Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 2 de 2
1.
Clin Cancer Res ; 28(14): 3156-3169, 2022 07 15.
Article En | MEDLINE | ID: mdl-35552677

PURPOSE: Paclitaxel (PTX) is one of the most potent and commonly used chemotherapies for breast and pancreatic cancer. Several ongoing clinical trials are investigating means of enhancing delivery of PTX across the blood-brain barrier for glioblastomas. Despite the widespread use of PTX for breast cancer, and the initiative to repurpose this drug for gliomas, there are no predictive biomarkers to inform which patients will likely benefit from this therapy. EXPERIMENTAL DESIGN: To identify predictive biomarkers for susceptibility to PTX, we performed a genome-wide CRISPR knockout (KO) screen using human glioma cells. The genes whose KO was most enriched in the CRISPR screen underwent further selection based on their correlation with survival in the breast cancer patient cohorts treated with PTX and not in patients treated with other chemotherapies, a finding that was validated on a second independent patient cohort using progression-free survival. RESULTS: Combination of CRISPR screen results with outcomes from patients with taxane-treated breast cancer led to the discovery of endoplasmic reticulum (ER) protein SSR3 as a putative predictive biomarker for PTX. SSR3 protein levels showed positive correlation with susceptibility to PTX in breast cancer cells, glioma cells, and in multiple intracranial glioma xenografts models. KO of SSR3 turned the cells resistant to PTX while its overexpression sensitized the cells to PTX. Mechanistically, SSR3 confers susceptibility to PTX through regulation of phosphorylation of ER stress sensor IRE1α. CONCLUSIONS: Our hypothesis generating study showed SSR3 as a putative biomarker for susceptibility to PTX, warranting its prospective clinical validation.


Antineoplastic Agents, Phytogenic , Biomarkers, Pharmacological , Brain Neoplasms , Breast Neoplasms , Calcium-Binding Proteins , Drug Resistance, Neoplasm , Glioblastoma , Membrane Glycoproteins , Paclitaxel , Receptors, Cytoplasmic and Nuclear , Receptors, Peptide , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Endoribonucleases/metabolism , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Membrane Glycoproteins/genetics , Mice , Paclitaxel/therapeutic use , Prospective Studies , Protein Serine-Threonine Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Peptide/genetics , Xenograft Model Antitumor Assays
2.
Mol Neurobiol ; 56(7): 5032-5040, 2019 Jul.
Article En | MEDLINE | ID: mdl-30460615

The immunosuppressive microenvironment is one of the major factors promoting the growth of glioblastoma multiforme (GBM). Infiltration of CD4+CD25+Foxp3+ regulatory T cells (Tregs) into the tumor microenvironment plays a significant role in the suppression of the anti-tumor immunity and portends a dismal prognosis for patients. Glioma-mediated secretion of chemo-attractant C-C motif ligand 2 and 22 (CCL2/22) has previously been shown by our group to promote Treg migration in vitro. In this study, we show that a local implantation of platelet-rich fibrin patch (PRF-P) into the brain of GL261 glioma-bearing mice prolonged the survival of affected animals by 42.85% (p = 0.0011). Analysis performed on brain tumor tissue harvested from PRF-P-treated mice revealed a specific decrease in intra-tumoral lymphocytes with a preferential depletion of immunosuppressive Tregs. Importantly, co-culture of GL261 or chemo-attractants (CCL2/22) with PRF-P abrogated Treg migration. Pharmacological blockade of the CCL2/22 interaction with their receptors potentiated the inhibitory effect of PRF-P on Tregs recruitment in culture. Moreover, our findings revealed the soluble CD40 ligand (sCD40L) as a major Treg inhibitory player produced by activated platelets entrapped within the fibrin matrix of the PRF-P. Blockade of sCD40L restored the migratory capacity of Tregs, emphasizing the role of PRF-P in preventing the Treg migration to glioma tissue. Our findings highlight autologous PRF-P as a personalized, Treg-selective suppression platform that can potentially supplement and enhance the efficacy of glioma therapies.


Autografts , Brain Neoplasms/therapy , Glioma/therapy , Platelet-Rich Fibrin/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cells, Cultured , Craniotomy/methods , Glioma/immunology , Glioma/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism , Tumor Microenvironment/immunology
...