Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-38687022

ABSTRACT

BACKGROUND AND OBJECTIVES: Postoperative intracranial hemorrhage (POH) is a serious neurosurgical complication occurring in approximately 1.4% of patients after intracranial tumor resection. The convention across the United States is to maintain an immediate postoperative systolic blood pressure (SBP) of < 140 mm Hg to minimize this risk; however, this SBP goal lacks support in the literature despite widespread adoption. This study aims to investigate the safety of SBP liberalization to 160 mm Hg in the immediate postoperative setting after intracranial tumor resection. METHODS: A retrospective review was conducted on consecutive patients, aged 18 to 75 years, undergoing craniotomy for intracranial tumor resection from October 2020 until June 2023. Data were gathered from the electronic medical record per Institutional Review Board guidelines regarding demographics, operative details, perioperative vital signs, resource utilization, and complications. Pharmaceutical prices and insurance charges were approximated from costs provided by the institution's pharmacy. POH was defined as symptomatic hemorrhage within 48 hours requiring intervention. RESULTS: The study included 147 patients, with 104 in the liberalized cohort (SBP <160 mm Hg) and 43 in the standard cohort (SBP <140 mm Hg). The average age was 54.5 ± 14.9 years and 57.6 ± 10.6 years in the liberalized and standard groups, respectively (P = .23). Intensive care unit and hospital length of stay were not significantly different between groups. The liberalized group used $81.88 ± $280.19 (95% CI $53.01-$110.75) on as-needed antihypertensive medications vs $108.39 ± $215.91 (95% CI $75.96-$140.82) in the standard (P = .29), with significantly lower labetalol (P = .04). There was no POH in either cohort. CONCLUSION: Liberalization of SBP goals to <160 mm Hg appears safe in the immediate postoperative period after craniotomy for tumor resection without an increased POH risk. Liberalized SBP parameters may allow reduced antihypertensive medication usage, thereby avoiding excess hospital cost and medication side effects.

2.
Cells ; 13(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474373

ABSTRACT

The mechanistic target of rapamycin (mTOR) is a serine/threonine kinase that functions via its discrete binding partners to form two multiprotein complexes, mTOR complex 1 and 2 (mTORC1 and mTORC2). Rapamycin-sensitive mTORC1, which regulates protein synthesis and cell growth, is tightly controlled by PI3K/Akt and is nutrient-/growth factor-sensitive. In the brain, mTORC1 is also sensitive to neurotransmitter signaling. mTORC2, which is modulated by growth factor signaling, is associated with ribosomes and is insensitive to rapamycin. mTOR regulates stem cell and cancer stem cell characteristics. Aberrant Akt/mTOR activation is involved in multistep tumorigenesis in a variety of cancers, thereby suggesting that the inhibition of mTOR may have therapeutic potential. Rapamycin and its analogues, known as rapalogues, suppress mTOR activity through an allosteric mechanism that only suppresses mTORC1, albeit incompletely. ATP-catalytic binding site inhibitors are designed to inhibit both complexes. This review describes the regulation of mTOR and the targeting of its complexes in the treatment of cancers, such as glioblastoma, and their stem cells.


Subject(s)
Glioblastoma , Neoplastic Stem Cells , Sirolimus , Humans , Glioblastoma/metabolism , Intercellular Signaling Peptides and Proteins/therapeutic use , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Neoplastic Stem Cells/metabolism
3.
Anticancer Res ; 44(1): 13-22, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38160007

ABSTRACT

BACKGROUND/AIM: Brain metastasis (BM) is a complex multi-step process involving various immune checkpoint proteins. Mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinases 1/2 (ERK1/2), and signal transducer and activator of transcription 3 (STAT3) are implicated in tumorigenesis and are critical upstream regulators of Programmed Death Ligand 1 (PD-L1), an immunotherapy target. Tumor suppressor p53, dysregulated in cancers, regulates STAT3 and ERK1/2 signaling. This study examined the roles of STAT3, MAPK and p53 status in BM initiation and maintenance. MATERIALS AND METHODS: Twenty-six BM, with various primary malignancies, were used (IRB-approved) to determine mutant p53 (p53mt), pSTAT3Tyr705, pERK1/2Thr202/Tyr204, and PD-L1 expression using immunohistochemistry. cDNA microarray was used for gene expression analysis. Brain-metastatic breast cancer cells (MDA-MB-231) were treated with STAT3 (NSC74859) or MAPK/ERK1/2 (U0126) inhibitors in regular or astrocytic media. ERK1/2 pathway was assessed using western blotting, and cell proliferation and migration were determined using MTT and scratch-wound assays, respectively. RESULTS: pSTAT3Tyr705 and pERK1/2Thr202/Tyr204 were expressed at tumor margins, whereas p53mt and PD-L1 were uniformly expressed, with significant overlap between expression of these proteins. Gene expression analysis identified alterations in 18 p53- and 32 STAT3- or MAPK-associated genes contributing to dysregulated immune responses and cell cycle regulation. U0126 and NSC74859 reduced pERK1/2Thr202/Tyr204 expression. Cell proliferation decreased following each treatment (p≤0.01). Migration stagnated following U0126 treatment in astrocytic media (p≤0.01). CONCLUSION: Activation of STAT3 and ERK1/2 promotes BM and provides compelling evidence for use of STAT3, ERK1/2 and p53 status as potential immunotherapeutic targets in BM.


Subject(s)
B7-H1 Antigen , Brain Neoplasms , Humans , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Extracellular Signal-Regulated MAP Kinases/metabolism , Brain Neoplasms/genetics , Cell Line, Tumor
4.
Bioorg Med Chem ; 25(12): 3077-3086, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28416100

ABSTRACT

Histone acetylation is an extensively investigated post-translational modification that plays an important role as an epigenetic regulator. It is controlled by histone acetyl transferases (HATs) and histone deacetylases (HDACs). The overexpression of HDACs and consequent hypoacetylation of histones have been observed in a variety of different diseases, leading to a recent focus of HDACs as attractive drug targets. The natural product largazole is one of the most potent natural HDAC inhibitors discovered so far and a number of largazole analogs have been prepared to define structural requirements for its HDAC inhibitory activity. However, previous structure-activity relationship studies have heavily investigated the macrocycle region of largazole, while there have been only limited efforts to probe the effect of various zinc-binding groups (ZBGs) on HDAC inhibition. Herein, we prepared a series of largazole analogs with various ZBGs and evaluated their HDAC inhibition and cytotoxicity. While none of the analogs tested were as potent or selective as largazole, the Zn2+-binding affinity of each ZBG correlated with HDAC inhibition and cytotoxicity. We expect that our findings will aid in building a deeper understanding of the role of ZBGs in HDAC inhibition as well as provide an important basis for the future development of new largazole analogs with non-thiol ZBGs as novel therapeutics for cancer.


Subject(s)
Depsipeptides/chemistry , Depsipeptides/pharmacology , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Thiazoles/chemistry , Thiazoles/pharmacology , Zinc/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Depsipeptides/chemical synthesis , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Humans , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/metabolism , Structure-Activity Relationship , Thiazoles/chemical synthesis
5.
ACS Med Chem Lett ; 5(8): 905-10, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25147612

ABSTRACT

Largazole is a potent and class I-selective histone deacetylase (HDAC) inhibitor purified from marine cyanobacteria and was demonstrated to possess antitumor activity. Largazole employs a unique prodrug strategy, via a thioester moiety, to liberate the bioactive species largazole thiol. Here we report alternate prodrug strategies to modulate the pharmacokinetic and pharmacodynamics profiles of new largazole-based compounds. The in vitro effects of largazole analogues on cancer cell proliferation and enzymatic activities of purified HDACs were comparable to the natural product. However, in vitro and in vivo histone hyperacetylation in HCT116 cells and implanted tumors, respectively, showed differences, particularly in the onset of action and oral bioavailability. These results indicate that, by employing a different approach to disguise the "warhead" moiety, the functional consequence of these prodrugs can be significantly modulated. Our data corroborate the role of the pharmacokinetic properties of this class of compounds to elicit the desired and timely functional response.

6.
Bioorg Med Chem Lett ; 24(16): 3728-31, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25070421

ABSTRACT

Largazole is a potent class I selective histone deacetylase (HDAC) inhibitor. The majority of largazole analogues to date have modified the thiazole-thiazoline and the warhead moiety. In order to elucidate class I-specific structure-activity relationships, a series of analogues with modifications in the valine or the linker region were prepared and evaluated for their class I isoform selectivity. The inhibition profile showed that the C2 position of largazole has an optimal steric requirement for efficient HDAC inhibition and that substitution of the trans-alkene in the linker with an aromatic group results in complete loss of activity. This data will aid the design of class I isoform selective HDAC inhibitors.


Subject(s)
Depsipeptides/pharmacology , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Thiazoles/pharmacology , Depsipeptides/chemical synthesis , Depsipeptides/chemistry , Dose-Response Relationship, Drug , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Molecular Structure , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...