Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Head Neck ; 45(11): 2789-2797, 2023 11.
Article in English | MEDLINE | ID: mdl-37682116

ABSTRACT

OBJECTIVES: To explore the impact of pre-existing comorbidities on immunotherapy response, overall and progression-free survival, and immune-related adverse events (irAEs) of patients with advanced head and neck cancer (HNC) treated with immunotherapy. PATIENTS AND METHODS: Ninety-three patients treated with immunotherapy were identified and stratified into comorbidity absent or present (CCI < 1 and CCI ≥ 1, respectively) cohorts, and clinical outcomes were compared between these two groups. RESULTS: Patients with no comorbidities had longer overall survival (aHR = 2.74, 95% CI [1.18, 6.40], p = 0.02) and progression-free survival (aHR = 2.07, 95% CI [1.03, 4.16], p = 0.04) and a higher tumor response rate (32% in CCI < 1 vs. 14% in CC ≥ 1, p = 0.05). Risk for irAEs was higher in the comorbidity absent group (p = 0.05). CONCLUSION: Comorbidity should be considered as a significant prognostic factor in clinical decision-making for patients with advanced HNC undergoing immunotherapy.


Subject(s)
Head and Neck Neoplasms , Humans , Prognosis , Head and Neck Neoplasms/therapy , Comorbidity , Progression-Free Survival , Immunotherapy/adverse effects , Retrospective Studies
2.
J Immunother Cancer ; 9(5)2021 05.
Article in English | MEDLINE | ID: mdl-34021032

ABSTRACT

BACKGROUND: Current therapy for osteosarcoma pulmonary metastases (PMs) is ineffective. The mechanisms that prevent successful immunotherapy in osteosarcoma are incompletely understood. We investigated the tumor microenvironment of metastatic osteosarcoma with the goal of harnessing the immune system as a therapeutic strategy. METHODS: 66 osteosarcoma tissue specimens were analyzed by immunohistochemistry (IHC) and immune markers were digitally quantified. Tumor-infiltrating lymphocytes (TILs) from 25 specimens were profiled by functional cytometry. Comparative transcriptomic studies of distinct tumor-normal lung 'PM interface' and 'PM interior' regions from 16 PMs were performed. Clinical follow-up (median 24 months) was available from resection. RESULTS: IHC revealed a statistically significantly higher concentration of TILs expressing immune checkpoint and immunoregulatory molecules in PMs compared with primary bone tumors (including programmed cell death 1 (PD-1), programmed death ligand 1 (PD-L1), lymphocyte-activation gene 3 (LAG-3), T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), and indoleamine 2,3-dioxygenase (IDO1). Remarkably, these lymphocytes are excluded at the PM interface compared with PM interior. TILs from PMs exhibited significantly higher amounts of PD-1 and LAG-3 and functional cytokines including interferon-γ (IFNγ) by flow cytometry. Gene expression profiling further confirmed the presence of CD8 and CD4 lymphocytes concentrated at the PM interface, along with upregulation of immunoregulatory molecules and IFNγ-driven genes in the same region. We further discovered a strong alternatively activated macrophage signature throughout the entire PMs along with a polymorphonuclear myeloid-derived suppressor cell signature focused at the PM interface. Expression of PD-L1, LAG-3, and colony-stimulating factor 1 receptor (CSF1R) at the PM interface was associated with significantly worse progression-free survival (PFS), while gene sets indicative of productive T cell immune responses (CD8 T cells, T cell survival, and major histocompatibility complex class 1 expression) were associated with significantly improved PFS. CONCLUSIONS: Osteosarcoma PMs exhibit immune exclusion characterized by the accumulation of TILs at the PM interface. These TILs produce effector cytokines, suggesting their capability of activation and recognition of tumor antigens. Our findings suggest cooperative immunosuppressive mechanisms in osteosarcoma PMs including immune checkpoint molecule expression and the presence of immunosuppressive myeloid cells. We identify cellular and molecular signatures that are associated with patient outcomes, which could be exploited for successful immunotherapy.


Subject(s)
Biomarkers, Tumor/analysis , Bone Neoplasms/therapy , Cytokines/analysis , Immune Checkpoint Proteins/analysis , Immunotherapy , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Osteosarcoma/therapy , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Cytokines/genetics , Electronic Health Records , Humans , Immune Checkpoint Proteins/genetics , Immunotherapy/adverse effects , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lymphocyte Activation , Macrophage Activation , Myeloid-Derived Suppressor Cells , Osteosarcoma/genetics , Osteosarcoma/immunology , Osteosarcoma/secondary , Progression-Free Survival , Retrospective Studies , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...