Modulated electro-hyperthermia (mEHT) is a complementary antitumor therapy applying capacitive radiofrequency at 13.56 MHz. Here we tested the efficiency of mEHT treatment in a BALB/c mouse isograft model using the firefly luciferase-transfected triple-negative breast cancer cell line, 4T1. Tumors inoculated orthotopically were treated twice using a novel ergonomic pole electrode and an improved mEHT device (LabEHY 200) at 0.7 ± 0.3 W for 30 min. Tumors were treated one, two, or three times every 48 h. Tumor growth was followed by IVIS, caliper, and ultrasound. Tumor destruction histology and molecular changes using immunohistochemistry and RT-qPCR were also revealed. In vivo, mEHT treatment transitionally elevated Hsp70 expression in surviving cells indicating heat shock-related cell stress, while IVIS fluorescence showed a significant reduction of viable tumor cell numbers. Treated tumor centers displayed significant microscopic tumor damage with prominent signs of apoptosis, and major upregulation of cleaved/activated caspase-3-positive tumor cells. Serial sampling demonstrated substantial elevation of heat shock (Hsp70) response twelve hours after the treatment which was exhausted by twenty-four hours after treatment. Heat shock inhibitors Quercetin or KRIBB11 could synergistically amplify mEHT-induced tumor apoptosis in vitro. In conclusion, modulated electro-hyperthermia exerted a protective heat shock response as a clear sign of tumor cell stress. Exhaustion of the HSR manifested in caspase-dependent apoptotic tumor cell death and tissue damage of triple-negative breast cancer after mEHT monotherapy. Inhibiting the HSR synergistically increased the effect of mEHT. This finding has great translational potential.
Modulated electro-hyperthermia (mEHT) is a selective cancer treatment used in human oncology complementing other therapies. During mEHT, a focused electromagnetic field (EMF) is generated within the tumor inducing cell death by thermal and nonthermal effects. Here we investigated molecular changes elicited by mEHT using multiplex methods in an aggressive, therapy-resistant triple negative breast cancer (TNBC) model. 4T1/4T07 isografts inoculated orthotopically into female BALB/c mice were treated with mEHT three to five times. mEHT induced the upregulation of the stress-related Hsp70 and cleaved caspase-3 proteins, resulting in effective inhibition of tumor growth and proliferation. Several acute stress response proteins, including protease inhibitors, coagulation and heat shock factors, and complement family members, were among the most upregulated treatment-related genes/proteins as revealed by next-generation sequencing (NGS), Nanostring and mass spectrometry (MS). pathway analysis demonstrated that several of these proteins belong to the response to stimulus pathway. Cell culture treatments confirmed that the source of these proteins was the tumor cells. The heat-shock factor inhibitor KRIBB11 reduced mEHT-induced complement factor 4 (C4) mRNA increase. In conclusion, mEHT monotherapy induced tumor growth inhibition and a complex stress response. Inhibition of this stress response is likely to enhance the effectiveness of mEHT and other cancer treatments.
There is growing interest in the role of nerve-driven mechanisms in tumorigenesis and tumor growth. Capsaicin-sensitive afferents have been previously shown to possess antitumoral and immune-regulatory properties, the mechanism of which is currently poorly understood. In this study, we have assessed the role of these terminals in the triple negative 4T1 orthotopic mouse model of breast cancer. The ultrapotent capsaicin-analogue resiniferatoxin (RTX) was used for the selective, systemic desensitization of capsaicin-sensitive afferents. Growth and viability of orthotopically implanted 4T1 tumors were measured by caliper, in vivo MRI, and bioluminescence imaging, while tumor vascularity and protease enzyme activity were assessed using fluorescent in vivo imaging. The levels of the neuropeptides Calcitonin Gene-Related Peptide (CGRP), Substance P (SP), and somatostatin were measured from tumor tissue homogenates using radioimmunoassay, while tumor structure and peritumoral inflammation were evaluated by conventional use of CD31, CD45 and CD3 immunohistology. RTX-pretreated mice demonstrated facilitated tumor growth in the early phase measured using a caliper, which was coupled with increased tumor vascular leakage demonstrated using fluorescent vascular imaging. The tumor size difference dissipated by day seven. The MRI tumor volume was similar, while the intratumoral protease enzyme activity measured by fluorescence imaging was also comparable in RTX-pretreated and non-pretreated animals. Tumor viability or immunohistopathological profile was measured using CD3, CD31, and CD45 stains and did not differ significantly from the non-pretreated control group. Intratumoral somatostatin, CGRP, and SP levels were similar in both groups. Our results underscore the beneficial, antitumoral properties of capsaicin sensitive nerve terminals in this aggressive model of breast cancer, which is presumed to be due to the inhibition of tumor vascular bed disruption. The absence of any difference in intratumoral neuropeptide levels indicates non-neural sources playing a substantial part in their expression.
Introduction Modulated electro‐hyperthermia (mEHT) is a complementary antitumor therapy, based on selective tumor cell killing by a 13.56 MHz radiofrequency induced electric field. The H19 long non coding RNA (lncRNA) is involved in tumor progression and metastasis. It’s overexpression is associated with poor prognosis and therapy resistance in breast cancer. We observed previously significant tumor inhibitory effects of mEHT. Aims Our aim was to investigate the hypothesis, that mEHT effects are related to H19 lncRNA inhibition in triple negative breast cancer (TNBC) spheroids and in a TNBC bearing mouse model. Methods 4T1 spheroids were embedded in Matrigel® and treated with a single mEHT, conventional hyperthermia (cHT) or normothermia (Ctr) for 30 minutes. Spheroids were collected 24 hours after treatment, RNA was isolated and processed for Real‐Time PCR (RT‐PCR). TNBC cells (highly aggressive 4T1 or less aggressive 4T07) were inoculated orthotopically in female BALB/c mice. Tumor growth was monitored in vivo by digital caliper and ultrasound (Phillips Sonos 5500), mice were randomized into two groups based on tumor size. Mice were treated with mEHT in monotherapy or in combination with methotrexate (MTX) 2 or 3 times for 30 minutes with 0.7±0.3W power to achieve 40°C skin temperature above the tumor. At the end of the experiments mice were euthanized, the tumors were dissected and processed for molecular biologic techniques. H19 expression was measured with RT‐PCR, results were normalized to GAPDH. Results Single mEHT or cHT treatment of 4T1 spheroids reduced significantly H19 expression compared to a normothermic control (Ctr:0.004±0.0004, cHT:0.001±0.0001 mEHT:0.0006±0.0002, p<0.0001). mEHT decreased H19 expression more effectively, than conventional HT (cHT:0.001±0.0001 mEHT:0.0006±0.0002, p<0.01). There was a significant decrease in H19 expression of 4T1 tumors in vivo after two (sham:0.068±0.044, mEHT:0.033±0.024, p<0.05) and three mEHT treatments (sham:0.097±0.059 vs mEHT:0.050±0.030, p<0.05) compared to the sham group. In case of combination treatments H19 expression was significantly lower in the mEHT+MTX group compared to MTX only (MTX:0.104±0,038 vs mEHT+MTX:0.056±0.025, p<0.01). The basic expression of H19 was significantly lower in 4T07 tumors compared to the more aggressive 4T1 tumors (4T07:0.006±0.004 vs 4T1:0.399±0.071, p<0.0001). In 4T07 tumors H19 expression did not change after three mEHT treatments (sham:0.404±0.334 vs mEHT:1.391±1.840, p>0.10) compared to the sham group. Conclusion Our results demonstrate, that modulated electro‐hyperthermia can reduce the expression of tumor promoting H19 lncRNA in vitro and in vivo both in monotherapy and in combination with chemotherapy. Our findings suggest, that mEHT as an alternative complementary treatment could promote antitumor therapy by inhibiting the tumor progression mediating H19 lncRNA expression. More effective therapy against the more aggressive 4T1 line may be related to higher baseline expression and more significant effect on H19. Suppo...
As our society ages inexorably, geroscience and research focusing on healthy aging is becoming increasingly urgent. Macroautophagy (referred to as autophagy), a highly conserved process of cellular clearance and rejuvenation has attracted much attention due to its universal role in organismal life and death. Growing evidence points to autophagy process as being one of the key players in the determination of lifespan and health. Autophagy inducing interventions show significant improvement in organismal lifespan demonstrated in several experimental models. In line with this, preclinical models of age-related neurodegenerative diseases demonstrate pathology modulating effect of autophagy induction, implicating its potential to treat such disorders. In humans this specific process seems to be more complex. Recent clinical trials of drugs targeting autophagy point out some beneficial effects for clinical use, although with limited effectiveness, while others fail to show any significant improvement. We propose that using more human-relevant preclinical models for testing drug efficacy would significantly improve clinical trial outcomes. Lastly, the review discusses the available cellular reprogramming techniques used to model neuronal autophagy and neurodegeneration while exploring the existing evidence of autophagy’s role in aging and pathogenesis in human-derived in vitro models such as embryonic stem cells (ESCs), induced pluripotent stem cell derived neurons (iPSC-neurons) or induced neurons (iNs).
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.
customersupport@researchsolutions.com
10624 S. Eastern Ave., Ste. A-614
Henderson, NV 89052, USA
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.
Copyright © 2024 scite LLC. All rights reserved.
Made with 💙 for researchers
Part of the Research Solutions Family.