scispace - formally typeset
Search or ask a question
Topic

Hyperthermia therapy

About: Hyperthermia therapy is a research topic. Over the lifetime, 404 publications have been published within this topic receiving 7842 citations. The topic is also known as: induced hyperthermia & hyperthermia, induced.


Papers
More filters
Journal ArticleDOI
TL;DR: The main feature of this review is to present the recent advances in the development of multifunctional therapeutic nanosystems incorporating both magnetic nanoparticles and drugs, and their superior efficacy in treating cancer compared to either hyperthermia or chemotherapy as standalone therapies.
Abstract: Magnetic nanoparticles have been widely investigated for their great potential as mediators of heat for localised hyperthermia therapy. Nanocarriers have also attracted increasing attention due to the possibility of delivering drugs at specific locations, therefore limiting systematic effects. The enhancement of the anti-cancer effect of chemotherapy with application of concurrent hyperthermia was noticed more than thirty years ago. However, combining magnetic nanoparticles with molecules of drugs in the same nanoformulation has only recently emerged as a promising tool for the application of hyperthermia with combined chemotherapy in the treatment of cancer. The main feature of this review is to present the recent advances in the development of multifunctional therapeutic nanosystems incorporating both magnetic nanoparticles and drugs, and their superior efficacy in treating cancer compared to either hyperthermia or chemotherapy as standalone therapies. The principle of magnetic fluid hyperthermia is also presented.

461 citations

Journal ArticleDOI
01 Jan 2017-Small
TL;DR: The results indicate that compared with chemotherapy, magnetichyperthermia or photothermal therapy alone, the combined chemo-magnetic hyperthermia therapy or chemo -photothermal therapy with the DOX-loaded MMSN/GQDs nanosystem exhibits a significant synergistic effect, resulting in a higher efficacy to kill cancer cells.
Abstract: A multifunctional platform is reported for synergistic therapy with controlled drug release, magnetic hyperthermia, and photothermal therapy, which is composed of graphene quantum dots (GQDs) as caps and local photothermal generators and magnetic mesoporous silica nanoparticles (MMSN) as drug carriers and magnetic thermoseeds. The structure, drug release behavior, magnetic hyperthermia capacity, photothermal effect, and synergistic therapeutic efficiency of the MMSN/GQDs nanoparticles are investigated. The results show that monodisperse MMSN/GQDs nanoparticles with the particle size of 100 nm can load doxorubicin (DOX) and trigger DOX release by low pH environment. Furthermore, the MMSN/GQDs nanoparticles can efficiently generate heat to the hyperthermia temperature under an alternating magnetic field or by near infrared irradiation. More importantly, breast cancer 4T1 cells as a model cellular system, the results indicate that compared with chemotherapy, magnetic hyperthermia or photothermal therapy alone, the combined chemo-magnetic hyperthermia therapy or chemo-photothermal therapy with the DOX-loaded MMSN/GQDs nanosystem exhibits a significant synergistic effect, resulting in a higher efficacy to kill cancer cells. Therefore, the MMSN/GQDs multifunctional platform has great potential in cancer therapy for enhancing the therapeutic efficiency.

364 citations

Journal Article
TL;DR: Radio-frequency hyperthermia appears to be a safe and potentially useful form of therapy for selected cancer patients, and while other cancer treatments are more effective for small tumors,hyperthermia may be uniquely beneficial against larger lesions.
Abstract: Localized hyperthermia therapy by high-energy radio-frequency waves was evaluated in malignant and adjacent normal tissue of 30 patients with 10 types of cancer Hyperthermia was delivered to superficial and deep visceral cancers in awake patients who had refractory disease Histological and clinical responses were recorded serially Toxicity tests in dogs, sheep, and pigs showed that progressive necrosis of normal and cancer tissue occurred at temperatures above 45 degrees C (113 degrees F) However, as normal tissues approached this temperature, intrinsic heat dissipation occurred (possibly due to augmented blood flow) so that temperatures below 45 degrees C could be maintained, whereas most solid tumors did not have this adaptive capacity and could be heated to 50 degrees C (122 degrees F) with virtually no injury to normal organs, sc tissue, or skin To date, 69 treatments have been administered to 36 tumors in the 30 patients Selective heating was observed in both primary and metastatic tumors located in surface tissues and internal organs Response appeared to be related to tumor size in that differential heating was possible more often in the larger lesions In tumors successfully heated, moderate to marked necrosis occurred Radio-frequency hyperthermia appears to be a safe and potentially useful form of therapy for selected cancer patients While other cancer treatments are more effective for small tumors, hyperthermia may be uniquely beneficial against larger lesions

274 citations

Journal ArticleDOI
TL;DR: Different mechanisms by which heating tumours can elicit anti-tumour immune responses, including tumour cell damage, tumour surface molecule changes, heat shock proteins, exosomes, direct effects on immune cells, and changes in the tumour vasculature are described.
Abstract: Local tumour hyperthermia for cancer treatment is currently used either for ablation purposes as an alternative to surgery or less frequently, in combination with chemotherapy and/or radiation therapy to enhance the effects of those traditional therapies. As it has become apparent that activating the immune system is crucial to successfully treat metastatic cancer, the potential of boosting anti-tumour immunity by heating tumours has become a growing area of cancer research. After reviewing the history of hyperthermia therapy for cancer and introducing methods for inducing local hyperthermia, this review describes different mechanisms by which heating tumours can elicit anti-tumour immune responses, including tumour cell damage, tumour surface molecule changes, heat shock proteins, exosomes, direct effects on immune cells, and changes in the tumour vasculature. We then go over in vivo studies that provide promising results showing that local hyperthermia therapy indeed activates various systemic anti-tumour immune responses that slow growth of untreated tumours. Finally, future research questions that will help bring the use of local hyperthermia as systemic immunotherapy closer to clinical application are discussed.

235 citations

Journal ArticleDOI
TL;DR: In this review, the application of MHT as a therapeutic modality for GBM will be discussed, its therapeutic efficacy, technical details, and major experimental and clinical findings will be reviewed and analysed.
Abstract: Hyperthermia therapy (HT) is the exposure of a region of the body to elevated temperatures to achieve a therapeutic effect. HT anticancer properties and its potential as a cancer treatment have been studied for decades. Techniques used to achieve a localised hyperthermic effect include radiofrequency, ultrasound, microwave, laser and magnetic nanoparticles (MNPs). The use of MNPs for therapeutic hyperthermia generation is known as magnetic hyperthermia therapy (MHT) and was first attempted as a cancer therapy in 1957. However, despite more recent advancements, MHT has still not become part of the standard of care for cancer treatment. Certain challenges, such as accurate thermometry within the tumour mass and precise tumour heating, preclude its widespread application as a treatment modality for cancer. MHT is especially attractive for the treatment of glioblastoma (GBM), the most common and aggressive primary brain cancer in adults, which has no cure. In this review, the application of MHT as a therapeutic modality for GBM will be discussed. Its therapeutic efficacy, technical details, and major experimental and clinical findings will be reviewed and analysed. Finally, current limitations, areas of improvement, and future directions will be discussed in depth.

234 citations


Network Information
Related Topics (5)
Nanoparticle
85.9K papers, 2.6M citations
72% related
Cancer cell
93.4K papers, 3.5M citations
69% related
Mesenchymal stem cell
53.6K papers, 2M citations
68% related
In vivo
61.3K papers, 1.9M citations
67% related
Carbon nanotube
109K papers, 3.6M citations
67% related
Performance
Metrics
No. of papers in the topic in previous years
YearPapers
20234
20228
202130
202031
201930
201819