Modulated Electro-Hyperthermia Accelerates Tumor Delivery and Improves Anticancer Activity of Doxorubicin Encapsulated in Lyso-Thermosensitive Liposomes in 4T1-Tumor-Bearing Mice

Modulated electro-hyperthermia (mEHT) is an adjuvant cancer therapy that enables tumor-selective heating (+2.5 °C). In this study, we investigated whether mEHT accelerates the tumor-specific delivery of doxorubicin (DOX) from lyso-thermosensitive liposomal doxorubicin (LTLD) and improves its antican...

Full description

Bibliographic Details
Main Authors: Kenan Aloss, Syeda Mahak Zahra Bokhari, Pedro Henrique Leroy Viana, Nino Giunashvili, Csaba András Schvarcz, Gábor Szénási, Dániel Bócsi, Zoltán Koós, Gert Storm, Zsuzsanna Miklós, Zoltán Benyó, Péter Hamar
Format: Article
Language:English
Published: MDPI AG 2024-03-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/25/6/3101
_version_ 1797240779882627072
author Kenan Aloss
Syeda Mahak Zahra Bokhari
Pedro Henrique Leroy Viana
Nino Giunashvili
Csaba András Schvarcz
Gábor Szénási
Dániel Bócsi
Zoltán Koós
Gert Storm
Zsuzsanna Miklós
Zoltán Benyó
Péter Hamar
author_facet Kenan Aloss
Syeda Mahak Zahra Bokhari
Pedro Henrique Leroy Viana
Nino Giunashvili
Csaba András Schvarcz
Gábor Szénási
Dániel Bócsi
Zoltán Koós
Gert Storm
Zsuzsanna Miklós
Zoltán Benyó
Péter Hamar
author_sort Kenan Aloss
collection DOAJ
description Modulated electro-hyperthermia (mEHT) is an adjuvant cancer therapy that enables tumor-selective heating (+2.5 °C). In this study, we investigated whether mEHT accelerates the tumor-specific delivery of doxorubicin (DOX) from lyso-thermosensitive liposomal doxorubicin (LTLD) and improves its anticancer efficacy in mice bearing a triple-negative breast cancer cell line (4T1). The 4T1 cells were orthotopically injected into Balb/C mice, and mEHT was performed on days 9, 12, and 15 after the implantation. DOX, LTLD, or PEGylated liposomal DOX (PLD) were administered for comparison. The tumor size and DOX accumulation in the tumor were measured. The cleaved caspase-3 (cC3) and cell proliferation were evaluated by cC3 or Ki67 immunohistochemistry and Western blot. The LTLD+mEHT combination was more effective at inhibiting tumor growth than the free DOX and PLD, demonstrated by reductions in both the tumor volume and tumor weight. LTLD+mEHT resulted in the highest DOX accumulation in the tumor one hour after treatment. Tumor cell damage was associated with cC3 in the damaged area, and with a reduction in Ki67 in the living area. These changes were significantly the strongest in the LTLD+mEHT-treated tumors. The body weight loss was similar in all mice treated with any DOX formulation, suggesting no difference in toxicity. In conclusion, LTLD combined with mEHT represents a novel approach for DOX delivery into cancer tissue.
first_indexed 2024-04-24T18:12:52Z
format Article
id doaj.art-d628f93e5d2f4bd0bac481bc6d86149a
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-04-24T18:12:52Z
publishDate 2024-03-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-d628f93e5d2f4bd0bac481bc6d86149a2024-03-27T13:45:01ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672024-03-01256310110.3390/ijms25063101Modulated Electro-Hyperthermia Accelerates Tumor Delivery and Improves Anticancer Activity of Doxorubicin Encapsulated in Lyso-Thermosensitive Liposomes in 4T1-Tumor-Bearing MiceKenan Aloss0Syeda Mahak Zahra Bokhari1Pedro Henrique Leroy Viana2Nino Giunashvili3Csaba András Schvarcz4Gábor Szénási5Dániel Bócsi6Zoltán Koós7Gert Storm8Zsuzsanna Miklós9Zoltán Benyó10Péter Hamar11Institute of Translational Medicine, Semmelweis University, 1094 Budapest, HungaryInstitute of Translational Medicine, Semmelweis University, 1094 Budapest, HungaryInstitute of Translational Medicine, Semmelweis University, 1094 Budapest, HungaryInstitute of Translational Medicine, Semmelweis University, 1094 Budapest, HungaryInstitute of Translational Medicine, Semmelweis University, 1094 Budapest, HungaryInstitute of Translational Medicine, Semmelweis University, 1094 Budapest, HungaryInstitute of Translational Medicine, Semmelweis University, 1094 Budapest, HungaryInstitute of Translational Medicine, Semmelweis University, 1094 Budapest, HungaryDepartment of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3508 Utrecht, The NetherlandsInstitute of Translational Medicine, Semmelweis University, 1094 Budapest, HungaryInstitute of Translational Medicine, Semmelweis University, 1094 Budapest, HungaryInstitute of Translational Medicine, Semmelweis University, 1094 Budapest, HungaryModulated electro-hyperthermia (mEHT) is an adjuvant cancer therapy that enables tumor-selective heating (+2.5 °C). In this study, we investigated whether mEHT accelerates the tumor-specific delivery of doxorubicin (DOX) from lyso-thermosensitive liposomal doxorubicin (LTLD) and improves its anticancer efficacy in mice bearing a triple-negative breast cancer cell line (4T1). The 4T1 cells were orthotopically injected into Balb/C mice, and mEHT was performed on days 9, 12, and 15 after the implantation. DOX, LTLD, or PEGylated liposomal DOX (PLD) were administered for comparison. The tumor size and DOX accumulation in the tumor were measured. The cleaved caspase-3 (cC3) and cell proliferation were evaluated by cC3 or Ki67 immunohistochemistry and Western blot. The LTLD+mEHT combination was more effective at inhibiting tumor growth than the free DOX and PLD, demonstrated by reductions in both the tumor volume and tumor weight. LTLD+mEHT resulted in the highest DOX accumulation in the tumor one hour after treatment. Tumor cell damage was associated with cC3 in the damaged area, and with a reduction in Ki67 in the living area. These changes were significantly the strongest in the LTLD+mEHT-treated tumors. The body weight loss was similar in all mice treated with any DOX formulation, suggesting no difference in toxicity. In conclusion, LTLD combined with mEHT represents a novel approach for DOX delivery into cancer tissue.https://www.mdpi.com/1422-0067/25/6/3101modulated electro-hyperthermiadoxorubicinlyso-thermosensitive liposomePEGylated liposome
spellingShingle Kenan Aloss
Syeda Mahak Zahra Bokhari
Pedro Henrique Leroy Viana
Nino Giunashvili
Csaba András Schvarcz
Gábor Szénási
Dániel Bócsi
Zoltán Koós
Gert Storm
Zsuzsanna Miklós
Zoltán Benyó
Péter Hamar
Modulated Electro-Hyperthermia Accelerates Tumor Delivery and Improves Anticancer Activity of Doxorubicin Encapsulated in Lyso-Thermosensitive Liposomes in 4T1-Tumor-Bearing Mice
International Journal of Molecular Sciences
modulated electro-hyperthermia
doxorubicin
lyso-thermosensitive liposome
PEGylated liposome
title Modulated Electro-Hyperthermia Accelerates Tumor Delivery and Improves Anticancer Activity of Doxorubicin Encapsulated in Lyso-Thermosensitive Liposomes in 4T1-Tumor-Bearing Mice
title_full Modulated Electro-Hyperthermia Accelerates Tumor Delivery and Improves Anticancer Activity of Doxorubicin Encapsulated in Lyso-Thermosensitive Liposomes in 4T1-Tumor-Bearing Mice
title_fullStr Modulated Electro-Hyperthermia Accelerates Tumor Delivery and Improves Anticancer Activity of Doxorubicin Encapsulated in Lyso-Thermosensitive Liposomes in 4T1-Tumor-Bearing Mice
title_full_unstemmed Modulated Electro-Hyperthermia Accelerates Tumor Delivery and Improves Anticancer Activity of Doxorubicin Encapsulated in Lyso-Thermosensitive Liposomes in 4T1-Tumor-Bearing Mice
title_short Modulated Electro-Hyperthermia Accelerates Tumor Delivery and Improves Anticancer Activity of Doxorubicin Encapsulated in Lyso-Thermosensitive Liposomes in 4T1-Tumor-Bearing Mice
title_sort modulated electro hyperthermia accelerates tumor delivery and improves anticancer activity of doxorubicin encapsulated in lyso thermosensitive liposomes in 4t1 tumor bearing mice
topic modulated electro-hyperthermia
doxorubicin
lyso-thermosensitive liposome
PEGylated liposome
url https://www.mdpi.com/1422-0067/25/6/3101
work_keys_str_mv AT kenanaloss modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice
AT syedamahakzahrabokhari modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice
AT pedrohenriqueleroyviana modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice
AT ninogiunashvili modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice
AT csabaandrasschvarcz modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice
AT gaborszenasi modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice
AT danielbocsi modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice
AT zoltankoos modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice
AT gertstorm modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice
AT zsuzsannamiklos modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice
AT zoltanbenyo modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice
AT peterhamar modulatedelectrohyperthermiaacceleratestumordeliveryandimprovesanticanceractivityofdoxorubicinencapsulatedinlysothermosensitiveliposomesin4t1tumorbearingmice