Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Neurosurg Case Lessons ; 6(21)2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-37992311

RESUMO

BACKGROUND: Syringomyelia is defined as dilation of the spinal cord's central canal and is often precipitated by skull base herniation disorders. Although respiratory failure (RF) can be associated with skull base abnormalities due to brainstem compression, most cases occur in pediatric patients and quickly resolve. The authors report the case of an adult patient with global spinal syringomyelia and Chiari malformation who developed refractory RF after routine administration of diazepam. OBSERVATIONS: A 31-year-old female presented with malnutrition, a 1-month history of right-sided weakness, and normal respiratory dynamics. After administration of diazepam prior to magnetic resonance imaging (MRI), she suddenly developed hypercapnic RF followed MRI and required intubation. MRI disclosed a Chiari malformation type I and syrinx extending from C1 to the conus medullaris. After decompressive surgery, her respiratory function progressively returned to baseline status, although 22 months after initial benzodiazepine administration, the patient continues to require nocturnal ventilation. LESSONS: Administration of central nervous system depressants should be closely monitored in patients with extensive syrinx formation given the potential to exacerbate diminished central respiratory drive. Early identification of syrinx in the context of Chiari malformation and hemiplegia should prompt clinical suspicion of underlying respiratory compromise and early involvement of intensive care consultants.

2.
J Gen Physiol ; 155(6)2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-37130264

RESUMO

The large-conductance, Ca2+-, and voltage-activated K+ (BK) channel consists of the pore-forming α (BKα) subunit and regulatory ß and γ subunits. The γ1-3 subunits facilitate BK channel activation by shifting the voltage-dependence of channel activation toward the hyperpolarization direction by about 50-150 mV in the absence of Ca2+. We previously found that the intracellular C-terminal positively charged regions of the γ subunits play important roles in BK channel modulation. In this study, we found that the intracellular C-terminal region of BKα is indispensable in BK channel modulation by the γ1 subunit. Notably, synthetic peptide mimics of the γ1-3 subunits' C-terminal positively charged regions caused 30-50 mV shifts in BKα channel voltage-gating toward the hyperpolarization direction. The cationic cell-penetrating HIV-1 Tat peptide exerted a similar BK channel-activating effect. The BK channel-activating effects of the synthetic peptides were reduced in the presence of Ca2+ and markedly ablated by both charge neutralization of the Ca2+-bowl site and high ionic strength, suggesting the involvement of electrostatic interactions. The efficacy of the γ subunits in BK channel modulation was reduced by charge neutralization of the Ca2+-bowl site. However, BK channel modulation by the γ1 subunit was little affected by high ionic strength and the positively charged peptide remained effective in BK channel modulation in the presence of the γ1 subunit. These findings identify positively charged peptides as BK channel modulators and reveal a role for the Ca2+-bowl site in BK channel modulation by positively charged peptides and the C-terminal positively charged regions of auxiliary γ subunits.


Assuntos
Cálcio , Canais de Potássio Ativados por Cálcio de Condutância Alta , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Subunidades Proteicas/metabolismo , Ativação do Canal Iônico/fisiologia , Peptídeos/farmacologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo
3.
MedEdPORTAL ; 16: 10886, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32206702

RESUMO

Introduction: Sepsis is a major cause of morbidity and mortality in medicine and is managed in ICUs daily. Critical care training is a vital part of anesthesiology residency, and understanding the presentation, management, and treatment of septic shock is fundamental to intraoperative patient care. Methods: This simulation involved a 58-year-old man undergoing surgical debridement of a peripancreatic cyst with hemodynamic instability and septic shock. We conducted the simulation yearly for clinical anesthesia year 2 residents (n = 26) in 1-hour sessions with three to five learners at a time. The simulation covered the six Anesthesiology Milestones related to sepsis and septic shock as outlined in the Anesthesiology Milestones Project. Results: To date, 155 anesthesiology residents have completed the simulation. Commonly missed critical actions included failure to recognize the need for invasive lines, provide appropriate volumes of fluid resuscitation, inquire about blood cultures and antibiotics, and recognize the need for the patient to remain intubated. Most participants could appropriately diagnose and treat intraoperative septic shock, but all had moments of action or inaction to discuss and improve upon, and all learned from this scenario. Discussion: Simulation is an optimal way to practice the more rare and life-threatening clinical events in medicine. Even though septic shock is commonly managed in the ICU, it is relatively uncommon for it to develop acutely in the OR. This simulation is an effective and educational way to discuss the most recent sepsis/septic shock definition and review evidence-based guidelines for treatment.


Assuntos
Anestesiologia/educação , Treinamento com Simulação de Alta Fidelidade , Internato e Residência , Salas Cirúrgicas , Cisto Pancreático/cirurgia , Sepse/terapia , Cuidados Críticos , Educação Médica , Humanos
4.
Oncotarget ; 7(15): 19620-30, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-26934000

RESUMO

We generated afatinib resistant clones of H1975 lung cancer cells by transient exposure of established tumors to the drug and collected the re-grown tumors. Afatinib resistant H1975 clones did not exhibit any additional mutations in proto-oncogenes when compared to control clones. Afatinib resistant H1975 tumor clones expressed less PTEN than control clones and in afatinib resistant clones this correlated with increased basal SRC Y416, ERBB3 Y1289, AKT T308 and mTOR S2448 phosphorylation, decreased expression of ERBB1, ERBB2 and ERBB3 and increased total expression of c-MET, c-KIT and PDGFRß. Afatinib resistant clones were selectively killed by knock down of [ERBB3 + c-MET + c-KIT] but not by the individual or doublet knock down combinations. The combination of the ERBB1/2/4 inhibitor afatinib with the SRC family inhibitor dasatinib killed afatinib resistant H1975 cells in a greater than additive fashion; other drugs used in combination with dasatinib such as sunitinib, crizotinib and amufatinib were less effective. [Afatinib + dasatinib] treatment profoundly inactivated ERBB3, AKT and mTOR in the H1975 afatinib resistant clones and increased ATG13 S318 phosphorylation. Knock down of ATG13, Beclin1 or eIF2α strong suppressed killing by [ERBB3 + c-MET + c-KIT] knock down, but were only modestly protective against [afatinib + dasatinib] lethality. Thus afatinib resistant H1975 NSCLC cells rely on ERBB1- and SRC-dependent hyper-activation of residual ERBB3 and elevated signaling, due to elevated protein expression, from wild type c-MET and c-KIT to remain alive. Inhibition of ERBB3 signaling via both blockade of SRC and ERBB1 results in tumor cell death.


Assuntos
Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Quinazolinas/farmacologia , Receptor ErbB-3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/metabolismo , Afatinib , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Dasatinibe/administração & dosagem , Dasatinibe/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Camundongos Nus , Mutação , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-met/genética , Quinazolinas/administração & dosagem , Interferência de RNA , Receptor ErbB-3/genética , Transdução de Sinais/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases da Família src/genética
5.
Cancer Biol Ther ; 16(11): 1660-70, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26417912

RESUMO

The present studies sought to determine whether the lethality of the drug combination [sorafenib + sildenafil] could be enhanced by the anti-inflammatory agent celecoxib, using ovarian cancer and other tumor cell lines as models. Also, in a dose dependent fashion celecoxib enhanced [sorafenib + sildenafil] lethality in multiple ovarian cancer cell lines. In a dose dependent fashion celecoxib enhanced the ability of [sorafenib + sildenafil] to reduce expression of multiple chaperone proteins in parallel with lower levels of the drug efflux pumps ABCB1 and ABCG2. Over-expression of GRP78 and HSP27 maintained pump expression in the presence of drugs. Cell killing by the 3 drug combination was mediated by mitochondrial / caspase 9 -dependent apoptotic signaling and by RIP-1 / caspases 2 and 4 / AIF -dependent necroptotic signaling. Pre-treatment of intrinsically resistant primary ovarian cancer cells with [celecoxib + sorafenib + sildenafil] significantly enhanced tumor cell killing by a subsequent cisplatin exposure. Similar data were obtained in some cancer cell lines, but not all, using the related platinum containing drugs, oxaliplatin and carboplatin. As our prior publications have also validated in vivo the combinations of [celecoxib + sildenafil] and [sorafenib + sildenafil] as cytotoxic to multiple tumor cell types, combined with the present findings, we would argue that the combination of celecoxib/sorafenib/sildenafil should be explored in a new phase I trial in ovarian cancer.


Assuntos
Antineoplásicos/farmacologia , Celecoxib/farmacologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Niacinamida/análogos & derivados , Compostos de Fenilureia/farmacologia , Citrato de Sildenafila/farmacologia , Carboplatina/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Chaperona BiP do Retículo Endoplasmático , Feminino , Humanos , Concentração Inibidora 50 , Niacinamida/farmacologia , Compostos Organoplatínicos/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Oxaliplatina , Sorafenibe
6.
J Cell Physiol ; 230(5): 1115-27, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25303541

RESUMO

The present studies determined whether clinically relevant phosphodiesterase 5 (PDE5) inhibitors interacted with a clinically relevant NSAID, celecoxib, to kill tumor cells. Celecoxib and PDE5 inhibitors interacted in a greater than additive fashion to kill multiple tumor cell types. Celecoxib and sildenafil killed ex vivo primary human glioma cells as well as their associated activated microglia. Knock down of PDE5 recapitulated the effects of PDE5 inhibitor treatment; the nitric oxide synthase inhibitor L-NAME suppressed drug combination toxicity. The effects of celecoxib were COX2 independent. Over-expression of c-FLIP-s or knock down of CD95/FADD significantly reduced killing by the drug combination. CD95 activation was dependent on nitric oxide and ceramide signaling. CD95 signaling activated the JNK pathway and inhibition of JNK suppressed cell killing. The drug combination inactivated mTOR and increased the levels of autophagy and knock down of Beclin1 or ATG5 strongly suppressed killing by the drug combination. The drug combination caused an ER stress response; knock down of IRE1α/XBP1 enhanced killing whereas knock down of eIF2α/ATF4/CHOP suppressed killing. Sildenafil and celecoxib treatment suppressed the growth of mammary tumors in vivo. Collectively our data demonstrate that clinically achievable concentrations of celecoxib and sildenafil have the potential to be a new therapeutic approach for cancer.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias/patologia , Inibidores da Fosfodiesterase 5/farmacologia , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Animais , Autofagia/efeitos dos fármacos , Celecoxib , Linhagem Celular Tumoral , Sinergismo Farmacológico , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Humanos , Neoplasias Mamárias Experimentais/patologia , Camundongos Nus , Piperazinas , Purinas , Transdução de Sinais/efeitos dos fármacos , Citrato de Sildenafila
7.
Channels (Austin) ; 8(4): 308-20, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24642630

RESUMO

Recent studies have shown that transmembrane protein 16 A (TMEM16A) is a subunit of calcium-activated chloride channels (CACCs). Pharmacological agents have been used to probe the functional role of CACCs, however their effect on TMEM16A currents has not been systematically investigated. In the present study, we characterized the voltage and concentration-dependent effects of 2 traditional CACC inhibitors (niflumic acid and anthracene-9-carboxcylic acid) and 2 novel CACC / TMEM16A inhibitors (CACC(inh)A01 and T16A(inh)A01) on TMEM16A currents. The whole cell patch clamp technique was used to record TMEM16A currents from HE K 293 cells that stably expressed human TMEM16A. Niflumic acid, A-9-C, CACC(inh)A01 and T16A(inh)A01 inhibited TMEM16A currents with IC50 values of 12, 58, 1.7 and 1.5 µM, respectively, however, A-9-C and niflumic acid were less efficacious at negative membrane potentials. A-9-C and niflumic acid reduced the rate of TMEM16A tail current deactivation at negative membrane potentials and A-9-C (1 mM) enhanced peak TMEM16A tail current amplitude. In contrast, the inhibitory effects of CACC(inh)A01 and T16A(inh)A01 were independent of voltage and they did not prolong the rate of TMEM16A tail current deactivation. The effects of niflumic acid and A-9-C on TMEM16A currents were similar to previous observations on CACCs in vascular smooth muscle, strengthening the hypothesis that they are encoded by TMEM16A. However, CACC(inh)A01 and T16A(inh)A01 were more potent inhibitors of TMEM16A channels and their effects were not diminished at negative membrane potentials making them attractive candidates to interrogate the functional role of TMEM16A channels in future studies.


Assuntos
Canais de Cloreto/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Ácido Niflúmico/farmacologia , Anoctamina-1 , Antracenos/farmacologia , Células HEK293 , Humanos , Cinética , Técnicas de Patch-Clamp , Pirimidinas/farmacologia , Tiazóis/farmacologia
8.
Clin Cancer Res ; 19(11): 3068-77, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23553849

RESUMO

PURPOSE: Ganetespib is a novel inhibitor of the heat shock protein 90 (Hsp90), a chaperone protein critical to tumor growth and proliferation. In this phase II study, we evaluated the activity and tolerability of ganetespib in previously treated patients with non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN: Patients were enrolled into cohort A (mutant EGFR), B (mutant KRAS), or C (no EGFR or KRAS mutations). Patients were treated with 200 mg/m(2) ganetespib by intravenous infusion once weekly for 3 weeks followed by 1 week of rest, until disease progression. The primary endpoint was progression-free survival (PFS) at 16 weeks. Secondary endpoints included objective response (ORR), duration of treatment, tolerability, median PFS, overall survival (OS), and correlative studies. RESULTS: Ninety-nine patients with a median of 2 prior systemic therapies were enrolled; 98 were assigned to cohort A (n = 15), B (n = 17), or C (n = 66), with PFS rates at 16 weeks of 13.3%, 5.9%, and 19.7%, respectively. Four patients (4%) achieved partial response (PR); all had disease that harbored anaplastic lymphoma kinase (ALK) gene rearrangement, retrospectively detected by FISH (n = 1) or PCR-based assays (n = 3), in crizotinib-naïve patients enrolled to cohort C. Eight patients (8.1%) experienced treatment-related serious adverse events (AE); 2 of these (cardiac arrest and renal failure) resulted in death. The most common AEs were diarrhea, fatigue, nausea, and anorexia. CONCLUSIONS: Ganetespib monotherapy showed a manageable side effect profile as well as clinical activity in heavily pretreated patients with advanced NSCLCs, particularly in patients with tumors harboring ALK gene rearrangement.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Triazóis/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Quinase do Linfoma Anaplásico , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Feminino , Rearranjo Gênico , Genótipo , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Estadiamento de Neoplasias , Receptores Proteína Tirosina Quinases/genética , Resultado do Tratamento , Triazóis/administração & dosagem , Triazóis/efeitos adversos
9.
J Biol Chem ; 286(51): 43913-43924, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22033924

RESUMO

Ivermectin is an anthelmintic drug that works by activating glutamate-gated chloride channel receptors (GluClRs) in nematode parasites. GluClRs belong to the Cys-loop receptor family that also includes glycine receptor (GlyR) chloride channels. GluClRs and A288G mutant GlyRs are both activated by low nanomolar ivermectin concentrations. The crystal structure of the Caenorhabditis elegans α GluClR complexed with ivermectin has recently been published. Here, we probed ivermectin sensitivity determinants on the α1 GlyR using site-directed mutagenesis and electrophysiology. Based on a mutagenesis screen of transmembrane residues, we identified Ala288 and Pro230 as crucial sensitivity determinants. A comparison of the actions of selamectin and ivermectin suggested the benzofuran C05-OH was required for high efficacy. When taken together with docking simulations, these results supported a GlyR ivermectin binding orientation similar to that seen in the GluClR crystal structure. However, whereas the crystal structure shows that ivermectin interacts with the α GluClR via H-bonds with Leu218, Ser260, and Thr285 (α GluClR numbering), our data indicate that H-bonds with residues homologous to Ser260 and Thr285 are not important for high ivermectin sensitivity or direct agonist efficacy in A288G α1 GlyRs or three other GluClRs. Our data also suggest that van der Waals interactions between the ivermectin disaccharide and GlyR M2-M3 loop residues are unimportant for high ivermectin sensitivity. Thus, although our results corroborate the ivermectin binding orientation as revealed by the crystal structure, they demonstrate that some of the binding interactions revealed by this structure do not pertain to other highly ivermectin-sensitive Cys-loop receptors.


Assuntos
Canais de Cloreto/química , Ivermectina/química , Receptores de Glicina/química , Animais , Antiparasitários/química , Antiparasitários/farmacologia , Benzofuranos/química , Caenorhabditis elegans/metabolismo , Cristalografia por Raios X/métodos , Cisteína/química , Eletrofisiologia/métodos , Glicina/química , Ligação de Hidrogênio , Ivermectina/análogos & derivados , Ivermectina/farmacologia , Mutagênese Sítio-Dirigida , Mutação
10.
Aviat Space Environ Med ; 81(8): 719-27, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20681231

RESUMO

INTRODUCTION: Fatigue in military aviation is a significant safety and operational problem resulting in diminished alertness and performance. Research demonstrates that stimulant medications maintain alertness and performance in sleep-deprived aircrew. However, these studies control many of the variables present during combat operations. Few studies have evaluated fatigue or the factors and effects associated with stimulant use in fighter aircrew during combat operations. METHODS: The study consisted of three questionnaires administered to 29 deployed F-1 5E aircrew participants. An initial questionnaire compiled demographic and sleep behavior data. Pre- and postflight questionnaires for each sortie collected substance use, fatigue, and physical symptoms data. Regression analysis identified variables associated with in-flight stimulant use. RESULTS: Surveys were completed for 111 sorties averaging 7.6 h in duration. Stimulants were used on 35% of sorties an average of 2.8 h after takeoff. Stimulant use was associated with a decrease in in-flight and postflight fatigue without significant differences in postflight symptoms. Sorties airborne during the circadian trough, longer sortie durations, and preflight hypnotic use displayed statistically significant associations with in-flight stimulant use. CONCLUSIONS: In-flight stimulants decrease fatigue during combat operations without significant postflight symptoms. During combat, stimulants were used earlier, at lower doses, and on shorter sorties than previously thought. The factors associated with stimulant use are potentially modifiable by improving training and aircrew scheduling practices. Furthermore, current policies authorizing stimulant use, based primarily on sortie duration, should also consider hypnotic use, inconsistent aircrew scheduling, and circadian disruption.


Assuntos
Compostos Benzidrílicos/uso terapêutico , Estimulantes do Sistema Nervoso Central/uso terapêutico , Dextroanfetamina/uso terapêutico , Fadiga/prevenção & controle , Militares , Guerra , Adulto , Medicina Aeroespacial , Cafeína/uso terapêutico , Ritmo Circadiano , Humanos , Hipnóticos e Sedativos/uso terapêutico , Masculino , Modafinila , Nicotina/uso terapêutico , Tolerância ao Trabalho Programado
11.
Bioconjug Chem ; 21(5): 903-10, 2010 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-20420372

RESUMO

Membrane receptors for steroid hormones are currently a subject of considerable debate. One approach to selectively target these putative receptors has been to couple ligands to substances that restrict cell permeability. Using this approach, an analogue of the estrogen receptor ligand 4-hydroxytamoxifen was attached to fluorescent dyes with differing degrees of predicted cell permeability. The conjugates bound to estrogen receptor in vitro, but all three conjugates, including one predicted to be cell-impermeable, inhibited estradiol-induced transcriptional activation. Fluorescence microscopy revealed cytoplasmic localization for all three conjugates. We further characterized a 4-hydroxytamoxifen analogue conjugated to a BODIPY fluorophore in breast cancer cell lines. Those experiments suggested a similar, but not identical, mode of action to 4-hydroxytamoxifen, as the fluorescent conjugate was equally effective at inhibiting proliferation of both tamoxifen-sensitive and tamoxifen-resistant breast cancer cell lines. While these findings point to significant complicating factors in designing steroid hormone mimics targeted to the plasma membrane, the results also reveal a possible new direction for designing estrogen receptor modulators.


Assuntos
Antagonistas de Estrogênios/química , Antagonistas de Estrogênios/farmacologia , Corantes Fluorescentes/química , Receptores de Estrogênio/metabolismo , Tamoxifeno/análogos & derivados , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Antagonistas de Estrogênios/síntese química , Antagonistas de Estrogênios/farmacocinética , Corantes Fluorescentes/síntese química , Humanos , Tamoxifeno/síntese química , Tamoxifeno/química , Tamoxifeno/farmacocinética , Tamoxifeno/farmacologia
12.
Support Care Cancer ; 18(4): 423-31, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19568773

RESUMO

PURPOSE: Aprepitant was shown previously to be effective for prevention of chemotherapy-induced nausea and vomiting (CINV) with moderately emetogenic chemotherapy (MEC) in breast cancer patients receiving an anthracycline and cyclophosphamide (AC)-based regimen. This study assessed aprepitant in patients receiving a broad range of MEC regimens with a variety of tumor types. METHODS: This phase III, randomized, gender-stratified, double-blind trial enrolled patients with confirmed malignancies, naïve to MEC or highly emetogenic chemotherapy, who were scheduled to receive a single dose of at least one MEC agent. Patients received an aprepitant triple-therapy regimen (aprepitant, ondansetron, and dexamethasone) or a control regimen (ondansetron and dexamethasone) administered orally. Primary and key secondary efficacy endpoints were proportions of patients with no vomiting and complete response (no vomiting and no rescue medication), respectively, during the 120 h post-chemotherapy. RESULTS: Of 848 randomized patients, 77% were female, and 52% received non-AC-based antineoplastic regimens. Significantly, more patients in the aprepitant group achieved no vomiting and complete response, regardless of whether they received AC or non-AC regimens, in the 120 h after chemotherapy. Overall, the incidences of adverse events were generally similar in the aprepitant (62.8%) and control groups (67.2%). CONCLUSIONS: The aprepitant regimen provided superior efficacy in the treatment of CINV in a broad range of patients receiving MEC (non-AC or AC) in both no vomiting and complete response endpoints. Aprepitant was generally well tolerated. These results show the benefit of including aprepitant as part of the standard antiemetic regimen for cancer patients receiving MEC.


Assuntos
Antieméticos/uso terapêutico , Antineoplásicos/efeitos adversos , Morfolinas/uso terapêutico , Náusea/prevenção & controle , Vômito/prevenção & controle , Adulto , Idoso , Antieméticos/administração & dosagem , Antieméticos/efeitos adversos , Antineoplásicos/uso terapêutico , Aprepitanto , Dexametasona/administração & dosagem , Dexametasona/efeitos adversos , Dexametasona/uso terapêutico , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Morfolinas/administração & dosagem , Morfolinas/efeitos adversos , Náusea/induzido quimicamente , Neoplasias/tratamento farmacológico , Ondansetron/administração & dosagem , Ondansetron/efeitos adversos , Ondansetron/uso terapêutico , Estudos Prospectivos , Resultado do Tratamento , Vômito/induzido quimicamente
13.
Curr Pharm Des ; 13(23): 2350-67, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17692006

RESUMO

The glycine receptor (GlyR) Cl(-) channel belongs to the cysteine-loop family of ligand-gated ion channel receptors. It is best known for mediating inhibitory neurotransmission in motor and sensory reflex circuits of the spinal cord, although glycinergic synapses are also present in the brain stem, cerebellum and retina. Extrasynaptic GlyRs are widely distributed throughout the central nervous system and they are also found in sperm and macrophages. A total of 5 GlyR subunits (alpha1-4 and beta) have been identified. Embryonic receptors comprise alpha2 homomers whereas adult receptors comprise predominantly alpha beta heteromers in a 2:3 stoichiometry. Notably, the alpha3 subunit is present in synaptic GlyRs that mediate inhibitory neurotransmission onto spinal nociceptive neurons. These receptors are specifically inhibited by inflammatory mediators, implying a role for alpha3-containing GlyRs in inflammatory pain sensitisation. Because molecules that increase GlyR current may have clinical potential as muscle relaxant and peripheral analgesic drugs, this review focuses on the molecular pharmacology of GlyR potentiating substances. Of all GlyR potentiating substances identified to date, we conclude that 5HT(3)R antagonists such as tropisetron offer the most promise as therapeutic lead compounds. However, one problem is that that virtually all known GlyR potentiating compounds, including tropisetron analogues, lack specificity for the GlyR. Another is that almost nothing is known about the pharmacological properties of alpha3-containing GlyRs, which is the subtype of choice for targeting by novel antinociceptive agents. These issues need to be addressed before GlyR-specific therapeutics can be developed.


Assuntos
Canais de Cloreto/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Moduladores de Transporte de Membrana/farmacologia , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptores de Glicina/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Sequência de Aminoácidos , Analgésicos/farmacologia , Animais , Canais de Cloreto/química , Canais de Cloreto/metabolismo , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Ligantes , Moduladores de Transporte de Membrana/química , Moduladores de Transporte de Membrana/uso terapêutico , Dados de Sequência Molecular , Estrutura Molecular , Neurônios/metabolismo , Conformação Proteica , Subunidades Proteicas , Receptores de Glicina/química , Receptores de Glicina/metabolismo , Relação Estrutura-Atividade
14.
J Biol Chem ; 282(23): 16803-10, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17405880

RESUMO

The pore structural changes associated with Cys-loop receptor gating are currently the subject of considerable interest. Several functional approaches have shown that surface exposure of pore-lining side chains does not change significantly during activation. However, a disulfide trapping study on alpha1(T6'C)beta1(T6'C) gamma-aminobutyric acid type A (GABA(A)) receptors (GABA(A)Rs), which showed that adjacent beta subunits cross-link in the open state only, concluded that channel gating is mediated by beta subunits contra-rotating through a summed angle of approximately 120 degrees. Such a large rotation is not easily reconciled with other evidence. The present study initially sought to investigate an observation that appeared inconsistent with the rotation model: namely that alpha1(T6'C)beta1(T6'C) GABA(A)Rs expressed in HEK293 cells form 6' cysteine-mediated disulfide bonds in the closed state. On the basis of electrophysiological and Western blotting experiments, we conclude that adjacent beta(T6'C) subunits dimerise efficiently in the closed state via cross-links between their respective 6' cysteines and that this locks the channels closed. This questions the beta subunit contra-rotation model of activation and raises the question of how the closed state cross-links form. We propose that beta subunit 6' cysteines move into sufficiently close proximity for disulfide formation via relatively large amplitude random thermal motions that appear to be a unique feature of beta subunits. Because dimerized channels are locked closed, we conclude either that the spontaneous beta subunit movements or asymmetries in the movements of adjacent beta subunits during activation are essential for pore opening. Our results identify a novel feature of GABA(A)R gating that may be important for understanding its activation mechanism.


Assuntos
Cisteína/metabolismo , Receptores de GABA-A/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Linhagem Celular , Humanos , Mutagênese Sítio-Dirigida , Ratos , Receptores de GABA-A/química , Receptores de GABA-A/genética , Receptores de GABA-A/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA