Abstract
Cancer stem-like cells (CSCs) are a subset of cancer cells that are resistant to conventional radiotherapy and chemotherapy. As such, CSCs have been recognized as playing a large role in tumor initiation and recurrence. Although hyperthermia is broadly used in cancer treatment either alone or in combination with radio- or chemo-therapy, its potential to target CSCs is not well understood. In this review, we discuss different types of hyperthermia and potential mechanisms of action in cancer treatment, particularly in regards to killing CSCs.
Keywords: Hyperthermia, Cancer stem-like cells, Chemotherapy, Cytotoxic treatment, Radiation, Nanoparticle, Ablation, LITT
Introduction
Despite advances in understanding the molecular changes underpinning cancer and improved technology and treatments, cancer remains a leading cause of death in America. The cancer stem-like cell (CSC) hypothesis posits that a subset of tumor cells have a high capacity for self-renewal, have the ability to differentiate into multiple lineages and can give rise to tumors [1–4]. These CSCs are highly malignant and can persist or proliferate in spite of cytotoxic treatment [1–4]. Therefore, these CSCs play a large role in tumor progression. Development of new treatment modalities that are able to target and kill CSCs may provide more durable cancer control [1–4].
Hyperthermia is a potent radiosensitizer that has been shown in numerous clinical trials to improve tumor control. Importantly, the efficacy of hyperthermia is seen across many cancer types, including breast cancer, prostate cancer, melanoma, sarcoma, rectal cancer, bladder cancer, esophageal cancer, cervical cancer and glioblastoma suggesting that it has broad clinical applicability [5–24]. Recently, combined hyperthermia and radiation has also been shown to improve pain palliation in patients with bone metastases compared to radiation alone [25]. Therefore, hyperthermia has widespread usage for patients with both locoregional disease and advanced cancers and can be used for patients with a variety of cancer types. The value of hyperthermia as a treatment has in fact been observed for centuries. Hippocrates, the father of modern medicine, is known to have said, “Those who cannot be cured by medicine can be cured by surgery. Those who cannot be cured by surgery can be cured by heat. Those who cannot be cured by heat, they are indeed incurable”. Over the years, medicine and surgery have seen significant advances, and hyperthermia fell by the wayside. However, in modern times, hyperthermia is making a resurgence due to improved technology in delivering hyperthermia and in non-invasive thermometry techniques.
Hyperthermia is classified into two broad categories based on the target heating temperature. Thermal ablation refers to treatments with target temperatures above 50°C and mild temperature hyperthermia refers to treatments with temperatures between 39 and 43°C [26]. While thermal ablation largely kills tumor cells due to the direct cytotoxic effects of heat, mild temperature hyperthermia uses heat as an adjunct treatment to enhance the cytotoxic effects of radiation and chemotherapy [26–28]. The biologic effects of thermal therapy are dependent on time and temperature. The mechanisms underlying the biologic effects are multi-factorial and impact the tumor population itself, the tumor microenvironment and immune system.
Methods for Administering Hyperthermia
Radio-frequency hyperthermia is the most widely used hyperthermia technique worldwide and is typically used for ablative heating [28–30]. To achieve heating, radio-frequency electrodes are passed into the tumor tissue under image guidance. A high-frequency alternating current is then passed through the electrodes to cause the rapid oscillation of ions in nearby cells, resulting in frictional heating [27,31]. The range of heating is limited to the millimeter range because it relies on heated tissue to conduct current to surrounding areas [32]. The short range of heating also limits the ability to heat tumors near blood vessels because the heat is dissipated too quickly [32,33].
Microwave hyperthermia is an alternate method of delivery that can overcome some of the limitations of radio-frequency hyperthermia. Microwave heating uses waves of higher frequency to kill cells. Unlike radio-frequency thermal therapy, microwave hyperthermia does not pass an electrical current through tissue, but rather creates an oscillating electromagnetic field that forces ions and dipoles to align with the field, causing them to rotate as the field oscillates [31,32,34]. This rotation causes friction that heats the tissue. Microwave hyperthermia presents several advantages compared to radio-frequency hyperthermia. While radio-frequency hyperthermia relies on ions inside tissue to conduct current, microwave hyperthermia creates an electric field, the effective range of which is larger without risking damage to tissue closer to the antenna or probe [32]. Microwave hyperthermia has a much higher effective range of up to 3 cm [32].
Laser interstitial thermal therapy (LITT) is a relatively new method of administering hyperthermia that uses a stereotactically placed laser probe to heat surrounding tissue with a low power (10–15 Watts) infrared laser (at Nd-YAG range) [35,36]. Heat essentially is produced after absorption of laser in the tissue and transferred up to 1.5–2 cm from the laser probe by conduction. To control the extent of thermal ablation, a specific sequence of MRI (MR-thermometry) is used to measure relative changes of temperature within the magnetic field. For deep seated lesions, including brain tumors, LITT is used in conjunction with MR-thermometry to give accurate thermal ablation of the target lesion [35,36]. The minimally invasive nature of LITT under MR-thermometry guidance has permitted the expansion of hyperthermia to deep and difficult to access tumors including intracranial and retroperitoneal tumors [35,36].
High intensity focused ultrasound (HIFU) (also called focused ultrasound surgery (FUS)) utilizes an ultrasound beam with very high energy to increase the temperature rapidly in the target tissues [37–39]. A single HIFU exposure usually treats a very small volume along the ultrasound axis. Multiple exposures can be used side by side to achieve coverage of a large volume of tumor [37–39]. One advantage of HIFU is that it creates a steep temperature gradient in a small focused area and effectively creates a sharp boundary of damage in the target tissue while sparing adjacent normal tissues [37–39].
Nanoparticles can also be used to augment heating within a tissue when exposed to electromagnetic energy [40–42]. These particles include magnetic nanoparticles (such as iron oxide), gold-silica nanoshells, solid gold nanoparticles and carbon nanotubes [40,41]. The outer shell of nanoparticles can be modified molecularly to facilitate their dissemination and uptake by specific cell types, including tumor cells [42]. Additionally, nanoparticles may be loaded with cargo including cytotoxic drugs or oncolytic viruses that can be released upon disruption by a heat source [43]. Nanoparticles can be administered systemically to exploit the leaky vasculature of primary tumors to enhance intra-tumoral delivery [44]. However, nanoparticles often display a patchy, near perivascular deposition within the well-vascularized regions of tumors [45]. Some blood vessels such as those associated with brain tumors are not as leaky as blood vessels found in other solid tumors. While the blood-brain barrier is partially breached in regions with glioma cells, the ‘compromised’ blood-brain barrier still presents a major challenge, especially in hypoxic and avascular regions of glioma dispersion [46]. Since high spatial concentrations of nanoparticles are required for hyperthermia, direct intratumoral delivery of 12 nm magnetic nanoparticles has been used in clinical trials for hyperthermia treatment of prostate tumors and recurrent glioblastoma [47,48]. When subjected to an external alternating magnetic field, the nanoparticles vibrate and heat up to kill surrounding cells. Because non-ionizing electromagnetic radiation can be applied remotely to heat the nanoparticles, this technology is considered noninvasive but requires good visualization of the target tumor [42,49]. A typical drawback of the application of iron oxide nanoparticles is associated with the indefinite exclusion of MRI for subsequent monitoring of tumor progression after initial injection of nanoparticles and the residual MR signals that interfere with follow-up MR imaging.
Hyperthermia in Cancer Therapy
When hyperthermia is applied to a tumor, three different reaction zones can be distinguished based on the temperature and duration of heating: a central zone that is directly and immediately beyond the application site, a peripheral zone that is around the central zone and is heated to a lower temperature, and an outer region which is not directly affected by the heat [28,42,50]. Hyperthermia causes cellular injury directly and indirectly in these different zones via different mechanisms, although some overlap may exist. The extent and type of cellular damage varies as a function of temperature and time. A high temperature for a short period of time can achieve similar levels of cell kill as lower temperature heating for a longer period of time.
Direct Effects to Tumor Cells
Hyperthermia causes membrane dysfunction to contribute to cell death. Rising temperature affects the stability fluidity and permeability of cellular membranes, including the plasma membrane, mitochondrial membranes, and other cytosolic membranes [50,51]. These membrane changes can compromise the function of transmembrane transport proteins, ion channels, cell surface receptors and other membrane-associated proteins and disrupt lipid rafts or signal transduction hubs [50,51]. However, the degree of membrane dysfunction strongly influences cell fate. For example, some changes in membrane potential, intracellular sodium and calcium content do not correlate well with the rate of cell death [52–54]. On the other hand, mitochondrial dysfunction induced by hyperthermia can lead to cell death [50,55].
Another direct effect of heat is the denaturation of proteins, especially under high temperatures. Denaturation and inactivation of these proteins can impact a broad range of cellular processes including cellular metabolism, protein synthesis, nucleic acid synthesis and DNA/RNA polymerization [50,56]. After mild hyperthermia, some cellular functions can recover. Proteins may refold, and RNA and protein synthesis may recover. However, DNA replication and repair typically remain repressed [57]. This is thought to be due to the aggregation of denatured proteins in the nuclear matrix and irreversible changes to chromatin structure that impair DNA synthesis and repair [57]. Hyperthermia can inhibit the function of DNA-polymerases-α and -β and can also facilitate degradation of the DNA repair protein BRCA2 to inhibit homologous recombination [57,58]. Hyperthermia itself is believed not to cause severe DNA damage, but rather indirectly contributes to DNA damage by reducing the efficiency of the DNA damage repair machinery [59].
Hyperthermia can induce cell death by necrosis and apoptosis. The cells in central application zones, which are confronted with high temperatures, usually die by necrosis. However, some subpopulations of cells may escape immediate hyperthermic killing. These resistant cells in the central zone and the cells in the peripheral zone, which receive lower temperature hyperthermia, may die within hours of heat cessation [50,51]. Mild temperature hyperthermia can induce apoptosis through both the intrinsic and extrinsic pathways. Hyperthermia can activate Caspase-2 which then binds to the adaptor protein RAIDD to cleave and activate Bim, which promotes mitochondria-dependent apoptosis [60]. Hyperthermia can also activate Bim to induce Caspase-2 independent apoptosis. In addition, hyperthermia can activate Fas, tumor necrosis factor α (TNF-α) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to trigger the extrinsic apoptotic pathway [61,62]. Additionally, heat treatment can increase the production of reactive oxygen species (ROS) by activation of xanthine oxidase and/or facilitating mitochondria respiration to produce O2−. Moreover, hyperthermia negatively affects SOD1 expression and the enzymatic activities of SOD1 and SOD2, whereas it is able to activate NADPH oxidase [63–65]. This increase in ROS can facilitate apoptosis [66,67].
Alterations to the Tumor Microenvironment
Hyperthermia can also modify the tumor microenvironment to modulate tumor growth and recurrence. Hyperthermia is well known to increase perfusion within tissue and compromise the integrity of blood vessels [33,68–70]. Hyperthermia can damage endothelial cells, alter the adhesiveness of the vessel wall, and increase the leakiness of blood vessels and viscosity of blood [68]. These changes in perfusion can influence local pH, oxygen and nutrient supply in the tumor, rendering them more stressed and more susceptible to cytotoxic therapy [71]. Hyperthermia can also improve tumor oxygenation, making cells more susceptible to radiation, and may improve the penetration of chemotherapy into the tumors [69,72].
Local hyperthermia can also affect the immune system and augment the anti-tumor immune response. With increased perfusion, hyperthermia can increase immune cell penetration into the tumor [73,74]. Hyperthermia-mediated tumor cell death releases the intracellular contents of tumors including RNA, DNA, metabolites and proteins that are immunogenic [75]. The tumor- specific molecules are taken up by dendritic cells and macrophages and trigger specific anti-tumor innate and adapted immune responses. Hyperthermia also promotes the production and release of pro-inflammatory cytokines from tumor cells and increases serum levels of interleukin-1β (IL-1β), IL-6, IL-8 and tumor necrosis factor-α (TNFα) [76–78]. Induction of heat shock proteins (HSPs) can also enhance anti-tumor immunity. Extracellular HSPs function as chaperones to bind antigens released by dying tumor cells. The protein complexes are recognized and internalized by antigen-presenting cells (APCs) [79–81]. HSPs also activate NK cells, T-lymphocyte and macrophages to provide a multi-pronged anti-tumor response [80–82].
Thermotolerance
When tumor cells are exposed to sublethal thermal treatment, they can develop tolerance to sequential thermal stress. This phenomenon is called thermotolerance [83]. Numerous biochemical and molecular changes are involved in this process. Thermotolerance may be due to the accumulation of heat shock proteins (HSPs), which are preferentially upregulated by transcriptional and translational mechanisms upon hyperthermic stress [83]. Hsp90, Hsp70, Hsp60, and Hsp27 families play a critical role in thermotolerance by maintaining protein structure, preventing protein aggregation and facilitating re-folding of proteins under heat stress [83,84]. HSPs also regulate apoptosis through caspase dependent and independent pathways [85]. Hsp27 and Hsp70 can also inhibit the translocation of pro-apoptotic factor Bid to mitochondria. Hsp70 can inhibit the cleavage and activation of Bid to repress mitochondria-mediated apoptosis [86,87]. HSPs can also suppress Fas, TNF and TRAIL-mediated apoptosis through binding to and inhibiting Daxx and ASK-1 or modulating IKK complex stability and activity. Hyperthermia can also affect other signaling pathways, including Integrin-linked kinase, JNK and p38 MAPK kinase activities that contribute to thermotolerance [86,87]. While thermotolerance is seen in vitro, its impact on clinical efficacy of hyperthermia is controversial.
Side Effects of Hyperthermia
In addition to thermotolerance, another factor to be considered during hyperthermia is its potential side effects. These adverse effects are mainly due to normal cell damage and are dependent on the area being treated. In whole-body hyperthermia, which has been used to treat metastatic cancers with chemotherapy in clinical trials, whole body hyperthermia can cause side effects including diarrhea, nausea, vomiting and more rarely results in cardiac and vascular dysfunction [6,88]. In regional and local hyperthermia, normal cells in the proximity of the targeted cancer cells can be damaged by heat. Limitations in directing heat to the tumor volume or suboptimal thermometry in the heated area can cause local areas of heat deposition that can damage normal tissue [6,88,89]. These hyperthermia-associated side effects include tissue swelling, erythema, blistering of skin, or burning pain in the heated volume, in the case of superficial hyperthermia. Most of these side effects are reversible and heal after conservative measures [6,9,88,89]. Side effects of hyperthermia for deeper tissue heating are dependent on the organ being treated. For example, patients undergoing hyperthermia treatment for locally advanced prostate cancer may develop mild urinary symptoms due to swelling of the prostate gland and mild proctitis [19,20]. This can be mitigated with a short course of steroids. Deep tissue hyperthermia can also increase sweating and vasodilation, causing a drop in blood pressure and contributing to postural hypotension [90]. The utilization of specific hyperthermia methods may also minimize side effects. For example, HIFU causes a very steep temperature gradient and damages tumor cells in a small area around the probe. Cancer biomarker-based nanoparticles selectively deliver heat to cancer cells. Overall, hyperthermia is relatively safe when administered properly, and as seen in many clinical trials, offers clinical tumor control benefits that exceed the side effects of hyperthermia [6].
The Effects of Hyperthermia on Cancer Stem-Like Cells
Cancer stem-like cells (CSCs) are a rare population of cancer cells that can self-renew and differentiate into progeny with limited proliferative potential. CSCs sit at the apex of hierarchically organized tumors. CSCs have strong tumorigenic activity compared to non-stem cancer cells and can establish an entire tumor [1,3,56]. CSCs usually reside in specific niches that orchestrate their fate. Niche components that support the undifferentiated state of CSCs include communication with contacting cells such as other stromal cells and endothelial cells, extracellular matrix components, soluble factors including Wnt, TGFβ and other cytokines, and physical states such as hypoxia and low pH [91,92].
CSCs are resistant to ionizing radiation and chemotherapy. CSCs preferentially activate the DNA damage checkpoint in response to radiation and repair DNA damage more efficiently than non-CSC cancer cells [93]. Inhibition of DNA damage checkpoint kinases Chk1 and Chk2 sensitizes CSCs to radiation [93]. CSCs can also overexpress ROS scavenger proteins to limit the amount of DNA damage after radiotherapy, which is largely mediated by ROS. Pharmacological depletion of ROS scavengers can improve the radiosensitivity of CSCs [94].
CSCs can develop resistance to chemotherapy via different mechanisms, including enhancing efflux of chemotherapy from tumor cells or interfering with chemotherapy metabolism. CSCs can overexpress ABC transporters to pump out chemotherapy [95,96]. CSCs can express enzymes to inactivate drugs, for example, aldehyde dehydrogenase (ALDH) which inactivates cyclophosphamide, temozolomide and doxorubicin [97]. CSCs can also epigenetically silence the enzymes that are needed to catalyze prodrugs into their active forms [97]. Additionally, CSCs can augment pro-survival signaling pathways, including upregulating BCL2 family proteins and activating Notch and Wnt pathways, to protect themselves from the cytotoxic effects of chemotherapy [98-101]. Some CSCs can also exit the cell cycle and remain in a quiescent state, reducing their sensitivity to chemotherapy and radiation, which preferentially targets rapidly proliferating cells [102,103].
The CSC niche also plays a major role in therapeutic resistance. The hypoxic regions where many CSCs reside are devoid of functional blood vessels and chemotherapy is unable to penetrate to these areas in appreciable concentrations. Furthermore, low ROS levels in these areas render CSCs more resistant to free radical damage induced by radiation and some chemotherapies [104,105]. Other components of the CSC niche including the extracellular matrix, cancer-associated fibroblasts and immune cells may provide signaling stimuli including hepatocyte growth factor (HGF), interleukin 6 (IL-6), fibroblast growth factor (FGF), neuregulin 1 to further facilitate therapeutic resistance [106,107].
Based on its effects on CSCs and their microenvironment, hyperthermia may have the potential to eliminate cancer stem cells and sensitize them to radiotherapy and chemotherapy. First, hyperthermia can denature proteins that pump out or inactivate chemotherapy. Second, hyperthermia can impede DNA damage repair pathways and thus sensitize CSCs to radiation and some types of chemotherapy. Third, hyperthermia can kill tumor cells independent of their cell cycle status, permitting killing of quiescent CSCs. Fourth, heat treatment can increase perfusion inside the tumor and so reduce hypoxia and alter ROS levels that support CSCs. Alternatively, hyperthermia can also cause collapse of blood vessels and damage endothelial cells to reduce blood flow to create local pockets of hypoxia. Therefore, hyperthermia can have complex effects within the tumor and in particular, CSCs within the perivascular or hypoxic niches. In addition, hyperthermia can enhance the anti-tumor immune response to target CSCs. This can be mediated in part by increasing cell kill, exposing CSC-specific antigens to APCs and improving immune cell recruitment to the tumor.
Although hyperthermia is broadly used in cancer treatment alone or in combination with surgery, radiotherapy and chemotherapy, whether and how hyperthermia can help to eliminate chemo- and radio-resistant CSCs has not been extensively studied. Some studies suggest that CSCs may be more sensitive to hyperthermia than normal stem/progenitor cells [108]. Wierenga, et al. compared the sensitivity of normal and acute myeloid leukemia (AML) stem cells to mild temperature hyperthermia (43°C). While heat treatment had only a mild effect on normal stem and progenitor cells, it greatly reduced AML stem cell survival and leukemia initiation ability [108]. This suggests that hyperthermia can be used therapeutically to purge leukemic stem cells with minimal adverse effects on normal stem/progenitor cells.
However, in some other tumors, hyperthermia alone may have little effect or even negative effects on CSCs in terms of cell kill. In a model of breast cancer, breast CSCs can scan survive better than bulk breast cancer cells to water bath hyperthermia, and the CD44high/CD24low stem cell fraction in bulk breast cancer cells was increased after hyperthermia [109]. This increased survival of breast CSCs was attributed to overexpression of Hsp90. Interestingly, the addition of an Hsp90 inhibitor sensitized breast CSCs but not bulk cancer cells to hyperthermia, suggesting that under thermal stress, breast CSCs are dependent on Hsp90 for survival and that combining Hsp90 inhibition with hyperthermia can target the breast CSC population [109].
However, hyperthermia mediated by nanoparticles may overcome CSC resistance to conventional heat treatment. Burke, et al. showed significantly reduced tumor size and prolonged mouse survival when treated with multi-walled carbon nanotubes, which were heated by near-infrared radiation [109]. Breast CSCs, as well as bulk tumor cells, died from necrosis in their study. In another report, superparamagnetic iron oxide nanoparticles have been used to induce magnetic hyperthermia in A549 lung cancer and MDA- MB-231 breast cancer cell lines [110]. This magnetic hyperthermia compromised stem-cell properties, including reduced side population phenotype, mammosphere formation ability, ALDH activity and tumor initiation ability [110]. In contrast, conventional water bath thermal treatment was less efficacious [110]. Notably, magnetic nanoparticle based hyperthermia promoted higher ROS generation for a prolonged time compared to conventional hyperthermia [110], suggesting the method of hyperthermia delivery can influence the extent of cell death.
Hyperthermia has also been combined with radiotherapy, chemotherapy or antibody-based targeting technology to improve CSC elimination. Atkinson, et al. reported that optically activated gold nanoshell-based hyperthermia sensitized breast cancer stem cells to radiation and reduced the percentage of Lin−CD29HCD24H CSC subpopulation, which are resistant to radiation [111]. Hyperthermia reduced DNA damage repair in breast CSCs, rendering them vulnerable to radiation-induced DNA damage [111]. Similarly, in glioblastoma, hyperthermia alone had no appreciable effect on the survival or proliferation of glioma CSCs [112]. However, when combined with radiation, hyperthermia preferentially reduced glioma CSC self-renewal, viability and proliferation. This was mediated by suppression of radiation-induced AKT signaling and impaired DNA damage response. Combined hyperthermia, delivered by a microwave applicator, and radiation reduced tumor growth and extended mouse survival in a CSC-derived mouse model of glioblastoma [112].
Hyperthermia can also induce metabolic stress and enhance the effects of chemotherapy. This strategy has been used to sensitize CSCs to metformin, a diabetes drug that also has anticancer activity [113]. Thermal therapy can activate AMPK and suppress the mTOR/S6K pathway to enhance the cytotoxic effects of metformin [113]. Wierenga, et al. also reported that hyperthermia can improve the leukemic stem cell killing mediated by ET-18-OCH3, an anti-tumor lipid chemotherapy that is toxic to leukemic cells in a temperature-dependent manner [108].
The combination of hyperthermia treatment with CSC targeting technology shows promise. Single-walled carbon nanotubes (SWNTs) conjugated with CD133 monoclonal antibody (anti-CD133) were used to target glioma stem cells, which frequently express the cell surface marker CD133 [114]. Upon nanotube internalization and heating with a near infrared laser, CD133+ but not CD133− cells were killed. Additionally, heating significantly suppressed the tumor initiation ability of CD133+ cells [114]. These data suggest that hyperthermia combined with antibody-based strategies can be used to kill CSCs efficiently.
Conclusion
The use of hyperthermia to treat cancer has a long history. Hyperthermia has consistently improved the efficacy of radiotherapy and chemotherapy for many types of cancers. The CSC model sheds light on another potential therapeutic benefit of hyperthermia. Strategies that combine hyperthermia with cytotoxic agents, metabolic stressors or immune therapies may improve CSC kill by targeting the cancer cells themselves and modulating their microenvironment. The method of administering heat may also influence cell kill. More work is needed to define the optimal modes of hyperthermia to kill CSCs safely and efficiently. The combination of hyperthermia and immunotherapy to target CSCs also holds great potential, and further studies are needed to understand how best to integrate hyperthermia with immuno-oncology. Heat therapy was recognized for its therapeutic effects by ancient physicians. It is once again emerging as an important treatment modality that fights cancer through multiple mechanisms.
Acknowledgments
We thank Dr. Ping Xia for critical review of the manuscript. This work was supported by NIH/NINDS NS092641 and NS094199.
Footnotes
Conflict of Interest
Authors declare no conflict of interest.
References
- 1.Dalerba P, Cho RW, Clarke MF. Cancer stem cells: models and concepts. Annu Rev Med. 2007;58:267–84. doi: 10.1146/annurev.med.58.062105.204854. [DOI] [PubMed] [Google Scholar]
- 2.Batlle E, Clevers H. Cancer stem cells revisited. Nat Med. 2017;23(10):1124–1134. doi: 10.1038/nm.4409. [DOI] [PubMed] [Google Scholar]
- 3.Beck B, Blanpain C. Unravelling cancer stem cell potential. Nat Rev Cancer. 2013;13(10):727–38. doi: 10.1038/nrc3597. [DOI] [PubMed] [Google Scholar]
- 4.Nassar D, Blanpain C. Cancer Stem Cells: Basic Concepts and Therapeutic Implications. Annu Rev Pathol. 2016;11:47–76. doi: 10.1146/annurev-pathol-012615-044438. [DOI] [PubMed] [Google Scholar]
- 5.Mallory M, Gogineni E, Jones GC, Greer L, Simone CB. Therapeutic hyperthermia: The old, the new, and the upcoming. Crit Rev Oncol Hematol. 2016;97:56–64. doi: 10.1016/j.critrevonc.2015.08.003. [DOI] [PubMed] [Google Scholar]
- 6.Wust P, Hildebrandt B, Sreenivasa G, Rau B, Gellermann J, Riess H, et al. Hyperthermia in combined treatment of cancer. Lancet Oncol. 2002;3(8):487–97. doi: 10.1016/s1470-2045(02)00818-5. [DOI] [PubMed] [Google Scholar]
- 7.Van Valenberg H, Colombo R, Witjes F. Intravesical radiofrequency-induced hyperthermia combined with chemotherapy for non-muscle-invasive bladder cancer. Int J Hyperthermia. 2016;32(4):351–62. doi: 10.3109/02656736.2016.1140232. [DOI] [PubMed] [Google Scholar]
- 8.Hu Y, Li Z, Mi DH, Cao N, Zu SW, Wen ZZ, et al. Chemoradiation combined with regional hyperthermia for advanced oesophageal cancer: a systematic review and meta-analysis. J Clin Pharm Ther. 2017;42(2):155–164. doi: 10.1111/jcpt.12498. [DOI] [PubMed] [Google Scholar]
- 9.Jones EL, Oleson JR, Prosnitz LR, Samulski TV, Vujaskovic Z, Yu D, et al. Randomized trial of hyperthermia and radiation for superficial tumors. J Clin Oncol. 2005;23(13):3079–3085. doi: 10.1200/JCO.2005.05.520. [DOI] [PubMed] [Google Scholar]
- 10.Zagar TM, Higgins KA, Miles EF, Vujaskovic Z, Dewhirst MW, Clough RW, et al. Durable palliation of breast cancer chest wall recurrence with radiation therapy, hyperthermia, and chemotherapy. Radiotherapy and Oncology. 2010;97(3):535–540. doi: 10.1016/j.radonc.2010.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Datta NR, Puric E, Klingbiel D, Gomez S, Bodis S. Hyperthermia and Radiation Therapy in Locoregional Recurrent Breast Cancers: A Systematic Review and Meta-analysis. Int J Radiat Oncol Biol Phys. 2016;94(5):1073–1087. doi: 10.1016/j.ijrobp.2015.12.361. [DOI] [PubMed] [Google Scholar]
- 12.Overgaard J, Gonzalez Gonzalez D, Hulshof MC, Arcangeli G, Dahl O, Mella O, et al. Hyperthermia as an adjuvant to radiation therapy of recurrent or metastatic malignant melanoma. A multicentre randomized trial by the European Society for Hyperthermic Oncology. Int J Hyperthermia. 1996;12(1):3–20. doi: 10.3109/02656739609023685. [DOI] [PubMed] [Google Scholar]
- 13.Overgaard J, Gonzalez Gonzalez D, Hulshof MC, Arcangeli G, Dahl O, Mella O, et al. Randomized Trial of Hyperthermia as Adjuvant to Radiotherapy for Recurrent or Metastatic Malignant-Melanoma. Lancet. 1995;345(8949):540–543. doi: 10.1016/s0140-6736(95)90463-8. [DOI] [PubMed] [Google Scholar]
- 14.Eckert F, Braun LH, Traub F, Kopp HG, Sipos В, Lamprecht U, et al. Radiotherapy and hyperthermia with curative intent in recurrent high risk soft tissue sarcomas. Int J Hyperthermia. 2017:1–8. doi: 10.1080/02656736.2017.1369174. [DOI] [PubMed] [Google Scholar]
- 15.Issels RD, Lindner LH, Verweij J, Wust P, Reichardt P, Schem BC, et al. Neo-adjuvant chemotherapy alone or with regional hyperthermia for localised high-risk soft-tissue sarcoma: a randomised phase 3 multicentre study. Lancet Oncol. 2010;11(6):561–70. doi: 10.1016/S1470-2045(10)70071-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lutgens LC, Koper PC, Jobsen JJ, van der Steen-Banasik EM, Creutzberg CL, van den Berg HA, et al. Radiation therapy combined with hyperthermia versus cisplatin for locally advanced cervical cancer: Results of the randomized RADCHOC trial. Radiother Oncol. 2016;120(3):378–382. doi: 10.1016/j.radonc.2016.02.010. [DOI] [PubMed] [Google Scholar]
- 17.Datta NR, Rogers S, Klingbiel D, Gómez S, Puric E, Bodis S. Hyperthermia and radiotherapy with or without chemotherapy in locally advanced cervical cancer: a systematic review with conventional and network meta-analyses. International Journal of Hyperthermia. 2016;32(7):809–821. doi: 10.1080/02656736.2016.1195924. [DOI] [PubMed] [Google Scholar]
- 18.Harima Y, Ohguri T, Imada H, Sakurai H, Ohno T, Hiraki Y. A multicentre randomised clinical trial of chemoradiotherapy plus hyperthermia versus chemoradiotherapy alone in patients with locally advanced cervical cancer. Int J Hyperthermia. 2016;32(7):801–808. doi: 10.1080/02656736.2016.1213430. [DOI] [PubMed] [Google Scholar]
- 19.Diederich CJ, Wootton J, Prakash P, Salgaonkar V, Juang T, Scott S, et al. Catheter-based ultrasound hyperthermia with HDR brachytherapy for treatment of locally advanced cancer of the prostate and cervix. Proc SPIE Int Soc Opt Eng. 2011;7901:79010O. doi: 10.1117/12.876401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hurwitz MD, Hansen JL, Prokopios-Davos S, Manola J, Wang Q, Bornstein BA, et al. Hyperthermia Combined With Radiation for the Treatment of Locally Advanced Prostate Cancer Long-Term Results From Dana-Farber Cancer Institute Study 94–153. Cancer. 2011;117(3):510–516. doi: 10.1002/cncr.25619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hurwitz MD, Kaplan ID, Hansen JL, Prokopios-Davos S, Topulos GP, Wishnow K, et al. Hyperthermia combined with radiation in treatment of locally advanced prostate cancer is associated with a favourable toxicity profile. Int J Hyperthermia. 2005;21(7):649–56. doi: 10.1080/02656730500331967. [DOI] [PubMed] [Google Scholar]
- 22.Zwirner K, Bonomo P, Lamprecht U, Zips D, Gani C. External validation of a rectal cancer outcome prediction model with a cohort of patients treated with preoperative radiochemotherapy and deep regional hyperthermia. Int J Hyperthermia. 2017:1–6. doi: 10.1080/02656736.2017.1338364. [DOI] [PubMed] [Google Scholar]
- 23.Gani C, Schroeder C, Heinrich V, Spillner P, Lamprecht U, Berger B. Longterm local control and survival after preoperative radiochemotherapy in combination with deep regional hyperthermia in locally advanced rectal cancer. Int J Hyperthermia. 2016;32(2):187–92. doi: 10.3109/02656736.2015.1117661. [DOI] [PubMed] [Google Scholar]
- 24.Rasulov AO, Gordeyev SS, Barsukov YA, Tkachev SI, Malikhov AG, Balyasnikova SS, et al. Short-course preoperative radiotherapy combined with chemotherapy, delayed surgery and local hyperthermia for rectal cancer: a phase II study. Int J Hyperthermia. 2017;2:1–9. doi: 10.1080/02656736.2016.1272138. [DOI] [PubMed] [Google Scholar]
- 25.Chi MS, Yang KL, Chang YC, Ko HL, Lin YH, Huang SC. Comparing the Effectiveness of Combined External Beam Radiation and Hyperthermia Versus External Beam Radiation Alone in Treating Patients With Painful Bony Metastases: A Phase 3 Prospective, Randomized, Controlled Trial. Int J Radiat Oncol Biol Phys. 2017 doi: 10.1016/j.ijrobp.2017.09.030. [DOI] [PubMed] [Google Scholar]
- 26.Dewhirst MW, Vujaskovic Z, Jones E, Thrall D. Re-setting the biologic rationale for thermal therapy. Int J Hyperthermia. 2005;21(8):779–90. doi: 10.1080/02656730500271668. [DOI] [PubMed] [Google Scholar]
- 27.Diederich CJ. Thermal ablation and high-temperature thermal therapy: overview of technology and clinical implementation. Int J Hyperthermia. 2005;21(8):745–53. doi: 10.1080/02656730500271692. [DOI] [PubMed] [Google Scholar]
- 28.Chu KF, Dupuy DE. Thermal ablation of tumours: biological mechanisms and advances in therapy. Nat Rev Cancer. 2014;14(3):199–208. doi: 10.1038/nrc3672. [DOI] [PubMed] [Google Scholar]
- 29.Friedman M, Mikityansky I, Kam A, Libutti SK, Walther MM, Neeman Z, et al. Radiofrequency ablation of cancer. Cardiovasc Intervent Radiol. 2004;27(5):427–34. doi: 10.1007/s00270-004-0062-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Curley SA. Radiofrequency ablation of malignant liver tumors. Oncologist. 2001;6(1):14–23. doi: 10.1634/theoncologist.6-1-14. [DOI] [PubMed] [Google Scholar]
- 31.Brace CL. Radiofrequency and microwave ablation of the liver, lung, kidney, and bone: what are the differences? Curr Probl Diagn Radiol. 2009;38(3):135–43. doi: 10.1067/j.cpradiol.2007.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lencioni R, Crocetti L, Cioni D, Della Pina C, Bartolozzi C. Percutaneous radiofrequency ablation of hepatic colorectal metastases: technique, indications, results, and new promises. Invest Radiol. 2004;39(11):689–97. doi: 10.1097/00004424-200411000-00007. [DOI] [PubMed] [Google Scholar]
- 33.Zorbas G, Samaras T. A study of the sink effect by blood vessels in radiofrequency ablation. Comput Biol Med. 2015;57:182–6. doi: 10.1016/j.compbiomed.2014.12.014. [DOI] [PubMed] [Google Scholar]
- 34.Lubner MG, Brace CL, Hinshaw JL, Lee FT., Jr Microwave Tumor Ablation: Mechanism of Action, Clinical Results, and Devices. J Vasc Interv Radiol. 2010;21(8 Suppl):S192–203. doi: 10.1016/j.jvir.2010.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lee I, Kalkanis S, Hadjipanayis CG. Stereotactic Laser Interstitial Thermal Therapy for Recurrent High-Grade Gliomas. Neurosurgery. 2016;79(6):S24–S34. doi: 10.1227/NEU.0000000000001443. [DOI] [PubMed] [Google Scholar]
- 36.Missios S, Bekelis K, Barnett GH. Renaissance of laser interstitial thermal ablation. Neurosurg Focus. 2015;38(3):E13. doi: 10.3171/2014.12.FOCUS14762. [DOI] [PubMed] [Google Scholar]
- 37.Zhou YF. High intensity focused ultrasound in clinical tumor ablation. World J Clin Oncol. 2011;2(1):8–27. doi: 10.5306/wjco.v2.i1.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhang L, Wang ZB. High-intensity focused ultrasound tumor ablation: review of ten years of clinical experience. Front Med China. 2010;4(3):294–302. doi: 10.1007/s11684-010-0092-8. [DOI] [PubMed] [Google Scholar]
- 39.Maloney E, Hwang JH. Emerging HIFU applications in cancer therapy. Int J Hyperthermia. 2015;31(3):302–9. doi: 10.3109/02656736.2014.969789. [DOI] [PubMed] [Google Scholar]
- 40.Chatterjee DK, Diagaradjane P, Krishnan S. Nanoparticle-mediated hyperthermia in cancer therapy. Ther Deliv. 2011;02(8):1001–14. doi: 10.4155/tde.11.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Cherukuri P, Glazer ES, Curley SA. Targeted hyperthermia using metal nanoparticles. Adv Drug Deliv Rev. 2010;62(3):339–45. doi: 10.1016/j.addr.2009.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Singh R, Torti SV. Carbon nanotubes in hyperthermia therapy. Adv Drug Deliv Rev. 2013;65(15):2045–60. doi: 10.1016/j.addr.2013.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hervault A, Thanh NT. Magnetic nanoparticle-based therapeutic agents for thermo-chemotherapy treatment of cancer. Nanoscale. 2014;6(20):11553–73. doi: 10.1039/c4nr03482a. [DOI] [PubMed] [Google Scholar]
- 44.Peiris PM, Bauer L, Toy R, Tran E, Pansky J, Doolittle E, et al. Enhanced Delivery of Chemotherapy to tumors Using a Multicomponent Nanochain with Radio-Frequency-Tunable Drug Release. Acs Nano. 2012;6(5):4157–4168. doi: 10.1021/nn300652p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Toy R, Bauer L, Hoimes C, Ghaghada KB, Karathanasis E. Targeted nanotechnology for cancer imaging. Adv Drug Deliv Rev. 2014;76:79–97. doi: 10.1016/j.addr.2014.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Baumann BC, Kao GD, Mahmud A, Harada T, Swift J, Chapman C, et al. Enhancing the efficacy of drug-loaded nanocarriers against brain tumors by targeted radiation therapy. Oncotarget. 2013;4(1):64–79. doi: 10.18632/oncotarget.777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Maier-Hauff K, Ulrich F, Nestler D, Niehoff H, Wust P, Thiesen B, et al. Efficacy and safety of intratumoral thermotherapy using magnetic iron-oxide nanoparticles combined with external beam radiotherapy on patients with recurrent glioblastoma multiforme. J Neurooncol. 2011;103(2):317–324. doi: 10.1007/s11060-010-0389-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Johannsen M, Gneveckow U, Eckelt L, Feussner A, Waldöfner N, Scholz R, et al. Clinical hyperthermia of prostate cancer using magnetic nanoparticles: Presentation of a new interstitial technique. Int J Hyperthermia. 2005;21(7):637–47. doi: 10.1080/02656730500158360. [DOI] [PubMed] [Google Scholar]
- 49.Bañobre-López M, Teijeiro A, Rivasa J. Magnetic nanoparticle-based hyperthermia for cancer treatment. Rep Pract Oncol Radiother. 2013;18(6):397–400. doi: 10.1016/j.rpor.2013.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Nikfarjam M, Muralidharan V, Christophi C. Mechanisms of focal heat destruction of liver tumors. J Surg Res. 2005;127(2):208–23. doi: 10.1016/j.jss.2005.02.009. [DOI] [PubMed] [Google Scholar]
- 51.Fajardo LF, Egbert B, Marmor J, Hahn GM. Effects of Hyperthermia in a Malignant-Tumor. Cancer. 1980;45(3):613–623. doi: 10.1002/1097-0142(19800201)45:3<613::aid-cncr2820450331>3.0.co;2-e. [DOI] [PubMed] [Google Scholar]
- 52.Nishida T, Akagi K, Tanaka Y. Correlation between cell killing effect and cell membrane potential after heat treatment: Analysis using fluorescent dye and flow cytometry. International Journal of Hyperthermia. 1997;13(2):227–234. doi: 10.3109/02656739709012385. [DOI] [PubMed] [Google Scholar]
- 53.Ruifrok AC, Kanon B, Konings AW. Correlation between Cellular- Survival and Potassium-Loss in Mouse Fibroblasts after Hyperthermia Alone and after a Combined Treatment with X-Rays. Radiation Research. 1985;101(2):326–331. [PubMed] [Google Scholar]
- 54.Vidair CA, Dewey WC. Evaluation of a role for intracellular Na+, K+, Ca2+, and Mg2+ in hyperthermic cell killing. Radiat Res. 1986;105(2):187–200. [PubMed] [Google Scholar]
- 55.Willis WT, Jackman MR, Bizeau ME, Pagliassotti MJ, Hazel JR. Hyperthermia impairs liver mitochondrial function in vitro. Am J Physiol Regul Integr Comp Physiol. 2000;278(5):R1240–6. doi: 10.1152/ajpregu.2000.278.5.R1240. [DOI] [PubMed] [Google Scholar]
- 56.Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell. 2014;14(3):275–91. doi: 10.1016/j.stem.2014.02.006. [DOI] [PubMed] [Google Scholar]
- 57.Warters RL, Roti Roti JL. Hyperthermia and the cell nucleus. Radiat Res. 1982;92(3):458–62. [PubMed] [Google Scholar]
- 58.Krawczyk PM, Eppink B, Essers J, Stap J, Rodermond H, Odijk H, et al. Mild hyperthermia inhibits homologous recombination, induces BRCA2 degradation, and sensitizes cancer cells to poly (ADP-ribose) polymerase-1 inhibition. Proc Natl Acad Sci U S A. 2011;108(24):9851–6. doi: 10.1073/pnas.1101053108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Oei AL, Vriend LEM, Crezee J, Franken NAP, Krawczyk PM. Effects of hyperthermia on DNA repair pathways: one treatment to inhibit them all. Radiat Oncol. 2015;10:165. doi: 10.1186/s13014-015-0462-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Milleron RS, Bratton SB. Heat shock induces apoptosis independently of any known initiator caspase-activating complex. J Biol Chem. 2006;281(25):16991–7000. doi: 10.1074/jbc.M512754200. [DOI] [PubMed] [Google Scholar]
- 61.Morlé A, Garrido C, Micheau O. Hyperthermia restores apoptosis induced by death receptors through aggregation-induced c-FLIP cytosolic depletion. Cell Death Dis. 2015;6:e1633. doi: 10.1038/cddis.2015.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Vertrees RA, Das GC, Coscio AM, Xie J, Zwischenberger JB, Boor PJ. A mechanism of hyperthermia-induced apoptosis in ras-transformed lung cells. Molecular Carcinogenesis. 2005;44(2):111–121. doi: 10.1002/mc.20124. [DOI] [PubMed] [Google Scholar]
- 63.Skibba JL, Quebbeman EJ, Kalbfleisch JH. Nitrogen-Metabolism and Lipid-Peroxidation during Hyperthermic Perfusion of Human Livers with Cancer. Cancer Research. 1986;46(11):6000–6003. [PubMed] [Google Scholar]
- 64.Wang Z, Cai F, Chen X, Luo M, Hu L, Lu Y. The Role of Mitochondria-Derived Reactive Oxygen Species in Hyperthermia-Induced Platelet Apoptosis. Plos One. 2013;8(9) doi: 10.1371/journal.pone.0075044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.El-Orabi NF, Rogers CB, Edwards HG. Schwartz DD. Heat-induced inhibition of superoxide dismutase and accumulation of reactive oxygen species leads to HT-22 neuronal cell death. Journal of Thermal Biology. 2011;36(1):49–56. [Google Scholar]
- 66.Dörthe M. Katschinski, Kristina Boos, Susann G. Schindler, Joachim Fandrey. Pivotal role of reactive oxygen species as intracellular mediators of hyperthermia-induced apoptosis. Journal of Biological Chemistry. 2000;275(28):21094–21098. doi: 10.1074/jbc.M001629200. [DOI] [PubMed] [Google Scholar]
- 67.Matés JM, Sánchez-Jiménez FM. Role of reactive oxygen species in apoptosis: implications for cancer therapy. Int J Biochem Cell Biol. 2000;32(2):157–170. doi: 10.1016/s1357-2725(99)00088-6. [DOI] [PubMed] [Google Scholar]
- 68.Song CW. Effect of local hyperthermia on blood flow and microenvironment: a review. Cancer Res. 1984;44:4721s–4730s. [PubMed] [Google Scholar]
- 69.Sun X, Xing L, Ling CC, Li GC. The effect of mild temperature hyperthermia on tumour hypoxia and blood perfusion: relevance for radiotherapy, vascular targeting and imaging. Int J Hyperthermia. 2010;26(3):224–31. doi: 10.3109/02656730903479855. [DOI] [PubMed] [Google Scholar]
- 70.Song CW, Park HJ, Lee CK, Griffin R. Implications of increased tumor blood flow and oxygenation caused by mild temperature hyperthermia in tumor treatment. Int J Hyperthermia. 2005;21(8):761–767. doi: 10.1080/02656730500204487. [DOI] [PubMed] [Google Scholar]
- 71.Song CW, Kang MS, Rhee JG, Levitt SH. The effect of hyperthermia on vascular function, pH, and cell survival. Radiology. 1980;137(3):795–803. doi: 10.1148/radiology.137.3.7444064. [DOI] [PubMed] [Google Scholar]
- 72.Sun X, Li XF, Russell J, Xing L, Urano M, Li GC, et al. Changes in tumor hypoxia induced by mild temperature hyperthermia as assessed by dual-tracer immunohistochemistry. Radiother Oncol. 2008;88(2):269–276. doi: 10.1016/j.radonc.2008.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Evans SS, Wang WC, Bain MD, Burd R, Ostberg JR, Repasky EA. Fever-range hyperthermia dynamically regulates lymphocyte delivery to high endothelial venules. Blood. 2001;97(9):2727–2733. doi: 10.1182/blood.v97.9.2727. [DOI] [PubMed] [Google Scholar]
- 74.Muthana M, Multhoff G, Pockley AG. Tumour infiltrating host cells and their significance for hyperthermia. Int J Hyperthermia. 2010;26(3):247–255. doi: 10.3109/02656730903413375. [DOI] [PubMed] [Google Scholar]
- 75.den Brok MH, Sutmuller RP, van der Voort R, Bennink EJ, Figdor CG, Ruers TJ, et al. In situ tumor ablation creates an antigen source for the generation of antitumor immunity. Cancer Research. 2004;64(11):4024–4029. doi: 10.1158/0008-5472.CAN-03-3949. [DOI] [PubMed] [Google Scholar]
- 76.Mikucki ME, Fisher DT, Ku AW, Appenheimer MM, Muhitch JB, Evans SS. Preconditioning thermal therapy: Flipping the switch on IL-6 for antitumour immunity. Int J Hyperthermia. 2013;29(5):464–473. doi: 10.3109/02656736.2013.807440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ali MY, Grimm CF, Ritter M, Mohr F, Allgaier HP, Weth R, et al. Activation of dendritic cells by local ablation of hepatocellular carcinoma. J Hepatol. 2005;43(5):817–22. doi: 10.1016/j.jhep.2005.04.016. [DOI] [PubMed] [Google Scholar]
- 78.Hausner PF. Image-guided thermal ablation of tumors increases the plasma level of interleukin-6 and interleukin-10. J Vasc Interv Radiol. 2013;24(8):1105–12. doi: 10.1016/j.jvir.2013.05.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Milani V, Noessner E, Ghose S, Kuppner M, Ahrens B, Scharner A, et al. Heat shock protein 70: role in antigen presentation and immune stimulation. Int J Hyperthermia. 2002;18(6):563–75. doi: 10.1080/02656730210166140. [DOI] [PubMed] [Google Scholar]
- 80.Multhoff G, Hightower LE. Hightower Cell surface expression of heat shock proteins and the immune response. Cell Stress Chaperones. 1996;1(3):167–76. doi: 10.1379/1466-1268(1996)001<0167:cseohs>2.3.co;2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Frey B, Weiss EM, Rubner Y, Wunderlich R, Ott OJ, Sauer R, et al. Old and new facts about hyperthermia-induced modulations of the immune system. Int J Hyperthermia. 2012;28(6):528–42. doi: 10.3109/02656736.2012.677933. [DOI] [PubMed] [Google Scholar]
- 82.Srivastava PK, Udono H, Blachere NE, Li Z. Heat shock proteins transfer peptides during antigen processing and CTL priming. Immunogenetics. 1994;39(2):93–8. doi: 10.1007/BF00188611. [DOI] [PubMed] [Google Scholar]
- 83.Carper SW, Duffy JJ, Gerner EW. Heat shock proteins in thermotolerance and other cellular processes. Cancer Res. 1987;47(20):5249–55. [PubMed] [Google Scholar]
- 84.Landry J, Chrétien P, Bernier D, Nicole LM, Marceau N, Tanguay RM. Thermotolerance and heat shock proteins induced by hyperthermia in rat liver cells. Int J Radiat Oncol Biol Phys. 1982;8(1):59–62. doi: 10.1016/0360-3016(82)90385-6. [DOI] [PubMed] [Google Scholar]
- 85.Fanneau D, Brunet M, Frisan E, Solary E, Fontenay M, Garrido C. Heat shock proteins: essential proteins for apoptosis regulation. J Cell Mol Med. 2008;12(3):743–761. doi: 10.1111/j.1582-4934.2008.00273.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Sreedhar AS, Csermely P. Heat shock proteins in the regulation of apoptosis: new strategies in tumor therapy: a comprehensive review. Pharmacol Ther. 2004;101(3):227–57. doi: 10.1016/j.pharmthera.2003.11.004. [DOI] [PubMed] [Google Scholar]
- 87.Beere HM. The stress of dying”: the role of heat shock proteins in the regulation of apoptosis. J Cell Sci. 2004;117:2641–51. doi: 10.1242/jcs.01284. [DOI] [PubMed] [Google Scholar]
- 88.van der Zee J. Heating the patient: a promising approach? Ann Oncol. 2002;13(8):1173–84. doi: 10.1093/annonc/mdf280. [DOI] [PubMed] [Google Scholar]
- 89.Sharma S, Sandhu AP, Patel FD, Ghoshal S, Gupta BD, Yadav NS. Side- Effects of Local Hyperthermia - Results of a Prospectively Randomized Clinical-Study. Int J Hyperthermia. 1990;6(2):279–285. doi: 10.3109/02656739009141138. [DOI] [PubMed] [Google Scholar]
- 90.Izukura R, Imada H, Hashiguchi N, Sawatari H, Ohguri T, Miyazono M, et al. Cardiac and respiratory effects of deep regional hyperthermia using an 8MHz radiofrequency-capacitive device on patients with cancer. International Journal of Hyperthermia. 2017;33(4):428–434. doi: 10.1080/02656736.2017.1283064. [DOI] [PubMed] [Google Scholar]
- 91.Borovski T, De Sousa E, Melo F, Vermeulen F, Medema JP. Cancer stem cell niche: the place to be. Cancer Res. 2011;71(3):634–9. doi: 10.1158/0008-5472.CAN-10-3220. [DOI] [PubMed] [Google Scholar]
- 92.Plaks V, Kong N, Werb Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 2015;16(3):225–38. doi: 10.1016/j.stem.2015.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756–760. doi: 10.1038/nature05236. [DOI] [PubMed] [Google Scholar]
- 94.Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 2009;458(7239):780–3. doi: 10.1038/nature07733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zhou S, Schuetz JD, Bunting KD, Colapietro AM, Sampath J, Morris JJ, et al. The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med. 2001;7(9):1028–34. doi: 10.1038/nm0901-1028. [DOI] [PubMed] [Google Scholar]
- 96.Golebiewska A, Brons NH, Bjerkvig R, Niclou SP. Critical appraisal of the side population assay in stem cell and cancer stem cell research. Cell Stem Cell. 2011;8(2):136–47. doi: 10.1016/j.stem.2011.01.007. [DOI] [PubMed] [Google Scholar]
- 97.Zhao J. Cancer stem cells and chemoresistance: The smartest survives the raid. Pharmacol Ther. 2016;160:145–58. doi: 10.1016/j.pharmthera.2016.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Konopleva M, Zhao S, Hu W, Jiang S, Snell V, Weidner D, et al. The anti-apoptotic genes Bcl-X(L) and Bcl-2 are over-expressed and contribute to chemoresistance of non-proliferating leukaemic CD34+ cells. Br J Haematol. 2002;118(2):521–34. doi: 10.1046/j.1365-2141.2002.03637.x. [DOI] [PubMed] [Google Scholar]
- 99.Wang J, Wakeman TP, Fathia JD, Hjelmeland AB, Wang XF, White RR, et al. Notch promotes radioresistance of glioma stem cells. Stem Cells. 2010;28(1):17–28. doi: 10.1002/stem.261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Todaro M, Alea MP, Di Stefano AB, Cammareri P, Vermeulen F, Iovino F, et al. Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4. Cell Stem Cell. 2007;1(4):389–402. doi: 10.1016/j.stem.2007.08.001. [DOI] [PubMed] [Google Scholar]
- 101.Zhao C, Blum J, Chen A, Kwon HY, Jung SH, Cook JM, et al. Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell. 2007;12(6):528–41. doi: 10.1016/j.ccr.2007.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Chen J, Li Y, Yu T, McKay RM, Burns DK, Kernie SG, et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012;488(7412):522–6. doi: 10.1038/nature11287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Saito Y, Uchida N, Tanaka S, Suzuki N, Tomizawa-Murasawa M, Sone A, et al. Induction of cell cycle entry eliminates human leukemia stem cells in a mouse model of AML. Nat Biotechnol. 2010;28(3):275–80. doi: 10.1038/nbt.1607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Liu L, Wise DR, Diehl JA, Simon MC. Hypoxic reactive oxygen species regulate the integrated stress response and cell survival. J Biol Chem. 2008;283(45):31153–62. doi: 10.1074/jbc.M805056200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Harrison H, Rogerson L, Gregson HJ, Brennan KR, Clarke RB, Landberg G. Contrasting hypoxic effects on breast cancer stem cell hierarchy is dependent on ER-alpha status. Cancer Res. 2013;73(4):1420–33. doi: 10.1158/0008-5472.CAN-12-2505. [DOI] [PubMed] [Google Scholar]
- 106.Malanchi I, Santamaria-Martínez A, Susanto E, Peng H, Lehr HA, Delaloye JF, et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 2011;481(7379):85–9. doi: 10.1038/nature10694. [DOI] [PubMed] [Google Scholar]
- 107.Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton F, Chan E, et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature. 2012;487(7408):505–9. doi: 10.1038/nature11249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Wierenga PK, Setroikromo R, Kamps G, Kampinga HH, Vellenga E. Differences in heat sensitivity between normal and acute myeloid leukemic stem cells: Feasibility of hyperthermic purging of leukemic cells from autologous stem cell grafts. Experimental Hematology. 2003;31(5):421–427. doi: 10.1016/s0301-472x(03)00049-3. [DOI] [PubMed] [Google Scholar]
- 109.Burke AR, Singh RN, Carroll DL, Wood JC, DAgostino RB, Jr, Ajayan PM, et al. The resistance of breast cancer stem cells to conventional hyperthermia and their sensitivity to nanoparticle-mediated photothermal therapy. Biomaterials. 2012;33(10):2961–70. doi: 10.1016/j.biomaterials.2011.12.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Sadhukha T, Niu L, Wiedmann TS, Panyam J. Effective elimination of cancer stem cells by magnetic hyperthermia. Mol Pharm. 2013;10(4):1432–41. doi: 10.1021/mp400015b. [DOI] [PubMed] [Google Scholar]
- 111.Atkinson RL, Zhang M, Diagaradjane P, Sirisha P, Contreras A, Hilsenbeck SG, et al. Thermal enhancement with optically activated gold nanoshells sensitizes breast cancer stem cells to radiation therapy. Sci Transl Med. 2010;2(55):55ra79. doi: 10.1126/scitranslmed.3001447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Man J, Shoemake JD, Ma T, Rizzo AE, Godley AR, Wu Q, et al. Hyperthermia Sensitizes Glioma Stem-like Cells to Radiation by Inhibiting AKT Signaling. Cancer Res. 2015;75(8):1760–9. doi: 10.1158/0008-5472.CAN-14-3621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Lee H, Park HJ, Park CS, Oh ET, Choi BH, Williams B, et al. Response of breast cancer cells and cancer stem cells to metformin and hyperthermia alone or combined. PLoS One. 2014;9(2):e87979. doi: 10.1371/journal.pone.0087979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Wang CH, Chiou SH, Chou CP, Chen YC, Huang YJ, Peng CA. Photothermolysis of glioblastoma stem-like cells targeted by carbon nanotubes conjugated with CD133 monoclonal antibody. Nanomedicine. 2011;7(1):69–79. doi: 10.1016/j.nano.2010.06.010. [DOI] [PubMed] [Google Scholar]