Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
JCI Insight ; 4(22)2019 11 14.
Article in English | MEDLINE | ID: mdl-31600167

ABSTRACT

BACKGROUNDMyeloid-derived suppressor cells (MDSCs) are elevated in the circulation of patients with glioblastoma (GBM), present in tumor tissue, and associated with poor prognosis. While low-dose chemotherapy reduces MDSCs in preclinical models, the use of this strategy to reduce MDSCs in GBM patients has yet to be evaluated.METHODSA phase 0/I dose-escalation clinical trial was conducted in patients with recurrent GBM treated 5-7 days before surgery with low-dose chemotherapy via capecitabine, followed by concomitant low-dose capecitabine and bevacizumab. Clinical outcomes, including progression-free and overall survival, were measured, along with safety and toxicity profiles. Over the treatment time course, circulating MDSC levels were measured by multiparameter flow cytometry, and tumor tissue immune profiles were assessed via time-of-flight mass cytometry.RESULTSEleven patients total were enrolled across escalating dose cohorts of 150, 300, and 450 mg bid. No serious adverse events related to the drug combination were observed. Compared with pretreatment baseline, circulating MDSCs were found to be higher after surgery in the 150-mg treatment arm and lower in the 300-mg and 450-mg treatment arms. Increased cytotoxic immune infiltration was observed after low-dose capecitabine compared with untreated GBM patients in the 300-mg and 450-mg treatment arms.CONCLUSIONSLow-dose, metronomic capecitabine in combination with bevacizumab was well tolerated in GBM patients and was associated with a reduction in circulating MDSC levels and an increase in cytotoxic immune infiltration into the tumor microenvironment.TRIAL REGISTRATIONClinicalTrials.gov NCT02669173.FUNDINGThis research was funded by the Cleveland Clinic, Case Comprehensive Cancer Center, the Musella Foundation, B*CURED, the NIH, the National Cancer Institute, the Sontag Foundation, Blast GBM, the James B. Pendleton Charitable Trust, and the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation. Capecitabine was provided in kind by Mylan Pharmaceuticals.


Subject(s)
Antineoplastic Agents, Immunological , Capecitabine , Glioblastoma/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Adult , Aged , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Bevacizumab/administration & dosage , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Capecitabine/administration & dosage , Capecitabine/pharmacology , Capecitabine/therapeutic use , Female , Humans , Male , Middle Aged , Tumor Microenvironment/drug effects
2.
JCI Insight ; 3(21)2018 11 02.
Article in English | MEDLINE | ID: mdl-30385717

ABSTRACT

Glioblastoma (GBM) remains uniformly lethal, and despite a large accumulation of immune cells in the microenvironment, there is limited antitumor immune response. To overcome these challenges, a comprehensive understanding of GBM systemic immune response during disease progression is required. Here, we integrated multiparameter flow cytometry and mass cytometry TOF (CyTOF) analysis of patient blood to determine changes in the immune system among tumor types and over disease progression. Utilizing flow cytometry analysis in a cohort of 259 patients ranging from benign to malignant primary and metastatic brain tumors, we found that GBM patients had a significant elevation in myeloid-derived suppressor cells (MDSCs) in peripheral blood but not immunosuppressive Tregs. In GBM patient tissue, we found that increased MDSC levels in recurrent GBM portended poor prognosis. CyTOF analysis of peripheral blood from newly diagnosed GBM patients revealed that reduced MDSCs over time were accompanied by a concomitant increase in DCs. GBM patients with extended survival also had reduced MDSCs, similar to the levels of low-grade glioma (LGG) patients. Our findings provide a rationale for developing strategies to target MDSCs, which are elevated in GBM patients and predict poor prognosis.


Subject(s)
Brain Neoplasms/immunology , Cell Line/immunology , Glioblastoma/immunology , Myeloid-Derived Suppressor Cells/immunology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Cell Line/drug effects , Disease Progression , Female , Flow Cytometry/methods , Glioblastoma/pathology , Humans , Longitudinal Studies , Male , Myeloid-Derived Suppressor Cells/drug effects , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Survival Analysis , Tumor Microenvironment/drug effects
3.
J Neurooncol ; 122(2): 293-301, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25579983

ABSTRACT

Myeloid derived suppressor cells (MDSCs) are bone marrow derived cells with immunosuppressive properties. We have shown previously that MDSCs numbers are elevated in the circulation of GBM patients and that they produce reversible T cell dysfunction. Here, we evaluated whether MDSCs infiltrate human GBM tissues, and whether a commonly used mouse model of GBM reproduces the biology of MDSCs that is observed in patients. We evaluated tumor specimens from patients with newly diagnosed GBM. We harvested and evaluated normal brain, tumors and hematopoietic tissues from control, vehicle and sunitinib-treated mice. In human GBM tumors, MDSCs represented 5.4 ± 1.8 % of total cells. The majority of MDSCs (CD33+HLADR-) were lineage negative (CD14-CD15-), followed by granulocytic (CD15+CD14-) and monocytic (CD15-CD14+) subtypes. In murine GBM tumors, MDSCs were 8.06 ± 0.78 % of total cells, of which more were monocytic (M-MDSC, CD11b+ Gr1-low) than granulocytic (G-MDSC, CD11b+ Gr1-high). Treatment with the tyrosine kinase inhibitor sunitinib decreased the infiltration of both granulocytic and monocytic MDSCs in murine GBM tumors. In the hematopoietic tissues, circulating G-MDSC blood levels were reduced after sunitinib treatment. In tumors, both CD3(+) and CD4(+) T cell counts increased following sunitinib treatment (p ≤ 0.001). Total T cell proliferation (p < 0.001) and interferon gamma production (p = 0.004) were increased in the spleens of sunitinib treated mice. Sunitinib-treated mice survived longer than vehicle-treated mice (p = 0.002). MDSCs are present in both human and mouse GBM tumors. Sunitinib may have an immunostimulatory effect, as its use is associated with a reduction in G-MDSCs and improvement in anti-tumor immune function.


Subject(s)
Brain Neoplasms/physiopathology , Brain/physiopathology , Glioma/physiopathology , Lymphocytes, Tumor-Infiltrating/physiology , Myeloid Cells/physiology , Animals , Antineoplastic Agents/pharmacology , Bone Marrow/drug effects , Bone Marrow/physiopathology , Brain/drug effects , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line , Chickens , Glioma/drug therapy , Glioma/pathology , Humans , Indoles/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/pathology , Mice, Transgenic , Myeloid Cells/drug effects , Myeloid Cells/pathology , Neoplasm Transplantation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Pyrroles/pharmacology , Spleen/drug effects , Spleen/physiopathology , Sunitinib , Survival Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , T-Lymphocytes/physiology , Treatment Outcome
4.
Adv Exp Med Biol ; 765: 351-355, 2013.
Article in English | MEDLINE | ID: mdl-22879055

ABSTRACT

In this study we investigated the effect of early life conditioning (hypoxia ± hypercapnia) on adult acute ventilatory sensitivity to hypoxia and hypercapnia. Mice were exposed to either hypoxia (5% O(2)) or hypoxia/hypercapnia (5% O(2)/8% CO(2)) in a normobaric chamber for 2 h at postnatal day 2 (P2), and then returned to normoxia. At 3 months of age, hypoxic ventilatory response (HVR) and hypercapnic ventilatory response (HCVR) were measured using a plethysmograph system. Results showed that HVR was significantly decreased in the P2-hypoxia mice but not in the P2 hypoxia/hypercapnia mice as compared to the P2-normoxic mice, respectively. However, HCVR was significantly decreased in the P2 hypoxia-hypercapnia group but not in the P2-hypoxia group. These data suggest early postnatal hypoxic stress vs. hypoxic/hypercapnic stress plays different roles in fetal programming of the respiratory control system as shown by altered adult acute ventilatory sensitivity.


Subject(s)
Aging/pathology , Hypercapnia/physiopathology , Hypoxia/physiopathology , Pulmonary Ventilation/physiology , Adaptation, Physiological , Animals , Animals, Newborn , Mice , Plethysmography
SELECTION OF CITATIONS
SEARCH DETAIL
...