Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 19(3): e0299499, 2024.
Article in English | MEDLINE | ID: mdl-38527041

ABSTRACT

Chemo-immunotherapy uses combined systemic therapies for resectable and unresectable tumors. This approach is gaining clinical momentum, but survival increases leave considerable room for improvement. A novel form of Pulsed Electric Field (PEF) ablation combines focal tissue destruction with immune activation in preclinical settings. The PEFs induce lethal cell damage without requiring thermal processes, leaving cellular proteins intact. This affords PEF a favorable safety profile, improved antigenicity, and significant immunostimulatory damage-associated molecular pattern release compared to other focal therapies. Preclinical investigations demonstrate a combinatorial benefit of PEF with immunostimulation. This study evaluates whether this proprietary PEF therapy induces an immunostimulatory effect sufficient to augment systemic neoadjuvant chemotherapy and immunotherapy to reverse metastatic disease in an immune-cold murine tumor model. To determine whether PEF improves a neoadjuvant chemo-immunotherapy standard-of-care, partial PEF ablation was delivered to orthotopically inoculated 4T1 metastatic tumors in addition to combinations of cisplatin chemotherapy and/or αPD-1 immunotherapy, followed by resection. In addition, to determine whether PEF combined with chemo-immunotherapy improves local and metastatic response in unresectable populations, partial PEF ablation was added to chemo-immunotherapy in mice that did not receive resection. Blood cytokines and flow cytometry evaluated immune response. Partial PEF ablation generates an immunostimulatory tumor microenvironment, increases systemic immune cell populations, slows tumor growth, and prolongs survival relative to neoadjuvant systemic therapies-alone. These data suggest the addition of this proprietary PEF locoregional therapy may synergize with systemic standard-of-care paradigms to improve outcomes with potential or demonstrated metastatic disease in both resectable and unresectable patient cohorts.


Subject(s)
Neoadjuvant Therapy , Neoplasms , Mice , Humans , Animals , Disease Models, Animal , Neoplasms/pathology , Immunotherapy , Cisplatin/therapeutic use , Tumor Microenvironment
2.
J Vasc Interv Radiol ; 35(3): 442-451.e7, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38042523

ABSTRACT

PURPOSE: To compare the immune response and survival after size-matched radiofrequency (RF) ablation and a proprietary form of pulsed electric field (PEF) ablation in murine tumors. MATERIAL AND METHODS: Orthotopically inoculated EMT6 or 4T1 murine tumors received sham, RF ablation, or PEF ablation. 4T1 tumor ablations included subgroups with intraperitoneal checkpoint inhibition immunotherapy (αPD-1). Blood was collected for cytokine profiling and flow cytometry. Tumor size was measured and survival was monitored. Tumor samples were processed for histology, immunohistochemistry, flow cytometry, and cytokine profiling. Lungs were collected from 4T1-bearing mice for hematoxylin and eosin histology to assess metastatic spread and abscopal effect induced by ablation. RESULTS: PEF elicited distinct immunomodulatory effects, with clear differences in serum and tumor cytokine profiles compared with RF ablation, including intratumoral downregulation of vascular endothelial growth factor, hypoxia-inducible factor 1α, c-MET, interleukin-10, Ki67, and tumor necrosis factor-α (all P < .05). PEF increased innate immune activation, with enhanced recruitment of dendritic cells, M1 macrophages, and natural killer cells coupled with a reduction in M2 macrophages and myeloid-derived suppressor cells (all P < .05). Concurrently, PEF strengthened adaptive immunity compared with RF ablation, characterized by increased antigen-specific T cells and decreased regulatory T cells (all P < .05). PEF stalled tumor growth and increased survival at the end of the study (≥4× versus RFA). Finally, PEF promoted an abscopal effect of clearing metastases in the lungs, which was stronger in combination with αPD-1 than with PEF alone. CONCLUSIONS: The proprietary form of PEF used in this study evoked a preferential immunostimulatory profile versus RF ablation thermal ablation in mice, with implications for enhancing the therapeutic effectiveness of checkpoint inhibition immunotherapy for immunotherapy-unresponsive tumors.


Subject(s)
Neoplasms , Radiofrequency Ablation , Mice , Animals , Vascular Endothelial Growth Factor A/metabolism , T-Lymphocytes/metabolism , Cytokines/metabolism
3.
Anticancer Agents Med Chem ; 18(15): 2187-2192, 2018.
Article in English | MEDLINE | ID: mdl-30198441

ABSTRACT

BACKGROUND: Bone Marrow (BM) has the self-renovation capacity and has been used recently in tumor medicine. Chlorambucil [CHB] is ordinarily utilized chemotherapy to treat varieties of malignancy patients. This investigation intended to gauge the effectiveness of BM as an in-vivo antimutagenic against CHB. METHODS: The experimental design relies upon four classes; each class contains ten adult male albino rats as follows: control, rats infused orally with CHB for fourteen days, rats intravenously infused with BM through a tail vein one time, rats infused the mix of CHB and BM.The Anticancer capability of BM was assessed by cytogenetic assay and mitotic index. The declarations of the apoptosis- related genes were examined by RT-qPCR examination. RESULTS: The present experiment demonstrated a curative effect of BM against the cytotoxic impact of CHB. Infusion of BM after chemotherapy helps to diminish the chromosomal aberration; increment mitotic index and decline the Bax/Bcl2 proportion compared with [CHB] class gather that prompts expanding the survival rate of influenced cells with chemotherapy cytotoxicity. CONCLUSION: The present study shows that bone marrow transplantation together after CHB infusion helps to increase genomic stability by diminishing structural chromosome abnormalities, diminishing the Bax/Bcl2 proportion and increasing the mitotic index.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Bone Marrow Cells/drug effects , Cell Survival/drug effects , Chlorambucil/pharmacology , Animals , Apoptosis/drug effects , Bone Marrow Cells/metabolism , Chromosome Aberrations , Male , Mutation , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , bcl-2-Associated X Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...