1
|
Dewhirst MW. A translational review of hyperthermia biology. Int J Hyperthermia 2025; 42:2447952. [PMID: 39799944 DOI: 10.1080/02656736.2024.2447952] [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: 09/14/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025] Open
Abstract
This review was written to be included in the Special Collection 'Therapy Ultrasound: Medicine's Swiss Army Knife?' The purpose of this review is to provide basic presentation and interpretation of the fundamentals of hyperthermia biology, as it pertains to uses of therapeutic ultrasound. The fundamentals are presented but in the setting of a translational interpretation and a view toward the future. Subjects that require future research and development are highlighted. The effects of hyperthermia are time and temperature dependent. Because intra-tumoral temperatures are non-uniform in tumors, one has to account for differential biologic effects in different parts of a tumor that occur simultaneously during and after hyperthermia.
Collapse
Affiliation(s)
- Mark W Dewhirst
- Gustavo S. Montana Distinguished Professor Emeritus of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
2
|
Wang Y, Xun X, Luan WY, Zhang Z, Xu ZX, Lin SX, Miao YD. Hyperthermia combined with opioid therapy: Enhancing cancer pain management and reducing surgical stress in gastrointestinal cancer patients. World J Gastrointest Surg 2025; 17:101060. [PMID: 40162416 PMCID: PMC11948118 DOI: 10.4240/wjgs.v17.i3.101060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/17/2025] [Accepted: 02/07/2025] [Indexed: 02/24/2025] Open
Abstract
In this article, we evaluate the findings of the study by Qian et al, which explores the efficacy of combining hyperthermia with opioid therapy for enhanced cancer pain management in patients with middle and late-stage gastrointestinal tumors. The study undertakes a retrospective analysis comparing traditional opioid therapy to an integrated approach of hyperthermia and opioids across 70 patients, highlighting significant benefits in pain control, reduction of opioid dosage, and minimization of adverse reactions. In our article, we not only discuss these findings but also emphasize the broader implications for clinical practice, particularly in enhancing patient outcomes through innovative pain management strategies. We advocate for further research to establish more robust data supporting this approach and to explore the mechanistic insights that enable these benefits. This discussion reflects on the potential paradigm shift in managing debilitating cancer-related pain, urging a reevaluation of current practices to incorporate these findings effectively.
Collapse
Affiliation(s)
- Yue Wang
- Cancer Center, People’s Hospital of Yuxi City, Yuxi 653100, Yunnan Province, China
| | - Xin Xun
- Department of Oncology, 920th Hospital of People’s Liberation Army Joint Logistics Support Force, Kunming 650118, Yunnan Province, China
| | - Wen-Yu Luan
- Department of Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The Second Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Zheng Zhang
- Department of Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The Second Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Zhen-Xi Xu
- Department of Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The Second Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Si-Xiang Lin
- Department of Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The Second Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Yan-Dong Miao
- Department of Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The Second Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| |
Collapse
|
3
|
Müller M, Ludwig L, Englert H, Riedl KA, Müller MC, Hemkemeyer SA, Beerens M, Mailer RK, Renné T, Lang S, Baumann-Zumstein P, Frye M. A novel stent flow chamber system demonstrates reduced thrombogenicity of bioresorbable magnesium scaffolds. Sci Rep 2024; 14:26691. [PMID: 39496698 PMCID: PMC11535548 DOI: 10.1038/s41598-024-77266-0] [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: 06/24/2024] [Accepted: 10/21/2024] [Indexed: 11/06/2024] Open
Abstract
Coronary artery disease (CAD) is characterized by narrowing and subsequent blockade of coronary arteries, and imposes a significant health and economic burden. Stent and scaffold devices are introduced in advanced CAD to improve vascular stability and restore blood flow. Although in vitro flow systems like the Chandler loop have been developed to enhance the understanding of interactions between device materials, their coatings, and vascular cells, imaging-based in vitro analysis of device performance is limited. In this study, we established a novel stent flow chamber system designed to assess the thrombogenicity of bioresorbable magnesium scaffold (RMS) and stent materials in vitro. Additionally, we compared the thrombogenicity - an important clinical parameter in stent performance - of the Magmaris-316 L stainless steel stent with its predecessors, Magmaris RMS and a prototype of the third-generation RMS (DREAMS 3G). Analysis of platelet adhesion and coverage of the different devices under flow conditions demonstrated that the Magmaris RMS exhibits reduced thrombogenicity compared to the Magmaris-316 L stainless steel stent. Moreover, thrombogenicity of the DREAMS 3G prototype, composed of BIOmag material, is further decreased compared to its predecessors. The observed reduction in thrombogenicity of the DREAMS 3G prototype in vitro suggests additional improvements in clinical safety and efficacy, highlighting its promise for treating CAD. Future research on this prototype may thus open avenues for analyzing other blood components and patient-derived endothelial cells. In line with the 3R principles, this approach may also help reduce the need for animal testing.
Collapse
Affiliation(s)
- Monja Müller
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Ludwig
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hanna Englert
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina A Riedl
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Interdisciplinary Cardiac Imaging, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - May Cathleen Müller
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra A Hemkemeyer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Manu Beerens
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Reiner K Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | | | - Maike Frye
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany.
| |
Collapse
|
4
|
Scarmelotto A, Delprat V, Michiels C, Lucas S, Heuskin AC. The oxygen puzzle in FLASH radiotherapy: A comprehensive review and experimental outlook. Clin Transl Radiat Oncol 2024; 49:100860. [PMID: 39381632 PMCID: PMC11458961 DOI: 10.1016/j.ctro.2024.100860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
FLASH radiotherapy is attracting increasing interest because it maintains tumor control while inflicting less damage to normal tissues compared to conventional radiotherapy. This sparing effect, the so-called FLASH effect, is achieved when radiation is delivered at ultra-high dose rates (≥40 Gy/s). Although the FLASH effect has already been demonstrated in several preclinical models, a complete mechanistic description explaining why tumors and normal tissues respond differently is still missing. None of the current hypotheses fully explains the experimental evidence. A common point between many of these is the role of oxygen, which is described as a major factor, either through transient hypoxia in the form of dissolved molecules, or reactive oxygen species (ROS). Therefore, this review focuses on both forms of this molecule, retracing old and more recent theories, while proposing new mechanisms that could provide a complete description of the FLASH effect based on preclinical and experimental evidence. In addition, this manuscript describes a set of experiments designed to provide the FLASH community with new tools for exploring the post-irradiation fate of ROS and their potential biological implications.
Collapse
Affiliation(s)
- Andrea Scarmelotto
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Victor Delprat
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Carine Michiels
- Unité de Recherche en Biologie Cellulaire (URBC), Namur Research Institute For Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Stéphane Lucas
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
- Ion Beam Application (IBA), Chemin du Cyclotron, 6, B-1348 Louvain-La-Neuve, Belgium
| | - Anne-Catherine Heuskin
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| |
Collapse
|
5
|
Ifijen IH, Christopher AT, Lekan OK, Aworinde OR, Faderin E, Obembe O, Abdulsalam Akanji TF, Igboanugo JC, Udogu U, Ogidi GO, Iorkula TH, Osayawe OJK. Advancements in tantalum based nanoparticles for integrated imaging and photothermal therapy in cancer management. RSC Adv 2024; 14:33681-33740. [PMID: 39450067 PMCID: PMC11498270 DOI: 10.1039/d4ra05732e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024] Open
Abstract
Tantalum-based nanoparticles (TaNPs) have emerged as promising tools in cancer management, owing to their unique properties that facilitate innovative imaging and photothermal therapy applications. This review provides a comprehensive overview of recent advancements in TaNPs, emphasizing their potential in oncology. Key features include excellent biocompatibility, efficient photothermal conversion, and the ability to integrate multifunctional capabilities, such as targeted drug delivery and enhanced imaging. Despite these advantages, challenges remain in establishing long-term biocompatibility, optimizing therapeutic efficacy through surface modifications, and advancing imaging techniques for real-time monitoring. Strategic approaches to address these challenges include surface modifications like PEGylation to improve biocompatibility, precise control over size and shape for effective photothermal therapy, and the development of biodegradable TaNPs for safe elimination from the body. Furthermore, integrating advanced imaging modalities-such as photoacoustic imaging, magnetic resonance imaging (MRI), and computed tomography (CT)-enable real-time tracking of TaNPs in vivo, which is crucial for clinical applications. Personalized medicine strategies that leverage biomarkers and genetic profiling also hold promise for tailoring TaNP-based therapies to individual patient profiles, thereby enhancing treatment efficacy and minimizing side effects. In conclusion, TaNPs represent a significant advancement in nanomedicine, poised to transform cancer treatment paradigms while expanding into various biomedical applications.
Collapse
Affiliation(s)
- Ikhazuagbe H Ifijen
- Department of Research Outreach, Rubber Research Institute of Nigeria Iyanomo Benin City Nigeria
| | - Awoyemi Taiwo Christopher
- Laboratory Department, Covenant University Medical Centre Canaan land, KM 10, Idiroko Road Ota Ogun State Nigeria
| | - Ogunnaike Korede Lekan
- Department of Chemistry, Wichita State University 1845 Fairmount, Box 150 Wichita KS 67260-0150 USA
| | | | - Emmanuel Faderin
- Department of Pharmaceutical Sciences, Southern Illinois University Edwardsville, 1 Hairpin Drive Edwardsville IL 62026-001 USA
| | | | | | - Juliet C Igboanugo
- Department of Health, Human Performance, and Recreation 155 Stadium Drive Arkansas 72701 USA
| | - Uzochukwu Udogu
- Department of Chemistry, Federal University of Technology Owerri Nigeria
| | | | - Terungwa H Iorkula
- Department of Chemistry and Biochemistry, Brigham Young University Provo Utah USA
| | | |
Collapse
|
6
|
Aparicio-Lopez CB, Timmerman S, Lorino G, Rogers T, Pyle M, Shrestha TB, Basel MT. Thermosensitive Liposomes for Gemcitabine Delivery to Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:3048. [PMID: 39272906 PMCID: PMC11394165 DOI: 10.3390/cancers16173048] [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: 07/26/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Treatment of pancreatic ductal adenocarcinoma with gemcitabine is limited by an increased desmoplasia, poor vascularization, and short plasma half-life. Heat-sensitive liposomes modified by polyethylene glycol (PEG; PEGylated liposomes) can increase plasma stability, reduce clearance, and decrease side effects. Nevertheless, translation of heat-sensitive liposomes to the clinic has been hindered by the low loading efficiency of gemcitabine and by the difficulty of inducing hyperthermia in vivo. This study was designed to investigate the effect of phospholipid content on the stability of liposomes at 37 °C and their release under hyperthermia conditions; this was accomplished by employing a two-stage heating approach. First the liposomes were heated at a fast rate, then they were transferred to a holding bath. Thermosensitive liposomes formulated with DPPC: DSPC: PEG2k (80:15:5, mole%) exhibited minimal release of carboxyfluorescein at 37 °C over 30 min, indicating stability under physiological conditions. However, upon exposure to hyperthermic conditions (43 °C and 45 °C), these liposomes demonstrated a rapid and significant release of their encapsulated content. The encapsulation efficiency for gemcitabine was calculated at 16.9%. Additionally, fluorescent analysis during the removal of unencapsulated gemcitabine revealed an increase in pH. In vitro tests with BxPC3 and KPC cell models showed that these thermosensitive liposomes induced a heat-dependent cytotoxic effect comparable to free gemcitabine at temperatures above 41 °C. This study highlights the effectiveness of the heating mechanism and cell models in understanding the current challenges in developing gemcitabine-loaded heat-sensitive liposomes.
Collapse
Affiliation(s)
- Cesar B Aparicio-Lopez
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Sarah Timmerman
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Gabriella Lorino
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Tatiana Rogers
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Marla Pyle
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Tej B Shrestha
- Nanotechnology Innovation Center of Kansas State (NICKS), Kansas State University, Manhattan, KS 66506, USA
| | - Matthew T Basel
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
7
|
Righini MF, Durham A, Tsoutsou PG. Hyperthermia and radiotherapy: physiological basis for a synergistic effect. Front Oncol 2024; 14:1428065. [PMID: 39165690 PMCID: PMC11333208 DOI: 10.3389/fonc.2024.1428065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024] Open
Abstract
In cancer treatment, mild hyperthermia (HT) represents an old, but recently revived opportunity to increase the efficacy of radiotherapy (RT) without increasing side effects, thereby widening the therapeutic window. HT disrupts cellular homeostasis by acting on multiple targets, and its combination with RT produces synergistic antitumoral effects on specific pathophysiological mechanisms, associated to DNA damage and repair, hypoxia, stemness and immunostimulation. HT is furthermore associated to direct tumor cell kill, particularly in higher temperature levels. A phenomenon of temporary resistance to heat, known as thermotolerance, follows each HT session. Cancer treatment requires innovative concepts and combinations to be tested but, for a meaningful development of clinical trials, the understanding of the underlying mechanisms of the tested modalities is essential. In this mini-review, we aimed to describe the synergistic effects of the combination of HT with RT as well as the phenomena of thermal shock and thermotolerance, in order to stimulate clinicians in new, clinically relevant concepts and combinations, which become particularly relevant in the era of technological advents in both modalities but also cancer immunotherapy.
Collapse
Affiliation(s)
| | - André Durham
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
- Department of Radiation Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Pelagia G. Tsoutsou
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
- Department of Radiation Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| |
Collapse
|
8
|
Xiang Y, Tang L, Pang H, Xu H, He Y, Feng Y, Ju L, Zhang L, Wang D. Ultrasound -Induced Thermal Effect Enhances the Efficacy of Chemotherapy and Immunotherapy in Tumor Treatment. Int J Nanomedicine 2024; 19:6677-6692. [PMID: 38975322 PMCID: PMC11227868 DOI: 10.2147/ijn.s464830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/11/2024] [Indexed: 07/09/2024] Open
Abstract
Background The inadequate perfusion, frequently resulting from abnormal vascular configuration, gives rise to tumor hypoxia. The presence of this condition hinders the effective delivery of therapeutic drugs and the infiltration of immune cells into the tumor, thereby compromising the efficacy of treatments against tumors. The objective of this study is to exploit the thermal effect of ultrasound (US) in order to induce localized temperature elevation within the tumor, thereby facilitating vasodilation, augmenting drug delivery, and enhancing immune cell infiltration. Methods The selection of US parameters was based on intratumor temperature elevation and their impact on cell viability. Vasodilation and hypoxia improvement were investigated using enzyme-linked immunosorbent assay (ELISA) and immunofluorescence examination. The distribution and accumulation of commercial pegylated liposomal doxorubicin (PLD) and PD-L1 antibody (anti-PD-L1) in the tumor were analyzed through frozen section analysis, ELISA, and in vivo fluorescence imaging. The evaluation of tumor immune microenvironment was conducted using flow cytometry (FCM). The efficacy of US-enhanced chemotherapy in combination with immunotherapy was investigated by monitoring tumor growth and survival rate after various treatments. Results The US irradiation condition of 0.8 W/cm2 for 10 min effectively elevated the tumor temperature to approximately 40 °C without causing any cellular or tissue damage, and sufficiently induced vasodilation, thereby enhancing the distribution and delivery of PLD and anti-PD-L1 in US-treated tumors. Moreover, it effectively mitigated tumor hypoxia while significantly increasing M1-phenotype tumor-associated macrophages (TAMs) and CD8+ T cells, as well as decreasing M2-phenotype TAMs. By incorporating US irradiation, the therapeutic efficacy of PLD and anti-PD-L1 was substantially boosted, leading to effective suppression of tumor growth and prolonged survival in mice. Conclusion The application of US (0.8 W/cm2 for 10 min) can effectively induce vasodilation and enhance the delivery of PLD and anti-PD-L1 into tumors, thereby reshaping the immunosuppressive tumor microenvironment and optimizing therapeutic outcomes.
Collapse
Affiliation(s)
- Yuting Xiang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Li Tang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Hua Pang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Han Xu
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yiman He
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yuyue Feng
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Linjun Ju
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Liang Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Dong Wang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
9
|
Aloss K, Hamar P. Augmentation of the EPR effect by mild hyperthermia to improve nanoparticle delivery to the tumor. Biochim Biophys Acta Rev Cancer 2024; 1879:189109. [PMID: 38750699 DOI: 10.1016/j.bbcan.2024.189109] [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: 02/12/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
The clinical translation of the nanoparticle (NP)-based anticancer therapies is still unsatisfactory due to the heterogeneity of the enhanced permeability and retention (EPR) effect. Despite the promising preclinical outcome of the pharmacological EPR enhancers, their systemic toxicity can limit their clinical application. Hyperthermia (HT) presents an efficient tool to augment the EPR by improving tumor blood flow (TBF) and vascular permeability, lowering interstitial fluid pressure (IFP), and disrupting the structure of the extracellular matrix (ECM). Furthermore, the HT-triggered intravascular release approach can overcome the EPR effect. In contrast to pharmacological approaches, HT is safe and can be focused to cancer tissues. Moreover, HT conveys direct anti-cancer effects, which improve the efficacy of the anti-cancer agents encapsulated in NPs. However, the clinical application of HT is challenging due to the heterogeneous distribution of temperature within the tumor, the length of the treatment and the complexity of monitoring.
Collapse
Affiliation(s)
- Kenan Aloss
- Institute of Translational Medicine - Semmelweis University - 1094, Tűzoltó utca, 37-49, Budapest, Hungary
| | - Péter Hamar
- Institute of Translational Medicine - Semmelweis University - 1094, Tűzoltó utca, 37-49, Budapest, Hungary.
| |
Collapse
|
10
|
Liu P, Wu J, Chen L, Wu Z, Wu Y, Zhang G, Yu B, Zhang B, Wei N, Shi J, Zhang C, Lei L, Yu S, Lai J, Guo Z, Zheng Y, Jing Z, Jiang H, Wang T, Zhou J, Wu Y, Sun C, Shen J, Zhang J, Wu Z. Water-filtered infrared A radiation hyperthermia combined with immunotherapy for advanced gastrointestinal tumours. Cancer Med 2024; 13:e70024. [PMID: 39049187 PMCID: PMC11269209 DOI: 10.1002/cam4.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 06/18/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
This study pioneered the use of WIRA whole-body infrared hyperthermia combined with ICI therapy to treat GIT and verified the feasibility and safety of HIT. The final results showed a DCR of 55.6%, with a median PFS of 53.5 days, median OS of 134 days, and an irAE incidence of 22.2%. Therefore, we believe that HIT can exert multiple synergistic sensitisation effects, thereby providing clinical benefits to patients with advanced GITs, increasing overall safety, and improving patients' QOL.
Collapse
Affiliation(s)
- Pengyuan Liu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jing Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Liting Chen
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhenhai Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Yufei Wu
- ACS (International) School of SingaporeSingapore
| | - Ganlu Zhang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Bingqi Yu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Beibei Zhang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Nan Wei
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jinan Shi
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | | | - Lan Lei
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Shuhuan Yu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jianjun Lai
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhen Guo
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Yuli Zheng
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhao Jing
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Hao Jiang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | | | - Jueyi Zhou
- Department of OncologyLishui People's HospitalLishuiChina
| | - Yajun Wu
- TCM Dispensary, Zhejiang HospitalHangzhouChina
| | - Chuan Sun
- Geriatrics Institute of Zhejiang ProvinceDepartment of Geriatrics, Zhejiang HospitalHangzhouChina
| | - Jie Shen
- Department of Medical Oncology, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jian Zhang
- Department of Gastrointestinal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Zhibing Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
- Department of Radiation Oncology, Affiliated Zhejiang HospitalZhejiang University School of MedicineHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| |
Collapse
|
11
|
de Arriba M, Borel N, LeibundGut-Landmann S. Water-filtered infrared A irradiation exerts antifungal effects on the skin fungus Malassezia. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 255:112909. [PMID: 38669741 DOI: 10.1016/j.jphotobiol.2024.112909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024]
Abstract
Many common skin diseases are associated with changes in the microbiota. This applies for the commensal yeast Malassezia, which is linked to a wide range of skin disorders ranging from mild dandruff to severe seborrheic and atopic dermatitis, all of which have a detrimental impact on the individuals' quality of life. While antifungal medications offer relief in many cases, the challenges of disease recurrence and the emergence of resistance to the limited range of available antifungal drugs poses a pressing need for innovative therapeutic options. Here we examined the activity of water-filtered infrared A (wIRA) irradiation against Malassezia. wIRA's antimicrobial and wound healing properties make it an attractive option for localized, non-invasive, and contact-free treatment of superficial skin infections. Irradiation of Malassezia furfur with wIRA (570-1400 nm) resulted in a reduction of the yeast's metabolic activity. When put in contact with immune cells, wIRA-irradiated M. furfur was recovered at lower counts than non-irradiated M. furfur. Likewise, wIRA irradiation of M. furfur put in contact with keratinocytes, the primary host interface of the fungus in the skin, reduced the fungal counts, while the keratinocytes were not affected by the irradiation. The combination of wIRA with the photosensitizer methyl aminolevulinate exerted an additional antifungal effect on M. furfur, irrespective of the presence or absence of keratinocytes, suggesting an enhancement of the treatment effect when used in combination. These findings suggest that wIRA holds promise as a potential therapy for skin disorders associated with Malassezia.
Collapse
Affiliation(s)
- Magdalena de Arriba
- Section of Immunology, Vetsuisse-Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057 Zürich, Switzerland; Institute of Veterinary Pathology, Vetsuisse-Faculty, University of Zurich, Winterthurerstrasse 268, CH 8057 Zürich, Switzerland
| | - Nicole Borel
- Institute of Veterinary Pathology, Vetsuisse-Faculty, University of Zurich, Winterthurerstrasse 268, CH 8057 Zürich, Switzerland
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse-Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057 Zürich, Switzerland; Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland.
| |
Collapse
|
12
|
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
|
13
|
Logghe T, van Zwol E, Immordino B, Van den Cruys K, Peeters M, Giovannetti E, Bogers J. Hyperthermia in Combination with Emerging Targeted and Immunotherapies as a New Approach in Cancer Treatment. Cancers (Basel) 2024; 16:505. [PMID: 38339258 PMCID: PMC10854776 DOI: 10.3390/cancers16030505] [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: 11/30/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Despite significant advancements in the development of novel therapies, cancer continues to stand as a prominent global cause of death. In many cases, the cornerstone of standard-of-care therapy consists of chemotherapy (CT), radiotherapy (RT), or a combination of both. Notably, hyperthermia (HT), which has been in clinical use in the last four decades, has proven to enhance the effectiveness of CT and RT, owing to its recognized potency as a sensitizer. Furthermore, HT exerts effects on all steps of the cancer-immunity cycle and exerts a significant impact on key oncogenic pathways. Most recently, there has been a noticeable expansion of cancer research related to treatment options involving immunotherapy (IT) and targeted therapy (TT), a trend also visible in the research and development pipelines of pharmaceutical companies. However, the potential results arising from the combination of these innovative therapeutic approaches with HT remain largely unexplored. Therefore, this review aims to explore the oncology pipelines of major pharmaceutical companies, with the primary objective of identifying the principal targets of forthcoming therapies that have the potential to be advantageous for patients by specifically targeting molecular pathways involved in HT. The ultimate goal of this review is to pave the way for future research initiatives and clinical trials that harness the synergy between emerging IT and TT medications when used in conjunction with HT.
Collapse
Affiliation(s)
- Tine Logghe
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
| | - Eke van Zwol
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
| | - Benoît Immordino
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, 56017 Pisa, Italy
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, 56127 Pisa, Italy
| | | | - Marc Peeters
- Department of Oncology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Elisa Giovannetti
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, 56017 Pisa, Italy
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Johannes Bogers
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
- Laboratory of Cell Biology and Histology, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
14
|
Orel VE, Diedkov AG, Ostafiichuk VV, Lykhova OO, Kolesnyk DL, Orel VB, Dasyukevich OY, Rykhalskyi OY, Diedkov SA, Prosvietova AB. Combination Treatment with Liposomal Doxorubicin and Inductive Moderate Hyperthermia for Sarcoma Saos-2 Cells. Pharmaceuticals (Basel) 2024; 17:133. [PMID: 38276006 PMCID: PMC10819935 DOI: 10.3390/ph17010133] [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: 11/30/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Despite efforts in osteosarcoma (OS) research, the role of inductive moderate hyperthermia (IMH) in delivering and enhancing the antitumor effect of liposomal doxorubicin formulations (LDOX) remains unresolved. This study investigated the effect of a combination treatment with LDOX and IMH on Saos-2 human OS cells. We compared cell viability using a trypan blue assay, apoptosis and reactive oxygen species (ROS) measured by flow cytometry and pro-apoptotic Bax protein expression examined by immunocytochemistry in response to IMH (42 MHz frequency, 15 W power for 30 min), LDOX (0.4 μg/mL), and LDOX plus IMH. The lower IC50 value of LDOX at 72 h indicated increased accumulation of the drug in the OS cells. LDOX plus IMH resulted in a 61% lower cell viability compared to no treatment. Moreover, IMH potentiated the LDOX action on the Saos-2 cells by promoting ROS production at temperatures of <42 °C. There was a 12% increase in cell populations undergoing early apoptosis with a less heterogeneous distribution of Bax after combination treatment compared to those treated with LDOX (p < 0.05). Therefore, we determined that IMH could enhance LDOX delivery and its antitumor effect via altered membrane permeabilization, ROS generation, and a lower level of visualized Bax heterogeneity in the Saos-2 cells, suggesting the potential translation of these findings into in vivo studies.
Collapse
Affiliation(s)
- Valerii E. Orel
- National Cancer Institute, 33/43 Zdanovska Str., 03022 Kyiv, Ukraine
- National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, 16/2 Yangel Str., 03056 Kyiv, Ukraine
| | | | | | - Oleksandra O. Lykhova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, 45 Vasylkivska Str., 03022 Kyiv, Ukraine
| | - Denys L. Kolesnyk
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, 45 Vasylkivska Str., 03022 Kyiv, Ukraine
| | - Valerii B. Orel
- National Cancer Institute, 33/43 Zdanovska Str., 03022 Kyiv, Ukraine
- National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, 16/2 Yangel Str., 03056 Kyiv, Ukraine
| | | | | | - Serhii A. Diedkov
- National Cancer Institute, 33/43 Zdanovska Str., 03022 Kyiv, Ukraine
| | - Anna B. Prosvietova
- National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, 16/2 Yangel Str., 03056 Kyiv, Ukraine
| |
Collapse
|
15
|
Lukácsi S, Munkácsy G, Győrffy B. Harnessing Hyperthermia: Molecular, Cellular, and Immunological Insights for Enhanced Anticancer Therapies. Integr Cancer Ther 2024; 23:15347354241242094. [PMID: 38818970 PMCID: PMC11143831 DOI: 10.1177/15347354241242094] [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: 12/28/2023] [Revised: 02/25/2024] [Accepted: 03/11/2024] [Indexed: 06/01/2024] Open
Abstract
Hyperthermia, the raising of tumor temperature (≥39°C), holds great promise as an adjuvant treatment for cancer therapy. This review focuses on 2 key aspects of hyperthermia: its molecular and cellular effects and its impact on the immune system. Hyperthermia has profound effects on critical biological processes. Increased temperatures inhibit DNA repair enzymes, making cancer cells more sensitive to chemotherapy and radiation. Elevated temperatures also induce cell cycle arrest and trigger apoptotic pathways. Furthermore, hyperthermia modifies the expression of heat shock proteins, which play vital roles in cancer therapy, including enhancing immune responses. Hyperthermic treatments also have a significant impact on the body's immune response against tumors, potentially improving the efficacy of immune checkpoint inhibitors. Mild systemic hyperthermia (39°C-41°C) mimics fever, activating immune cells and raising metabolic rates. Intense heat above 50°C can release tumor antigens, enhancing immune reactions. Using photothermal nanoparticles for targeted heating and drug delivery can also modulate the immune response. Hyperthermia emerges as a cost-effective and well-tolerated adjuvant therapy when integrated with immunotherapy. This comprehensive review serves as a valuable resource for the selection of patient-specific treatments and the guidance of future experimental studies.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
| | - Gyöngyi Munkácsy
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
| | - Balázs Győrffy
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
- University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| |
Collapse
|
16
|
Thomsen AR, Sahlmann J, Bronsert P, Schilling O, Poensgen F, May AM, Timme-Bronsert S, Grosu AL, Vaupel P, Gebbers JO, Multhoff G, Lüchtenborg AM. Protocol of the HISTOTHERM study: assessing the response to hyperthermia and hypofractionated radiotherapy in recurrent breast cancer. Front Oncol 2023; 13:1275222. [PMID: 38169879 PMCID: PMC10759986 DOI: 10.3389/fonc.2023.1275222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/20/2023] [Indexed: 01/05/2024] Open
Abstract
Introduction Breast cancer is globally the leading cancer in women, and despite the high 5-year survival rate the most frequent cause of cancer related deaths. Surgery, systemic therapy and radiotherapy are the three pillars of curative breast cancer treatment. However, locoregional recurrences frequently occur after initial treatment and are often challenging to treat, amongst others due to high doses of previous radiotherapy treatments. Radiotherapy can be combined with local hyperthermia to sensitize tumor cells to radiation and thereby significantly reduce the required radiation dose. Therefore, the combination treatment of mild local hyperthermia, i.e. locally heating of the tissue to 39-43°C, and re-irradiation with a reduced total dose is a relevant treatment option for previously irradiated patients. The mechanisms of this effect in the course of the therapy are to date not well understood and will be investigated in the HISTOTHERM study. Methods and analyses Patients with local or (loco)regional recurrent breast cancer with macroscopic tumors are included in the study. Local tumor control is evaluated clinically and histologically during the course of a combination treatment of 60 minutes mild superficial hyperthermia (39 - 43°C) using water-filtered infrared A (wIRA) irradiation, immediately followed by hypofractionated re-irradiation with a total dose of 20-24 Gy, administered in weekly doses of 4 Gy. Tumor and tumor stroma biopsies as well as blood samples will be collected prior to treatment, during therapy (at a dose of 12 Gy) and in the follow-up to monitor therapy response. The treatment represents the standard operating procedure for hyperthermia plus re-irradiation. Various tissue and blood-based markers are analyzed. We aim at pinpointing key mechanisms and markers for therapy response which may help guiding treatment decisions in future. In addition, quality of life in the course of treatment will be assessed and survival data will be evaluated. Registration The study is registered at the German Clinical Trials Register, Deutsches Register Klinischer Studien (DRKS00029221).
Collapse
Affiliation(s)
- Andreas R. Thomsen
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
| | - Jörg Sahlmann
- Institute for Medical Biometry and Statistics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Bronsert
- Tumorbank Comprehensive Cancer Center Freiburg, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felicia Poensgen
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Pediatric Department, Black Forest Baar Clinic, Villingen-Schwenningen, Germany
| | - Annette M. May
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Medizinisches Versorgungszentrum Laaff, Freiburg, Germany
| | - Sylvia Timme-Bronsert
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
| | - Peter Vaupel
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
| | - Jan-Olaf Gebbers
- Department of Pathology, Working Group Digital Pathology, University of Berne, Bern, Switzerland
| | - Gabriele Multhoff
- Center for Translational Cancer Research, Klinikum rechts der Isar, Department of Radiation Oncology, Technical University Munich (TUM), Munich, Germany
| | - Anne-Marie Lüchtenborg
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
| |
Collapse
|
17
|
Priyam J, Saxena U. Therapeutic applications of carbon nanomaterials in renal cancer. Biotechnol Lett 2023; 45:1395-1416. [PMID: 37864745 DOI: 10.1007/s10529-023-03429-0] [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: 06/07/2023] [Revised: 08/27/2023] [Accepted: 08/31/2023] [Indexed: 10/23/2023]
Abstract
Carbon nanomaterials (CNMs), including carbon nanotubes (CNTs), graphene, and nanodiamonds (NDs), have shown great promise in detecting and treating numerous cancers, including kidney cancer. CNMs can increase the sensitivity of diagnostic techniques for better kidney cancer identification and surveillance. They enable targeted medicine delivery specifically to tumour locations, with little effect on healthy tissue. Because of their unique chemical and physical characteristics, they can avoid the body's defence mechanisms, making it easier to accumulate where tumours exist. Consequently, CNMs provide more effective drug delivery to kidney cancer cells. It also helps in improving the efficacy of treatment. This review explores the potential of several CNMs in improving therapeutic strategies for kidney cancer. We briefly covered the physicochemical properties and therapeutic applications of CNMs. Additionally, we discussed how structural modifications in CNMs enhance their precision in treating renal cancer. A thorough overview of CNM-based gene, peptide, and drug delivery strategies for the treatment of renal cancer is presented in this review. It covers information on other CNM-based therapeutic approaches, such as hyperthermia, photodynamic therapy, and photoacoustic therapy. Also, the interactions of CNMs with the tumour microenvironment (TME) are explored, including modulation of the immune response, regulation of tumour hypoxia, interactions between CNMs and TME cells, effects of TME pH on CNMs, and more. Finally, potential side effects of CNMs, such as toxicity, bio corona formation, enzymatic degradation, and biocompatibility, are also discussed.
Collapse
Affiliation(s)
- Jyotsna Priyam
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, 506004, India
| | - Urmila Saxena
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, 506004, India.
| |
Collapse
|
18
|
Ferioli M, Perrone AM, Buwenge M, Arcelli A, Vadala’ M, Fionda B, Malato MC, De Iaco P, Zamagni C, Cammelli S, Tagliaferri L, Morganti AG. Combination of Electrochemotherapy with Radiotherapy: A Comprehensive, Systematic, PRISMA-Compliant Review of Efficacy and Potential Radiosensitizing Effects in Tumor Control. Curr Oncol 2023; 30:9895-9905. [PMID: 37999139 PMCID: PMC10670517 DOI: 10.3390/curroncol30110719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/25/2023] Open
Abstract
Radiotherapy (RT) and electrochemotherapy (ECT) are established local treatments for cancer. While effective, both therapies have limitations, especially in treating bulky and poorly oxygenated tumors. ECT has emerged as a promising palliative treatment, raising interest in exploring its combination with RT to enhance tumor response. However, the potential benefits and challenges of combining these treatments remain unclear. A systematic review was conducted following PRISMA guidelines. PubMed, Scopus, and Cochrane libraries were searched. Studies were screened and selected based on predefined inclusion and exclusion criteria. Ten studies were included, comprising in vitro and in vivo experiments. Different tumor types were treated with ECT alone or in combination with RT. ECT plus RT demonstrated superior tumor response compared to that under single therapies or other combinations, regardless of the cytotoxic agent and RT dose. However, no study demonstrated a clear superadditive effect in cell survival curves, suggesting inconclusive evidence of specific ECT-induced radiosensitization. Toxicity data were limited. In conclusion, the combination of ECT and RT consistently improved tumor response compared to that with individual therapies, supporting the potential benefit of their combination. However, evidence for a specific ECT-induced radiosensitization effect is currently lacking. Additional investigations are necessary to elucidate the potential benefits of this combination therapy.
Collapse
Affiliation(s)
- Martina Ferioli
- Radiation Oncology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.M.P.); (M.B.); (A.A.); (M.C.M.); (P.D.I.); (S.C.); (A.G.M.)
| | - Anna M. Perrone
- Radiation Oncology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.M.P.); (M.B.); (A.A.); (M.C.M.); (P.D.I.); (S.C.); (A.G.M.)
- Division of Oncologic Gynaecology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Milly Buwenge
- Radiation Oncology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.M.P.); (M.B.); (A.A.); (M.C.M.); (P.D.I.); (S.C.); (A.G.M.)
| | - Alessandra Arcelli
- Radiation Oncology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.M.P.); (M.B.); (A.A.); (M.C.M.); (P.D.I.); (S.C.); (A.G.M.)
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Maria Vadala’
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Bruno Fionda
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, UOC di Radioterapia Oncologica, 00168 Roma, Italy; (B.F.); (L.T.)
| | - Maria C. Malato
- Radiation Oncology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.M.P.); (M.B.); (A.A.); (M.C.M.); (P.D.I.); (S.C.); (A.G.M.)
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Pierandrea De Iaco
- Radiation Oncology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.M.P.); (M.B.); (A.A.); (M.C.M.); (P.D.I.); (S.C.); (A.G.M.)
- Division of Oncologic Gynaecology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Claudio Zamagni
- Oncologia Medica Addarii, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Silvia Cammelli
- Radiation Oncology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.M.P.); (M.B.); (A.A.); (M.C.M.); (P.D.I.); (S.C.); (A.G.M.)
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Luca Tagliaferri
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, UOC di Radioterapia Oncologica, 00168 Roma, Italy; (B.F.); (L.T.)
| | - Alessio G. Morganti
- Radiation Oncology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.M.P.); (M.B.); (A.A.); (M.C.M.); (P.D.I.); (S.C.); (A.G.M.)
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
19
|
Shestovskaya MV, Luss AL, Bezborodova OA, Makarov VV, Keskinov AA. Iron Oxide Nanoparticles in Cancer Treatment: Cell Responses and the Potency to Improve Radiosensitivity. Pharmaceutics 2023; 15:2406. [PMID: 37896166 PMCID: PMC10610190 DOI: 10.3390/pharmaceutics15102406] [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: 08/11/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
The main concept of radiosensitization is making the tumor tissue more responsive to ionizing radiation, which leads to an increase in the potency of radiation therapy and allows for decreasing radiation dose and the concomitant side effects. Radiosensitization by metal oxide nanoparticles is widely discussed, but the range of mechanisms studied is not sufficiently codified and often does not reflect the ability of nanocarriers to have a specific impact on cells. This review is focused on the magnetic iron oxide nanoparticles while they occupied a special niche among the prospective radiosensitizers due to unique physicochemical characteristics and reactivity. We collected data about the possible molecular mechanisms underlying the radiosensitizing effects of iron oxide nanoparticles (IONPs) and the main approaches to increase their therapeutic efficacy by variable modifications.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| | - Anna L. Luss
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
- The Department of Technology of Chemical, Pharmaceutical and Cosmetic Products Mendeleev of University of Chemical Technology of Russia, Miusskaya sq. 9, Moscow 125047, Russia
| | - Olga A. Bezborodova
- P. Hertsen Moscow Oncology Research Institute of the National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinskiy p. 3, Moscow 125284, Russia;
| | - Valentin V. Makarov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| | - Anton A. Keskinov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| |
Collapse
|
20
|
Sedky NK, Braoudaki M, Mahdy NK, Amin K, Fawzy IM, Efthimiadou EK, Youness RA, Fahmy SA. Box-Behnken design of thermo-responsive nano-liposomes loaded with a platinum(iv) anticancer complex: evaluation of cytotoxicity and apoptotic pathways in triple negative breast cancer cells. NANOSCALE ADVANCES 2023; 5:5399-5413. [PMID: 37767043 PMCID: PMC10521260 DOI: 10.1039/d3na00368j] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
Herein, thermo-responsive liposomes (TLs) loaded with Asp (Asp/TLs) were produced by self-assembling DPPC, DSPE-PEG2000, and cholesterol. The preparation variables were optimized using the Box-Behnken design (BBD). The optimized Asp/TLs exhibited an average particle size of 114.05 ± 1.56 nm, PDI of 0.15 ± 0.015, zeta potential of -15.24 ± 0.65 mV, and entrapment efficiency (EE%) of 84.08 ± 2.75%. In addition, under physiological conditions, Asp/TLs showed spherical shape, outstanding stability and thermo-triggered the release of Asp at 38 °C, reaching the maximum Asp release at 40 °C. The MTT assay showed that the optimal Asp/TLs exhibited the highest cytotoxic activity upon exposure to mild hyperthermia (40 °C) against the invasive triple-negative breast cancer cell line (MDA-MB-231) when compared to other preparations. The IC50 of Asp/TLs (40 °C) was estimated at 0.9 μg mL-1, while that of free Asp (40 °C) was 3.83 μg mL-1. As such, the optimal Asp/TLs were shown to increase the cytotoxic activity of Asp by 4-fold upon exposure to mild hyperthermia. The IC50 values of Asp and Asp/TLs without exposure to 40 °C were 6.6 μg mL-1 and 186 μg mL-1, respectively. This indicated that Asp was released only when placed at 40 °C. The apoptosis assay revealed that Asp/TLs (40 °C) caused a remarkable increase in the percentage of cell population among both the late apoptosis and necrosis quartiles, as well as a significant decline in the viable cell quartile (P ≤ 0.001) when compared to Asp (40 °C). Asp/TLs (40 °C) and Asp (40 °C) could stimulate the intrinsic apoptosis pathway by upregulating the apoptotic genes Bak and Bax, while downregulating the anti-apoptotic genes, BCL-xL and BCL-2. The free Asp (40 °C) increased the gene expression of Bak and Bax by 4.4- and 5.2-folds, while reducing the expression of BCL-xL and BCL-2 by 50% and 73%, respectively. The optimal Asp TLs (40 °C) manifested more potent effects as demonstrated by the upregulation of Bak, Bax, and P53 by 5.6-, 7.2-, and 1.3-folds, as well as the downregulation of BCL-xL and BCL-2 by 70% and 85%, respectively. As such, the optimal Asp TLs (40 °C) treatment displayed the most potent cytotoxic profile and induced both apoptosis and necrosis in MDA-MB-231.
Collapse
Affiliation(s)
- Nada K Sedky
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Administrative Capital Cairo Egypt
| | - Maria Braoudaki
- Department of Clinical, Pharmaceutical, and Biological Science, School of Life and Medical Sciences, University of Hertfordshire Hatfield AL10 9AB UK
| | - Noha Khalil Mahdy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University Kasr El-Aini Street 11562 Cairo Egypt
| | - Kenzy Amin
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Capital Cairo 11835 Egypt +20-1222613344
| | - Iten M Fawzy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Future University in Egypt 11835 Cairo Egypt
| | - Eleni K Efthimiadou
- Inorganic Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens Panepistimiopolis Zografou 157 71 Greece
| | - Rana A Youness
- Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU) New Administrative Capital Cairo Egypt
- Department of Biology and Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Administrative Capital Cairo Egypt
| | - Sherif Ashraf Fahmy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Capital Cairo 11835 Egypt +20-1222613344
| |
Collapse
|
21
|
Dhungel L, Harris C, Romine L, Sarkaria J, Raucher D. Targeted c-Myc Inhibition and Systemic Temozolomide Therapy Extend Survival in Glioblastoma Xenografts. Bioengineering (Basel) 2023; 10:718. [PMID: 37370649 DOI: 10.3390/bioengineering10060718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/26/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023] Open
Abstract
Glioblastoma is a highly aggressive disease with poor patient outcomes despite current treatment options, which consist of surgery, radiation, and chemotherapy. However, these strategies present challenges such as resistance development, damage to healthy tissue, and complications due to the blood-brain barrier. There is therefore a critical need for new treatment modalities that can selectively target tumor cells, minimize resistance development, and improve patient survival. Temozolomide is the current standard chemotherapeutic agent for glioblastoma, yet its use is hindered by drug resistance and severe side effects. Combination therapy using multiple drugs acting synergistically to kill cancer cells and with multiple targets can provide increased efficacy at lower drug concentrations and reduce side effects. In our previous work, we designed a therapeutic peptide (Bac-ELP1-H1) targeting the c-myc oncogene and demonstrated its ability to reduce tumor size, delay neurological deficits, and improve survival in a rat glioblastoma model. In this study, we expanded our research to the U87 glioblastoma cell line and investigated the efficacy of Bac-ELP1-H1/hyperthermia treatment, as well as the combination treatment of temozolomide and Bac-ELP1-H1, in suppressing tumor growth and extending survival in athymic mice. Our experiments revealed that the combination treatment of Bac-ELP1-H1 and temozolomide acted synergistically to enhance survival in mice and was more effective in reducing tumor progression than the single components. Additionally, our study demonstrated the effectiveness of hyperthermia in facilitating the accumulation of the Bac-ELP1-H1 protein at the tumor site. Our findings suggest that the combination of targeted c-myc inhibitory biopolymer with systemic temozolomide therapy may represent a promising alternative treatment option for glioblastoma patients.
Collapse
Affiliation(s)
- Laxmi Dhungel
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| | - Cayla Harris
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| | - Lauren Romine
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| | - Jan Sarkaria
- Division of Radiation Oncology, Mayo Clinic and Foundation, 200 First Street, SW, Rochester, MN 55905, USA
| | - Drazen Raucher
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| |
Collapse
|
22
|
Obrador E, Montoro A. Ionizing Radiation, Antioxidant Response and Oxidative Damage: Radiomodulators. Antioxidants (Basel) 2023; 12:1219. [PMID: 37371949 DOI: 10.3390/antiox12061219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Ionizing radiation (IR) is the energy released by atoms in the form of electromagnetic waves (e [...].
Collapse
Affiliation(s)
- Elena Obrador
- Elena Obrador Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
| | - Alegría Montoro
- Alegría Montoro, Radiation Protection Service, University and Polytechnic Hospital La Fe, 46021 Valencia, Spain
| |
Collapse
|
23
|
Zhu Y, Yang D, Guo T, Lin M. Use of S2.2/DOX Magnetic Nanoliposomes in MR Molecule Imaging and Targeted Thermochemotherapy for Breast Cancer In Vitro. Technol Cancer Res Treat 2023; 22:15330338231194498. [PMID: 37563954 PMCID: PMC10422896 DOI: 10.1177/15330338231194498] [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] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE To prepare S2.2/DOX magnetic nanoliposomes by combining the potential benefits of MNPs in MRI and the targeted performance of nano-drugs as an innovative method for integrated diagnosis and treatment of breast cancer (BC). METHODS We created a S2.2-PEG-MZF/DOX molecular probe by using a lipid material to encapsulate PEG-MZF-NPs and doxorubicin (DOX), and a S2.2 aptamer to target MUC1 to conjugate with PEG-MZF/DOX nanoliposomes. The potential of probe for cell-specific targeting and magnetic resonance (MR) molecular imaging was evaluated by MR scanner and Prussian blue staining. Additionally, we explored the feasibility by using nanoliposome magnetic induction heating to interfere with MCF-7 (MUC1+) BC cells under the influence of an alternating magnetic field (AMF). RESULTS PEG-MZF-NPs were biologically safe. The T2 relaxation rate of PEG-MZF-NPs was found to inhibit T2 signal in a concentration-dependent manner, and the T2 signal of the S2.2-PEG-MZF molecular probe in MCF-7 cells was significantly lower than that in PEG-MZF-NPs group. Moreover, the T2 signal reduction was more pronounced in MCF-7 cells than in the hepatoma cell line HepG2 (MUC1-), suggesting a strong MRI potential of the S2.2-PEG-MZF molecular probe. The S2.2-PEG-MZF/DOX nanoliposome was able to achieve the desired temperature range for tumor hyperthermia (42-44 °C) in vitro. The S2.2-PEG-MZF/DOX nanoliposome accompanied by magnetic fluid hyperthermia (MFH) could inhibit proliferation and invasion and induce apoptosis of MCF-7 cells. The effects of this approach were significantly higher than those observed in the other groups. CONCLUSION We successfully developed a novel technique for BC diagnosis and treatment using thermochemotherapy under the guidance of MR molecular imaging. This approach holds great potential for improving the management of this devastating disease in the future.
Collapse
Affiliation(s)
- Yinxing Zhu
- Taizhou School of Clinical Medicine, Nanjing Medical University, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Dazhuang Yang
- Imaging Department, General Hospital of Xuzhou Mining Group, Xuzhou, Jiangsu, China
| | - Ting Guo
- Institute of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Mei Lin
- Clinical Laboratory, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| |
Collapse
|