Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Int J Mol Sci ; 24(14)2023 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-37511180

RESUMEN

Despite a multimodal radical treatment, mortality of advanced epithelial ovarian cancer (AEOC) remains high. Host-related factors, such as systemic inflammatory response and its interplay with the immune system, remain underexplored. We hypothesized that the prognostic impact of this response could vary between patients undergoing primary debulking surgery (PDS) and those undergoing interval debulking surgery (IDS). Therefore, we evaluated the outcomes of two surgical groups of newly diagnosed AEOC patients according to the neutrophil, monocyte and platelet to lymphocyte ratios (NLR, MLR, PLR), taking median ratio values as cutoffs. In the PDS group (n = 61), low NLR and PLR subgroups showed significantly better overall survival (not reached (NR) vs. 72.7 months, 95% confidence interval [CI]: 40.9-95.2, p = 0.019; and NR vs. 56.1 months, 95% CI: 40.9-95.2, p = 0.004, respectively) than those with high values. Similar results were observed in progression free survival. NLR and PLR-high values resulted in negative prognostic factors, adjusting for residual disease, BRCA1/2 status and stage (HR 2.48, 95% CI: 1.03-5.99, p = 0.043, and HR 2.91, 95% CI: 1.11-7.64, p = 0.03, respectively). In the IDS group (n = 85), ratios were not significant prognostic factors. We conclude that NLR and PLR may have prognostic value in the PDS setting, but none in IDS, suggesting that time of surgery can modulate the prognostic impact of baseline complete blood count (CBC).


Asunto(s)
Neutrófilos , Neoplasias Ováricas , Humanos , Femenino , Carcinoma Epitelial de Ovario , Monocitos , Proteína BRCA1 , Pronóstico , Procedimientos Quirúrgicos de Citorreducción , Estudios Retrospectivos , Proteína BRCA2 , Linfocitos , Neoplasias Ováricas/diagnóstico
2.
Gynecol Oncol ; 172: 121-129, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37030280

RESUMEN

BACKGROUND: The open-label, single-arm, multicenter ORZORA trial (NCT02476968) evaluated the efficacy and safety of maintenance olaparib in patients with platinum-sensitive relapsed ovarian cancer (PSR OC) who had tumor BRCA mutations (BRCAm) of germline (g) or somatic (s) origin or non-BRCA homologous recombination repair mutations (HRRm) and were in response to their most recent platinum-based chemotherapy after ≥2 lines of treatment. METHODS: Patients received maintenance olaparib capsules (400 mg twice daily) until disease progression. Prospective central testing at screening determined tumor BRCAm status and subsequent testing determined gBRCAm or sBRCAm status. Patients with predefined non-BRCA HRRm were assigned to an exploratory cohort. The co-primary endpoints were investigator-assessed progression-free survival (PFS; modified Response Evaluation Criteria in Solid Tumors v1.1) in BRCAm and sBRCAm cohorts. Secondary endpoints included health-related quality of life (HRQoL) and tolerability. RESULTS: 177 patients received olaparib. At the primary data cut-off (17 April 2020), the median follow-up for PFS in the BRCAm cohort was 22.3 months. The median PFS (95% CI) in BRCAm, sBRCAm, gBRCAm and non-BRCA HRRm cohorts was 18.0 (14.3-22.1), 16.6 (12.4-22.2), 19.3 (14.3-27.6) and 16.4 (10.9-19.3) months, respectively. Most patients with BRCAm reported improvements (21.8%) or no change (68.7%) in HRQoL and the safety profile was as expected. CONCLUSIONS: Maintenance olaparib had similar clinical activity in PSR OC patients with sBRCAm and those with any BRCAm. Activity was also observed in patients with a non-BRCA HRRm. ORZORA further supports use of maintenance olaparib in all patients with BRCA-mutated, including sBRCA-mutated, PSR OC.


Asunto(s)
Antineoplásicos , Neoplasias Ováricas , Humanos , Femenino , Antineoplásicos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Calidad de Vida , Reparación del ADN por Recombinación , Estudios Prospectivos , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Carcinoma Epitelial de Ovario/genética , Ftalazinas/efectos adversos , Mutación , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Mutación de Línea Germinal
3.
Eur J Cancer ; 182: 3-14, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36706655

RESUMEN

AIM: To describe patient characteristics, effectiveness and safety in a real-world population treated with niraparib in the Spanish expanded-access programme. PATIENTS AND METHODS: This retrospective observational study included women with platinum-sensitive recurrent high-grade serous ovarian cancer who received maintenance niraparib within the Spanish niraparib expanded-access programme. Eligible patients had received ≥2 previous lines of platinum-containing therapy, remained platinum-sensitive after the penultimate line of platinum and had responded to the most recent platinum-containing therapy. Niraparib dosing was at the treating physician's discretion (300 mg/day fixed starting dose or individualised starting dose [ISD] according to baseline body weight and platelet count). Safety, impact of dose adjustments, patient characteristics and effectiveness were analysed using data extracted from medical records. RESULTS: Among 316 eligible patients, 80% had BRCA wild-type tumours and 66% received an ISD. Median niraparib duration was 7.8 months. The most common adverse events typically occurred within 3 months of starting niraparib. Median progression-free survival was 8.6 (95% confidence interval [CI] 7.6-10.0) months. One- and 2-year overall survival rates were 86% (95% CI 81-89%) and 65% (95% CI 59-70%), respectively. Dose interruptions, dose reductions, haematological toxicities and asthenia/fatigue were less common with ISD than fixed starting dose niraparib, but progression-free survival was similar irrespective of dosing strategy. Subsequent therapy included platinum in 71% of patients who received further treatment. CONCLUSION: Outcomes in this large real-world dataset of niraparib-treated patients are consistent with phase III trials, providing reassuring evidence of the tolerability and activity of niraparib maintenance therapy for platinum-sensitive recurrent ovarian cancer. GOV REGISTRATION: NCT04546373.


Asunto(s)
Neoplasias Ováricas , Humanos , Femenino , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Indazoles , Recurrencia Local de Neoplasia/tratamiento farmacológico
4.
Cancers (Basel) ; 14(18)2022 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-36139574

RESUMEN

Background: Despite impressive progression-free survival (PFS) results from PARP inhibitors (PARPi) in ovarian cancer, concerns about their effect on post-progression treatment outcomes have recently arisen, particularly when administered in the relapsed setting. Overlapping mechanisms of resistance between PARPi and platinum have been described, and optimal therapies upon progression to PARPi are unknown. We communicate real-world data (RWD) on outcomes of subsequent chemotherapy upon progression to PARPi used as maintenance in ovarian cancer relapses, particularly focusing on platinum rechallenge, according to BRCA status. Methods: Data from high-grade serous or endometrioid ovarian cancer patients who received subsequent chemotherapy after progression to maintenance PARPi in the relapsed setting, in 16 Catalan hospitals between August 2016 and April 2021, and who were followed-up until July 2021, were included. Endpoints were overall response rate (ORR), and PFS and overall survival (OS) measured from the subsequent chemotherapy starting date. Results: 111 patients were included [46 (41.4%) presented pathological BRCA1/2 mutations, 8 (7.5%) in other homologous recombination-related genes]. Sixty-four patients (57.7%) had received two prior chemotherapy lines, including the one immediately prior to PARPi. PARPi were niraparib (n = 60, 54.1%), olaparib (n = 49, 44.1%), and rucaparib (n = 2, 1.8%). A total of 81 patients remained platinum-sensitive (PS population) after progression to PARPi (when progression-free interval [PFI] was >6 months after the last cycle of prior platinum) [median PFI 12.0 months (interquartile range, IQR, 8.8−17.1)]. Of those, 74 were treated with subsequent platinum regimens, with the following results: ORR of 41.9%, median PFS (mPFS) of 6.6 months (95% CI 6−9.2), and median OS (mOS) of 20.6 months (95% CI 13.6−28.9). Analysis of these 74 patients according to BRCA status showed that PFIs for BRCA mutant and non BRCA-mutant patients were 13.6 [IQR11.2−22.2] and 10.3 [IQR 7.4−14.9] months, respectively (p = 0.010); ORR were 40.0% versus 43.6%, respectively; Rates of progression (as best response) to subsequent platinum were 45.7% versus 17.9%, respectively (p = 0.004); mPFS and mOS were 3.5 (95% CI 2.5−8.6) versus 7.5 months (95% CI 6.5−10.1, p = 0.03), and 16.4 (95% CI 9.3−27.5) versus 24.2 months (95% CI 17.2−NR, p = 0.036), respectively. Conclusion: This is the largest series of real-world data on ovarian cancer patients retreated with platinum in the post-PARPi scenario, separately analyzing BRCA mutant and non-mutant patients, to our knowledge. In our platinum-sensitive population, rechallenge with platinum after progression upon PARPi in the 3rd or later lines for ovarian cancer relapses shows relevant ORR and similar PFS outcomes to historical series of the prePARPi era. However, BRCA mutant patients presented significantly higher rates of progression under subsequent platinum and worse survival outcomes associated with subsequent platinum than non-BRCA-mutant patients.

5.
J Neurosci ; 42(19): 3879-3895, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35387872

RESUMEN

Calcium is an important second messenger regulating a bioenergetic response to the workloads triggered by neuronal activation. In embryonic mouse cortical neurons using glucose as only fuel, activation by NMDA elicits a strong workload (ATP demand)-dependent on Na+ and Ca2+ entry, and stimulates glucose uptake, glycolysis, pyruvate and lactate production, and oxidative phosphorylation (OXPHOS) in a Ca2+-dependent way. We find that Ca2+ upregulation of glycolysis, pyruvate levels, and respiration, but not glucose uptake, all depend on Aralar/AGC1/Slc25a12, the mitochondrial aspartate-glutamate carrier, component of the malate-aspartate shuttle (MAS). MAS activation increases glycolysis, pyruvate production, and respiration, a process inhibited in the presence of BAPTA-AM, suggesting that the Ca2+ binding motifs in Aralar may be involved in the activation. Mitochondrial calcium uniporter (MCU) silencing had no effect, indicating that none of these processes required MCU-dependent mitochondrial Ca2+ uptake. The neuronal respiratory response to carbachol was also dependent on Aralar, but not on MCU. We find that mouse cortical neurons are endowed with a constitutive ER-to-mitochondria Ca2+ flow maintaining basal cell bioenergetics in which ryanodine receptors, RyR2, rather than InsP3R, are responsible for Ca2+ release, and in which MCU does not participate. The results reveal that, in neurons using glucose, MCU does not participate in OXPHOS regulation under basal or stimulated conditions, while Aralar-MAS appears as the major Ca2+-dependent pathway tuning simultaneously glycolysis and OXPHOS to neuronal activation.SIGNIFICANCE STATEMENT Neuronal activation increases cell workload to restore ion gradients altered by activation. Ca2+ is involved in matching increased workload with ATP production, but the mechanisms are still unknown. We find that glycolysis, pyruvate production, and neuronal respiration are stimulated on neuronal activation in a Ca2+-dependent way, independently of effects of Ca2+ as workload inducer. Mitochondrial calcium uniporter (MCU) does not play a relevant role in Ca2+ stimulated pyruvate production and oxygen consumption as both are unchanged in MCU silenced neurons. However, Ca2+ stimulation is blunt in the absence of Aralar, a Ca2+-binding mitochondrial carrier component of Malate-Aspartate Shuttle (MAS). The results suggest that Ca2+-regulated Aralar-MAS activation upregulates glycolysis and pyruvate production, which fuels mitochondrial respiration, through regulation of cytosolic NAD+/NADH ratio.


Asunto(s)
Ácido Aspártico , Fosforilación Oxidativa , Adenosina Trifosfato/metabolismo , Animales , Ácido Aspártico/metabolismo , Calcio/metabolismo , Glucosa/metabolismo , Glucólisis , Malatos/metabolismo , Ratones , Neuronas/fisiología , Piruvatos/metabolismo
6.
Hum Mol Genet ; 30(24): 2441-2455, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34274972

RESUMEN

Charcot-Marie-Tooth (CMT) disease is a neuropathy that lacks effective therapy. CMT patients show degeneration of peripheral nerves, leading to muscle weakness and loss of proprioception. Loss of mitochondrial oxidative phosphorylation proteins and enzymes of the antioxidant response accompany degeneration of nerves in skin biopsies of CMT patients. Herein, we followed a drug-repurposing approach to find drugs in a Food and Drug Administration-approved library that could prevent development of CMT disease in the Gdap1-null mouse model. We found that the antibiotic florfenicol is a mitochondrial uncoupler that prevents the production of reactive oxygen species and activates respiration in human GDAP1-knockdown neuroblastoma cells and in dorsal root ganglion neurons of Gdap1-null mice. Treatment of CMT-affected Gdap1-null mice with florfenicol has no beneficial effect in the course of the disease. However, administration of florfenicol, or the antioxidant MitoQ, to pre-symptomatic GDAP1-null mice prevented weight gain and ameliorated the motor coordination deficiencies that developed in the Gdap1-null mice. Interestingly, both florfenicol and MitoQ halted the decay in mitochondrial and redox proteins in sciatic nerves of Gdap1-null mice, supporting that oxidative damage is implicated in the etiology of the neuropathy. These findings support the development of clinical trials for translation of these drugs for treatment of CMT patients.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth , Animales , Enfermedad de Charcot-Marie-Tooth/tratamiento farmacológico , Enfermedad de Charcot-Marie-Tooth/genética , Humanos , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética
7.
PLoS Biol ; 19(5): e3001252, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33983919

RESUMEN

The mitochondrial ATP synthase emerges as key hub of cellular functions controlling the production of ATP, cellular signaling, and fate. It is regulated by the ATPase inhibitory factor 1 (IF1), which is highly abundant in neurons. Herein, we ablated or overexpressed IF1 in mouse neurons to show that IF1 dose defines the fraction of active/inactive enzyme in vivo, thereby controlling mitochondrial function and the production of mitochondrial reactive oxygen species (mtROS). Transcriptomic, proteomic, and metabolomic analyses indicate that IF1 dose regulates mitochondrial metabolism, synaptic function, and cognition. Ablation of IF1 impairs memory, whereas synaptic transmission and learning are enhanced by IF1 overexpression. Mechanistically, quenching the IF1-mediated increase in mtROS production in mice overexpressing IF1 reduces the increased synaptic transmission and obliterates the learning advantage afforded by the higher IF1 content. Overall, IF1 plays a key role in neuronal function by regulating the fraction of ATP synthase responsible for mitohormetic mtROS signaling.


Asunto(s)
Mitocondrias/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Proteínas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Encéfalo/metabolismo , Línea Celular , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , ATPasas de Translocación de Protón Mitocondriales/fisiología , Cultivo Primario de Células , Proteínas/fisiología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteína Inhibidora ATPasa
8.
Anticancer Drugs ; 32(1): 88-94, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33332891

RESUMEN

The objective of this observational study was to evaluate the efficacy and safety of duloxetine in a cohort of 100 cancer survivors with chemotherapy-induced peripheral neurotoxicity (CIPN). CIPN was graded employing the TNSc and the NCI-CTCv4. The Patient Global Impression of Change (PGIC) scale measured the efficacy of duloxetine (1: no benefit; to 7: excellent response). A clinically meaningful response was considered a PGIC > 4. Median age was 62 (29-81) years and 42% were male. CIPN was graded as grades 1, 2 and 3 in 20, 66, and 14% of patients, respectively. Median time to duloxetine initiation was 6 (1-63) months after chemotherapy. Fifty-seven patients early dropped out from duloxetine, due to lack of efficacy (20%) or side effects (37%). Male patients more frequently discontinued duloxetine due to lack of efficacy (35.7 vs. 8.6% P = 0.001). PGIC scores were higher in female patients (4 vs. 1, P = 0.001), taxane-treated patients (4 vs. 1, P = 0.042) and with short-lasting (<6 months) CIPN (4 vs. 1, P = 0.008). Patients with long-lasting CIPN had a higher rate of adverse events (47 vs. 27%, P = 0.038) and discontinuation (54.8 vs. 45.1%, P = 0.023). In the multivariate analysis, female gender and short-lasting CIPN were independently associated with a favorable response to duloxetine. Low tolerability, male gender, and long-lasting CIPN significantly limited duloxetine use in daily practice setting. A minority of cancer survivors with CIPN treated with duloxetine had a meaningful CIPN improvement, and tolerability was overall low. Female gender and short-term CIPN were independently associated with a favorable response to duloxetine.


Asunto(s)
Supervivientes de Cáncer/estadística & datos numéricos , Clorhidrato de Duloxetina/efectos adversos , Neoplasias/tratamiento farmacológico , Síndromes de Neurotoxicidad/patología , Enfermedades del Sistema Nervioso Periférico/patología , Calidad de Vida , Inhibidores de Captación de Serotonina y Norepinefrina/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/patología , Síndromes de Neurotoxicidad/etiología , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Pronóstico , Estudios Prospectivos
9.
EJC Suppl ; 15: 96-103, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33240448

RESUMEN

AIM: The description of rare malignant ovarian tumours and the most suitable treatments. Alternative therapies different from intravenous chemotherapy are also explained. METHODS: Literature review and ongoing trial information have been used to elaborate this guide. RESULTS: Each ovarian cancer type must be identified and treated properly from diagnostic to surgery, adjuvant treatment and metastatic disease. Hormonotherapy can be useful as an alternative treatment, especially in low-grade ovarian cancer and endometrioid subtype. Tumour characterisation is appropriated for treatment selection when targeted therapy is indicated. MEK inhibitors, tyrosine-kinase inhibitors, EGFR inhibitors, therapies against integrins, antibody-drug conjugates and other strategies are described. Antiangiogenics, PARP inhibitors and immunotherapy are discussed in other parts of this publication. CONCLUSION: Different ovarian cancer types must receive the appropriated treatment. Alternative therapies may be evaluated beyond the standard therapy, frequently in a clinical trial, and an individualised molecular study may help to find the best treatment.

10.
Neurochem Int ; 131: 104541, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31472174

RESUMEN

The elevated energy demands in the brain are fulfilled mainly by glucose catabolism. In highly polarized neurons, about 10-50% of mitochondria are transported along microtubules using mitochondrial-born ATP to locations with high energy requirements. In this report, we have investigated the impact of Aralar deficiency on mitochondrial transport in cultured cortical neurons. Aralar/slc25a12/AGC1 is the neuronal isoform of the aspartate-glutamate mitochondrial carrier, a component of the malate-aspartate shuttle (MAS) which plays an important role in redox balance, which is essential to maintain glycolytic pyruvate supply to neuronal mitochondria. Using live imaging microscopy we observed that the lack of Aralar does not affect the number of moving mitochondria nor the Ca2+-induced stop, the only difference being a 10% increase in mitochondrial velocity in Aralar deficient neurons. Therefore, we evaluated the possible fuels used in each case by studying the relative contribution of oxidative phosphorylation and glycolysis to mitochondrial movement using specific inhibitors. We found that the ATP synthase inhibitor oligomycin caused a smaller inhibition of mitochondrial movement in Aralar-KO than control neurons, whereas the glycolysis inhibitor iodoacetate had similar effects in neurons from both genotypes. In line with these findings, the decrease in cytosolic ATP/ADP ratio caused by oligomycin was more pronounced in control than in Aralar-KO neurons, but no differences were observed with iodoacetate. Oligomycin effect was reverted by aralar re-expression in knock out cultures. As mitochondrial movement is not reduced in Aralar-KO neurons, these results suggest that these neurons may use an additional pathway for mitochondria movement and ATP/ADP ratio maintenance.


Asunto(s)
Agrecanos/metabolismo , Corteza Cerebral/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Neuronas/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Agrecanos/genética , Animales , Ácido Aspártico/metabolismo , Transporte Biológico Activo , Calcio/metabolismo , Calcio/farmacología , Corteza Cerebral/citología , Glucólisis/efectos de los fármacos , Inmunohistoquímica , Malatos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Transporte de Membrana Mitocondrial/genética , Fosforilación Oxidativa , Ácido Pirúvico/metabolismo
11.
Int J Gynecol Cancer ; 29(7): 1141-1147, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31420414

RESUMEN

INTRODUCTION: The PENELOPE trial evaluated pertuzumab added to chemotherapy for biomarker-selected platinum-resistant ovarian cancer. As previously reported, pertuzumab did not statistically significantly improve progression-free survival (primary end point: HR 0.74, 95% CI 0.50 to 1.11), although results in the paclitaxel and gemcitabine cohorts suggested activity. Here, we report final overall survival and patient-reported outcomes. PATIENTS AND METHODS: Eligible patients had ovarian carcinoma that progressed during/within 6 months of completing ≥4 platinum cycles, low tumor human epidermal growth factor receptor 3 (HER3) mRNA expression, and ≤2 prior chemotherapy lines. Investigators selected single-agent topotecan, gemcitabine or weekly paclitaxel before patients were randomized to either placebo or pertuzumab (840→420 mg every 3 weeks), stratified by selected chemotherapy, prior anti-angiogenic therapy, and platinum-free interval. Final overall survival analysis (key secondary end point) was pre-specified after 129 deaths. Patient-reported outcomes (secondary end point) were assessed at baseline and every 9 weeks until disease progression. RESULTS: At database lock (June 9, 2016), 130 (83%) of 156 randomized patients had died. Median follow-up was 27 months in the pertuzumab arm versus 26 months in the control arm. In the intent-to-treat population there was no overall survival difference between treatment arms (stratified HR 0.90, 95% CI 0.61 to 1.32; p=0.60). Results in subgroups defined by stratification factors indicated heterogeneity similar to previous progression-free survival results. Updated safety was similar to previously published results. Compliance with patient-reported outcomes questionnaire completion was >75% for all validated patient-reported outcomes measures. Pertuzumab demonstrated neither beneficial nor detrimental effects on patient-reported outcomes compared with placebo, except for increased diarrhea symptoms. DISCUSSION: Consistent with the primary results, adding pertuzumab to chemotherapy for low tumor HER3 mRNA-expressing platinum-resistant ovarian cancer did not improve overall survival, but showed trends in some cohorts. Except for increased diarrhea symptoms, pertuzumab had no impact on patient-reported outcomes. ClinicalTrials.gov: ClinicalTrials.gov: NCT01684878.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , ARN Mensajero/biosíntesis , Receptor ErbB-3/genética , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Inmunológicos/administración & dosificación , Carcinoma Epitelial de Ovario/enzimología , Carcinoma Epitelial de Ovario/genética , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Método Doble Ciego , Resistencia a Antineoplásicos , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Paclitaxel/administración & dosificación , Medición de Resultados Informados por el Paciente , Supervivencia sin Progresión , ARN Mensajero/genética , Receptor ErbB-3/biosíntesis , Topotecan/administración & dosificación , Gemcitabina
12.
Int J Mol Sci ; 18(8)2017 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-28758950

RESUMEN

There have been no major improvements in the overall survival of ovarian cancer patients in recent decades. Even though more accurate surgery and more effective treatments are available, the mortality rate remains high. Given the differences in origin and the heterogeneity of these tumors, research to elucidate the signaling pathways involved is required. The Transforming Growth Factor (TGFß) family controls different cellular responses in development and cell homeostasis. Disruption of TGFß signaling has been implicated in many cancers, including ovarian cancer. This article considers the involvement of TGFß in ovarian cancer progression, and reviews the various mechanisms that enable the TGFß signaling pathway to control ovarian cancer cell proliferation. These mechanistic explanations support the therapeutic use of TGFß inhibitors in ovarian cancer, which are currently in the early phases of development.


Asunto(s)
Proliferación Celular , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Femenino , Humanos , Proteínas de Neoplasias/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Factor de Crecimiento Transformador beta/genética
13.
Biochim Biophys Acta ; 1857(8): 1158-1166, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27060251

RESUMEN

Glutamate elicits Ca(2+) signals and workloads that regulate neuronal fate both in physiological and pathological circumstances. Oxidative phosphorylation is required in order to respond to the metabolic challenge caused by glutamate. In response to physiological glutamate signals, cytosolic Ca(2+) activates respiration by stimulation of the NADH malate-aspartate shuttle through Ca(2+)-binding to the mitochondrial aspartate/glutamate carrier (Aralar/AGC1/Slc25a12), and by stimulation of adenine nucleotide uptake through Ca(2+) binding to the mitochondrial ATP-Mg/Pi carrier (SCaMC-3/Slc25a23). In addition, after Ca(2+) entry into the matrix through the mitochondrial Ca(2+) uniporter (MCU), it activates mitochondrial dehydrogenases. In response to pathological glutamate stimulation during excitotoxicity, Ca(2+) overload, reactive oxygen species (ROS), mitochondrial dysfunction and delayed Ca(2+) deregulation (DCD) lead to neuronal death. Glutamate-induced respiratory stimulation is rapidly inactivated through a mechanism involving Poly (ADP-ribose) Polymerase-1 (PARP-1) activation, consumption of cytosolic NAD(+), a decrease in matrix ATP and restricted substrate supply. Glutamate-induced Ca(2+)-activation of SCaMC-3 imports adenine nucleotides into mitochondria, counteracting the depletion of matrix ATP and the impaired respiration, while Aralar-dependent lactate metabolism prevents substrate exhaustion. A second mechanism induced by excitotoxic glutamate is permeability transition pore (PTP) opening, which critically depends on ROS production and matrix Ca(2+) entry through the MCU. By increasing matrix content of adenine nucleotides, SCaMC-3 activity protects against glutamate-induced PTP opening and lowers matrix free Ca(2+), resulting in protracted appearance of DCD and protection against excitotoxicity in vitro and in vivo, while the lack of lactate protection during in vivo excitotoxicity explains increased vulnerability to kainite-induced toxicity in Aralar +/- mice. This article is part of a Special Issue entitled 'EBEC 2016: 19th European Bioenergetics Conference, Riva del Garda, Italy, July 2-6, 2016', edited by Prof. Paolo Bernardi.


Asunto(s)
Antiportadores/metabolismo , Canales de Calcio/metabolismo , Calcio/metabolismo , Ácido Glutámico/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antiportadores/genética , Canales de Calcio/genética , Respiración de la Célula/efectos de los fármacos , Expresión Génica , Ácido Glutámico/farmacología , Ratones , Mitocondrias/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/genética , Poro de Transición de la Permeabilidad Mitocondrial , Proteínas Mitocondriales/genética , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
14.
Biochim Biophys Acta ; 1863(10): 2413-21, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27033520

RESUMEN

Mitochondrial function is regulated by calcium. In addition to the long known effects of matrix Ca(2+), regulation of metabolite transport by extramitochondrial Ca(2+) represents an alternative Ca(2+)-dependent mechanism to regulate mitochondrial function. The Ca(2+) regulated mitochondrial transporters (CaMCs) are well suited for that role, as they contain long N-terminal extensions harboring EF-hand Ca(2+) binding domains facing the intermembrane space. They fall in two groups, the aspartate/glutamate exchangers, AGCs, major components of the NADH malate aspartate shuttle (MAS) and urea cycle, and the ATP-Mg(2+)/Pi exchangers or short CaMCs (APCs or SCaMCs). The AGCs are activated by relatively low Ca(2+) levels only slightly higher than resting Ca(2+), whereas all SCaMCs studied so far require strong Ca(2+) signals, above micromolar, for activation. In addition, AGCs are not strictly Ca(2+) dependent, being active even in Ca(2+)-free conditions. Thus, AGCs are well suited to respond to small Ca(2+) signals and that do not reach mitochondria. In contrast, ATP-Mg(2+)/Pi carriers are inactive in Ca(2+) free conditions and activation requires Ca(2+) signals that will also activate the calcium uniporter (MCU). By changing the net content of adenine nucleotides of the matrix upon activation, SCaMCs regulate the activity of the permeability transition pore, and the Ca(2+) retention capacity of mitochondria (CRC), two functions synergizing with those of the MCU. The different Ca(2+) activation properties of the two CaMCs are discussed in relation to their newly obtained structures. This article is part of a Special Issue entitled: Mitochondrial Channels edited by Pierre Sonveaux, Pierre Maechler and Jean-Claude Martinou.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/fisiología , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Animales , Antiportadores/fisiología , Proteínas de Arabidopsis/metabolismo , Transporte Biológico Activo , Proteínas de Unión al Calcio/fisiología , Respiración de la Célula , Humanos , Transporte Iónico , Mamíferos/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/deficiencia , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/fisiología , Modelos Moleculares , Transportadores de Anión Orgánico/fisiología , Conformación Proteica , Proteínas de Saccharomyces cerevisiae/metabolismo
15.
J Clin Oncol ; 34(7): 706-13, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26755507

RESUMEN

PURPOSE: Volasertib is a potent and selective cell-cycle kinase inhibitor that induces mitotic arrest and apoptosis by targeting Polo-like kinase. This phase II trial evaluated volasertib or single-agent chemotherapy in patients with platinum-resistant or -refractory ovarian cancer who experienced failure after treatment with two or three therapy lines. PATIENTS AND METHODS: Patients were randomly assigned to receive either volasertib 300 mg by intravenous infusion every 3 weeks or an investigator's choice of single-agent, nonplatinum, cytotoxic chemotherapy. The primary end point was 24-week disease control rate. Secondary end points included best overall response, progression-free survival (PFS), safety, quality of life, and exploratory biomarker analyses. RESULTS: Of the 109 patients receiving treatment, 54 received volasertib and 55 received chemotherapy; demographics were well balanced. The 24-week disease control rates for volasertib and chemotherapy were 30.6% (95% CI, 18.0% to 43.2%) and 43.1% (95% CI, 29.6% to 56.7%), respectively, with partial responses in seven (13.0%) and eight (14.5%) patients, respectively. Median PFS was 13.1 weeks and 20.6 weeks for volasertib and chemotherapy (hazard ratio, 1.01; 95% CI, 0.66 to 1.53). Six patients (11%) receiving volasertib achieved PFS fore more than 1 year, whereas no patient receiving chemotherapy achieved PFS greater than 1 year. No relationship between the expression of the biomarkers tested and their response was determined. Patients treated with volasertib experienced more grade 3 and 4 drug-related hematologic adverse events (AEs) and fewer nonhematologic AEs than did patients receiving chemotherapy. Discontinuation resulting from AEs occurred in seven (13.0%) and 15 (27.3%) patients in the volasertib and chemotherapy arms, respectively. Both arms showed similar effects on quality of life. CONCLUSION: Single-agent volasertib showed antitumor activity in patients with ovarian cancer. AEs in patients receiving volasertib were mainly hematologic and manageable.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Pteridinas/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Carboplatino/uso terapéutico , Cisplatino/uso terapéutico , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Femenino , Humanos , Infusiones Intravenosas , Persona de Mediana Edad , Pteridinas/administración & dosificación , Calidad de Vida , Resultado del Tratamiento
16.
J Neurosci ; 35(8): 3566-81, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25716855

RESUMEN

Glutamate excitotoxicity is caused by sustained activation of neuronal NMDA receptors causing a large Ca(2+) and Na(+) influx, activation of poly(ADP ribose) polymerase-1 (PARP-1), and delayed Ca(2+) deregulation. Mitochondria undergo early changes in membrane potential during excitotoxicity, but their precise role in these events is still controversial. Using primary cortical neurons derived from mice, we show that NMDA exposure results in a rapid fall in mitochondrial ATP in neurons deficient in SCaMC-3/Slc25a23, a Ca(2+)-regulated mitochondrial ATP-Mg/Pi carrier. This fall is associated with blunted increases in respiration and a delayed decrease in cytosolic ATP levels, which are prevented by PARP-1 inhibitors or by SCaMC-3 activity promoting adenine nucleotide uptake into mitochondria. SCaMC-3 KO neurons show an earlier delayed Ca(2+) deregulation, and SCaMC-3-deficient mitochondria incubated with ADP or ATP-Mg had reduced Ca(2+) retention capacity, suggesting a failure to maintain matrix adenine nucleotides as a cause for premature delayed Ca(2+) deregulation. SCaMC-3 KO neurons have higher vulnerability to in vitro excitotoxicity, and SCaMC-3 KO mice are more susceptible to kainate-induced seizures, showing that early PARP-1-dependent fall in mitochondrial ATP levels, counteracted by SCaMC-3, is an early step in the excitotoxic cascade.


Asunto(s)
Adenosina Trifosfato/metabolismo , Antiportadores/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Potenciales de Acción , Animales , Antiportadores/genética , Calcio/metabolismo , Respiración de la Célula , Células Cultivadas , Corteza Cerebral/citología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Convulsiones/metabolismo
17.
Biochim Biophys Acta ; 1837(10): 1617-24, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24820519

RESUMEN

Calcium is thought to regulate respiration but it is unclear whether this is dependent on the increase in ATP demand caused by any Ca(2+) signal or to Ca(2+) itself. [Na(+)]i, [Ca(2+)]i and [ATP]i dynamics in intact neurons exposed to different workloads in the absence and presence of Ca(2+) clearly showed that Ca(2+)-stimulation of coupled respiration is required to maintain [ATP]i levels. Ca(2+) may regulate respiration by activating metabolite transport in mitochondria from outer face of the inner mitochondrial membrane, or after Ca(2+) entry in mitochondria through the calcium uniporter (MCU). Two Ca(2+)-regulated mitochondrial metabolite transporters are expressed in neurons, the aspartate-glutamate exchanger ARALAR/AGC1/Slc25a12, a component of the malate-aspartate shuttle, and the ATP-Mg/Pi exchanger SCaMC-3/APC2/Slc25a23, with S0.5 for Ca(2+) of 300nM and 3.4µM, respectively. The lack of SCaMC-3 results in a smaller Ca(2+)-dependent stimulation of respiration only at high workloads, as caused by veratridine, whereas the lack of ARALAR reduced by 46% basal OCR in intact neurons using glucose as energy source and the Ca(2+)-dependent responses to all workloads: a reduction of about 65-70% in the response to the high workload imposed by veratridine, and completely suppression of the OCR responses to moderate (K(+)-depolarization) and small (carbachol) workloads, effects reverted by pyruvate supply. For K(+)-depolarization, this occurs in spite of the presence of large [Ca(2+)]mit signals and increased formation of mitochondrial NAD(P)H. These results show that ARALAR-MAS is a major contributor of Ca(2+)-stimulated respiration in neurons by providing increased pyruvate supply to mitochondria. In its absence and under moderate workloads, matrix Ca(2+) is unable to stimulate pyruvate metabolism and entry in mitochondria suggesting a limited role of MCU in these conditions. This article was invited for a Special Issue entitled: 18th European Bioenergetic Conference.


Asunto(s)
Calcio/fisiología , Mitocondrias/fisiología , Neuronas/fisiología , Adenosina Trifosfato/metabolismo , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animales , Antiportadores/metabolismo , Citosol/metabolismo , Humanos , Transporte Iónico , Mitocondrias/metabolismo , Neuronas/metabolismo , Ácido Pirúvico/metabolismo
18.
J Neurosci ; 33(35): 13957-71, 13971a, 2013 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-23986233

RESUMEN

Neuronal respiration is controlled by ATP demand and Ca2+ but the roles played by each are unknown, as any Ca2+ signal also impacts on ATP demand. Ca2+ can control mitochondrial function through Ca2+-regulated mitochondrial carriers, the aspartate-glutamate and ATP-Mg/Pi carriers, ARALAR/AGC1 and SCaMC-3, respectively, or in the matrix after Ca2+ transport through the Ca2+ uniporter. We have studied the role of Ca2+ signaling in the regulation of mitochondrial respiration in intact mouse cortical neurons in basal conditions and in response to increased workload caused by increases in [Na+]cyt (veratridine, high-K+ depolarization) and/or [Ca2+]cyt (carbachol). Respiration in nonstimulated neurons on 2.5-5 mm glucose depends on ARALAR-malate aspartate shuttle (MAS), with a 46% drop in aralar KO neurons. All stimulation conditions induced increased OCR (oxygen consumption rate) in the presence of Ca2+, which was prevented by BAPTA-AM loading (to preserve the workload), or in Ca2+-free medium (which also lowers cell workload). SCaMC-3 limits respiration only in response to high workloads and robust Ca2+ signals. In every condition tested Ca2+ activation of ARALAR-MAS was required to fully stimulate coupled respiration by promoting pyruvate entry into mitochondria. In aralar KO neurons, respiration was stimulated by veratridine, but not by KCl or carbachol, indicating that the Ca2+ uniporter pathway played a role in the first, but not in the second condition, even though KCl caused an increase in [Ca2+]mit. The results suggest a requirement for ARALAR-MAS in priming pyruvate entry in mitochondria as a step needed to activate respiration by Ca2+ in response to moderate workloads.


Asunto(s)
Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Respiración de la Célula/genética , Homeostasis , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Neuronas/metabolismo , Animales , Señalización del Calcio , Carbacol/farmacología , Respiración de la Célula/efectos de los fármacos , Corteza Cerebral/citología , Glucosa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Translocasas Mitocondriales de ADP y ATP/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Oxígeno/metabolismo , Cloruro de Potasio/farmacología , Piruvatos/metabolismo , Sodio/metabolismo , Veratridina/farmacología
19.
Bol. méd. Hosp. Infant. Méx ; 70(3): 195-208, may.-jun. 2013. ilus, tab
Artículo en Español | LILACS | ID: lil-701238

RESUMEN

Introducción. En México, no se contaba con una prueba de evaluación del desarrollo infantil con propiedades psicométricas. La prueba de evaluación del desarrollo infantil (EDI) se desarrolló con este fin. El objetivo de este trabajo fue determinar las propiedades psicométricas de la EDI como prueba de tamizaje para los problemas de desarrollo infantil en menores de 5 años. Métodos. Se realizó un estudio transversal que incluyó pacientes menores de 5 años en tres entidades de la República Mexicana: Chihuahua, Yucatán y Distrito Federal. El espectro de la población incluyó niños con factores de riesgo biológico, ambiental y sin riesgo para retraso en el desarrollo. Se excluyeron los pacientes con alteraciones neurológicas evidentes. Se consideró, como prueba diagnóstica, el Inventario de Desarrollo de Battelle-2 en las tres entidades. En el Distrito Federal, adicionalmente, se aplicó Bayley-III. A cada participante se le aplicaron la prueba de tamizaje en dos versiones y la prueba diagnóstica, el mismo día o en un lapso no mayor a una semana. La persona que aplicó la prueba diagnóstica no conocía el resultado de la prueba de tamizaje. Se definió retraso cuando el cociente total de desarrollo resultó menor a 90. Resultados. Se incluyeron, en total, 438 niños menores de 5 años provenientes del Distrito Federal (n =152, 34.7%), Yucatán (n =151, 34.5%) y Chihuahua (n =135, 30.8%). Del total, 43.4% fueron del sexo femenino (n =190). La clasificación por tipo de riesgo fue biológico (n =197, 45%), ambiental (n =137, 31.3%) y sin riesgo (n =104, 23.7%). Se encontró una sensibilidad de 0.81 (IC 95%: 0.75-0.86), especificidad de 0.61 (IC 95%: 0.54-0.67), concordancia 0.70 (IC 95%: 0.66-0.74). La correlación parcial de las áreas del desarrollo entre la prueba de tamizaje y la prueba Bayley III (n =87) ajustada por grupo de edad del tamizaje fue la siguiente: área motor fino 0.468, motor grueso 0.441, lenguaje 0.508, social 0.336 y adaptativo 0.355 (p ≤0.001). Conclusiones. La prueba EDI posee propiedades adecuadas y similares a las pruebas más utilizadas en América.


Background. The ''Evaluación del Desarrollo Infantil'' (EDI) test was developed as an screening tool for the developmental evaluation of Mexican children younger than 5 years old. The objective of this study was to evaluate the psychometric properties of EDI as a screening tool for children with developmental problems. Methods. We carried out a cross-sectional study including patients from urban and rural areas in three locations: Mexico City, Yucatan and Chihuahua. The disease spectrum was defined according to biological risk, environmental risk or without risk for developmental problems. Patients with obvious neurological disabilities or genetic syndromes were excluded. The gold standards were the Battelle Developmental Inventory 2 (in Spanish) and Bayley-III. Each participant had two complete applications of the EDI test (all interrogated and all observed) and the gold standard (Bayley-III only in Mexico City). Developmental delay was defined as a total development quotient <90. Results. The study included 438 children <5 years old. Distribution by site includes Mexico City (n =152, 34.7%), Yucatan (n =151, 34.5%), Chihuahua (n =135, 30.8%); female gender (n =190, 43.4%). Classification by risk includes biological (n =197, 45%), environmental (n =137, 31.3%), without risk (n =104, 23.7%). With BDI-II as the gold standard, the modified version of EDI (interrogated plus observation) has a sensitivity of 0.81 (95% CI: 0.75-0.86), specificity 0.61 (95% CI: 0.54-0.67), and concordance 0.70 (95% CI: 0.66-0.74). The partial correlation between EDI areas and Bayley-III areas (n =87) was adjusted by test group: fine motor 0.468, gross motor 0.441, language 0.508, social 0.336 and adaptive 0.355 (p ≤0.001). Conclusions. The modified version of EDI has similar properties as the various developmental screening tools available in the U.S. or Latin America and could be a good screening tool in Spanish.

20.
J Plant Physiol ; 170(3): 265-71, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23246027

RESUMEN

The aims of this work were to investigate the microlocalisation of cadmium (Cd) in Lupinus albus L. cv. Multolupa nodules, and to determine its effects on carbon and nitrogen metabolism. Nodulated white lupin plants were grown in a growth chamber with or without Cd (150 µM). Energy-dispersive X-ray microanalysis showed the walls of the outer nodule cortex cells to be the main area of Cd retention, helping to reduce the harmful effect Cd might have on the amount of N(2) fixed by the bacteroids. Sucrose synthase activity declined by 33% in the nodules of the Cd-treated plants, and smaller reductions were recorded in glutamine synthetase, aspartate aminotransferase, alkaline invertase and NADP-dependent isocitrate dehydrogenase activities. The Cd treatment also sharply reduced nodule concentrations of malate, succinate and citrate, while that of starch doubled, but that of sucrose experienced no significant change. In summary, the present results show that white lupins accumulate significant amounts of Cd in their root nodules. However, the activity of some enzymes involved in ammonium assimilation did decline, promoting a reduction in the plant N content. The downregulation of sucrose synthase limits the availability of carbon to the bacteroids, which might interfere with their respiration. Carbon metabolism therefore plays a primary role in the impaired function of the white lupin root nodule caused by Cd, while N metabolism appears to have a more secondary involvement.


Asunto(s)
Bradyrhizobium/metabolismo , Cadmio/toxicidad , Carbono/metabolismo , Lupinus/metabolismo , Lupinus/microbiología , Fijación del Nitrógeno , Nódulos de las Raíces de las Plantas/metabolismo , Cadmio/metabolismo , Pared Celular/química , Glucosiltransferasas/metabolismo , Glutamato-Amoníaco Ligasa/metabolismo , Lupinus/efectos de los fármacos , Nitrógeno/metabolismo , Reguladores del Crecimiento de las Plantas/metabolismo , Nódulos de las Raíces de las Plantas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA