Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Ann Vasc Surg ; 46: 368.e9-368.e12, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28890067

RESUMEN

Thyroid cancer causing superior vena cava (SVC) syndrome is a very rare clinical entity with only 26 cases reported worldwide. Metastatic thyroid cancer causing SVC syndrome several years following resection of the primary tumor is extremely rare with only 2 cases reported; one of them was of the papillary variety. We report the second case worldwide of metastatic papillary thyroid cancer causing SVC syndrome 2 years after total thyroidectomy of the original tumor in a 62-year-old Indian female pilgrim. Unfortunately, the patient died on the third day of intensive care unit admission. The severity of the clinical condition in addition to the late presentation resulted in a catastrophic outcome, which made all the possible resuscitative efforts very difficult.


Asunto(s)
Carcinoma Papilar/complicaciones , Síndrome de la Vena Cava Superior/etiología , Neoplasias de la Tiroides/complicaciones , Biopsia con Aguja Fina , Carcinoma Papilar/diagnóstico por imagen , Carcinoma Papilar/secundario , Carcinoma Papilar/cirugía , Resultado Fatal , Femenino , Humanos , Persona de Mediana Edad , Síndrome de la Vena Cava Superior/diagnóstico por imagen , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/diagnóstico por imagen , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/cirugía , Tiroidectomía , Factores de Tiempo , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
2.
BMC Cancer ; 14: 317, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24886434

RESUMEN

BACKGROUND: Current treatment of ovarian cancer patients with chemotherapy leaves behind a residual tumor which results in recurrent ovarian cancer within a short time frame. We have previously demonstrated that a single short-term treatment of ovarian cancer cells with chemotherapy in vitro resulted in a cancer stem cell (CSC)-like enriched residual population which generated significantly greater tumor burden compared to the tumor burden generated by control untreated cells. In this report we looked at the mechanisms of the enrichment of CSC-like residual cells in response to paclitaxel treatment. METHODS: The mechanism of survival of paclitaxel-treated residual cells at a growth inhibitory concentration of 50% (GI50) was determined on isolated tumor cells from the ascites of recurrent ovarian cancer patients and HEY ovarian cancer cell line by in vitro assays and in a mouse xenograft model. RESULTS: Treatment of isolated tumor cells from the ascites of ovarian cancer patients and HEY ovarian cancer cell line with paclitaxel resulted in a CSC-like residual population which coincided with the activation of Janus activated kinase 2 (JAK2) and signal transducer and activation of transcription 3 (STAT3) pathway in paclitaxel surviving cells. Both paclitaxel-induced JAK2/STAT3 activation and CSC-like characteristics were inhibited by a low dose JAK2-specific small molecule inhibitor CYT387 (1 µM) in vitro. Subsequent, in vivo transplantation of paclitaxel and CYT387-treated HEY cells in mice resulted in a significantly reduced tumor burden compared to that seen with paclitaxel only-treated transplanted cells. In vitro analysis of tumor xenografts at protein and mRNA levels demonstrated a loss of CSC-like markers and CA125 expression in paclitaxel and CYT387-treated cell-derived xenografts, compared to paclitaxel only-treated cell-derived xenografts. These results were consistent with significantly reduced activation of JAK2 and STAT3 in paclitaxel and CYT387-treated cell-derived xenografts compared to paclitaxel only-treated cell derived xenografts. CONCLUSIONS: This proof of principle study demonstrates that inhibition of the JAK2/STAT3 pathway by the addition of CYT387 suppresses the 'stemness' profile in chemotherapy-treated residual cells in vitro, which is replicated in vivo, leading to a reduced tumor burden. These findings have important implications for ovarian cancer patients who are treated with taxane and/or platinum-based therapies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Janus Quinasa 2/antagonistas & inhibidores , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Adulto , Anciano , Animales , Benzamidas/farmacología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Janus Quinasa 2/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Terapia Molecular Dirigida , Recurrencia Local de Neoplasia , Neoplasia Residual , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Paclitaxel/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Factor de Transcripción STAT3/metabolismo , Factores de Tiempo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Cell Biochem ; 114(1): 21-34, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22887554

RESUMEN

Chemotherapy with platinum and taxanes is the first line of treatment for all epithelial ovarian cancer (EOC) patients after debulking surgery. Even though the treatment is initially effective in 80% of patients, recurrent cancer is inevitable in the vast majority of cases. Emerging evidence suggests that some tumor cells can survive chemotherapy by activating the self-renewal pathways resulting in tumor progression and clinical recurrence. These defined population of cells commonly termed as "cancer stem cells" (CSC) may generate the bulk of the tumor by using differentiating pathways. These cells have been shown to be resistant to chemotherapy and, to have enhanced tumor initiating abilities, suggesting CSCs as potential targets for treatment. Recent studies have introduced a new paradigm in ovarian carcinogenesis which proposes in situ carcinoma at the fimbrial end of the fallopian tube to generate high-grade serous ovarian carcinomas, in contrast to ovarian cortical inclusion cysts (CIC) which produce borderline and low grade serous, mucinous, endometrioid, and clear cell carcinomas. This review summarizes recent advances in our understanding of the cellular origin of EOC and the molecular mechanisms defining the basis of CSC in EOC progression and chemoresistance. Using a model ovarian cancer cell line, we highlight the role of CSC in response to chemotherapy, and relate how CSCs may impact on chemoresistance and ultimately recurrence. We also propose the molecular targeting of CSCs and suggest ways that may improve the efficacy of current chemotherapeutic regimens needed for the management of this disease.


Asunto(s)
Adenocarcinoma de Células Claras/terapia , Antineoplásicos/uso terapéutico , Carcinoma in Situ/terapia , Cistadenocarcinoma Seroso/terapia , Neoplasias Glandulares y Epiteliales/terapia , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Ováricas/terapia , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patología , Antineoplásicos/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patología , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patología , Trompas Uterinas/efectos de los fármacos , Trompas Uterinas/metabolismo , Trompas Uterinas/patología , Femenino , Humanos , Terapia Molecular Dirigida , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Ovario/efectos de los fármacos , Ovario/metabolismo , Ovario/patología
4.
Mol Cancer ; 12: 24, 2013 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-23537295

RESUMEN

Over 80% of women diagnosed with advanced-stage ovarian cancer die as a result of disease recurrence due to failure of chemotherapy treatment. In this study, using two distinct ovarian cancer cell lines (epithelial OVCA 433 and mesenchymal HEY) we demonstrate enrichment in a population of cells with high expression of CSC markers at the protein and mRNA levels in response to cisplatin, paclitaxel and the combination of both. We also demonstrate a significant enhancement in the sphere forming abilities of ovarian cancer cells in response to chemotherapy drugs. The results of these in vitro findings are supported by in vivo mouse xenograft models in which intraperitoneal transplantation of cisplatin or paclitaxel-treated residual HEY cells generated significantly higher tumor burden compared to control untreated cells. Both the treated and untreated cells infiltrated the organs of the abdominal cavity. In addition, immunohistochemical studies on mouse tumors injected with cisplatin or paclitaxel treated residual cells displayed higher staining for the proliferative antigen Ki67, oncogeneic CA125, epithelial E-cadherin as well as cancer stem cell markers such as Oct4 and CD117, compared to mice injected with control untreated cells. These results suggest that a short-term single treatment of chemotherapy leaves residual cells that are enriched in CSC-like traits, resulting in an increased metastatic potential. The novel findings in this study are important in understanding the early molecular mechanisms by which chemoresistance and subsequent relapse may be triggered after the first line of chemotherapy treatment.


Asunto(s)
Antineoplásicos/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Carga Tumoral/efectos de los fármacos , Animales , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Antineoplásicos/administración & dosificación , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Polaridad Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Cisplatino/farmacología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Endonucleasas/genética , Endonucleasas/metabolismo , Femenino , Humanos , Inmunofenotipificación , Ratones , Metástasis de la Neoplasia , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Fenotipo , ARN Mensajero/genética , Esferoides Celulares , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas
5.
J Cell Biochem ; 112(10): 2850-64, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21618587

RESUMEN

Epithelial mesenchymal transition (EMT) and cancer stem cells (CSC) have been associated with resistance to chemotherapy. Eighty percent of ovarian cancer patients initially respond to platinum-based combination therapy but most return with recurrence and ultimate demise. To better understand such chemoresistance we have assessed the potential role of EMT in tumor cells collected from advanced-stage ovarian cancer patients and the ovarian cancer cell line OVCA 433 in response to cisplatin in vitro. We demonstrate that cisplatin-induced transition from epithelial to mesenchymal morphology in residual cancer cells correlated with reduced E-cadherin, and increased N-cadherin and vimentin expression. The mRNA expression of Snail, Slug, Twist, and MMP-2 were significantly enhanced in response to cisplatin and correlated with increased migration. This coincided with increased cell surface expression of CSC-like markers such as CD44, α2 integrin subunit, CD117, CD133, EpCAM, and the expression of stem cell factors Nanog and Oct-4. EMT and CSC-like changes in response to cisplatin correlated with enhanced activation of extracellular signal-regulated kinase (ERK)1/2. The selective MEK inhibitor U0126 inhibited ERK2 activation and partially suppressed cisplatin-induced EMT and CSC markers. In vivo xenotransplantation of cisplatin-treated OVCA 433 cells in zebrafish embryos demonstrated significantly enhanced migration of cells compared to control untreated cells. U0126 inhibited cisplatin-induced migration of cells in vivo, suggesting that ERK2 signaling is critical to cisplatin-induced EMT and CSC phenotypes, and that targeting ERK2 in the presence of cisplatin may reduce the burden of residual tumor, the ultimate cause of recurrence in ovarian cancer patients.


Asunto(s)
Cisplatino/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Ováricas/metabolismo , Butadienos/farmacología , Línea Celular Tumoral , Femenino , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Nitrilos/farmacología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
6.
Br J Neurosurg ; 25(1): 142-3, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21158504

RESUMEN

Intra-extracranial schwannomas arising unrelated to major cranial nerves are exceedingly rare neoplasms. We report the case of a 23-year-old male who presented with a 9 month history of progressive temporal swelling which was excised and found histologically to be a schwannoma. A succinct review of the relevant literature is presented.


Asunto(s)
Fosa Craneal Posterior/diagnóstico por imagen , Neoplasias de los Nervios Craneales/diagnóstico por imagen , Neurilemoma/diagnóstico por imagen , Neoplasias de los Nervios Craneales/patología , Humanos , Masculino , Neurilemoma/patología , Neoplasias de la Base del Cráneo/diagnóstico por imagen , Neoplasias de la Base del Cráneo/patología , Tomografía Computarizada por Rayos X , Resultado del Tratamiento , Adulto Joven
7.
Br J Neurosurg ; 25(1): 94-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21323404

RESUMEN

OBJECTIVES: Idiopathic intracranial hypertension (IIH) is an uncommon but important cause of headache that can lead to visual loss. This study was undertaken to review our experience in the treatment of IIH by neuronavigation-assisted ventriculoperitoneal (VP) shunts with programmable valves as compared to lumboperitoneal (LP) shunts. METHODS: A retrospective chart review was conducted on 25 patients treated for IIH between 2001 and 2009. Age, sex, clinical presentation, methods of treatment and failure rates were recorded. RESULTS: Seventy-two per cent were treated initially with LP shunts. Failure rate was 11% in this group. Neuronavigation-assisted VP shunts were used to treat 28%. In this group, the failure rate was 14%. CONCLUSION: Our experience indicates that both LP shunts and VP shuts are effective in controlling all the clinical manifestations of IIH in the immediate postoperative period. Failure rates are slightly higher for VP shunts (14%) than LP shunts (11%). However, revision rates are higher with LP shunts (60%) than with VP shunts (30%).


Asunto(s)
Derivaciones del Líquido Cefalorraquídeo/métodos , Cefalea/cirugía , Seudotumor Cerebral/cirugía , Trastornos de la Visión/cirugía , Adulto , Anciano , Niño , Femenino , Cefalea/etiología , Humanos , Masculino , Persona de Mediana Edad , Seudotumor Cerebral/complicaciones , Seudotumor Cerebral/fisiopatología , Reoperación , Estudios Retrospectivos , Resultado del Tratamiento , Derivación Ventriculoperitoneal/métodos , Trastornos de la Visión/etiología , Trastornos de la Visión/prevención & control
8.
Mol Aspects Med ; 39: 110-25, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23811037

RESUMEN

In spite of recent progress in cancer therapeutics and increased knowledge about the cellular and molecular biology of cancer, ovarian cancer still remains a clinical challenge. Chemoresistance followed by tumor recurrence are major causes of poor survival rates of ovarian cancer patients. In recent years, ovarian cancer has been described as a stem cell disease. In this scenario, a small percentage of ovarian tumor cells with cancer stem cell-like properties should survive therapeutic treatments by activating the self-renewal and differentiating pathways resulting in tumor progression and clinical recurrence. The mere concept that a small subset of cells in the tumor population drives tumor formation and recurrence after therapies has major implications for therapeutic development. This review focuses on the current understanding of normal and malignant ovarian stem cells in an attempt to contribute to our understanding the mechanisms responsible for tumor development as well as recurrence after chemotherapy. We also discuss recent findings on the cancer stem cell niche and how tumor and associated cells in the niche may respond to chemotherapeutic stress by activating autocrine and paracrine programs which may opt as survival mechanisms for residual cells in response to frontline chemotherapy. Using mouse ovarian cancer models we highlight the role of cancer stem cells in response to chemotherapy, and relate how cancer stem cells may impact on recurrence. Understanding the distinct mechanisms that facilitate cancer stem cell survival and propagation are likely to reveal opportunities for improving the treatment outcomes for ovarian cancer patients.


Asunto(s)
Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Animales , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , MicroARNs/genética , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/genética
9.
J Cereb Blood Flow Metab ; 34(8): e1-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24865999

RESUMEN

In addition to delayed vasospasm also early brain injury, which occurs during the first few days after subarachnoid hemorrhage (SAH) when large cerebral arteries are still fully functional, plays an important role for the outcome after SAH. In the current study, we investigated the hypothesis that carbon dioxide (CO2), a strong cerebral vasodilator, has a therapeutic potential against early posthemorrhagic microvasospasm. C57BL/6 mice (n=36) and Sprague-Dawley rats (n=23) were subjected to sham surgery or SAH by filament perforation. The pial microcirculation in the mice was visualized 3 and 24 hours after SAH using intravital fluorescence microscopy. Partial pressure of CO2 (PaCO2) was modulated by hyper- or hypoventilation or by inhalation of 10% CO2. In rats, CO2-mediated changes in cerebral blood flow (CBF) were measured at the same time points using laser Doppler fluxmetry. Increased PaCO2 caused vasodilatation in sham-operated animals. Following SAH, however, cerebral arterioles were nonreactive to CO2. This lack of microvascular CO2 reactivity was accompanied by a complete loss of CO2-induced hyperemia. Our data show that CO2 is not able to dilate spastic microvessels and to increase CBF early after SAH. Future therapeutic approaches will therefore need to address mechanisms beyond CO2.


Asunto(s)
Dióxido de Carbono/uso terapéutico , Microvasos/fisiopatología , Hemorragia Subaracnoidea/tratamiento farmacológico , Vasoespasmo Intracraneal/tratamiento farmacológico , Administración por Inhalación , Animales , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Dióxido de Carbono/administración & dosificación , Arterias Cerebrales/fisiopatología , Circulación Cerebrovascular/efectos de los fármacos , Modelos Animales de Enfermedad , Flujometría por Láser-Doppler , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Microvasos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/fisiopatología , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasoespasmo Intracraneal/etiología , Vasoespasmo Intracraneal/fisiopatología
10.
Front Oncol ; 4: 75, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24782986

RESUMEN

Chemotherapy resistance associated with recurrent disease is the major cause of poor survival of ovarian cancer patients. We have recently demonstrated activation of the JAK2/STAT3 pathway and the enhancement of a cancer stem cell (CSC)-like phenotype in ovarian cancer cells treated in vitro with chemotherapeutic agents. To elucidate further these mechanisms in vivo, we used a two-tiered paclitaxel treatment approach in nude mice inoculated with ovarian cancer cells. In the first approach, we demonstrate that a single intraperitoneal administration of paclitaxel in mice 7 days after subcutaneous transplantation of the HEY ovarian cancer cell line resulted in a significant increase in the expression of CA125, Oct4, and CD117 in mice xenografts compared to control mice xenografts which did not receive paclitaxel. In the second approach, mice were administered once weekly with paclitaxel and/or a daily dose of the JAK2-specific inhibitor, CYT387, over 4 weeks. Mice receiving paclitaxel only demonstrated a significant decrease in tumor volume compared to control mice. At the molecular level, mouse tumors remaining after paclitaxel administration showed a significant increase in the expression of Oct4 and CD117 coinciding with a significant activation of the JAK2/STAT3 pathway compared to control tumors. The addition of CYT387 with paclitaxel resulted in the suppression of JAK2/STAT3 activation and abrogation of Oct4 and CD117 expression in mouse xenografts. This coincided with significantly smaller tumors in mice administered CYT387 in addition to paclitaxel, compared to the control group and the group of mice receiving paclitaxel only. These data suggest that the systemic administration of paclitaxel enhances Oct4- and CD117-associated CSC-like marker expression in surviving cancer cells in vivo, which can be suppressed by the addition of the JAK2-specific inhibitor CYT387, leading to a significantly smaller tumor burden. These novel findings have the potential for the development of CSC-targeted therapy to improve the treatment outcomes of ovarian cancer patients.

11.
Curr Cancer Drug Targets ; 10(3): 268-78, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20370691

RESUMEN

Overcoming intrinsic and acquired chemoresistance is the major challenge in treating ovarian cancer patients. Initially nearly 75% of ovarian cancer patients respond favourably to chemotherapy, but subsequently the majority gain acquired resistance resulting in recurrence, cancer dissemination and death. This review summarizes recent advances in our understanding of the cellular origin and the molecular mechanisms defining the basis of cancer initiation and malignant transformation with respect to epithelial-mesenchymal transition (EMT) of ovarian cancer cells. We discuss the critical role of EMT frequently encountered in different phases of ovarian cancer progression and its involvement in regulating cancer growth, survival, migration, invasion and drug resistance. Using a model ovarian cancer cell line we highlight the relationship between EMT and the 'migrating cancer stem (MCS) cell-like phenotype' in response to drug treatment, and relate how these processes can impact on chemoresistance and ultimately recurrence. We propose the molecular targeting of distinct 'EMT transformed cancer stem-like cells' and suggest ways that may improve the efficacy of current chemotherapeutic regimens much needed for the management of this disease.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Células Epiteliales/efectos de los fármacos , Mesodermo/efectos de los fármacos , Recurrencia Local de Neoplasia , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Mesodermo/metabolismo , Mesodermo/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/secundario , Fenotipo , Transducción de Señal/efectos de los fármacos , Insuficiencia del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA