Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nature ; 615(7953): 697-704, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36890230

RESUMEN

Neoantigens are peptides derived from non-synonymous mutations presented by human leukocyte antigens (HLAs), which are recognized by antitumour T cells1-14. The large HLA allele diversity and limiting clinical samples have restricted the study of the landscape of neoantigen-targeted T cell responses in patients over their treatment course. Here we applied recently developed technologies15-17 to capture neoantigen-specific T cells from blood and tumours from patients with metastatic melanoma with or without response to anti-programmed death receptor 1 (PD-1) immunotherapy. We generated personalized libraries of neoantigen-HLA capture reagents to single-cell isolate the T cells and clone their T cell receptors (neoTCRs). Multiple T cells with different neoTCR sequences (T cell clonotypes) recognized a limited number of mutations in samples from seven patients with long-lasting clinical responses. These neoTCR clonotypes were recurrently detected over time in the blood and tumour. Samples from four patients with no response to anti-PD-1 also demonstrated neoantigen-specific T cell responses in the blood and tumour to a restricted number of mutations with lower TCR polyclonality and were not recurrently detected in sequential samples. Reconstitution of the neoTCRs in donor T cells using non-viral CRISPR-Cas9 gene editing demonstrated specific recognition and cytotoxicity to patient-matched melanoma cell lines. Thus, effective anti-PD-1 immunotherapy is associated with the presence of polyclonal CD8+ T cells in the tumour and blood specific for a limited number of immunodominant mutations, which are recurrently recognized over time.


Asunto(s)
Antígenos de Neoplasias , Linfocitos T CD8-positivos , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia , Melanoma , Humanos , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Antígenos HLA/inmunología , Metástasis de la Neoplasia , Medicina de Precisión , Edición Génica , Sistemas CRISPR-Cas , Mutación
2.
Semin Oncol Nurs ; 37(2): 151131, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33785232

RESUMEN

OBJECTIVE: It is an exciting time in oncology nursing as new treatments are continually emerging to improve oncological outcomes for people affected by cancer. Immunotherapy is one such innovation in contemporary oncology practice, but the side effects of its treatment can be life threatening. Oncology nurses must be educated in the early identification of life-threatening side effects, and deliver evidence-based interventions to manage consequences of therapy such as, immune checkpoint inhibitor-related adrenal insufficiency. DATA SOURCE: This article utilizes a case study to emphasize the need for early identification and interventions for clinical management of immunotherapy related adrenal insufficiency. A review of the current evidence is included to provide recommendations for oncology nurses who are involved in providing care for people affected by cancer receiving immunotherapy. IMPLICATIONS FOR NURSING PRACTICE: Oncology nurses are the frontline caregivers to assess, recognize, so that early intervention and treament can lead to optimal patient outcomes. CONCLUSION: Adrenal insufficiency is a life-threatening condition that is difficult to diagnose but if recognized early, morbidity can be prevented, quality of life improved, treatment initiated and life saving immunotherapy treatment can safely continue. Oncology nurses play an active role in ensuring the safe administration of immunotherapy by recognizing this important immune related adverse event. I would like to give credit to Patricia Jakel, RN, MSN, UCLA CNS and Dr. Melissa Lechner, UCLA endocrinologist for their invaluable input in this article.


Asunto(s)
Insuficiencia Suprarrenal , Inhibidores de Puntos de Control Inmunológico , Insuficiencia Suprarrenal/inducido químicamente , Insuficiencia Suprarrenal/diagnóstico , Insuficiencia Suprarrenal/terapia , Humanos , Inmunoterapia/efectos adversos , Enfermería Oncológica , Calidad de Vida
3.
Immunotherapy ; 13(4): 283-288, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33397120

RESUMEN

Renal toxicity from immune checkpoint inhibitors (ICIs) is an increasingly recognized cause of acute kidney injury among patients with cancer. ICI-associated acute kidney injuries typically present as acute interstitial nephritis and the timing of onset is highly variable. Herein, we present a case of a patient with relapsed metastatic melanoma previously treated with pembrolizumab who developed grade 3 immune-related renal toxicity after reintroduction of the same ICI, secondary to acute interstitial nephritis with accompanying high PR3-antineutrophil cytoplasmic antibody titer. The patient improved after steroid treatment and discontinuation of pembrolizumab. This case highlights the importance of not excluding ICI-related nephrotoxicity as a possible cause of renal failure, including in those who previously tolerated ICI treatment, since it is a treatable entity.


Asunto(s)
Anticuerpos Anticitoplasma de Neutrófilos/efectos de los fármacos , Anticuerpos Monoclonales Humanizados/efectos adversos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Nefritis Intersticial/inducido químicamente , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Anciano , Anticuerpos Anticitoplasma de Neutrófilos/sangre , Antígeno B7-H1/metabolismo , Femenino , Humanos , Inmunoterapia/efectos adversos , Melanoma/tratamiento farmacológico , Melanoma/patología , Nefritis Intersticial/tratamiento farmacológico , Nefritis Intersticial/metabolismo , Nefritis Intersticial/patología , Recurrencia , Esteroides/uso terapéutico , Resultado del Tratamiento
4.
Ther Adv Med Oncol ; 10: 1758834017751634, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29383039

RESUMEN

BACKGROUND: The anti-programmed cell death-1 (PD-1) and anti-programmed cell death ligand-1 (PD-L1) immunotherapies have shown exceptional activity in many cancers. However, these immunotherapies can also result in diverse adverse cutaneous eruptions that need to be better characterized for ongoing management. The objective was to provide clinical and histopathologic descriptions of the variety of cutaneous adverse events seen in patients who received anti-PD-1/PD-L1 treatment and discuss their management. METHODS: Patients with advanced cancers in clinical trials at University of California Los Angeles (UCLA), receiving anti-PD-1/PD-L1 treatment between 2012 and 2016 who developed cutaneous eruptions and were evaluated in the dermatology clinic were included in this retrospective case series study. A total of 16 patients were included in this study; of these, five were treated with pembrolizumab alone, two with avelumab alone, eight with nivolumab plus ipilimumab and one with nivolumab plus T-Vec. Of these 16 patients, eight had received systemic chemotherapy, six had received radiotherapy, and one had received trememlimumab prior to the immunotherapies described in this study. RESULTS: Cutaneous eruptions occurred at variable times, from week 1 to 88, with a median of 11.5 weeks; the morphologies included lichenoid, bullous, psoriasiform, macular, morbiliform appearances, and alopecia which were confirmed histopathologically in several of the cases. All cutaneous immune-related adverse events were either grade 1 or 2. Ten patients were treated with topical corticosteroids, and one also received NBUVB. Four patients eventually required systemic steroids. Three required discontinuation of their anti-PD-1/PD-L1 therapy secondary to the cutaneous eruptions. CONCLUSIONS: There are several different types of adverse cutaneous morphologies that may be seen with administration of PD-1 and PD-L1 inhibitors. Identifying the patterns of eruption may assist in prompt treatment. Most eruptions could be managed with topical corticosteroids and without discontinuation of the systemic treatment.

5.
Clin Endocrinol (Oxf) ; 88(2): 327-332, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28941311

RESUMEN

BACKGROUND: Immunomodulatory therapies, including CTLA-4 and PD-1 inhibitors, provide a directed attack against cancer cells by preventing T cell deactivation. However, these drugs also prevent the downregulation of auto-reactive T cells, resulting in immune-related adverse events (IRAEs). Reports show a varied incidence of endocrine IRAEs, ranging from 0% to 63%. OBJECTIVE: To describe the frequency and clinical characteristics of endocrine IRAEs in patients taking cancer immunomodulatory therapies. DESIGN: Retrospective cohort study. PATIENTS: A total of 388 patients aged ≥18 years who were prescribed ipilimumab, nivolumab and/or pembrolizumab between 2009 and 2016 at our institution. MEASUREMENTS: Biochemical criteria were used to define endocrine IRAEs, including thyroid, pituitary, pancreas and adrenal dysfunction, following use of immunomodulatory therapies. RESULTS: Fifty endocrine IRAEs occurred in our cohort, corresponding to a rate of 12.9%. The most common endocrine IRAEs were thyroid dysfunction (11.1%), with a lower incidence of pituitary dysfunction (1.8% of patients). CONCLUSIONS: Over 12% of patients receiving ipilimumab, nivolumab and/or pembrolizumab in our study sample developed an endocrine IRAE. Patients who undergo treatment with immunomodulatory therapies should be monitored for the development of endocrine IRAEs.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Enfermedades del Sistema Endocrino/terapia , Inmunoterapia/métodos , Anciano , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígeno CTLA-4/antagonistas & inhibidores , Enfermedades del Sistema Endocrino/sangre , Enfermedades del Sistema Endocrino/tratamiento farmacológico , Femenino , Humanos , Hipotiroidismo/sangre , Hipotiroidismo/tratamiento farmacológico , Hipotiroidismo/terapia , Ipilimumab/uso terapéutico , Masculino , Persona de Mediana Edad , Nivolumab/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Estudios Retrospectivos
7.
Cancer Discov ; 7(2): 188-201, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27903500

RESUMEN

Loss-of-function mutations in JAK1/2 can lead to acquired resistance to anti-programmed death protein 1 (PD-1) therapy. We reasoned that they may also be involved in primary resistance to anti-PD-1 therapy. JAK1/2-inactivating mutations were noted in tumor biopsies of 1 of 23 patients with melanoma and in 1 of 16 patients with mismatch repair-deficient colon cancer treated with PD-1 blockade. Both cases had a high mutational load but did not respond to anti-PD-1 therapy. Two out of 48 human melanoma cell lines had JAK1/2 mutations, which led to a lack of PD-L1 expression upon interferon gamma exposure mediated by an inability to signal through the interferon gamma receptor pathway. JAK1/2 loss-of-function alterations in The Cancer Genome Atlas confer adverse outcomes in patients. We propose that JAK1/2 loss-of-function mutations are a genetic mechanism of lack of reactive PD-L1 expression and response to interferon gamma, leading to primary resistance to PD-1 blockade therapy. SIGNIFICANCE: A key functional result from somatic JAK1/2 mutations in a cancer cell is the inability to respond to interferon gamma by expressing PD-L1 and many other interferon-stimulated genes. These mutations result in a genetic mechanism for the absence of reactive PD-L1 expression, and patients harboring such tumors would be unlikely to respond to PD-1 blockade therapy. Cancer Discov; 7(2); 188-201. ©2016 AACR.See related commentary by Marabelle et al., p. 128This article is highlighted in the In This Issue feature, p. 115.


Asunto(s)
Resistencia a Antineoplásicos , Janus Quinasa 1/genética , Janus Quinasa 2/genética , Mutación , Neoplasias/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/farmacología , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Interferón gamma/farmacología , Melanoma/tratamiento farmacológico , Melanoma/genética , Neoplasias/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos
8.
N Engl J Med ; 375(9): 819-29, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27433843

RESUMEN

BACKGROUND: Approximately 75% of objective responses to anti-programmed death 1 (PD-1) therapy in patients with melanoma are durable, lasting for years, but delayed relapses have been noted long after initial objective tumor regression despite continuous therapy. Mechanisms of immune escape in this context are unknown. METHODS: We analyzed biopsy samples from paired baseline and relapsing lesions in four patients with metastatic melanoma who had had an initial objective tumor regression in response to anti-PD-1 therapy (pembrolizumab) followed by disease progression months to years later. RESULTS: Whole-exome sequencing detected clonal selection and outgrowth of the acquired resistant tumors and, in two of the four patients, revealed resistance-associated loss-of-function mutations in the genes encoding interferon-receptor-associated Janus kinase 1 (JAK1) or Janus kinase 2 (JAK2), concurrent with deletion of the wild-type allele. A truncating mutation in the gene encoding the antigen-presenting protein beta-2-microglobulin (B2M) was identified in a third patient. JAK1 and JAK2 truncating mutations resulted in a lack of response to interferon gamma, including insensitivity to its antiproliferative effects on cancer cells. The B2M truncating mutation led to loss of surface expression of major histocompatibility complex class I. CONCLUSIONS: In this study, acquired resistance to PD-1 blockade immunotherapy in patients with melanoma was associated with defects in the pathways involved in interferon-receptor signaling and in antigen presentation. (Funded by the National Institutes of Health and others.).


Asunto(s)
Resistencia a Antineoplásicos/genética , Inmunoterapia , Janus Quinasa 1/genética , Janus Quinasa 2/genética , Melanoma/genética , Mutación , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Microglobulina beta-2/genética , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Biopsia , Exoma , Regulación Neoplásica de la Expresión Génica , Genes MHC Clase I , Humanos , Interferón gamma/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/secundario , Receptor de Muerte Celular Programada 1/metabolismo , Recurrencia , Análisis de Secuencia de ADN , Transducción de Señal
9.
Cell ; 165(1): 35-44, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-26997480

RESUMEN

PD-1 immune checkpoint blockade provides significant clinical benefits for melanoma patients. We analyzed the somatic mutanomes and transcriptomes of pretreatment melanoma biopsies to identify factors that may influence innate sensitivity or resistance to anti-PD-1 therapy. We find that overall high mutational loads associate with improved survival, and tumors from responding patients are enriched for mutations in the DNA repair gene BRCA2. Innately resistant tumors display a transcriptional signature (referred to as the IPRES, or innate anti-PD-1 resistance), indicating concurrent up-expression of genes involved in the regulation of mesenchymal transition, cell adhesion, extracellular matrix remodeling, angiogenesis, and wound healing. Notably, mitogen-activated protein kinase (MAPK)-targeted therapy (MAPK inhibitor) induces similar signatures in melanoma, suggesting that a non-genomic form of MAPK inhibitor resistance mediates cross-resistance to anti-PD-1 therapy. Validation of the IPRES in other independent tumor cohorts defines a transcriptomic subset across distinct types of advanced cancer. These findings suggest that attenuating the biological processes that underlie IPRES may improve anti-PD-1 response in melanoma and other cancer types.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos , Melanoma/tratamiento farmacológico , Metástasis de la Neoplasia/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados/efectos adversos , Antineoplásicos/efectos adversos , Proteína BRCA2/genética , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/genética , Metástasis de la Neoplasia/genética , Nivolumab , Transcriptoma
10.
Nature ; 515(7528): 568-71, 2014 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-25428505

RESUMEN

Therapies that target the programmed death-1 (PD-1) receptor have shown unprecedented rates of durable clinical responses in patients with various cancer types. One mechanism by which cancer tissues limit the host immune response is via upregulation of PD-1 ligand (PD-L1) and its ligation to PD-1 on antigen-specific CD8(+) T cells (termed adaptive immune resistance). Here we show that pre-existing CD8(+) T cells distinctly located at the invasive tumour margin are associated with expression of the PD-1/PD-L1 immune inhibitory axis and may predict response to therapy. We analysed samples from 46 patients with metastatic melanoma obtained before and during anti-PD-1 therapy (pembrolizumab) using quantitative immunohistochemistry, quantitative multiplex immunofluorescence, and next-generation sequencing for T-cell antigen receptors (TCRs). In serially sampled tumours, patients responding to treatment showed proliferation of intratumoral CD8(+) T cells that directly correlated with radiographic reduction in tumour size. Pre-treatment samples obtained from responding patients showed higher numbers of CD8-, PD-1- and PD-L1-expressing cells at the invasive tumour margin and inside tumours, with close proximity between PD-1 and PD-L1, and a more clonal TCR repertoire. Using multivariate analysis, we established a predictive model based on CD8 expression at the invasive margin and validated the model in an independent cohort of 15 patients. Our findings indicate that tumour regression after therapeutic PD-1 blockade requires pre-existing CD8(+) T cells that are negatively regulated by PD-1/PD-L1-mediated adaptive immune resistance.


Asunto(s)
Inmunidad Adaptativa/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunoterapia , Melanoma/terapia , Modelos Biológicos , Anciano , Anciano de 80 o más Años , Biomarcadores , Linfocitos T CD8-positivos/citología , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Melanoma/diagnóstico , Melanoma/inmunología , Melanoma/patología , Persona de Mediana Edad , Análisis Multivariante , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...