Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Clin Transl Sci ; 15(6): 1507-1518, 2022 06.
Article in English | MEDLINE | ID: mdl-35301810

ABSTRACT

This study aimed to define the clinically relevant supratherapeutic dose of rilzabrutinib, an oral Bruton tyrosine kinase (BTK) inhibitor, and evaluate potential effects of therapeutic and supratherapeutic exposures on cardiac repolarization in healthy subjects. This was a two-part phase I study (anzctr.org.au ACTRN12618001036202). Part A was a randomized, open-label, three-period, single-dose crossover study (n = 12) with rilzabrutinib 100 mg ± ritonavir 100 mg or rilzabrutinib 1200 mg. Part B was a randomized, double-blind, placebo-controlled, four-way, single-dose crossover study (n = 39) with matched placebo, rilzabrutinib 400 mg ± ritonavir 100 mg, or moxifloxacin (positive control). Primary objectives: part A - pharmacokinetics (PK) of rilzabrutinib ± ritonavir, safety, and optimal dose for Part B; Part B - effect of rilzabrutinib therapeutic and supratherapeutic concentration on electrocardiogram (ECG) parameters. ECGs and PK samples were serially recorded before and post-dose. In part A, rilzabrutinib 100 mg + ritonavir led to 17-fold area under the concentration-time curve (AUC0-∞ ) and 7-fold maximum plasma concentration (Cmax ) increases over rilzabrutinib alone. Rilzabrutinib 1200 mg was discontinued due to mild-to-moderate gastrointestinal intolerance. In Part B, rilzabrutinib 400 mg + ritonavir increased rilzabrutinib mean AUC0-∞ from 454 to 3800 ng h/mL and Cmax from 144 to 712 ng/mL. The concentration-QTc relationship was slightly negative, shallow (-0.01 ms/ng/mL [90% CI -0.016 to -0.001]), and an effect >10 ms on QTcF could be excluded within the observed range of plasma concentrations, up to 2500 ng/mL. Safety was similar to other studies of rilzabrutinib. In conclusion, rilzabrutinib, even at supratherapeutic doses, had no clinically relevant effects on ECG parameters, including the QTc interval.


Subject(s)
Protein Kinase Inhibitors , Ritonavir , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Electrocardiography , Healthy Volunteers , Heart Rate , Humans , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Ritonavir/therapeutic use
3.
Immunohorizons ; 5(7): 581-589, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326199

ABSTRACT

The expression of Bruton tyrosine kinase (BTK) in B cells and innate immune cells provides essential downstream signaling for BCR, Fc receptors, and other innate immune cell pathways. The topical covalent BTK inhibitor PRN473 has shown durable, reversible BTK occupancy with rapid on-rate and slow off-rate binding kinetics and long residence time, resulting in prolonged, localized efficacy with low systemic exposure in vivo. Mechanisms of PRN473 include inhibition of IgE (FcεR)-mediated activation of mast cells and basophils, IgG (FcγR)-mediated activation of monocytes, and neutrophil migration. In vivo, oral PRN473 was efficacious and well tolerated in the treatment of canine pemphigus foliaceus. In this study, we evaluated in vitro selectivity and functionality, in vivo skin Ab inflammatory responses, and systemic pharmacology with topically administered PRN473. Significant dose-dependent inhibition of IgG-mediated passive Arthus reaction in rats was observed with topical PRN473 and was maintained when given 16 h prior to challenge, reinforcing extended activity with once-daily administration. Similarly, topical PRN473 resulted in significant dose-dependent inhibition of the mouse passive cutaneous anaphylaxis IgE-mediated reaction. Multiday treatment with topical PRN473 in rodents resulted in low-to-no systemic accumulation, suggesting that efficacy was mainly due to localized exposure. Reduced skin Ab inflammatory activity was also confirmed with oral PRN473. These preclinical studies provide a strong biologic basis for targeting innate immune cell responses locally in the skin, with rapid onset of action following once-daily topical PRN473 administration and minimal systemic exposure. Dose-dependent inhibition in these preclinical models of immune-mediated skin diseases support future clinical studies.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Arthus Reaction , Passive Cutaneous Anaphylaxis , Protein Kinase Inhibitors , Skin Diseases , Animals , Female , Humans , Mice , Rats , Administration, Cutaneous , Administration, Oral , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Arthus Reaction/drug therapy , Arthus Reaction/immunology , Arthus Reaction/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Passive Cutaneous Anaphylaxis/drug effects , Protein Kinase Inhibitors/administration & dosage , Skin/drug effects , Skin/immunology , Skin/pathology , Skin Diseases/drug therapy , Skin Diseases/immunology , Skin Diseases/pathology
4.
Cancer Res ; 69(10): 4277-85, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19417133

ABSTRACT

Reactivation of silenced tumor suppressor genes by 5-azacytidine (Vidaza) and its congener 5-aza-2'-deoxycytidine (decitabine) has provided an alternate approach to cancer therapy. We have shown previously that these drugs selectively and rapidly induce degradation of the maintenance DNA methyltransferase (DNMT) 1 by a proteasomal pathway. Because the toxicity of these compounds is largely due to their incorporation into DNA, it is critical to explore novel, nonnucleoside compounds that can effectively reactivate the silenced genes. Here, we report that a quinoline-based compound, designated SGI-1027, inhibits the activity of DNMT1, DNMT3A, and DNMT3B as well M. SssI with comparable IC(50) (6-13 micromol/L) by competing with S-adenosylmethionine in the methylation reaction. Treatment of different cancer cell lines with SGI-1027 resulted in selective degradation of DNMT1 with minimal or no effects on DNMT3A and DNMT3B. At a concentration of 2.5 to 5 micromol/L (similar to that of decitabine), complete degradation of DNMT1 protein was achieved within 24 h without significantly affecting its mRNA level. MG132 blocked SGI-1027-induced depletion of DNMT1, indicating the involvement of proteasomal pathway. Prolonged treatment of RKO cells with SGI-1027 led to demethylation and reexpression of the silenced tumor suppressor genes P16, MLH1, and TIMP3. Further, this compound did not exhibit significant toxicity in a rat hepatoma (H4IIE) cell line. This study provides a novel class of DNA hypomethylating agents that have the potential for use in epigenetic cancer therapy.


Subject(s)
Azacitidine/toxicity , DNA Methylation/drug effects , Gene Silencing/drug effects , Genes, Tumor Suppressor/drug effects , Quinolines/toxicity , Animals , Breast Neoplasms , Carcinoma, Hepatocellular , Cell Line, Tumor , Colonic Neoplasms , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/drug effects , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/isolation & purification , DNA Methyltransferase 3A , DNA Modification Methylases/metabolism , Female , HeLa Cells , Humans , Liver Neoplasms , Mice , Reverse Transcriptase Polymerase Chain Reaction , DNA Methyltransferase 3B
5.
Am J Hematol ; 82(11): 981-5, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17696208

ABSTRACT

The silencing of tumor suppressor genes associated with increased DNA methylation of the promoter regions is a frequent observation in many forms of cancer. Reactivation of these genes using pharmacological inhibitors of DNA methyltransferase such as 5-aza-2'-deoxycytidine (decitabine) is a worthwhile therapeutic goal. The effectiveness and tolerability of low-dose intravenous and subcutaneous decitabine regimens to demethylate and reactivate expression of the methylated gamma-globin gene in baboons and in patients with sickle cell disease led to successful trials of low-dose regimens of this drug in patients with myelodysplastic syndrome. Since these low-dose regimens are well-tolerated with minimal toxicity, they are suitable for chronic dosing to maintain promoter hypomethylation and expression of target genes. The development of an orally administered therapy using DNA methyltransferase inhibitors would facilitate such chronic approaches to therapy. We tested the ability of decitabine and a new salt derivative, decitabine mesylate, to reactivate the methylated gamma-globin gene in baboons when administered orally. Our results demonstrate that oral administration of these drugs at doses 17-34 times optimal subcutaneous doses of decitabine reactivates fetal hemoglobin, demethylates the epsilon- and gamma-globin gene promoters, and increases histone acetylation of these promoters in baboons (Papio anubis).


Subject(s)
Azacitidine/analogs & derivatives , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Fetal Hemoglobin , Gene Silencing/drug effects , Administration, Oral , Animals , Azacitidine/administration & dosage , Azacitidine/pharmacokinetics , Decitabine , Fetal Hemoglobin/drug effects , Fetal Hemoglobin/genetics , Gene Expression Regulation/drug effects , Globins/drug effects , Globins/genetics , Globins/metabolism , Papio anubis
6.
Cancer Res ; 67(13): 6400-8, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17616700

ABSTRACT

The major goal of epigenetic therapy is to reverse aberrant promoter hypermethylation and restore normal function of tumor suppressor genes by the use of chromatin-modifying drugs. Decitabine, or 5-aza-2'-deoxycytidine (5-aza-CdR), is a well-characterized drug that is now Food and Drug Administration approved for the treatment of myelodysplastic syndrome. Although 5-aza-CdR is an extremely potent inhibitor of DNA methylation, it is subject to degradation by hydrolytic cleavage and deamination by cytidine deaminase. We show that short oligonucleotides containing a 5-aza-CdR can also inhibit DNA methylation in cancer cells at concentrations comparable with 5-aza-CdR. Detailed studies with S110, a dinucleotide, showed that it works via a mechanism similar to that of 5-aza-CdR after incorporation of its aza-moiety into DNA. Stability of the triazine ring in aqueous solution was not improved in the S110 dinucleotide; however, deamination by cytidine deaminase was dramatically decreased. This is the first demonstration of the use of short oligonucleotides to provide effective delivery and cellular uptake of a nucleotide drug and protection from enzymatic degradation. This approach may pave the way for more stable and potent inhibitors of DNA methylation as well as provide means for improving existing therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Azacitidine/analogs & derivatives , Drug Delivery Systems , Genetic Therapy/methods , Oligonucleotides/pharmacology , Azacitidine/administration & dosage , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Methylation , Decitabine , Dose-Response Relationship, Drug , Epigenesis, Genetic , Humans , Models, Chemical , Myelodysplastic Syndromes/drug therapy , Oligonucleotides/chemistry , Triazines/chemistry , Urinary Bladder Neoplasms/drug therapy
7.
Org Lett ; 4(26): 4587-90, 2002 Dec 26.
Article in English | MEDLINE | ID: mdl-12489936

ABSTRACT

[reaction: see text] Synthesis of glycosyl cyanides was optimized with a new catalyst system. Reduction of tri-O-acetyl-beta-L-fucopyranosyl cyanide with Pd-hydrogen, in the presence of Ac(2)O and Boc(2)O, gave N-protected-mono- and -di-(2,3,4-tri-O-acetyl-beta-L-fucopyranosylmethyl)-amines, which allow for the syntheses of small cluster oligosaccharide mimetics of fucopyranosylomethyl-substituted ureas. From di-(2,3,4-tri-O-acetyl-beta-L-fucopyranosylmethyl) amine was also prepared a carbamoyl chloride as a potentially useful synthon for preparation of more complex C-glycosidic conjugates.


Subject(s)
Fucose/chemistry , Molecular Mimicry , Oligosaccharides/chemical synthesis , Catalysis , Charcoal/chemistry , Cyanides/chemistry , Oxidation-Reduction , Palladium/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...