaria/drug therapy , Plasmodium berghei/drug effects , Sulfhydryl Compounds/therapeutic use , Animals , Antimalarials/administration & dosage , Antimalarials/chemistry , Carbonates/administration & dosage , Carbonates/chemistry , Dimerization , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship , Sulfhydryl Compounds/administration & dosage , Sulfhydryl Compounds/chemistry
6.
J Parasitol ; 100(4): 516-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24524228

ABSTRACT

Toxoplasmosis continues to be a public health problem, causing significant morbidity worldwide. Currently available medications, effective for acute toxoplasmosis, are nonetheless problematic due to adverse side effects in many patients. In addition, no medication is able to completely eradicate the parasite cysts, rendering infected individuals at risk for reactivation upon becoming immunocompromised. We examined the anti- T. gondii activity of 2 derivatives of artemisinin. In vitro metabolic stability tests revealed that both derivatives are stable in mouse plasma but only the thiazole CPH4-136 is stable in the presence of mouse microsomes. When tested in a mouse model of acute toxoplasmosis, both derivatives showed modest efficacy dependent upon the compound dose and the solvent vehicle. Finally, in a mouse model of chronic T. gondii infection, CPH4-136 at 3 mg/kg once per day for 32 days moderately but significantly decreased mouse brain cyst burden. Collectively, our findings suggest that artemisinin derivatives are partially effective in treating experimental T. gondii infections.


Subject(s)
Antiprotozoal Agents/pharmacology , Artemisinins/pharmacology , Toxoplasma/drug effects , Toxoplasmosis, Animal/drug therapy , Animals , Antiprotozoal Agents/chemistry , Artemether , Artemisinins/chemistry , Brain/parasitology , Chronic Disease , Drug Stability , Female , Mice , Mice, Inbred CBA , Thiazoles/chemistry , Toxoplasma/growth & development , Toxoplasmosis, Animal/mortality , Toxoplasmosis, Animal/parasitology
7.
Bioorg Med Chem Lett ; 24(5): 1285-9, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24508128

ABSTRACT

Sixteen new artemisinin-derived 2-carbon-linked trioxane dimers were prepared to study chemical structure/antimalarial activity relationships (SAR). Administering a very low single oral dose of only 5mg/kg of dimer secondary alcohol 6a or 6b plus 15 mg/kg of mefloquine hydrochloride prolonged the lives of Plasmodium berghei-infected mice to an average of 25 days after infection. This ACT chemotherapy result is of high medicinal significance because the antimalarial efficacy of the popular trioxane drug artemether (2) plus mefloquine under the same conditions was significantly lower (only 20 day average survival). NH-aryl carbamate derivatives 7e, 7i, and 7j of 2-carbon-linked dimer alcohol 6b also significantly outperformed artemether (2) in prolonging the survival times (25-27 days) of malaria-infected mice.


Subject(s)
Antimalarials/chemistry , Artemisinins/chemistry , Carbamates/chemistry , Animals , Antimalarials/chemical synthesis , Antimalarials/pharmacology , Artemether , Artemisinins/pharmacology , Artemisinins/therapeutic use , Carbamates/pharmacology , Carbamates/therapeutic use , Carbon/chemistry , Crystallography, X-Ray , Dimerization , Drug Therapy, Combination , Malaria/drug therapy , Malaria/mortality , Mefloquine/pharmacology , Mefloquine/therapeutic use , Mice , Molecular Conformation , Plasmodium berghei/drug effects , Structure-Activity Relationship , Survival Analysis
8.
Antimicrob Agents Chemother ; 58(2): 986-94, 2014.
Article in English | MEDLINE | ID: mdl-24277030

ABSTRACT

Conventional therapy for human cytomegalovirus (CMV) relies on inhibition of the viral DNA polymerase. Ganciclovir (GCV) is the first-line therapy, but when GCV-resistant strains emerge, alternative therapies are extremely limited and are associated with significant toxicities. Combination of anti-CMV agents that act on different targets or stages of virus replication has not been well studied, mostly because of the limited number of anti-CMV agents. We report our investigation of combinations of agents that inhibit CMV by targeting the viral DNA polymerase, cellular kinases, or other cell/virus mechanisms yet to be discovered. The selected compounds differed by the slopes of their dose-response curve: compounds with a slope of 1 (GCV) representing one target or noncooperativity and compounds with high slopes indicating positive cooperativity. Analysis of anti-CMV drug combinations using the Bliss model (which accounts for the slope parameter) distinguished between combinations with synergistic, antagonistic, and additive activities. The combination of GCV and foscarnet was slightly synergistic; strong synergism was found when GCV was used with artemisinin-derived monomers or dimers or the MEK inhibitor U0126. The combination of GCV and cardiac glycosides (digoxin, digitoxin, and ouabain) was additive. The monomeric artemisinin artesunate was synergistic when combined with U0126 or the multikinase inhibitor sunitinib. However, the combination of artemisinin-derived dimers (molecular weights, 606 and 838) and U0126 or sunitinib was antagonistic. These results demonstrate that members of a specific drug class show similar patterns of combination with GCV and that the slope parameter plays an important role in the evaluation of drug combinations. Lastly, antagonism between different classes of CMV inhibitors may assist in target identification and improve the understanding of CMV inhibition by novel compounds.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Fibroblasts/drug effects , Virus Replication/drug effects , Artemisinins/pharmacology , Butadienes/pharmacology , Cell Line , Cytomegalovirus/growth & development , Digitoxin/pharmacology , Digoxin/pharmacology , Drug Combinations , Drug Interactions , Fibroblasts/virology , Foscarnet/pharmacology , Ganciclovir/pharmacology , Humans , Indoles/pharmacology , Nitriles/pharmacology , Ouabain/pharmacology , Pyrroles/pharmacology , Sunitinib
9.
Expert Rev Endocrinol Metab ; 9(3): 283-292, 2014 May.
Article in English | MEDLINE | ID: mdl-30736167

ABSTRACT

BACKGROUND: Human cultured osteoblasts from pre-Ob, post-Ob or m-Ob express different mRNAs and respond to different hormones. AIMS: To test expression and hormonal modulation of VDR and 1OHase and 1,25D production in hObs. METHODS: hObs obtained from bone explants were prepared, treated and analyzed as before. RESULTS: (i) VDR and 1OHase were expressed in all hObs. (ii) 1,25D was produced similarly. (iii) Treatment with E2, DPN (ER agonist), but not PPT (ER agonist) increased VDR. (iv) These hormones increased 1OHase. (v) Vitamin D analog JKF and PTH 1-84 increased similarly mRNAs. (vi) Treatment with E2, DPN and PPT increased 1,25D. (vii) JKF and PTH increased similarly 1,25D. (viii) DNA synthesis and CK were stimulated by all hormones in hObs. CONCLUSIONS: Hormonal modulation of VDR and 1OHase and 1,25D production is important for bone physiology but their correlation, activity and bone physiology is not yet clear.

10.
Antimicrob Agents Chemother ; 57(9): 4208-14, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23774439

ABSTRACT

We report that the artemisinin-derived dimer diphenyl phosphate (DPP; dimer 838) is the most selective inhibitor of human cytomegalovirus (CMV) replication among a series of artemisinin-derived monomers and dimers. Dimer 838 was also unique in being an irreversible CMV inhibitor. The peroxide unit within artemisinins' chemical structures is critical to their activities, and its absence results in loss of anti-CMV activities. Surprisingly, the deoxy dimer of 838 retained modest anti-CMV activity, suggesting that the DPP moiety of dimer 838 contributes to its anti-CMV activities. DPP alone did not inhibit CMV replication, but triphenyl phosphate (TPP) had modest CMV inhibition, although its selectivity index was low. Artemisinin DPP derivatives dimer 838 and monomer diphenyl phosphate (compound 558) showed stronger CMV inhibition and a higher selectivity index than their analogs lacking the DPP unit. An add-on and removal assay revealed that removing DPP derivatives (compounds 558 and 838) but not the non-DPP backbones (artesunate and compound 606) at 24 h postinfection (hpi) already resulted in dominant CMV inhibition. CMV inhibition was fully irreversible with 838 and partially irreversible with 558, while non-DPP artemisinins were reversible inhibitors. While all artemisinin derivatives and TPP reduced the expression of the CMV immediate early 2 (IE2), UL44, and pp65 proteins at or after 48 hpi, only TPP inhibited the expression of both IE1 and IE2. Combination of a non-DPP dimer (compound 606) with TPP was synergistic in CMV inhibition, while ganciclovir and TPP were additive. Although TPP shared structural similarity with monomer DPP (compound 558) and dimer DPP (compound 838), its pattern of CMV inhibition was significantly different from the patterns of the artemisinins. These findings demonstrate that the DPP group contributes to the unique activities of compound 838.


Subject(s)
Antiviral Agents/pharmacology , Artemisinins/pharmacology , Cytomegalovirus/drug effects , DNA, Viral/antagonists & inhibitors , Organophosphates/chemistry , Virus Replication/drug effects , Antiviral Agents/chemistry , Artemisinins/chemistry , Cell Survival/drug effects , Cells, Cultured , Cytomegalovirus/growth & development , DNA, Viral/metabolism , Dimerization , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/virology , Genes, Reporter , HeLa Cells , Humans , Inhibitory Concentration 50 , Luciferases/genetics , Luciferases/metabolism , Quantitative Structure-Activity Relationship
11.
Bioorg Med Chem ; 21(13): 3702-7, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23673218

ABSTRACT

We recently reported the anti-cancer and anti-cytomegalovirus (CMV) activity of artemisinin-derived trioxane diphenylphosphate dimer 838. To probe the relationship between chemical structure and anti-CMV and anti-cancer activities, we now report synthesis and evaluation of a series of eight new dimer phosphate ester analogs of 838. This series of novel molecules was screened against human foreskin fibroblasts (HFFs) infected with CMV and against the human Jurkat T cell acute lymphoblastic leukemia cell line. This SAR study confirms the very high anti-CMV and anti-cancer potencies of dimer diphenyl phosphate ester 838 without its being toxic to normal cells.


Subject(s)
Antineoplastic Agents/chemistry , Antiviral Agents/chemistry , Artemisinins/chemistry , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , Leukemia/drug therapy , Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Artemisinins/pharmacology , Dimerization , Esters , Humans , Jurkat Cells , Phosphates/chemistry , Phosphates/pharmacology
12.
J Med Chem ; 56(6): 2630-41, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23425037

ABSTRACT

Malaria continues to be a difficult disease to eradicate largely because of the widespread populations it affects and the resistance that malaria parasites have developed against once very potent therapies. The natural product artemisinin has been a boon for antimalarial chemotherapy, as artemisinin combination therapy (ACT) has become the first line of chemotherapy. Because the threat of resistance is always on the horizon, it is imperative to continually identify new treatments, comprising both advanced analogues of all antimalarial drugs, especially artemisinin, and the exploration of novel combinations, ideally with distinct mechanisms of action. Here we report for the first time the synthesis of a series of two-carbon-linked artemisinin-derived dimers, their unique structural features, and demonstration of their antimalarial efficacy via single oral dose administration in two 60-day survival studies of Plasmodium berghei infected mice. Several of the new endoperoxide chemical entities consistently demonstrated excellent antimalarial efficacy, and combinations with two non-peroxide antimalarial drugs have been studied.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Artemisinins/chemistry , Artemisinins/pharmacology , Carbon/chemistry , Dimerization , Animals , Antimalarials/chemical synthesis , Artemisinins/chemical synthesis , Chemistry Techniques, Synthetic , Drug Interactions , Male , Mice , Models, Molecular , Molecular Conformation , Plasmodium berghei/drug effects , Stereoisomerism
13.
Medchemcomm ; 3(12): 1505-1511, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23205265

ABSTRACT

Parasitic diseases continue to have a devastating impact on human populations worldwide. Lack of effective treatments, the high cost of existing ones, and frequent emergence of resistance to these agents provide a strong argument for the development of novel therapies. Here we report the results of a hybrid approach designed to obtain a dual acting molecule that would demonstrate activity against a variety of parasitic targets. The antimalarial drug amodiaquine has been covalently joined with a nitric oxide-releasing furoxan to achieve multiple mechanisms of action. Using in vitro and ex vivo assays, the hybrid molecule shows activity against three parasites - Plasmodium falciparum, Schistosoma mansoni, and Ancylostoma ceylanicum.

14.
J Med Chem ; 55(17): 7892-9, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22891714

ABSTRACT

In only three steps and in 21-67% overall yields from the natural trioxane artemisinin, a series of 21 new trioxane C-10 thioacetals was prepared. Upon receiving a single oral dose of only 6 mg/kg of the monomeric trioxane 12c combined with 18 mg/kg of mefloquine hydrochloride, Plasmodium berghei-infected mice survived on average 29.8 days after infection. Two of the four mice in this group had no parasites detectable in their blood on day 30 after infection, and they behaved normally and appeared healthy. One of the mice had 11% blood parasitemia on day 30, and one mouse in this group died on day 29. Of high medicinal importance, the efficacy of this ACT chemotherapy is much better than (almost double) the efficacy under the same conditions using as a positive control the popular trioxane drug artemether plus mefloquine hydrochloride (average survival time of only 16.5 days).


Subject(s)
Acetals/therapeutic use , Antimalarials/therapeutic use , Artemisinins/therapeutic use , Malaria/drug therapy , Mefloquine/therapeutic use , Acetals/administration & dosage , Administration, Oral , Animals , Antimalarials/administration & dosage , Artemisinins/administration & dosage , Magnetic Resonance Spectroscopy , Mefloquine/administration & dosage , Mice , Mice, Inbred C57BL , Spectrometry, Mass, Fast Atom Bombardment
15.
Photochem Photobiol Sci ; 11(12): 1825-30, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22907250

ABSTRACT

We have previously demonstrated that the active vitamin D hormone, 1α,25-dihydroxyvitamin D3 (1,25(OH)(2)D(3)) and a cis-locked non-genomic analogue, protect skin cells from ultraviolet radiation (UV)-induced skin cell loss, DNA damage, immunosuppression and skin carcinogenesis. Herein, we used a low-calcaemic analogue, 1α-hydroxymethyl-16-ene-24,24-difluoro-25-hydroxy-26,27-bis-homovitamin D3 (QW), which has some transactivating capacity and is approximately 80-100 times less calcaemic than 1,25(OH)(2)D(3). QW (0.1-10 nM) significantly (p < 0.05-0.01) reduced UV-induced DNA lesions (CPD) in skin fibroblasts and keratinocytes and reduced cell death after UV exposure. Moreover, both 1,25(OH)(2)D(3) and QW (1 nM) were equally effective in significantly (p < 0.01) increasing levels of tumour suppressive p53 in cultured human keratinocytes at 3 and 6 h after UV exposure. In a hairless mouse model, both 1,25(OH)(2)D(3) and QW (22.8 ρmol cm(-2)) reduced UV-immunosuppression from 13.7 ± 1.3% to 0.1 ± 1.1% (p < 0.01) and 5.4 ± 1.5% (p < 0.01) respectively. When tested alongside 1,25(OH)(2)D(3) in a murine model of skin carcinogenesis. QW (22.8 ρmol cm(-2)) was not as effective as 1α,25(OH)(2)D(3) or a cis-locked analogue in reducing tumour formation or inhibiting tumour progression. It is possible that the dose required for QW to be effective as an anti-photocarcinogenesis agent in vivo is higher than for protection against the acute effects of UV exposure, but the dissociation between clear acute photo-protective effects and limited long term photoprotection is as yet unexplained.


Subject(s)
Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Keratinocytes/drug effects , Radiation-Protective Agents/pharmacology , Skin/drug effects , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/radiation effects , Cells, Cultured , DNA Damage/drug effects , DNA Damage/radiation effects , Humans , Immunosuppression Therapy , Keratinocytes/cytology , Keratinocytes/radiation effects , Mice , Mice, Hairless , Skin/radiation effects , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays
16.
Antimicrob Agents Chemother ; 56(7): 3508-15, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22547612

ABSTRACT

We previously reported that among a series of artemisinin-derived monomers and dimers, dimer diphenyl phosphate (838) was the most potent inhibitor of human cytomegalovirus (CMV) replication. Our continued investigation of a prototypic artemisinin monomer (artesunate [AS]) and dimer (838) now reveals that both compounds have specific activity against CMV but do not inhibit lytic replication of human herpesvirus 1 or 2 or Epstein-Barr virus. AS and 838 inhibited CMV replication during the first 24 h of the virus replication cycle, earlier than the time of ganciclovir (GCV) activities and prior to DNA synthesis. Neither compound inhibited virus entry. Quantification of DNA replication and virus yield revealed a similar level of inhibition by GCV, but AS and 838 had a 10-fold-higher inhibition of virus yield than of DNA replication, suggesting that artemisinins could inhibit CMV through multiple steps: a predominant early inhibition and possibly an additional step following DNA replication. During the strong early CMV inhibition, the transcription of immediate-early genes was not significantly downregulated, and viral protein expression was reduced only after 48 h. AS and GCV were reversible CMV inhibitors, but the inhibition of CMV replication by 838 was irreversible. Combinations of GCV and 838 as well as GCV and AS were highly synergistic. Finally, treatment with 838, but not AS, prior to CMV infection demonstrated strong anti-CMV activity. These findings illustrate the unique activities of dimer 838, including early and irreversible CMV inhibition, possibly by tight binding to its target.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Artemisinins/chemistry , Cytomegalovirus/drug effects , Virus Replication/drug effects , Animals , Blotting, Western , Cell Line , Chlorocebus aethiops , Cytomegalovirus/genetics , DNA Replication/drug effects , Fluorescent Antibody Technique , Humans , Vero Cells
17.
J Med Chem ; 55(1): 291-6, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22128829

ABSTRACT

Sixteen new anilide derivatives of the natural trioxane artemisinin were prepared and evaluated for antimalarial efficacy in Plasmodium berghei infected mice. Of these 16 new anilides administered orally as one 6 mg/kg dose combined with 18 mg/kg mefloquine hydrochloride, only sulfide 3-arteSanilide 12d was completely curative: on day 30 after infection, all mice in this group had no detectable parasitemia, gained as much weight as the uninfected control mice, and behaved normally.


Subject(s)
Anilides/chemical synthesis , Antimalarials/chemical synthesis , Malaria/drug therapy , Mefloquine/therapeutic use , Sulfides/chemical synthesis , Administration, Oral , Anilides/chemistry , Anilides/therapeutic use , Animals , Antimalarials/chemistry , Antimalarials/therapeutic use , Drug Therapy, Combination , Mice , Parasitemia/drug therapy , Plasmodium berghei , Stereoisomerism , Structure-Activity Relationship , Sulfides/chemistry , Sulfides/therapeutic use , Sulfones/chemical synthesis , Sulfones/chemistry , Sulfones/therapeutic use , Sulfoxides/chemical synthesis , Sulfoxides/chemistry , Sulfoxides/therapeutic use
18.
Org Lett ; 13(19): 5358-61, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21894954

ABSTRACT

Carboxylic acids were converted directly in 56-99% yields into methyl, n-butyl, and isopropyl ketones using excess cyanocuprates R(2)CuLi·LiCN. A substrate with a stereocenter α to the carboxylic acid was converted into ketones with very little loss of enantiomeric purity. A variety of functional groups were tolerated including aryl bromides. This direct transformation of a carboxylic acid into ketone with minimal tertiary alcohol formation is proposed to involve a relatively stable copper ketal tetrahedral intermediate.

19.
PLoS One ; 6(8): e24334, 2011.
Article in English | MEDLINE | ID: mdl-21904628

ABSTRACT

We recently reported that two artemisinin-derived dimers (dimer primary alcohol 606 and dimer sulfone 4-carbamate 832-4) are significantly more potent in inhibiting human cytomegalovirus (CMV) replication than artemisinin-derived monomers. In our continued evaluation of the activities of artemisinins in CMV inhibition, twelve artemisinin-derived dimers and five artemisinin-derived monomers were used. Dimers as a group were found to be potent inhibitors of CMV replication. Comparison of CMV inhibition and the slope parameter of dimers and monomers suggest that dimers are distinct in their anti-CMV activities. A deoxy dimer (574), lacking the endoperoxide bridge, did not have any effect on CMV replication, suggesting a role for the endoperoxide bridge in CMV inhibition. Differences in anti-CMV activity were observed among three structural analogs of dimer sulfone 4-carbamate 832-4 indicating that the exact placement and oxidation state of the sulfur atom may contribute to its anti-CMV activity. Of all tested dimers, artemisinin-derived diphenyl phosphate dimer 838 proved to be the most potent inhibitor of CMV replication, with a selectivity index of approximately 1500, compared to our previously reported dimer sulfone 4-carbamate 832-4 with a selectivity index of about 900. Diphenyl phosphate dimer 838 was highly active against a Ganciclovir-resistant CMV strain and was also the most active dimer in inhibition of cancer cell growth. Thus, diphenyl phosphate dimer 838 may represent a lead for development of a highly potent and safe anti-CMV compound.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Artemisinins/chemistry , Cytomegalovirus/drug effects , Cell Proliferation/drug effects , Cells, Cultured , HeLa Cells , Humans
20.
J Org Chem ; 76(19): 8121-5, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21853986

ABSTRACT

5-(Z)-alkylidene-2-thioxo-1,3-thiazolidin-4-ones (rhodanine derivatives) were prepared by reaction of in situ generated dithiocarbamates with recently reported racemic α-chloro-ß,γ-alkenoate esters. This multicomponent sequential transformation performed in one reaction flask represents a general route to this medicinally valuable class of sulfur/nitrogen heterocycles. Using this convergent procedure, we prepared an analogue of the drug epalrestat, an aldose reductase inhibitory rhodanine.


Subject(s)
Chemistry Techniques, Synthetic/methods , Enzyme Inhibitors/chemical synthesis , Rhodanine/chemical synthesis , Aldehyde Reductase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Rhodanine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...