1
|
Minnaar CA, Szigeti GP, Szasz A. The Synergy of Thermal and Non-Thermal Effects in Hyperthermic Oncology. Cancers (Basel) 2024; 16:3908. [PMID: 39682096 DOI: 10.3390/cancers16233908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Modulated electro-hyperthermia (mEHT) is unique due to its combination of thermal and non-thermal effects. METHOD This report summarizes the literature on the effects of mEHT observed in vitro and in vivo. RESULTS The thermal and electrical heterogeneity of tissues allows the radiofrequency signal to selectively target malignant tissue. The applied modulation appears to activate various apoptotic pathways, predominantly leading to immunogenic cell death (ICD). ICD promotes the release of damage-associated molecular patterns, potentially producing tumour-specific antigen-presenting cells. This abscopal-type effect may target distant metastases while treating the primary tumour locally. This immune memory effect is like vaccination mechanisms. CONCLUSIONS The application of mEHT has the potential to expand from local to systemic disease, enabling the simultaneous treatment of micro- and macro-metastases.
Collapse
Affiliation(s)
- Carrie Anne Minnaar
- Department of Radiation Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Gyula Peter Szigeti
- John von Neumann Faculty of Informatics, Óbuda University, 1034 Budapest, Hungary
- MedTech Innovation and Education Center, University Research and Innovation Center, Óbuda University, 1034 Budapest, Hungary
| | - Andras Szasz
- Department of Biotechnics, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| |
Collapse
|
2
|
Romani A, Lodi G, Casciano F, Gonelli A, Secchiero P, Zauli G, Bortolini O, Valacchi G, Ragno D, Bondi A, Benedusi M, Esposito E, Voltan R. Enhanced Anti-Melanoma Activity of Nutlin-3a Delivered via Ethosomes: Targeting p53-Mediated Apoptosis in HT144 Cells. Cells 2024; 13:1678. [PMID: 39451196 PMCID: PMC11506859 DOI: 10.3390/cells13201678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
This study evaluated ethosomes as a novel nanodelivery system for nutlin-3a, a known MDM2 inhibitor and activator of the p53 pathway, to improve nutlin-3a's poor solubility, limiting its bio-distribution and therapeutic efficacy. The potential of nutlin-3a-loaded ethosomes was investigated on two in vitro models of melanoma: the HT144 cell line p53wild-type and the SK-MEL-28 cell line p53mutated. Nutlin-3a-loaded ethosomes were characterized for their physicochemical properties and used to treat melanoma cells at different concentrations, considering nutlin-3a solution and empty ethosomes as controls. The biological effects on cells were evaluated 24 and 48 h after treatment by analyzing the cell morphology and viability, cell cycle, and apoptosis rate using flow cytometry and the p53 pathway's activation via Western blotting. The results indicate that ethosomes are delivery systems able to maintain nutlin-3a's functionality and specific biological action, as evidenced by the molecular activation of the p53 pathway and the biological events leading to cell cycle block and apoptosis in p53wild-type cells. Nutlin-3a-loaded ethosomes induced morphological changes in the HT144 cell line, with evident apoptotic cells and a reduction in the number of viable cells of over 80%. Furthermore, nutlin-3a-loaded ethosomes successfully modulated two p53-regulated proteins involved in survival/apoptosis, with up to a 2.5-fold increase in membrane TRAIL-R2 and up to an 8.2-fold decrease in Notch-1 (Notch intracellular domain, NICD) protein expression. The expression of these molecules is known to be altered or dysfunctional in a large percentage of melanoma tumors. Notably, ethosomes, regardless of their nutlin-3a loading, exhibited the ability to reduce HT144 melanoma cellular migration, as assessed in real time using xCELLigence, likely due to the modification of lipid rafts, suggesting their potential antimetastatic properties. Overall, nutlin-3a delivery using ethosomes appears to be a significantly effective means for upregulating the p53 pathway and downregulating active Notch-1, while also taking advantage of their unexpected ability to reduce cellular migration. The findings of this study could pave the way for the development of specific nutlin-3a-loaded ethosome-based medicinal products for cutaneous use.
Collapse
Affiliation(s)
- Arianna Romani
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (A.R.); (P.S.)
| | - Giada Lodi
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (G.L.); (F.C.)
| | - Fabio Casciano
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (G.L.); (F.C.)
| | - Arianna Gonelli
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.G.); (O.B.); (G.V.)
| | - Paola Secchiero
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (A.R.); (P.S.)
| | - Giorgio Zauli
- Research Department, King Khaled Eye Specialistic Hospital, Riyadh 12329-8139, Saudi Arabia;
| | - Olga Bortolini
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.G.); (O.B.); (G.V.)
| | - Giuseppe Valacchi
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.G.); (O.B.); (G.V.)
- Plants for Human Health Institute, Animal Sciences Department, NC Research Campus, NC State University, Kannapolis, NC 28081, USA
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Daniele Ragno
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (D.R.); (A.B.)
| | - Agnese Bondi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (D.R.); (A.B.)
| | - Mascia Benedusi
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Elisabetta Esposito
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (D.R.); (A.B.)
| | - Rebecca Voltan
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (G.L.); (F.C.)
| |
Collapse
|
3
|
Viana PHL, Schvarcz CA, Danics LO, Besztercei B, Aloss K, Bokhari SMZ, Giunashvili N, Bócsi D, Koós Z, Benyó Z, Hamar P. Heat shock factor 1 inhibition enhances the effects of modulated electro hyperthermia in a triple negative breast cancer mouse model. Sci Rep 2024; 14:8241. [PMID: 38589452 PMCID: PMC11002009 DOI: 10.1038/s41598-024-57659-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Female breast cancer is the most diagnosed cancer worldwide. Triple negative breast cancer (TNBC) is the most aggressive type and there is no existing endocrine or targeted therapy. Modulated electro-hyperthermia (mEHT) is a non-invasive complementary cancer therapy using an electromagnetic field generated by amplitude modulated 13.56 MHz frequency that induces tumor cell destruction. However, we have demonstrated a strong induction of the heat shock response (HSR) by mEHT, which can result in thermotolerance. We hypothesized that inhibition of the heat shock factor 1 (HSF1) can synergize with mEHT and enhance tumor cell-killing. Thus, we either knocked down the HSF1 gene with a CRISPR/Cas9 lentiviral construct or inhibited HSF1 with a specific small molecule inhibitor: KRIBB11 in vivo. Wild type or HSF1-knockdown 4T1 TNBC cells were inoculated into the mammary gland's fat pad of BALB/c mice. Four mEHT treatments were performed every second day and the tumor growth was followed by ultrasound and caliper. KRIBB11 was administrated intraperitoneally at 50 mg/kg daily for 8 days. HSF1 and Hsp70 expression were assessed. HSF1 knockdown sensitized transduced cancer cells to mEHT and reduced tumor growth. HSF1 mRNA expression was significantly reduced in the KO group when compared to the empty vector group, and consequently mEHT-induced Hsp70 mRNA upregulation diminished in the KO group. Immunohistochemistry (IHC) confirmed the inhibition of Hsp70 upregulation in mEHT HSF1-KO group. Demonstrating the translational potential of HSF1 inhibition, combined therapy of mEHT with KRIBB11 significantly reduced tumor mass compared to either monotherapy. Inhibition of Hsp70 upregulation by mEHT was also supported by qPCR and IHC. In conclusion, we suggest that mEHT-therapy combined with HSF1 inhibition can be a possible new strategy of TNBC treatment with great translational potential.
Collapse
Affiliation(s)
- Pedro H L Viana
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
| | - Csaba A Schvarcz
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
- HUN-REN-SU Cerebrovascular and Neurocognitive Diseases Research Group, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
| | - Lea O Danics
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
| | - Balázs Besztercei
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
| | - Kenan Aloss
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
| | - Syeda M Z Bokhari
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
| | - Nino Giunashvili
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
| | - Dániel Bócsi
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
| | - Zoltán Koós
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
- HUN-REN-SU Cerebrovascular and Neurocognitive Diseases Research Group, Tűzoltó Utca 37-47, Budapest, 1094, Hungary
| | - Péter Hamar
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, Budapest, 1094, Hungary.
| |
Collapse
|
4
|
Viana P, Hamar P. Targeting the heat shock response induced by modulated electro-hyperthermia (mEHT) in cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189069. [PMID: 38176599 DOI: 10.1016/j.bbcan.2023.189069] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
The Heat Shock Response (HSR) is a cellular stress reaction crucial for cell survival against stressors, including heat, in both healthy and cancer cells. Modulated electro-hyperthermia (mEHT) is an emerging non-invasive cancer therapy utilizing electromagnetic fields to selectively target cancer cells via temperature-dependent and independent mechanisms. However, mEHT triggers HSR in treated cells. Despite demonstrated efficacy in cancer treatment, understanding the underlying molecular mechanisms for improved therapeutic outcomes remains a focus. This review examines the HSR induced by mEHT in cancer cells, discussing potential strategies to modulate it for enhanced tumor-killing effects. Approaches such as HSF1 gene-knockdown and small molecule inhibitors like KRIBB11 are explored to downregulate the HSR and augment tumor destruction. We emphasize the impact of HSR inhibition on cancer cell viability, mEHT sensitivity, and potential synergistic effects, addressing challenges and future directions. This understanding offers opportunities for optimizing treatment strategies and advancing precision medicine in cancer therapy.
Collapse
Affiliation(s)
- Pedro Viana
- Institute of Translational Medicine, Semmelweis University, Tűzoltó utca 37-49, 1094 Budapest, Hungary.
| | - Péter Hamar
- Institute of Translational Medicine, Semmelweis University, Tűzoltó utca 37-49, 1094 Budapest, Hungary.
| |
Collapse
|
5
|
Bokhari SZ, Aloss K, Leroy Viana PH, Schvarcz CA, Besztercei B, Giunashvili N, Bócsi D, Koós Z, Balogh A, Benyó Z, Hamar P. Digoxin-Mediated Inhibition of Potential Hypoxia-Related Angiogenic Repair in Modulated Electro-Hyperthermia (mEHT)-Treated Murine Triple-Negative Breast Cancer Model. ACS Pharmacol Transl Sci 2024; 7:456-466. [PMID: 38357275 PMCID: PMC10863435 DOI: 10.1021/acsptsci.3c00296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 02/16/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer type with no targeted therapy and hence limited treatment options. Modulated electrohyperthermia (mEHT) is a novel complementary therapy where a 13.56 MHz radiofrequency current targets cancer cells selectively, inducing tumor damage by thermal and electromagnetic effects. We observed severe vascular damage in mEHT-treated tumors and investigated the potential synergism between mEHT and inhibition of tumor vasculature recovery in our TNBC mouse model. 4T1/4T07 isografts were orthotopically inoculated and treated three to five times with mEHT. mEHT induced vascular damage 4-12 h after treatment, leading to tissue hypoxia detected at 24 h. Hypoxia in treated tumors induced an angiogenic recovery 24 h after the last treatment. Administration of the cardiac glycoside digoxin with the potential hypoxia-inducible factor 1-α (HIF1-α) and angiogenesis inhibitory effects could synergistically augment mEHT-mediated tumor damage and reduce tissue hypoxia signaling and consequent vascular recovery in mEHT-treated TNBC tumors. Conclusively, repeated mEHT induced vascular damage and hypoxic stress in TNBC that promoted vascular recovery. Inhibiting this hypoxic stress signaling enhanced the effectiveness of mEHT and may potentially enhance other forms of cancer treatment.
Collapse
Affiliation(s)
| | - Kenan Aloss
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | | | - Csaba András Schvarcz
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
- Cerebrovascular
and Neurocognitive Disorders Research Group, Eötvös, Loránd Research Network and Semmelweis
University (ELKH-SE), Tűzoltó utca 37-47, Budapest 1094, Hungary
| | - Balázs Besztercei
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Nino Giunashvili
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Dániel Bócsi
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Zoltán Koós
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Andrea Balogh
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Zoltán Benyó
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Péter Hamar
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| |
Collapse
|
6
|
Forcing the Antitumor Effects of HSPs Using a Modulated Electric Field. Cells 2022; 11:cells11111838. [PMID: 35681533 PMCID: PMC9180583 DOI: 10.3390/cells11111838] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 12/10/2022] Open
Abstract
The role of Heat Shock Proteins (HSPs) is a “double-edged sword” with regards to tumors. The location and interactions of HSPs determine their pro- or antitumor activity. The present review includes an overview of the relevant functions of HSPs, which could improve their antitumor activity. Promoting the antitumor processes could assist in the local and systemic management of cancer. We explore the possibility of achieving this by manipulating the electromagnetic interactions within the tumor microenvironment. An appropriate electric field may select and affect the cancer cells using the electric heterogeneity of the tumor tissue. This review describes the method proposed to effect such changes: amplitude-modulated radiofrequency (amRF) applied with a 13.56 MHz carrier frequency. We summarize the preclinical investigations of the amRF on the HSPs in malignant cells. The preclinical studies show the promotion of the expression of HSP70 on the plasma membrane, participating in the immunogenic cell death (ICD) pathway. The sequence of guided molecular changes triggers innate and adaptive immune reactions. The amRF promotes the secretion of HSP70 also in the extracellular matrix. The extracellular HSP70 accompanied by free HMGB1 and membrane-expressed calreticulin (CRT) form damage-associated molecular patterns encouraging the dendritic cells’ maturing for antigen presentation. The process promotes killer T-cells. Clinical results demonstrate the potential of this immune process to trigger a systemic effect. We conclude that the properly applied amRF promotes antitumor HSP activity, and in situ, it could support the tumor-specific immune effects produced locally but acting systemically for disseminated cells and metastatic lesions.
Collapse
|
7
|
Filipiak-Duliban A, Brodaczewska K, Majewska A, Kieda C. Spheroid culture models adequately imitate distinctive features of the renal cancer or melanoma microenvironment. In Vitro Cell Dev Biol Anim 2022; 58:349-364. [PMID: 35536385 DOI: 10.1007/s11626-022-00685-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/24/2022] [Indexed: 11/05/2022]
Abstract
Tumor development studies should adapt to cancer cells' specific mechanisms in connection with their microenvironment. Standard two-dimensional cultures and gas composition are not relevant to the real cancer environment. Existing three-dimensional models are often requiring sophisticated conditions. Here, we propose and characterize, in two cancer models, melanoma (B16F10) and kidney cancer (RenCa), a three-dimensional culture method, reporting the presence of hypoxia-related genes/proteins and aggressiveness mechanisms (epithelial mesenchymal transition and cancer stem cells). We validate the designed three-dimensional method by comparing it with in vivo growing tumors. The developed method brings simplicity and data reproducibility. Melanoma spheroid-growing cells reached a cell cycle arrest at the G0/G1 phase and showed induction of hypoxia. Spheroid-recovered RenCa cells were enriched in proliferating cells and displayed delayed hypoxia. Moreover, the responses to hypoxia observed in spheroids were validated by in vivo tumor studies for both lines. Three-dimensional shapes induced cancer stem cells in renal cancer, whereas epithelial to mesenchymal transition occurred in the melanoma model. Such distinction in the use of different aggressiveness-leading pathways was observed in in vivo melanoma vs kidney tumors. Thus, this 3D culture model approach is adequate to uncover crucial molecular pathways using distinct mechanisms to reach aggressiveness; i.e., B16F10 cells perform epithelial to mesenchymal transition while RenCa cells dedifferentiate into cancer stem cells. Such three-dimensional models help mimic the in vivo tumor features, i.e., hypoxia and aggressiveness mechanisms as validated here by next-generation sequencing analysis, and are proposed for further alternative methods to in vivo studies.
Collapse
Affiliation(s)
- Aleksandra Filipiak-Duliban
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland. .,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.
| | - Klaudia Brodaczewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Aleksandra Majewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland.,Center for Molecular Biophysics UPR 4301 CNRS, 45071, Orleans, France
| |
Collapse
|
8
|
Heterogeneous Heat Absorption Is Complementary to Radiotherapy. Cancers (Basel) 2022; 14:cancers14040901. [PMID: 35205649 PMCID: PMC8870118 DOI: 10.3390/cancers14040901] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/20/2022] [Accepted: 01/30/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary This review shows the advantages of heterogeneous heating of selected malignant cells in harmonic synergy with radiotherapy. The main clinical achievement of this complementary therapy is its extreme safety and minimal adverse effects. Combining the two methods opens a bright perspective, transforming the local radiotherapy to the antitumoral impact on the whole body, destroying the distant metastases by “teaching” the immune system about the overall danger of malignancy. Abstract (1) Background: Hyperthermia in oncology conventionally seeks the homogeneous heating of the tumor mass. The expected isothermal condition is the basis of the dose calculation in clinical practice. My objective is to study and apply a heterogenic temperature pattern during the heating process and show how it supports radiotherapy. (2) Methods: The targeted tissue’s natural electric and thermal heterogeneity is used for the selective heating of the cancer cells. The amplitude-modulated radiofrequency current focuses the energy absorption on the membrane rafts of the malignant cells. The energy partly “nonthermally” excites and partly heats the absorbing protein complexes. (3) Results: The excitation of the transmembrane proteins induces an extrinsic caspase-dependent apoptotic pathway, while the heat stress promotes the intrinsic caspase-dependent and independent apoptotic signals generated by mitochondria. The molecular changes synergize the method with radiotherapy and promote the abscopal effect. The mild average temperature (39–41 °C) intensifies the blood flow for promoting oxygenation in combination with radiotherapy. The preclinical experiences verify, and the clinical studies validate the method. (4) Conclusions: The heterogenic, molecular targeting has similarities with DNA strand-breaking in radiotherapy. The controlled energy absorption allows using a similar energy dose to radiotherapy (J/kg). The two therapies are synergistically combined.
Collapse
|
9
|
Yan Y, Yao S, Jia Z, Zhao J, Wang L. Iso-suillin-induced DNA damage leading to cell cycle arrest and apoptosis arised from p53 phosphorylation in A549 cells. Eur J Pharmacol 2021; 907:174299. [PMID: 34217708 DOI: 10.1016/j.ejphar.2021.174299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 06/22/2021] [Accepted: 06/29/2021] [Indexed: 01/10/2023]
Abstract
Extensive investigations have revealed that iso-suillin, a secondary metabolite isolated from Suillus flavus, could induce cell cycle arrest and apoptosis in human chronic myeloid leukemia K562 cells, human hepatocellular carcinoma SMMC-7721 cell line, and human small cell lung cancer H446 cell line in vitro. In the present study, human lung cancer A549 cells were used to reveal the mechanism of iso-suillin's effects on lung adenocarcinoma, which were detected both in vitro and in vivo. Results showed that iso-suillin potently inhibited A549 cell proliferation through an early G1 arrest. Iso-suillin also induced A549 cell apoptosis in vitro. Phosphorylation of p53 at serines 15 and 20 may be one of the pivotal factors for cell cycle arrest and apoptosis after treatment of iso-suillin in A549 cells. Moreover, in an A549 xenograft model, tumor growth and progression could be inhibited by iso-suillin. Body weight change and some vital organs toxicity was also roughly examined, no significant toxic effects of iso-suillin were shown (at a dose of 5 mg/kg for each administration). The in vitro and in vivo anti-tumor effects implied that iso-suillin may act as a tumor growth inhibitor, and its induction of p53 phosphorylation is pivotal for cell cycle arrest and apoptosis in A549 cells.
Collapse
Affiliation(s)
- Yongxin Yan
- The Basic Medical College, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Shengjie Yao
- The Basic Medical College, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zhiqiang Jia
- The Basic Medical College, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Junxia Zhao
- The Basic Medical College, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Li'an Wang
- The Life Science College, Hebei Normal University, Shijiazhuang, 050024, PR China
| |
Collapse
|
10
|
Wust P, Stein U, Ghadjar P. Non-thermal membrane effects of electromagnetic fields and therapeutic applications in oncology. Int J Hyperthermia 2021; 38:715-731. [PMID: 33910472 DOI: 10.1080/02656736.2021.1914354] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The temperature-independent effects of electromagnetic fields (EMF) have been controversial for decades. Here, we critically analyze the available literature on non-thermal effects of radiofrequency (RF) and microwave EMF. We present a literature review of preclinical and clinical data on non-thermal antiproliferative effects of various EMF applications, including conventional RF hyperthermia (HT, cRF-HT). Further, we suggest and evaluate plausible biophysical and electrophysiological models to decipher non-thermal antiproliferative membrane effects. Available preclinical and clinical data provide sufficient evidence for the existence of non-thermal antiproliferative effects of exposure to cRF-HT, and in particular, amplitude modulated (AM)-RF-HT. In our model, transmembrane ion channels function like RF rectifiers and low-pass filters. cRF-HT induces ion fluxes and AM-RF-HT additionally promotes membrane vibrations at specific resonance frequencies, which explains the non-thermal antiproliferative membrane effects via ion disequilibrium (especially of Ca2+) and/or resonances causing membrane depolarization, the opening of certain (especially Ca2+) channels, or even hole formation. AM-RF-HT may be tumor-specific owing to cancer-specific ion channels and because, with increasing malignancy, membrane elasticity parameters may differ from that in normal tissues. Published literature suggests that non-thermal antiproliferative effects of cRF-HT are likely to exist and could present a high potential to improve future treatments in oncology.
Collapse
Affiliation(s)
- Peter Wust
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Centrum (MDC), Berlin, Germany
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
11
|
Hannon G, Tansi FL, Hilger I, Prina‐Mello A. The Effects of Localized Heat on the Hallmarks of Cancer. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Gary Hannon
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
| | - Felista L. Tansi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Ingrid Hilger
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Adriele Prina‐Mello
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute Trinity College Dublin Dublin 2 Ireland
| |
Collapse
|
12
|
Vancsik T, Máthé D, Horváth I, Várallyaly AA, Benedek A, Bergmann R, Krenács T, Benyó Z, Balogh A. Modulated Electro-Hyperthermia Facilitates NK-Cell Infiltration and Growth Arrest of Human A2058 Melanoma in a Xenograft Model. Front Oncol 2021; 11:590764. [PMID: 33732640 PMCID: PMC7959784 DOI: 10.3389/fonc.2021.590764] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 01/13/2021] [Indexed: 11/13/2022] Open
Abstract
Modulated electro-hyperthermia (mEHT), induced by 13.56 MHz radiofrequency, has been demonstrated both in preclinical and clinical studies to efficiently induce tumor damage and complement other treatment modalities. Here, we used a mouse xenograft model of human melanoma (A2058) to test mEHT (~42°C) both alone and combined with NK-cell immunotherapy. A single 30 min shot of mEHT resulted in significant tumor damage due to induced stress, marked by high hsp70 expression followed by significant upregulation of cleaved/activated caspase-3 and p53. When mEHT was combined with either primary human NK cells or the IL-2 independent NK-92MI cell line injected subcutaneously, the accumulation of NK cells was observed at the mEHT pretreated melanoma nodules but not at the untreated controls. mEHT induced the upregulation of the chemoattractant CXCL11 and increased the expression of the matrix metalloproteinase MMP2 which could account for the NK-cell attraction into the treated melanoma. In conclusion, mEHT monotherapy of melanoma xenograft tumors induced irreversible heat and cell stress leading to caspase dependent apoptosis to be driven by p53. mEHT could support the intratumoral attraction of distantly injected NK-cells, contributed by CXCL11 and MMP2 upregulation, resulting in an additive tumor destruction and growth inhibition. Therefore, mEHT may offer itself as a good partner for immunotherapy.
Collapse
Affiliation(s)
- Tamás Vancsik
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Ildikó Horváth
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | | | - Anett Benedek
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Ralf Bergmann
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Tibor Krenács
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Andrea Balogh
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
13
|
Suppression of Metastatic Melanoma Growth in Lung by Modulated Electro-Hyperthermia Monitored by a Minimally Invasive Heat Stress Testing Approach in Mice. Cancers (Basel) 2020; 12:cancers12123872. [PMID: 33371498 PMCID: PMC7767533 DOI: 10.3390/cancers12123872] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/05/2020] [Accepted: 12/17/2020] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The lung is the most frequent site of distant melanoma metastases. Metastases of melanoma in the lungs offer a very poor prognosis, with a 5-year survival rate of below 10%. Hyperthermic therapies including modulated electro-hyperthermia (mEHT) in clinical settings have been used to improve the efficacy of radiotherapy, chemotherapy, and immunotherapy of tumors. In this study, we focused primarily on the optimization of mEHT for targeted lung treatment of mice lungs burdened with B16F10 melanoma pulmonary metastases, with a particular focus on elucidating the mechanism of action of mEHT on treated melanoma cells while investigating any potential treatment-related side effects on normal lung tissue. mEHT showed evidence of significant anti-tumor effects as demonstrated by the reduced number of pulmonary metastatic nodules, DNA damage response, downregulation of Ki67 expression, higher immune cell infiltration, and upregulation of p21waf1 expression in mEHT-treated tumors. Abstract Modulated electro-hyperthermia (mEHT) is a novel complementary therapy in oncology which is based on the higher conductivity and permittivity of cancerous tissues due to their enhanced glycolytic activity and ionic content compared to healthy normal tissues. We aimed to evaluate the potential of mEHT, inducing local hyperthermia, in the treatment of pulmonary metastatic melanoma. Our primary objective was the optimization of mEHT for targeted lung treatment as well as to identify the mechanism of its potential anti-tumor effect in the B16F10 mouse melanoma pulmonary metastases model while investigating the potential treatment-related side effects of mEHT on normal lung tissue. Repeated treatment of tumor-bearing lungs with mEHT induced significant anti-tumor effects as demonstrated by the lower number of tumor nodules and the downregulation of Ki67 expression in treated tumor cells. mEHT treatment provoked significant DNA double-strand breaks indicated by the increased expression of phosphorylated H2AX protein in treated tumors, although treatment-induced elevation of cleaved/activated caspase-3 expression was insignificant, suggesting the minimal role of apoptosis in this process. The mEHT-related significant increase in p21waf1 positive tumor cells suggested that p21waf1-mediated cell cycle arrest plays an important role in the anti-tumor effect of mEHT on melanoma metastases. Significantly increased CD3+, CD8+ T-lymphocytes, and F4/80+CD11b+ macrophage density in the whole lung and tumor of treated animals emphasizes the mobilizing capability of mEHT on immune cells. In conclusion, mEHT can reduce the growth potential of melanoma, thus offering itself as a complementary therapeutic option to chemo- and/or radiotherapy.
Collapse
|
14
|
Danics L, Schvarcz CA, Viana P, Vancsik T, Krenács T, Benyó Z, Kaucsár T, Hamar P. Exhaustion of Protective Heat Shock Response Induces Significant Tumor Damage by Apoptosis after Modulated Electro-Hyperthermia Treatment of Triple Negative Breast Cancer Isografts in Mice. Cancers (Basel) 2020; 12:cancers12092581. [PMID: 32927720 PMCID: PMC7565562 DOI: 10.3390/cancers12092581] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Breast cancer is one of the most frequent cancer types among women worldwide. Triple-negative breast cancer is a highly aggressive breast cancer type with very poor survival due to the lack of targeted therapy. Modulated electro-hyperthermia (mEHT) is a newly emerging form of adjuvant, electromagnetic cancer-treatment. Capacitive energy delivery and frequency modulation enable the application of non-thermal effects. Furthermore, selective energy absorption by the tumor (as demonstrated in our present paper) enables 2.5 °C selective heating of the tumor. In the present study, we demonstrate in an in vivo syngeneic Balb/c TNBC mouse model that mEHT caused a remarkable reduction in the number of viable tumor cells accompanied by significant cleaved caspase-3-related apoptotic tumor tissue destruction and a transitional heat shock response. Furthermore, we demonstrated in vitro that the tumor cell killing effect of mEHT was amplified by inhibitors of the protective heat shock response such as Quercetin and KRIBB11. Abstract 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.
Collapse
Affiliation(s)
- Lea Danics
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.D.); (C.A.S.); (P.V.); (T.V.); (Z.B.); (T.K.)
| | - Csaba András Schvarcz
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.D.); (C.A.S.); (P.V.); (T.V.); (Z.B.); (T.K.)
| | - Pedro Viana
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.D.); (C.A.S.); (P.V.); (T.V.); (Z.B.); (T.K.)
| | - Tamás Vancsik
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.D.); (C.A.S.); (P.V.); (T.V.); (Z.B.); (T.K.)
| | - Tibor Krenács
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary;
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.D.); (C.A.S.); (P.V.); (T.V.); (Z.B.); (T.K.)
| | - Tamás Kaucsár
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.D.); (C.A.S.); (P.V.); (T.V.); (Z.B.); (T.K.)
| | - Péter Hamar
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.D.); (C.A.S.); (P.V.); (T.V.); (Z.B.); (T.K.)
- Correspondence:
| |
Collapse
|
15
|
Krenacs T, Meggyeshazi N, Forika G, Kiss E, Hamar P, Szekely T, Vancsik T. Modulated Electro-Hyperthermia-Induced Tumor Damage Mechanisms Revealed in Cancer Models. Int J Mol Sci 2020; 21:E6270. [PMID: 32872532 PMCID: PMC7504298 DOI: 10.3390/ijms21176270] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/18/2022] Open
Abstract
The benefits of high-fever range hyperthermia have been utilized in medicine from the Ancient Greek culture to the present day. Amplitude-modulated electro-hyperthermia, induced by a 13.56 MHz radiofrequency current (mEHT, or Oncothermia), has been an emerging means of delivering loco-regional clinical hyperthermia as a complementary of radiation-, chemo-, and molecular targeted oncotherapy. This unique treatment exploits the metabolic shift in cancer, resulting in elevated oxidative glycolysis (Warburg effect), ion concentration, and electric conductivity. These promote the enrichment of electric fields and induce heat (controlled at 42 °C), as well as ion fluxes and disequilibrium through tumor cell membrane channels. By now, accumulating preclinical studies using in vitro and in vivo models of different cancer types have revealed details of the mechanism and molecular background of the oncoreductive effects of mEHT monotherapy. These include the induction of DNA double-strand breaks, irreversible heath and cell stress, and programmed cells death; the upregulation of molecular chaperones and damage (DAMP) signaling, which may contribute to a secondary immunogenic tumor cell death. In combination therapies, mEHT proved to be a good chemosensitizer through increasing drug uptake and tumor reductive effects, as well as a good radiosensitizer by downregulating hypoxia-related target genes. Recently, immune stimulation or intratumoral antigen-presenting dendritic cell injection have been able to extend the impact of local mEHT into a systemic "abscopal" effect. The complex network of pathways emerging from the published mEHT experiments has not been overviewed and arranged yet into a framework to reveal links between the pieces of the "puzzle". In this paper, we review the mEHT-related damage mechanisms published in tumor models, which may allow some geno-/phenotype treatment efficiency correlations to be exploited both in further research and for more rational clinical treatment planning when mEHT is involved in combination therapies.
Collapse
Affiliation(s)
- Tibor Krenacs
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (N.M.); (G.F.); (T.S.)
| | - Nora Meggyeshazi
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (N.M.); (G.F.); (T.S.)
| | - Gertrud Forika
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (N.M.); (G.F.); (T.S.)
| | - Eva Kiss
- Institute of Oncology at 1st Department of Internal Medicine, Semmelweis University, H-1083 Budapest, Hungary;
| | - Peter Hamar
- Institute of Translational Medicine, Semmelweis University, H-1094 Budapest, Hungary; (P.H.); (T.V.)
| | - Tamas Szekely
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (N.M.); (G.F.); (T.S.)
| | - Tamas Vancsik
- Institute of Translational Medicine, Semmelweis University, H-1094 Budapest, Hungary; (P.H.); (T.V.)
| |
Collapse
|
16
|
Modulated Electro-Hyperthermia Resolves Radioresistance of Panc1 Pancreas Adenocarcinoma and Promotes DNA Damage and Apoptosis In Vitro. Int J Mol Sci 2020; 21:ijms21145100. [PMID: 32707717 PMCID: PMC7404126 DOI: 10.3390/ijms21145100] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
The poor outcome of pancreas ductal adenocarcinomas (PDAC) is frequently linked to therapy resistance. Modulated electro-hyperthermia (mEHT) generated by 13.56 MHz capacitive radiofrequency can induce direct tumor damage and promote chemo- and radiotherapy. Here, we tested the effect of mEHT either alone or in combination with radiotherapy using an in vivo model of Panc1, a KRAS and TP53 mutant, radioresistant PDAC cell line. A single mEHT shot of 60 min induced ~50% loss of viable cells and morphological signs of apoptosis including chromatin condensation, nuclear shrinkage and apoptotic bodies. Most mEHT treatment related effects exceeded those of radiotherapy, and these were further amplified after combining the two modalities. Treatment related apoptosis was confirmed by a significantly elevated number of annexin V single-positive and cleaved/activated caspase-3 positive tumor cells, as well as sub-G1-phase tumor cell fractions. mEHT and mEHT+radioterapy caused the moderate accumulation of γH2AX positive nuclear foci, indicating DNA double-strand breaks and upregulation of the cyclin dependent kinase inhibitor p21waf1 besides the downregulation of Akt signaling. A clonogenic assay revealed that both mono- and combined treatments affected the tumor progenitor/stem cell populations too. In conclusion, mEHT treatment can contribute to tumor growth inhibition and apoptosis induction and resolve radioresistance of Panc1 PDAC cells.
Collapse
|