Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Oncologist ; 27(2): 89-96, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35641208

RESUMEN

PURPOSE: Provide real-world data regarding the risk for SARS-CoV-2 infection and mortality in breast cancer (BC) patients on active cancer treatment. METHODS: Clinical data were abstracted from the 3778 BC patients seen at a multisite cancer center in New York between February 1, 2020 and May 1, 2020, including patient demographics, tumor histology, cancer treatment, and SARS-CoV-2 testing results. Incidence of SARS-CoV-2 infection by treatment type (chemotherapy [CT] vs endocrine and/or HER2 directed therapy [E/H]) was compared by Inverse Probability of Treatment Weighting. In those diagnosed with SARS-CoV-2 infection, Mann-Whitney test was used to a assess risk factors for severe disease and mortality. RESULTS: Three thousand sixty-two patients met study inclusion criteria with 641 patients tested for SARS-COV-2 by RT-PCR or serology. Overall, 64 patients (2.1%) were diagnosed with SARS-CoV-2 infection by either serology, RT-PCR, or documented clinical diagnosis. Comparing matched patients who received chemotherapy (n = 379) with those who received non-cytotoxic therapies (n = 2343) the incidence of SARS-CoV-2 did not differ between treatment groups (weighted risk; 3.5% CT vs 2.7% E/H, P = .523). Twenty-seven patients (0.9%) expired over follow-up, with 10 deaths attributed to SARS-CoV-2 infection. Chemotherapy was not associated with increased risk for death following SARS-CoV-2 infection (weighted risk; 0.7% CT vs 0.1% E/H, P = .246). Advanced disease (stage IV), age, BMI, and Charlson's Comorbidity Index score were associated with increased mortality following SARS-CoV-2 infection (P ≤ .05). CONCLUSION: BC treatment, including chemotherapy, can be safely administered in the context of enhanced infectious precautions, and should not be withheld particularly when given for curative intent.


Asunto(s)
Neoplasias de la Mama , COVID-19 , Terapia Biológica , Neoplasias de la Mama/tratamiento farmacológico , COVID-19/epidemiología , Prueba de COVID-19 , Femenino , Humanos , Pandemias , SARS-CoV-2 , Espera Vigilante
3.
Cancers (Basel) ; 16(1)2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38201446

RESUMEN

BACKGROUND: We previously reported a higher incidence of a pathogenic germline variant in the kinase insert domain receptor (KDR) in melanoma patients compared to the general population. Here, we dissect the impact of this genotype on melanoma tumor growth kinetics, tumor phenotype, and response to treatment with immune checkpoint inhibitors (ICIs) or targeted therapy. METHODS: The KDR genotype was determined and the associations between the KDR Q472H variant (KDR-Var), angiogenesis, tumor immunophenotype, and response to MAPK inhibition or ICI treatment were examined. Melanoma B16 cell lines were transfected with KDR-Var or KDR wild type (KDR-WT), and the differences in tumor kinetics were evaluated. We also examined the impact of KDR-Var on the response of melanoma cells to a combination of VEGFR inhibition with MAPKi. RESULTS: We identified the KDR-Var genotype in 81/489 (37%) patients, and it was associated with a more angiogenic (p = 0.003) and immune-suppressive tumor phenotype. KDR-Var was also associated with decreased PFS to MAPKi (p = 0.022) and a trend with worse PFS to anti-PD1 therapy (p = 0.06). KDR-Var B16 murine models had increased average tumor volume (p = 0.0027) and decreased CD45 tumor-infiltrating lymphocytes (p = 0.0282). The anti-VEGFR treatment Lenvatinib reduced the tumor size of KDR-Var murine tumors (p = 0.0159), and KDR-Var cells showed synergistic cytotoxicity to the combination of dabrafenib and lenvatinib. CONCLUSIONS: Our data demonstrate a role of germline KDR-Var in modulating melanoma behavior, including response to treatment. Our data also suggest that anti-angiogenic therapy might be beneficial in patients harboring this genotype, which needs to be tested in clinical trials.

4.
JAMA Netw Open ; 6(8): e2327145, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37535354

RESUMEN

Importance: Anti-programmable cell death-1 (anti-PD-1) improves relapse-free survival when used as adjuvant therapy for high-risk resected melanoma. However, it can lead to immune-related adverse events (irAEs), which become chronic in approximately 40% of patients with high-risk melanoma treated with adjuvant anti-PD-1. Objective: To determine the incidence, characteristics, and long-term outcomes of chronic irAEs from adjuvant anti-PD-1 therapy. Design, Setting, and Participants: This retrospective multicenter cohort study analyzed patients treated with adjuvant anti-PD-1 therapy for advanced and metastatic melanoma between 2015 and 2022 from 6 institutions in the US and Australia with at least 18 months of evaluable follow-up after treatment cessation (range, 18.2 to 70.4 months). Main Outcomes and Measures: Incidence, spectrum, and ultimate resolution vs persistence of chronic irAEs (defined as those persisting at least 3 months after therapy cessation). Descriptive statistics were used to analyze categorical and continuous variables. Kaplan-Meier curves assessed survival, and Wilson score intervals were used to calculate CIs for proportions. Results: Among 318 patients, 190 (59.7%) were male (median [IQR] age, 61 [52.3-72.0] years), 270 (84.9%) had a cutaneous primary, and 237 (74.5%) were stage IIIB or IIIC at presentation. Additionally, 226 patients (63.7%) developed acute irAEs arising during treatment, including 44 (13.8%) with grade 3 to 5 irAEs. Chronic irAEs, persisting at least 3 months after therapy cessation, developed in 147 patients (46.2%; 95% CI, 0.41-0.52), of which 74 (50.3%) were grade 2 or more, 6 (4.1%) were grade 3 to 5, and 100 (68.0%) were symptomatic. With long-term follow-up (median [IQR], 1057 [915-1321] days), 54 patients (36.7%) experienced resolution of chronic irAEs (median [IQR] time to resolution of 19.7 [14.4-31.5] months from anti-PD-1 start and 11.2 [8.1-20.7] months from anti-PD-1 cessation). Among patients with persistent irAEs present at last follow-up (93 [29.2%] of original cohort; 95% CI, 0.25-0.34); 55 (59.1%) were grade 2 or more; 41 (44.1%) were symptomatic; 24 (25.8%) were using therapeutic systemic steroids (16 [67%] of whom were on replacement steroids for hypophysitis (8 [50.0%]) and adrenal insufficiency (8 [50.0%]), and 42 (45.2%) were using other management. Among the 54 patients, the most common persistent chronic irAEs were hypothyroid (38 [70.4%]), arthritis (18 [33.3%]), dermatitis (9 [16.7%]), and adrenal insufficiency (8 [14.8%]). Furthermore, 54 [17.0%] patients experienced persistent endocrinopathies, 48 (15.1%) experienced nonendocrinopathies, and 9 (2.8%) experienced both. Of 37 patients with chronic irAEs who received additional immunotherapy, 25 (67.6%) experienced no effect on chronic irAEs whereas 12 (32.4%) experienced a flare in their chronic toxicity. Twenty patients (54.1%) experienced a distinct irAE. Conclusions and Relevance: In this cohort study of 318 patients who received adjuvant anti-PD-1, chronic irAEs were common, affected diverse organ systems, and often persisted with long-term follow-up requiring steroids and additional management. These findings highlight the likelihood of persistent toxic effects when considering adjuvant therapies and need for long-term monitoring and management.


Asunto(s)
Insuficiencia Suprarrenal , Antineoplásicos Inmunológicos , Enfermedades del Sistema Inmune , Melanoma , Femenino , Humanos , Masculino , Persona de Mediana Edad , Insuficiencia Suprarrenal/inducido químicamente , Antineoplásicos Inmunológicos/efectos adversos , Estudios de Cohortes , Estudios de Seguimiento , Enfermedades del Sistema Inmune/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Melanoma/cirugía , Estudios Retrospectivos , Anciano
5.
J Immunother Cancer ; 11(6)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37328287

RESUMEN

BACKGROUND: Management of immune-related adverse events (irAEs) is important as they cause treatment interruption or discontinuation, more often seen with combination immune checkpoint inhibitor (ICI) therapy. Here, we retrospectively evaluated the safety and effectiveness of anti-interleukin-6 receptor (anti-IL-6R) as therapy for irAEs. METHODS: We performed a retrospective multicenter study evaluating patients diagnosed with de novo irAEs or flare of pre-existing autoimmune disease following ICI and were treated with anti-IL-6R. Our objectives were to assess the improvement of irAEs as well as the overall tumor response rate (ORR) before and after anti-IL-6R treatment. RESULTS: We identified a total of 92 patients who received therapeutic anti-IL-6R antibodies (tocilizumab or sarilumab). Median age was 61 years, 63% were men, 69% received anti-programmed cell death protein-1 (PD-1) antibodies alone, and 26% patients were treated with the combination of anti-cytotoxic T lymphocyte antigen-4 and anti-PD-1 antibodies. Cancer types were primarily melanoma (46%), genitourinary cancer (35%), and lung cancer (8%). Indications for using anti-IL-6R antibodies included inflammatory arthritis (73%), hepatitis/cholangitis (7%), myositis/myocarditis/myasthenia gravis (5%), polymyalgia rheumatica (4%), and one patient each with autoimmune scleroderma, nephritis, colitis, pneumonitis and central nervous system vasculitis. Notably, 88% of patients had received corticosteroids, and 36% received other disease-modifying antirheumatic drugs (DMARDs) as first-line therapies, but without adequate improvement. After initiation of anti-IL-6R (as first-line or post-corticosteroids and DMARDs), 73% of patients showed resolution or change to ≤grade 1 of irAEs after a median of 2.0 months from initiation of anti-IL-6R therapy. Six patients (7%) stopped anti-IL-6R due to adverse events. Of 70 evaluable patients by RECIST (Response Evaluation Criteria in Solid Tumors) V.1.1 criteria; the ORR was 66% prior versus 66% after anti-IL-6R (95% CI, 54% to 77%), with 8% higher complete response rate. Of 34 evaluable patients with melanoma, the ORR was 56% prior and increased to 68% after anti-IL-6R (p=0.04). CONCLUSION: Targeting IL-6R could be an effective approach to treat several irAE types without hindering antitumor immunity. This study supports ongoing clinical trials evaluating the safety and efficacy of tocilizumab (anti-IL-6R antibody) in combination with ICIs (NCT04940299, NCT03999749).


Asunto(s)
Antirreumáticos , Neoplasias Pulmonares , Melanoma , Receptores de Interleucina-6 , Femenino , Humanos , Masculino , Persona de Mediana Edad , Corticoesteroides/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Estudios Retrospectivos , Receptores de Interleucina-6/antagonistas & inhibidores
6.
J Clin Invest ; 132(13)2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35552271

RESUMEN

The inability of CD8+ effector T cells (Teffs) to reach tumor cells is an important aspect of tumor resistance to cancer immunotherapy. The recruitment of these cells to the tumor microenvironment (TME) is regulated by integrins, a family of adhesion molecules that are expressed on T cells. Here, we show that 7HP349, a small-molecule activator of lymphocyte function-associated antigen-1 (LFA-1) and very late activation antigen-4 (VLA-4) integrin cell-adhesion receptors, facilitated the preferential localization of tumor-specific T cells to the tumor and improved antitumor response. 7HP349 monotherapy had modest effects on anti-programmed death 1-resistant (anti-PD-1-resistant) tumors, whereas combinatorial treatment with anti-cytotoxic T lymphocyte-associated protein 4 (anti-CTLA-4) increased CD8+ Teff intratumoral sequestration and synergized in cooperation with neutrophils in inducing cancer regression. 7HP349 intratumoral CD8+ Teff enrichment activity depended on CXCL12. We analyzed gene expression profiles using RNA from baseline and on treatment tumor samples of 14 melanoma patients. We identified baseline CXCL12 gene expression as possibly improving the likelihood or response to anti-CTLA-4 therapies. Our results provide a proof-of-principle demonstration that LFA-1 activation could convert a T cell-exclusionary TME to a T cell-enriched TME through mechanisms involving cooperation with innate immune cells.


Asunto(s)
Antígeno-1 Asociado a Función de Linfocito , Melanoma , Linfocitos T CD8-positivos , Antígeno CTLA-4 , Humanos , Inmunoterapia/métodos , Antígeno-1 Asociado a Función de Linfocito/genética , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Linfocitos Infiltrantes de Tumor , Melanoma/tratamiento farmacológico , Melanoma/genética , Receptor de Muerte Celular Programada 1 , Linfocitos T/metabolismo , Microambiente Tumoral
7.
Cancer Cell ; 40(5): 509-523.e6, 2022 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-35537412

RESUMEN

Immune checkpoint blockade (ICB) therapy frequently induces immune-related adverse events. To elucidate the underlying immunobiology, we performed a deep immune analysis of intestinal, colitis, and tumor tissue from ICB-treated patients with parallel studies in preclinical models. Expression of interleukin-6 (IL-6), neutrophil, and chemotactic markers was higher in colitis than in normal intestinal tissue; T helper 17 (Th17) cells were more prevalent in immune-related enterocolitis (irEC) than T helper 1 (Th1). Anti-cytotoxic T-lymphocyte-associated antigen 4 (anti-CTLA-4) induced stronger Th17 memory in colitis than anti-program death 1 (anti-PD-1). In murine models, IL-6 blockade associated with improved tumor control and a higher density of CD4+/CD8+ effector T cells, with reduced Th17, macrophages, and myeloid cells. In an experimental autoimmune encephalomyelitis (EAE) model with tumors, combined IL-6 blockade and ICB enhanced tumor rejection while simultaneously mitigating EAE symptoms versus ICB alone. IL-6 blockade with ICB could de-couple autoimmunity from antitumor immunity.


Asunto(s)
Colitis , Neoplasias , Animales , Colitis/inducido químicamente , Humanos , Factores Inmunológicos/uso terapéutico , Inmunoterapia , Interleucina-6 , Ratones , Células Mieloides , Neoplasias/tratamiento farmacológico
8.
J Immunother Cancer ; 9(2)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33637601

RESUMEN

BACKGROUND: Immune checkpoint inhibitors (ICIs) are being used after allogeneic hematopoietic stem cell transplantation (alloHCT) to reverse immune dysfunction. However, a major concern for the use of ICIs after alloHCT is the increased risk of graft-versus-host disease (GVHD). We analyzed the association between GVHD prophylaxis and frequency of GVHD in patients who had received ICI therapy after alloHCT. METHODS: A retrospective study was performed in 21 patients with acute myeloid leukemia (n=16) or myelodysplastic syndromes (n=5) who were treated with antiprogrammed cell death protein 1 (16 patients) or anticytotoxic T lymphocyte-associated antigen 4 (5 patients) therapy for disease relapse after alloHCT. Associations between the type of GVHD prophylaxis and incidence of GVHD were analyzed. RESULTS: Four patients (19%) developed acute GVHD. The incidence of acute GVHD was associated only with the type of post-transplantation GVHD prophylaxis; none of the other variables included (stem cell source, donor type, age at alloHCT, conditioning regimen and prior history of GVHD) were associated with the frequency of acute GVHD. Twelve patients received post-transplantation cyclophosphamide (PTCy) for GVHD prophylaxis. Patients who received PTCy had a significantly shorter median time to initiation of ICI therapy after alloHCT compared with patients who did not receive PTCy (median 5.1 months compared with 26.6 months). Despite early ICI therapy initiation, patients who received PTCy had a lower observed cumulative incidence of grades 2-4 acute GVHD compared with patients who did not receive PTCy (16% compared with 22%; p=0.7). After controlling for comorbidities and time from alloHCT to ICI therapy initiation, the analysis showed that PTCy was associated with a 90% reduced risk of acute GVHD (HR 0.1, 95% CI 0.02 to 0.6, p=0.01). CONCLUSIONS: ICI therapy for relapsed acute myeloid leukemia/myelodysplastic syndromes after alloHCT may be a safe and feasible option. PTCy appears to decrease the incidence of acute GVHD in this cohort of patients.


Asunto(s)
Ciclofosfamida/administración & dosificación , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Inmunosupresores/administración & dosificación , Leucemia Mieloide Aguda/terapia , Síndromes Mielodisplásicos/terapia , Adulto , Anciano , Ciclofosfamida/efectos adversos , Bases de Datos Factuales , Esquema de Medicación , Femenino , Enfermedad Injerto contra Huésped/epidemiología , Enfermedad Injerto contra Huésped/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inmunosupresores/efectos adversos , Incidencia , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/inmunología , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/inmunología , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Texas/epidemiología , Trasplante Homólogo/efectos adversos , Resultado del Tratamiento
9.
Blood Adv ; 4(8): 1746-1755, 2020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32337583

RESUMEN

This review summarizes the evidence on antiphospholipid (aPL) antibodies and related thromboembolic events in patients with solid tumors. Data sources included Medline, EMBASE, Web of Science, PubMed ePubs, and the Cochrane Central Register of Controlled Trials through August 2019 without restrictions. Observational studies that evaluated patients with solid tumors for the presence of aPL antibodies were included. Data were extracted and quality was assessed by one reviewer and cross-checked by another. Thirty-three studies were identified. Gastrointestinal (GI) and genitourinary (GU) cancers were the most frequently reported. Compared with healthy patients, patients with GI cancer were more likely to develop anticardiolipin antibodies (risk ratio [RR], 5.1; 95% confidence interval [CI], 2.6-9.95), as were those with GU (RR, 7.3; 95% CI, 3.3-16.2) and lung cancer (RR, 5.2; 95% CI, 1.3-20.6). The increased risk for anti-ß2-glycoprotein I or lupus anticoagulant was not statistically significant. Patients with lung cancer who had positive aPL antibodies had higher risk of developing thromboembolic events than those who had negative antibodies (RR, 3.8%; 95% CI, 1.2-12.2), while the increased risk in patients with GU cancer was not statistically significant. Deaths due to thromboembolic events were more common among patients with lung cancer who had elevated aPL antibodies. A limitation of this review is that the results are contingent on the reported information. We found an increased risk of developing aPL antibodies in patients with GI, GU, and lung cancers resulting in thromboembolic events and death. Further studies are needed to better understand the pathogenesis and development of aPL antibodies in cancer.


Asunto(s)
Síndrome Antifosfolípido , Neoplasias , Tromboembolia , Anticuerpos Antifosfolípidos , Humanos , Inhibidor de Coagulación del Lupus , Neoplasias/epidemiología , Estudios Observacionales como Asunto , Tromboembolia/epidemiología , Tromboembolia/etiología
10.
Nat Commun ; 11(1): 661, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-32005826

RESUMEN

High dose interleukin-2 (IL-2) is active against metastatic melanoma and renal cell carcinoma, but treatment-associated toxicity and expansion of suppressive regulatory T cells (Tregs) limit its use in patients with cancer. Bempegaldesleukin (NKTR-214) is an engineered IL-2 cytokine prodrug that provides sustained activation of the IL-2 pathway with a bias to the IL-2 receptor CD122 (IL-2Rß). Here we assess the therapeutic impact and mechanism of action of NKTR-214 in combination with anti-PD-1 and anti-CTLA-4 checkpoint blockade therapy or peptide-based vaccination in mice. NKTR-214 shows superior anti-tumor activity over native IL-2 and systemically expands anti-tumor CD8+ T cells while inducing Treg depletion in tumor tissue but not in the periphery. Similar trends of intratumoral Treg dynamics are observed in a small cohort of patients treated with NKTR-214. Mechanistically, intratumoral Treg depletion is mediated by CD8+ Teff-associated cytokines IFN-γ and TNF-α. These findings demonstrate that NKTR-214 synergizes with T cell-mediated anti-cancer therapies.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Interleucina-2/análogos & derivados , Melanoma/tratamiento farmacológico , Polietilenglicoles/administración & dosificación , Profármacos/administración & dosificación , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/inmunología , Estudios de Cohortes , Quimioterapia Combinada , Femenino , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-2/administración & dosificación , Interleucina-2/agonistas , Interleucina-2/inmunología , Ipilimumab/administración & dosificación , Activación de Linfocitos/efectos de los fármacos , Melanoma/genética , Melanoma/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-2/genética , Receptores de Interleucina-2/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
11.
Am J Case Rep ; 20: 785-789, 2019 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-31160547

RESUMEN

BACKGROUND Advanced urothelial carcinoma has been associated with poor prognosis due to high resistance to chemotherapy and radiation until immunotherapeutic agents, such as atezolizumab, emerged as an option and have shown improved survival. However, atezolizumab is associated with side effects, which were mainly autoimmune. In this case study, we report on a rare case of atezolizumab-induced tumor lysis syndrome. CASE REPORT A 67-year-old female with a primary diagnosis of metastatic urothelial carcinoma who presented to the emergency department with generalized weakness associated with nausea and vomiting 8 days after her first cycle of atezolizumab. Laboratory values showed hyperphosphatemia, hyperuricemia, hypocalcemia, and acute kidney injury consistent with tumor lysis syndrome. CONCLUSIONS In our report, we highlight tumor lysis syndrome as a potential reaction to atezolizumab; a condition that requires prophylaxis and close laboratory monitoring.


Asunto(s)
Anticuerpos Monoclonales Humanizados/efectos adversos , Carcinoma de Células Transicionales/patología , Síndrome de Lisis Tumoral/etiología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología , Anciano , Anticuerpos Monoclonales Humanizados/administración & dosificación , Biopsia con Aguja , Carcinoma de Células Transicionales/diagnóstico por imagen , Carcinoma de Células Transicionales/tratamiento farmacológico , Progresión de la Enfermedad , Servicio de Urgencia en Hospital , Femenino , Cuidados Paliativos al Final de la Vida , Humanos , Inmunohistoquímica , Monitoreo Fisiológico , Invasividad Neoplásica/patología , Estadificación de Neoplasias , Tomografía Computarizada por Rayos X/métodos , Síndrome de Lisis Tumoral/fisiopatología , Neoplasias de la Vejiga Urinaria/diagnóstico por imagen
12.
J Immunother Cancer ; 7(1): 319, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31753014

RESUMEN

BACKGROUND: Myasthenia gravis (MG) is a rare but life-threatening adverse event of immune checkpoint inhibitors (ICI). Given the limited evidence, data from a large cohort of patients is needed to aid in recognition and management of this fatal complication. METHODS: We reviewed our institutional databases to identify patients who had cancer and MG in the setting of ICI. We systematically reviewed the literature through August 2018 to identify all similar reported patients. We collected data on clinical and diagnostic features, management, and outcomes of these cases. RESULTS: Sixty-five patients were identified. Median age was 73 years; 42 (65%) were males, 31 (48%) had metastatic melanoma, and 13 (20%) had a preexisting MG before ICI initiation. Most patients received anti-PD-1 (82%). Sixty-three patients (97%) developed ICI-related MG (new onset or disease flare) after a median of 4 weeks (1 to 16 weeks) of ICI initiation. Twenty-four patients (37%) experienced concurrent myositis, and respiratory failure occurred in 29 (45%). ICI was discontinued in 61 patients (97%). Death was reported in 24 patients (38%); 15 (23%) due to MG complication. A better outcome was observed in patients who received intravenous immunoglobulin (IVIG) or plasmapheresis (PLEX) as first-line therapy than in those who received steroids alone (95% vs 63% improvement of MG symptoms, p = 0.011). CONCLUSIONS: MG is a life-threatening adverse event of acute onset and rapid progression after ICI initiation. Early use of IVIG or PLEX, regardless of initial symptoms severity, may lead to better outcomes than steroids alone. Our data suggest the need to reassess the current recommendations for management of ICI-related MG until prospective longitudinal studies are conducted to establish the ideal management approach for these patients.


Asunto(s)
Antineoplásicos Inmunológicos/efectos adversos , Miastenia Gravis/etiología , Neoplasias/complicaciones , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Inmunológicos/uso terapéutico , Biomarcadores , Progresión de la Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida/efectos adversos , Miastenia Gravis/diagnóstico , Miastenia Gravis/epidemiología , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Evaluación de Síntomas
14.
Tex Heart Inst J ; 43(6): 503-506, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28100968

RESUMEN

Left atrial pressure indicates the left ventricular filling pressure in patients who have systolic or diastolic left ventricular dysfunction or valvular heart disease. The use of indirect surrogate methods to determine left atrial pressure has been essential in the modern evaluation and treatment of cardiovascular disease because of the difficulty and inherent risks associated with direct methods (typically the transseptal approach). One method that has been widely used to determine left atrial pressure indirectly is Swan-Ganz catheterization, in which a balloon-flotation technique is applied to measure pulmonary capillary wedge pressure; however, this approach has been associated with several limitations and potential risks. Measuring left ventricular end-diastolic pressure has also been widely used as a simple means to estimate filling pressures but remains a surrogate for the gold standard of directly measuring left atrial pressure. We describe a simple, low-risk method to directly measure left atrial pressure that involves the use of standard coronary catheterization techniques during a transradial procedure.


Asunto(s)
Función del Atrio Izquierdo , Presión Atrial , Cateterismo Cardíaco/métodos , Cateterismo Periférico/métodos , Insuficiencia Cardíaca/diagnóstico , Arteria Radial , Cateterismo Cardíaco/instrumentación , Catéteres Cardíacos , Angiografía Coronaria , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Presión Esfenoidal Pulmonar , Punciones , Reproducibilidad de los Resultados , Transductores de Presión , Función Ventricular Izquierda , Presión Ventricular
15.
Dis Model Mech ; 9(3): 335-45, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26935106

RESUMEN

HIRA is the histone chaperone responsible for replication-independent incorporation of histone variant H3.3 within gene bodies and regulatory regions of actively transcribed genes, and within the bivalent promoter regions of developmentally regulated genes. The HIRA gene lies within the 22q11.2 deletion syndrome critical region; individuals with this syndrome have multiple congenital heart defects. Because terminally differentiated cardiomyocytes have exited the cell cycle, histone variants should be utilized for the bulk of chromatin remodeling. Thus, HIRA is likely to play an important role in epigenetically defining the cardiac gene expression program. In this study, we determined the consequence of HIRA deficiency in cardiomyocytes in vivo by studying the phenotype of cardiomyocyte-specific Hira conditional-knockout mice. Loss of HIRA did not perturb heart development, but instead resulted in cardiomyocyte hypertrophy and susceptibility to sarcolemmal damage. Cardiomyocyte degeneration gave way to focal replacement fibrosis and impaired cardiac function. Gene expression was widely altered in Hira conditional-knockout hearts. Significantly affected pathways included responses to cellular stress, DNA repair and transcription. Consistent with heart failure, fetal cardiac genes were re-expressed in the Hira conditional knockout. Our results suggest that transcriptional regulation by HIRA is crucial for cardiomyocyte homeostasis.


Asunto(s)
Cardiomegalia/metabolismo , Cardiomegalia/patología , Proteínas de Ciclo Celular/deficiencia , Chaperonas de Histonas/deficiencia , Miocitos Cardíacos/metabolismo , Sarcolema/metabolismo , Sarcolema/patología , Factores de Transcripción/deficiencia , Animales , Apoptosis/genética , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Daño del ADN/genética , Reparación del ADN/genética , Feto/metabolismo , Regulación de la Expresión Génica , Pruebas de Función Cardíaca , Chaperonas de Histonas/metabolismo , Ratones Noqueados , Miocitos Cardíacos/patología , Especificidad de Órganos , Estrés Oxidativo/genética , Reproducibilidad de los Resultados , Estrés Fisiológico/genética , Factores de Transcripción/metabolismo , Transcriptoma/genética
16.
J Immunother Cancer ; 4: 55, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27660709

RESUMEN

BACKGROUND: Immune related adverse events (irAEs) are common side effects of checkpoint inhibitory (CPI) therapies targeting CTLA-4 and PD-1/PD-L1. Grover's disease is an uncommon dermatologic condition with unclear pathogenesis previously reported as an irAE with ipilimumab. CASE PRESENTATION: We report an additional case of ipilimumab-induced Grover's disease. Interestingly, this dermatologic side effect did not appear with use of anti-PD-1 therapy in our patient. Immune analysis was performed and suggests a possible role of Th2 cells in its patholgenesis. CONCLUSION: This case suggests that Grover's disease is an irAE induced by Ipilimumab. Our immune analysis suggests that Th2 cells may be pathogenic mediators which warrants further study.

17.
Sci Rep ; 6: 31457, 2016 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-27538477

RESUMEN

Mesp1 directs multipotential cardiovascular cell fates, even though it's transiently induced prior to the appearance of the cardiac progenitor program. Tracing Mesp1-expressing cells and their progeny allows isolation and characterization of the earliest cardiovascular progenitor cells. Studying the biology of Mesp1-CPCs in cell culture and ischemic disease models is an important initial step toward using them for heart disease treatment. Because of Mesp1's transitory nature, Mesp1-CPC lineages were traced by following EYFP expression in murine Mesp1(Cre/+); Rosa26(EYFP/+) ES cells. We captured EYFP+ cells that strongly expressed cardiac mesoderm markers and cardiac transcription factors, but not pluripotent or nascent mesoderm markers. BMP2/4 treatment led to the expansion of EYFP+ cells, while Wnt3a and Activin were marginally effective. BMP2/4 exposure readily led EYFP+ cells to endothelial and smooth muscle cells, but inhibition of the canonical Wnt signaling was required to enter the cardiomyocyte fate. Injected mouse pre-contractile Mesp1-EYFP+ CPCs improved the survivability of injured mice and restored the functional performance of infarcted hearts for at least 3 months. Mesp1-EYFP+ cells are bona fide CPCs and they integrated well in infarcted hearts and emerged de novo into terminally differentiated cardiac myocytes, smooth muscle and vascular endothelial cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Infarto del Miocardio/terapia , Trasplante de Células Madre , Células Madre/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteína Morfogenética Ósea 2/farmacología , Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Corazón/diagnóstico por imagen , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones SCID , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal/efectos de los fármacos , Células Madre/citología , Transcriptoma , Proteína Wnt3A/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA