Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Neuro Oncol ; 26(8): 1421-1437, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38506351

RESUMEN

BACKGROUND: Cellular functions hinge on the meticulous orchestration of protein transport, both spatially and temporally. Central to this process is retrograde trafficking, responsible for targeting proteins to the nucleus. Despite its link to many diseases, the implications of retrograde trafficking in glioblastoma (GBM) are still unclear. METHODS: To identify genetic drivers of TMZ resistance, we conducted comprehensive CRISPR-knockout screening, revealing ADP-ribosylation factor 4 (ARF4), a regulator of retrograde trafficking, as a major contributor. RESULTS: Suppressing ARF4 significantly enhanced TMZ sensitivity in GBM patient-derived xenograft (PDX) models, leading to improved survival rates (P < .01) in both primary and recurrent lines. We also observed that TMZ exposure stimulates ARF4-mediated retrograde trafficking. Proteomics analysis of GBM cells with varying levels of ARF4 unveiled the influence of this pathway on EGFR signaling, with increased nuclear trafficking of EGFR observed in cells with ARF4 overexpression and TMZ treatment. Additionally, spatially resolved RNA-sequencing of GBM patient tissues revealed substantial correlations between ARF4 and crucial nuclear EGFR (nEGFR) downstream targets, such as MYC, STAT1, and DNA-PK. Decreased activity of DNA-PK, a DNA repair protein downstream of nEGFR signaling that contributes to TMZ resistance, was observed in cells with suppressed ARF4 levels. Notably, treatment with DNA-PK inhibitor, KU-57788, in mice with a recurrent PDX line resulted in prolonged survival (P < .01), highlighting the promising therapeutic implications of targeting proteins reliant on ARF4-mediated retrograde trafficking. CONCLUSIONS: Our findings demonstrate that ARF4-mediated retrograde trafficking contributes to the development of TMZ resistance, cementing this pathway as a viable strategy to overcome chemoresistance in GBM.


Asunto(s)
Factores de Ribosilacion-ADP , Neoplasias Encefálicas , Resistencia a Antineoplásicos , Glioblastoma , Ensayos Antitumor por Modelo de Xenoinjerto , Humanos , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Animales , Ratones , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Factores de Ribosilacion-ADP/metabolismo , Factores de Ribosilacion-ADP/genética , Temozolomida/farmacología , Antineoplásicos Alquilantes/farmacología , Transporte de Proteínas , Células Tumorales Cultivadas , Receptores ErbB/metabolismo , Receptores ErbB/genética , Proliferación Celular , Línea Celular Tumoral , Transducción de Señal , Regulación Neoplásica de la Expresión Génica
2.
J Neurosurg Case Lessons ; 5(21)2023 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-37218733

RESUMEN

BACKGROUND: Gliosarcoma is a rare and highly malignant cancer of the central nervous system with the ability to metastasize. Secondary gliosarcoma, or the evolution of a spindle cell-predominant tumor after the diagnosis of a World Health Organization grade IV glioblastoma, has also been shown to metastasize. There is little information on metastatic secondary gliosarcoma. OBSERVATIONS: The authors present a series of 7 patients with previously diagnosed glioblastoma presenting with recurrent tumor and associated metastases with repeat tissue diagnosis consistent with gliosarcoma. The authors describe the clinical, imaging, and pathological characteristics in addition to carrying out a systematic review on metastases in secondary gliosarcoma. LESSONS: The present institutional series and the systematic review of the literature show that metastatic secondary gliosarcoma is a highly aggressive disease with a poor prognosis.

3.
Sci Adv ; 9(20): eade7236, 2023 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-37196077

RESUMEN

During therapy, adaptations driven by cellular plasticity are partly responsible for driving the inevitable recurrence of glioblastoma (GBM). To investigate plasticity-induced adaptation during standard-of-care chemotherapy temozolomide (TMZ), we performed in vivo single-cell RNA sequencing in patient-derived xenograft (PDX) tumors of GBM before, during, and after therapy. Comparing single-cell transcriptomic patterns identified distinct cellular populations present during TMZ therapy. Of interest was the increased expression of ribonucleotide reductase regulatory subunit M2 (RRM2), which we found to regulate dGTP and dCTP production vital for DNA damage response during TMZ therapy. Furthermore, multidimensional modeling of spatially resolved transcriptomic and metabolomic analysis in patients' tissues revealed strong correlations between RRM2 and dGTP. This supports our data that RRM2 regulates the demand for specific dNTPs during therapy. In addition, treatment with the RRM2 inhibitor 3-AP (Triapine) enhances the efficacy of TMZ therapy in PDX models. We present a previously unidentified understanding of chemoresistance through critical RRM2-mediated nucleotide production.


Asunto(s)
Neoplasias Encefálicas , Resistencia a Antineoplásicos , Glioblastoma , Ribonucleótido Reductasas , Humanos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Ribonucleótido Reductasas/genética , Ribonucleótido Reductasas/uso terapéutico , Temozolomida/farmacología , Temozolomida/uso terapéutico , Resistencia a Antineoplásicos/genética
4.
Stem Cell Rev Rep ; 18(1): 228-240, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34244971

RESUMEN

Tumor invasion and metastasis remain the leading causes of mortality for patients with cancer despite current treatment strategies. In some cancer types, recurrence is considered inevitable due to the lack of effective anti-metastatic therapies. Recent studies across many cancer types demonstrate a close relationship between cancer-initiating cells (CICs) and metastasis, as well as general cancer progression. First, this review describes CICs' contribution to cancer progression. Then we discuss our recent understanding of mechanisms through which CICs promote tumor invasion and metastasis by examining the role of CICs in each stage. Finally, we examine the current understanding of CICs' contribution to therapeutic resistance and recent developments in CIC-targeting drugs. We believe this understanding is key to advancing anti-CIC clinical therapeutics.


Asunto(s)
Neoplasias , Células Madre Neoplásicas , Línea Celular Tumoral , Humanos , Neoplasias/patología , Células Madre Neoplásicas/patología
5.
Brain ; 144(4): 1230-1246, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33855339

RESUMEN

Glioblastoma is a primary brain cancer with a near 100% recurrence rate. Upon recurrence, the tumour is resistant to all conventional therapies, and because of this, 5-year survival is dismal. One of the major drivers of this high recurrence rate is the ability of glioblastoma cells to adapt to complex changes within the tumour microenvironment. To elucidate this adaptation's molecular mechanisms, specifically during temozolomide chemotherapy, we used chromatin immunoprecipitation followed by sequencing and gene expression analysis. We identified a molecular circuit in which the expression of ciliary protein ADP-ribosylation factor-like protein 13B (ARL13B) is epigenetically regulated to promote adaptation to chemotherapy. Immuno-precipitation combined with liquid chromatography-mass spectrometry binding partner analysis revealed that that ARL13B interacts with the purine biosynthetic enzyme inosine-5'-monophosphate dehydrogenase 2 (IMPDH2). Further, radioisotope tracing revealed that this interaction functions as a negative regulator for purine salvaging. Inhibition of the ARL13B-IMPDH2 interaction enhances temozolomide-induced DNA damage by forcing glioblastoma cells to rely on the purine salvage pathway. Targeting the ARLI3B-IMPDH2 circuit can be achieved using the Food and Drug Administration-approved drug, mycophenolate mofetil, which can block IMPDH2 activity and enhance the therapeutic efficacy of temozolomide. Our results suggest and support clinical evaluation of MMF in combination with temozolomide treatment in glioma patients.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Resistencia a Antineoplásicos/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Glioblastoma/metabolismo , Purinas/biosíntesis , Animales , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/patología , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Ácido Micofenólico/farmacología , Temozolomida/farmacología , Células Tumorales Cultivadas
6.
Cancers (Basel) ; 12(12)2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-33255632

RESUMEN

Glioblastoma (GBM) is the most common primary brain malignancy in adults, with a 100% recurrence rate and 21-month median survival. Our lab and others have shown that GBM contains a subpopulation of glioma stem cells (GSCs) that expand during chemotherapy and may contribute to therapeutic resistance and recurrence in GBM. To investigate the mechanism behind this expansion, we applied gene set expression analysis (GSEA) to patient-derived xenograft (PDX) cells in response to temozolomide (TMZ), the most commonly used chemotherapy against GBM. Results showed significant enrichment of cancer stem cell and cell cycle pathways (False Discovery Rate (FDR) < 0.25). The ligand of numb protein 1 (LNX1), a known regulator of Notch signaling by targeting negative regulator Numb, is strongly upregulated after TMZ therapy (p < 0.0001) and is negatively correlated with survival of GBM patients. LNX1 is also upregulated after TMZ therapy in multiple PDX lines with concomitant downregulations in Numb and upregulations in intracellular Notch1 (NICD). Overexpression of LNX1 results in Notch1 signaling activation and increased GSC populations. In contrast, knocking down LNX1 reverses these changes, causing a significant downregulation of NICD, reduction in stemness after TMZ therapy, and resulting in more prolonged median survival in a mouse model. Based on this, we propose that during anti-GBM chemotherapy, LNX1-regulated Notch1 signaling promotes stemness and contributes to therapeutic resistance.

7.
World Neurosurg ; 142: 1-12, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32522637

RESUMEN

BACKGROUND: There has been much attention recently on whether the involvement of neurosurgical residents during surgery impacts patient outcomes. Our goal was to perform a meta-analysis of all existing studies in order to determine the true effect of resident involvement. METHODS: We performed a systematic review and identified studies that compared resident involvement during surgery to attending neurosurgeons alone. Event rates and adjusted odds ratios were collected and pooled to generate estimates. RESULTS: Eleven studies were identified, of which 9 reported adjusted odds ratios. Meta-analysis showed that there were no significant differences in patient baseline characteristics (age, gender, the majority of medical comorbidities). Analysis of operative variables showed increases in a number of complications. However, adjustment of odds ratios for confounders eliminated most of these effects but continued to show a mild increase in overall complications with an odds ratio of 1.14 (P = 0.02). Notably, for both adjusted and unadjusted estimates, no significant differences were seen in 30-day mortality. CONCLUSIONS: We found that, when adjusted for comorbidities, complexity, and procedure type, there was no difference in outcomes in terms of surgical complications, reoperation, length of stay more than 5 days, and mortality. While these results suggest that our apprenticeship teaching model is safe for developing independent physicians, using new educational modalities such as simulation and resident-directed labs may be useful to attenuate potential patient complications in higher-risk procedures and in patients with comorbidities.


Asunto(s)
Competencia Clínica/normas , Internado y Residencia/normas , Procedimientos Neuroquirúrgicos/normas , Humanos , Internado y Residencia/tendencias , Tiempo de Internación/tendencias , Procedimientos Neuroquirúrgicos/tendencias , Tempo Operativo , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/epidemiología , Reoperación/tendencias , Resultado del Tratamiento
8.
Stem Cells Int ; 2019: 6107456, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31316566

RESUMEN

Glioblastoma (GBM) is the most common and aggressive primary malignant brain tumor affecting adults, with a median survival of approximately 21 months. One key factor underlying the limited efficacy of current treatment modalities is the remarkable plasticity exhibited by GBM cells, which allows them to effectively adapt to changes induced by anticancer therapeutics. Moreover, GBM tumors are highly vascularized with aberrant vessels that complicate the delivery of antitumor agents. Recent research has demonstrated that GBM cells have the ability to transdifferentiate into endothelial cells (ECs), illustrating that GBM cells may use plasticity in concert with vascularization leading to the creation of tumor-derived blood vessels. The mechanism behind this transdifferentiation, however, remains unclear. Here, we show that treatment with temozolomide (TMZ) chemotherapy induces time-dependent expression of markers for glioma stem cells (GSCs) and immature and mature ECs. In addition, GBM tumors growing as orthotopic xenografts in nude mice showed increased expression of GSC and EC markers after TMZ treatment. Ex vivo FACS analysis showed the presence of immature and mature EC populations. Furthermore, immunofluorescence analysis revealed increased tumor-derived vessels in TMZ-recurrent tumors. Overall, this study identifies chemotherapeutic stress as a new driver of transdifferentiation of tumor cells to endothelial cells and highlights cellular plasticity as a key player in therapeutic resistance and tumor recurrence.

9.
Cell Death Dis ; 10(4): 292, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30926789

RESUMEN

Emerging evidence reveals enrichment of glioma-initiating cells (GICs) following therapeutic intervention. One factor known to contribute to this enrichment is cellular plasticity-the ability of glioma cells to attain multiple phenotypes. To elucidate the molecular mechanisms governing therapy-induced cellular plasticity, we performed genome-wide chromatin immunoprecipitation sequencing (ChIP-Seq) and gene expression analysis (gene microarray analysis) during treatment with standard of care temozolomide (TMZ) chemotherapy. Analysis revealed significant enhancement of open-chromatin marks in known astrocytic enhancers for interleukin-8 (IL-8) loci as well as elevated expression during anti-glioma chemotherapy. The Cancer Genome Atlas and Ivy Glioblastoma Atlas Project data demonstrated that IL-8 transcript expression is negatively correlated with GBM patient survival (p = 0.001) and positively correlated with that of genes associated with the GIC phenotypes, such as KLF4, c-Myc, and HIF2α (p < 0.001). Immunohistochemical analysis of patient samples demonstrated elevated IL-8 expression in about 60% of recurrent GBM tumors relative to matched primary tumors and this expression also positively correlates with time to recurrence. Exposure to IL-8 significantly enhanced the self-renewing capacity of PDX GBM (average threefold, p < 0.0005), as well as increasing the expression of GIC markers in the CXCR2 population. Furthermore, IL-8 knockdown significantly delayed PDX GBM tumor growth in vivo (p < 0.0005). Finally, guided by in silico analysis of TCGA data, we examined the effect of therapy-induced IL-8 expression on the epigenomic landscape of GBM cells and observed increased trimethylation of H3K9 and H3K27. Our results show that autocrine IL-8 alters cellular plasticity and mediates alterations in histone status. These findings suggest that IL-8 signaling participates in regulating GBM adaptation to therapeutic stress and therefore represents a promising target for combination with conventional chemotherapy in order to limit GBM recurrence.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Carcinogénesis/genética , Plasticidad de la Célula/efectos de los fármacos , Plasticidad de la Célula/genética , Glioblastoma/metabolismo , Interleucina-8/metabolismo , Receptores de Interleucina-8B/metabolismo , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Técnicas de Silenciamiento del Gen , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Histonas/metabolismo , Humanos , Interleucina-8/genética , Factor 4 Similar a Kruppel , Ratones , Ratones Desnudos , Recurrencia Local de Neoplasia/genética , Temozolomida/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Neurosci ; 39(11): 1982-1993, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30651332

RESUMEN

Glioblastoma (GBM) is one of the most aggressive and lethal tumor types. Evidence continues to accrue indicating that the complex relationship between GBM and the brain microenvironment contributes to this malignant phenotype. However, the interaction between GBM and neurotransmitters, signaling molecules involved in neuronal communication, remains incompletely understood. Here we examined, using human patient-derived xenograft lines, how the monoamine dopamine influences GBM cells. We demonstrate that GBM cells express dopamine receptor 2 (DRD2), with elevated expression in the glioma-initiating cell (GIC) population. Stimulation of DRD2 caused a neuron-like hyperpolarization exclusively in GICs. In addition, long-term activation of DRD2 heightened the sphere-forming capacity of GBM cells, as well as tumor engraftment efficiency in both male and female mice. Mechanistic investigation revealed that DRD2 signaling activates the hypoxia response and functionally alters metabolism. Finally, we found that GBM cells synthesize and secrete dopamine themselves, suggesting a potential autocrine mechanism. These results identify dopamine signaling as a potential therapeutic target in GBM and further highlight neurotransmitters as a key feature of the pro-tumor microenvironment.SIGNIFICANCE STATEMENT This work offers critical insight into the role of the neurotransmitter dopamine in the progression of GBM. We show that dopamine induces specific changes in the state of tumor cells, augmenting their growth and shifting them to a more stem-cell like state. Further, our data illustrate that dopamine can alter the metabolic behavior of GBM cells, increasing glycolysis. Finally, this work demonstrates that GBM cells, including tumor samples from patients, can synthesize and secrete dopamine, suggesting an autocrine signaling process underlying these results. These results describe a novel connection between neurotransmitters and brain cancer, further highlighting the critical influence of the brain milieu on GBM.


Asunto(s)
Glioblastoma/metabolismo , Receptores de Dopamina D2/metabolismo , Transcriptoma , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular Tumoral , Dopamina/biosíntesis , Epigénesis Genética , Femenino , Xenoinjertos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Transducción de Señal
12.
Am J Ophthalmol ; 182: 133-140, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28784553

RESUMEN

PURPOSE: To analyze trends in utilization and payment of ophthalmic services in the Medicare population for years 2012 and 2013. DESIGN: Retrospective, cross-sectional study. METHODS: A retrospective cross-sectional observational analysis was performed using publicly available Medicare Physician and Other Supplier aggregate file and the Physician and Other Supplier Public Use File. Variables analyzed included aggregate beneficiary demographics, Medicare payments to ophthalmologists, ophthalmic medical services provided, and the most common Medicare-reimbursed ophthalmic services. RESULTS: In 2013, total Medicare Part B reimbursement for ophthalmology was $5.8 billion, an increase of 3.6% from the previous year. From 2012 to 2013, the total number of ophthalmology services rendered increased by 2.2%, while average dollar amount reimbursed per ophthalmic service decreased by 5.4%. The top 5 highest reimbursed services accounted for 85% of total ophthalmic Medicare payments in 2013, an 11% increase from 2012. During 2013, drug reimbursement represented 32.8% of the total Medicare payments to ophthalmologists. Ranibizumab and aflibercept alone accounted for 95% of the entire $1.9 billion in drug reimbursements ophthalmologists in 2013. CONCLUSION: Medicare Part B reimbursement for ophthalmologists was primarily driven by use of anti-vascular endothelial growth factor (anti-VEGF) injections from 2012 to 2013. Of the total drug payments to ophthalmologists, biologic anti-VEGF agents ranibizumab and aflibercept accounted for 95% of all drug reimbursement. This is in contrast to other specialties, in which drug reimbursement represented only a small portion of Medicare reimbursement.


Asunto(s)
Atención a la Salud/tendencias , Reembolso de Seguro de Salud/tendencias , Medicare Part B/economía , Oftalmólogos/economía , Anciano , Anciano de 80 o más Años , Inhibidores de la Angiogénesis/economía , Estudios Transversales , Utilización de Medicamentos/tendencias , Femenino , Investigación sobre Servicios de Salud , Humanos , Masculino , Medicare Part B/tendencias , Oftalmólogos/tendencias , Ranibizumab/economía , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión/economía , Estudios Retrospectivos , Estados Unidos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
13.
Transl Cancer Res ; 6(Suppl 2): S321-S327, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30662831

RESUMEN

It remains incumbent on researchers to conceive novel treatments for the most common primary malignancy of the brain in adults, glioblastoma multiforme (GBM), as the standard of care for patients today fails to yield a median survival beyond two years following diagnosis. Recent studies have tended towards appreciating the cellular heterogeneity of GBM tumors, focusing on the subpopulation of highly plastic glioblastoma stem cells (GSCs). In the November 2016 issue of Cell, Hu and colleagues developed a de nova GBM model derived from immortalized neural stem cells and, using this model, they demonstrated that GSCs can generate CD133+/CD144+ cells with endothelial cell-like characteristics. Contrasts between the epigenetic state and gene expression level before and after oncogenic transformation of this utilized de novo model for GBM implicated WNT5A, which has been previously shown to play a role in endothelial cell proliferation and migration via non-canonical Wnt signaling, as a mediator of the process. The transdifferentiation was accompanied by alterations in the histone marks at the gene loci of WNT5A, and its transcription factors PAX6 and DXL5. The authors hypothesize that activation of AKT, an aberration of the RTK/PTEN/PI3K pathway observed in the majority of GBM cases, triggers these epigenetic changes causing WNT5A expression. This phenomenon is of obvious clinical significance, as it provides an insight into how GBM may circumvent therapies targeting angiogenesis to achieve the neovascularization required to sustain invasive growth. The unveiling of this atypical differentiation process also raises questions about its interaction with the radiotherapy and chemotherapy commonly used to counter GBM progression. Here, we review the recent efforts to understand the complex mechanisms behind the plasticity of GSCs.

14.
Ophthalmic Surg Lasers Imaging Retina ; 47(6): 555-62, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27327285

RESUMEN

BACKGROUND AND OBJECTIVE: To evaluate variation in physician use of vascular endothelial growth factor (VEGF) inhibitors. PATIENTS AND METHODS: Population-based analysis of comprehensive, publicly available 2012 Medicare claims, aggregated by physician specialty and service type - including intravitreal injections of bevacizumab (Avastin; Genentech, South San Francisco, CA), ranibizumab (Lucentis; Genetech, South San Francisco, CA), and aflibercept (Eylea; Regeneron, Tarrytown, NY). Physicians were characterized by total patients treated, proportion treated with each drug, total intravitreal injection payments, and proportion of total payments for each drug. RESULTS: The authors identified 2,869 ophthalmologists. On average, each treated 203 patients with VEGF-inhibitors, 75.9% of which were treated with bevacizumab. Using all three agents was the most common practice (1,121 physicians), closely followed by using bevacizumab only (1,061 physicians). Ranibizumab accounted for most payments, but bevacizumab was the largest payment source for a sizeable proportion of physicians who used only/mostly bevacizumab. CONCLUSION: Most ophthalmologists use multiple VEGF inhibitors, but vary in their relative use. A subset of ophthalmologists predominantly use ranibizumab, but ophthalmologists overall use more bevacizumab despite financial incentives favoring ranibizumab. [Ophthalmic Surg Lasers Imaging Retina. 2016;47:555-562.].


Asunto(s)
Bevacizumab/uso terapéutico , Utilización de Medicamentos/tendencias , Medicare/estadística & datos numéricos , Oftalmólogos , Ranibizumab/uso terapéutico , Receptores de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/provisión & distribución , Humanos , Inyecciones Intravítreas , Vigilancia de la Población/métodos , Estudios Retrospectivos , Tomografía de Coherencia Óptica , Estados Unidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA